Protein Protein Interactions Methods and
Protein Protein Interactions Methods and
Protein Protein Interactions Methods and
Cheryl L. Meyerkord
Haian Fu
Editors
Protein-Protein
Interactions
Methods and Applications
Second Edition
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Second Edition
Edited by
Cheryl L. Meyerkord
Department of Pharmacology and Emory Chemical Biology Discovery Center,
Emory University School of Medicine, Atlanta, GA, USA
Haian Fu
Department of Pharmacology, Department of Hematology & Medical
Oncology, and Emory Chemical Biology Discovery Center, Emory University
School of Medicine, Atlanta, GA, USA
Editors
Cheryl L. Meyerkord Haian Fu
Department of Pharmacology Department of Pharmacology
and Emory Chemical Biology Department of Hematology & Medical
Discovery Center, Emory University Oncology, and Emory Chemical Biology
School of Medicine Discovery Center, Emory University
Atlanta, GA, USA School of Medicine
Atlanta, GA, USA
v
vi Preface
Our hope is that this book will serve as a valuable resource for all readers, from students
to experienced scientists. In the chapters of this book, experienced researchers describe in
detail the underlying theory and practical application of widely used methods for monitor-
ing protein-protein interactions. In keeping with the series tradition, the Notes section of
the chapters contains valuable explanations to sensitive procedures and potential pitfalls,
with tips on how to avoid them. This book is expected to empower readers in their quest to
elucidate the mechanisms of protein-protein interactions and the role of these interactions
in diverse biological processes, and to target protein-protein interactions for therapeutic
discovery.
We are grateful to the contributors of each chapter for their effort, commitment, and
willingness to share their knowledge and valuable experience with the scientific commu-
nity. We sincerely thank the series editor, John Walker, for all of his guidance. Together, we
present an extensively updated and expanded version of this valuable resource to all those
who study protein-protein interactions.
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
PART I OVERVIEWS
Index. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 615
Contributors
xi
xii Contributors
SAM LIEVENS Department of Medical Protein Research, VIB, Ghent, Belgium; Department
of Biochemistry, Faculty of Medicine and Health Sciences, Ghent University, Ghent,
Belgium
LIU LIU Comprehensive Cancer Center and Departments of Internal Medicine,
Pharmacology and Medicinal Chemistry, University of Michigan, Ann Arbor, MI, USA
ADAM I. MARCUS Department of Hematology and Medical Oncology, Winship Cancer
Institute of Emory University, Atlanta, GA, USA
ALEXA L. MATTHEYSES Department of Cell Biology, Emory University School of Medicine,
Atlanta, GA, USA
ALISON R. MERCER-SMITH Department of Chemistry, Pomona College, Claremont,
CA, USA
SAMY O. MEROUEH Department of Biochemistry and Molecular Biology, Center for
Computational Biology and Bioinformatics, Indiana University School of Medicine,
Indianapolis, IN, USA; Department of Chemistry and Chemical Biology, Indiana
University Purdue University Indianapolis, Indianapolis, IN, USA
CHERYL L. MEYERKORD Department of Pharmacology, Emory University School
of Medicine, Atlanta, GA, USA; Emory Chemical Biology Discovery Center,
Emory University, Atlanta, GA, USA
STEPHEN W. MICHNICK Département de Biochimie, Université de Montréal, Montréal,
QC, Canada
ASHUTOSH MISHRA St. Jude Proteomics Facility, St. Jude Children’s Research Hospital,
Memphis, TN, USA
TOSHIYUKI MIYASHITA Department of Molecular Genetics, Kitasato University School
of Medicine, Sagamihara, Japan
JEREMY MOGRIDGE Department of Laboratory Medicine and Pathology, University
of Toronto, Toronto, ON, Canada
TERRY W. MOORE Department of Chemistry, University of Illinois at Urbana-
Champaign, Urbana, IL, USA; Department of Medicinal Chemistry and
Pharmacognosy, University of Illinois Hospital and Health Sciences System Cancer Center,
Chicago, IL, USA
ZANETA NIKOLOVSKA-COLESKA Department of Pathology, University of Michigan
Medical School, Ann Arbor, MI, USA
VISHWAJEETH R. PAGALA St. Jude Proteomics Facility, St. Jude Children’s Research
Hospital, Memphis, TN, USA
NELI I. PANOVA-ELEKTRONOVA Department of Pharmacology and Toxicology, The
University of Texas Medical Branch, Galveston, TX, USA
JUNMIN PENG St. Jude Proteomics Facility, Department of Structural Biology,
Department of Neurodevelopmental Biology, St. Jude Children’s Research Hospital,
Memphis, TN, USA
SAMUEL J. PFAFF Small Molecule Discovery Center, Department of Pharmaceutical
Chemistry, University of California San Francisco, San Francisco, CA, USA
CAU D. PHAM Department of Pharmacology, Emory University School of Medicine,
Atlanta, GA, USA
MARY C. PUCKETT Department of Pharmacology, Emory University School of Medicine,
Atlanta, GA, USA
RONALD T. RAINES Department of Biochemistry, University of Wisconsin-Madison,
Madison, WI, USA; Department of Chemistry, University of Wisconsin-Madison,
Madison, WI, USA
xiv Contributors
Overviews
Chapter 1
Abstract
Regulated interactions between proteins govern signaling pathways within and between cells. Structural
studies on protein complexes formed reversibly and/or transiently illustrate the remarkable diversity of
interactions, both in terms of interfacial size and nature. In recent years, “domain–peptide” interactions
have gained much greater recognition and may be viewed as both pre-translational and posttranslational-
dependent functional switches. Our understanding of the multistep regulation of auto-inhibited multi-
domain proteins has also grown. Their activity may be understood as the “combinatorial” output of
multiple input signals, including phosphorylation, location, and mechanical force. The prospects for
bridging the gap between the new “systems biology” data and the traditional “reductionist” data are also
discussed.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_1, © Springer Science+Business Media New York 2015
3
4 Robert C. Liddington
actually optimal for larger particles (>500 kDa) and can provide
“sub-molecular” (5–15 Å resolution) maps or envelopes [21], into
which atomic models can be fitted with high overall precision in
the case of rigid-body docking, or when quaternary reorganization
of domains occur [22]. Another major technique is Small-Angle
X-ray Scattering (SAXS), which is performed in solution and can
discriminate (with high precision) between competing models of
quaternary organization [23]. Many other complementary techni-
ques are discussed in Parts II, III, and IV of this book.
In this chapter, I begin by classifying and describing the proper-
ties of binary complexes, and then review the regulatory mechan-
isms of binary interactions as well as large multidomain signaling
proteins. While the diversity of interactions may be finite, we should
continue to expect surprises. For example, over the past 10 years,
the profound role of “domain–peptide” interactions in higher
eukaryotes (arising from “pre-translational modification”) has
been established [24]; and giant steps have been made in defining
the roles of mechanical force, which can be considered as an addi-
tional (and often decisive) allosteric effector in many signaling
pathways [25]. Clearly, much remains to be learned.
3.1 The Standard Most domain–domain interfaces have a typical layout, which
Patch defines a standard “patch” (Fig. 2a). A central solvent-excluded
region (“core”) is surrounded by a partly buried outer ring (“rim”)
that includes water-mediated interactions. The core, on average,
Fig. 2 The standard patch and interface size. (a) Slice through an ideal domain–domain interfacial “patch”
(side view). All the atoms that lose full or partial accessibility to solvent when ligand binds are interface atoms.
Dark-shaded atoms, which were solvent-exposed but become fully buried constitute the core, which
is surrounded by a partly buried rim that forms a water-tight “O-ring.” “Hot spot” residues typically comprise
a subset of the core (see text). (b) Distribution of buried surface area (Å2) among a selection of mostly
reversible heterodimeric complexes, including transient redox complexes, signaling, enzyme–inhibitor, and
antibody–antigen complexes. Interfaces larger than 2,000 Å generally comprise more than one patch. Adapted
from ref. 35
8 Robert C. Liddington
contributes ~75 % of the buried surface area and the majority of the
binding energy. It is typically more hydrophobic than the surface in
general but less than the interior of the protein. The rim has a
similar composition to the protein surface; it contributes ~25 % of
the buried surface, and its major role is thought to be the exclusion
of water molecules from the core, analogous to the “O-ring” model
proposed by Bogan and Thorn [38]. Good shape and charge
complementarity is important, such that the packing density is
not very different from the interior of the protein. Most interfaces
are rather flat, with a typical RMS deviation from planarity of ~2 Å.
The interfacial “patch” varies in size, but most are in the range of
1,200–2,000 Å2 (Fig. 2b). Interfaces larger than this typically
comprise two or more patches.
Bringing two hydrophobic surfaces into close apposition in
water is generally favorable (the “hydrophobic effect”), although
its physical basis is still debated (see Chapters 2, 4, and 7). An
entropic origin, arising from the release of water molecules into
bulk solvent, is often cited; but careful thermodynamic measure-
ments of mutated interfaces point to an enthalpic driving force;
moreover, the free energy change (ΔΔG) scales linearly with the
change in buried surface, ~20 cal/Å2 at the rim and ~45 cal/Å2 at
the center of the core [39]. A minimal interface of 1,200 Å2 would
therefore equate to a free energy of ~4 kcal/mol or a Kd ~ 1 μM.
For a 2,000 Å2 interface, this estimate rises to Kd ~ 10 nM. Both
values are commensurate with the local concentrations of interact-
ing proteins in vivo.
A priori, the energetics of polar interactions at interfaces are
much harder to assess, because complex formation involves the
displacement of bound water molecules from both sides of the
interface (gain in entropy, loss in enthalpy), which must be com-
pensated by a new set of ionic, polar, and van der Waals contacts
between the protein partners (gain in enthalpy, loss in entropy), so
that the net energetic effect of polar interactions may be positive,
negative, or negligible. However, a simplifying feature may be the
presence of energetic “hot spots” (see below).
4.1 Residue Count Perhaps the simplest method of describing an interface is to count
the number of residues of a given type, and express them as a
percentage of the total. Sometimes, residues counts are weighted
by their size (i.e., their Å2 contribution to the buried surface).
Recently, Levy and colleagues collated a comprehensive “mixed”
set of protein-protein interfaces from human, yeast, and E. coli
[41]. Figure 3a plots, for human complexes, the relative incidence
Fig. 3 Residue usage at interfaces. (a) Residues are color-coded by type: hydrophobic (black); polar (orange);
charged (positive ¼ blue, negative ¼ red); or aromatic (green), and sorted within their groups by contribution
to interface (blue bars) and surface (gray bars (stacked)). Average interface incidence by type is given as %
below residues. Values above bars are total contributions for each type. Plot derived from data on human
complexes [41]. (b) The difference in residue incidence between reversible and irreversible heterodimeric
complexes. Bars above zero indicate an increase in residue usage at reversible interfaces. An enrichment in
charged and aromatic residues at the expense of hydrophobic and small polar residues is evident. Plot derived
from data in ref. [40]. (c) Residues ordered by increasing “Residue Propensity,” p (red bars; scale on right
ordinate), where p ¼ ln (fi/fs) (see main text). Blue bars (scale on left ordinate) show the actual incidence (%)
of residues at the core. Plot derived from data in ref. [41]. (d) Residues ranked in decreasing order of their
energetic contribution to hot spots (vertical bars), based on a data set of >2,000 alanine mutations [38]. Also
indicated, as connecting brackets (numbers refer to boxed inset), are the most common pairwise interactions
reported for a large set of reversible heterodimeric interfaces, ranked by incidence at the core [44]. Refer to
Table 1 for further information
10 Robert C. Liddington
4.3 Residue Pairs A third method focuses on the frequency with which specific pairs
of residues interact across interfaces [43]. In a recent study of a
diverse set of reversible heterodimeric complexes [44], with inter-
faces ranging from 700 to 8,500 Å2 and an average size of
2,100 Å2, the results are quite striking. The authors also divided
residue distribution into categories of total interface and core,
making the results more directly comparable with the approaches
above. The ten most abundant interactions are listed in Table 1 and
Structural Basis 11
Table 1
Common pairwise interactions at interfaces
Rank
Residue
Core Interface pair Interaction type(s)
1 1 Arg–Glu Mostly salt bridges
2 3 Lys–Asp Mostly salt bridges
3 9 Trp–Arg Π–cation (75 %)
4 5 Tyr–Arg Π–cation (40 %); s/c H-bond 40 %
5 – Phe–Leu Hydrophobic s/c packing
6 7 Tyr–Lys s/c H-bond (55 %); s/c-m/c (33 %)
7 6 Tyr–Asn Π–cation (15 %); s/c H-bond (55 %)
8 – Trp–Ile Hydrophobic s/c packing
9 2 Arg–Asp Mostly salt bridges
– 4 Lys–Glu Mostly salt bridges
The table lists the top ten most common pairwise interactions in a large collection
of reversible heterodimeric complexes [44]. They are ranked according to their incidence
at the core. Incidence at the interface is also shown. Residues are color-coded as in Fig. 3.
s/c side chain, m/c main chain, H-bond hydrogen bond
Fig. 3d. All four charge-charge pairs are present; two of them
(Arg–Glu and Lys–Asp) are highly abundant at both the rim and
core, while the other two are abundant at the rim but not the core.
The other six pairs involve aromatic residues: two of them involve
purely hydrophobic side-chain packing (Phe–Leu and Trp–Leu),
and are restricted to the core; the other four are polar interactions,
three of which are often mediated by Π–cation bonding (see
below).
4.4 Hot Spots and A fourth descriptor of interfaces focuses on the energetics of the
the Special Role of interaction and relies on the results of alanine scanning mutagene-
Π–Cation Interactions sis. Thus, while many mutations at the interface have a limited
effect on binding (ΔΔG), certain clusters, called “hot spots” [38],
have a large/dominant effect. Hot spots are nearly always buried
near the center of the core and well shielded from solvent. They
pack against hot spots on their cognate domains, forming an intri-
cate cooperative network of interactions. Sequence conservation
(between orthologous complexes) is highest at hot spots [45].
Residue preferences at hot spots are a subset of those described
in the previous section (Fig. 3d). Trp, Arg, and Tyr are the most
common, accounting for more than half of the total. These three
residues are versatile in being able to form hydrophobic, aromatic,
12 Robert C. Liddington
A very useful starting point for probing the linkage between com-
plex formation and conformational changes is the “benchmark”
collection of high-resolution heterodimeric reversible complexes
last updated in 2010 [48]. The set is restricted to those complexes
for which structures of both isolated components are also known. A
plot of the RMS change in structure of the component domains
upon complex formation versus interface size points first to a tight
cluster with differences of ~1 0.5 Å; these approximate to “rigid-
body” docking, and account for about half of the total set (colored
red in Fig. 4a). Most of these interfaces conform to the standard
“patch” model described above. Large RMS differences (3–7 Å) are
almost exclusively associated with reversible signaling complexes, in
which conformational changes, including disorder–order transi-
tions and quaternary changes, are commonplace.
A particularly well-studied example is the interactions between
the small G-proteins (GTPases) and their effectors, activators, and
inhibitors (reviewed in ref. [49]). For example, guanine nucleotide
exchange factors (“GEFs”), which activate small G-proteins by
recognizing the inactive GDP-bound form and promoting GDP
release and GTP binding, show a great variety in their structures
and modes of interaction, and several distinct nucleotide exchange
mechanisms (Fig. 4b). And while some GEFs are specific for a
single G-protein, others are quite promiscuous. Although they all
utilize at least part of one or both of the “switch” regions (regions
that undergo conformational changes upon hydrolysis of GTP),
different GEFs bind to different parts of the G-protein surface and
display a large range of interface sizes that extend well beyond the
“rigid-body” set.
Structural Basis 13
Fig. 4 Conformational changes and complex formation. (a) RMS change in protein structure following complex
formation, as a function of buried surface area. Red diamonds approximate to “rigid-body” docking. Black
diamonds are typically signaling complexes. See main text for details. Based on data from ref. 48. (b) The
remarkable variety of interactions made between small G proteins (shown in gray, in approximately the same
orientation) and their GTP Exchange Factors (GEFs). All GEFs recognize the GDP-bound form by binding to at
least one of the switch regions of the G protein (highlighted in red). The complexes shown are: 1 Ras:SOS,
2 Sec4p:Sec2p, 3 Cdc42:Dock9, 4 Rab21:Rabex5, 5 Cdc42:Dbs, 6 Rab35:DENN1B, 7 Rab8:MSS4, 8 Rop4:
RopGEF8, 9 Ypt1p:TRAPP1, 10 Arf1:Gea2, 11 Ran:RCC1. Adapted from ref. 49, with permission
Fig. 6 Homologous domains may bind the same ligand but at different locations. Both domains are members
of the same Pfam family (vWF A domain). The five central β-strands superpose with an RMS difference of
0.69 Å for 35 Cα positions. At right, the integrin α2 “I” domain engages triple-helical collagen via the MIDAS
motif (labeled “M”) at the “top” of the domain (see Fig. 5). At left, the von Willebrand Factor A3 domain lacks
one of the metal-coordinating residues that comprise the MIDAS motif, and does not bind metal (PDB code
4DMU [58]). It also binds collagen, but on the “front” face of the A domain, and no conformational changes are
observed
9 Concluding Remarks
how cells work. We must learn the nature of each switch (e.g.,
phosphorylation, phospholipid-binding, mechanical stress), and
precisely how each of the binding functions or catalytic activity of
each protein is turned on/off by each switch, both singly and in
combination.
But this is only the end of the beginning. Next, we need to
observe multiprotein complexes in their native environment (see
Part IV). Recent advances in EM (“Tomography”) have enabled
direct visualization of protein complexes at molecular resolution
(20–60 Å) in flash-frozen (fixed, but unstained/undamaged) thin
cell sections [90]. And finally, cells are most definitely not static
structures: we must observe single molecules as they go about
their business in living cells. For example, during cell migration,
multiprotein complexes and higher subsystems (e.g., the cytoske-
leton–membrane–matrix linkage) assemble, work, and then disas-
semble, on a rapid timescale. Ongoing developments that combine
light microcopy of living cells (using fluorescently labeled protein)
with electron tomography of the same (subsequently frozen) cell
(termed “correlative microscopy”) provide a glimpse into a future
in which we may truly begin to understand how protein-protein
interactions drive the organization and dynamics of living
cells [91].
References
1. Schreiber G, Keating AE (2011) Protein bind- viruses from electron cryo-microscopy. Curr
ing specificity versus promiscuity. Curr Opin Opin Struct Biol 21:265–273
Struct Biol 21:50–61 9. Almo SC, Garforth SJ, Hillerich BS et al
2. Levy ED, De S, Teichmann SA (2012) Cellular (2013) Protein production from the structural
crowding imposes global constraints on the genomics perspective: achievements and future
chemistry and evolution of proteomes. Proc needs. Curr Opin Struct Biol 23:335–344
Natl Acad Sci U S A 109:20461–20466 10. Cuff AL, Sillitoe I, Lewis T et al (2011)
3. Finn RD, Tate J, Mistry J et al (2008) The Extending CATH: increasing coverage of the
Pfam protein families database. Nucleic Acids protein structure universe and linking structure
Res 36:D281–D288 with function. Nucleic Acids Res 39:
4. Mosca R, Ceol A, Stein A et al (2014) 3did: a D420–D426
catalog of domain-based interactions of known 11. Kundrotas PJ, Vakser IA, Janin J (2013) Struc-
three-dimensional structure. Nucleic Acids Res tural templates for modeling homodimers.
42:D374–D379 Protein Sci 22:1655–1663
5. Stumpf MP, Thorne T, de Silva E et al (2008) 12. Kundrotas PJ, Zhu Z, Janin J et al (2012)
Estimating the size of the human interactome. Templates are available to model nearly all
Proc Natl Acad Sci U S A 105:6959–6964 complexes of structurally characterized pro-
6. Venkatesan K, Rual JF, Vazquez A et al (2009) teins. Proc Natl Acad Sci U S A 109:
An empirical framework for binary interactome 9438–9441
mapping. Nat Methods 6:83–90 13. Zhang QC, Petrey D, Norel R et al (2010)
7. Voorhees RM, Weixlbaumer A, Loakes D et al Protein interface conservation across
(2009) Insights into substrate stabilization structure space. Proc Natl Acad Sci U S A
from snapshots of the peptidyl transferase cen- 107:10896–10901
ter of the intact 70S ribosome. Nat Struct Mol 14. Gao M, Skolnick J (2010) Structural space of
Biol 16:528–533 protein-protein interfaces is degenerate, close
8. Grigorieff N, Harrison SC (2011) Near-atomic to complete, and highly connected. Proc Natl
resolution reconstructions of icosahedral Acad Sci U S A 107:22517–22522
20 Robert C. Liddington
15. Zhang QC, Petrey D, Garzon JI et al (2013) 31. Tourigny DS, Fernandez IS, Kelley AC et al
PrePPI: a structure-informed database of (2013) Elongation factor G bound to the ribo-
protein–protein interactions. Nucleic Acids some in an intermediate state of translocation.
Res 41:D828–D833 Science 340:1235490
16. Aloy P, Ceulemans H, Stark A et al (2003) The 32. Brown CJ, Johnson AK, Dunker AK et al
relationship between sequence and interaction (2011) Evolution and disorder. Curr Opin
divergence in proteins. J Mol Biol Struct Biol 21:441–446
332:989–998 33. Dunker AK, Silman I, Uversky VN et al (2008)
17. Janin J (2013) The targets of CAPRI rounds Function and structure of inherently disor-
20–27. Proteins 81:2075–2081 dered proteins. Curr Opin Struct Biol
18. Janin J, Rodier F, Chakrabarti P et al (2007) 18:756–764
Macromolecular recognition in the Protein 34. Janin J, Sternberg MJ (2013) Protein flexibil-
Data Bank. Acta Crystallogr D Biol Crystallogr ity, not disorder, is intrinsic to molecular rec-
63:1–8 ognition. F1000 Biol Rep 5:2
19. Janin J (2010) The targets of CAPRI Rounds 35. Lo CL, Chothia C, Janin J (1999) The atomic
13–19. Proteins 78:3067–3072 structure of protein-protein recognition sites. J
20. Wass MN, David A, Sternberg MJ (2011) Mol Biol 285:2177–2198
Challenges for the prediction of macromolecu- 36. Wodak SJ, Janin J (2003) Structural basis of
lar interactions. Curr Opin Struct Biol macromolecular recognition. Adv Protein
21:382–390 Chem 61:9–73
21. Lander GC, Saibil HR, Nogales E (2012) Go 37. Levy ED, Teichmann S (2013) Structural, evo-
hybrid: EM, crystallography, and beyond. Curr lutionary, and assembly principles of protein
Opin Struct Biol 22:627–635 oligomerization. Prog Mol Biol Transl Sci
22. Rouiller I, Xu XP, Amann KJ et al (2008) The 117:25–51
structural basis of actin filament branching 38. Bogan AA, Thorn KS (1998) Anatomy of hot
by the Arp2/3 complex. J Cell Biol spots in protein interfaces. J Mol Biol 280:1–9
180:887–895 39. Li Y, Huang Y, Swaminathan CP et al (2005)
23. Rambo RP, Tainer JA (2013) Super-resolution Magnitude of the hydrophobic effect at central
in solution X-ray scattering and its applications versus peripheral sites in protein-protein inter-
to structural systems biology. Annu Rev Bio- faces. Structure 13:297–307
phys 42:415–441 40. Ofran Y, Rost B (2003) Analysing six types of
24. Stein A, Aloy P (2010) Novel peptide- protein-protein interfaces. J Mol Biol
mediated interactions derived from high- 325:377–387
resolution 3-dimensional structures. PLoS 41. Levy ED (2010) A simple definition of struc-
Comput Biol 6:e1000789 tural regions in proteins and its use in analyzing
25. Seong J, Tajik A, Sun J et al (2013) Distinct interface evolution. J Mol Biol 403:660–670
biophysical mechanisms of focal adhesion 42. Dey S, Pal A, Chakrabarti P et al (2010) The
kinase mechanoactivation by different extracel- subunit interfaces of weakly associated homo-
lular matrix proteins. Proc Natl Acad Sci U S A dimeric proteins. J Mol Biol 398:146–160
110:19372–19377 43. Glaser F, Steinberg DM, Vakser IA et al (2001)
26. Hunter T (2012) Why nature chose phosphate Residue frequencies and pairing preferences at
to modify proteins. Philos Trans R Soc Lond B protein-protein interfaces. Proteins 43:89–102
Biol Sci 367:2513–2516 44. Headd JJ, Ban YE, Brown P et al (2007)
27. Wolfenson H, Lavelin I, Geiger B (2013) Protein–protein interfaces: properties, prefer-
Dynamic regulation of the structure and func- ences, and projections. J Proteome Res
tions of integrin adhesions. Dev Cell 6:2576–2586
24:447–458 45. Keskin O, Ma B, Nussinov R (2005) Hot
28. Rawlings ND, Tolle DP, Barrett AJ (2004) regions in protein–protein interactions: the
Evolutionary families of peptidase inhibitors. organization and contribution of structurally
Biochem J 378:705–716 conserved hot spot residues. J Mol Biol
29. Dice JF (1990) Peptide sequences that target 345:1281–1294
cytosolic proteins for lysosomal proteolysis. 46. Crowley PB, Golovin A (2005) Cation-Π inter-
Trends Biochem Sci 15:305–309 actions in protein-protein interfaces. Proteins
30. Pawson T, Nash P (2003) Assembly of cell 59:231–239
regulatory systems through protein interaction 47. Chen P, Li J, Wong L et al (2013) Accurate
domains. Science 300:445–452 prediction of hot spot residues through
Structural Basis 21
physicochemical characteristics of amino acid 63. Eck MJ, Shoelson SE, Harrison SC (1993)
sequences. Proteins 81:1351–1362 Recognition of a high-affinity phosphotyrosyl
48. Hwang H, Vreven T, Janin J et al (2010) peptide by the Src homology-2 domain of
Protein-protein docking benchmark version p56lck. Nature 362:87–91
4.0. Proteins 78:3111–3114 64. Doyle DA, Lee A, Lewis J et al (1996) Crystal
49. Cherfils J, Zeghouf M (2013) Regulation of structures of a complexed and peptide-free
small GTPases by GEFs, GAPs, and GDIs. membrane protein-binding domain: molecular
Physiol Rev 93:269–309 basis of peptide recognition by PDZ. Cell
50. Lee JO, Rieu P, Arnaout MA et al (1995) 85:1067–1076
Crystal structure of the A domain from the 65. Hayashi I, Vuori K, Liddington RC (2002)
alpha subunit of integrin CR3 (CD11b/ The focal adhesion targeting (FAT) region of
CD18). Cell 80:631–638 focal adhesion kinase is a four-helix bundle that
51. Emsley J, Knight CG, Farndale RW et al binds paxillin. Nat Struct Biol 9:101–106
(2000) Structural basis of collagen recognition 66. Pokutta S, Weis WI (2000) Structure of the
by integrin α2β1. Cell 101:47–56 dimerization and beta-catenin-binding region
52. Hogg N, Harvey J, Cabanas C et al (1993) of alpha-catenin. Mol Cell 5:533–543
Control of leukocyte integrin activation. Am 67. Musacchio A, Saraste M, Wilmanns M (1994)
Rev Respir Dis 148:S55–S59 High-resolution crystal structures of tyrosine
53. Shimaoka M, Xiao T, Liu JH et al (2003) kinase SH3 domains complexed with proline-
Structures of the αL I domain and its complex rich peptides. Nat Struct Biol 1:546–551
with ICAM-1 reveal a shape-shifting pathway 68. Nilsen TW, Graveley BR (2010) Expansion of
for integrin regulation. Cell 112:99–111 the eukaryotic proteome by alternative splic-
54. Kuhlmann UC, Pommer AJ, Moore GR et al ing. Nature 463:457–463
(2000) Specificity in protein–protein interac- 69. Weatheritt RJ, Jehl P, Dinkel H et al (2012)
tions: the structural basis for dual recognition iELM – a web server to explore short linear
in endonuclease colicin-immunity protein motif-mediated interactions. Nucleic Acids
complexes. J Mol Biol 301:1163–1178 Res 40:W364–W369
55. Xiong JP, Stehle T, Zhang R et al (2002) Crys- 70. Van Roey K, Gibson TJ, Davey NE (2012)
tal structure of the extracellular segment of Motif switches: decision-making in cell regula-
integrin αVβ3 in complex with an Arg-Gly- tion. Curr Opin Struct Biol 22:378–385
Asp ligand. Science 296:151–155 71. Harrison SC (1996) Peptide-surface associa-
56. Luo BH, Carman CV, Springer TA (2007) tion: the case of PDZ and PTB domains. Cell
Structural basis of integrin regulation and sig- 86:341–343
naling. Annu Rev Immunol 25:619–647 72. Van Roey K, Dinkel H, Weatheritt RJ et al
57. Huizinga EG, Tsuji S, Romijn RA et al (2002) (2013) The switches ELM resource: a compen-
Structures of glycoprotein Ibα and its complex dium of conditional regulatory interaction
with von Willebrand factor A1 domain. Science interfaces. Sci Signal 6:rs7
297:1176–1179 73. Stein A, Aloy P (2008) Contextual specificity in
58. Brondijk TH, Bihan D, Farndale RW et al peptide-mediated protein interactions. PLoS
(2012) Implications for collagen I chain regis- One 3:e2524
try from the structure of the collagen von Will- 74. Garcia-Alvarez B, de Pereda JM, Calderwood
ebrand factor A3 domain complex. Proc Natl DA et al (2003) Structural determinants
Acad Sci U S A 109:5253–5258 of integrin recognition by talin. Mol Cell
59. Weatheritt RJ, Gibson TJ (2012) Linear 11:49–58
motifs: lost in (pre)translation. Trends Bio- 75. Cowan KJ, Law DA, Phillips DR (2000) Iden-
chem Sci 37:333–341 tification of Shc as the primary protein binding
60. Weatheritt RJ, Davey NE, Gibson TJ (2012) to the tyrosine-phosphorylated β3 subunit of
Linear motifs confer functional diversity onto αIIbβ3 during outside-in integrin platelet sig-
splice variants. Nucleic Acids Res 40:7123–7131 naling. J Biol Chem 275:36423–36429
61. Yaffe MB, Rittinger K, Volinia S et al (1997) 76. Di Paolo G, Pellegrini L, Letinic K et al (2002)
The structural basis for 14-3-3:phosphopep- Recruitment and regulation of phosphatidyli-
tide binding specificity. Cell 91:961–971 nositol phosphate kinase type 1-γ by the FERM
62. Lee JO, Russo AA, Pavletich NP (1998) Struc- domain of talin. Nature 420:85–89
ture of the retinoblastoma tumour-suppressor 77. Schill NJ, Anderson RA (2009) Two novel
pocket domain bound to a peptide from HPV phosphatidylinositol-4-phosphate 5-kinase
E7. Nature 391:859–865 type Igamma splice variants expressed in
22 Robert C. Liddington
human cells display distinctive cellular target- localization and action. Curr Opin Struct Biol
ing. Biochem J 422:473–482 21:808–813
78. Reyes A, Anders S, Weatheritt RJ et al (2013) 86. Toutant M, Costa A, Studler JM et al (2002)
Drift and conservation of differential exon Alternative splicing controls the mechanisms of
usage across tissues in primate species. Proc FAK autophosphorylation. Mol Cell Biol
Natl Acad Sci U S A 110:15377–15382 22:7731–7743
79. Hui S, Xing X, Bader GD (2013) 87. Margadant F, Chew LL, Hu X et al (2011)
Predicting PDZ domain mediated protein Mechanotransduction in vivo by repeated talin
interactions from structure. BMC Bioinfor- stretch-relaxation events depends upon vincu-
matics 14:27 lin. PLoS Biol 9:e1001223
80. Bakolitsa C, Cohen DM, Bankston LA et al 88. Hu K, Ji L, Applegate KT et al (2007) Differ-
(2004) Structural basis for vinculin activation ential transmission of actin motion within focal
at sites of cell adhesion. Nature 430:583–586 adhesions. Science 315:111–115
81. Balla T (2005) Inositol-lipid binding motifs: 89. Seifert C, Grater F (2013) Protein mechanics:
signal integrators through protein-lipid and how force regulates molecular function. Bio-
protein–protein interactions. J Cell Sci chim Biophys Acta 1830:4762–4768
118:2093–2104 90. Yahav T, Maimon T, Grossman E et al (2011)
82. Carlton JG, Cullen PJ (2005) Coincidence Cryo-electron tomography: gaining insight
detection in phosphoinositide signaling. into cellular processes by structural approaches.
Trends Cell Biol 15:540–547 Curr Opin Struct Biol 21:670–677
83. Schiller HB, Fassler R (2013) Mechanosensi- 91. Zhang P (2013) Correlative cryo-electron
tivity and compositional dynamics of cell- tomography and optical microscopy of cells.
matrix adhesions. EMBO Rep 14:509–519 Curr Opin Struct Biol 23:763–770
84. Lietha D, Cai X, Ceccarelli DF et al (2007) 92. Baker ML, Hryc CF, Zhang Q et al (2013)
Structural basis for the autoinhibition of focal Validated near-atomic resolution structure of
adhesion kinase. Cell 129:1177–1187 bacteriophage epsilon15 derived from cryo-
85. Arold ST (2011) How focal adhesion kinase EM and modeling. Proc Natl Acad Sci U S A
achieves regulation by linking ligand binding, 110:12301–12306
Chapter 2
Abstract
Numerous authors, including contributors to this volume, have described methods to detect protein-protein
interactions. Many of these approaches are now accessible to the inexperienced investigator thanks to core
facilities and/or affordable instrumentation. This chapter discusses some common design considerations that
are necessary to obtain valid measurements, as well as the assumptions and analytical methods that are relevant
to the quantitation of these interactions.
Key words Ligand binding, Protein-protein interaction, Fluorescence, Binding equations, Binding
equilibria
1 Introduction
1.1 Assumptions There are several assumptions inherent to any analysis of a simple
ligand–receptor interaction (https://tools.lifetechnologies.com/
downloads/FP7.pdf).
1. The interactions are assumed to be reversible. In the simplest
case, the association reaction is bimolecular while the dissocia-
tion reaction in unimolecular.
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_2, © Springer Science+Business Media New York 2015
23
24 Ziad M. Eletr and Keith D. Wilkinson
½R f ½L f
½RL Kd
¼ ; ð3Þ
½R t ½Rf þ ½RL
½RL ½L f
¼ : ð4Þ
½R t K d þ ½L f
a b c
1 1 nRt 1
-1/Kd
0.8 0.8 0.8
0.6
[Lb]/[Lf]
0.6 0.6
[Lb]/[Rt]
[Lb]/[Rt]
0.4 0.4
0.4
0.2 0.2
0.2
Kd
0
0 0
Fig. 1 Plots of simulated data for simple binding. In all cases n ¼ 1 and Kd ¼ 1 μM. (a) Direct plot of
fractional saturation vs. free ligand; (b) Klotz plot of the same data. Note the log scale; (c) Scatchard plot of the
same data. The parameters nRt and nRt/Kd are estimated from the intercepts
26 Ziad M. Eletr and Keith D. Wilkinson
1.3 Binding to It should be noted that if more than one ligand molecule binds to R
Multiple Sites then the behavior may be more complex. For n multiple binding
sites we get:
½RL ¼ ½RL 1 þ ½RL 2 . . . ½RL n
½Rt ½L f ½R t ½L f ½R t ½L f
¼ þ ... ; ð7Þ
K d1 þ ½L f K d2 þ ½L f K dn þ ½L f
where n different sites can be occupied by ligand with the
corresponding binding constants.
1.3.1 Identical, Non- If all binding sites are identical and non-interacting (i.e., all bind
interacting Binding Site(s) with the same Kd) then Eq. 7 reduces to:
½L b ½L f
¼ ; ð8Þ
n ½R t K d þ ½L f
1.3.2 Non-identical While the most common reason for observing multiple non-
Binding Sites identical binding sites in a protein-protein interaction is likely to
be nonspecific binding (see below), it is always possible that there
are two independent and non-interacting sites with different affi-
nities. Either case will manifest itself as a deviation from the
expected behavior for a simple binding model. The Scatchard plot
is a useful diagnostic tool to point out such deviations (Fig. 2). A
Scatchard plot that is concave upward is indicative of nonspecific
binding, negative cooperativity, or multiple classes of binding sites.
A concave downward plot suggests either positive cooperativity or
instability of the ligand. In any case, proper analysis of this behavior
requires other information (for instance stoichiometry, stability)
and the data are best fitted using nonlinear least squares fitting of
the data according to an appropriate model.
28 Ziad M. Eletr and Keith D. Wilkinson
a b c
1 1 1
[Lb]/[Lf]
[Lb]/[Lf]
0.4 0.4 0.4
0 0 0
0 0.2 0.4 0.6 0.8 1 0 0.2 0.4 0.6 0.8 1 0 0.2 0.4 0.6 0.8 1
[Lb] μM [Lb] μM [Lb] μM
Simple binding Two different sites, Positive cooperativity
Negative cooperativity, or
or Unstable ligand
Non-specific binding
Fig. 2 Effects of complexities on the appearance of the Scatchard plot. (a) Represents the expected behavior in
the simple case; (b) a concave upward deviation as shown in this panel could be caused by the presence
of two different sites, the presence of negative cooperativity or a significant nonspecific binding component;
(c) Positive cooperativity or ligand instability would lead to the curvature shown in this panel
1.4 Cooperativity Cooperativity is the term used to describe the situation where occu-
pancy of one site changes the affinity for ligand at another site. There
have been many treatments of cooperative binding interactions,
including analysis by Scatchard and Hill plots, but these are beyond
the scope of this discussion. In general, models explaining coopera-
tivity invoke subunit–subunit interactions in oligomeric protein
structures and may well be important in cases where multiple pro-
teins are being assembled into a multimeric complex. The reader is
referred to any of several other treatments of such binding if com-
plications of this sort are indicated [9–11]. However, it may be
simpler to restrict the measurements to conditions where individual
subcomplexes are assembled at saturating concentrations before
measuring the binding of a subsequent protein.
2 Materials
available for all platforms, i.e., Prism (GraphPad Software, Inc., San
Diego, CA), SigmaPlot (SPSS Science, Chicago, IL), Mathematica
(Wolfram Research Inc., Champaign, IL), DynaFit (BioKin, Ltd.,
Pullman, WA), MATLAB (MathWorks, Natick, MA), and others.
There are also published solutions using the popular spreadsheet
Microsoft Excel [12, 13]. The choice is largely up to personal
preference.
3 Methods
3.1 Direct Direct methods require that we accurately determine the concen-
Measurement of Free trations of free and bound ligand. Examples of techniques that yield
Ligand such information include gel filtration, ultracentrifugation, ultrafil-
tration, or equilibrium dialysis. For binding with slow dissociation
rates pull-downs, band shift or electrophoresis techniques may be
appropriate. If the process of separating the bound and free ligand
is fast compared to the rate of dissociation of the complex such
methods can yield directly the concentrations of bound and free
ligand. If dissociation and separation of bound and free reactants
occur on similar time scales such methods are not appropriate for
quantitation as the equilibrium will be disturbed by the separation
of the reactants. For the same reasons techniques such as cross-
linking may overestimate the concentration of RL since the equi-
librium will be disturbed by the removal of RL from the
equilibrium.
monitored it is assumed that there are only two states, the bound
and the free, and that each has a characteristic value. If So is the
signal in the absence of binding, SL the signal in the presence of
total ligand concentration L, and S1 is the value at saturation, then:
ðS L S o Þ
fraction saturation ¼ : ð11Þ
ðS 1 S o Þ
The concentration of free ligand can be calculated by assuming a
stoichiometry n and using the expression [L f] ¼ [L t] n[Rt] ¼
(SL So)/(S1 So). Note that if n is incorrect, then the calcu-
lated [L f] will be incorrect also and this will be apparent in the
deviation of the data from the theoretical rectangular hyperbola.
This is one reason why the determination of n is an important
exercise in most binding studies. Alternatively, and preferably,
data are fitted using nonlinear least squares methods and n is
determined directly from this analysis.
3.3 Competition Direct methods measure either bound ligand [RL] or free ligand
Methods [L f] as a function of [L t] and indirect methods usually involve
measuring fractional saturation [RL]/n[Rt] as a function of [L t].
However, one of the most useful variations of the binding experi-
ment is the use of competitive binding assays where a single labeled
indicator ligand can be bound and subsequently displaced by any of
a variety of competitive inhibitors [14–19]. Such experiments are
particularly useful if the affinity of a series of inhibitors is to be
determined. Methods such as fluorescence polarization or flores-
cence resonance energy transfer are particularly well suited for such
measurements. A small amount of the labeled ligand is first bound
to the receptor and subsequently displaced by titrating with unla-
beled inhibitor. The Ki of the unlabeled inhibitor is then calculated.
The labeled ligand does not have to be physiological or bound with
a physiological affinity since we are always comparing the Ki of the
unlabeled inhibitor. Thus, any adverse effects of labeling the indi-
cator ligand will be unimportant.
The IC50 is the concentration of inhibitor necessary to displace
half the labeled ligand. If [Rt] Kd, IC50 is related to Ki, the
affinity of the unlabeled ligand by:
IC50
Ki ¼ ; ð12Þ
1 þ Lt =K d
a ¼ K a þ K b þ ½A t þ ½B t ½P t
b ¼ K b ð½A t ½P t Þ þ K a ð½B t ½P t Þ þ K a K b
c ¼ K a K b ½P t :
The experiment requires the measurement of the fractional satura-
tion at various concentrations of At, Bt, and Pt. Only a small range
of measurements are useful: the ones where fractional saturation is
>0.05 and <0.95. Fractional saturation of P with the probe A is
determined by indirect measurements where it is the fluorescence
or the anisotropy of AP which gives rise to the signal. The usual
experiment is to measure the full binding curve, i.e., (S So)/
(S1 So) as a function of Pt. This experiment should then be
repeated at three or more concentrations of Bt to calculate Kb.
Although this may seem like its only giving you three data points,
if the curve is fitted, the actual number of useful data points is equal
to the total measurements made where fractional saturation is in a
useful range.
32 Ziad M. Eletr and Keith D. Wilkinson
3.4.2 Kinetics The analysis of binding requires that we conduct the measurements
after binding has reached equilibrium or that we measure individu-
ally the rate constants involved. The binding constant can then be
calculated from the relationship Kd ¼ k2/k1. From a practical
standpoint, assuring that the reaction has reached equilibrium
often involves measuring a time course for binding at low ligand
concentrations and making all measurements after sufficient time to
allow attainment of equilibrium. Several examples of each type of
analysis are given in later chapters.
In any case it is instructive to consider the magnitudes of
association and dissociation rates. The association rate constants
expected for protein-protein interactions are limited by diffusion. If
we assume reasonable numbers for the diffusion rate of an average
protein, the diffusion limit in aqueous solution is around
108–109 M1 s1. There are also additional steric constraints as
only a fraction of the collisions occurring at this rate are oriented
properly, and it is commonly assumed that the rate limiting associa-
tion rate (k1) for two proteins binding to each other is around
108 M1 s1.
It can be shown that the rate of approach to equilibrium is
determined by the sum of the association rate and the dissociation
rate constants. Further, the concentrations of reagents must be at
or near the binding constant for accurate determination of both
stoichiometry and affinity in the same experiment (see below). If
the dissociation constant (Kd) for such an interaction is moderate
(106 M) then the dissociation rate for such a complex will be
k2 ¼ k1 Kd ¼ 102 s1. Thus, binding will be complete in
seconds and the half-life of the bound state will be tens of milli-
seconds. If, however, the binding constant is very tight, as may
occur in antibody–antigen interactions, the overall equilibrium may
take some time. Consider a binding interaction with a free energy of
16 kcal/mol, an affinity exhibited by many antibodies and other
protein-protein interactions [20]. This represents a dissociation
Quantitation of Binding 33
3.5 Concentrations Equation 8 is the equation for the familiar rectangular hyperbola
of Components to Use with a horizontal asymptote corresponding to 100 % saturation and
half-maximal saturation occurring at L f ¼ Kd. This equation points
3.5.1 Ligand out that the concentrations of free ligand present must be similar to
Concentration the dissociation constant in order to vary the fractional saturation of
receptor, i.e., to measure the strength of binding. The most com-
mon form of the experiment then is to titrate a fixed amount of
receptor with variable amounts of ligand and to fit the experimental
data to the appropriate binding equation to determine the stoichi-
ometry n and the binding constant Kd.
3.5.2 Receptor If we consider the concentration of the fixed protein in this binding
Concentration equation, i.e., [Rt], we can define three limiting conditions: [Rt]
Kd, [Rt] Kd, and [Rt] ~ Kd. Figure 3 illustrates the interre-
lationships between [Kd] and [Rt] in such experiments.
a b
1 Kd=.01μM
1 Kd=.01 μM
=1μM
=1 μM
0.8 0.8
=2 μM
=5 μM
0.6 =2μM
0.6
[Lb]/[Rt]
[Lb]/[Rt]
=10 μM
Rt=1 μM Rt=1 μM
=5μM
0.4 =10μM
0.4
0.2 0.2
0 0
Fig. 3 Binding isotherms for a simple binding equilibrium where n ¼ 1 and the total concentration of receptor
is 1 μM. (a) Direct plot of fractional saturation vs. total ligand added and (b) the same data plotted on a log
scale. Note that as the Kd approaches [Rt] there is a significant deviation from the rectangular hyperbolic
behavior
34 Ziad M. Eletr and Keith D. Wilkinson
½L b ½L t
¼ : ð14Þ
n ½R t K d þ ½L t
In the first part of the titration curve, when [L f] is less than Kd (and
much less than n[Rt] in this example), the ratio of bound/free
ligand is determined solely by the ratio of n[Rt]/[Kd].
If we only measure [L t] the limiting slope for a plot of satura-
tion vs. [L t] is n[Rt]/[Kd]. For example, if n[Rt]/[Kd] ¼ 100,
then only about 1 % of the added ligand is free at low ligand
concentrations. In order to saturate binding [L t] must exceed
100 Kd. When [Rt] Kd, the saturation curve is really an end-
point determination consisting of two lines (first a slope of ~n and
then 0 intersecting at [L t] ¼ n[Rt]) with little curvature (Fig. 3).
Under these conditions we can accurately determine n, but not Kd.
If direct methods to measure free ligand are used, we can, in
theory, calculate Kd, but in practical terms the curve will only
deviate from its biphasic nature near L t ¼ Rt, and generally there
will not be enough data in this region to obtain accurate estimates
of Kd.
[Rt] ~ Kd The most useful conditions for determining both Kd and n are
when [Rt] ~ Kd. The binding curve still resembles a rectangular
hyperbola but with small deviations due to the fact that [L t] ¼
[L f] + n[RL]. Since [L f] is similar in magnitude to [Lb] each can
be measured (or calculated) with good accuracy. Under these con-
ditions we can determine both Kd and n with a good degree of
accuracy from the same experiment.
Quantitation of Binding 35
4 Notes
References
their E1 enzymes before E3-dependent ubiqui- signal originating from a reference macromole-
tin and ubiquitin-like transfer. Nat Struct Mol cule. Application to Escherichia coli replicative
Biol. 12:933–934 helicase DnaB protein nucleic acid interactions.
6. Munson PJ, Rodbard D (1983) Number of Biochemistry 35:2117–2128
receptor sites from Scatchard and Klotz 15. Schwarz G (2000) A universal thermodynamic
graphs: a constructive critique. Science approach to analyze biomolecular binding
220:979–981 experiments. Biophys Chem 86:119–129
7. Swillens S (1995) Interpretation of binding 16. van Zoelen EJ (1992) Analysis of receptor
curves obtained with high receptor concentra- binding displacement curves by a nonhomol-
tions: practical aid for computer analysis. Mol ogous ligand, on the basis of an equivalent
Pharmacol 47:1197–1203 competition principle. Anal Biochem
8. Motulsky HJ, Ransnas LA (1987) Fitting 200:393–399
curves to data using nonlinear regression: a 17. van Zoelen EJ, Kramer RH, van Moerkerk HT
practical and nonmathematical review. FASEB et al (1998) The use of nonhomologous
J 1:365–374 Scatchard analysis in the evaluation of ligand-
9. Tuk B, van Oostenbruggen MF (1996) Solving protein interactions. Trends Pharmacol Sci
inconsistencies in the analysis of receptor- 19:487–490
ligand interactions. Trends Pharmacol Sci 18. van Zoelen EJ, Kramer RH, van Reen MM et al
17:403–409 (1993) An exact general analysis of ligand
10. Koshland DE Jr (1996) The structural basis of binding displacement and saturation curves.
negative cooperativity: receptors and enzymes. Biochemistry 32:6275–6280
Curr Opin Struct Biol 6:757–761 19. Wang ZX (1995) An exact mathematical
11. Forsen S, Linse S (1995) Cooperativity: over expression for describing competitive binding
the Hill. Trends Biochem Sci 20:495–497 of two different ligands to a protein molecule.
12. Hedlund PB, von Euler G (1999) EasyBound – FEBS Lett 360:111–114
a user-friendly approach to nonlinear regres- 20. Brooijmans N, Sharp KA, Kuntz ID (2002)
sion analysis of binding data. Comput Methods Stability of macromolecular complexes. Pro-
Programs Biomed 58:245–249 teins 48:645–653
13. Brown AM (2001) A step-by-step guide to 21. Rovati GE, Rodbard D, Munson PJ (1988)
non-linear regression analysis of experimental DESIGN: computerized optimization of
data using a Microsoft Excel spreadsheet. Com- experimental design for estimating Kd and
put Methods Programs Biomed 65:191–200 Bmax in ligand binding experiments. I.
14. Jezewska MJ, Bujalowski W (1996) A general Homologous and heterologous binding to
method of analysis of ligand binding to com- one or two classes of sites. Anal Biochem
peting macromolecules using the spectroscopic 174:636–649
Chapter 3
Abstract
Years of meticulous curation of scientific literature and increasingly reliable computational predictions have
resulted in creation of vast databases of protein interaction data. Over the years, these repositories have
become a basic framework in which experiments are analyzed and new directions of research are explored.
Here we present an overview of the most widely used protein-protein interaction databases and the
methods they employ to gather, combine, and predict interactions. We also point out the trade-off between
comprehensiveness and accuracy and the main pitfall scientists have to be aware before adopting protein
interaction databases in any single-gene or genome-wide analysis.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_3, © Springer Science+Business Media New York 2015
39
40 Damian Szklarczyk and Lars Juhl Jensen
Much like the term “function” does not only encompass molecular
functions such as enzymatic catalysis, the term “interaction” in
addition to direct physical binding covers a variety of indirect
links such as complex co-membership, regulatory relationships,
and genetic interactions. These are collectively referred to as “func-
tional associations”; however, the terms “interaction” and
“functional association” are often used interchangeably in the liter-
ature and in databases too.
Protein-Protein Interaction Networks 41
Fig. 1 Default visualizations of JAK1 interactions by different databases. (a) BioGRID HTML table showing
proteins that physically interact with JAK1 (indicated by the yellow color of “experimental evidence”). (b) JAK1
interactions shown in the IntAct “graph viewer” with small molecules being depicted as triangles. The network
view shows only the interactions with JAK1 and not between any other two nodes. (c) JAK1 STRING network.
Each different colored line that connects proteins indicates a separate evidence channel for the particular
interaction, such as text mining (green), experiments (magenta), and databases (blue). (d) InnateDB’s
Cytoscape with Cerebral plug-in view uses “cellular component” GO annotation to lay out proteins based
on their localization inside the cell with the horizontal lines indicating boundaries between different
components
3 Pathway Databases
6 Interolog Prediction
7 Predicted PPIs
Table 1
Comparison of different interaction databases
Number of
Total number interactions Number Built in
of interactions in human of Computational network Unified
Database (thousands) (thousands) species predictions view scoring IMEx
BioGRID 372 75,9 40 No No No Yes (C)
DIP 68 3,4 485 No Yes No Yes
GeneMANIA 96208 24220,1 7 Yes Yes Yes No
HPRD 37 37,1 1 No No No Yes
I2D 666 152,4 6 Yes (A) No No Yes
InnateDB 104 84,0 3 Yes (A) Yes No Yes
IntAct 2331 44,0 397 No Yes Yes Yes
MINT 95 21,3 434 Yes (A, B) Yes Yes Yes
PIPs 78 78,4 1 Yes No Yes No
STRING 224346 1540,7 1133 Yes Yes Yes No
Reactome 2161 121,9 28 No Yes No No
KEGG (D) 5880 63,3 1509 Yes (B) Yes No No
Binary protein interaction count does not include self-associations. For databases focused on computational predictions,
the total number of interactions stored is not a valid predictor of quality of comprehensiveness of the database as most of
the interactions are low-scoring and not relevant for noncomputational analysis. A only interologs prediction, B only
homoMINT, C observing member. D the version of KEGG PATHWAY included in this breakdown is from July 2011,
which was the last set available from KEGG under free license
48 Damian Szklarczyk and Lars Juhl Jensen
10 Data Integration
11 Bias
References
1. Croft D, O’Kelly G, Wu G, Haw R et al (2011) 10. Goodman N, McCormick K, Goldowitz D,
Reactome: a database of reactions, pathways Hockly E et al (2003) Plans for HDBase—a
and biological processes. Nucleic Acids Res research community website for Huntington’s
39:D691–D697 Disease. Clin Neurosci Res 3:197–217
2. Kanehisa M, Goto S, Furumichi M, Tanabe M 11. Lechner M, Höhn V, Brauner B, Dunger I et al
et al (2010) KEGG for representation and anal- (2012) CIDeR: multifactorial interaction net-
ysis of molecular networks involving diseases works in human diseases. Genome Biol 13:R62
and drugs. Nucleic Acids Res 38:D355–D360 12. Dinkel H, Chica C, Via A, Gould CM et al
3. Kerrien S, Aranda B, Breuza L, Bridge A et al (2011) Phospho.ELM: a database of phos-
(2012) The IntAct molecular interaction data- phorylation sites–update 2011. Nucleic Acids
base in 2012. Nucleic Acids Res 40: Res 39:D261–D267
D841–D846 13. Caspi R, Foerster H, Fulcher CA, Kaipa P et al
4. Stark C, Breitkreutz B-J, Chatr-Aryamontri A, (2008) The MetaCyc Database of metabolic
Boucher L et al (2011) The BioGRID Interac- pathways and enzymes and the BioCyc collec-
tion Database: 2011 update. Nucleic Acids Res tion of Pathway/Genome Databases. Nucleic
39:D698–D704 Acids Res 36:D623–D631
5. Szklarczyk D, Franceschini A, Kuhn M, Simo- 14. Smoot ME, Ono K, Ruscheinski J, Wang P-L
novic M et al (2011) The STRING database in et al (2011) Cytoscape 2.8: new features for
2011: functional interaction networks of pro- data integration and network visualization.
teins, globally integrated and scored. Nucleic Bioinformatics 27:431–432
Acids Res 39:D561–D568 15. Brown KR, Otasek D, Ali M, McGuffin MJ
6. Warde-Farley D, Donaldson SL, Comes O, et al (2009) NAViGaTOR: Network Analysis,
Zuberi K et al (2010) The GeneMANIA pre- Visualization and Graphing Toronto. Bioinfor-
diction server: biological network integration matics 25:3327–3329
for gene prioritization and predicting gene 16. Gene T., Consortium O. (2010) The Gene
function. Nucleic Acids Res 38:W214–W220 Ontology Consortium in 2010: extensions and
7. Goel R, Harsha HC, Pandey A, Prasad TSK refinements. Nucleic Acids Res 38:D331–D335
(2012) Human Protein Reference Database 17. Hakes L, Robertson DL, Oliver SG (2005)
and Human Proteinpedia as resources for phos- Effect of dataset selection on the topological
phoproteome analysis. Mol Biosyst 8:453–463 interpretation of protein interaction networks.
8. Cherry JM, Hong EL, Amundsen C, Balakrish- BMC Genomics 6:131
nan R et al (2012) Saccharomyces Genome 18. Salwinski L, Miller CS, Smith AJ, Pettit FK et al
Database: the genomics resource of budding (2004) The Database of Interacting Proteins:
yeast. Nucleic Acids Res 40:D700–D705 2004 update. Nucleic Acids Res 32:
9. Murali T, Pacifico S, Yu J, Guest S et al (2011) D449–D451
DroID 2011: a comprehensive, integrated 19. Salwinski L, Eisenberg D (2007) The MiSink
resource for protein, transcription factor, Plugin: cytoscape as a graphical interface to the
RNA and gene interactions for Drosophila. Database of Interacting Proteins. Bioinformat-
Nucleic Acids Res 39:D736–D743 ics 23:2193–2195
Protein-Protein Interaction Networks 55
20. Deane CM, Salwiński Ł, Xenarios I, Eisenberg 33. Barsky A, Gardy JL, Hancock REW, Munzner
D (2002) Protein interactions: two methods T (2007) Cerebral: a Cytoscape plugin for lay-
for assessment of the reliability of high out of and interaction with biological networks
throughput observations. Mol Cell Proteomics using subcellular localization annotation. Bio-
1:349–356 informatics 23:1040–1042
21. Deng M, Mehta S, Sun F, Chen T (2002) 34. Fu W, Sanders-Beer BE, Katz KS, Maglott DR
Inferring domain-domain interactions from et al (2009) Human immunodeficiency virus
protein-protein interactions. Genome Res type 1, human protein interaction database at
12:1540–1548 NCBI. Nucleic Acids Res 37:D417–D422
22. Graeber TG, Eisenberg D (2001) Bioinfor- 35. Resource Coordinators NCBI (2013) Data-
matic identification of potential autocrine sig- base resources of the National Center for Bio-
naling loops in cancers from gene expression technology Information. Nucleic Acids Res 41:
profiles. Nat Genet 29:295–300 D8–D20
23. Hastings J, de Matos P, Dekker A, Ennis M 36. Chen R, Jeong SS (2000) Functional predic-
et al (2013) The ChEBI reference database tion: identification of protein orthologs and
and ontology for biologically relevant chemis- paralogs. Protein Sci 9:2344–2353
try: enhancements for 2013. Nucleic Acids Res 37. Niu Y, Otasek D, Jurisica I (2010) Evaluation
41:D456–D463 of linguistic features useful in extraction of
24. Ceol A, Chatr Aryamontri A, Licata L, Peluso interactions from PubMed; application to
D et al (2010) MINT, the molecular interac- annotating known, high-throughput and pre-
tion database: 2009 update. Nucleic Acids Res dicted interactions in I2D. Bioinformatics
38:D532–D539 26:111–119
25. Persico M, Ceol A, Gavrila C, Hoffmann R et al 38. Hermjakob H, Montecchi-Palazzi L, Bader G,
(2005) HomoMINT: an inferred human net- Wojcik J et al (2004) The HUPO PSI’s molec-
work based on orthology mapping of protein ular interaction format–a community standard
interactions discovered in model organisms. for the representation of protein interaction
BMC Bioinformatics 6(Suppl 4):S21 data. Nat Biotechnol 22:177–183
26. Chatr-aryamontri A, Ceol A, Peluso D, Nar- 39. Hart GT, Ramani AK, Marcotte EM (2006)
dozza A et al (2009) VirusMINT: a viral pro- How complete are current yeast and human
tein interaction database. Nucleic Acids Res 37: protein-interaction networks? Genome Biol
D669–D673 7:120
27. Ceol A, Chatr-aryamontri A, Santonico E, 40. Burns DM, Horn V, Paluh J, Yanofsky C
Sacco R et al (2007) DOMINO: a database of (1990) Evolution of the tryptophan synthetase
domain-peptide interactions. Nucleic Acids of fungi. Analysis of experimentally fused
Res 35:D557–D560 Escherichia coli tryptophan synthetase alpha
28. Amberger J, Bocchini C, Hamosh A (2011) A and beta chains. J Biol Chem 265:2060–2069
new face and new challenges for Online Men- 41. Enright AJ, Iliopoulos I, Kyrpides NC, Ouzou-
delian Inheritance in Man (OMIM®). Hum nis CA (1999) Protein interaction maps for
Mutat 32:564–567 complete genomes based on gene fusion
29. Kandasamy K, Mohan SS, Raju R, Keerthiku- events. Nature 402:86–90
mar S et al (2010) NetPath: a public resource 42. Marcotte EM, Pellegrini M, Ng HL, Rice DW
of curated signal transduction pathways. et al (1999) Detecting protein function and
Genome Biol 11:R3 protein-protein interactions from genome
30. Breuer K, Foroushani AK, Laird MR, Chen C sequences. Science 285:751–753
et al (2013) InnateDB: systems biology of 43. Dandekar T, Snel B, Huynen M, Bork P (1998)
innate immunity and beyond–recent updates Conservation of gene order: a fingerprint of
and continuing curation. Nucleic Acids Res proteins that physically interact. Trends Bio-
41:D1228–D1233 chem Sci 23:324–328
31. Bader GD, Donaldson I, Wolting C, Ouellette 44. Overbeek R, Fonstein M, D’Souza M, Pusch
BF et al (2001) BIND–The Biomolecular GD et al (1999) Use of contiguity on the chro-
Interaction Network Database. Nucleic Acids mosome to predict functional coupling. In
Res 29:242–245 Silico Biol 1:93–108
32. Royer L, Reimann M, Andreopoulos B, 45. Eisen MB, Spellman PT, Brown PO, Botstein
Schroeder M (2008) Unraveling protein net- D (1998) Cluster analysis and display of
works with power graph analysis. PLoS Com- genome-wide expression patterns. Proc Natl
put Biol 4:e1000108 Acad Sci U S A 95:14863–14868
56 Damian Szklarczyk and Lars Juhl Jensen
46. Barrett T, Troup DB, Wilhite SE, Ledoux P PSISCORE: accessing and scoring molecular
et al (2011) NCBI GEO: archive for functional interactions. Nat Methods 8:528–529
genomics data sets–10 years on. Nucleic Acids 56. Sambourg L, Thierry-Mieg N (2010) New
Res 39:D1005–D1010 insights into protein-protein interaction data
47. Hirschman L, Park JC, Tsujii J, Wong L et al lead to increased estimates of the S cerevisiae
(2002) Accomplishments and challenges in lit- interactome size. BMC Bioinformatics 11:605
erature data mining for biology. Bioinformatics 57. Nakayama M, Kikuno R, Ohara O (2002)
18:1553–1561 Protein-protein interactions between large
48. Kuhn M, Szklarczyk D, Franceschini A, von proteins: two-hybrid screening using a func-
Mering C et al (2012) STITCH 3: zooming tionally classified library composed of long
in on protein-chemical interactions. Nucleic cDNAs. Genome Res 12:1773–1784
Acids Res 40:D876–D880 58. Jeong H, Tombor B, Albert R, Oltvai ZN et al
49. Powell S, Szklarczyk D, Trachana K, Roth A (2000) The large-scale organization of meta-
et al (2012) eggNOG v3.0: orthologous bolic networks. Nature 407:651–654
groups covering 1133 organisms at 41 differ- 59. Wuchty S, Oltvai ZN, Barabási A-L (2003)
ent taxonomic ranges. Nucleic Acids Res 40: Evolutionary conservation of motif constitu-
D284–D289 ents in the yeast protein interaction network.
50. McDowall MD, Scott MS, Barton GJ (2009) Nat Genet 35:176–179
PIPs: human protein-protein interaction pre- 60. Von Mering C, Krause R, Snel B, Cornell M
diction database. Nucleic Acids Res 37: et al (2002) Comparative assessment of large-
D651–D656 scale data sets of protein-protein interactions.
51. Chautard E, Fatoux-Ardore M, Ballut L, Nature 417:399–403
Thierry-Mieg N et al (2011) MatrixDB, the 61. Ioannidis JPA (2005) Why most published
extracellular matrix interaction database. research findings are false. PLoS Med 2:e124
Nucleic Acids Res 39:D235–D240 62. Tang JL (2005) Selection bias in meta-analyses
52. Goll J, Rajagopala SV, Shiau SC, Wu H et al of gene-disease associations. PLoS Med 2:e409
(2008) MPIDB: the microbial protein interac- 63. Pál C, Papp B, Hurst LD (2003) Genomic
tion database. Bioinformatics 24:1743–1744 function: rate of evolution and gene dispens-
53. Lynn DJ, Winsor GL, Chan C, Richard N et al ability. Nature 421:496–497, discussion 497–8
(2008) InnateDB: facilitating systems-level 64. Bloom JD, Adami C (2003) Apparent depen-
analyses of the mammalian innate immune dence of protein evolutionary rate on number
response. Mol Syst Biol 4:218 of interactions is linked to biases in protein-
54. The UniProt Consortium (2011) Ongoing and protein interactions data sets. BMC Evol Biol
future developments at the Universal Protein 3:21
Resource. Nucleic Acids Res 39:D214–D219 65. Brito GC, Andrews DW (2011) Removing bias
55. Aranda B, Blankenburg H, Kerrien S, Brink- against membrane proteins in interaction net-
man FSL et al (2011) PSICQUIC and works. BMC Syst Biol 5:169
Chapter 4
Abstract
The prediction of protein-protein interactions and kinase-specific phosphorylation sites on individual
proteins is critical for correctly placing proteins within signaling pathways and networks. The importance
of this type of annotation continues to increase with the continued explosion of genomic and proteomic
data, particularly with emerging data categorizing posttranslational modifications on a large scale. A variety
of computational tools are available for this purpose. In this chapter, we review the general methodologies
for these types of computational predictions and present a detailed user-focused tutorial of one such
method and computational tool, Scansite, which is freely available to the entire scientific community over
the Internet.
Key words Scansite, Protein-protein interaction prediction, Sequence motif, PSSM, Binding motif,
Phosphorylation sites, Bioinformatics
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_4, © Springer Science+Business Media New York 2015
57
58 Tobias Ehrenberger et al.
prediction engine based on known data [8]. The problem with this
approach—as with any other machine learning approach used in
this manner—is that the resulting predictor does not provide
easily decipherable information about why the proteins are likely
to interact. This means that, although it may yield useful results, it
is hard to reconstruct and understand exactly why a prediction is
made by this black-box predictor. A very specific type of machine
learning method tries to find a pattern based on features at the
interaction interface of the proteins involved. A method closely
related to this approach is described later in this chapter. Other
prediction methods are based on genomics. Gene fusion methods
predict that discrete proteins are likely to interact if their homo-
logues are fused into single genomic entities in other species.
Other techniques based on gene neighborhood conservation are
built on the hypothesis that gene pairs within such neighborhoods
that are evolutionary conserved across different species are likely to
interact.
No matter which method is used, it is important to keep the
caveats of the method in mind. First, no prediction can guarantee
either biological correctness or relevance. This is especially impor-
tant if prediction tools are used to design and plan further experi-
ments without first confirming the initial prediction. Failure of a
method to predict an interaction may not reflect a fundamental
problem with the method but may instead reflect limitations of the
data that the method is based on. The data, be it experimentally
verified interaction sites, 3D data of proteins, or other information,
originates from experiments which are all error-prone, though in
some cases the extent of the error may be difficult to estimate. This
also applies to methods that use machine learning to train a predic-
tor, as these methods are highly dependent on the quality of the
underlying training dataset. Obviously, a large set of training data is
necessary to create a good predictor and, indeed, large databases of
experimentally verified protein-protein interactions are now avail-
able. However, training a predictor also requires a negative dataset
that provides information of what interactions are very unlikely to
happen. Experimental data of this type are typically not published, at
least in part due to difficulties in distinguishing whether the lack of an
observed interaction is the result of a technically failed experiment
or because there is no biologically relevant interaction [9]. The
end result is a lack of reliable negative training data for computational
method development. The quality and nature of the training data
should therefore be one important consideration in the user’s choice
of whether to trust a predictor trained on these kinds of data types,
including the species of the proteins in the training dataset, the type of
experiments used to verify the sites, and of course the number of sites
and proteins included. Thus, it is very important for prediction tools
to explicitly (1) give information about how the method works
and what information it uses, (2) provide some type of quantitative
60 Tobias Ehrenberger et al.
manner, such as SH2 and SH3 domains, FHA and BRCT domains,
14-3-3 proteins, etc.
Specific amino acid preferences can be described in two ways.
One is to describe them in a strict combinatorial regular expression-
like pattern (Boolean matching model). This approach was origi-
nally used in PROSITE [14] to search for patterns in a sequence
database. However, these patterns are very inflexible and do not
allow for including differently weighted preferences for amino
acids. A more flexible and powerful approach is the use of
position-specific scoring matrices (PSSMs) to describe patterns/
motifs in this form. This approach was implemented in Scansite
[15, 16], an application to predict short linear sequence motif sites.
A PSSM matrix like this contains a probability value for each amino
acid (columns) at each position of a sequence window of certain
size (rows), where each value in a column and row of the matrix
describes the binding partner’s preference for that amino acid at
that position in the motif. Scansite is a web application that uses
PSSMs to predict interaction sites that are important in cellular
signaling and includes more than 120 kinases and proteins that
recognize specific short linear binding motifs. It can be used to
show all potential sites in a given protein or all proteins in a database
that contain sites for one or more motifs. Directions for both uses
are provided in the following sections.
2 Materials
2.2 Searching a Database Searches only require information about the motif that is
Sequence Database for searched for in a particular sequence database. All of the standard
Motifs Scansite matrices for kinases and modular binding domains are
available. In addition, you may enter more specific information to
restrict the search to a smaller number of proteins.
3 Methods
3.1 Scanning a The key feature of Scansite is the prediction of motif-relevant sites
Protein for Motifs in a given protein. This feature is referred to as Protein Scan or Scan
Proteins for Motifs and allows a range of different inputs.
1. Navigate to Input Page. To get to the Protein Scan input screen
from anywhere in Scansite, click the “Scan Proteins for Motifs”
button in the navigation section on the left-hand side of the
web page.
2. Choose the Protein to Scan. There are two different ways of
choosing proteins in Scansite: by protein identifier (default
option) and by sequence.
To choose a protein by accession number, select “Protein
Accession” from the “Choose Protein by. . .” drop-down list.
Below, select a protein sequence database and enter a protein
ID. Links on the right-hand side of the text boxes refer to the
different sequence databases that Scansite currently supports
and where you can search for protein identifiers. After entering
at least three characters in the text box entitled “Protein Acces-
sion”, Scansite searches for protein IDs that start with these
characters and presents a list of options below the text box. The
same happens when the “Check!” button next to the text box is
clicked or the Enter key is pressed. You can either continue
typing or select an ID from the list. The text box turns green
for valid and red for invalid protein identifiers.
In order to enter a peptide sequence, select “Input
Sequence” from the drop-down list. The area below this menu
will change accordingly. Then, enter or paste a name and an
Protein-Protein Interactions using Scansite 3 63
Fig. 1 The results of a high stringency protein scan for all mammalian motifs using the SwissProt protein
P53_HUMAN and the default reference proteome are shown. The section entitled “Scan Overview” which
summarizes the parameters of the scan of the page is collapsed to better fit this figure on the page
Protein-Protein Interactions using Scansite 3 65
especially helpful if your search did not return any results. The next
part in the page (“Download Results”) offers a link to a download-
able version of the table shown above (tabulator-separated file).
At the bottom of the result page (“Additional Analyses”) users can
directly submit the current protein’s sequence to DisPhos [27], a
Disorder-Enhanced Phosphorylation Site Predictor (see Note 9).
3.2 Searching a The Scansite feature Search Sequence Database for Motifs or short
Sequence Database for Database Search performs a broader search than single protein scans.
Motifs Given a motif (or a set of motifs) and a sequence database, it searches
the database for sequences that contain motif-relevant sites. One of
the most powerful parts of this tool is the option of targeting a
search to specific experimental requirements by restricting searches
to proteins of a specific organism class, species, molecular weight
and isoelectric point range, annotation, and sequence property. For
example, this tool can be used to help identify unknown bands in
two-dimensional (2D) gel electrophoresis experiments.
1. Navigate to Input Page. To get to the Database Search input
screen from anywhere in Scansite, click the button “Search a
Sequence Database for Motifs” in the navigation section on the
left-hand side of the web page.
2. Choose the Search Method. The area below this drop-down list
will change dependent on what you select. Searches for single
“Database motifs” from the Scansite database are the easiest
option to choose. Alternatively, you can search for your own
motifs (see Note 3) or so-called “Quick Motifs” (see Note 10).
It is also possible to search for sequences that match up to five
motifs. These searches can include either database motifs, user-
defined motifs, or a combination of both. The score of a multi-
motif site is the mean (average) of all the scores of the sites
involved. Co-occurrences of different motif sites in proteins
can be filtered in different ways. First of all, it is possible to
penalize gaps between sites of different motifs. Gap penalty
settings are either high, medium, low, or none. Penalties p are
then added to the score according to the maximum distance
dmax between the involved sites (i.e., position of site closest to
C-terminus minus position of site closest to N-terminus). The
penalty values are calculated as follows: plow ¼ 0.001 dmax;
pmedium ¼ 0.01 dmax; phigh ¼ 0.1 dmax. Secondly, it is
possible to define up to three strict minimum and maximum
distance bounds between motif-specific sites. This can be used
if you know which motifs to expect and how far apart you
expect them to be in the protein sequence. If you just want to
get an overview of peptides that have multiple motif sites, it is
recommended to use a gap penalty. Using distance bounds is
the better option for very specific searches.
Protein-Protein Interactions using Scansite 3 67
Fig. 2 The results of a Database Search for ATM in human proteins of SwissProt that are annotated with “cell
cycle” and contain the sequence “ARATT”. Here, only one protein matched the given restrictions and this
protein also contains the motif that was searched for
4 Notes
page that gives information about the gene that recognizes this
motif. Mammalian motifs and yeast-specific motifs are sup-
ported by information from GeneCards [30] and SGD,
respectively.
5. Interpreting Scansite Scores. Scansite’s scores range from 0 to
(theoretically) 1. However, you will never see scores higher
than 5 because sites with scores that high are discarded in the
scoring process. Please be aware that scores in Scansite are
always motif-dependent. This means that scores for different
motifs should not be directly compared to each other. For
example, knowing that one motif’s optimal score is 0.001 and
another motif’s best score is 0.4 it is easy to say that these are
the best possible scores, so hits with these scores are equally
good. However, the only way to extend this knowledge to
slightly poorer scores is to know how likely other scores are to
occur. To make this possible and allow a comparison among
motifs, Scansite offers percentile values. The percentiles used in
Scansite are calculated from the so-called reference proteomes
which are proteomes that are commonly used in research. In
the process of adding a motif to Scansite, it is scored against
every single peptide in the reference proteome and the scores
are stored to create a score distribution. This distribution is
then used to calculate percentile values from scores calculated
when users run certain searches. Using these values it is possible
to rank sites from different motifs.
6. Domains in Scansite 3. Scansite uses InterProScan [28] to
predict a protein’s PFAM domains [31]. Therefore the domain
positions displayed in Scansite may vary by a few amino acids
from the positional assignments seen on the PFAM homepage.
This is mentioned because these variations may cause confusion
but do not pose a problem since all these positions are predic-
tions and there is no way to tell which numbers are more
correct in the absence of clear structural data from crystallo-
graphic or NMR experiments.
7. Previously Mapped Sites in Scansite. Displaying previously
mapped sites in Scansite is only possible for proteins from public
protein databases and works best with proteins from SwissProt.
Please note that these references are only site-specific but not
motif-specific. This means that if a previously mapped site
shows up in the list, the site is reported in the linked databases;
however, this does not imply that the Scansite motif that was
found at this site is related to the site reported in the database.
It could be that a completely different gene is responsible for
this site. Wherever possible, the hyperlinks refer directly to the
external databases page about this site. If a database does not
support direct linking, the link just takes you to the database’s
homepage.
72 Tobias Ehrenberger et al.
References
1. Berman HM, Westbrook J, Feng Z et al (2000) 13. Pinna LA, Maria Ruzzene M (1996) How do
The Protein Data Bank. Nucleic Acids Res protein kinases recognize their substrates? Bio-
28:235–242 chim Biophys Acta 13143:191–225
2. Mathivanan S, Periaswamy B, Gandhi T et al 14. Bairoch A (1992) PROSITE: a dictionary of
(2006) An evaluation of human protein- sites and patterns in proteins. Nucleic Acids
protein interaction data in the public domain. Res 20(Suppl):2013–2018
BMC Bioinformatics 7:S19 15. Yaffe M, Leparc G, Lai J et al (2001) A motif-
3. Turinsky A, Razick S, Turner B, et al. (2010) based profile scanning approach for genome-
Literature curation of protein interactions: wide prediction of signaling pathways. Nat
measuring agreement across major public data- Biotechnol 19:348–353
bases. Database (Oxford) 2010: baq026 16. Obenauer J, Cantley L, Yaffe M (2003) Scan-
4. Shoemaker B, Panchenko A (2007) Decipher- site 2.0: proteome-wide prediction of cell
ing protein-protein interactions. Part II. signaling interactions using short sequence
Computational methods to predict protein motifs. Nucleic Acids Res 31:3635–3641
and domain interaction partners. PLoS Com- 17. M. M, UniProt-consortium (2011) UniProt
put Biol 3:e43 Knowledgebase: a hub of integrated protein
5. Pitre S, Alamgir M, Green J, et al. (2008) data. Database: bar009
Computational methods for predicting 18. Cherry J, Hong E, Amundsen C et al (2011)
protein–protein interactions. In: Advances in Saccharomyces Genome Database: the geno-
biochemical engineering/biotechnology: mics resource of budding yeast. Nucleic Acids
protein-protein interaction. Springer, Res 40(Database issue):D700–D705
Heidelberg, pp 247–267 19. Flicek P, Amode M, Barrell D et al (2011)
6. Andrusier N, Mashiach E, Nussinov R et al Ensembl 2011. Nucleic Acids Res 39(Suppl
(2008) Principles of flexible protein–protein 1):D800–D806
docking. Proteins 73:271–289 20. Burks C, Cassidy M, Cinkosky MJ et al (1991)
7. Janin J (2002) Welcome to CAPRI: a Critical GenBank. Nucleic Acids Res 19:221–225
Assessment of PRedicted Interactions. Proteins 21. Boeckmann B, Bairoch A, Apweiler R et al
Struct Funct Genet 47:257 (2003) The SWISS-PROT protein knowledge-
8. Rhodes DR, Tomlins SA, Varambally S et al base and its supplement TrEMBL in 2003.
(2005) Probabilistic model of the human Nucleic Acids Res 31:365–370
protein-protein interaction network. Nat Bio- 22. Bjellqvist B, Hughes G, Pasquali C et al (1993)
technol 23:951–959 The focusing positions of polypeptides in
9. Trost B, Kusalik A (2011) Computational pre- immobilized pH gradients can be predicted
diction of eukaryotic phosphorylation sites. from their amino acid sequences. Electropho-
Bioinformatics 27:2927–2935 resis 14:1023–1031
10. Hutti J, Jarrell E, Chang J et al (2004) A rapid 23. Hornbeck P, Kornhauser J, Tkachev S et al
method for determining protein kinase (2012) PhosphoSitePlus: a comprehensive
phosphorylation specificity. Nat Methods resource for investigating the structure and
1:27–29 function of experimentally determined
11. Songyang Z, Blechner S, Hoagland N et al post-translational modifications in man and
(1994) Use of an oriented peptide library to mouse. Nucleic Acids Res 40:D261–D270
determine the optimal substrates of protein 24. Dinkel H, Chica C, Via A et al (2011) (2011)
kinases. Curr Biol 4:973–982 Phospho.ELM: a database of phosphorylation
12. Kemp BE, Pearson RB (1990) Protein kinase sites—update 2011. Nucleic Acids Res 39:
recognition sequence motifs. Trends Biochem D261–D267
Sci 15:342–346 25. Gnad F, Ren S, Cox J et al (2007) PHOSIDA
(phosphorylation site database): management,
structural and evolutionary investigation, and
Protein-Protein Interactions using Scansite 3 75
Abstract
Three-dimensional structures of proteins offer an opportunity for the rational design of small molecules to
modulate protein-protein interactions. The presence of a well-defined binding pocket on the surface of
protein complexes, particularly at their interface, can be used for docking-based virtual screening of
chemical libraries. Several approaches have been developed to identify binding pockets that are implemen-
ted in programs such as SiteMap, fpocket, and FTSite. These programs enable the scoring of these pockets
to determine whether they are suitable to accommodate high-affinity small molecules. Virtual screening of
commercial or combinatorial libraries can be carried out to enrich these libraries and select compounds for
further experimental validation. In virtual screening, a compound library is docked to the target protein.
The resulting structures are scored and ranked for the selection and experimental validation of top
candidates. Molecular docking has been implemented in a number of computer programs such as Auto-
Dock Vina. We select a set of protein-protein interactions that have been successfully inhibited with small
molecules in the past. Several computer programs are applied to identify pockets on the surface, and
molecular docking is conducted in an attempt to reproduce the binding pose of the inhibitors. The results
highlight the strengths and limitations of computational methods for the design of PPI inhibitors.
Key words Protein-protein interactions, Molecular docking, Structure-based drug design, Virtual
screening, Small molecules, Inhibitors
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_5, © Springer Science+Business Media New York 2015
77
78 David Xu et al.
2 Methods
2.1 Identifying Starting with the sequence of a gene that encodes for PPI proteins,
Three-Dimensional the sequence of one of the binding partners can be used in a BLAST
Structures of Protein search using the advanced search tool at the Protein Data Bank
Interactions (PDB) [4] (see Note 1). The FASTA format [5] for the protein
sequence can be used in the search. A threshold value known as the
E-value limits the search to proteins that possess significant
sequence identity and coverage of the query sequence. Based on
previous studies [6, 7], a value of 10-6 is expected to identify
protein domains or very close homologs of the query protein
(see Note 2). In addition, the search parameter “Number of Enti-
ties” can be used to filter for protein complexes by setting the Entity
Type to “Protein” with a minimum bound of 2. Different structures
that overlap the same protein sequence can also be removed by
checking the “Remove similar sequences at 90 % identity” box.
Similarly, structures of protein complexes can be identified by
using the Web server Interactome3D [8]. This tool provides struc-
tural annotations of individual proteins as well as protein complexes
in a variety of model organisms, which can be queried by using
UniProt accession IDs. If the protein complex is known, then the
interaction pair can be used in the search. Otherwise, searching for
individual proteins results in a network with all the interactions of
that protein. Clicking on any of the edges in this network yields a
ranked list showing structures from PDB which contain this partic-
ular interaction, as well as other information such as: (1) resolution
of the structure; (2) chain of each of the binding partners in the
Structure-Based Computational Design 79
2.3 Scoring Pockets Various metrics can aid in determining whether a pocket can
accommodate a small molecule. Two metrics have been developed
and implemented in SiteMap for this purpose: SiteScore and Drug-
Score [14]. SiteScore is a measure of whether a pocket can accom-
modate a small molecule, while DrugScore provides information
about the suitability of the pocket for the development of thera-
peutics. Both SiteScore and DrugScore use the weighted sums of
the same parameters, namely (1) the number of site points in the
binding pocket; (2) enclosure score that is a measure of how
accessible the pocket is to solvents; and (3) hydrophilic character
of the binding pocket (hydrophilic score). Unlike DrugScore,
SiteScore limits the impact of hydrophilicity in charged and highly
polar sites. On the other hand, fpocket provides only one metric
known as the Druggability Score [15]. The Druggability Score is a
general logistical model based on the local hydrophobic density of
the binding site, the hydrophobicity score, and the normalized
polarity score. Generally, a SiteMap SiteScore above 0.8, corre-
sponds to a pocket that can accommodate a high-affinity small
molecule. A SiteMap DrugScore above 0.9 [14], or an fpocket
Druggability Score above 0.7 [15] correspond to druggable pock-
ets. FTSite identifies and ranks the detected pockets by the number
of amino acids that are in contact with the probes, but does not give
a quantitative assessment on the druggability of a pocket.
It is often the case that pockets will be identified outside a
protein-protein interface or on the surface of a protein whose
complex with its binding partner has yet to be solved by crystallog-
raphy. In many cases, the binding partners may not even be known.
Potential binding partners can be identified from various datasets
that report PPIs such as BioGRID [16] or MINT [17]. These
datasets typically provide experimental data from yeast two-hybrid
studies or other techniques in tab-delimited or PSI-MI XML for-
mats, or through their web interface. The data can be readily
converted to protein sequence by first retrieving a common identi-
fier, such as the UniProt ID, and then retrieving the FASTA format
sequence from UniProt [18]. Pockets may also potentially be an
enzyme active site. Binding of a small molecule to an enzyme active
site located on a protein–protein complex may potentially act in an
Structure-Based Computational Design 81
2.4 Virtual Screening Structure-based virtual screening is widely used to enrich large
of Chemical Libraries chemical databases to generate focused libraries that can be experi-
to Target Pockets mentally validated [20–22]. To date, only a handful of cases have
been reported whereby compounds that inhibit PPIs emerged
directly from virtual screening [23–26]. In most cases, virtual
screening provides moderate to low affinity compounds that can
serve as a starting point for the development of PPI inhibitors.
Structure-based virtual screening consists of two steps, namely
docking and scoring. Docking corresponds to the series of compu-
tational steps to predict the binding mode of a ligand to a target
protein. The resulting protein–ligand complex provides coordi-
nates that can be used to score and rank-order the interactions.
In virtual screening, it is not uncommon that hundreds of
thousands of compounds are docked to a target, followed by
scoring for the selection of the top ~100 compounds.
Several computer programs have been developed for docking,
such as DOCK [27, 28], AutoDock [29], GOLD [30, 31], FlexX
[32], Glide [33, 34] among others [35–39]. AutoDock is one of
the most cited open-source molecular modeling simulation dock-
ing programs. It was designed, implemented, and maintained by
The Scripps Research Institute. Its latest version, Vina [40], was
released in 2010. Vina inherits major ideas and approaches from
AutoDock. By using a new source code, scoring function, and
algorithms, Vina significantly improves the average accuracy of
the binding mode prediction compared to AutoDock.
As described above, docking requires a three-dimensional
structure of a protein as well as a pocket on the surface. Typically,
a simple visualization of the structure can identify the location of
the pocket of interest. For PPIs, pockets located at the interface are
the most desirable since compounds that bind to these pockets are
expected to disrupt the interaction. Pockets that include hot-spot
residues are particularly attractive. The hot-spots can be located
within the binding partner that contains the pocket or on the
partner that occupies the pocket [25, 41–44]. If the location of
the pocket is unknown, it can be identified using programs such as
SiteMap or fpocket (see above) if necessary. Once a pocket is iden-
tified, a grid box based at the center of the pocket is created. The
pre-calculated grid maps obviate the need to calculate the interac-
tion energies at each step of the docking process. This results in a
significantly faster docking run (see Note 4). Vina generates the
map and carries out the energy calculations.
82 David Xu et al.
3 Case Studies
Table 1
Pockets identified by the various detection programs
4 Notes
PDB Compound structure Compound name Binding energy (kcal/mol) RMSD (Å)
1PY2 SP4206 7.7 1.06
87
(continued)
Table 2
88
(continued)
David Xu et al.
PDB Compound structure Compound name Binding energy (kcal/mol) RMSD (Å)
1R6N Compound 23 7.5 6.65
2. The cutoff used in the BLAST search will only identify PDB
entries that are nearly identical to the initial sequence query. If
structures that you expect to see are not returned, lowering the
cutoff will result in a larger number of potential structures.
However, a cutoff that is too low will often yield structures
that have very low sequence identity to the original.
3. Sometimes there are no atomic coordinates for residues within
a PDB entry. Often, these missing residues occur at the begin-
ning or end of the PDB sequence, or in intrinsically disordered
regions, which are difficult to crystallize. If these missing resi-
dues are within the proximity of a known binding cavity, using
an alternative crystal structure from the BLAST search can yield
a PDB structure that has these coordinates available. Other-
wise, homology modeling can be used to thread the missing
residues into a homologous structure. This can be accom-
plished with software packages such as MODELLER or Prime.
4. During docking, the ligands can dock outside of the specified
binding pocket if the grid is too large. In this case, either visual
inspection or a computational approach can determine which
ligands, if any, are outside the binding pocket. In the computa-
tional approach, the distance between the center of mass of the
binding pocket and the center of mass of the docked ligand can
be used to determine which ligands are outside the binding
cavity.
References
10. Halgren T (2007) New method for fast 26. Geppert T, Bauer S, Hiss JA et al (2012)
and accurate binding-site identification and Immunosuppressive small molecule discovered
analysis. Chem Biol Drug Des 69:146–148 by structure-based virtual screening for inhibi-
11. Le Guilloux V, Schmidtke P, Tuffery P (2009) tors of protein–protein interactions. Angew
Fpocket: an open source platform for ligand Chem Int Edit 51:258–261
pocket detection. BMC Bioinformatics 10:168 27. Kuntz ID, Blaney JM, Oatley SJ et al (1982) A
12. Kuhn D, Weskamp N, Hullermeier E et al geometric approach to macromolecule-ligand
(2007) Functional classification of protein interactions. J Mol Biol 161:269–288
kinase binding sites using cavbase. ChemMed- 28. Makino S, Kuntz ID (1997) Automated flexi-
Chem 2:1432–1447 ble ligand docking method and its application
13. Ngan CH, Hall DR, Zerbe B et al (2012) for database search. J Comput Chem
FTSite: high accuracy detection of ligand bind- 18:1812–1825
ing sites on unbound protein structures. Bioin- 29. Goodsell DS, Olson AJ (1990) Automated
formatics 28:286–287 docking of substrates to proteins by simulated
14. Halgren TA (2009) Identifying and character- annealing. Proteins 8:195–202
izing binding sites and assessing druggability. J 30. Jones G, Willett P, Glen RC et al (1997) Devel-
Chem Inf Model 49:377–389 opment and validation of a genetic algorithm
15. Schmidtke P, Barril X (2010) Understanding for flexible docking. J Mol Biol 267:727–748
and predicting druggability. A high- 31. Jones G, Willett P, Glen RC (1995) Molecular
throughput method for detection of drug recognition of receptor sites using a genetic
binding sites. J Med Chem 53:5858–5867 algorithm with a description of desolvation.
16. Stark C, Breitkreutz BJ, Reguly T et al (2006) J Mol Biol 245:43–53
BioGRID: a general repository for interaction 32. Rarey M, Kramer B, Lengauer T et al (1996) A
datasets. Nucleic Acids Res 34:D535–D539 fast flexible docking method using an incre-
17. Licata L, Briganti L, Peluso D et al (2012) mental construction algorithm. J Mol Biol
MINT, the molecular interaction database: 261:470–489
2012 update. Nucleic Acids Res 40:D857–D861 33. Friesner RA, Banks JL, Murphy RB et al (2004)
18. UniProt C (2012) Reorganizing the protein Glide: a new approach for rapid, accurate dock-
space at the Universal Protein Resource (Uni- ing and scoring. 1. Method and assessment of
Prot). Nucleic Acids Res 40:D71–D75 docking accuracy. J Med Chem 47:1739–1749
19. Porter CT, Bartlett GJ, Thornton JM (2004) 34. Halgren TA, Murphy RB, Friesner RA et al
The catalytic site atlas: a resource of catalytic (2004) Glide: a new approach for rapid, accu-
sites and residues identified in enzymes using rate docking and scoring. 2. Enrichment fac-
structural data. Nucleic Acids Res 32: tors in database screening. J Med Chem
D129–D133 47:1750–1759
20. Leach AR, Gillet VJ, Lewis RA et al (2009) 35. Pierce BG, Hourai Y, Weng Z (2011) Acceler-
Three-dimensional pharmacophore methods ating protein docking in ZDOCK using an
in drug discovery. J Med Chem 53:539–558 advanced 3D convolution library. PLoS One
21. Hubbard RE (2011) Structure-based drug dis- 6:e24657
covery and protein targets in the CNS. Neuro- 36. McGann M (2011) FRED pose prediction and
pharmacology 60:7–23 virtual screening accuracy. J Chem Inf Model
22. Cheng T, Li Q, Zhou Z et al (2012) Structure- 51:578–596
based virtual screening for drug discovery: a 37. Pedretti A, Villa L, Vistoli G (2004) VEGA –
problem-centric review. AAPS J 14:133–141 an open platform to develop chemo-bio-infor-
23. Khanna M, Wang F, Jo I et al (2011) Targeting matics applications, using plug-in architecture
multiple conformations leads to small molecule and script programming. J Comput-Aided Mol
inhibitors of the uPAR·uPA protein–protein Des 18:167–173
interaction that block cancer cell invasion. 38. Thomsen R, Christensen MH (2006) MolDock:
ACS Chem Biol 6:1232–1243 a new technique for high-accuracy molecular
24. Scheper J, Guerra-Rebollo M, Sanclimens G docking. J Med Chem 49:3315–3321
et al (2010) Protein–protein interaction 39. Abagyan R, Totrov M, Kuznetsov D (1994)
antagonists as novel inhibitors of non-canonical ICM – a new method for protein modeling
polyubiquitylation. PLoS One 5:e11403 and design: applications to docking and
25. Koes D, Khoury K, Huang Y et al (2012) structure prediction from the distorted native
Enabling large-scale design, synthesis and vali- conformation. J Comp Chem 15:488–506
dation of small molecule protein–protein 40. Trott O, Olson AJ (2010) AutoDock Vina:
antagonists. PLoS One 7:e32839 improving the speed and accuracy of docking
Structure-Based Computational Design 91
with a new scoring function, efficient optimiza- model for HIV-1 integrase. J Med Chem
tion, and multithreading. J Comput Chem 43:2100–2114
31:455–461 54. Kukol A (2011) Consensus virtual screening
41. Obiol-Pardo C, Alcarraz-Vizán G, Cascante M approaches to predict protein ligands. Eur J
et al (2012) Diphenyl urea derivatives as inhi- Med Chem 46:4661–4664
bitors of transketolase: a structure-based virtual 55. Irwin JJ, Sterling T, Mysinger MM et al (2012)
screening. PLoS One 7:e32276 ZINC: a free tool to discover chemistry for
42. Dessal AL, Prades R, Giralt E et al (2011) biology. J Chem Inf Model 52:1757–1768
Rational design of a selective covalent modifier 56. Greenwood JR, Calkins D, Sullivan AP et al
of G protein βγ subunits. Mol Pharm 79:24–33 (2010) Towards the comprehensive, rapid,
43. Trosset J-Y, Dalvit C, Knapp S et al (2006) and accurate prediction of the favorable tauto-
Inhibition of protein–protein interactions: the meric states of drug-like molecules in aqueous
discovery of druglike β-catenin inhibitors by solution. J Comput Aid Mol Des 24:591–604
combining virtual and biophysical screening. 57. Cramer CJ, Truhlar DG (1992) An SCF solva-
Proteins 64:60–67 tion model for the hydrophobic effect and
44. Gr€uneberg S, Stubbs MT, Klebe G (2002) absolute free energies of aqueous solvation.
Successful virtual screening for novel inhibitors Science 256:213–217
of human carbonic anhydrase: strategy and 58. Cramer CJ, Truhlar DG (1992) AM1-SM2 and
experimental confirmation. J Med Chem PM3-SM3 parameterized SCF solvation mod-
45:3588–3602 els for free energies in aqueous solution.
45. Elokely KM, Doerksen RJ (2013) Docking J Comput Aided Mol Des 6:629–666
Challenge: Protein Sampling and Molecular 59. Hawkins PCD, Skillman AG, Nicholls A
Docking Performance. J Chem Inf Model (2006) Comparison of shape-matching and
53:1934–1945 docking as virtual screening tools. J Med
46. Lill MA, Winiger F, Vedani A et al (2005) Chem 50:74–82
Impact of Induced Fit on Ligand Binding to 60. Tetko IV, Gasteiger J, Todeschini R et al
the Androgen Receptor: A Multidimensional (2005) Virtual computational chemistry
QSAR Study To Predict Endocrine-Disrupting laboratory-design and description. J Comput-
Effects of Environmental Chemicals. J Med Aided Mol Des 19:453–463
Chem 48:5666–5674 61. Gaulton A, Bellis LJ, Bento AP et al (2012)
47. Sherman W, Day T, Jacobson MP et al (2005) ChEMBL: a large-scale bioactivity database for
Novel procedure for modeling ligand/receptor drug discovery. Nucleic Acids Res 40:
induced fit effects. J Med Chem 49:534–553 D1100–D1107
48. Arooj M, Sakkiah S, Kim S et al (2013) A com- 62. Li Q, Cheng T, Wang Y et al (2010) PubChem
bination of receptor-based pharmacophore as a public resource for drug discovery. Drug
modeling & QM techniques for identification Discov Today 15:1052–1057
of human chymase inhibitors. PLoS One 8: 63. Knox C, Law V, Jewison T et al (2011) Drug-
e63030 Bank 3.0: a comprehensive resource for
49. Zhou S, Li Y, Hou T (2013) Feasibility of “omics” research on drugs. Nucleic Acids Res
using molecular docking-based virtual screen- 39:D1035–D1041
ing for searching dual target kinase inhibitors. J 64. Liu T, Lin Y, Wen X et al (2007) BindingDB: a
Chem Inf Model 53:982–996 web-accessible database of experimentally
50. Li Y, Kim DJ, Ma W et al (2011) Discovery of determined protein–ligand binding affinities.
novel checkpoint kinase 1 inhibitors by virtual Nucleic Acids Res 35:D198–D201
screening based on multiple crystal structures. 65. Still WC, Tempczyk A, Hawley RC et al (1990)
J Chem Inf Model 51:2904–2914 Semianalytical treatment of solvation for
51. Isvoran A, Badel A, Craescu C et al (2011) molecular mechanics and dynamics. J Am
Exploring NMR ensembles of calcium binding Chem Soc 112:6127–6129
proteins: perspectives to design inhibitors of 66. Luo R, David L, Gilson MK (2002) Acceler-
protein–protein interactions. BMC Struct Biol ated Poisson–Boltzmann calculations for static
11:24 and dynamic systems. J Comput Chem
52. Knegtel RMA, Kuntz ID, Oshiro CM (1997) 23:1244–1253
Molecular docking to ensembles of protein 67. Eldridge MD, Murray CW, Auton TR et al
structures. J Mol Biol 266:424–440 (1997) Empirical scoring functions: I. The
53. Carlson HA, Masukawa KM, Rubins K et al development of a fast empirical scoring func-
(2000) Developing a dynamic pharmacophore tion to estimate the binding affinity of ligands
92 David Xu et al.
in receptor complexes. J Comput Aided Mol 73. Malek TR (2003) The main function of IL-2 is
Des 11:425–445 to promote the development of T regulatory
68. Li L, Wang B, Meroueh SO (2011) Support cells. J Leukoc Biol 74:961–965
vector regression scoring of receptor–ligand 74. Willis S, Day CL, Hinds MG et al (2003) The
complexes for rank-ordering and virtual Bcl-2-regulated apoptotic pathway. J Cell Sci
screening of chemical libraries. J Chem Inf 116:4053–4056
Model 51:2132–2138 75. Moll UM, Petrenko O (2003) The MDM2-
69. Muegge I, Martin YC (1999) A general and p53 interaction. Mol Cancer Res 1:1001–1008
fast scoring function for protein ligand 76. Muller M, Demeret C (2012) The HPV E2-
interactions: a simplified potential approach. host protein–protein interactions: a complex
J Med Chem 42:791–804 hijacking of the cellular network. Open Virol J
70. Gohlke H, Hendlich M, Klebe G (2000) 6:173–189
Knowledge-based scoring function to predict pro- 77. Hughes FJ, Romanos MA (1993) E1 protein
tein–ligand interactions. J Mol Biol 295:337–356 of human papillomavirus is a DNA helicase/
71. Li L, Khanna M, Jo I et al (2011) Target- ATPase. Nucleic Acids Res 21:5817–5823
specific support vector machine scoring in 78. Pazos M, Natale P, Vicente M (2013) A specific
structure-based virtual screening: computa- role for the ZipA protein in cell division: stabi-
tional validation, in vitro testing in kinases, lization of the FtsZ protein. J Biol Chem
and effects on lung cancer cell proliferation. 288:3219–3226
J Chem Inf Model 51:755–759 79. Locksley RM, Killeen N, Lenardo MJ (2001)
72. Wells JA, McClendon CL (2007) Reaching for The TNF and TNF receptor superfamilies:
high-hanging fruit in drug discovery at protein– integrating mammalian biology. Cell 104:
protein interfaces. Nature 450:1001–1009 487–501
Chapter 6
Abstract
Protein-protein interactions are associated with key activities and pathways in the cell, and in that regard are
promising targets for drug discovery. However, in terms of small molecule drugs, this promise has not been
realized. The physical nature of many protein-protein interaction surfaces renders them unable to support
binding of small drug-like molecules. In addition, there are other unique hurdles presented by this class that
make the drug development process difficult and risky. Nevertheless, success stories have begun to steadily
appear in this field. These experiences are starting to provide general strategies and tools to help overcome
the problems inherent in pursuing protein‐protein interaction targets. These lessons should improve the
rate of success as these systems are pursued in the future.
Key words Protein-protein interactions, Drug discovery, MDM2, Nutlins, Druggability, Protein
NMR, NMR screening, Screening library, Fragment-based drug discovery
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_6, © Springer Science+Business Media New York 2015
93
94 David C. Fry
bind there [3–5]. These surfaces are typically large and flat, and
devoid of significant subpockets. Binding affinity is achieved by
summing up a large number of weak interactions. The interactions
are so widely spaced that a small molecule cannot adequately dupli-
cate them. However, this situation is a generalization, and not all
protein‐protein systems adopt such a strategy. Pioneering work by
Jim Wells and colleagues demonstrated that, in some cases, a lim-
ited number of amino acids mediate nearly all of the key interac-
tions that produce binding affinity. These sub-regions have been
referred to as “hot spots,” and their dimensions can be comparable
to the size of a small organic molecule [6].
Therefore, protein‐protein interaction systems that are candi-
dates for drug discovery must be judged for druggability on a
case-by-case basis. The approaches and techniques for making
such a judgment are still being refined and are not fully reliable
yet. As a consequence, embarking on a drug discovery program
targeting a protein‐protein interaction system is still a high-risk
undertaking. An unwillingness to take on such risk has meant that
protein‐protein targets have not received the same degree of active
pursuit as other target classes. This is one of the major reasons for
the paucity of marketed drugs with a mode of action that involves
modulation of a protein‐protein interaction.
Another reason for lack of success against this target class is the
inability to overcome the unique hurdles that are presented by this
class during the drug discovery and development process. The field
is gaining an awareness of, and an appreciation for, these hurdles,
and is acquiring an understanding of how to overcome them, as
success stories steadily appear in this area. It has been helpful to
examine, at a molecular level, cases in which a drug-like small
molecule has been successfully developed against a protein‐protein
interaction target, and a collective consideration of these examples
has led to important lessons that should improve the means by
which these targets are addressed in the future.
Fig. 1 Series of one single and four superimposed X-ray structures showing how the p53 peptide binds to
MDM2 [8] (a), and how various small molecule inhibitors [7, 34–36] are able to mimic its binding strategy
(b–e). MDM2 is depicted as a tan surface. The p53 peptide backbone is shown as a green ribbon, and key side
chains (Phe19, Trp23, and Leu26) as green sticks. The inhibitors are shown in stick form. All structural figures
in this chapter were prepared using the PyMol Molecular Graphics System (Schroedinger, LLC)
Given the experience gained from many attempts over the years at
drug discovery involving protein‐protein systems, and the
recent availability of numerous successful examples in this area, a
generalized optimal approach can be laid out for an attack on
a protein‐protein target:
3.1 Choosing The target will be chosen based on biology considerations, but then
Targets/Assessing must be quickly put under scrutiny at a molecular level [24]. If a
Druggability drug-like small molecule modulator is already known for the system
in question, then by definition it is druggable. Otherwise, if struc-
tural information is available for the target protein, a druggability
assessment can be made by computational methods [25–28]. These
methods have not yet proven to be completely accurate, but should
be a reasonable guide at the extremes—for example, a target with a
complete lack of subpockets at the interface is almost certain to be
highly challenging. Given the unreliability of the computational
methods, an inexpensive experimental method for assessing drugg-
ability is advisable. The most established approach is to perform a
fragment screen. It has been shown in two retrospective studies that
there is a correlation between the inability to find fragment hits for
a target and its eventual failure in a small molecule drug discovery
program.
3.2 The Role of A helpful first step is identification of a peptide that can serve as a
Peptides fully competent surrogate for the entire partner protein. This can
be accomplished via design, based on structural or mutagenesis
information; by phage display; or by a screen of a peptide library.
Once obtained, a surrogate peptide can serve as a useful tool for
assay development and calibration, and for structural studies. One
should even consider developing the peptide itself into a drug. The
methodologies for doing this represent an area of intense current
interest [29], but an exposition is beyond the scope of this article.
3.3 Computational Experimental screens are expensive, and any method that can effi-
Screening and De Novo ciently focus such a screen, or bypass it altogether, is worthwhile. If
Design sufficient structural information is available on the target, a virtual
screen can be carried out. This requires the preparation of a virtual
compound library, where choices must be made about the confor-
mations of the small molecules that will be sampled. Also, it is
100 David C. Fry
3.5 Protein-Protein It has already been noted that modulators of protein‐protein inter-
Focused Screening actions have unique properties, in particular that they tend to be
Library larger and more three-dimensional than typical drugs. We have
observed in screens of protein‐protein targets that the hits are
statistically more three-dimensional than the hits from screens
involving other target classes [23]. These extremes of size and
shape tend to be underrepresented in typical corporate compound
libraries. To remedy this, we have built a “PPI Library,” which is a
collection of compounds for screening that have substantial three-
dimensionality. This library has already shown promise by produc-
ing authenticated hits against protein‐protein interaction targets
that had yielded very low hits rates in earlier screens using our
conventional library [30].
102 David C. Fry
Fig. 3 Deconstruction of a Nutlin into fragments. The parent molecule is shown at the top as a chemical
structure and, to the immediate right, as a simplified schematic depiction. The various fragments are shown
below in schematic form, and their binding behavior to MDM2 as assessed by protein-observe NMR is given,
expressed as “Yes” or “No”
4 Future Outlook
References
1. Stumpf MPH, Thorne T, de Silva E, Stewart R, Adams PD (eds) Protein‐protein interactions:
An HJ, Lappe M, Wiuf C (2008) Estimating a molecular cloning manual. Cold Spring
the size of the human interactome. Proc Nat Harbor Laboratory, Cold Spring Harbor, NY,
Acad Sci 105:6959–6964 pp 893–906
2. Overington JP, Al-Lazikani B, Hoopkins AL 10. Fry DC, Graves BJ, Vassilev LT (2005) Devel-
(2006) How many drug targets are there? Nat opment of E3-substrate (MDM2-p53) binding
Rev Drug Disc 5:993–996 inhibitors: structural aspects. Methods Enzy-
3. Fry DC (2006) Protein‐protein interactions as mol 399C:622–633
targets for small molecule drug discovery. Bio- 11. Vu BT, Vassilev L (2011) Small-molecule inhi-
polymers 84:535–552 bitors of the p53-MDM2 interaction. In: Vas-
4. Wells JA, McClendon CL (2007) Reaching for silev L, Fry D (eds) Small-molecule inhibitors
high-hanging fruit in drug discovery at protein‐ of protein‐protein interactions. Springer, Ber-
protein interfaces. Nature 450:1001–1009 lin, pp 151–172
5. Doemling A (2008) Small molecular weight 12. Fry DC, Emerson SD, Palme S, Vu BT, Liu
protein‐protein interaction antagonists – an CM, Podlaski F (2004) NMR structure of a
insurmountable challenge? Curr Opin Chem complex between MDM2 and a small molecule
Biol 12:281–291 inhibitor. J Biomol NMR 30:163–173
6. Clackson T, Wells JA (1995) A hot spot of 13. Wang S, Zhao Y, Bernard D, Aguilar A,
binding energy in a hormone-receptor inter- Kumar S (2012) Targeting the MDM2-p53
face. Science 267:383–386 protein‐protein interaction for new cancer
7. Vassilev LT, Vu BT, Graves B, Carvajal D, therapies. Top Med Chem 8:57–80
Podlaski F, Filipovic Z, Kong N, Kammlott 14. Graves B, Thompson T, Xia M, Janson C,
U, Lukacs C, Klein C, Fotouhi N, Liu E Lukacs C, Deo D, DiLello P, Fry D, Garvie
(2004) In vivo activation of the p53 pathway C, Huang K, Gao L, Tovar C, Lovey A, Wanner
by small-molecule antagonists of MDM2. Sci- J, Vassilev L (2012) Activation of the p53 path-
ence 303:844–848 way by small-molecule induced MDM2 and
8. Kussie PH, Gorina S, Marechal V, Elenbaas B, MDMX dimerization. Proc Nat Acad Sci
Moreau J, Levine AJ, Pavletich NP (1996) 109:11788–11793
Structure of the MDM2 oncoprotein bound 15. Arkin MR, Wells JA (2004) Small-molecule
to the p53 tumor suppressor transactivation inhibitors of protein‐protein interactions:
domain. Science 274:948–953 progressing towards the dream. Nat Rev Drug
9. Fry DC, Graves BJ, Vassilev LT (2005) Disc 3:301–317
Exploiting protein‐protein interactions to 16. Fry DC (2008) Drug-like inhibitors of protein‐
design an activator of p53. In: Golemis EA, protein interactions: a structural examination
106 David C. Fry
of effective protein mimicry. Curr Prot Pep Sci binding pockets for drug discovery. Nucleic
9:240–247 Acids Res 38:W407–W411
17. Sperandio O, Reynes CH, Camproux AC, 28. Sugaya N, Kanai S, Furuya T (2012) Dr. PIAS
Villoutreix BO (2010) Rationalizing the chem- 2.0: an update of a database of predicted drug-
ical space of protein‐protein interaction inhibi- gable protein‐protein interactions. Database
tors. Drug Disc Today 15:220–229 2012:bas034
18. Higueruelo AP, Schreyer A, Bickerton GRJ, 29. Vlieghe P, Lisowski V, Martinez J, Khrestcha-
Pitt WR, Groom CR, Blundell TL (2009) tisky M (2009) Synthetic therapeutic peptides:
Atomic interactions and profile of small mole- science and market. Drug Disc Today 15:40–56
cules disrupting protein‐protein interfaces: the 30. Fry D, Huang K-S, Di Lello P, Mohr P, Mueller
TIMBAL database. Chem Biol Drug Des 74: K, So S-S, Harada T, Stahl M, Vu B, Mauser H
457–467 (2013) Design of libraries targeting protein‐pro-
19. Morelli X, Bourgeas R, Roche P (2011) Chem- tein interfaces. Chem Med Chem 8:726–732
ical and structural lessons from recent successes 31. Murray CW, Rees DC (2009) The rise of
in protein‐protein interaction inhibition fragment-based drug discovery. Nat Chem 1:
(2P2I). Curr Opin Chem Biol 15:1–7 187–192
20. Fry DC (2012) Small-molecule inhibitors of 32. Hajduk PJ (2006) Fragment-based drug
protein‐protein interactions: how to mimic a design: how big is too big? J Med Chem
protein partner. Curr Pharm Des 18: 49:6972–6976
4679–4684 33. Fry DC, Wartchow C, Graves B, Janson C,
21. Cerchietti LC, Ghetu AF, Zhu X, DaSilva GF, Lukacs C, Kammlott U, Beluins C, Palme S,
Zhong S, Matthews M, Bunting KL, Plol JM, Klein C, Vu B (2013) Deconstruction of a
Fares C, Arrowsmith CH, Yang SN, Garcia M, nutlin: dissecting the binding determinants of
Coop A, MacKerell AD, Prive GG, Melnick A a potent protein‐protein interaction inhibitor.
(2010) A small-molecule inhibitor of BCL6 ACS Med Chem Lett 4(7):660–665
kills DLBCL cells in vitro and in vivo. Cancer 34. Grasberger BL, Lu T, Schubert C, Parks DJ,
Cell 17:400–411 Carver TE, Koblish HK, Cummings MD,
22. Buckley DL, Van Molle I, Gareiss PC, Tae HS, LaFrance LV, Milkiewicz KL, Calvo RR,
Michel J, Noblin DJ, Jorgensen WL, Ciulli A, Maguire D, Lattanze J, Franks CF, Zhao S,
Crews CM (2012) Targeting the von Hippel- Ramachandren K, Bylebyi GR, Zhang M, Man-
Lindau E3 ubiquitin ligase using small mole- they CL, Petrella EC, Pantoliano MW, Deck-
cules to disrupt the VHL/HIF-1alpha interac- man IC, Spurlino JC, Maroney AC, Tomczuk
tion. J Am Chem Soc 134:4465–4468 BE, Molloy CJ, Bone RF (2005) Discovery and
23. Fry DC, So S-S (2012) Modulators of protein‐ cocrystal structure of benzodiazepinedione
protein interactions: the importance of three- HDM2 antagonists that activate p53 in cells.
dimensionality. In: Doemling A (ed) Protein‐ J Med Chem 48:909–912
protein interactions in drug discovery. Wiley- 35. Allen JG, Bourbeau MP, Wohlhieter GE, Bart-
VCH, Weinhelm, pp 55–62 berger MD, Michelsen K, Hungate R, Gad-
24. Wanner J, Fry DC, Peng Z, Roberts J (2011) wood RC, Gaston RD, Evans B, Mann LW,
Druggability assessment of protein‐protein Matison ME, Schneider S, Huang X, Yu D,
interfaces. Future Med Chem 3:2021–2038 Andrews PS, Reichelt A, Long AM, Yakowec
25. Fuller JC, Burgoyne NJ, Jackson RM (2008) P, Yang E, Lee TA, Oliner JD (2009) Discovery
Predicting druggable binding sites at the pro- and optimization of chromenotriazolopyrimi-
tein‐protein interface. Drug Disc Today 14: dines as potent inhibitors of the mouse double
155–161 minute 2 – tumor protein 53 protein interac-
26. Bourgeas R, Basse MJ, Morelli X, Roche P tion. J Med Chem 52:7044–7053
(2010) Atomic analysis of protein‐protein 36. Popowicz GM, Czarna A, Wolf S, Wang K,
interfaces with known inhibitors: the 2P2I Wang W, Doemling A, Holak T (2010) Struc-
database. PLoS One 5:e9598 tures of low molecular weight inhibitors bound
27. Meireles LMC, Doemling AS, Camacho CJ to MDMX and MDM2 reveal new approaches
(2010) ANCHOR: a web server and database for p53-MDMX/MDM2 antagonist drug dis-
for analysis of protein‐protein interaction covery. Cell Cycle 9:1–8
Part II
Abstract
Protein-protein interactions regulate many important cellular processes, including carbohydrate and lipid
metabolism, cell cycle and cell death regulation, protein and nucleic acid metabolism, signal transduction,
and cellular architecture. A complete understanding of cellular function depends on full characterization of
the complex network of cellular protein-protein interactions, including measurements of their kinetic and
binding properties. Surface plasmon resonance (SPR) is one of the commonly used technologies for
detailed and quantitative studies of protein-protein interactions and determination of their equilibrium
and kinetic parameters. SPR provides excellent instrumentation for a label-free, real-time investigation of
protein-protein interactions. This chapter details the experimental design and proper use of the instrumen-
tation for a kinetic experiment. It will provide readers with basic theory, assay setup, and the proper way of
reporting this type of results with practical tips useful for SPR-based studies. A generic protocol for
immobilizing ligands using amino coupling chemistry, also useful if an antibody affinity capture approach
is used, performing kinetic studies, and collecting and analyzing data is described.
Key words Protein-protein interactions, Surface plasmon resonance, Real-time and label-free
biosensor applications, Kinetics, Affinity, Association, Dissociation
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_7, © Springer Science+Business Media New York 2015
109
110 Zaneta Nikolovska-Coleska
1.1 Surface Plasmon SPR is an optical-based real-time detection method for monitoring
Resonance: Principle specific binding events between two or more biomolecules, without
the use of labels, and provides quantitative information on the
specificity, kinetics and affinity of the biomolecular interactions
[11–13]. SPR can be also used for thermodynamic analysis, epitope
mapping, and to determine analyte concentration [14–16].
A unique approach to protein investigations is enabled by the
combination of SPR and mass spectrometry, which increases limits
Studying Protein-Protein Interactions 111
Flow
Analyte
Flow channel
Ligand
Carboxymethylated Sensor chip
dextran
surface
Gold surface
dq
Dissociation
Association
Time (sec)
Injection of analyte Running buffer
1.2 Sensor Surface The first step in the interaction analysis is the surface preparation
Preparation/ and immobilization of one of the binding partners (the “ligand”)
Immobilization on the sensor chip surface. Several factors should be considered for
this procedure and include selecting (1) the binding partner for the
immobilization, (2) the appropriate immobilization level, and (3)
the method to be used for immobilization, and consequently, the
sensor chip that will be selected for use.
1.2.1 Selection of Ligand Several properties can influence the decision as to which ligand will
be immobilized on the chip. These include the molecular weight of
binding partners, their purity, the number of binding sites, the
functionality of the immobilized ligand, the isoelectric point (pI)
of the protein(s), the amount of ligand available, and the assay
conditions.
When direct coupling is used as an immobilization method, it is
important that the purity of the ligand be >95 %. The immobilized
ligand should be stable and the binding activity must survive the
coupling procedure as well as the regeneration protocol. During
SPR experiments, binding is measured as a change in the refractive
index on the surface of the sensor chip caused by accumulation of
mass within the surface dextran layer and the measured response is
related to the mass of the bound analyte. Therefore, when possible,
the smallest and most stable molecule in the system should be used
as the immobilized component. The larger molecule of the pair
Studying Protein-Protein Interactions 113
1.2.2 Immobilization The binding capacity of the sensor chip surface depends on the
Levels immobilization level and the optimal amount of the immobilized
ligand depends on the experimental goal. For example, higher
levels of immobilized ligand are needed in experiments designed
to determine the concentration of analytes in solution, while for
kinetic analysis the immobilization level of the ligand should be low.
The maximum binding capacity (Rmax) of the immobilized ligand
recommended in the literature is typically in the range of 50–150
resonance units (RU), depending on the relative molecular weights
and sizes of ligand and analyte. An overloaded surface causes
problems of steric hindrance, aggregation, rebinding and diffu-
sional limitations such as mass transport. Mass transport limitations
can result in calculated binding kinetics that are slower than the
true binding parameters. Problems due to mass transport effects are
complex and they are best avoided by use of a low density surface to
provide a maximum binding response level (Rmax) of no more than
50–100 RU, and high flow rates, preferably 100 μL/min.
The maximum response (Rmax) that can be obtained in experi-
ments depends directly on the molecular weight of the protein-protein
complex that will be formed on the surface after flowing the analyte
molecule. The theoretical Rmax describes the maximum binding capac-
ity of the surface, which is usually higher than the experimental Rmax.
The theoretical Rmax can be calculated using the following equation:
MW analyte
Rmax ¼ RL Sm
MW ligand
Experimental Rmax
% ligand activity ¼ 100 %
Theoretical Rmax
Poor surface activity can result from denaturation of the ligand as a
result of the pre-concentration solution, blocking and inactivating
the binding site by immobilization chemistry, or impurities present
in the ligand which can also be immobilized, lowering the surface
activity. In these cases, changes in the coupling chemistry or ligand
capture approach should be considered in order to improve the
surface activity.
1.2.3 Immobilization Direct covalent coupling and affinity capturing techniques are two
Method general methods for immobilization of the ligand. Covalent cou-
pling is the most commonly used means of immobilizing the
ligand. In this technique, the ligand is directly immobilized on
the sensor surface using established coupling chemistry depending
on the available reactive groups, for example amine, thiol, or alde-
hyde groups in the protein. In such cases, direct immobilization
does not require any modification of the ligand. The sensor chip,
CM5, is the most versatile available chip for covalent coupling of
ligands with high binding capacity (Table 1). It gives high response
and excellent chemical stability providing accuracy and allowing
repeated analyses on the same surface. The immobilization level is
easily controlled and ligand consumption is low. The disadvantage
of this immobilization method is that it can be associated with
multiple attachment sites leading to randomized coupling and
heterogeneous orientation of the ligand.
In contrast to direct immobilization, the affinity capturing
method offers steric orientation of the immobilized interaction
partner for optimum site exposure and assurance that all immobi-
lized molecules will be in the same orientation, resulting in a highly
active and homogeneous surface (Fig. 2a). The method relies on
non-covalent protein-protein interactions and provides oriented
coupling, because binding occurs at a well-defined site on the
target. Another advantage of this approach is that it is fully regen-
erable and allows immobilization from crude protein mixtures,
such as cell lysates. A disadvantage of the capturing method is
that it may not produce a stable surface, thus complicating the
analysis of analyte binding.
Three major coupling classes can be used for the affinity cap-
turing method: artificially introduced affinity tags (e.g., biotin or
hexahistidine), antibody–antigen systems (e.g., antibodies against
Studying Protein-Protein Interactions 115
Table 1
Available sensor chips, their characteristics and applications
Sensor
chip Surface Characteristics and applications
CM5 Standard surface—Carboxymethylated dextran Excellent chemical stability; Versatile chip
matrix suitable for most applications
CM4 Low carboxylation—Carboxymethylated Reduces nonspecific binding of highly
dextran matrix with lower degree of positively charged molecules that may be
carboxylation than CM5, i.e., less negatively found, for example, in crude samples
charged Convenient for measurement of low Rmax
needed in kinetic applications
CM3 Short dextran—Carboxymethylated dextran For low immobilization levels and work with
matrix, with the same level of carboxylation cells, viruses, and multicomponent
as CM5, but shorter matrix complexes with high molecular weight
analytes
CM7 High Carboxylation—Comparable with the Gives higher immobilization capacity; Ideal
CM5 chip but has a higher density of for applications that use small molecules
carboxymethylated dextran and fragments
C1 Flat carboxymethylated surface For work with particles such as cells and
viruses, and in applications where dextran
matrix is undesirable
SA Carboxymethylated dextran matrix pre- Captures biotinylated ligands such as
immobilized with streptavidin carbohydrates, peptides, proteins, and
DNA fragments
NTA Carboxymethylated dextran matrix pre- Designed to bind histidine-tagged ligands via
immobilized with NTA (nitrilotriacetic acid) metal chelation; Allows control of steric
orientation of ligand component for
optimal site exposure
HPA Flat hydrophobic surface For studying lipid monolayers interacting
with membrane binding biomolecules and
membrane-associated interactions
L1 Carboxymethylated dextran matrix modified For rapid and reproducible capture of
with lipophilic substances liposomes with retention of lipid bilayer
structure
a
Analyte
Tagged protein
Anti-tag
antibody
CM5 Sensor chip
b
1250
1000
750
RU
pCDNA3-FLAG-DOT1L (2.4µg/µl)
500
pCDNA3-FLAG-DOT1L (1.2µg/µl)
250 pCDNA3-FLAG-DOT1L (0.6µg/µl)
0 pCDNA3 (2.4µg/µl)
0 100 200 300 400
time (s)
Fig. 2 Affinity based immobilization method. (a) Illustration of the capture affinity immobilization method using
an antibody–antigen interaction. (b) Dose-dependent and specific immobilization of Flag-tagged full length
histone methyltransferase, DOT1L, obtained from crude cell lysate of 293 cells transfected with pCDNA3-
FLAG-DOT1L, using anti-flag antibody as a capture molecule20 (pCDNA3—empty vector)
1.2.4 Coupling Chemistry Different covalent coupling chemistries have been developed for
immobilization of ligands: coupling through primary amines, thiol
groups, and aldehyde coupling (Fig. 3). Choosing an immobiliza-
tion method depends mostly on the nature of the ligand.
Amine coupling is the most generally applicable coupling
chemistry because of its universality, stability, speed and most macro-
molecules contain amine groups. This coupling uses primary amine
groups (the N-terminus and lysine residues), which are often
solvent-exposed due to their hydrophilicity. They react directly
with active esters generated by N-hydroxysuccinimide–1-ethyl-3-
(3-dimethylaminopropyl)carbodiimide (NHS/EDC) activation
(Fig. 3). The second type of covalent coupling chemistry, thiol
coupling, provides an alternative approach when amine coupling
cannot be performed or the immobilization level will not be suffi-
cient, such as immobilization of acidic proteins. Thiol coupling uti-
lizes exchange reactions between thiols and active disulphide groups
which can be introduced either on the dextran matrix to exchange
with a thiol group on the ligand (ligand thiol approach) or on the
ligand molecule to exchange with a thiol group introduced on the
dextran matrix (surface thiol approach). A recommended reagent for
introducing active disulphide groups is 2-(2-pyridinyldithio)ethanea-
mine (PDEA). Thiol coupling chemistry is not suitable for experi-
ments where the surface is exposed to reducing agents or high pH,
since the coupling bond is unstable under such conditions.
Ligands containing aldehyde groups (either native or introduced by
oxidation of cis-diols) can be immobilized after activating the surface
118 Zaneta Nikolovska-Coleska
NHS/EDC
a)
b)
Sensor chip CM5 Amine functionalized
ligands
PDEA
c) d)
Cystamine/DTE
c)
Aldehyde
functionalized ligands
Ligand thiol
coupling Surface thiol
coupling
Fig. 3 Steps in coupling chemistry for covalently attaching biomolecules to the sensor surface: (a ) Activation
of the matrix-based carboxyl groups by NHS (N-hydroxysuccinimide) and EDC (1-ethyl-3-(3-dimethylamino-
propyl)-carbodiimide); (b) Direct immobilization of amine functionalized ligands; (c ) Thiol coupling utilizing
disulphide exchange (ligand and surface thiol coupling); (d ) Immobilization of aldehyde functionalized ligands
using reductive ammination. (PDEA Thiol coupling reagent: 2-(2-pyridinyldithio)ethaneamine hydrochloride;
H2N-NH2 Hydrazine; NaCNBH4 Cyanoborohydride)
1.2.5 Determining of The immobilization process is based upon the principle of electro-
Optimal Immobilization pH static interaction of the ligand and the activated surface molecules,
(pH Scouting) which results in a covalent binding. In order to reach the highest
efficiency of the amine coupling reaction, proteins have to be pre-
concentrated on the sensor chip surface. A ligand can be concen-
trated at the sensor surface by electrostatic attraction. The pKa of
the dextran matrix is 3.5 and at pH > 3.5 the dextran matrix will
have a net negative charge. Efficient pre-concentration requires that
the pH be between the pKa of the sensor surface and the isoelectric
point (pI) of the ligand (the pH at which there is no net charge on
the protein). Such surface attraction will be reached by using
immobilization buffer with pH > 3.5, but lower than the pI of
the ligand. As a general rule, the pH of the buffer should be at least
Studying Protein-Protein Interactions 119
20000
pH 4.0
0 100 200
Time (sec)
0.5 units below the pI of the ligand. In addition, the lower the ionic
strength of the coupling buffer, the more ligand will be pre-
concentrated and immobilized. This will allow strong electrostatic
attractions between the negatively charged carboxyl group of the
matrix and the positively charged amine groups of the ligand.
Therefore, acidic proteins with pI values <3.5 cannot be immobi-
lized by amine coupling. Buffers such as Tris buffer and reagents
bearing primary amines should not be used due to possible compe-
tition with the amino groups of the protein. To determine the
appropriate pH for immobilization, an experimental procedure
known as pre-concentration pH-scouting, should be performed.
The pre-concentration is driven by electrostatic interactions, which
are most pronounced in low ionic strength buffers. For this pur-
pose 10 mM sodium acetate buffer with different pH values, in a
range from 4.0 to 5.5, is tested in order to determine the most
optimal pH (Fig. 4). The magnitude and slope of the response
generated due to the increased mass on the surface provide infor-
mation about the ligand density. By comparing the pre-
concentration responses of each immobilization buffer, the mildest
pH solution that can achieve the targeted level for immobilization
should be selected in order to minimize possible denaturation. In
general, for many proteins, 10 mM sodium acetate buffer (pH 4.5)
is optimal for their preconcentration.
1.2.6 Reference Surface During an SPR experiment the choice of an appropriate reference
surface is crucial. Usually the surface on the chip is divided into
several flow cells, depending on the instrument, which can be used
individually or in a number of combinations, and at least one flow
120 Zaneta Nikolovska-Coleska
Binding + Bulk
Reference
subtraction True binding
Fig. 5 Bulk effects and reference surface subtraction. Bulk effects are due to differences in the refractive index
of the running buffer and sample solution. The bulk contribution should be subtracted using a reference
surface
2 Materials
3 Methods
3.1 Amine Coupling The coupling procedure consists, in general, of three steps: (1)
activation resulting in an active group that can be further modified
or used to couple the ligand; (2) coupling, when the ligand is
injected over the activated surface until sufficient ligand is bound;
and (3) deactivation to quench remaining activated sites and obtain
the final level of immobilized ligand (Fig. 6). The covalently immo-
bilized ligand either participates directly in the interaction under
study or is used for affinity capture of one of the interacting mole-
cules. The methods presented below are the same if an interacting
ligand is immobilized or a corresponding antibody will be used for
affinity capturing of the ligand.
Ethanolamine
40000
Deactivation
Ligand
Resonance Unit (RU)
30000 Immobilization
NHS/EDC
Activation
20000 x
RU of
immobilization
x
10000
0
0 500 1000 1500 2000
Time(sec)
3.1.2 Preconditioning of 1. Allow the sealed sensor chip pouch to equilibrate at room
the Sensor Chip CM5 temperature for 30 min in order to prevent condensation on
the chip surface. Insert Sensor Chip CM5 in the instrument
and dock the chip.
2. Run three times Prime with running buffer (HBS-P) to fill the
syringes and compartments.
3. Set the flow rate to 100 μL/min and sequentially using
“Quickinject” command inject two times 10 mM HCl,
50 mM NaOH and 0.1 % SDS to precondition the chip surface.
This procedure allows washing and hydrating the surface
simultaneously, before starting the immobilization procedure.
4. “Rinse,” “Flush,” and “Prime” to wash the microfluidic system
to be ready for the next step of experiments.
3.1.3 Ligand 1. Start a sensorgram, choose the flow cell where the immobiliza-
Pre-concentration tion will take place and decide on a control, reference surface
for blank subtraction. Usually, flow cell 1 is used as the refer-
ence surface.
2. Set flow rate to 10 μL/mL.
3. Dilute and prepare the ligand solution (protein sample for
immobilization) (see Note 2) to a constant concentration in a
range from 5 to 20 μg/mL using a variety of 10 mM NaOAc
immobilization buffers with different pH values (e.g., 4.0, 4.5,
5.0, and 5.5 where the pH is adjusted using 10 % (v/v) acetic
acid) (see Note 3). Place the solutions into propylene tubes and
cap the tubes with rubber caps.
4. Inject 10 μL of the prepared protein in NaOAc for 60 s contact
time.
5. Inject 50 mM NaOH for 15 s after each pre-concentration as a
wash solution to remove the ligand from the surface, if the
response does not return to baseline.
6. Analyze the level of RU and the slope of the pre-concentration
response to determine the ligand density that can be achieved
(Fig. 4). Compare all responses to identify the mildest pH
Studying Protein-Protein Interactions 125
3.1.4 Ligand 1. Start new sensorgram with a 10 μL/min flow rate and select the
Immobilization surface upon which the ligand protein will be immobilized
(Fc2, Fc3, and/or Fc4).
2. Prepare fresh solution for activation of the CM5 surface by
mixing equal volumes of 100 mM NHS and 400 mM EDC
and inject 70 μL (7 min contact time) (see Note 5).
3. Prepare the ligand in the NaOAc immobilization buffer (with
pH determined in the pre-concentration procedure) at a con-
centration between 10 and 50 μg/mL and inject it over the
activated surface until the target levels for ligand immobiliza-
tion are obtained (see Note 6).
4. Inject 70 μL of 1 M ethanolamine hydrochloride, pH 8.5, to
deactivate excess reactive groups (see Note 7).
5. Repeat steps 1–4 until each flow cell has been treated and the
ligand has been immobilized.
6. Prepare the reference surface on flow cell 1 (Fc1) to be used for
reference-subtracted data necessary for kinetic and affinity mea-
surements. Repeat steps 1–4, replacing the ligand for immobi-
lization with an inactive protein or omit immobilization of the
control protein so that the control surface is treated only with
the activation and deactivation reagents.
7. Wash the surface(s) with a washing solution (i.e., a regenera-
tion solution that is tolerated by the ligand, such as two times
15 s injections of 20 mM NaOH). This allows determination of
stably immobilized ligand and the net ligand immobilization
level.
8. The sensor chip is now ready to be used or stored. To ensure a
stable baseline for the interaction analysis, running a sensor
chip overnight in running buffer is recommended (see Note 8).
3.2 Sample Preparation of the samples for testing and injection over a prepared
Preparation immobilized surface is a critical factor for obtaining high-quality
results and accurate kinetic measurements. Similar to the ligand,
the analyte should also be pure, homogenous and unaggregated
since differently sized binding species will give different responses.
The buffer used in experiments can make a significant difference in
the binding of the analyte to the ligand and additions such as
detergents, chelating agents or denaturing chemicals can influence
the binding characteristics and the stability of the complex. The
standard running buffer, HBS-P, contains: 10 mM HEPES, pH
7.4, 150 mM NaCl, and 0.005 % P20 (polyoxyethylene sorbitan),
with or without EDTA (3 mM). The NaCl is necessary to reduce
nonspecific binding, P20 is a non-ionic detergent used to avoid
126 Zaneta Nikolovska-Coleska
then the regeneration step will not be necessary. In this case, one
should just wash the surface until all of the analyte dissociates from
it. Recently, a detailed method for a systematic, seven-step experi-
mental approach to efficiently determine the optimal regeneration
conditions for SPR surfaces with covalently coupled proteins was
reported [25]. Successful selection of a regeneration solution is a
very important step ensuring that the surface chip will have intact
immobilized ligand and no build-up of bound analyte protein. This
will allow multiple uses of the chip and accurate KD determinations.
3.3.1 Identification of 1. Run sensorgrams using the prepared analyte solutions and
Suitable Regeneration various regeneration solutions, under constant contact time
Solution and flow rate (see Note 9).
2. Insert report points and analyze the results through obtained
responses to determine that regeneration is within 10 % of the
first injection (see Note 10).
100
RU
50
3.4.1 Kinetic Analysis 1. Run Desorb (step 1, Subheading 3.1), then dock the CM5
of PPIs chip with ligand and prime with running buffer.
2. Prepare a twofold serial dilution of the analyte into running
buffer based on the optimum concentration range. The recom-
mended range is 10–0.1 times the expected KD value. How-
ever, it is important to avoid analyte concentrations that can
introduce artifacts such as nonspecific binding, steric hindrance
or aggregation. Prepare at least one set of duplicate analyte
132 Zaneta Nikolovska-Coleska
spread and Chi2 values should fall within the defined accep-
tance criteria.
4 Summary
5 Notes
flow cell that will be used for the immobilization, not on the
reference surface.
4. As a general rule, pH values down to about 3.5 can be used and
for pH 3.5–4.0 apply citrate buffer. If pre-concentration is
inadequate even at pH 3.5, the ligand may be too acidic and a
different immobilization approach should be considered. Some
ligands can be immobilized at pH values above 5.5; maleate
buffers are suitable for immobilization at pH values in the range
5–6. Furthermore, low ionic strength buffer should be used,
and the ligand should be sufficiently diluted or desalted from
salt-containing stock solutions. The total ion concentration
should be 10 mM or less.
5. After 7 min of activation of the sensor chip surface with EDC/
NHS, approximately 40 % of the carboxyl groups will be acti-
vated leaving a net negative charge during the immobilization
procedure, which enables pre-concentration. Ligand contact
should be completed within 15 min after surface activation to
ensure coupling before the reactive esters on the surface can be
hydrolyzed. If high-density surfaces are required, the injection
time of EDC/NHS can be increased.
6. If the immobilization level is low, increase the contact time if
the immobilization sensorgram indicates that more ligand can
bind. Also, increase the ligand concentration. Make sure that
the EDC and NHS are fresh solutions. Immobilization and
running buffers should not contain primary amines, for exam-
ple Tris or sodium azide, since they can compete with the ligand
for reactive groups on the surface.
7. Immobilization can be performed on each flow cell of the
sensor chip simultaneously or only on the flow cell intended
for use. Experience shows that the activation level of each flow
cell might not be the same if the activation solution is run over
all flow cells at the same time, however, deactivation of all flow
cells together is satisfactory.
8. Once the immobilization is finished, the sensor chip with
bound ligand can be undocked and kept in a 50 mL conical
tube at 4 C. Before using the sensor chip again and docking it
in the instrument, allow the sensor chip to equilibrate to room
temperature.
9. Initial regeneration steps should be performed with mild regen-
eration solutions for a minimal contact time to ensure that the
immobilized ligand will not be affected or otherwise damaged.
If necessary, the concentration of the regeneration solution can
be increased during the procedure. NaOH regeneration solu-
tion should be kept in glass vials and should be freshly prepared
because its efficiency as a regeneration solution decreases over
the time.
Studying Protein-Protein Interactions 135
References
1. Cusick ME, Klitgord N, Vidal M et al (2005) 7. Ivanov AA, Khuri FR, Fu H (2013) Targeting
Interactome: gateway into systems biology. protein-protein interactions as an anticancer
Hum Mol Genet 14(Spec No. 2):R171–R181 strategy. Trends Pharmacol Sci 34:393–400
2. Ge H, Walhout AJ, Vidal M (2003) Integrating 8. Fagerstam LG, Frostell-Karlsson A, Karlsson R
‘omic’ information: a bridge between geno- et al (1992) Biospecific interaction analysis
mics and systems biology. Trends Genet using surface plasmon resonance detection
19:551–560 applied to kinetic, binding site and concentra-
3. Wells JA, McClendon CL (2007) Reaching for tion analysis. J Chromatogr 597:397–410
high-hanging fruit in drug discovery at protein- 9. Myszka DG (2000) Kinetic, equilibrium, and
protein interfaces. Nature 450:1001–1009 thermodynamic analysis of macromolecular
4. Blazer LL, Neubig RR (2009) Small molecule interactions with BIACORE. Methods Enzy-
protein-protein interaction inhibitors as mol 323:325–340
CNS therapeutic agents: current progress and 10. Rich RL, Myszka DG (2008) Survey of the year
future hurdles. Neuropsychopharmacology 2007 commercial optical biosensor literature. J
34:126–141 Mol Recognit 21:355–400
5. Ryan DP, Matthews JM (2005) Protein- 11. Hunter MC, O’Hagan KL, Kenyon A et al
protein interactions in human disease. Curr (2014) Hsp90 binds directly to fibronectin
Opin Struct Biol 15:441–446 (FN) and inhibition reduces the extracellular
6. Gerrard JA, Hutton CA, Perugini MA (2007) fibronectin matrix in breast cancer cells. PLoS
Inhibiting protein-protein interactions as an One 9:e86842
emerging paradigm for drug discovery. Mini 12. Chow CR, Suzuki N, Kawamura T et al (2013)
Rev Med Chem 7:151–157 Modification of p115RhoGEF Ser(330)
138 Zaneta Nikolovska-Coleska
regulates its RhoGEF activity. Cell Signal biosensor assays. A multivariate cocktail
25:2085–2092 approach. Anal Chem 71:2475–2481
13. Pal A, Huang W, Li X et al (2012) CCN6 25. Drake AW, Klakamp SL (2011) A strategic and
modulates BMP signaling via the Smad- systematic approach for the determination of
independent TAK1/p38 pathway, acting to biosensor regeneration conditions. J Immunol
suppress metastasis of breast cancer. Cancer Methods 371:165–169
Res 72:4818–4828 26. Karlsson R, Larsson A (2004) Affinity measure-
14. Day YS, Baird CL, Rich RL et al (2002) Direct ment using surface plasmon resonance. Meth-
comparison of binding equilibrium, thermody- ods Mol Biol 248:389–415
namic, and rate constants determined by sur- 27. Karlsson R, Katsamba PS, Nordin H et al (2006)
face- and solution-based biophysical methods. Analyzing a kinetic titration series using affinity
Protein Sci 11:1017–1025 biosensors. Anal Biochem 349:136–147
15. Mehand MS, Srinivasan B, De Crescenzo G 28. Rich RL, Papalia GA, Flynn PJ et al (2009) A
(2011) Estimation of analyte concentration by global benchmark study using affinity-based
surface plasmon resonance-based biosensing biosensors. Anal Biochem 386:194–216
using parameter identification techniques. 29. Cornish-Bowden A (2001) Detection of errors
Anal Biochem 419:140–144 of interpretation in experiments in enzyme
16. Towne V, Zhao Q, Brown M et al (2013) kinetics. Methods 24:181–190
Pairwise antibody footprinting using surface 30. Cannon MJ, Papalia GA, Navratilova I et al
plasmon resonance technology to characterize (2004) Comparative analyses of a small mole-
human papillomavirus type 16 virus-like parti- cule/enzyme interaction by multiple users of
cles with direct anti-HPV antibody immobili- Biacore technology. Anal Biochem 330:98–113
zation. J Immunol Methods 388:1–7
31. Rich RL, Myszka DG (2000) Survey of the
17. Nedelkov D, Nelson RW (2003) Surface plas- 1999 surface plasmon resonance biosensor lit-
mon resonance mass spectrometry: recent erature. J Mol Recognit 13:388–407
progress and outlooks. Trends Biotechnol
21:301–305 32. Rich RL, Myszka DG (2001) Survey of the year
2000 commercial optical biosensor literature.
18. Williams C, Addona TA (2000) The integra- J Mol Recognit 14:273–294
tion of SPR biosensors with mass spectrometry:
possible applications for proteome analysis. 33. Rich RL, Myszka DG (2002) Survey of the year
Trends Biotechnol 18:45–48 2001 commercial optical biosensor literature.
J Mol Recognit 15:352–376
19. Davis TM, Wilson WD (2000) Determination
of the refractive index increments of small 34. Rich RL, Myszka DG (2003) A survey of the
molecules for correction of surface plasmon year 2002 commercial optical biosensor litera-
resonance data. Anal Biochem 284:348–353 ture. J Mol Recognit 16:351–382
20. Shen C, Jo SY, Liao C et al (2013) Targeting 35. Rich RL, Myszka DG (2005) Survey of the year
recruitment of disruptor of telomeric silencing 2004 commercial optical biosensor literature.
1-like (DOT1L): characterizing the interac- J Mol Recognit 18:431–478
tions between DOT1L and mixed lineage leu- 36. Rich RL, Myszka DG (2005) Survey of the year
kemia (MLL) fusion proteins. J Biol Chem 2003 commercial optical biosensor literature.
288:30585–30596 J Mol Recognit 18:1–39
21. Drake AW, Tang ML, Papalia GA et al (2012) 37. Rich RL, Myszka DG (2006) Survey of the year
Biacore surface matrix effects on the binding 2005 commercial optical biosensor literature.
kinetics and affinity of an antigen/antibody J Mol Recognit 19:478–534
complex. Anal Biochem 429:58–69 38. Rich RL, Myszka DG (2007) Survey of the year
22. Pace CN, Vajdos F, Fee L et al (1995) How 2006 commercial optical biosensor literature.
to measure and predict the molar absorption J Mol Recognit 20:300–366
coefficient of a protein. Protein Sci 39. Rich RL, Myszka DG (2010) Grading the
4:2411–2423 commercial optical biosensor literature-Class
23. Andersson K, Areskoug D, Hardenborg E of 2008: ‘The Mighty Binders’. J Mol Recognit
(1999) Exploring buffer space for molecular 23:1–64
interactions. J Mol Recognit 12:310–315 40. Katsamba PS, Navratilova I, Calderon-Cacia M
24. Andersson K, Hamalainen M, Malmqvist M et al (2006) Kinetic analysis of a high-affinity
(1999) Identification and optimization of antibody/antigen interaction performed by mul-
regeneration conditions for affinity-based tiple Biacore users. Anal Biochem 352:208–221
Chapter 8
Abstract
Label-free detection technologies have been widely used to characterize biomolecular interactions without
having to label the target molecules. These technologies exhibit considerable potential in facilitating assay
development and enabling new integrated readouts. When combined with high-throughput capability,
label-free detection may be applied to small molecule screens for drug candidates. Based on the resonant
waveguide grating biosensors, a label-free high-throughput detection system, the Epic® System, has been
applied to monitor molecular interactions. Here we describe a generic label-free assay to quantitatively
measure phospho-specific interactions between a trafficking signal—phosphorylated SWTY peptide and
14-3-3 proteins or anti-phosphopeptide antibodies. Compared with the solution-based fluorescence
anisotropy assay, our results support that the high-throughput resonant waveguide grating biosensor
system has shown the capability not only for high-throughput characterization of binding rank and affinity
but also for the exploration of potential interacting kinases for the substrates. Hence, it provides a new
generic HTS platform for phospho-detection.
Key words Resonant waveguide grating, Label-free, Epic, High throughput, Phospho-specific
interactions, SWTY peptide, 14-3-3 proteins, Kinase
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_8, © Springer Science+Business Media New York 2015
139
140 Meng Wu and Min Li
1.1 Assay Principle The Epic® system is based on a RWG biosensor, which exploits the
of Biosensors evanescent wave generated by the resonant coupling of light into a
waveguide via a diffraction grating. The guided light can be viewed
as one or more mode(s) of light that all have directions of propaga-
tion parallel with the waveguide, due to the confinement by total
internal reflection at the substrate–film and medium–film interfaces.
The waveguide has a higher refractive index value than its surround-
ing medium. Because the guided light mode has a transversal ampli-
tude profile that covers all layers, the effective refractive index N of
each mode is a weighted sum of the refractive indices of all layers:
N ¼ f N ðnF ; nS ; nC ; nad ; d F ; d ad ; λ; m; σ Þ ð1Þ
Here, nF, nS, nC, and nad is a refractive index of the waveguide, the
substrate, the cover medium, and the protein adlayer, respectively.
dF and dad are the effective thicknesses of the film and the protein
adlayer, respectively. λ is the vacuum wavelength of the light used.
m ¼ 0, 1, 2, is the mode number; and σ is the mode type number,
which equals 1 for TE (transverse electric or s-polarized) and 0 for
TM modes (transverse magnetic or p-polarized). Because of its
higher sensitivity, generally the TM (σ ¼ 0) is used for measuring
the binding of biomolecules to the probe proteins immobilized on
the surface of the waveguide substrate.
When a laser illuminates the waveguide at varying angles or
wavelengths, light is coupled into the waveguide only at
corresponding specific angles or wavelengths. This coupling is deter-
mined by the effective refractive index of the guided mode, denoted
as N. The value of N can be calculated numerically from the mode
equation for a given mode of a four-layer waveguide configuration:
" #σ !
2 2 0:5
n2A n2C ðN =nC Þ2 þ ðN =nA Þ2 1
0 ffi πm k nF N dF þ dA 2
nF n2C ðN =nC Þ2 þ ðN =nF Þ2 1
2 !0:5 3
2 2 2
n F N n S
þarctan4 5
nS n2F N 2
2 !0:5 3
2 2 2
n F N n C
þarctan4 5 ð2Þ
nC n2F N 2
Here, k ¼ 2π/λ.
Since the laser light is coupled to, and propagates parallel to the
surface in the plane of a waveguide film, this creates an
Label-free Detection of Biomolecular Interactions 141
1.2 Resonant A beta version of the Corning® Epic® System is comprised of three
Waveguide Grating major components for bioassay applications: an Epic® sensor
Biosensor Detection microplate, an RWG detector, and a liquid handling system. The
sensor microplate consists of a glass bottom plate attached to a
holey plastic 384-well plate, which enables high-throughput
screening. Each well in the 384-well Epic® microplate contains an
RWG sensor, which consists of an optical grating and a high index
of refraction waveguide coating [6, 7]. When illuminated with
broadband light at a fixed angle of incidence, these sensors reflect
only a narrow band of wavelengths that is a sensitive function of the
effective index of refraction of the waveguide. The sensors are
coated with a surface chemistry layer that enables covalent attach-
ment (via peptide bond formation) of peptides/proteins or other
biomolecules. Binding of molecular recognition partners to the
immobilized target induces a change in the effective index of
refraction of the waveguide, and this is manifest as a shift in the
wavelength of light that is reflected from the sensor. The magnitude
of this wavelength shift is proportional to the amount of analyte
that binds to the immobilized target. Unlike most commercial SPR
biosensors, which generally use a continuous flow system for the
determination of the kinetics of binding, the Epic® System does not
utilize flow channels. RWG sensors are evanescent in nature which
means that the magnitude of the electric field in the medium
adjacent to the sensor surface decays exponentially from the sensor
surface. The distance from the sensor surface at which the electric
field strength has decreased to 1/e of its initial value is the penetra-
tion depth. For the Epic® System, the penetration depth is
~150 nm. Thus, the system is selective and sensitive to binding
events that take place within this penetration. For most of the
142 Meng Wu and Min Li
Chemistry
Waveguide
Substrate
2 Materials
2.2 Preparation and 1. 14-3-3ζ protein expressed as a GST-tagged fusion and purified
Purification of from E. coli strain BL21-SI as previously described [8].
Recombinant 14-3-3 2. Recombinant GST-tagged 14-3-3 proteins purified using Glu-
Proteins tathione Sepharose 4B beads.
Fluorescence anisotropy measurements were used to determine
the binding affinity of 14-3-3ζ for SWpTY [8].
3 Methods
3.1 14-3-3 Detection As a common protocol for 14-3-3 detection [9] (Fig. 2), the first
Protocol on the Epic® step is to immobilize NH2-SWpTY peptide (50 μg/ml, pH 7.5), as
System a 14-3-3 binding motif, on the Epic® plate. This was followed by
the addition of ethanolamine to quench the remaining active sites
on the surface. Reference wells were made by the addition of PEG-
amine (50 μg/ml, pH 9.2) or the non-phosphorylated peptide
NH2-SWTY (50 μg/ml, pH 7.5). The second step is the addition
of 14-3-3 protein solutions into both the sample wells and the
reference wells. As seen in Fig. 2c, the wavelength shift after the
subtraction of the signals from the reference wells is termed
the referenced signals for the 14-3-3 binding event. The signal
without subtraction of the reference is termed the unreferenced
signal. Usually six sample wells and two reference wells were used
for each binding reaction unless otherwise described.
To evaluate the specificity of the interaction, 14-3-3 was added
into wells modified with either NH2-SWpTY or non-
phosphorylated NH2-SWTY (Fig. 3a). Only wells immobilized
Label-free Detection of Biomolecular Interactions 145
a b
O O O O O
X
X
=
=
|
|
C C C C C GST-14-3-3ζ
| | | | |
NH-SWpTY
Sample
well
NH2-Peptide, pH 7.5
or
PEG-amine, pH 9.0
GST-14-3-3ζ
NH-Peptide
NH-Peptide
NH-Peptide
NH-Peptide
PEG-amine
Reference
O O O O O well
X
=
=
|
C C C C |
C
| | | | |
OH
GST-14-3-3ζ NH-SWpTY PEG-amine
NH 2 pH 9.0
NH-Peptide
NH-Peptide
NH-Peptide
on SWpTY
120
OH
Net Signal (pm)
80
S3
O O O NH O O 40 S1
=
=
|
C C C C C
| | | | | 0
on PEG-amine
-40
S2
0 1000 2000 3000 4000
Time (s)
Fig. 2 Scheme of Epic® detection. (a) Surface reaction scheme for immobilization of N-terminal amino group
modified peptides. X, reactive group; NH2-Peptide, N-terminal amino group modified peptides. (b) Binding
scheme for 14-3-3. Left two panels show the immobilized substrate (NH2-SWpTY) and reference (PEG-amine).
Right two panels show GST-14-3-3ζ binding. In the sample well, where GST-14-3-3ζ is added, GST-14-3-3ζ
binds to the immobilized NH2-SWpTY peptide. In the reference well containing immobilized PEG-amine, there
is no binding of GST-14-3-3ζ. (c) The time-course charts of the respective responses from sample wells and
reference wells, with S1 as an unreferenced signal from GST-14-3-3ζ on NH2-SWpTY (50 μg/ml at pH 5.5.
GST14-3-3 at 1 μM) S2 as an unreferenced signal from GST-14-3-3ζ on PEG-amine (50 μg/ml, pH 9.0 GST14-
3-3 at 1 μM), and S3 as a referenced signal obtained by subtraction of S1 with S2. From: Wu, M Long S, Frutos
AG, Eichelberger M, Li M, Fang Y., Journal of Receptors and Signal Transduction, 2009; 29 (3–4): 202–210,
copyright © 2009, Informa Healthcare. Reproduced with permission from Informa
Fig. 3 Specificity of detection. (a) The referenced signals for the binding of 14-3-3 to immobilized SWpTY, and
SWTY. SWpTY, and SWTY were immobilized at 50 μg/ml, pH 5.5, and GST-14-3-3ζ was added at 0.4 μM. (b)
Competition assay with competitors and control. 1 μM of competitors (SWpTY and R18) and control peptide
(SWTY) were pre-incubated with 0.4 μM GST-14-3-3ζ and applied on the immobilized NH2-SWpTY (50 μg/ml).
(c) Comparison of the referenced signals from the GST-14-3-3ζ and its validated binding mutant GST-14-3-3ζ
(K49E). GST-14-3-3 was used at 0.4 μM
3.2 Validation of 14- To further validate the 14-3-3 binding event on the Epic® plates,
3-3 Detection by In the same Epic® plates above were then subjected to an ELISA assay.
Situ ELISA After washing five times with PBS buffer, 50 μl of the anti-14-3-3
antibody (1:200 dilution) was added and incubated at 4 C for
0.5 h. After washing with PBS buffer with 0.1 % Tween, 50 μl of
Label-free Detection of Biomolecular Interactions 147
Fig. 4 Binding detection of 14-3-3 to immobilized NH2-SWpTY peptide. NH2-SWpTY in different concentra-
tions (pH 5.5) was immobilized on the Epic™ plate. After addition of GST-14-3-3ζ (0.4 μM), the referenced
signals were plotted on the left axis against the concentrations of NH2-SWpTY. The same plate was used for
ELISA detection as described in Subheading 3 and the luminescence signals were plotted on the right axis
3.3 Affinity Ranking The rank order of affinity of the peptide motifs was determined by
by Competitive competition assays. After immobilization of NH2-SWpTY on the
Replacement Epic® plate, a pre-mixed solution of 14-3-3 and varying concentra-
tions of the competitive peptides were added into the Epic® wells.
The referenced signals were applied in Eq. 5 to determine the
relative affinities of the corresponding competitive peptides.
148 Meng Wu and Min Li
( !
K D NH‐SWpTY
y¼B x þ K D NH‐SWpTY þ D 0 þ P 0
K D competitor
vffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi!
ffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
u 2
u KD
NH‐SWpTY
t x þ K D NH‐SWpTY þ D 0 þ P 0 4 D 0 P 0 g
K D competitor
ð5Þ
The fitting was done with Eq. 5 using Origin 7.0 (OriginLab), with
y, the referenced binding signal; B as a constant; D0, the concentra-
tion of NH2-SWpTY that conferred 90 % of saturated GST-14-3-3ζ
response; P0, the concentration of GST-14-3-3; K D NH‐SWpTY , the KD
(¼2.1 μM) for NH2-SWpTY binding to GST-14-3-3ζ as deter-
mined by the concentration response of GST-14-3-3ζ with 50 μg/ml
of NH2-SWpTY immobilized at pH 7.5; K D competitor as KD for competi-
tive peptides, and x as the concentration of competitor peptide.
In contrast to the direct detection using the immobilized bind-
ing partner, the competition assay uses the binding competitors to
measure binding affinity in solution. Affinity detection through
competitive replacement has been a routine method [10, 11].
The distinct signals by competitor peptides can be observed with
considerably different affinities using the Epic® System. The fitting
results for KD values of the competitors SWpTY, SWTY, SWpTD,
and SWpTP are 0.12 0.05 μM, >50 μM, 1.75 0.14 μM, and
6 1.67 μM, respectively. Direct comparison of this method with
the previous detection of SWTY-14-3-3 interaction with fluores-
cence anisotropy suggests a similar rank order of the affinity
(Table 1).
Table 1
Comparison of the 14-3-3–SWpTY interaction detection from fluorescence anisotropy and Epic®
label-free detection
3.4 Characterization To characterize the capability of the Epic® System for high-
for High-Throughput throughput screening of 14-3-3 phospho-specific interactions,
Screening 50 μg/ml of NH2-SWpTY at pH 7.5 was immobilized in one set
of rows and PEG-amine was immobilized in a second set of rows on
the Epic® plate with PEG-amine immobilized wells used as a refer-
ence. GST-14-3-3ζ at 1 μM with varying DMSO amounts was
added to test the compatibility of adding small molecule library in
DMSO solution. GST-14-3-3ζ at 1 μM with or without pre-
incubated 20 μM SWpTY (as a positive inhibition control) was
added into the wells of one Epic® plate with intrawell self-
referencing for the statistic data. The Z factor, Z0 factor, and S/N
ratio [12] were calculated from Eqs. 6 and 7, respectively, where Av
is the average and SD is the standard deviation of the referenced
signals.
Z ¼ 1 3ðSDWith 14‐3‐3 þ SDWithout 14‐3‐3 Þ=ðAvWith 14‐3‐3 AvWithout 14‐3‐3 Þ
ð6Þ
S=N ¼ AvWith 14‐3‐3 AvWith 14‐3‐3 and SWpTY =
qffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
2
ð7Þ
ðSDWith 14‐3‐3 Þ2 þ SDWith 14‐3‐3 and SWpTY
4 Notes
3. Wild type and mutant 14-3-3 proteins: The assay was tested
with wild type 14-3-3 and 14-3-3 K49E, which has a mutation
at the binding site. Consistently, the binding signal was
obtained only in wild type 14-3-3 proteins.
4. Two different SWpTY binding proteins: Different phosphor-
recognizing modules, 14-3-3 protein and an anti-SWpTY anti-
body, have been applied using the same detection format. Both
gave detectable signals, where BSA gave only minimal back-
ground signals.
5. Validation with alternative assays: Direct and indirect schemes
of the Epic assay have been validated by the alternative assays,
by in situ ELISA and fluorescent polarization assay respectively.
The ranking and affinity data have further quantitatively ver-
ified the specificity of the current Epic detection method.
In contrast to peptides, proteins, and nucleic acids, small mole-
cules are quite challenging for label-free detection, because of the
small molecular weight and theoretically much smaller signals. In
addition, the nonspecific interactions by small molecules, especially
those promiscuous hydrophobic ones, require additional control
tests to discriminate artifacts from real signals. To overcome this
issue, a self-reference Epic® plate has been designed, as shown in
Fig. 5. One half of the sensor was coated with active chemical, good
for immobilization; while the other half was not coated, and resis-
tant to the immobilization, and consequently serves as the control.
The Epic® system can detect two areas and normalize the
5 Conclusion
References
1. Halai R, Cooper MA (2012) Using label-free 8. Wu M, Coblitz B, Shikano S, Long S, Spieker
screening technology to improve efficiency in M, Frutos AG, Mukhopadhyay S, Li M (2006)
drug discovery. Expert Opin Drug Discov Phospho-specific recognition by 14-3-3 pro-
7:123–131 teins and antibodies monitored by a high
2. Filiou MD, Martins-de-Souza D, Guest throughput label-free optical biosensor. FEBS
PC, Bahn S, Turck CW (2012) To label Lett 580:5681–5689
or not to label: applications of quantitative 9. Wu M, Long S, Frutos AG, Eichelberger M, Li
proteomics in neuroscience research. Proteo- M, Fang Y (2009) Interrogation of phosphor-
mics 12:736–747 specific interaction on a high-throughput label-
3. Saito A, Kawai K, Takayama H, Sudo T, Osada free optical biosensor system-Epic system.
H (2008) Improvement of photoaffinity SPR J Recept Signal Transduct Res 29:202–210
imaging platform and determination of the 10. Huang X (2003) Fluorescence polarization
binding site of p62/SQSTM1 to p38 MAP competition assay: the range of resolvable inhib-
kinase. Chem Asian J 3:1607–1612 itor potency is limited by the affinity of the
4. Landry JP, Gray J, O’Toole MK, Zhu XD fluorescent ligand. J Biomol Screen 8:34–38
(2006) Incidence-angle dependence of optical 11. Dai JG, Murakami K (2003) Constitutively and
reflectivity difference from an ultrathin film on autonomously active protein kinase C asso-
solid surface. Opt Lett 31:531–533 ciated with 14-3-3 zeta in the rodent brain. J
5. Li G, Lai F, Fang Y (2012) Modulating cell-cell Neurochem 84:23–34
communication with a high-throughput label- 12. Zhang L, Wang H, Masters SC, Wang B,
free cell assay. J Lab Autom 17:6–15 Barbieri JT, Fu H (1999) Residues of 14-3-
6. Fang Y, Ferrie AM, Fontaine NH, Yuen PK 3 zeta required for activation of exoenzyme
(2005) Characteristics of dynamic mass S of Pseudomonas aeruginosa. Biochemistry
redistribution of epidermal growth factor 38:12159–12164
receptor signaling in living cells measured 13. Sun H, Li M (2013) Antibody therapeutics
with label-free optical biosensors. Anal Chem targeting ion channels: are we there yet? Acta
77:5720–5725 Pharmacol Sin 34:199–204
7. Fang Y, Ferrie AM, Fontaine NH, Mauro J, 14. Zhang L, Wang H, Liu D, Liddington R, Fu H
Balakrishnan J (2006) Resonant waveguide (1997) Raf-1 kinase and exoenzyme S interact
grating biosensor for living cell sensing. Bio- with 14-3-3zeta through a common site involv-
phys J 91:1925–1940 ing Lysine 49. J Biol Chem 272:13717–13724
Chapter 9
Abstract
Quartz crystal microbalance with dissipation monitoring (QCM-D) is a useful technique for observing the
adsorption of molecules onto a protein-functionalized surface in real time. This technique is based on
relating changes in the frequency of a piezoelectric sensor chip, onto which molecules are adsorbing, to
changes in mass using the Sauerbrey equation. Here, we outline the cleaning, preparation, and analysis
involved in a typical QCM-D experiment, from which one can obtain mass adsorption and kinetic binding
information.
Key words QCM-D, Surface, Piezoelectric, Sauerbrey, Frequency, Mass, Adsorption, Deposition,
Kinetics
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_9, © Springer Science+Business Media New York 2015
153
154 Gabriella T. Heller et al.
2 Materials
2.4 Sensor Chip 1. Phosphate-buffered saline (PBS): Weigh 8 g NaCl, 0.2 g KCl,
Preparation Materials 1.44 g Na2HPO4, and 0.24 g KH2PO4. Transfer to a 1,000 mL
volumetric flask. Fill the flask with ultrapure water to a few
milliliters below the 1,000 mL mark and transfer to a bottled
1 L container. Adjust the pH to 7.4 with HCl or NaOH. Add
ultrapure water to obtain a final volume of 1 L as needed.
2. 10 mM salicylic acid solution in water.
3. 10 mM mercaptoundecanoic acid (MUA) solution in methanol.
4. 5 mM 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide/
N-Hydroxysuccinimide (EDC/NHS) solution in water.
5. 2 mg/mL metmyoglobin in PBS.
6. 50 mM ethanolamine in ultrapure water.
7. Borate buffer: Weigh 6.18 g boric acid and 1.3 g NaOH.
Transfer to a 500 mL graduated cylinder and fill with water to
a few milliliters below the 500 mL mark. Transfer to a bottled
500 mL container. Adjust the pH to 9.5 with NaOH and HCl,
as needed. Shake to mix thoroughly. Add ultrapure water to
obtain a final volume of 500 mL.
8. 0.2, 0.5, 0.7, 1 M sodium azide.
9. 1 mM Nα-(tert-Butoxycarbonyl)-L-asparagine, Boc-L-asparagine
(Boc-Asn-OH) solution in 200 proof ethanol.
10. 200 mM Nickel (NiSO4) solution in water.
11. 2 mg/mL bovine serum albumin (BSA) in PBS.
3 Methods
4. Rinse sensor chips with water and then rinse with ethanol.
5. Dry sensor chips with nitrogen gas (see Note 4).
6. Expose sensor chips to UV/Ozone treatment for another
10 min.
7. Place sensor chips in decontaminated QCM-D flow cells
immediately.
3.2 Flow Cell 1. Mount a “cleaning sensor chip” (see Note 6) in each flow cell,
Decontamination ensuring that a good seal is formed between the sensor chip and
the O-ring.
3.2.1 Regular Cleaning
(To Be Performed Before 2. Flow approximately 10 mL of alkaline liquid detergent solution
Each Use) [3] (see Note 7) through the measurement chamber.
3. Flow 20 mL of water through the flow cells.
4. Allow air to flow through the flow cells for 5 min.
5. Remove cleaning sensor chips.
6. Use nitrogen gas to blow extra liquid out of the tubing and dry
the flow cells.
3.2.2 Deep Cleaning (To 1. Disassemble tubing and measurement chambers. Remove
Be Performed as Needed) O-rings.
(See Note 8) [3] 2. Place any component that comes directly into contact with
liquid into a beaker filled with disposable, phosphate-free liquid
detergent. Sonicate for 1 h.
3. Rinse components with water and place in a beaker containing
water–ethanol mixture. Sonicate for at least 15 min.
4. Rinse components with water and place them in a beaker con-
taining water. Sonicate for at least 10 min.
158 Gabriella T. Heller et al.
3.3.1 Electrostatic It is often possible to form a densely packed layer of protein on the
Protein Layer Formation surface due to electrostatic interactions between the protein and a
charged surface. This method is very simple as it can be carried out
entirely within the QCM-D flow cell. It should be noted, however,
that as a layer of protein is formed, the tertiary structure of the
protein becomes compromised, making this method of attachment
less desirable than the others discussed in this chapter. Below, we
outline a procedure for creating a layer of charged protein on a SiO2
sensor chip (see Note 10) and measuring the deposition of salicylic
acid onto the surface.
1. Mount clean SiO2 sensors chips in decontaminated flow cells.
2. Flow PBS until the frequency stabilizes (see Note 11).
3. Flow BSA solution for 10 min.
4. Rinse with PBS until stable.
5. Flow over solution of interacting salicylic acid until stable.
6. Rinse with PBS until stable.
3.3.3 Histidine Tag When used appropriately, the His-tag capture method of protein-
Capture immobilization is perhaps the most desirable method because of its
ability to orient proteins on the surface. When a histidine tag is on
the opposite side of a protein than the binding site, this procedure
allows the binding site to be exposed during immobilization, thus
preventing results from becoming confounded by its inaccessibility.
Unfortunately, this method does not apply to all proteins, although
some analogs do exist.
1. Rinse Au crystal with ethanol (see Note 13) until frequency
stabilizes (see Note 11).
2. Flow Boc-Asn-OH solution through the systems for 15 min at
0.1 mL/min (see Note 14).
3. Flow ethanol until the frequency stabilizes.
4. Flow water until the frequency stabilizes.
5. Charge with nickel solution by flowing nickel solution through
the system (see Note 15).
6. Rinse with water until stable.
7. Rinse with PBS until stable.
8. Flow protein with an exposed histidine residue through the
system.
9. Rinse with PBS until stable.
10. Flow ligand through the system.
11. Rinse with PBS until stable.
3.4 Data Analysis Changes in frequency obtained from QCM-D can be converted to
changes in mass using either the Sauerbrey relation or viscoelastic
modeling, depending on the viscoelastic properties of the surface.
The Sauerbrey equation, Eq. 1, describes a linear relationship
between frequency shifts and mass changes in thin films under the
assumption that the film is rigidly attached to the sensor.
CΔf
Δm ¼ ð1Þ
n
Fig. 3 Sample QCM-D protein-protein binding data. After a stable baseline was achieved for the functionalized
sensor chip, interacting protein was introduced to the system at 150 s. Association was terminated at 425 s,
and the sensor chip was rinsed with buffer to measure dissociation rates until equilibrium was achieved
koff
KD ¼ ð4Þ
kon
where KD is in M1.
5. Repeat steps 1–4 for each ligand concentration and average KD
values.
QCM-D Methods 163
4 Notes
10. Local positive regions interact with the negatively charged SiO2
surface, thus allowing for layer formation.
11. We define stable frequency as a change no larger than 2 Hz over
10 min. Sometimes this can take 15 min, but sometimes it can
take as long as 3 h.
12. This website provides a great review of cross-linking:
http://www.piercenet.com/browse.cfm?fldID¼CE4D6C5C-
5946-4814-9904-C46E01232683.
13. Be sure to use tubing that is compatible with ethanol.
14. A decrease in frequency of 12–13 Hz should be observed.
15. A small change in frequency should be observed.
16. Theoretically, as soon as dissipation values are larger than zero,
the Voigt model or another viscoelastic model should be used.
When a surface displays viscoelastic behavior, the linear nature
of the frequency-mass relation fails, and true mass on the surface
will be underestimated using the Sauerbrey equation. Data
generally require viscoelastic modeling if the dissipation values
(in 1E6) are greater than 5 % of the frequency shifts (in Hz).
17. For best results, this segment should consist of at least 50 points.
Acknowledgements
References
1. Johal MS (2011) Quartz crystal microbalance. interactions: a molecular cloning manual. Cold
In: Press C (ed) Understanding nanomaterials, Spring Harbor Laboratory Press, Cold Spring
1st edn. CRC Press, Boca Raton, FL, pp 101–108 Harbor, NY, pp 273–284
2. Hauck S, Drost S, Prohaska E, Wolf H, D€ ubel S 3. www.qsense.com
(2005) Analysis of protein interactions using a 4. Vig JR (1985) UV/Ozone cleaning of surfaces.
quartz crystal microbalance biosensor. In: Gole- J Vac Sci Technol A 3:1027
mis EA, Adams PD (eds) Protein-protein
Chapter 10
Abstract
Biolayer Interferometry (BLI) is a powerful technique that enables direct measurement of biomolecular
interactions in real time without the need for labeled reagents. Here we describe the analysis of a high-
affinity binding interaction between a monoclonal antibody and purified antigen using BLI. A simple Dip-
and-Read™ format in which biosensors are dipped into microplate wells containing purified or complex
samples provides a highly parallel, user-friendly technique to study molecular interactions. A rapid rise in
publications citing the use of BLI technology in a wide range of applications, from biopharmaceutical
discovery to infectious diseases monitoring, suggests broad utility of this technology in the life sciences.
Key words Label-free, Antibody, Antigen, PSA, Biolayer interferometry, Affinity constant, Kinetic
analysis, Ligand, Analyte, Biosensor, Association, Dissociation
1 Introduction
1.1 Label-Free Label-free biosensors are in routine use for the analysis of the
Technology for kinetics of binding interactions between two biomolecules [1]. In
Analysis of Molecular biosensor-based analysis, one of the binding partners is immobi-
Interactions lized on the solid surface of the biosensor (ligand) while the other
molecule is present in solution (analyte). While traditional techni-
ques for measuring binding activity and affinity rely on a similar
format, they typically require enzymatic or fluorescent molecular
labeling. Generating labeled biomolecules not only consumes time
and material, but can lead to altered protein activity or steric
blocking of binding sites. Unlike standard endpoint assays such as
ELISA, label-free biosensor technology enables monitoring of
binding interactions in real time. Real-time kinetic measurements
provide more information on mechanisms of interaction, including
association rates (ka), dissociation rates (kd), and affinity constants
(KD) [2]. Label-free technology has greatly advanced in recent
years, enabling rapid, sensitive and accurate measurement of bind-
ing kinetics, affinity and activity of biomolecular complex formation
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_10, © Springer Science+Business Media New York 2015
165
166 Sriram Kumaraswamy and Renee Tobias
1.2 BioLayer BLI is an optical technique that utilizes disposable fiber-optic bio-
Interferometry sensors for measurement of biomolecular interactions. In BLI,
white light is directed down the length of the biosensor fiber
toward two interfaces separated by a thin layer at the tip: the
biocompatible surface of the tip, and an internal reference layer.
Light is reflected back to the detector from each of the two layers,
and the two reflected beams interfere constructively or destruc-
tively at different wavelengths in the spectrum. When the tip of a
biosensor is dipped into a sample, analyte binds to immobilized
ligand on the biosensor surface. This binding forms a molecular
layer which increases in thickness as more analyte molecules bind to
the surface. As the thickness at the biosensor tip increases, the
surface layer effectively moves away from the internal reference
layer and the detector, creating a shift in the interference pattern
of the reflected light (Fig. 1a). The spectral pattern changes as a
function of the optical thickness of the molecular layer, i.e., the
number of molecules bound to the biosensor surface. This shift is
monitored at the detector, and reported on a sensorgram as a
change in wavelength (nm shift) (Fig. 1b). Multiple layers can be
bound sequentially to the surface, providing accurate, comprehen-
sive kinetic data on the interactions between molecules [4].
1.3 Biosensors BLI biosensors are made of glass, and coated with a proprietary
biocompatible matrix which minimizes nonspecific binding to the
surface. This matrix is pre-coated with one of a wide selection of
capture chemistries for specific binding of analyte molecules in a
sample. Using a dip-and-read format, biosensors are moved
a b
Reflected Beams
Incident
white light
nm shift
Biocompatible
surface
Bound
molecule
Un-bound molecules
have no effect
Time
Fig. 1 BLI diagram. (a) As more molecules bind to the surface of the biosensor, the interference pattern of the
reflected light changes, creating a wavelength shift that is reported in real time on a sensorgram (b)
Kinetic Analysis Using Biolayer Interferometry 167
where A represents the ligand, and B is the analyte [5]. This binding
model assumes a 1:1 interaction, where one ligand molecule inter-
acts with one analyte molecule, and binding is independent and of
equal strength for all binding sites. The rate of complex (AB)
formation during the association step of an assay is a function of
the association constant, ka, which is expressed in M1 s1. When
the biosensor is dipped into buffer in the dissociation step, the
complex dissociates back to A and B. The dissociation constant,
kd, identifies the fraction of complexes decaying per second, and is
expressed in units of s1. The dissociation constant is a measure of
the stability of the interaction; the smaller the kd, the more stable
the complex. The affinity constant, KD, is a measure of how tightly
a ligand binds to its analyte. It is the ratio of the on-rate and
off-rate:
168 Sriram Kumaraswamy and Renee Tobias
a
Baseline Loading Baseline Association Dissociation
b 4.0
3.6
Dissociation
3.2
Association
2.8
2.4
Binding, nm
2.0
Loading
1.6
1.2
0.8
0.4
Baseline
0
0 400 800 1,200 1,600 2,000 2,400
Time (seconds)
Fig. 2 Binding kinetics experiment on Streptavidin biosensors. (a) Biotinylated ligand is immobilized on the
streptavidin-coated surface. After a baseline step, an association step is performed where analyte binding
occurs, followed by a dissociation step in buffer. (b) A typical sensorgram trace showing loading, baseline,
association, and dissociation assay steps for replicate samples
½A ½B kd
KD ¼ ¼ ð2Þ
½AB ka
1.5 Considerations Maintaining the structure and activity of the immobilized ligand is
for Designing a the most important consideration for biosensor selection. Direct
Successful Assay immobilization of a target protein to a biosensor can be accom-
plished by (1) covalent linkage to free lysine residues in the target
1.5.1 Choosing an protein via a carboxyl group on the biosensor or by (2) biotinylated
Immobilization Strategy ligand binding to streptavidin-coated biosensors. These methods
are compatible with most proteins and enable creation of “custom”
biosensor surfaces with virtually any protein. However, these
approaches require purified protein and creation of a covalent
bond, either directly to the surface or to biotin. Site-directed or
capture-based biosensors provide an alternative immobilization
strategy that can be used to maximize activity of the ligand on the
surface without covalent bonding. Capture biosensors are pre-
immobilized with a high affinity capture antibody or protein
which binds to the protein ligand via a known motif or tag,
enabling favorable orientation of the ligand and improved homo-
geneity on the surface. Because of the high specificity of these
interactions, ligand protein can be captured directly from crude
samples such as culture media without need for purification.
1.5.2 Assay Orientation The choice of which molecule in a binding pair to use as ligand or
analyte depends on several factors [6]. Primary factors include (1)
binding valency (use the lower valency molecule as analyte to
reduce avidity effects and simplify curve fitting; Fig. 3), (2) avail-
ability of purified protein to use as ligand, (3) stability of the protein
when immobilized (if unstable, use the other molecule as ligand)
and (4) sensitivity of detection (BLI signal is a function of molecu-
lar size and packing density, so use the larger molecule among the
binding pair as analyte to maximize detection sensitivity). Regard-
less of orientation or assay format, proper assay development is a
necessity for obtaining reliable kinetic data with any system.
Biosensor
Surface
Immobilized
Ligand
Bivalent
Analyte
1.5.3 Assay Optimization The amount of ligand to immobilize needs to be optimized for each
assay and reagent lot. Typically, increasing the loading density will
lead to increased signal in the analyte association step. However, it
is a common observation that loading biosensors with very high
density of ligand may lead to artifacts and secondary binding
effects, such as analyte rebinding, that alter observed binding
rates [7]. Hence, a “scouting” experiment must be performed to
find the balance between maximizing sensitivity for analyte detec-
tion and minimizing secondary binding effects. As a rule, the lowest
concentration of immobilized ligand that yields an acceptable signal
in the analyte association step should be used.
Analyte concentration is another important consideration for
obtaining accurate kinetic and affinity constants. A dilution series of
at least four analyte concentrations should be measured in a kinetic
assay. The analyte dilution series ideally should range from a con-
centration of about tenfold higher to tenfold lower than the
expected affinity constant. For screening purposes or qualitative
analyses, a single concentration is often sufficient. If the approxi-
mate KD of the interaction is not known, performing an analyte
concentration scouting step is recommended. In this case, choose a
few concentrations that span a wide range to obtain an approxima-
tion of the affinity constant [8].
1.6 BLI to Study an Here we describe how to perform kinetic characterization of the
Antibody–Antigen interaction between an antibody and antigen using the BLI-based
Interaction Octet analytical system. The reagents used in this example are
purified human prostate-specific antigen (PSA) and purified anti-
PSA monoclonal IgG. This is a previously characterized binding
pair of low nanomolar affinity [12]. Anti-PSA antibody is first
biotinylated for loading onto streptavidin biosensors. The PSA
antigen in solution then is bound to the antibody in an association
step, followed by a dissociation step in buffer. The kinetics of the
binding interaction are monitored in real time. Data processing and
curve fitting parameters are described.
2 Materials
3 Methods
3.2 Optimization of 1. Pipet 200 μl per well of 1 Kinetics buffer into wells of a 96-well
Antibody Loading black flat-bottom microplate corresponding to the number and
Concentration on position of biosensors to be used. The buffer used for hydration
Biosensors should be the same as that used throughout the assay.
3.2.1 Hydrate Biosensors
2. Insert the hydration plate into the biosensor tray. Align the
biosensor rack over the hydration plate and lower the biosen-
sors into the wells, taking care not to scrape or touch the tips of
the biosensors. Allow biosensors to hydrate for at least 10 min.
3.2.3 Load Sample Plate 1. Ensure that the Octet instrument lamp is warmed up for at least
and Biosensors onto Octet 40 min prior to starting the assay.
Instrument and Run Assay 2. Set the sample plate temperature to 30 C in the Octet software
(see Note 13).
3. Place the sample plate on the sample plate stage inside the Octet
system with well A1 toward the back right corner. Place the
biosensor hydration assembly on the biosensor stage. Ensure
that both the tray and the sample plate are securely in place.
4. Equilibrate the plates in the instrument for 10 min prior to
starting the experiment. The delay timer can be used to auto-
matically start the assay after 10 min (600 s).
5. Set up the assay in the instrument software. For details, refer to
the Octet Data Acquisition User Guide. Table 1 shows an
example of the settings for a kinetic assay, which can be used
for the ligand loading experiment. Steps consist of equilibra-
tion, ligand loading, baseline, association, and dissociation
steps (see Note 14).
Assay Step
1 2 3 4 5 6 7 8 9 10 11 12
A 50 Buffer
B 25 Biotin-anti-PSA (μg/mL)
C 10 PSA antigen
D 5
E 2.5
F
G
H
Table 1
Recommended settings for kinetic assay
Sample plate
Step# Step name Time (s) Flow (RPM) column
1 Equilibration 60 1,000 1
2 Loading 300–600 1,000 2
3 Baseline 180–600 1,000 3
4 Association 300–600 1,000 4
5 Dissociation 300–3,600 1,000 3
174 Sriram Kumaraswamy and Renee Tobias
100 μg/mL
3.0 50 μg/mL
25 μg/mL
2.5
2.0
Binding, nm
1.5
10 μg/mL
1.0
5 μg/mL
0.5
2.5 μg/mL
0 0 μg/mL
0 100 200 300 400 500 600 700 800 900 1000 1100 1200
Time (sec)
6. Run the assay. An example of the sensorgram output for the ligand
loading optimization is shown in Fig. 5. The shape of individual
binding curves can be observed. Note that the initial slope of the
binding curve corresponds to concentration of ligand.
7. In Octet Data Analysis software, process the data so that the
association step is aligned to the baseline. In this view, the
relative signal of the analyte binding at each corresponding
ligand concentration can be clearly observed (Fig. 6). The
optimal concentration of biotin–anti-PSA for a binding kinetics
experiment can be selected based on these data. In this exam-
ple, the optimal loading concentration is estimated to be
25 μg/mL (see Note 15).
8. Check for nonspecific binding of analyte to the biosensor. The
non-ligand loaded sample should show a flat response even in
the presence of analyte.
3.3.2 Prepare Assay 1. Equilibrate reagents and samples to room temperature and mix
Samples thoroughly.
2. Dilute the biotin–anti-PSA in 1 Kinetics Buffer to a working
concentration of 25 μg/mL, based on results from the ligand
loading experiment above.
Kinetic Analysis Using Biolayer Interferometry 175
0.5 25 μg/mL
50 μg/mL
100 μg/mL
0.4
Binding, nm
0.3
10 μg/mL
0.2
5 μg/mL
0 0 μg/mL
0 100 200 300 400 500 600
Time (sec)
3.3.3 Load Sample Plate 1. Ensure that the instrument lamp is warmed up for at least
and Biosensors onto Octet 40 min prior to starting the assay.
Instrument and Set Up 2. Set the sample plate temperature in the Octet software.
Kinetic Assay in Octet
3. Open instrument door and place the sample plate on the
Software
sample plate stage with well A1 toward the back right corner.
Place the biosensor hydration assembly on the biosensor stage.
Ensure that both the tray and the sample plate are securely in
place. Close the instrument door.
4. Equilibrate the plates in the instrument for 10 min prior to
starting the experiment. The delay timer can be used to auto-
matically start the assay after 10 min (600 s).
176 Sriram Kumaraswamy and Renee Tobias
Assay Step
1 2 3 4 5 6 7 8 9 10 11 12
A 300 Buffer
B 100 Biotin-anti-PSA
C 33 PSA antigen (nM)
D 11
E 3.7
F
G
H
3.5
3.0
2.5
Binding, nm
2.0
1.5
1.0
0.5
0
0 200 400 600 800 1000 1200 1400 1600 1800
Time (sec)
3.4 Data Analysis 1. Load data into the Octet Data Analysis software. A sample of
the raw sensorgram output is shown in Fig. 8 (see Note 23).
2. Select parameters for data processing. Recommended para-
meters for a standard protein-protein interaction are:
(a) Y-axis alignment (select last 5–10 s)
Kinetic Analysis Using Biolayer Interferometry 177
0.7
300 nM
0.6
100 nM
0.5
Binding, nm
0.4
0.3
33 nM
0.2
0.1 11 nM
3.7 nM
0
0 100 200 300 400 500 600 700 800 900 1000 1100 1200
Time (sec)
Fig. 9 Processed data for anti-PSA–PSA binding, showing aligned association and dissociation steps
0.7 300 nM
0.6
100 nM
0.5
Binding, nm
0.4
0.3
33 nM
0.2
0.1 11 nM
3.7 nM
0
0 200 400 600 800 1000 1200
Time (sec)
Fig. 10 Analyzed data for anti-PSA–PSA binding using 1:1 binding model. Curve fit overlays are shown as thin
lines over traces
4 Notes
21. For a high affinity binding pair (KD < 1 nM), a dissociation
time of 30 min or more may be required. For accurate calcula-
tion of binding and affinity constants, at least 5 % of the
complex must dissociate. Do not run the assay longer than
3 h, as sample evaporation from the microplate wells may
begin to impact results.
22. When running the assay, be sure the baseline is stable before
proceeding to association step. If needed, use the Extend Step
function in the software to run the baseline for longer periods.
With biotin-streptavidin interactions, there should be minimal
signal drift.
23. Some variation in loading levels between individual streptavidin
biosensors during the loading step is normal. This variability
will not affect calculation of kinetic constants in a properly
designed experiment. As long as several concentrations of ana-
lyte are run and global curve fitting is performed, small differ-
ences in ligand loading level are compensated for.
24. Avoid using inter-step correction for binding pairs with very
fast association and dissociation rates.
25. “Model surfing”, or determining the type of interaction based
on the curve-fitting model that best fits your data, is not
recommended. Unless the interaction is known to be more
complex, use 1:1 binding model. If the interaction being stud-
ied is predicted to follow 1:1 binding, and the data do not fit
well, this indicates that further assay development is required.
26. Rmax may be linked if the same biosensor is used for every
sample concentration in the series. This selection is typically
made in small molecule analyses, where dissociation is rapid and
complete and allows for reuse of the biosensor in a new sample.
27. Note that steady state, or equilibrium analysis can also be
performed, but should be used only if the response at the
association step for each concentration has reached
equilibrium.
28. To determine the quality of the fit and accuracy of the calcu-
lated constants, consider the following general guidelines:
(a) Visually inspect the fit: do the fit lines conform well to the
data traces?
(b) Look at residuals, which are plotted below the data traces.
Residual values should not be greater than 1 % Rmax.
(c) The ka error and kd error values should be no greater than
one order of magnitude below the reported constant.
(d) R2 should be above 0.95 to indicate goodness of fit.
(e) Chi-squared value should be below 3.
182 Sriram Kumaraswamy and Renee Tobias
References
1. Rich RL, Myszka DG (2010) Grading the kinetics assays. (2012). http://www.fortebio.
commercial optical biosensor literature - Class com/literature.html
of 2008: ‘The Mighty Binders’. J Mol Recognit 9. Karlsson R, Falt A (1997) Experimental
23:1–64 design for kinetic analysis of protein-
2. Rich RL, Myszka DG (2007) Higher- protein interactions with surface plasmon
throughput, label-free, real-time molecular resonance biosensors. J Immunol Methods
interaction analysis. Anal Biochem 361:1–6 200:121–133
3. Cooper MA (2006) Optical biosensors: where 10. Pall ForteBio Technical Note 8: regeneration
next and how soon? Drug Discov Today strategies for amine reactive biosensors on the
11:1061–1067 octet system. (2007). http://www.fortebio.
4. Concepcion J et al (2009) Label-free detection of com/literature.html
biomolecular interactions using BioLayer Inter- 11. Pall ForteBio Technical Note 14: regeneration
ferometry for kinetic characterization. Comb strategies for streptavidin biosensors on the
Chem High Throughput Screen 12:791–800 octet platform. (2009). http://www.fortebio.
5. Elwing H (1998) Protein absorption and ellip- com/literature.html
sometry in biomaterial research. Biomaterials 12. Katsamba P et al (2006) Kinetic analysis of a
19:397–406 high-affinity antibody/antigen interaction per-
6. Markey F (2009) Macromolecular interactions. formed by multiple Biacore users. Anal Bio-
In: Cooper MA (ed) Label-free biosensors. chem 352:208–211
Cambridge University Press, Cambridge, 13. More information may be found at www.
pp 143–158 fortebio.com
7. Myszka DG (1999) Improving biosensor anal- 14. Pall ForteBio Technical Note 28: biotinylation
ysis. J Mol Recognit 12:279–284 of protein for immobilization onto streptavidin
8. Pall ForteBio Application Note 8: optimizing biosensors. (2011). http://www.fortebio.
protein-protein and protein-small molecule com/literature.html
Chapter 11
Abstract
The analysis of protein-protein interactions has attracted the attention of many researchers from both a
fundamental point of view and a practical point of view. From a fundamental point of view, the development
of an understanding of the signaling events triggered by the interaction of two or more proteins provides
key information to elucidate the functioning of many cell processes. From a practical point of view,
understanding protein-protein interactions at a quantitative level provides the foundation for the develop-
ment of antagonists or agonists of those interactions. Isothermal Titration Calorimetry (ITC) is the only
technique with the capability of measuring not only binding affinity but the enthalpic and entropic
components that define affinity. Over the years, isothermal titration calorimeters have evolved in sensitivity
and accuracy. Today, TA Instruments and MicroCal market instruments with the performance required to
evaluate protein-protein interactions. In this methods paper, we describe general procedures to analyze
heterodimeric (porcine pancreatic trypsin binding to soybean trypsin inhibitor) and homodimeric (bovine
pancreatic α-chymotrypsin) protein associations by ITC.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_11, © Springer Science+Business Media New York 2015
183
184 Adrian Velazquez-Campoy et al.
1.2 ITC of Protein- Two situations are possible when characterizing intermacromole-
Protein Interactions cular interactions in binding reactions: the binding partners are (a)
different (heterodimeric complex) or (b) identical (homodimeric
complex). Even though experimentally they require different meth-
odologies, the underlying principles are the same in both cases, that
is, they follow the same chemical scheme based on a reversible
association equilibrium:
M1 þ M2 ↔M1 M2 ð1Þ
½M 1 M2 1
Ka ¼ ¼ ð2Þ
½M1 ½M2 K d
where [M1] and [M2] are the concentrations of the free reactants
and [M1M2] is the concentration of the complex. These constants
are related to the Gibbs energy of association, ΔGa, and
Isothermal Titration Calorimetry 185
ΔG a ¼ RT ln K a ¼ ΔH a T ΔS a
ð3Þ
ΔG d ¼ RT ln K d ¼ ΔH d T ΔS d
1.2.1 Heterodimeric Typical ITC experiments allow for the characterization of the inter-
Interactions action between two different binding partners. A solution of one of
the reactants is placed in a syringe and a solution with the other
interacting macromolecule is located in a calorimetric cell (Fig. 1).
The stepwise addition of the macromolecule from the injection
syringe solution triggers the binding reaction, leading the system
through a sequence of equilibrium states, the composition of each
one being dictated by the association constant. Given the total
concentrations of reactants in the calorimetric cell, [M1]T and
[M2]T, the association constant, Ka, determines the partition
between the different chemical species:
½M1 M2
Ka ¼ ð4Þ
½M1 ½M2
ITC directly measures the heat, qi, associated with each change
of state after each injection, which is proportional to the increment
in the concentration of complex in the calorimetric cell after the
injection i:
q i ¼ V ΔH a ½M1 M2 i ½M1 M2 i1 ð5Þ
a b time (min)
0 30 60 90 120
0.0
dQ/dt (mcal/s)
-1.0
-2.0
-3.0
c
0.0
Q (kcal/mol of injectant)
-2.0
-4.0
DH Ka
-6.0
-8.0 n
-10.0
0.0 1.0 2.0 3.0
[M1]T/[M2]T
Fig. 1 (a) Illustration of the configuration of an ITC reaction cell. The cell volume
is 1.4 mL and initially is filled with the macromolecule solution (gray). The
injection syringe is filled with the ligand solution (black). At specified time
intervals (400 s), a small volume (10 μL) of the ligand solution is injected into
the cell triggering the binding reaction and producing the characteristic peak
sequence in the recorded signal (b). After saturating the macromolecule, the
residual heat effects (the so-called dilution peaks), if any, are due to mechanical
and dilution phenomena. After integration of the area under each peak (and
subtraction of the dilution heat effects and normalization per mol of injected
ligand) the individual heats are plotted against the molar ratio (Panel c) from
which, through nonlinear regression, it is possible to estimate the thermody-
namic parameters n, Ka, and ΔH
½M2
Kd ¼ ð6Þ
½M2
1.3 Information Every equilibrium binding technique requires the reactant concen-
Available by ITC and trations to be in an appropriate range in order to obtain reliable
Experimental Design estimations of the association constant. A practical rule of thumb
for ITC is given by the parameter c ¼ Ka [M2]T ¼ [M2]T/Kd,
1.3.1 Simultaneous which must lie between 0.1 and 1,000, thus, imposing a limit to the
Determination of the lowest and largest association constant measurable at a given mac-
Association Constant, romolecule concentration [44]. This phenomenon is illustrated in
the Enthalpy of Binding Fig. 2. As the c value increases, the transition from low to high total
titrant concentration is more abrupt. In the case of very high
association constants (macromolecule concentration much higher
than the dissociation constant), all the titrant added in any injection
will bind to the macromolecule until saturation occurs and, there-
fore, all the peaks, except the last ones after saturation, exhibit the
same heat effect. For low association constants (macromolecule
188 Adrian Velazquez-Campoy et al.
time (min)
0 30 60 90 120 0 30 60 90 120 0 30 60 90 120
3.0 a b c
dQ/dt (mcal/s)
2.0
1.0
0.0
Q (kcal/mol of injectant)
10.0
Ka = 104 M-1 Ka = 106 M-1 Ka = 108 M-1
8.0
Ka[M]T = 70 Ka[M]T = 7000
Ka[M]T= 0.7
6.0
4.0
2.0
0.0
0.0 0.5 1.0 1.5 2.0 2.5 3.0 0.0 0.5 1.0 1.5 2.0 2.5 3.0 0.0 0.5 1.0 1.5 2.0 2.5 3.0
[M1]T/[M2]T
Fig. 2 Heterodimer formation. Illustration of the effect of the association constant value on the shape of a
titration curve. The plots represent three titrations simulated using the same parameters (concentrations of
reactants and association enthalpy), but different association constants. Low (a), moderate (b), and high
affinity (c) binding processes are shown. In order to obtain accurate estimates of the association constant, an
intermediate case is desirable (1 < c ¼ [M2]T Ka < 1,000)
1.3.2 Simultaneous For self-associating systems that can be measured by ITC dissocia-
Determination of the tion experiments, another dimensionless parameter can be used to
Dissociation Constant and define a practical limit for the accurate measurement of the dissoci-
the Enthalpy of Dissociation ation constant for a dimeric system. Now, c ¼ [M]T/Kd, which
must lie between 10 and 10,000, imposes a limit to the lowest and
largest dissociation constant measurable at a given macromolecule
concentration. This phenomenon is illustrated in Fig. 3.
As the c value increases, the transition from low to high total
protein concentration in the cell is more abrupt. In the case of very
low dissociation constants (macromolecule concentration much
higher than the dissociation constant) most of the macromolecule
in the syringe is forming dimers and, when diluted in the cell, the
strength of the interactions determines that only a small fraction of
the dimers will dissociate. For that reason the observed heats are
very small. For very high dissociation constants (macromolecule
concentration far below the dissociation constant), the weak inter-
action within the dimer makes it possible for a large fraction of the
dimers to dissociate upon dilution; however, because the mono-
mer–monomer interaction is weak, the population of dimers in the
syringe will be small. In order to obtain accurate estimates of the
dissociation constant, an intermediate case is desirable: the curva-
ture of the plot should be enough for a reliable estimation of the
dissociation constant and the size of the peaks should give an
acceptable signal-to-noise ratio.
1.3.3 Determination of Binding enthalpy is usually determined from titrations in the opti-
the Enthalpy of Binding mal range of reactant concentrations. However, a greater accuracy
can be achieved if enthalpy is measured by performing injections of
190 Adrian Velazquez-Campoy et al.
time (min)
0 30 60 90 120 0 30 60 90 120 0 30 60 90 120
3.0 a b c
dQ/dt (mcal/s)
2.0
1.0
0.0
Q (kcal/mol of injectant)
Fig. 3 Homodimer dissociation. Illustration of the effect of the dissociation constant value on the shape of a
dissociation curve. The plots represent three titrations simulated using the same parameters (concentration of
reactant and dissociation enthalpy), but different dissociation constants. Low (a), moderate (b), and high (c)
dissociation constant processes are shown. In order to obtain accurate estimates of the association constant,
an intermediate case is desirable (10 < c ¼ [M]T/Kd < 10,000)
1.3.4 Blank Experiment In order to estimate the heat effect associated with dilution, mix-
ing, and other phenomena different from binding, blank experi-
ments can be performed. In this case, the experiment is similar to
the one described in Subheading 1.3.3: titrant is injected into a
buffer solution without macromolecule. However, this may be not
the ideal way to estimate such heat effects, since the degree of
solvation of the titrant and the chemical composition of the cell
solution would be different compared to the real titration with the
other interacting macromolecule present. For that reason, some
researchers usually consider the average effect of the last injection
peaks as the reference heat effect. If during the experiment it is not
possible to reach complete saturation, precluding such averaging, it
is possible to include in the fitting function a term accounting for
Isothermal Titration Calorimetry 191
1.3.5 Determination The temperature derivative (at constant pressure) of the enthalpy,
of Heat Capacity Change i.e., the heat capacity change in the process (association or dissocia-
of Binding tion), ΔCP, is defined as
∂ΔH
ΔC P ¼ ð8Þ
∂T P
and can be determined by performing the same experiment at
several temperatures. The slope of the enthalpy vs. temperature
plot gives the heat capacity of the reaction.
ΔCP has been shown to originate from surface desolvation upon
binding or solvation upon dissociation and, to a less extent, from the
difference in vibrational modes between the complex and the free
species [53, 54]. It provides information about the nature of the
interactions driving the binding, in addition to the main thermody-
namic functions (Gibbs energy, enthalpy, and entropy of binding).
ΔH ¼ ΔH 0 þ nH ΔH ion ð9Þ
2 Materials
2.2 Isothermal Modern ITC instruments are simple to use, compact, and
Titration Calorimeter computer-controlled. The calorimeter unit consists of two cells,
the reference cell and the sample cell embedded in an adiabatic
chamber. The system holds the reference cell at a constant temper-
ature. Initially, constant power is applied to the sample cell in order
to activate a feedback control mechanism whose purpose is to
maintain the temperature difference between the two cells as close
to zero as possible. As the reaction, initiated with the injection of
titrant, occurs in the sample cell, the system adjusts the power
applied to the sample cell up or down depending on whether an
endothermic or exothermic reaction is taking place. This power is
recorded by the computer and corresponds to the signal observed
in the characteristic form of a peak. The area under each peak
corresponds to the heat released or absorbed during the reaction
after each injection.
2.3 Biological Two different systems, but functionally and structurally closely
Systems related, have been selected to illustrate the two main types of
association reactions that can be studied by ITC: formation of
heterocomplexes (trypsin–trypsin inhibitor) and homocomplexes
(chymotrypsin dimer). These complexes have been characterized
enzymatically, energetically, and structurally.
Isothermal Titration Calorimetry 193
3 Methods
3.2 Experimental This general protocol was designed for most experiments. Depend-
Procedure ing on the calorimeter or the particular biological system, the
protocol may require some adjustments.
What follows is a step-by-step protocol for running an ITC
experiment to study heterodimer formation. When studying
homodimer dissociation the only differences are in step 5 (cell is
filled with buffer solution only), step 6 (syringe is filled with BP-α-
CT solution) and step 12 (data analysis (see Subheading 3.3)).
1. For typical experiments, allow the equipment to equilibrate one
degree below the experimental temperature.
2. Prepare 2.2 mL of STI solution, 0.5 mL of PPT (in a glass tube,
6 50 mm) and 10 mL of buffer solution. All solutions should
be degassed for 10 min with a vacuum pump (see Note 1).
3. Meanwhile, thoroughly clean the calorimetric reaction cell. The
cell can be washed with a 5 % Contrad 70™ solution or 0.5 M
NaOH if necessary, and rinsed thoroughly with water.
4. Fill the reference cell with water. Usually, water should be
replaced every week.
5. Rinse the reaction cell with buffer. Slowly load the STI solution
into the reaction cell, and carefully remove bubbles. The con-
centration of the sample should be determined again after
loading because some dilution can take place due to residual
buffer in the cell.
6. Fill the 250-μL injection syringe with PPT solution (see Note
2). Rinse the syringe tip with buffer or water and dry.
7. Carefully insert the injection syringe into the reaction cell.
Avoid bending the needle or touching any surface with the
needle tip.
8. Equilibrate the calorimeter at the experimental temperature.
9. Set the running parameters for the experiment: number of
injections (28), temperature (e.g., 25 C), reference power
(10 μcal/s), initial delay (180 s), concentration in syringe
(approx 300 μM), concentration in cell (approx 20 μM), stir-
ring speed (490 rpm), file name (*.itc), feedback mode (high),
equilibration options (fast), comments, injection volume (first
injection 3 μL, the rest 10 μL), duration (automatically set
according to the injection volume), spacing between injections
(400 s), filter (2 s) (see Notes 3–7).
10. Start the experiment (see Note 8). There will be thermal
and mechanical equilibration stages. Initiate the injection
sequence after a stable no-drift noise-free baseline (as seen in
the 1 μcal/s scale) (see Note 9).
11. At the end of the experiment the system should be cleaned
thoroughly with water and the syringe rinsed and dried.
Isothermal Titration Calorimetry 195
1.0 1.0
dQ/dt (mcal/s)
dQ/dt (mcal/s)
25°C 30°C
0.5 0.5
0.0 0.0
Q (kcal/mol of injectant)
Q (kcal/mol of injectant)
10.0 10.0
8.0 8.0
6.0 6.0
4.0 4.0
2.0 2.0
0.0 0.0
0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5
[PPT]T/[STI]T [PPT]T/[STI]T
Fig. 4 Titrations of STI with PPT. The experiments were performed in 25 mM potassium acetate, pH 4.25,
10 mM calcium chloride, at 25 C (left) and 30 C (right). The concentrations of reactants are 21 μM STI (in
cell) and 312 μM PPT (in syringe). The inhibitor was placed in the calorimetric cell due to its low solubility. The
solid lines correspond to theoretical curves with n ¼ 1.26, Ka ¼ 1.5 106 M1, and ΔHa ¼ 8.4 kcal/mol
(left) and n ¼ 1.28, Ka ¼ 2.2 106 M1, and ΔHa ¼ 7.6 kcal/mol (right)
3.3 Data Analysis Equilibrium equations are very simple when using free concentra-
tions of reactants (Eqs. 4 and 6). However, they become more
complex when expressed in terms of total concentrations of reac-
tants, the known independent variables in ITC. Data processing
through transformation of the experimental data in order to linear-
ize the equilibrium equations must be avoided, since it often pro-
vokes a systematic propagation of errors and uneven distribution of
statistical weights over the binding curve. Nonlinear fitting proce-
dures should be implemented to directly analyze the raw calorimet-
ric data.
The analysis should be performed with the individual or differ-
ence heat plot, not with the cumulative or total heat plot. The
analysis in terms of the individual heat (heat associated with each
injection) is more convenient since it eliminates error propagation
associated with cumulative data. See Notes 12–16 for additional
considerations.
196 Adrian Velazquez-Campoy et al.
3.3.1 Heterodimeric The heat, qi, released or absorbed during injection i is proportional
Interaction to the change in concentration of binary complex after that injec-
tion and is given by Eq. 5. The 1:1 stoichiometric model permits an
explicit analytical solution for the concentration of complex (1, 13):
rffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
2
1 þ ½M2 T, i K a þ K a ½M1 T, i 1 þ ½M2 T, i K a þ K a ½M1 T, i 4½M2 T, i K 2a ½M1 T, i
½M1 M2 i ¼
2K a
ð10Þ
where [M1]T,i and [M2]T,i are the total concentrations of macro-
molecule M1 and M2 in the cell after the injection i.
Even when assuming a model corresponding to 1:1 stoichiom-
etry, it is useful to introduce a parameter n representing the number
(or fraction) of binding sites, as in the general 1:n model. When
estimating the parameters, n should be equal to unity, within the
experimental error, otherwise the following statements would hold:
n > 1: There is an error in the determination of the reactants
concentration (actual titrating molecule M1 concentration is
lower and/or actual titrated macromolecule M2 concentration
is higher).
There is more than one binding site (specific binding or
not) per macromolecule M2.
n < 1: There is an error in the determination of the reactants
concentration (actual titrating macromolecule M1 concentra-
tion is higher and/or actual titrated macromolecule M2 con-
centration is lower).
There is less than one binding site per macromolecule M2, that
is, the sample is not chemically or conformationally homogeneous,
or there is more than one binding site (specific binding or not) per
macromolecule M1.
The 1:n stoichiometric model (n identical and independent
binding sites) also permits an explicit analytical solution for the
concentration of complex (1):
rffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
2
1 þ n½M2 T, i K a þ K a ½M1 T, i 1 þ n½M2 T, i K a þ K a ½M1 T, i 4n½M2 T, i K 2a ½M1 T, i
½M1 M2 i ¼
2K a
ð11Þ
v, the same volume of liquid is expelled from the cell. Therefore, the
total concentrations of reactants after the injection i are given by:
i
½M2 T, i ¼ ½M2 0 1 Vv
i ð12Þ
½M1 T, i ¼ ½M1 0 1 1 Vv
3.3.2 Homodimeric The heat, qi, released or absorbed during injection i is proportional
Interaction to the change in concentration of monomer after that injection and
is given by Eq. 7. As already stated, the third term in the parenthesis
accounts for a contribution to the increase in monomer concentra-
tion in the cell that does not contribute to the heat measured.
The monomer–dimer equilibrium model permits an explicit
analytical solution for the monomer concentration (12):
0sffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi 1
K d@ 8 ½ M T, i
½M i ¼ 1þ 1A ð14Þ
4 Kd
8.4 -7.2
3x106
DHa (kcal/mol)
DGa (kcal/mol)
8.2 -7.6
Slope = DCp
Ka(M-1)
6
2x10
8.0 -8.0
7.6 -8.8
0
25 26 27 28 29 30 0 20 40
T (°C) T (°C)
Fig. 5 (Left panel) Estimation of the change of heat capacity upon association. Once the enthalpy of
association is determined at different temperatures (see Fig. 4), these values can be plotted vs. temperature.
The slope of this plot corresponds to the heat capacity change. In the case of the PPT/STI interaction, the heat
capacity was determined to be 160 cal/K mol. This negative value reflects the desolvation of molecular
surfaces upon binding. (Right panel) Outline of the dependency of the association constant and the Gibbs
energy of association on the temperature according to Eq. 3 and ΔGa(T) ¼ ΔHa(T0) + ΔCP,a(T T0)-T
[ΔSa(T0) + ΔCP,aln(T/T0)], where T0 is a given reference temperature at which the enthalpy and the affinity
have been simultaneously measured. The curves were calculated using the thermodynamic parameters
obtained at 25 C (dashed) and 30 C (continuous). The estimation of the heat capacity change allows the
determination of the thermodynamic parameters (affinity, Gibbs energy, enthalpy, and entropy) at any
temperature
time (min)
0 30 60 90 120 150 180 210
0.9
dQ/dt (mcal/s)
0.6
0.3
0.0
Q (kcal/mol of injectant)
4.0
3.0
2.0
1.0
0.0
0 20 40 60 80 100 120
[a-Chymotrypsin]T (mM)
4 Notes
Acknowledgments
References
1. Wells JA, McClendon CL (2007) Reaching for 4. Falconer RJ, Collins BM (2011) Survey of the
high-hanging fruit in drug discovery at protein- year 2009: Applications of isothermal titration
protein interfaces. Nature 450:1001–1009 calorimetry. J Mol Recognit 24:1–16
2. Zinzalla G, Thurston DE (2009) Targeting 5. Ghai R, Falconer RJ, Collins BM (2012)
protein-protein interactions for therapeutic Applications of isothermal titration calorimetry
intervention: a challenge for future. Future in pure and applied research - Survey of the
Med Chem 1:65–93 literature from 2010. J Mol Recognit 25:32–52
3. Falconer RJ, Penkova A, Jelesarov I, Collins 6. Banner DW, D’Arcy A, Janes W, Gentz R,
BM (2010) Survey of the year 2008: Applica- Schoenfeld HJ, Broger C, Loetscher H, Les-
tions of isothermal titration calorimetry. J Mol slauer W (1993) Crystal structure of the solu-
Recognit 23:395–413 ble human 55 kd TNF receptor-human TNF
202 Adrian Velazquez-Campoy et al.
beta complex: implications for TNF receptor helicase in complex with its molecular match-
activation. Cell 73:431–445 maker E2. Genes Dev 18:1981–1996
7. Shibata H, Yoshioka Y, Ohkawa A, Minowa K, 17. Myszka DG, Sweet RW, Hensley P, Brigham-
Mukai Y, Abe Y, Taniai M, Nomura T, Kaya- Burke M, Kwong PD, Hendrickson WA, Wyatt
muro H, Nabeshi H, Sugita T, Imai S, Nagano R, Sodroski J, Doyle ML (1997) Energetics of
K, Yoshikawa T, Fujita T, Nakagawa S, Yama- the HIV gp120-CD4 binding reaction. Proc
moto A, Ohta T, Hayakawa T, Mayumi T, Natl Acad Sci U S A 97:9026–9031
Vandenabeele P, Aggarwal BB, Nakamura T, 18. Gift SK, Zentner IJ, Schön A, McFadden K,
Yamagata Y, Tsunoda S, Kamada H, Tsutsumi Umashankara M, Rajagopal S, Contarino M,
Y (2008) Creation and x-ray structure analysis Duffy C, Courter JR, Zhang MY, Gershoni
of the tumor necrosis factor receptor-1-selec- JM, Cocklin S, Dimitrov DS, Smith AB 3rd,
tive mutant of a tumor necrosis factor-alpha Freire E, Chaiken IM (2001) Conformational
antagonist. J Biol Chem 283:998–1007 and structural features of HIV-1 gp120 under-
8. Hage T, Sebald W, Reinemer P (1999) Crystal lying the dual receptor antagonism by cross-
structure of the interleukin-4/receptor alpha reactive neutralizing antibody m18. Biochem-
chain complex reveals a mosaic binding inter- istry 50:2756–2768
face. Cell 97:271–281 19. Kwong PD, Doyle ML, Casper DJ, Cicala C,
9. Rickert M, Wang X, Boulanger MJ, Goriatch- Leavitt SA, Majeed S, Steenbeke TD, Venturi
eva N, Garcia KC (2005) The structure of M, Chaiken I, Fung M, Katinger H, Parren
interleukin-2 complexed with its alpha recep- PW, Robinson J, Van Ryk D, Wang L, Burton
tor. Science 308:1477–1480 DR, Freire E, Wyatt R, Sodroski J, Hendrick-
10. Philo JS, Aoki KH, Arakawa T, Narhi LO, Wen son WA, Arthos J (2002) HIV-1 evades
J (1996) Dimerization of the extracellular antibody-mediated neutralization through
domain of the erythropoietin (EPO) receptor conformational masking of receptor-binding
by EPO: one high-affinity and one low-affinity sites. Nature 420:678–682
interaction. Biochemistry 35:1681–1691 20. Freire E, Mayorga OL, Straume M (1990) Iso-
11. Syed RS, Reid SW, Li C, Cheetham JC, Aoki thermal titration calorimetry. Anal Chem
KH, Liu B, Zhan H, Osslund TD, Chirino AJ, 62:950A–959A
Zhang J, Finer-Moore J, Elliott S, Sitney K, 21. Doyle ML (1997) Characterization of binding
Katz BA, Matthews DJ, Wendoloski JJ, Egrie interactions by isothermal titration. Curr Opin
J, Stroud RM (1998) Efficiency of signalling Biotechnol 8:31–35
through cytokine receptors depends critically 22. Jelessarov I, Bosshard HR (1999) Isothermal
on receptor orientation. Nature 395:511–516 titration calorimetry and differential scanning
12. Kelekar A, Chang BS, Harlan JE, Fesik SW, calorimetry as complementary tools to investi-
Thompson CB (1997) Bad is a BH3 domain- gate the energetics of biomolecular recogni-
containing protein that forms an inactivating tion. J Mol Recognit 12:3–18
dimer with Bcl-XL. Mol Cell Biol 23. Leavitt S, Freire E (2001) Direct measurement
17:7040–7046 of protein binding energetics by isothermal
13. Kussie PH, Gorina S, Marechal V, Elenbaas B, titration calorimetry. Curr Opin Struct Biol
Moreau J, Levine AJ, Pavletich NP (1996) 11:560–566
Structure of the MDM2 oncoprotein bound 24. Parker MH, Lunney EA, Ortwine DF, Pav-
to the p53 tumor suppressor transactivation lovsky AG, Humblet C, Brouillette CG
domain. Science 274:948–953 (1999) Analysis of the binding of hydroxamic
14. Sundstrom M, Lundqvist T, Rodin J, Giebel acid and carboxylic acid inhibitors to the
LB, Milligan D, Norstedt G (1996) Crystal stromelysin-1 (matrix metalloproteinase-3)
structure of an antagonist mutant of human catalytic domain by isothermal titration calo-
growth hormone, G120R, in complex with its rimetry. Biochemistry 38:13592–13601
receptor at 2.9 Å resolution. J Biol Chem 25. Velazquez-Campoy A, Todd MJ, Freire E
271:32197–32203 (2000) HIV-1 protease inhibitors: enthalpic
15. Walsh ST, Jevitts LM, Sylvester JE, Kossiakoff versus entropic optimization of the binding
AA (2003) Site2 binding energetics of the reg- affinity. Biochemistry 39:2201–2207
ulatory step of growth hormone-induced 26. Todd MJ, Luque I, Velazquez-Campoy A,
receptor homodimerization. Protein Sci Freire E (2000) Thermodynamic basis of resis-
12:1960–1970 tance to HIV-1 protease inhibition: calorimet-
16. Abbate EA, Berger JM, Botchan MR (2004) ric analysis of the V82F/I84V active site
The X-ray structure of the papillomavirus resistant mutant. Biochemistry
39:11876–11883
Isothermal Titration Calorimetry 203
titration calorimetry. Curr Protoc Cell Biol 59. Velazquez-Campoy A, Luque I, Todd MJ,
Chapter 17, Unit 17.8 Milutinovich M, Kiso Y, Freire E (2000) Ther-
53. Murphy KP, Freire E (1992) Thermodynamics modynamic dissection of the binding energet-
of structural stability and cooperative folding ics of KNI-272, a potent HIV-1 protease
behavior in proteins. Adv Protein Chem inhibitor. Protein Sci 9:1801–1809
43:313–361 60. Wyman J, Gill SJ (1990) Binding and linkage:
54. Gomez J, Hilser VJ, Freire E (1995) The heat functional chemistry of biological macromole-
capacity of proteins. Proteins 22:404–412 cules. University Science, Mill Valley, CA
55. Hinz HJ, Shiao DDF, Sturtevant JM (1971) 61. Edgcomb SP, Baker BM, Murphy KP (2000)
Calorimetric investigation of inhibitor binding The energetics of phosphate binding to a pro-
to rabbit muscle aldolase. Biochemistry tein complex. Protein Sci 9:927–933
10:1347–1352 62. Patel CN, Noble SM, Weatherly GT, Tripathy
56. Biltonen RL, Langerman N (1979) Microcalo- A, Winzor DJ, Pielak GJ (2002) Effects of
rimetry for biological chemistry: experimental molecular crowding by saccharides on α-chy-
design, data analysis and interpretation. Meth- motrypsin dimerization. Protein Sci
ods Enzymol 61:287–319 11:997–1003
57. Gomez J, Freire E (1995) Thermodynamic 63. Freire E, Kawasaki Y, Velazquez-Campoy A,
mapping of the inhibitor site of the aspartic pro- Schön A (2011) Characterisation of ligand
tease endothiapepsin. J Mol Biol 252:337–350 binding by calorimetry. In: Podjarny A, Dejae-
58. Baker BM, Murphy KP (1997) Dissecting the gere A, Kieffer B (eds) Biophysical approaches
energetics of a protein-protein interaction: the determining ligand binding to biomolecular
binding of ovomucoid third domain to elas- targets. Detection, measurement and model-
tase. J Mol Biol 268:557–569 ing. RSC Publishing, Cambridge
Chapter 12
Abstract
The reversible formation of protein-protein interactions plays a crucial role in many biological processes.
In order to carry out a thorough quantitative characterization of these interactions it is essential to establish
the oligomerization state of the individual components first. The sedimentation equilibrium method is
ideally suited to perform these studies because it allows a reliable, accurate, and absolute value of the
solution molecular weight of a macromolecule to be obtained. This technique is independent of the shape
of the macromolecule under investigation and allows the determination of equilibrium constants for a
monomer–multimer self-associating system.
1 Introduction
{
(Deceased)
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_12, © Springer Science+Business Media New York 2015
205
206 Ian A. Taylor et al.
1.1 Determination Popular methods for the determination of molecular weights are
of Solution Molecular size exclusion chromatography and dynamic laser light scattering.
Weight In these methods, estimates of the molecular weight of unknowns
are obtained by interpolation, using a curve generated by plotting
an experimentally determinable parameter against the molecular
weight of a set of standards. In size exclusion chromatography
Kav, the partition coefficient between a porous matrix and free
solution, is often used. In dynamic laser light scattering, a transla-
tional diffusion coefficient DT, derived from the measured autocor-
relation function, is often the parameter of choice. Along with the
fact that using these methods molecular weights have to be
obtained by interpolation, the main drawback is that no account
is taken of molecular shape. Consequently, there are often large
errors in the values obtained. The effects of molecular shape are not
accounted for because the experimentally measured parameter
reported by both methods is a function of DT rather than molecular
weight. Although DT is correlated with molecular weight, it is
directly related, through Eq. 1, to an important molecular parame-
ter, the frictional coefficient, f [8].
RT
DT ¼ ð1Þ
N 0f
M ω2 ð1 νpÞ 2
Cx
r x r 20
In ¼ ð6aÞ
C0 2RT
or
M ω2 ð1vρÞ
C x ¼ C 0e 2RT ðr 2x r 20 Þ ð6bÞ
1.3 Analysis There are currently two models of analytical ultracentrifuges pro-
of Molecular duced by Beckman-Coulter: the XL-A and the XL-I. Both contain
Weight Data an absorbance optical system able to measure the absorbance, at a
chosen wavelength, at many points in the cell between r0 and rx [9].
Providing data is collected within the usable linear range of the
optical system, absorbance can replace concentration in Eq. 6b
simply using the Beer–Lambert law, Eq. 7.
A xλ ¼ ελ c x l ð7Þ
2.3 Description Although there are two optical systems available for Beckman-
of the Instrument Coulter analytical ultracentrifuges, in this chapter we will concen-
trate mainly on the use of absorbance optics. There are some
advantages to the use of Rayleigh interference optics, but these
largely surround the speed, the quality, and the density of data
that can be collected and are more relevant to sedimentation veloc-
ity rather than equilibrium experiments. A good comparison of
the advantages and disadvantages of the two optical systems can
found in [9].
The basic feature of the absorbance optics is to allow absor-
bance, as a function of radial distance, to be measured while the
solution is being centrifuged. This is achieved by the use of cells in
which the solution is contained within quartz windows. A mono-
chromator placed above the cell allows it to be illuminated with
monochromatic light between 190 nm and 800 nm, while a slit
mechanism below the cell scans radially at 0.001 cm or greater step
sizes (Fig. 1).
Two types of centrifuge rotors that hold the cells are available,
the An-50 Ti rotor (maximum speed 50,000 rpm) and the An-60
Ti rotor (maximum speed 60,000 rpm). The An-50 rotor has eight
rotor holes, seven for sample cells and one for a counterbalance,
whereas the An-60 has four rotor holes, three for sample cells and
one for a counterbalance. The counterbalance is required in all
centrifuge runs in order to coordinate the flash of the lamp with
the cell position when it is above the detector.
210 Ian A. Taylor et al.
Fig. 1 A Schematic diagram of the optical system of the Beckman Optima XL-A analytical ultracentrifuge.
Figure courtesy of Beckman-Coulter
2.4 Software The Beckman Optima XL-A/XL-I is supplied with analysis soft-
and Data Analysis ware based on the Origin® software package (MicroCal). This
software has been used to fit all the data described in this chapter.
The calculations of buffer density (ρ) and the partial specific
volumes (ν) have been carried out using SEDNTERP [10] (see
Note 1). This and several other useful programs for the analysis
of equilibrium data have been developed by experts in the field and
are available by download over the Internet, in most cases as
Freeware. The Reversible Associations in Structural and Molecular
Biology group (RASMB) maintains a website that provides links to
most of these programs, some of which are available for different
platforms (http://www.bbri.org/RASMB/rasmb.html). Simula-
tions to determine optimal run conditions can be carried out
using the Beckman XL-A software and the freely available Sedfit
[11] and Sedphat [1] while the relative amounts of monomer and
n-mers for self-associating systems can be calculated using the
Ultrascan software (http://www.ultrascan.uthscsa.edu/).
2.5.1 Sample Buffer The sample buffer should not absorb more than 0.3 OD,
referenced against water at the wavelength chosen for the experi-
ment as this may otherwise interfere with the range and linearity of
absorbance measurements (see Note 2). Charge repulsion between
macromolecules will lead to problems with non-ideality, which is
best addressed by using a buffer with ionic strength >0.1 M and by
carrying out the experiments close to the pI of the protein in order
to minimize the charge on the surface of the molecule. Further-
more, it should be borne in mind that samples might be in the
centrifuge for long periods of time (>60 h), and therefore, buffer
components that have significant time-dependent changes in their
absorbance spectrum, for instance DTT, should be avoided.
2.5.2 Protein The sample concentration should not exceed 0.7 OD when
Concentration measured against the sample buffer, otherwise the linear range of
the detector (approximately 1.5 OD) will be exceeded when the
absorbance rises towards the bottom of the cell during the run (see
Fig. 1). If no information about the association state of the protein
is available, 0.5 OD is a good starting point. If self-association is
present, the loading concentration should be chosen such that
there are detectable amounts of monomers and multimers (see
Subheading 2.4). The protein solution and sample buffer have to
be in equilibrium, this is best achieved by gel filtration (using for
example NAP-5 columns, GE Healthcare) or exhaustive dialysis
(2–3 buffer changes). We routinely use D-Tube Dialyzer, Novagen
which is available for a range of different volumes starting at 10 μl.
Before every ultracentrifugation run, the sample should be
checked for aggregation. This might be done by a separate sedi-
mentation velocity experiment or by static and/or dynamic light-
scattering analysis if an instrument is available. Otherwise, a simple
test is to spin the sample in a microcentrifuge for 15–20 min at
maximum speed and to recheck the absorbance (see Note 3). If the
sample OD has significantly decreased, the sample might not be
suitable for equilibrium analysis at this time.
down to 0.5 μM (see Note 2). To extend the range upwards shorter
pathlength cells (3 mm) are available extending the range up to
50 μM. Finally, data can be collected away from the peak of the
absorbance in the 250 nm trough or at 295 nm to give around a two
to threefold decrease in sensitivity, expanding the range up to 150 μM.
3 Methods
3.1 Cell Assembly The cells themselves are assembled for each centrifugation run.
There are several different types of cell but all consist of the follow-
ing main components: a cylindrical cell housing that fits into the
rotor, two window assemblies consisting of a window holder, a
quartz window, a white Vinylite window gasket, and a Bakelite
window liner. These two window assemblies form a sandwich
around a centerpiece that contains the samples. This sandwich is
assembled in the cell housing and a screw ring and gasket is then
screwed into one or both ends of the cell housing and tightened
with a torque wrench (see Note 4). Full details of the cell assembly
are provided in the manufacturer’s instructions and should be
followed very carefully (https://www.beckmancoulter.com/
wsrportal/techdocs?docname¼LXLA-TB-003).
There are many types of centerpieces constructed of different
materials and dimensions. The simplest centerpieces are the double
sector centerpieces, which allow for a sample solution and a buffer
blank. These have a path length of 1.2 cm and are made in alumi-
num, aluminum filled Epon or charcoal filled Epon. The aluminum
double sector centerpiece has the advantage of having a maximum
speed of 60,000 rpm while the others are limited to 42,000 rpm.
The charcoal filled Epon centerpiece should be used if there is any
possibility of aluminum ions interacting with the sample, otherwise
all of the centerpieces are compatible with aqueous solutions but
softening of the Epon can occur if strong acids or organic solvents
are used (see Note 4). A 3 mm path length charcoal filled Epon
centerpiece is also available and requires the use of two spacers.
Charcoal filled Epon centrerpieces that contain six channels (three
samples and three buffer blanks) have a maximum speed of
48,000 rpm whereas the eight channel cells (four samples and
four buffer blanks) can be operated up to 50,000 rpm. The double
sector centerpieces that hold 450 μl per sector are generally used for
sedimentation velocity measurements because of their longer col-
umn length. However, they can also be used for equilibrium runs if
the volume is reduced to 100 μl. This is particularly useful if the
3 mm path length centerpiece is required since this is not available
in six channel centerpieces. In this case, the volume is reduces to
33 μl. The volumes required to fill a 6-sector cell are 110 μl of
protein and 120 μl of sample buffer.
214 Ian A. Taylor et al.
0:7ðh Þ2
T eqm ¼ ð9Þ
DT
From Eq. 9 it is apparent that smaller molecules reach equilib-
rium faster than larger ones. A 20 kD protein in about 16 h and a
100 kD protein in around 27 h. A value for DT can be determined
experimentally, but for these purposes it is reasonable to estimate a
value by assuming the macromolecule is spherical and applying
Eq. 10.
3 105
DT ¼ p
3
ffiffiffiffiffi ð10Þ
M
Whichever method is used, it is still necessary to show experi-
mentally that the system has reached equilibrium. The best way to
do this is to overlay or subtract successive scans, taken at two hourly
intervals. When equilibrium is established no further change in the
absorbance profile should occur, the only differences being due to
noise in the data.
11. When data has been collected at all the necessary speeds, set the
centrifuge to 42,000 rpm. After approximately 16 h, collect a
radial scan using a 0.001 cm step size with two replicates. This
“overspeed” scan gives a depleted boundary that should be
used to provide a reasonable estimate of the cell offset during
the data fitting procedure (see Note 10).
3.4 Analyzing All data analysis described in the following paragraph has been
the Data carried out using the Origin® software package. This program
allows the direct fitting of the equilibrium concentration gradients
to mathematical models for single species or associating systems
using nonlinear least squares algorithms [12, 13]. Furthermore, it
allows global fitting of multiple datasets covering a range of con-
centrations and rotor speeds. This is particularly important for self-
associating systems.
1. Once a whole dataset is collected, select the files that will be
used in the molecular weight analysis. Load the relevant “RA”
files into Origin, then cut out the data corresponding to the
sample absorbance profiles using the select subset command
(see Note 11).
2. First, use the Origin software to individually fit each “cut” file
assuming an ideal single species model. In this fitting proce-
dure, include the offset value obtained from the “overspeed”
scan along with the calculated values for ρ and ν (see Note 10).
3. Plot the molecular weights obtained against the initial protein
concentration. Is there any correlation between initial concen-
tration and apparent molecular weight? If so, this may be
indicative of an interacting system.
4. Plot ln(C) against r2 (under Utilities/Plot) (see Note 12).
5. If the fit of the individual data files provided any indication of
self-association carry out a simultaneous (global) fit, still
assuming a single species. Under these conditions there should
be clear systematic deviations in the residuals plot indicating
that the protein is not an ideal monomeric species.
6. Carry out global fits to monomer–dimer, monomer–trimer,
and monomer–tetramer equilibria and carefully inspect for
deviations between the fitted curve and the experimental data
as well as for systematic deviations in the residuals plot. At this
stage it is of great importance to have some previous knowl-
edge of the monomer molecular weight of the protein under
investigation as this allows the monomer molecular weight to
be fixed (see Note 13).
7. Convert the association constant expressed in absorbance units
into the commonly used association constant Ka (see Note 14).
Sedimentation Equilibrium Studies 217
Fig. 3 Analysis of the solution molecular weight of Swi6 by sedimentation equilibrium. The lower panel shows
the absorbance profile produced at equilibrium by Swi6 at a rotor speed of 12,000 rpm, T ¼ 293 K,
ν ¼ 0.727, ρ ¼ 1.003. The upper panel shows the residuals to the plot
218 Ian A. Taylor et al.
Fig. 4 Sedimentation equilibrium data of dynamin I fitted to different models: (a) monomer alone; (b) dimer
alone; (c) tetramer alone; (d) monomer/tetramer. Reproduced with permission from Journal of Protein
Chemistry (1999) 18, 277–290
4 Notes
∑ni M i vi
v¼ ð12Þ
∑ni M i
K aabs ðεm l Þ
Ka ¼ ð13aÞ
2
K aabs ðεm l Þ2
Ka ¼ ð13bÞ
3
K aabs ðεm l Þ3
Ka ¼ ð13cÞ
4
Acknowledgement
References
1. Vistica J, Dam J, Balbo A et al (2004) Sedi- SE, Rowe AJ, Horton JC (eds) Analytical
mentation equilibrium analysis of protein inter- Ultracentrifugation in Biochemistry and Poly-
actions with global implicit mass conservation mer Science. The Royal Society of Chemistry,
constraints and systematic noise decomposi- Cambridge United Kingdom, pp 90–125
tion. Anal Biochem 326:234–256 11. Brown PH, Schuck P (2006) Macromolecular
2. Howlett GJ, Minton AP, Rivas G (2006) Ana- size-and-shape distributions by sedimentation
lytical ultracentrifugation for the study of pro- velocity analytical ultracentrifugation. Biophys
tein association and assembly. Curr Opin Chem J 90:4651–4661
Biol 10:430–436 12. Johnson ML, Faunt LM (1992) Parameter
3. Rivas G, Stafford W, Minton AP (1999) Char- estimation by least-squares methods. Methods
acterization of heterologous protein-protein Enzymol 210:1–37
interactions using analytical ultracentrifuga- 13. Johnson ML, Correia JJ, Yphantis DA et al
tion. Methods 19:194–212 (1981) Analysis of data from the analytical
4. Laue TM, Stafford WF 3rd (1999) Modern ultracentrifuge by nonlinear least-squares tech-
applications of analytical ultracentrifugation. niques. Biophys J 36:575–588
Annu Rev Biophys Biomol Struct 28:75–100 14. Sedgwick SG, Taylor IA, Adam AC et al (1998)
5. Cole JL, Lary JW, PMoody T et al (2008) Structural and functional architecture of the
Analytical ultracentrifugation: sedimentation yeast cell-cycle transcription factor swi6. J Mol
velocity and sedimentation equilibrium. Meth- Biol 281:763–775
ods Cell Biol 84:143–179 15. Binns DD, Helms MK, Barylko B et al (2000)
6. Cole JL, Hansen JC (1999) Analytical ultra- The mechanism of GTP hydrolysis by dynamin
centrifugation as a contemporary biomolecular II: a transient kinetic study. Biochemistry
research tool. J Biomol Tech 10:163–176 39:7188–7196
7. Liu J, Shire SJ (1999) Analytical ultracentrifu- 16. Binns DD, Barylko B, Grichine N et al (1999)
gation in the pharmaceutical industry. J Pharm Correlation between self-association modes
Sci 88:1237–1241 and GTPase activation of dynamin. J Protein
8. Cantor CR, Schimmel PR (1980) Ultracentri- Chem 18:277–290
fugation. In: biophysical chemistry, Part II. 17. Silkowski H, Davis SJ, Barclay AN et al (1997)
Freeman Characterisation of the low affinity interaction
9. Laue TM (1996) Choosing which optical sys- between rat cell adhesion molecules CD2 and
tem of the Optima XL-I analytical centrifuge to CD48 by analytical ultracentrifugation. Eur
use. Beckman-Coulter Technical Application Biophys J 25:455–462
Information Bulletin A-1821-A 18. Philo JS (2000) Sedimentation equilibrium
10. Laue TM, Shah BD, Ridgeway TM et al (1992) analysis of mixed associations using numerical
Computer-aided interpretation of analytical constraints to impose mass or signal conserva-
sedimentation data for proteins. In: Harding tion. Methods Enzymol 321:100–120
Chapter 13
Abstract
Upon protein-protein interaction, the formed complex is subject to a change in size. A number of methods
can be utilized to detect such a change. Gel filtration technology is well recognized for its ability to monitor
and separate protein species of different sizes, and can greatly facilitate functional studies of protein
complexes. In addition, gel filtration can be performed in any buffer system that preserves the protein
complex formation and function. Therefore, it can be a significantly useful method for studying protein
interactions. In this chapter, a protocol for performing gel filtration is described in detail.
Key words Gel filtration, FPLC, Molecular weight estimation, Protein dimerization
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_13, © Springer Science+Business Media New York 2015
223
224 Yan Bai
Small
molecule
pores
large
molecule
Fig. 1 Illustration of the gel filtration principle. Protein molecules pass through
the gel medium via different routes based on their size. Small molecules take
longer to elute because they can get into small pores and big pores; big
molecules take less time because they are too large to travel through the pores
Intermediate MW
Injection point
High MW
Low MW
Vo Vt
Void volume Vo Total volume Vt Vs(Vt-Vo)
Ve
Vt-Vo
Fig. 2 Common terms in gel filtration. Vo (the void volume): space taken by solvent surrounding the gel beads,
usually about 30 % of the total bed volume. Vt: the total volume of a gel bed. Vs: space occupied by solvent
inside the medium particles
column and prepare protein standards and samples. (3) Run protein
standards to establish a calibration curve. (4) Run protein samples.
(5) Analyze collected protein fractions on an SDS-PAGE gel.
1.1 General Terms A set of terminology is employed to describe the gel filtration
and Concepts in Gel process. As shown in Fig. 2, the total volume of a gel bed, Vt,
Filtration includes space occupied by solvent inside the medium particles Vs,
and space taken by solvent surrounding the gel beads called Vo (the
void volume), which is about 30 % of the total bed volume.
Vt ¼ Vo þVs ð1Þ
The elution volume, Ve, is the volume of mobile phase entering
the column between the start of the elution and the emergence of
the peak maximum [8].
K av ¼ ðV e V o Þ=ðV t V o Þ ð2Þ
K av ¼ alogðMW Þ þ b ð3Þ
2 Materials
3 Methods
3.1 Chromatography It is advisable to read the manufacture’s user manual for detailed
System Setup instrument operation and maintenance. A simplified outline for the
AKTApurifier FPLC system is provided here.
Gel Filtration 227
Sample Sample
2 syringe 2 syringe
6 6
3 3
Sample Sample
loop loop
4 5 4 5
Fig. 4 Illustration of the injection valve at inject mode and load mode. After being manually injected through
port 3, the sample will be retained in the sample loop between port 2 and port 6. (a) Upon sample loading on
the column, the system automatically switches to the “Inject” mode, in which the flow path is 7-6–2-1. (b)
When elution starts, the liquid flows directly from 7 to 1, bypassing the sample loop
3.2 Sample Loading 1. Equilibrate the column (see Note 8). Wash the column using at
and Fractionating least 1CV (column volume) of filtered distilled water, and
equilibrate the column using at least 1CV of running buffer.
By the end of the equilibration, the conductivity trace should
show an “S” shape and level off in the end. The operation can
be set up and executed through Manual on the System control
module.
2. Wash the sample loop. The sample loop (0.5 ml) should be well
cleaned using running buffer. Use a needle syringe to inject 1 ml
of running buffer into the fill port three times (see Note 9).
3. Load the sample. Go to the UNICORN System Control inter-
face, under Manual/ flow path, set “Inject valve” to “Inject”
before taking out the syringe. Take out the syringe and fill the
syringe with 180–200 μl of protein sample (see Note 10).
Insert the syringe needle into the fill port. Set “Inject valve”
Gel Filtration 229
to “load”, and then slowly inject the sample into the loop.
Leave the syringe in position during the entire run. Go to the
System Control interface, click “End” on the operation tool
bar. Ensure that 1 mL of buffer volume is used to empty the
sample loop in order to load all of the sample onto the column.
4. Start a run. Right click the “Run” button on the top tool bar to
select a method for the run. Several dialog windows will be
displayed sequentially, on which the user can review/edit the
method settings and set up how the results file should be
generated. Click “Start” on the last page of the dialog window
to initiate the run.
5. Monitor the run. The gel filtration progress can be monitored
in the System Control module. The ongoing experiment can
be pulsed or ended at any time by clicking the “Pulse” or
“End” button on the top tool bar.
3.3 Create the Studies [9, 10] have shown that, for globular proteins, their parti-
Standard Curve and tion coefficients, Kav, are related to their corresponding molecular
Estimate the Molecular weights in a sigmoidal fashion on gel filtration columns. Therefore,
Weight a calibration curve, also called a selectivity curve, can be created by
plotting a group of known proteins’ partition coefficients against
the logarithm of their molecular weight. Other elution parameters,
such as Ve, Ve/Vo, Kd, have been used in the calibration curve,
however the use of Kav is recommended because it is insensitive to
errors that may be generated by column preparation and dimension
variations.
An unknown protein sample’s molecular weight can be esti-
mated according to its elution volume using a calibration curve. It
is important to make sure that the calibration curve’s linear range
covers the size of the protein of interest since only the linear region
of the plot is reliable [10]. When gel filtration is used to detect or
confirm protein complex formation, the protein complex’s molec-
ular weight is usually considered as the sum of each individual
protein. However, when the complex exhibits an irregular shape,
the molecular weight cannot be simply extrapolated from the cali-
bration curve. For example, an elongated protein complex will
behave like a bigger molecular on the gel chromatogram than it
actually is.
1. Determine the void volume (Vo). Make 2 mg/ml blue dextran
in running buffer (see Note 11).
2. Prepare the protein standards. Dissolve 3.5 mg of Albumin
bovine serum, 1 mg of Carbonic Anhydrase and 1 mg of
aprotinin in 500 μl of running buffer (see Note 12).
3. Equilibrate the column using running buffer and run the pro-
tein standards as described in Subheading 3.2.
4. Calculate Kav values for each protein maker.
230 Yan Bai
a b mAU
MW: KDa
Kav 200 14-3-3
0.5 75
160
0.4 37
25 14-3-3
120
0.3
80
0.2
y = -0.3185x + 0.6917 Void
0.1 R² = 0.9998 40
0 0
0.0 0.5 1.0 1.5 2.0 Log Mr 0 5 10 15 20 25 ml
Fig. 5 (a) A standard curve was created based on the experimentally determined Kav of the standard proteins.
(b) Chromatogram of the protein sample. The experimental Ve is 9.39 mL. Therefore, the estimated molecular
weight of the protein sample is 61.74 kDa, indicating a dimer form of the protein. The collected fractions were
pooled and run on 12.5 % SDS-PAGE, the protein monomer was observed on the gel at approximately 30 kDa
4 Notes
5. The user can control the UNICORN displays and manage files
through the UNICORN manager module. Method Editor is
for creating new methods. System Control is used to monitor
and control runs in real time. The results for each run will be
displayed and evaluated in Evaluation module.
6. Correct maximum pressure setup is the most critical parameter
to protect columns. Ensure that the back-pressure over the
column does not exceed the maximum recommended pressure
(1.8 M for a Superdex 75). The recommended maximum
pressure for a selected column can be found in manufacturer’s
instruction sheet. Pressure could gradually build up with
increasing system usage. One of the common reasons is that
impurities build up on the in-line filter. The filter can be cleaned
by soaking it in 0.1 N NaOH in a beaker with gentle agitation
overnight and rinsing it thoroughly with filtered deionized
water. In addition, the columns have to be washed completely
after each use. Once column clogging is observed, a stringent
cleaning operation has to be performed immediately according
to manufacturer’s instructions.
7. The process of setting up a fraction collector varies depending
on the type of collector used.
8. The column equilibration step can be incorporated into the
method through the Method Wizard so that the equilibration
will automatically end and switch to the elution step. However,
it may be more advantageous to have the equilibration run
separately through manual control, by which the user can
alter the equilibration volume as needed to make sure that the
column is perfectly equilibrated before starting a run.
9. If partial filling is used (see Note 10), at the end of the last loop
wash, do not remove the syringe from the fill port, otherwise
the buffer inside the sample loop will drain out.
10. There are two ways to load the sample, partial filling and
complete filling. When a high recovery rate is required, partial
filling is used. The maximum sample volume loaded cannot
exceed 50 % loop volume, 1/3 of the loop volume is usually
loaded. Make sure the sample loop is completely filled with
buffer before sample loading. Complete filling requires about
2–3 loop volumes of the sample to achieve 95 % maximum loop
volume. For example, in order to fill a 0.5 mL sample loop with
95 % protein sample without diluting it, 1–1.5 mL of protein
sample is required to be injected into the loop. When complete
filling is used, a buffer volume at least five times the sample loop
volume should be used to flush the loop during emptying the
loop.
11. 5 % glycerol can be contained in the running buffer to increase
the density of the solution, but it is optional.
232 Yan Bai
12. If more than three protein markers are used, dissolving all
markers together may cause unassignable peaks because BSA
may form a complex with other protein markers. It is advisable
to prepare multiple protein marker groups, 2–3 proteins for
each group.
References
1. Ackers GK (ed) (1975) The proteins, vol 1. assimilation in Haloferax mediterranei: inter-
Molecular sieve methods of analysis. Academic, action between glutamine synthetase and two
New York, NY GlnK proteins. Biochim Biophys Acta 1834:
2. Cooper TG (ed) (1977) Gel permeation chro- 16–23
matography. The tools of biochemistry. Wiley, 7. Kuo WY, Huang CH, Liu AC et al (2013)
New York CHAPERONIN 20 mediates iron superoxide
3. Freydell EJ, van der Wielen LA, Eppink MH dismutase (FeSOD) activity independent of its
et al (2010) Size-exclusion chromatographic co-chaperonin role in Arabidopsis chloroplasts.
protein refolding: fundamentals, modeling and New Phytol 197:99–110
operation. J Chromatogr A 1217:7723–7737 8. Wechsler DS, Dang CV (1992) Opposite
4. Hall D, Huang L (2012) On the use of size orientations of DNA bending by c-Myc and
exclusion chromatography for the resolution of Max. Proc Natl Acad Sci U S A 89:7635–7639
mixed amyloid aggregate distributions: I. 9. Porath J, Flodin P (1959) Gel filtration: a
Equilibrium partition models. Anal Biochem method for desalting and group separation.
426:69–85 Nature 183:1657–1659
5. Monti M, Cozzolino M, Cozzolino F et al 10. Abate C, Luk D, Gentz R et al (1990)
(2009) Puzzle of protein complexes in vivo: a Expression and purification of the leucine
present and future challenge for functional pro- zipper and DNA-binding domains of Fos
teomics. Expert Rev Proteomics 6:159–169 and Jun: both Fos and Jun contact DNA
6. Pedro-Roig L, Camacho M, Bonete MJ directly. Proc Natl Acad Sci U S A 87:
(2013) Regulation of ammonium 1032–1036
Chapter 14
Abstract
The stoichiometry of a protein complex can be calculated from an accurate measurement of the complex’s
molecular weight. Multiangle laser light scattering in combination with size exclusion chromatography and
interferometric refractometry provides a powerful means for determining the molecular weights of proteins
and protein complexes. In contrast to conventional size exclusion chromatography and analytical centrifu-
gation, measurements do not rely on the use of molecular weight standards and are not affected by the
shape of the proteins. The technique is based on the direct relationship between the amount of light
scattered by a protein in solution, and the product of its concentration and molecular weight. A typical
experimental configuration includes a size exclusion column to fractionate the sample, a light scattering
detector to measure scattered light, and an interferometric refractometer to measure protein concentration.
The determination of the molecular weight of an anthrax toxin complex will be used to illustrate how
multiangle laser light scattering can be used to determine the stoichiometry of protein complexes.
Key words Light scattering, Stoichiometry, Molecular weight, Size exclusion chromatography,
Interferometric refractometer, Protein complex
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_14, © Springer Science+Business Media New York 2015
233
234 Jeremy Mogridge
2 Materials
1. HPLC.
2. DAWN EOS 18-angle light scattering detector (Wyatt
Technology).
3. OPTILAB DSP interferometric refractometer (Wyatt
Technology).
4. ASTRA software (Wyatt Technology).
5. Superdex 200 HR 10/30 column.
6. Column buffer: 20 mM Tris–HCl, pH 8.2, 200 mM NaCl.
7. 0.2 μm filter units.
8. 0.02 μm Anotop 10 filters.
3 Methods
3.1 System Setup Before measurements are taken, the LS detector must be calibrated
with a pure solvent that has a known Rayleigh ratio, such as toluene.
Calibration relates the voltage detected by the 90 detector to the
measured intensity of scattered light. If the LS detector was cali-
brated by the manufacturer, it needs to be re-calibrated only if any
changes to the detector that affects the 90 signal, such as realign-
ment of the laser, have been made.
The HPLC is connected in series to the LS detector and the IR.
The IR should be placed after the LS detector to avoid subjecting
the IR to high back-pressure. Connections should be made with
Light Scattering 235
3.3 Injecting Do not stop the HPLC pumps after the equilibration step or
a Sample between measurements because starting and stopping the pumps
may cause particles from the column to dislodge and interfere with
the measurements. Start data collection by ASTRA software and
inject each component of the protein complex (EF and PA63; see
Note 5) individually and then a mixture of the proteins (see Note
6). The results of a chromatography run in which a molar excess of
EF was mixed with PA63 are shown in Fig. 1.
3.4 Analyzing The results can be analyzed using a computer program, such as
the Results ASTRA.
1. Set the baseline voltages of the signals detected by the LS
detector and IR.
2. When selecting values for the light scattering and refractive
index signals, use data relating to the middle of the protein
236 Jeremy Mogridge
Table 1
Molecular mass determinations using multiangle laser light scattering
4 Notes
References
1. Wyatt PJ (1993) Light scattering and the abso- 5. Zhao J, Milne JC, Collier RJ (1995) Effect of
lute characterization of macromolecules. Anal anthrax toxin’s lethal factor on ion channels
Chim Acta 272:1–40 formed by the protective antigen. J Biol Chem
2. Gell DA, Grant RP, Mackay JP (2012) The 270:18626–18630
detection and quantitation of protein oligomer- 6. Miller CJ, Elliott JL, Collier RJ (1999) Anthrax
ization. Adv Exp Med Biol 747:19–41 protective antigen: prepore-to-pore conversion.
3. Mogridge J, Cunningham K, Collier RJ (2002) Biochemistry 38:10432–10441
Stoichiometry of anthrax toxin complexes. Bio- 7. Folta-Stogniew E, Williams KR (1999) Deter-
chemistry 41:1079–1082 mination of molecular masses of proteins in
4. Young JA, Collier RJ (2007) Anthrax toxin: solution: implementation of an HPLC size
receptor binding, internalization, pore forma- exclusion chromatography and laser light scat-
tion, and translocation. Annu Rev Biochem tering service in a core laboratory. J Biomol Tech
76:243–265 10:51–63
Chapter 15
Abstract
Circular dichroism (CD) spectroscopy is a useful technique for studying protein-protein interactions in
solution. CD in the far ultraviolet region (178–260 nm) arises from the amides of the protein backbone and
is sensitive to the conformation of the protein. Thus, CD can determine whether there are changes in the
conformation of proteins when they interact. Changes in the conformation of the protein complexes as a
function of temperature or added denaturants, compared to the individual proteins, can be used to
determine binding constants. CD bands in the near ultraviolet (350–260 nm) and visible regions arise
from aromatic amino acid side chains and prosthetic groups. There are often changes in these regions when
proteins bind to each other. Because CD is a quantitative technique, these changes are directly proportional
to the amount of the protein–protein complexes formed and thus also can be used to estimate binding
constants.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_15, © Springer Science+Business Media New York 2015
239
240 Norma J. Greenfield
1.1 Spectra The intrinsic CD spectrum of a protein, arising from its amide
of the Peptide backbone, is sensitive to the secondary structure of the protein.
Backbone The amide backbone bonds absorb in the far UV. Most commercial
CD machines can collect far UV spectra between 260 and 178 nm.
To a first approximation, increases in negative ellipticity at 222 and
208 nm and positive ellipticity at 193 nm usually indicate an
increase in α-helical content, while increases in a single negative
band near 218 nm and positive band at 195 nm indicates an
increase in β-structure. Figure 1a shows typical CD curves obtained
for a polypeptide, Poly-L-lysine in the 100 % α-helical, 100 %
β-sheet, and 100 % denatured states [9]. Figure 1b illustrates the
increase in α-helix that occurs when a peptide from titin, a Z-repeat,
interacts with a fragment of the C-terminal domain of α-actinin
[10]. Figure 1c illustrates the increase in β-structure when two cell
cycle control proteins, Arf and Hdm2, interact [11]. Both of these
proteins are mainly disordered in the absence of the interactions.
1.2 Side Chain and Non-backbone intrinsic CD bands are due to the chromophores of
Extrinsic CD Spectra the aromatic amino acids, tryptophan, tyrosine, and phenylalanine
and disulfide bonds. Extrinsic bands are due to noncovalently
bound chromophores such as hemes, aromatic cofactors, and col-
ored metal ions. Figure 2a illustrates a change in the aromatic
spectrum of the catalytic domain of a cAMP-dependent protein
kinase when it binds to an artificial peptide substrate [12], and
Fig. 2b illustrates the change in the heme spectrum of cytochrome
C when it binds to cytochrome C oxidase [13].
1.3 Binding There are four methods that can be used to extract binding (or
Constants dissociation) constants from CD data. These include:
1. Direct titration of one protein with another.
2. Serial dilution of a protein complex.
3. Changes in susceptibility to denaturants or osmolytes of the
protein complex compared to the unbound components.
4. Changes in thermal stability of the complex compared to the
unbound components.
If formation of the complex changes the protein secondary
structure or causes changes in the environment of optically active
aromatic or prosthetic groups of one or both of the interacting
proteins, one protein can be titrated by the other. The change in
CD upon binding is directly proportional to the amount of com-
plex formed, and thus the binding constant can be directly
CD Spectroscopy 241
2 Materials
2.2 Buffers For the most precise estimates of protein secondary structure from
CD spectra, it is important to collect data to low wavelengths;
178 nm has been recommended [3, 14, 15]. In this case one should
use buffers with very low absorption, such as 10 mM potassium
CD Spectroscopy 243
2.4 Instrumentation The following is a list of companies that currently produce com-
mercially available CD machines. All are adequate to perform ther-
mal denaturations and titration experiments. They are listed
alphabetically:
1. Applied Photophysics Ltd, 21 Mole Business Park, Leather-
head, Surrey KT22 7BA, UK.
2. Aviv Biomedical, 750 Vassar 750 Vassar Avenue—Suite 2 Lake-
wood, NJ 08701-6929, USA.
3. JASCO Inc., 28600 Mary’s Court, Easton, MD 21601, USA.
4. Olis, Inc., 130 Conway Drive Suites A, B & C, Bogart, GA
30622, USA.
244 Norma J. Greenfield
3 Methods
3.1 Analyzing While changes at 222 or 218 nm can give an estimate of increases in
Changes in α-helical or β-structure content upon protein complex formation,
Conformation more precise estimates of changes in structure accompanying pro-
Accompanying tein-protein interactions can be made utilizing computer programs.
Protein-Protein Computer programs for analyzing CD spectra are not available
Interactions commercially, but many are available from their authors without
charge (see Note 1 for sources of software).
The use of the various methods to determine protein confor-
mation from CD spectra has been reviewed, with a detailed com-
parison of the advantages and disadvantages of each [5], so only a
brief description of each method is given below.
Fig. 3 (a) Circular dichroism spectra of (open circles) E-Tmod1-130, (inverted filled triangles) AcTM1bZip,
(inverted open triangles) the sum of the spectra of E-Tmod1-130 and AcTM1bZip, and (filled circles) the
spectrum of the mixture E-Tmod1-130 and AcTM1bZip. Data are the average and standard error of four to
seven measurements. (b) The mean residue ellipticity of (open circles) Tmod11-130, (inverted filled triangles)
AcTM1bZip, and (filled circles) the mixture of the two peptides. The data are fit by (dashed dotted line)
constrained and (dotted line) nonconstrained least squares fits. (c) (Filled circles) Difference between the
spectrum of the mixture of the components and the sum of the unmixed components in (a), (Solid line)
nonconstrained least squares fit of the data. The difference data are the average of four measurements. The
data are from Greenfield and Fowler [21] used with permission from Elsevier
the data of Brahms and Brahms [16] for the spectra of peptides
in the β-pleated sheet, β-turn, and disordered conformations.
Figure 3a shows the raw spectra of the two unmixed peptides, the
sum of the spectra of the unmixed peptides, and the spectrum of
the complex. Figure 3b shows the mean residue ellipticities of the
individual proteins and the complex and the best fits using
constrained and nonconstrained multilinear regression. Figure 3c
shows the difference spectrum of the complex minus the sum of
unmixed peptides and the best fit to the curves using non-
multilinear regression with the same peptide references. The per-
centage of the conformations for the nonconstrained fits were
normalized to a sum of 100 %. The results are compared in Table 1.
The results for the TM1bZip peptide are also compared to the
structures determined by X-ray crystallography [24] and NMR
[25]. The number of residues of each protein fragment in each
conformation was calculated by multiplying the percentage of each
conformation by the number of residues of each protein fragment,
74 for AcTM1bZip, 142 for E-Tmod (Tmod11-130), and 216 for
the complex. When the tropomyosin peptide binds to the tropo-
modulin fragment, there is an increase in ellipticity of about 10 %
relative to the unmixed peptides. When the data in Fig. 3b is
analyzed using LINCOMB, the results suggested that most of the
change is due to an increase in helical content of about 22 residues,
~10 %, and a small increase in β-sheet residues, 3, ~1 %. When the
nonconstrained least squares fitting program MLR was used, the
results suggested that there was an almost equal increase in both
helical and β-sheet content of approximately 6 %, 14 and 13 resi-
dues, respectively. When the difference spectrum (Fig. 3c) was
analyzed directly, the results suggested that the increase in ellipticity
upon binding was 44 % due to an increase in helix and 55 % increase
in β-sheet. NMR studies of a similar E-Tmod peptide containing
residues 1–92 [23] show only that 12 residues [21–35] of tropo-
modulin are α-helical in the absence of tropomyosin peptides, but
residues 1–35 become ordered when tropomodulin binds to the
N-terminus of tropomyosin, an increase of 23 residues, in reason-
able agreement with the calculations from the CD data. Mutation
experiments suggest the TMod and tropomyosin peptides bind via
formation of a multichain α-helical coiled coil, so the constrained
least squares fits of the CD data are more accurate than the non-
constrained fits.
3.1.3 Ridge Regression CONTIN, developed by Provencher and Glöckner [26], fits the
(CONTIN) CD of unknown proteins by a linear combination of the spectra of a
large database of proteins with known conformations. In this
method, the contribution of each reference spectrum is kept
small, unless it contributes to a good agreement between the
theoretical best fit curve and the raw data. The method usually
gives very good estimates of β-sheets and turns and gives good
fits with polypeptides [5].
Table 1
Characterization of the secondary structural changes induced by AcTM1bZip-Tmod11-130 complex formation
CD Spectroscopy
β-Sheet 16.72 % 20.56 % 36.1 44.4
β-turn 0.00 % 0.00 % 0.0 0.0
Remainder 29.29 % 30.63 % 63.3 66.2
Total sum 100.00 % 100.00 % 216.0 216.0
(continued)
247
Table 1
248
(continued)
Norma J. Greenfield
Number of residues in each conformation
3.1.4 Singular Value The application of singular value decomposition to determine the
Decomposition (SVD, secondary structure of proteins by comparing their spectra to the
VARSLC, CDSSTR, spectra of a large number of proteins with known structure was first
SELCON) developed by Hennessey and Johnson [14]. This method extracts
basis curves, with unique shapes, from a set of spectra of proteins with
known structures. Each basis curve is then related to a mixture of
secondary structures, which are then used to analyze the conforma-
tion of unknown proteins. The method is excellent for estimating the
α-helical content of proteins but is poor for sheets and turns unless
the data is collected to very low wavelengths, at least 184 nm [14].
Several newer programs have improved the method of singular
value decomposition by selecting references that have spectra that
closely match the protein of interest. They include VARSLC [15],
SELCON [27–31], and CDSSTR [32]. These give good estimates
of β-sheet and turns in proteins and work with data collected only
to 200 nm but give poor fits to polypeptides with high β-structure
content because reference sets for such compounds are not in their
database [5].
3.1.5 Neural Network A neural network is an artificial intelligence program which can
Programs (CDNN and K2D) detect patterns and correlations in data. Two widely used programs
are CDNN [33] and K2D [34]. A neural network is first trained
using a set of known proteins so that the input of the CD at each
wavelength results in the output of the correct secondary structure.
The trained network is then used to analyze unknown proteins. The
method works very well and the fits seem to be relatively indepen-
dent of the wavelength range that is analyzed [5].
3.1.6 Convex Constraint The CCA algorithm [19, 20, 35] deconvolutes a set of spectra into
Algorithm (CCA) basis spectra that when recombined generate the entire data set
with a minimum deviation between the original data set and recon-
structed curves. It is very useful for determining whether there are
intermediate states in thermal and denaturant induced unfolding.
The method has also been used to estimate protein conformation
but is poorer than least squares, SVD, or neural net analyses [5].
Table 2
Secondary structure analysis of fragments and cleaved and uncleaved thioredoxin
3.2 Determining When two proteins interact, there are often changes in either
Protein–Protein intrinsic or extrinsic circular dichroism. CD is a quantitative tech-
Association Constants nique and the change in CD, relative to the ellipticity of the
Using CD Data unmixed components, is directly proportional to the amount of
complex formed. The change can then be used to determine the
3.2.1 Determination of association or dissociation constant of the complex. Ideally, when
Binding Constants by Direct performing titrations, the concentration of the peptide being
Titration of One Protein by titrated, e.g., protein A, should remain constant. Thus, one should
Another at Constant titrate protein A with aliquots of a solution containing the starting
Temperature (Isothermal concentration of A and a large excess of protein B. In this case one
Titrations) does not have to correct for dilution of protein A when determining
the change in ellipticity upon complex formation.
When one titrates A with B to form a complex AB, the associa-
tion constant is K.
where [Ao] and [Bo] are the initial concentrations of A and B and
[AB] is the amount of complex formed. The saturation fraction is s.
s ¼ ½AB=½Ao ð2Þ
Weak Binding Constants If the binding of one protein to the other is relatively weak, under
conditions where the dissociation constant, 1/k, of the complex is
more than 100-fold the concentration of A, one can assume that
the concentration of unbound B is approximately equal to the total
amount of B added to A. In this case it is easy to determine the
binding constant and one can use linear plots to determine Δ[θ]max
and KD.
252 Norma J. Greenfield
For example one can fit the data to the Scatchard equation [38]
where
Δ½θ=½Ao ½B ¼ kðε Δ½θ=½Ao Þ ð5Þ
One plots Δ[θ]/[B] versus Δ[θ]. The y intercept ¼ kε[Ao] and the
slope ¼ k.
The Scatchard equation [38] described above works well for
studying interactions where there is one or multiple equivalent
binding sites of one protein for another. However, when there are
multiple sites, and the binding is cooperative, it is much more
difficult to estimate binding constants from spectroscopic data,
and usually only apparent constants can be estimated. The Hill
equation [39] may be used to fit the change in CD as a function
of titrant in a phenomenological fashion to give an estimate of the
apparent binding affinity. Here, the first protein A is titrated with a
second protein B, where [Bo] is the total concentration of added
protein giving an observed change in ellipticity Δ[θ].
Tight Binding Constants If the dissociation constant is close to the concentrations of the
proteins being studied, one cannot assume that the free concentra-
tion of the added protein, B, is the same as the total protein. One
must correct the concentration of the added protein B for the
amount that is bound. In this case [B] ¼ [Bo][AB]. From
Eq. 4, the fraction of protein A which is bound
½A bound ¼ ½A O Δ½θ=Δ½θmax ð7Þ
½B ¼ ½Bo ½Ao Δ½θ=Δ½θmax ð8Þ
Estimating Binding When two unfolded or partially unfolded proteins fold upon bind-
Constants When ing to each other, a simple method of determining the dissociation
Association of the Proteins constant is to follow the mean residue or molar ellipticity of a 1:1
Is Coupled with Folding mixture of the proteins as a function of concentration. In this case
h 1=2 i
kD þ k2D þ 4P t kD
½θobs ¼ ½θF þ ½θU ½θF =ð2P t Þ
ð10Þ
Fig. 5 Increase in ellipticity at 222 nm, 30 C, when fragments of SK- and E-tropomodulin, (a) Sk-Tmod1-130
and (b) E-Tmod1-130 bind to chimeric peptides containing the N-termini of long and short α-tropomyosins,
(open circles) AcTM1aZip and (filled squares) AcTM1bZip, respectively [21]. 10 nmol of the TMZip peptides in
0.5 ml of 100 mM NaCl, 10 mM sodium phosphate, pH 6.5 in 2 mm pathlength cells were titrated with
concentrated solutions of the Tmod peptides. The ellipticity changes were corrected for the ellipticity
of the Tmod peptides alone and for dilution. The data were fit (solid line) to the equation
2 1=2
Δ½θ ¼ Δ½θmax 1 þ k ½Bo þ k ½Ao =2 k ½Ao 1 þ k ½Bo þ k ½Ao =2k ½Ao ½Bo =½Ao
using the Levenberg–Marquardt algorithm [43] implemented in the commercial program SigmaPlot to yield
dissociation constants for Sk-TMod1-130 for AcTM1aZip and AcTM1bZip of 1.2 0.9 and 0.3 0.3 μM,
respectively and for E-Tmod1-130 for AcTM1aZip and AcTM1bZip of 2.8 1.2 and 0.3 0.2 μM, respec-
tively. Data from Greenfield and Fowler [21] and used with permission from Elsevier
Fig. 6 Association of two complementary fragments of the immunoglobulin binding domain B1 of streptococcal
protein G, PGB1(1–40) and PGB1(41–56), evaluated by molecular ellipticity at 222 nm. Equimolar
concentrations of the fragments were dissolved in 50 mM phosphate buffer (pH 5.5) at 298 K. The filled
circles and solid line hshow observed data and besti curve fitted to the equation
0:5
k D þ k 2D þ 4P t k D
½θobs = ½θF þ ½θU ½θF =ð2P t Þ to determine the apparent dissociation
constant of the two fragments (Kapp ¼ 9 106 M). Data from Honda et al. [41]. Used with permission from the
American Chemical Society
CD Spectroscopy 255
Chemical Induced In the simplest case, two proteins A and B form a complex with 1:1
Unfolding stoichiometry. The isolated proteins are unfolded but fold to form
the complex. When this complex is treated with denaturants, the
complex (native form) dissociates to give the disordered proteins. At
any concentration of denaturant, [D], the association constant is:
ð12Þ
ΔG ¼ RT lnk ð13Þ
ΔG D ¼ ΔG o þ m½D ð14Þ
where [θ]U and [θ]F are the ellipticities of the fully unfolded and
folded complexes, respectively, and mU and mF are any necessary
256 Norma J. Greenfield
k ¼ α=ð1 αÞ ð16Þ
ΔG ¼ RT lnk ð17Þ
k ¼ expðΔG=RT Þ ð20Þ
α ¼ k=ð1 þ kÞ ð21Þ
½θobs ¼ ½θF ½θU α þ ½θU ð22Þ
4 Notes
ΔG ¼ ΔG o þ m½D
k ¼ expðΔG=1:987 T Þ
n 2 o
2 P 2t k þ P t sqrt 2P 2t k P t 4P t4 k2 = 2P 2t k
α ¼
½θobs ¼ α ½θF m F ½D ½θU mU ½D þ ½θU
K ¼ expðΔG=RT Þ
α ¼ K =ð1 þ K Þ
½θobs ¼ ½θF ½θU α þ ½θU
(b) Homodimers
ΔG ¼ ΔH þ ðΔCpðT T m ÞÞ
T ððΔH =T m Þ þ ðΔCpðlnðT =T m ÞÞÞÞ
k ¼ expðΔG=ð1:987 T ÞÞ
a ¼ 4kP 2t
b ¼ 8kP 2t P t
c ¼ 4kP 2t
(c) Heterodimers
Here the equations are set up in terms of unfolding and Pt is
the total concentration of protein in terms of monomers,
262 Norma J. Greenfield
kD ¼ expðΔG=1:987 T Þ
a ¼ 2k
b ¼ 2P t 2 k
c ¼ Pt
b sqrt b 2 4 ac =2a
α ¼
½θobs ¼ α ½θF ½θU þ ½θU
z ¼ 3P 2t
q ¼ ðð3kz Þ þ 1Þ=ðkz Þ
e ¼ q3
d ¼ q3
n ð1=2Þ oð1=3Þ
½ð1Þðd=2Þ þ d 2 =4 þ e 3 =27
A ¼
n ð1=2Þ oð1=3Þ
B ¼ ð1Þ ðd=2Þ þ d 2 =4 þ e 3 =9
CD Spectroscopy 263
X ¼ Aþ B
α ¼ X þ 1
½θobs ¼ α ½θF ½θU þ ½θU
Q ¼ 1= 12 P 2t k
R ¼ 1= 8 P 2t k
ð1=3Þ
A ¼ R þ sqrt R2 Q 3
B ¼ Q =A
y ¼ A þ B
α ¼ 1y
½θobs ¼ α ½θF ½θU þ ½θU
References
1. Adler AJ, Greenfield NJ, Fasman GD (1973) 3. Johnson WC Jr (1990) Protein secondary
Circular dichroism and optical rotatory disper- structure and circular dichroism: a practical
sion of proteins and polypeptides. Methods guide. Proteins 7:205–214
Enzymol 27:675–735 4. Sreerama S, Woody RW (1995) Computation
2. Johnson WC Jr (1988) Secondary structure of and analysis of protein circular dichroism spec-
proteins through circular dichroism spectros- tra. Methods Enzymol 383(Part D):318–351
copy. Annu Rev Biophys Biophys Chem 5. Greenfield NJ (1996) Methods to estimate the
17:145–166 conformation of proteins and polypeptides
264 Norma J. Greenfield
from circular dichroism data. Anal Biochem 19. Perczel A, Park K, Fasman GD (1992) Analysis
235:1–10 of the circular dichroism spectrum of proteins
6. Greenfield NJ (2006) Using circular dichroism using the convex constraint algorithm: a prac-
spectra to estimate protein secondary struc- tical guide. Anal Biochem 203:83–93
ture. Nat Protoc 1:2876–2890 20. Perczel A, Park K, Fasman GD (1992) Decon-
7. Greenfield NJ (2006) Using circular dichroism volution of the circular dichroism spectra of pro-
collected as a function of temperature to deter- teins: the circular dichroism spectra of the
mine the thermodynamics of protein unfolding antiparallel beta-sheet in proteins. Proteins
and binding interactions. Nat Protoc 13:57–69
1:2527–2535 21. Greenfield NJ, Fowler VM (2002) Tropomyo-
8. Greenfield NJ (2006) Determination of the sin requires an intact N-terminal coiled coil to
folding of proteins as a function of denaturants, interact with tropomodulin. Biophys J
osmolytes or ligands using circular dichroism. 82:2580–2591
Nat Protoc 1:2733–2741 22. Greenfield NJ, DeGregori H (1993) Confor-
9. Greenfield N, Fasman GD (1969) Computed mational intermediates in the folding of a
circular dichroism spectra for the evaluation of coiled-coil model peptide of the N-terminus
protein conformation. Biochemistry of tropomyosin and αα-tropomyosin. Protein
8:4108–4116 Sci 2:1263–1273
10. Joseph C, Stier G, O’Brien R, Politou AS, 23. Greenfield NJ, Kostyukova A, Hitchcock-
Atkinson RA, Bianco A, Ladbury JE, Martin DeGregori SE (2005) Structure and tropomy-
SR, Pastore A (2001) A structural characteriza- osin binding properties of the N-terminal cap-
tion of the interactions between titin Z- repeats ping domain of tropomodulin 1. Biophys J
and the alpha-actinin C-terminal domain. Bio- 88:372–383
chemistry 40:4957–4965 24. Meshcheryakov VA, Krieger I, Kostyukova AS,
11. Bothner B, Lewis WS, DiGiammarino EL, Samatey FA (2011) Structure of a tropomyosin
Weber JD, Bothner SJ, Kriwacki RW (2001) N-terminal fragment at 0.98 Å resolution.
Defining the molecular basis of Arf and Hdm2 Acta Crystallogr D Biol Crystallogr
interactions. J Mol Biol 314:263–277 67:822–825
12. Reed J, Kinzel V (1984) Near- and far-ultraviolet 25. Greenfield NJ, Huang YJ, Palm T, Swapna GV,
circular dichroism of the catalytic subunit of Monleon D, Montelione GT, Hitchcock-
adenosine cyclic 50 -monophosphate dependent DeGregori SE (2001) Solution NMR structure
protein kinase. Biochemistry 23:1357–1362 and folding dynamics of the N terminus of a rat
13. Michel B, Proudfoot AE, Wallace CJ, Bosshard non-muscle alpha-tropomyosin in an engi-
HR (1989) The cytochrome c oxidase- neered chimeric protein. J Mol Biol
cytochrome c complex: spectroscopic analysis 312:833–847
of conformational changes in the protein–pro- 26. Provencher SW, Glockner J (1981) Estimation
tein interaction domain. Biochemistry of globular protein secondary structure from
28:456–462 circular dichroism. Biochemistry 20:33–37
14. Hennessey JP Jr, Johnson WC Jr (1981) Infor- 27. Sreerama N, Woody RW (2000) Estimation of
mation content in the circular dichroism of protein secondary structure from circular
proteins. Biochemistry 20:1085–1094 dichroism spectra: comparison of CONTIN,
15. Manavalan P, Johnson WC Jr (1987) Variable SELCON, and CDSSTR methods with an
selection method improves the prediction of expanded reference set. Anal Biochem
protein secondary structure from circular 287:252–260
dichroism spectra. Anal Biochem 167:76–85 28. Sreerama N, Woody RW (1994) Poly(pro)II
16. Brahms S, Brahms J (1980) Determination of helices in globular proteins: identification and
protein secondary structure in solution by vac- circular dichroic analysis. Biochemistry
uum ultraviolet circular dichroism. J Mol Biol 33:10022–10025
138:149–178 29. Sreerama N, Woody RW (1994) Protein sec-
17. Saxena VP, Wetlaufer DB (1971) A new basis ondary structure from circular dichroism spec-
for interpreting the circular dichroic spectra of troscopy. Combining variable selection
proteins. Proc Natl Acad Sci U S A principle and cluster analysis with neural net-
68:969–972 work, ridge regression and self-consistent
methods. J Mol Biol 242:497–507
18. Chang CT, Wu C-SC, Yang JT (1978) Circular
dichroic analysis of protein conformation: 30. Sreerama N, Woody RW (1993) A self-
inclusion of β-turns. Anal Biochem 91:13–31 consistent method for the analysis of protein
CD Spectroscopy 265
secondary structure from circular dichroism. 41. Honda S, Kobayashi N, Munekata E, Uedaira H
Anal Biochem 209:32–44 (1999) Fragment reconstitution of a small pro-
31. Sreerama N, Woody RW (2000) Estimation of tein: folding energetics of the reconstituted
protein secondary structure from CD spectra: immunoglobulin binding domain B1 of strepto-
comparison of CONTIN, SELCON and coccal protein G. Biochemistry 38:1203–1213
CDSSTR methods with an expanded reference 42. Santoro MM, Bolen DW (1988) Unfolding free
set. Anal Biochem 282:252–260 energy changes determined by the linear extrap-
32. Johnson WC (1999) Analyzing protein circular olation method. 1. Unfolding of phenylmetha-
dichroism spectra for accurate secondary struc- nesulfonyl alpha-chymotrypsin using different
tures. Proteins 35:307–312 denaturants. Biochemistry 27:8063–8068
33. Bohm G, Muhr R, Jaenicke R (1992) Quanti- 43. Marquardt DW (1963) An algorithm for the
tative analysis of protein far UV circular dichro- estimation of non-linear parameters. J Soc
ism spectra by neural networks. Protein Eng Indust Appl Math 11:431–441
5:191–195 44. Holmgren A, Soderberg BO, Eklund H, Bran-
34. Andrade MA, Chacon P, Merelo JJ, Moran F den CI (1975) Three-dimensional structure of
(1993) Evaluation of secondary structure of Escherichia coli thioredoxin-S2 to 2.8 A reso-
proteins from UV circular dichroism spectra lution. Proc Natl Acad Sci U S A
using an unsupervised learning neural network. 72:2305–2309
Protein Eng 6:383–390 45. Jeng MF, Campbell AP, Begley T, Holmgren
35. Perczel A, Hollosi M, Tusnady G, Fasman GD A, Case DA, Wright PE, Dyson HJ (1994)
(1991) Convex constraint analysis: a natural High-resolution solution structures of oxi-
deconvolution of circular dichroism curves of dized and reduced Escherichia coli thiore-
proteins. Protein Eng 4:669–679 doxin. Structure 2:853–868
36. Georgescu RE, Braswell EH, Zhu D, Tasayco 46. Koradi R, Billeter M, W€ uthrich K (1996)
ML (1999) Energetics of assembling an artifi- MOLMOL: a program for display and analysis
cial heterodimer with an alpha/beta motif: of macromolecular structures. J Mol Graph 14
cleaved versus uncleaved Escherichia coli thior- (51–5):29–32
edoxin. Biochemistry 38:13355–13366 47. Deléage G, Geourjon C (1993) An interactive
37. Katti SK, LeMaster DM, Eklund H (1990) graphic program for calculating the secondary
Crystal structure of thioredoxin from Escher- structure content of proteins from circular
ichia coli at 1.68 A resolution. J Mol Biol dichroism spectrum. Comput Appl Biosci
212:167–184 9:197–199
38. Scatchard G (1949) The attractions of proteins 48. Unneberg P, Merelo JJ, Chacón P, Morán F,
for small molecules and ions. Ann NY Acad Sci SOMCD (2001) Method for evaluating pro-
51:660–672 tein secondary structure from UV circular
39. Hill AV (1910) The possible effects of the dichroism spectra. Proteins 42:460–470
aggregation of the molecules of haemoglobin 49. Louis-Jeune C, Andrade MA, Perez-Iratxetal C
on its dissociation curves. J Physiol (Lond) 40: (2012) Prediction of protein secondary struc-
iv–vii ture from circular dichroism using theoretically
40. Engel G (1974) Estimation of binding para- derived spectra. Proteins 80:374–381
meters of enzyme-ligand complex from fluoro- 50. Wiedemann C, Bellstedt P, Görlach M (2013)
metric data by a curve fitting procedure: seryl- CAPITO – a web server-based analysis and
tRNA synthetase-tRNA Ser complex. Anal Bio- plotting tool for circular dichroism data. Bioin-
chem 61:184–191 formatics 29:1750–1757
Chapter 16
Abstract
Nuclear magnetic resonance (NMR) has continued to evolve as a powerful method, with an increase in the
number of pulse sequences and techniques available to study protein-protein interactions. In this chapter, a
straightforward method to map a protein–protein interface and design a structural model is described,
using chemical shift perturbation, paramagnetic relaxation enhancement, and data-driven docking.
Key words Nuclear magnetic resonance (NMR), Protein-protein interaction, Chemical-shift pertur-
bation (CSP), Paramagnetic relaxation enhancement (PRE), Docking
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_16, © Springer Science+Business Media New York 2015
267
268 Peter M. Thompson et al.
<100 μM), or weak binding (Kd > 100 μM), assuming diffusion-
limited association rates. Therefore, CSP is most useful when the
binding interaction is either in slow or fast exchange.
PRE is another NMR technique that involves changing the
chemical environment of nuclei involved in an interaction. Para-
magnetic ions have a free, unpaired electron and therefore a net
spin, which generates a magnetic field [11]. When the paramag-
netic ion is allowed to rotate freely (isotropically), it exposes nearby
nuclei to a constantly changing magnetic field. This increases the
rate of relaxation of the signal from the nucleus and is observed as a
decreased intensity of the signal and a significantly increased line-
width. This relaxation enhancement is related to the distance
between the ion and the magnetically susceptible nucleus by a
factor of 1/r6, where r is the distance between the two atoms.
Thus, if a paramagnetic ion is attached to a ligand, then nuclei in
the protein near the ligand-binding site should have increased
relaxation rates. More complex experiments can be run to measure
the rate of relaxation and determine the distance from the paramag-
netic ion, but it is more common to use PRE to identify nuclei with
decreased signal intensity and define them as being close to the
paramagnetic ion when the ligand is bound [12].
The final method we discuss is NMR data-driven docking,
which couples computational methods with limited NMR
restraints. When NMR restraints are too limited for structural
determination or a high-resolution structure is not critical, docking
procedures can be used to develop a testable ligand-binding model.
There are many available docking programs, each with its own
distinct methods for performing docking and scoring its structures
[13–18]. We used the program HADDOCK, which is designed to
allow for straightforward input of restraints from NMR data, as well
as other methods [13]. HADDOCK refers to these inputs as
Ambiguous Interaction Restraints (AIRs) and relies on them to
generate docked structures from the structures of the individual
binding partners. The structures are then scored, with those scor-
ing the highest being selected for semi-flexible simulated annealing.
Next, a four-step, iterative annealing process that allows for the
movement of amino acid residues at the interaction interface is
performed. Finally, the structures are refined in explicit solvent
(water), clustered into groups, and scored. The scoring function
is based upon physical parameters and experimental restraints, and
can be customized for each project.
In this chapter, we show how a combination of CSP, PRE, and
docking were used to generate a model for an interaction between
an EF-hand domain of α-actinin-2 and a short region of the protein
palladin [19]. These two proteins are involved in regulation of the
actin cytoskeleton and have been implicated in cancer. A structural
model of the interaction between the two provides a starting point
for understanding the functional role of the interaction. While
270 Peter M. Thompson et al.
2 Materials
15
1. N-enriched human α-actinin 2 EF-hand residues 823–894
(referred to as Act-EF34, numbered 1–73 herein).
15
2. NH4Cl.
3. Unlabeled palladin peptide (residues 235–252 of human palla-
din, numbered 1–19 herein).
4. Palladin peptide with an N-terminal cysteine labeled with
S-(2,2,5,5-tetramethyl-2,5-dihydro-1H-pyrrol-3-yl)methyl
methanesulfonothioate (MTSL).
5. Varian Inova 600 or 700 MHz NMR spectrometer.
6. 5.0 mm Wilmad standard NMR sample tube.
7. NMRPipe/NMRDraw (Delaglio, NIH).
8. NMRViewJ (One Moon Scientific, Inc.).
9. NMR Buffer: 20 mM 3-(N-morpholino)propanesulfonic acid,
pH 6.6, 10 mM NaCl, 2 mM tris(2-carboxyethyl)phosphine,
0.01 % NaN3, and 10 % D2O.
10. 1 M ascorbic acid.
3 Methods
3.3 Chemical Shift Proton and 15N resonance assignments of apo Act-EF34 were
Perturbation obtained from the Biological Magnetic Resonance Bank (BMRB),
entry 17627. Two-dimensional 1H–15N HSQC data were acquired
on the Act-EF34:palladin peptide (see Note 4) complex at molar
ratios of 1:0, 1:0.1, 1:0.5, 1:1, 1:2, 1:3, 1:4, and 1:5, with 220 μM
Act-EF34. At a ratio of 1:5, complete saturation of binding was
achieved as peaks no longer shifted. To prevent dilution effects,
samples were made by mixing the 1:0 and 1:5 samples in the
appropriate ratio. The Kd of the interaction, determined by iso-
thermal titration calorimetry (Part II Chapter 11) to be ~16 μM,
suggested that exchange between the free and bound state would
likely be intermediate or fast on the NMR time scale (see Note 5).
In this system, we see exchange on the fast time scale, which results
in a single peak for each nucleus with a chemical shift that is the
average of the weighted population of the free- and bound-state
chemical shifts.
3.4 Paramagnetic An NMR sample with 220 μM Act-EF34 and 1.1 mM MTSL-
Relaxation labeled palladin peptide was created with a final volume of
600 μL. MTSL-labeled peptide was generated by incubation of
MTSL and the palladin peptide with an N-terminal cysteine with
an MTSL:peptide ratio of 4:1 in a nonreducing buffer [19]. This
allows for formation of a disulfide bond between the MTSL label
and the thiol group on the cysteine. Then excess, unbound MTSL
label was removed by extensive dialysis into nonreducing NMR
buffer. A 2D 1H–15N HSQC was acquired on the sample, using
the same parameters as those given above. Then 2 μL of 1 M
ascorbic acid was added to the sample to reduce the MTSL label,
allowing it to diffuse into the solution, and a duplicate 1H–15N
HSQC was acquired on this sample.
3.5 NMR Data Both the chemical shift perturbation and paramagnetic relaxation
Analysis studies relied on changes in the chemical shifts or peak intensities,
respectively, as observed by 2D 1H–15N HSQC analyses. Addition-
ally, in both experiments, 15N-labeled Act-EF34 was used, meaning
that the signals observed come only from Act-EF34. The 2D
1
H–15N HSQC provides information on all 15N atoms bound to
a 1H. This includes one NH peak for the backbone amide, which
we expect for all residues except proline, giving a site-specific probe
for each residue. Side-chain amides also provide peaks, though their
assignment is more difficult. The signal from the proton is in the
first dimension, while the signal from the 15N nucleus is in the
second. The peaks in the resulting spectrum appear as spots with
contour lines to represent their height or intensity. Both the
272 Peter M. Thompson et al.
Fig. 2 Structural model of Act-EF34 complexed with WT palladin peptide. (a) CSP and PRE data mapped onto
the primary structure of Act-EF34. CSP data are shown in black, with PRE data in gray. The secondary
structure (from PDB 1H8B) of Act-EF34 is given by black bars (α-helices) and white bars (β-sheets).
Figure modified from Beck et al. [19] (b) Model structure of Act-EF34 when bound to WT palladin peptide
as created by PyMOL [28]. Sidechains shown as sticks belong to residues selected as AIRs for docking with
HADDOCK. Residues colored in pink showed significant relaxation when bound to the MTSL-labeled palladin
peptide. (c) Model of the ligand-bound complex. The color scheme is maintained, and the palladin peptide is
shown in blue. The sidechains of L9 and L12 are represented as sticks. The N-terminus (where the MTSL label
was added) is shown as spheres
3.6 Docking The docking program HADDOCK [13] was used to develop a
model for the interaction between the palladin peptide and Act-
EF34. HADDOCK starts with known structures of the two ligands
and docks using experimental restraints and a traditional energy
function; it does allow for small conformational changes, a com-
mon occurrence in protein-protein interactions. The starting struc-
ture for Act-EF34 was taken from the solution structure of
Mapping Protein-Protein Interactions by NMR 275
4 Notes
References
1. Akke M (2002) NMR methods for characteriz- approach based on biochemical or biophysical
ing microsecond to millisecond dynamics in information. J Am Chem Soc 125:1731–1737
recognition and catalysis. Curr Opin Struct 14. Lyskov S, Gray JJ (2008) The RosettaDock
Biol 12:642–647 server for local protein–protein docking.
2. Clore G, Gronenborn A (1991) Two-, three-, Nucleic Acids Res 36:W233–W238
and four-dimensional NMR methods for 15. Claussen H, Buning C, Rarey M et al (2001)
obtaining larger and more precise three- FlexE: efficient molecular docking considering
dimensional structures of proteins in solution. protein structure variations. J Mol Biol 308:
Annu Rev Biophys Biophys Chem 20:29–63 377–395
3. Kay LE (1997) NMR methods for the study of 16. Tovchigrechko A, Vakser IA (2006) GRAMM-
protein structure and dynamics. Biochem Cell X public web server for protein–protein dock-
Biol 75:1–15 ing. Nucleic Acids Res 34:W310–W314
4. Vinogradova O, Qin J (2012) NMR as a 17. Pierce BG, Hourai Y, Weng Z (2011) Acceler-
unique tool in assessment and complex deter- ating protein docking in ZDOCK using an
mination of weak protein–protein interactions. advanced 3D convolution library. PLoS One
Top Curr Chem 326:35–45 6:e24657
5. O’Connell MR, Gamsjaeger R, Mackay JP 18. Morris GM, Huey R, Lindstrom W et al (2009)
(2009) The structural analysis of protein–pro- AutoDock4 and AutoDockTools4: automated
tein interactions by NMR spectroscopy. Prote- docking with selective receptor flexibility.
omics 9:5224–5232 J Comput Chem 30:2785–2791
6. Jensen MR, Ortega-Roldan JL, Salmon L et al 19. Beck MR, Otey CA, Campbell SL (2011)
(2011) Characterizing weak protein–protein Structural characterization of the interactions
complexes by NMR residual dipolar couplings. between palladin and alpha-actinin. J Mol Biol
Eur Biophys J 40:1371–1381 413:712–725
7. Marintchev A, Frueh D, Wagner G (2007) 20. Joseph C, Stier G, O’Brien R et al (2001) A
NMR methods for studying protein–protein structural characterization of the interactions
interactions involved in translation initiation. between titin Z-repeats and the alpha-actinin C-
Methods Enzymol 430:283–331 terminal domain. Biochemistry 40:4957–4965
8. Wittekind M, Mueller L (1993) Hncacb, a high- 21. Bodenhausen G, Ruben DJ (1980) Natural
sensitivity 3d Nmr experiment to correlate abundance N-15 Nmr by enhanced heteronuc-
amide-proton and nitrogen resonances with lear spectroscopy. Chem Phys Lett 69:185–189
the alpha-carbon and beta-carbon resonances 22. Delaglio F, Grzesiek S, Vuister GW et al (1995)
in proteins. J Magn Reson Ser B 101:201–205 NMRPipe: a multidimensional spectral proces-
9. Ikura M, Kay LE, Bax A (1990) A novel- sing system based on UNIX pipes. J Biomol
approach for sequential assignment of H-1, NMR 6:277–293
C-13, and N-15 spectra of larger proteins – 23. Hubbard SJ, Thornton JM (1993) NACCESS
heteronuclear triple-resonance 3-dimensional Computer Program, Department of Biochem-
Nmr-spectroscopy – application to calmodulin. istry and Molecular Biology, University
Biochemistry 29:4659–4667 College London www.bionf.manchester.ac.
10. Grzesiek S, Bax A (1992) Correlating back- uk/naccess/nac_intro.html
bone amide and side-chain resonances in larger 24. de Vries SJ, van Dijk M, Bonvin AM (2010)
proteins by multiple relayed triple resonance The HADDOCK web server for data-driven
NMR. J Am Chem Soc 114:6291–6293 biomolecular docking. Nat Protoc 5:883–897
11. Solomon I (1955) Relaxation processes in a 25. Venters RA, Calderone TL, Spicer LD et al
system of two spins. Phys Rev 99:559–565 (1991) Uniform 13C isotope labeling of pro-
12. Jahnke W (2002) Spin labels as a tool to teins with sodium acetate for NMR studies:
identify and characterize protein-ligand inter- application to human carbonic anhydrase II.
actions by NMR spectroscopy. Chembiochem Biochemistry 30:4491–4494
3:167–173 26. Kuipers BJ, Gruppen H (2007) Prediction
13. Dominguez C, Boelens R, Bonvin AM (2003) of molar extinction coefficients of proteins
HADDOCK: a protein–protein docking and peptides using UV absorption of the
Mapping Protein-Protein Interactions by NMR 279
constituent amino acids at 214 nm to enable of related native and engineered high-alkaline
quantitative reverse phase high-performance Bacillus subtilisins. J Mol Biol 292:111–123
liquid chromatography-mass spectrometry 28. DeLano WL (2006) MacPyMOL: The
analysis. J Agric Food Chem 55:5445–5451 PyMOL Molecular Graphics System, Version
27. Mulder FA, Schipper D, Bott R et al (1999) 1.5.0.4 Schrödinger, LLC www.pymol.org
Altered flexibility in the substrate-binding site
Chapter 17
Abstract
Mass spectrometry is one of the most sensitive methods in analytical chemistry, and its application in
proteomics has been rapidly expanded after sequencing the human genome. Mass spectrometry is now the
mainstream approach for identification and quantification of proteins and posttranslational modifications,
either in small scale or in the entire proteome. Shotgun proteomics can analyze up to 10,000 proteins in a
comprehensive study, with detection sensitivity in the picogram range. In this chapter, we describe major
experimental steps in a shotgun proteomics platform, including sample preparation in the context of
studying protein-protein interaction, mass spectrometric data acquisition, and database search to identify
proteins and posttranslational modification analysis. Proteome quantification strategies and bioinformatics
analysis are also illustrated. Finally, we discuss the capabilities, limitations, and potential improvements of
current platforms.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_17, © Springer Science+Business Media New York 2015
281
282 Vishwajeeth R. Pagala et al.
Triple quadrupole Linear ion trap Q-TOF LTQ orbitrap (ELITE) Q exactive
Accuracy 0.01 % (100 ppm) 0.01 % (100 ppm) 0.001 % (10 ppm) <2 ppm <2 ppm
Resolution 4,000 4,000 10,000 240,000 140,000
m/z Range 4,000 4,000 10,000 6,000 4,000
Scan speed (s) ~1 ~1 ~1 ~1 ~0.5
Comments Good accuracy, Good accuracy, Excellent accuracy, Excellent accuracy, resolution, Excellent accuracy, resolution,
good resolution, good resolution, good resolution, and sensitivity; flexible and sensitivity; only HCD
low-energy low-energy low-energy collisions, fragmentation (e.g., CID, available; bench top and
collisions, low cost collisions, low cost high sensitivity HCD, and ETD) easy to use
Protein Analysis by MS 285
2 Materials
3 Methods
3.1 Sample Digestion Protein samples are usually separated by sodium dodecyl sulfate
and LC-MS/MS (SDS) polyacrylamide gel electrophoresis followed by protein stain-
Analysis for Protein ing with Coomassie blue or SYPRO Ruby (see Note 7). The SDS
Identification gel not only resolves proteins based on protein size, but also
removes contaminants that interfere with mass spectrometry (e.g.,
3.1.1 Protein In-Gel salt and detergents, Fig. 1). Total protein level of the samples may
Digestion be also evaluated according to staining intensity if known amount
of bovine serum albumin is titrated on the same gel (e.g., 10, 100,
1,000 ng). As most protein quantification kits need microgram
Protein Analysis by MS 287
3.1.2 Protein In-Solution If protein samples contain no detergents, the samples may be
Digestion directly digested in solution to avoid the lengthy in-gel digestion
protocol. In-solution digestion efficiency might be lower than in-
gel digestion, which may be due to residual protein folding that
reduces trypsin accessibility. Thus, a two-step digestion protocol is
Protein Analysis by MS 289
3.1.3 Standard LC-MS/ Successful LC-MS/MS analysis relies on a robust and reproducible
MS Analysis LC-MS/MS system, including a nanoscale HPLC system with
autosampler, and an inline modern tandem mass spectrometer.
Peptide samples are loaded on a reverse-phase C18 column by the
HPLC system and eluted by a gradient. Eluted peptides are ionized
and detected by the mass spectrometer (e.g., Thermo Q Exactive or
LTQ Orbitrap Elite). MS spectra are collected first (in ~0.5 s), and
the top 20 abundant ions are sequentially isolated for MS/MS
analysis (each in ~0.1 s, totaling ~2 s). This process (~2.5 s) is
cycled over the entire liquid chromatography gradient, and more
290 Vishwajeeth R. Pagala et al.
3.1.4 Analysis of Protein Approximately 300 forms of protein modifications are documen-
Posttranslational ted. In cells, the modifications range from the simple addition of
Modifications (PTM) small chemical groups (e.g., phosphorylation, methylation, and
acetylation), to more complex moieties (e.g., glycosylation, lipida-
tion, and ubiquitination). The modifications are reversible and
catalyzed by an array of enzymes, leading to regulation of protein
surfaces, structures, and interacting partners. Thus, protein mod-
ifications are central to signal transduction in cells.
Mapping modified residues by mass spectrometry is often chal-
lenging, which is highly different from protein identification analy-
sis that requires one unique peptide (matched to a single MS/MS
spectrum). For PTM mapping, approximately 100 % “sequence
coverage” by MS/MS is needed. As some digested peptides (with
extreme lengths or hydrophobicity) may not be compatible with
the standard LC-MS/MS method, custom methods may be
required, such as digestions with additional enzymes (e.g., Arg-C,
Glu-C, Asp-N) to improve peptide coverage. In addition, the stoi-
chiometry of PTM may be low and the modified sites may be
Protein Analysis by MS 291
3.2.2 Protein Profiling The SILAC approach can accurately compare the relative intensities
by SILAC of peptides from protein samples. First, cells are grown in medium
containing either light or heavy stable isotope labeled Lys and Arg.
Then the differentially labeled cells are treated under different
conditions, such as transfection with mock or a protein-expressing
plasmid in a co-IP experiment. The cells are harvested and lysed for
immunoprecipitation. Finally, the heavy and light IP samples are
pooled together for SDS PAGE and LC-MS/MS (Fig. 2, see Note
15). We continue to use the co-IP experiment to explain details in a
SILAC analysis.
1. Select appropriate cell lines and culture in light or heavy
medium to ensure full labeling.
2. Examine for cell viability in SILAC medium, as certain cell lines
are not able to thrive in SILAC medium.
3. Examine if the use of SILAC medium alters the biology of the
cells (e.g., expression of protein of interest).
4. Test for complete labeling of proteins. Certain cell types may
require extra passages to achieve full labeling of all proteins. It is
necessary to achieve >95 % labeling for accurate quantification.
Protein Analysis by MS 293
Fig. 2 Protein profiling by the SILAC strategy. Protein samples are differentially
labeled by light and heavy SILAC media. The two samples are harvested and
subjected to immunoprecipitation. The IP samples are then pooled together,
subjected to gel separation, in gel digestion and reverse phase LC-MS/MS
analysis. Biological replicates can be performed using a forward and reverse
labeling strategy
3.2.3 Protein Profiling The development of multiplex isobaric labeling methods (e.g.,
by iTRAQ/TMT iTRAQ and TMT) greatly improves the throughput of proteomics.
In this protocol, we describe how to perform a 6-plex TMT analysis
(Fig. 3). Every labeling reagent consists of three groups: an amine-
specific active ester group, a balance group, and a reporter group.
Six reagents can label six different samples by reacting with free
294 Vishwajeeth R. Pagala et al.
Fig. 3 Protein profiling by the TMT technology. Six samples can be differentially
labeled by 6-plex TMT reagents for a quantitative analysis. The samples are
first digested separately, labeled by TMT reagents, followed by pooling and
LC-MS/MS analysis. If necessary, two dimensional LC may be implemented,
including strong cation exchange (SCX) chromatography and reverse phase
chromatography
low quality spectra to save time and reduce risk of false identifica-
tions, post-processing filters database search results to maintain the
false discovery rate at a tolerable level (e.g., 1 %).
Preprocessing 1. Extract MS/MS data from raw files. Mass spectrometers from
different companies generate raw data in different formats. For
instance, Thermo MS instruments produce files in raw format.
The raw files are first converted to mzXML format with
“ReAdW”, then to dta files by “MzXML2Search”. Both pro-
grams are available from the Institute for Systems Biology
Protein Analysis by MS 297
Protein Profiling by Spectral Spectral count of a given protein is the summed number of its
Counting (SC) matching MS/MS spectra, similar to the number of reads in RNA-
seq analysis. Since peptide ions are selected for MS/MS based on
the rank of ion intensity, the spectral count of an identified protein
is correlated with its abundance. Some pitfalls are that the spectral
counting method is influenced by LC-MS/MS run-to-run varia-
tion, and small SC number (e.g., less than 5) markedly decreases
the reliability of quantitative comparison.
1. Acquire spectral counts for proteins assigned by database
search.
2. Normalize spectral counts to assume that the average SC per
protein should be the same in all samples. This assumption
works only when protein composition and concentration
among samples are highly similar. For IP samples, this assump-
tion may not apply well if the control and IP samples are vastly
different.
3. Use a G-test to determine statistical significance (p-value) of
protein abundance difference (see Note 19)
4. Calculate FDR based on null experiments (e.g., comparison
between biological replicates) and set up a threshold for filter-
ing the dataset.
Protein Analysis by MS 299
Protein Profiling by SILAC The SILAC method offers many benefits compared to label-free
approaches. It can detect relatively small changes in protein expres-
sion levels or posttranslational modifications during comparison.
Whereas SILAC quantification may be carried out prior to protein
identification, as in the MaxQuant program [33], we usually derive
SILAC data after identifying proteins [34].
1. Extract ion currents in MS scans for identified peptides. Define
peak intensity, area intensity, and single-to-noise (S/N) ratio.
2. Match light and heavy isotope labeled peaks using predicted
m/z difference and a defined mass tolerance (e.g., 6 ppm for
high resolution MS data).
3. Compare peak intensity of matched peptides to obtain abun-
dance ratios of the peptides/proteins, and convert to log2
(ratio).
4. Summarize peptide ratios into protein ratios. Remove outliers
by Dixon’s Q test. The S/N ratio of a protein is represented by
that of the most abundant peptide ion.
5. Fit the data to Gaussian distribution to obtain mean and stan-
dard deviation (SD). If the vast majority of proteins are not
expected to change, the mean can be used to correct for sys-
tematic bias (e.g., experimental loading bias).
6. Select proteins with significantly changes in expression by the
cutoff of log2(ratio) that is outside of a 95 % confidence inter-
val (~2 SD) of the Gaussian distribution. FDR is evaluated by
null experiments of biological replicates. We also notice that
increasing the protein S/N cutoff leads to less false positives.
Protein Profiling Current isobaric mass tags (e.g., iTRAQ and TMT) provide simul-
by iTRAQ/TMT taneously analysis of 4, 6, 8, or 10 biological samples. Quantitative
information is extracted from report ions detected in MS/MS
spectra.
1. Acquire ion intensity of reporter ions in MS/MS scans for all
matched peptides, and define the S/N ratio of the ions.
2. For every binary comparison, compare the peak intensity of
reporter ions to obtain abundance ratios, and convert to log2
(ratio). The S/N ratio of a protein is represented by that of the
most abundant peptide ion.
3. The remaining protocol is the same as that in the SILAC
analysis.
3.3.3 Validation of MS Theoretically, a computer program could match almost any MS/
Results MS spectrum to a putative peptide sequence. Post-processing is
crucial to reduce false positives. The widely used target-decoy
strategy provides a general estimation of the FDR of an entire
300 Vishwajeeth R. Pagala et al.
4 Notes
Acknowledgements
References
1. Cravatt BF, Simon GM, Yates JR 3rd (2007) 16. Picotti P, Aebersold R (2012) Selected reaction
The biological impact of mass-spectrometry- monitoring-based proteomics: workflows,
based proteomics. Nature 450:991–1000 potential, pitfalls and future directions. Nat
2. Choudhary C, Mann M (2010) Decoding sig- Methods 9:555–566
nalling networks by mass spectrometry-based 17. Aebersold R, Mann M (2003) Mass
proteomics. Nat Rev Mol Cell Biol spectrometry-based proteomics. Nature
11:427–439 422:198–207
3. Gstaiger M, Aebersold R (2009) Applying 18. Liu H, Sadygov RG, Yates JR 3rd (2004) A
mass spectrometry-based proteomics to genet- model for random sampling and estimation of
ics, genomics and network biology. Nat Rev relative protein abundance in shotgun proteo-
Genet 10:617–627 mics. Anal Chem 76:4193–4201
4. Mann M, Kulak NA, Nagaraj N et al (2013) 19. Zhou JY, Afjehi-Sadat L, Asress S et al (2010)
The coming age of complete, accurate, and Galectin-3 is a candidate biomarker for amyo-
ubiquitous proteomes. Mol Cell 49:583–590 trophic lateral sclerosis: discovery by a proteo-
5. Chien CT, Bartel PL, Sternglanz R et al (1991) mics approach. J Proteome Res 9:5133–5141
The two-hybrid system: a method to identify 20. Wang W, Zhou H, Lin H et al (2003) Quanti-
and clone genes for proteins that interact with a fication of proteins and metabolites by mass
protein of interest. Proc Natl Acad Sci U S A spectrometry without isotopic labeling or
88:9578–9582 spiked standards. Anal Chem 75:4818–4826
6. Behrends C, Sowa ME, Gygi SP et al (2010) 21. Ross PL, Huang YN, Marchese JN et al (2004)
Network organization of the human autophagy Multiplexed protein quantitation in Saccharo-
system. Nature 466:68–76 myces cerevisiae using amine-reactive isobaric
7. Vidal M, Cusick ME, Barabasi AL (2011) tagging reagents. Mol Cell Proteomics
Interactome networks and human disease. 3:1154–1169
Cell 144:986–998 22. Ong SE, Blagoev B, Kratchmarova I et al
8. Havugimana PC, Hart GT, Nepusz T et al (2002) Stable isotope labeling by amino acids
(2012) A census of human soluble protein in cell culture, SILAC, as a simple and accurate
complexes. Cell 150:1068–1081 approach to expression proteomics. Mol Cell
9. Eng JK, Searle BC, Clauser KR et al (2011) A Proteomics 1:376–386
face in the crowd: recognizing peptides 23. Dephoure N, Gygi SP (2012) Hyperplexing: a
through database search. Mol Cell Proteomics method for higher-order multiplexed quantita-
10(R111):009522 tive proteomics provides a map of the dynamic
10. Nesvizhskii AI, Vitek O, Aebersold R (2007) response to rapamycin in yeast. Sci Signal 5:rs2
Analysis and validation of proteomic data gen- 24. Eng J, McCormack AL, Yates JR 3rd (1994)
erated by tandem mass spectrometry. Nat An approach to correlate tandem mass spectral
Methods 4:787–797 data of peptides with amino acid sequences in a
11. Peng J, Gygi SP (2001) Proteomics: the move protein database. J Am Soc Mass Spectrom
to mixtures. J Mass Spectrom 36:1083–1091 5:976–989
12. Xie F, Smith RD, Shen Y (2012) Advanced 25. Perkins DN, Pappin DJ, Creasy DM et al
proteomic liquid chromatography. J Chroma- (1999) Probability-based protein identification
togr A 1261:78–90 by searching sequence databases using mass
spectrometry data. Electrophoresis
13. Duncan MW, Aebersold R, Caprioli RM 20:3551–3567
(2010) The pros and cons of peptide-centric
proteomics. Nat Biotechnol 28:659–664 26. Xu P, Duong DM, Peng J (2009) Systematical
optimization of reverse-phase chromatography
14. Michalski A, Cox J, Mann M (2011) More than for shotgun proteomics. J Proteome Res
100,000 detectable peptide species elute in sin- 8:3944–3950
gle shotgun proteomics runs but the majority is
inaccessible to data-dependent LC-MS/MS. J 27. Shevchenko A, Wilm M, Vorm O et al (1996)
Proteome Res 10:1785–1793 Mass spectrometric sequencing of proteins
silver-stained polyacrylamide gels. Anal Chem
15. Gerber SA, Rush J, Stemman O et al (2003) 68:850–858
Absolute quantification of proteins and
phosphoproteins from cell lysates by 28. Ficarro SB, McCleland ML, Stukenberg PT
tandem MS. Proc Natl Acad Sci U S A et al (2002) Phosphoproteome analysis by
100:6940–6945 mass spectrometry and its application to
Protein Analysis by MS 305
Abstract
Evaluating sites of protein-protein interactions can be an arduous task involving extensive mutagenesis
work and attempts to express and purify individual proteins in sufficient quantities. Peptide mapping is a
useful alternative to traditional methods as it allows rapid detection of regions and/or individual residues
important for binding, and it can be readily applied to numerous proteins at once. Here we describe the use
of the ResPep SL SPOT method to evaluate protein–protein binding interactions such as that between G-
protein βγ subunits and SNARE proteins, identifying both regions of interest and subsequently individual
residues which can then be manipulated in further biochemical assays to confirm their validity.
Key words ResPep SL, Fmoc chemistry, Peptide, Protein-protein interaction, Alanine screening
1 Introduction
1.1 History/Theory Automated peptide arrays such as that performed by the ResPep SL
enable protein-protein interactions to be easily studied. Using the
SPOT method, overlapping linear peptides are simultaneously
synthesized on cellulose membranes and used to predict the
biological activity of a protein. The SPOT method was first
described by Ronald Frank in 1992 [1] as a method for in situ
peptide synthesis. It creates short, immobilized peptides on a glass
plate or cellulose membrane, which are easy to handle and retain
some of the biological characteristics derived from the primary
sequence [2]. Cellulose membranes have become the preferred
medium for these arrays as the membranes are inexpensive, stable
in aqueous solutions, nontoxic to biological samples, and able to
withstand organic solvents and acids [3]. Further, for a given
membrane, the SPOT method allows the parallel synthesis of up
to 600 different peptides in a highly reliable manner. This is done
using Fmoc-chemistry whereby the amino group of each amino
acid is temporarily protected by a 9-fluorenylmethoxycarbonyl
group (Fmoc) to prevent polymerization and allow the addition
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_18, © Springer Science+Business Media New York 2015
307
308 Yun Young Yim et al.
1.2 The Applications Peptides arrays generated using the SPOT method have wide
of Peptide Arrays biological applications, including evaluating the actions of
enzymes, cell adhesion, and the binding of metals [2] but are
particularly useful for identifying sites of protein-protein inter-
actions and the development of inhibitors or activators of enzymes
[6]. To understand protein-protein interactions, investigators often
use co-immunoprecipitation, pull-down, or two hybrid assays, as
well as in vivo Förster resonance energy transfer (FRET) [7]. While
such assays can identify the presence of protein-protein interac-
tions, they cannot determine the interaction sites without conjunc-
tion to other methods such as X-ray crystallography. At present, X-
ray crystallography is widely used to evaluate sites of protein-
protein interaction; however, this process is both challenging and
time consuming, particularly with proteins that are difficult to
express, crystallize, or obtain diffraction. In such instances, peptide
arrays offer an easy alternative as they can rapidly highlight regions
of residues which may be involved in binding without the challenge
of expressing and crystallizing full-length proteins. While caution
must be taken as this method does not use full-length expressed
proteins, it offers a method to determine regions of interaction
which can then be followed up with alanine screening and other
biochemical methods to test identified residues in the context of the
full-length proteins for their functional role in the interaction. As
an example, Wells et al. [8] recently described this method to
characterize the interaction sites between G-protein βγ subunits
and a member of the SNARE complex, SNAP25. Following initial
peptide screens, they determined which specific amino acid residues
were important using Ala screening, before demonstrating that
those residues were important in the context of the whole protein
using biochemical and functional assays. Comparatively, peptide
Peptide Arrays 309
arrays generated using the SPOT method can also be used in a high
throughput manner to develop enzyme inhibitors, as well as active
substrates for various enzymes. As Thiele et al. shows, by generat-
ing peptide libraries on a glass plate or cellulose membrane, the
peptide-enzyme activity, such as phosphorylation of peptide, can be
detected using autoradiography, chemiluminescence, or enzymatic
color development [6]. By monitoring the phosphorylation of
peptides by a particular enzyme, researchers can select lead peptides
which have the potential to be developed as enzyme inhibitors or
activators. This is exemplified in Mukhija et al. [9], where the
authors successfully used this method to identify peptides inhibit-
ing enzyme I (EI) of the bacterial phosphotransferase system
(PTS). Using immobilized combinatorial peptide libraries and
phosphorimaging, they identified heptapeptides and octapeptides
which selectively inhibit EI in vitro. Similarly, Collet et al. [10] has
screened potential substrates of proteases by synthesizing peptides
on a fluorous-coated glass. For the first time, the authors describe
the use of fluorous to probe peptide sequences in protease screen-
ing. With the change in material where the peptide is immobilized
and the detection methods [5], the peptide array by SPOT method
was used as a screening method for the development of inhibitors
or activators of enzymes. Thus, peptide arrays generated by the
SPOT method have wide biological implications including signifi-
cantly enhancing protein-protein interaction studies, as well as
aiding in the development of reagents such as inhibitors and active
substrates of enzymes.
In this chapter, we demonstrate how to use peptide arrays
created by the SPOT method to understand protein-protein inter-
actions. We first describe how to prepare all the reagents and
program software for the SPOT method. With appropriate reagents
and correctly programmed software, peptides are synthesized on
cellulose membrane. Then, we show how to de-protect side chain
and use far-western technique to identify peptides involved in
protein-protein interaction. Lastly, we describe how to identify
amino acids involved in protein-protein interaction by alanine
screen.
2 Materials
2.2 Peptide Array 1. Amino acid derivatives: Remove amino acids derivatives from
Synthesis the 20 C freezer and thaw until they reach room tempera-
ture (approximately 2 h). Weigh out the appropriate amount of
each amino acid derivative into 2 ml microcentrifuge tubes and
dissolve in N-methyl pyrrolidone (NMP) using an oscillating
shaker (see Note 2).
2. 2 ml ResPep tubes with caps.
3. Membrane preparation: Pre-swell cellulose membrane from
INTAVIS in 30 ml of DMF in a black container covered with
a lid for 1 h. Agitate gently every 15 min.
4. 1.1 M hydroxybenzotriazole (HOBTlH2O): Weigh out the
appropriate amount of HOBTlH2O into a 30 ml conical tube
and dissolve in DMF to produce a 1.1 M solution (see Note 3).
5. 1.1 M diisopropylcarbodiimide (DIC): Using a glass pipette,
dilute the appropriate volume of DIC with DMF in a 50 ml
conical tube to produce a 1.1 M solution (see Note 3).
6. 20 % piperidine: In a 250 ml plastic bottle, add the appropriate
volume of piperidine to DMF to produce a 20 % solution (see
Note 3).
7. 5 % acetic anhydride: Using a glass pipette, transfer the appro-
priate volume of acetic anhydride to a 50 ml conical tube and
mix with DMF to produce a 5 % solution (see Note 3).
2.5 Alanine Screen 1. This process requires the same materials listed in
Subheadings 2.1–2.4.
3 Methods
3.1 SPOT Method 1. Turn on Auto SPOT robot such as ResPep SL and vacuum
Preparation (Fig. 1) pump by pressing the green ON/OFF switch on the side of the
AUTO spot robot.
2. Start the spotter software such as ResPep SL Spotter software
on the computer (see Note 8).
3. Change waste bottles as needed.
312 Yun Young Yim et al.
Fig. 1 ResPep SL and membrane design to study protein-protein interactions. (a) The ResPep SL is a peptide
synthesizer that needs to be connected to a computer, vacuum pump, and various solutions to synthesize
peptides on a membrane. The 4 L of anhydride Dimethylformamide and 2 L of ethanol are connected to the
equipment from the outside as dilutor 2 solvent 1 and solvent 2, respectively (permission from Intravis AG). (b)
Shown is a representative image of membrane design. White dots represent the location where peptides will
be synthesized. Grey dots represent negative controls which are left without peptide synthesis. On a
membrane, the positive control to the protein of interest and primary antibody are also synthesized. For
example, the sequences for the SIRK peptide, QEHA peptide, βARK peptide, the Gβγ binding domain of the
calcium channel CaV2.2, and the C-terminus of Gβ1 are used to synthesize the positive control peptides to
study the interaction of SNAP-25 and Gβ1γ2 (permission from Intravis AG)
14. Press the “SHOW PEPTIDES” tab and click any peptide to
ensure it is located at the right position.
15. Load the method: Click the “EDIT METHODS” tab, press “file”
and open the method called “SPOTpreactiv.” (see Note 11).
16. Click the “REPORT” tab and check the amount of amino acid
and reagents needed for the prep (see Note 12).
17. Mount and calibrate the spotter needle and 1 ml syringe (see
Note 13).
18. Test the vacuum pump by clicking the ON and OFF buttons
under the “MANUAL” tab in the ResPep software.
19. Fill the DMF reservoir bottle in the ResPep machine and
ensure that the ethanol and DMF bottles on the outside are
attached to the Auto SPOT robot as appropriate (see Note 14).
20. Prime dilutor 1 and 2 to remove air bubbles (see Note 15).
21. Wash the membrane three times with ethanol using the wash
protocol in the method folder.
3.2 Peptide Array 1. In the fumehood, transfer the dissolved amino acid derivatives
Synthesis (Fig. 2) into 2 ml ResPep tubes with caps (see Note 16).
2. Place the pre-swelled membrane onto the SPOT synthesis
frame in fumehood (see Note 17).
3. Load the SPOT synthesis frame into the ResPep SL equipment
and connect the vacuum tube to bottom of the SPOT synthesis
frame.
4. Wash the membrane three times with ethanol using the wash
protocol in methods (see Note 18).
5. Reload the “SPOTpreactiv” method for peptide generation.
Fig. 2 Generating peptide arrays. (a) The cellulose membrane is pre-swelled by DMF. (b) Each peptide is
generated as 14mers, and peptides overlap as shown. The blue residues are part of three different peptides.
(c) Linear peptides are generated on a membrane as shown. Each color dot represents amino acids, which are
synthesized into peptides
314 Yun Young Yim et al.
3.3 Side Chain 1. At the end of the run, use a pencil to mark the top and bottom
Deprotection corners of the membrane and any other location required for
cutting the membrane (see Note 24).
2. In the fumehood, remove the membrane from the SPOT
synthesis frame and place it in a container labeled TFA.
3. Add 10 ml of deprotection solution to the container and let
react for 1 h in the fumehood (see Note 25). Following incu-
bation, discard in a TFA waste container.
4. Wash the membrane four times in 20 ml dichloromethane
(DCM) for 10 min per wash. Discard each wash in dichloro
waste container.
5. Wash the membrane four times in 20 ml DMF for 2 min per
wash. Discard waste in dichloro waste container.
6. Wash the membrane two times in 20 ml EtOH for 2 min per
wash. Discard in dichloro waste container.
7. Allow the membrane to dry in the fumehood until completely
dry. Wrap loosely in tin foil, and place in a labeled container in
the refrigerator until ready to perform the far western
experiment.
Peptide Arrays 315
3.4 Far-Western 1. In a gel box, soak the membrane in ethanol for 5 min and wash
Development (Fig. 3) twice with distilled water for 5 min on a shaker table at room
temperature.
2. Block the membrane for 1 h on a shaker in 20 ml of 5 % milk in
TBS/0.1 % Tween.
3. Wash the membrane five times for 5 min per wash in TBS/
0.1 % Tween on shaker table.
4. Incubate the membrane overnight with an appropriate concen-
tration of the protein of interest in protein binding buffer at
4 C (see Note 26).
5. Wash the membrane three times for 5 min per wash with an
appropriate wash solution on a shaker table
6. Incubate the membrane in 20 ml of primary antibody in pri-
mary antibody solution for 1 h (see Note 7).
7. Wash the membrane three times for 5 min/wash with an
appropriate wash solution on shaker table.
8. Incubate the membrane in 20 ml of horseradish peroxidase
(HRP) conjugated secondary antibody in secondary antibody
solution for 1 h (see Note 7).
9. Wash the membrane two times for 5 min/wash in an appropri-
ate wash solution on shaker.
10. Wash the membrane two times for 10 min/wash in TBS with
OG.
11. Visualize using HRP: in a container, combine 3 ml of each
enhanced chemiluminescence (ECL) reagent and mix well.
Place membrane in the container and cover with tin foil.
Swish container back and forth for 1 min. Transfer the mem-
brane to a plastic sheet cover and use a pipette to smooth away
bubbles. Set up the camera and place the membrane in the
machine to focus as per instructions. Set number of pictures,
exposures, etc. and take pictures (see Note 27).
12. Analyze data using a densitometry program (see Note 28).
3.5 Alanine Screen For the alanine screen, use the methods listed in Subhead-
(Fig. 4) ings 3.1–3.4. At Subheading 3.1, step 5, command “.replace, A”
is needed for alanine screening. This will generate overlapping
peptides as shown in Fig. 2b. The amount of amino acid derivatives
and reagents needed for the experiment will change according to
the peptide sequence, but the protocol is identical to that of general
peptide array synthesis.
316 Yun Young Yim et al.
Fig. 3 Far-Western technique to screen for interactions with a protein of interest. Peptides are synthesized on
a cellulose membrane, and binding interactions assessed using a far-western technique as stated in
Subheading 3. (a) Peptides on the membrane are washed with ethanol and water, and then the blot is
exposed to the protein of interest overnight. The next day, membranes are incubated with a primary antibody
for the binding partner of interest, and a HRP-conjugated secondary antibody. (b) Using enhanced chemilumi-
nescence (ECL) reagents that detect horseradish peroxidase (HRP) enzyme activity, peptides on membranes
are screened for interaction with potential binding partners. Selective peptides will bind to their binding
partners. Shown is a representative image of SNAP-25 peptides on a membrane exposed to Gβ1γ2. Using
image J and Prism, densitometry analysis is used to quantify the intensity of each dot, and values are plotted
via bar graph. Shown are representative densitometry results from the membrane examining SNAP-25 and
Gβ1γ2 interactions. The x-axis reflects both the peptide number and residue number of SNAP-25. Circled
regions, representing regions that interact with of the protein of interest, are used for subsequent alanine
screening (reproduced from Molecular Pharmacology December 2012 82:1136–1149 with permission of
American Society for Pharmacology and Experimental therapeutics)
Peptide Arrays 317
Fig. 4 Alanine Screening. Clusters of SNAP-25 peptides that are circled in red in Fig. 3b are tested via alanine
screen. By monitoring the change in the intensity of each dot, the alanine screening suggests residues that are
important for the protein-protein interaction. (a) Shown is a representative image of alanine screening for the
SNAP-25 130–143 peptide that was synthesized on a membrane. The first spot is a wild type peptide, while
the following 14 are mutated with a single alanine replacement of the residue at position 1–14 from wild type.
Boxes highlight residues which are shown to interrupt the SNAP25-Gβ1γ2 interaction by alanine mutation. (b)
The membrane is developed using the enhanced chemiluminescence (ECL) reagents. Densitometry is
quantified and plotted into a graph as shown above. When arginine residues in the SNAP25 130–143 peptide
are mutated to alanine, a significant decrease in the intensity of the dots is observed (*** p < 0.001)
(reproduced from Molecular Pharmacology December 2012 82:1136–1149 with permission of American
Society for Pharmacology and Experimental therapeutics)
4 Notes
15. For priming, allow depressing five times and emptying out
waste reservoir by turning on the vacuum pump and completely
draining into waste. For dilutor 2, there is solvent 1 (DMF) and
solvent 2 (EtOH), both solutions need to be primed. By click-
ing the dilutor 2 solvent 1 prime button, you can prime the
dilutor 2 solvent 1. Once it is finished, click the dilutor 2 solvent
2 prime button to prime the dilutor 2 solvent 2.
16. Only 2 ml ResPep tubes will fit onto the amino acid holder in
the ResPep equipment. Remember to label each cap and tube
appropriately.
17. Ensure that there are no bubbles or wrinkles on the membrane
once mounted on the SPOT membrane frame.
18. If there are bubbles or the membrane dries unevenly, remove
the membrane from the frame and repeat the DMF and wash
steps.
19. Put a hole on foil for the needle to go through.
20. Before placing the amino acid tubes onto the rack, uncap all
tubes.
21. These tubes will be used as mixing tubes for the activation of
amino acid derivatives.
22. During refilling, synthesis is paused by clicking the PAUSE key.
This will suspend the operation. Only do this when the needle
is away from the reagent rack and NOT pressing down on the
membrane or in an activation tube. A good time to pause is
when the software is counting down between piperidine depro-
tection steps.
23. Waste should be collected following the hazardous waste rules
at each institution. A carboy is connected to the ResPep SL
equipment during peptide array synthesis. There should be
separate waste containers for the deprotection solution and
washes in side-deprotection steps.
24. Use a pencil to mark the top and bottom corners of the mem-
brane. This will be important to identify the orientation of the
membrane later.
25. Ensure the lid is on for the container. Every 15 min or so, swish
solution around.
26. Protein and detergent concentrations have to be determined
empirically. Individual proteins may need different concentra-
tions to obtain optimal signals for analysis. The purity of
expressed proteins may also affect the amount of protein
needed for detection. For example, 0.4 μM Gβγ protein is
needed to identify Gβγ-SNARE interactions. Similarly, for
these experiments, the membrane was determined to tolerate
0.1 % OG.
320 Yun Young Yim et al.
References
1. Frank R (2002) The SPOT synthesis technique 6. Thiele A, Zerweck J, Schutkowski M (2009)
– synthetic peptide arrays on membrane sup- Peptide arrays for enzyme profiling. Methods
ports – principles and applications. J Immunol Mol Biol 570:19–65
Methods 267:13–26 7. Phizicky EM, Fields S (1995) Protein–protein
2. Min DH, Mrksich M (2004) Peptide arrays: interactions – methods for detection and anal-
towards routine implementation. Curr Opin ysis. Microbiol Rev 59:94–123
Chem Biol 8:554–558 8. Wells CA, Zurawski Z, Betke KM et al (2012)
3. Winkler DF, Hilpert K, Brandt O et al (2009) G beta gamma inhibits exocytosis via interac-
Synthesis of peptide arrays using SPOT- tion with critical residues on soluble N-ethyl-
technology and the CelluSpots-method. Meth- maleimide-sensitive factor attachment protein-
ods Mol Biol 570:157–174 25. Mol Pharmacol 82:1136–1149
4. Frank R, Doring R (1988) Simultaneous mul- 9. Mukhija S, Germeroth L, Schneider-Mergener J
tiple peptide-synthesis under continuous-flow et al (1998) Identification of peptides inhibiting
conditions on cellulose paper disks as segmen- enzyme I of the bacterial phosphotransferase
tal solid supports. Tetrahedron 44:6031–6040 system using combinatorial cellulose-bound
5. Dikmans A, Beutling U, Schmeisser E et al peptide libraries. Eur J Biochem 254:433–438
(2006) SC2: a novel process for manufacturing 10. Collet BY, Nagashima T, Yu MS et al (2009)
multipurpose high-density chemical microar- Fluorous-based peptide microarrays for prote-
rays. Qsar Comb Sci 25:1069–1080 ase screening. J Fluor Chem 130:1042–1048
Part III
Tag/Affinity-Based Methods
Chapter 19
Abstract
A fluorescence polarization assay can be used to evaluate the strength of a protein-protein interaction. A
green fluorescent protein variant is fused to one of the protein partners. The formation of a complex is then
deduced from an increase in fluorescence polarization, and the equilibrium dissociation constant of the
complex is determined in a homogeneous aqueous environment. The assay is demonstrated by using the
interaction of the S-protein and S-peptide fragments of ribonuclease A as a case study.
Key words Fluorescence anisotropy, Fluorescence polarization, Fusion protein, Green fluorescent
protein, Protein-protein interaction
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_19, © Springer Science+Business Media New York 2015
323
324 Ronald T. Raines
2 Materials
3 Methods
3.1 Fluorescence 1. Mix protein GFP–X (0.50–1.0 nM) with various concentrations
Polarization Assay of protein Y in 1.0 mL of 20 mM Tris–HCl buffer, pH 8.0, with
or without NaCl at 20 C (see Note 4). Conditions such as
buffer, pH, temperature, and salt can be varied as desired.
2. After mixing, take five to seven polarization measurements at
each concentration of protein Y (see Notes 5 and 6). For a
blank measurement, use a mixture that contains the same com-
ponents except for protein GFP–X.
I jj I ⊥
P¼ ð1Þ
I jj I ⊥
ΔP F
P¼ þ P min ð2Þ
Kd þ F
In Eq. 2, P is the measured polarization, ΔP (¼Pmax – Pmin) is
the total change in polarization, and F is the concentration of
free protein Y (see Note 7).
2. Calculate the fraction of bound protein ( fB) by using the
equation
P P min F
fB ¼ ¼ ð3Þ
ΔP Kd þ F
Plot fB versus F to show the binding isotherms.
3.3 Case Study Fluorescence polarization was used to determine the effect of salt
concentration on the formation of a complex between the S15 and
S-protein fragments of ribonuclease A [11–15]. A GFP chimera of
S-peptide [S15–GFP(S65T)–His6] was produced from bacteria and
titrated with free S-protein. The value of Kd increased fourfold
when NaCl was added to a final concentration of 0.10 M (Fig. 1).
A similar salt dependence for the dissociation of RNase S had been
observed previously [16]. The added salt is likely to disturb the
4 Notes
3ηV
P /τ¼ ð4Þ
RT
In Eq. 4, rotational correlation time (τ) is the time taken for a
molecule to rotate 68.5 and is related to the solution viscosity
(η), molecular volume (V ), gas constant (R), and absolute
temperature (T ). Thus, under conditions of constant viscosity
and temperature, polarization is directly proportional to the
molecular volume, which increases upon complex formation.
3. Using the “superfolder” variant of GFP could facilitate the
creation of a GFP–X fusion protein [19].
4. In the assay solution, [GFP–X] should be significantly lower
than the value of Kd ([GFP–X] < < Kd) but still be high
enough to generate detectable fluorescence in the spectrome-
ter. In the case study, [GFP–X] ¼ 1 nM and Kd > 10 nM.
5. Data collection must be done at equilibrium. To estimate the
time to reach equilibrium, a pilot experiment can be performed
in which Y is added at [Y] ¼ Kd, and the polarization is moni-
tored until it reaches a stationary value.
6. At each [Y], the sample should be blanked with an identical
mixture that lacks GFP–X.
7. The change in polarization (ΔP) upon complex formation must
be detectable. For example, if the value of τ for GFP–X does
not change significantly upon formation of the GFP–X·Y com-
plex, then the value of ΔP is small and the data analysis is
difficult.
Fluorescence Polarization Assay 327
References
1. Jameson DM, Ross JA (2010) Fluorescence 11. Richards FM, Vithayathil PJ (1959) The
polarization/anisotropy in diagnostics and preparation of subtilisin modified ribonucle-
imaging. Chem Rev 110:2685–2708 ase and separation of the peptide and
2. Smith DS, Eremin SA (2008) Fluorescence protein components. J Biol Chem
polarization immunoassays and related methods 234:1459–1465
for simple, high-throughput screening of small 12. Watkins RW, Arnold U, Raines RT (2011)
molecules. Anal Bioanal Chem 391:1499–1507 Ribonuclease S redux. Chem Commun
3. Owicki JC (2000) Fluorescence polarization 47:973–975
and anisotropy in high throughput screening: 13. Richards FM (1958) On the enzymic activity of
perspectives and primer. J Biomol Screen subtilisin-modified ribonuclease. Proc Natl
5:297–306 Acad Sci U S A 44:162–166
4. Royer CA, Scarlata SF (2008) Fluorescence 14. Raines RT (1998) Ribonuclease A. Chem Rev
approaches to quantifying biomolecular inter- 98:1045–1065
actions. Methods Enzymol 450:79–106 15. Kim J-S, Raines RT (1993) Ribonuclease S-
5. Park S-H, Raines RT (1997) Green fluorescent peptide as a carrier in fusion proteins. Protein
protein as a signal for protein–protein interac- Sci 2:348–356
tions. Protein Sci 6:2344–2349 16. Schreier AA, Baldwin RL (1977) Mechanism of
6. Park SH, Raines RT (2000) Green fluorescent dissociation of S-peptide from ribonuclease S.
protein chimeras to probe protein–protein Biochemistry 16:4203–4209
interactions. Methods Enzymol 328:251–261 17. Baldwin RL (1996) How Hofmeister ion inter-
7. Park SH, Raines RT (2004) Fluorescence actions affect protein stability. Biophys J
polarization assay to quantify protein–protein 71:2056–2063
interactions. Methods Mol Biol 261:161–166 18. Connelly PR, Varadarajan R, Sturtevant JM
8. Jameson DM, Sawyer WH (1995) Fluores- et al (1990) Thermodynamics of protein–pep-
cence anisotropy applied to biomolecular inter- tide interactions in the ribonuclease S system
actions. Methods Enzymol 246:283–300 studied by titration calorimetry. Biochemistry
9. Ormö M, Cubitt AB, Kallio K et al (1996) Crys- 29:6108–6114
tal structure of the Aequorea victoria green fluo- 19. Pédelacq JD, Cabantous S, Tran T et al (2006)
rescent protein. Science 237:1392–1395 Engineering and characterization of a super-
10. Yang F, Moss LG, Phillips GN Jr (1996) The folder green fluorescent protein. Nat Biotech-
molecular structure of green fluorescent pro- nol 24:79–88
tein. Nat Biotechnol 14:1246–1251
Chapter 20
Abstract
Förster or Fluorescence resonance energy transfer (FRET) can be used to detect protein-protein interactions.
When combined with microscopy, FRET has high temporal and spatial resolution, allowing the interaction
dynamics of proteins within specific subcellular compartments to be detected in cells. FRET microscopy
has become a powerful technique to assay the direct binding interaction of two proteins in vivo. Here, we
describe a sensitized emission method to determine the presence and dynamics of protein-protein interactions
in living cells.
Key words Fluorescence, FRET, Sensitized emission, Dynamics, Microscopy, Live cell imaging
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_20, © Springer Science+Business Media New York 2015
329
330 Alexa L. Mattheyses and Adam I. Marcus
a No FRET FRET
Spectral Overlap
b
Distance
D A D A
>10nm <10nm
c
Orientation
A
A
D D
θ = 90˚ θ = 0˚
Fig. 1 Conditions for FRET include (a) A donor and acceptor pair where the emission spectra of the donor
overlaps with the excitation spectra of the acceptor (spectral overlap; grey shading). (b) The distance between
the donor and acceptor must be less than approximately 10 nm, at larger distances there is no FRET. (c) FRET
efficiency is highest when the excitation and emission dipoles are oriented parallel and there is no FRET if they
are perpendicular
Table 1
Common FRET pairs
Donor Acceptor
relative fluorescence
Idonor
Donor Excitation
Donor Emission
wavelength
relative fluorescence
Iacceptor
Acceptor Excitation
Acceptor Emission
wavelength
IFRET
Donor Excitation
Acceptor Emission
wavelength
Fig. 2 Microscope setup for FRET. Two lasers, 458 nm for donor excitation and
514 nm for acceptor excitation, are selected with an AOTF and directed into the
microscope by a polychroic mirror. After excitation of the sample, fluorescence is
collected by the objective lens and the fluorescence split based on wavelength
and imaged through a 480/20 filter (donor) or 535/30 filter (acceptor)
Table 2
Imaging parameters
2 Materials
2.1 Sample There are three required and two optional control samples and
one experimental sample. Before transfection with fluorescently
tagged protein constructs cells should be plated in an appropriate
live cell imaging dish, such as MatTek #1.5 coverslip bottom dish
(MatTek Corporation, Ashland, MA). Fixed cell samples should
be plated on #1.5 coverslips, fixed and mounted for imaging (see
Note 1).
1. Required Control sample, donor only. Transfect cells with
cytosolic CFP (or Protein-A-CFP, see Note 2).
2. Required Control sample, acceptor only. Transfect cells with
cytosolic Venus (or Protein-B-Venus, see Note 2).
3. Required Control sample, medium only. Prepare a dish con-
taining no cells with the imaging medium.
4. Required Experimental sample. Transfect cells with protein-A-
CFP and protein-B-Venus. Ideally the expression level of the
proteins should be similar (see Note 3).
5. Suggested Control sample, FRET positive. Transfect cells with
CFP-Venus tandem protein.
6. Suggested Control sample, FRET negative. Transfect cells with
cytoplasmic CFP and cytoplasmic Venus.
2.2 Microscope This protocol describes data acquisition on a laser scanning confo-
cal microscope (Fig. 3). It can be easily adapted for widefield
microscopy (see Note 4).
1. Inverted laser scanning confocal fluorescence microscope with
excitation lasers 458 nm (CFP) and 514 (Venus) and emission
filters 480/40 (CFP) and 535/30 (Venus).
2. Objective lens: 60 1.4 NA oil immersion or similar high
resolution objective lens.
3. Environmental control: heated stage and optional CO2 to
maintain live cells on the microscope (not required for fixed
cell imaging).
2.3 Image Analysis 1. Image analysis software: Fiji (ImageJ pre-packaged with plu-
gins) (free download: fiji.sc/Fiji) or equivalent commercial
software.
FRET Microscopy 335
cells expressing
FRET probes
excitation
458 laser
lasers
514 laser
AOTF
polychroic
458 and 514
beamsplitter
PMT
514 LP
480/20
PMT
fluorescence
detection
535/30
Fig. 3 The three acquisition conditions for sensitized emission FRET. (a) Excite at
the donor wavelength and image at the donor wavelength (Idonor). (b) Excite at
the acceptor wavelength and image at the acceptor wavelength (Iacceptor). (c)
Excite at the donor wavelength and image at the acceptor wavelength (IFRET)
3 Methods
3.1 Optimize The imaging parameters must be held constant for acquisition of all
Imaging Conditions control and experimental images. First, you will optimize these
parameters for all three imaging channels. Use the experimental
sample expressing protein-A-CFP and protein-B-Venus.
1. Prepare the microscope. Select the 60 1.4 NA oil high-
resolution objective. Place a drop of immersion oil on the
objective and focus on the doubly labeled sample. Set the
temperature and CO2 environmental control (see Note 5).
2. Set the image dimensions. The zoom/magnification and num-
ber of pixels should be selected to achieve optimum resolution
as defined by Nyquist [12]. For our 60 1.4 NA objective this
is 90 nm/pixel. The ideal pixel size is determined by the optics
and the NA of the objective (see Note 6).
3. Set the scan speed and line averaging. For live cell imaging the
scan speed should be relatively fast and the line averaging low
to minimize sample movement artifacts.
4. Sequentially for each channel, set the acquisition parameters for
laser power, and detector gain and offset. The signal levels
336 Alexa L. Mattheyses and Adam I. Marcus
3.2 Data Collection All images are collected with the parameters established in
Subheading 3.1.
1. Image the required control for donor bleed-through. Imaging
the CFP only sample, collect Idonor and IFRET for 10–20 indi-
vidual cells.
2. Image the required control for acceptor bleed-through. Imag-
ing the Venus only sample, collect Iacceptor and IFRET for 10–20
individual cells.
3. Image the required control for background. Imaging the
medium only sample, collect Idonor, Iacceptor, and IFRET from
20 independent positions. Ensure the microscope is focused in
the medium (see Note 7).
4. Image the experimental sample. Imaging the protein-A-CFP
and protein-B-YFP sample, collect Idonor, Iacceptor, and IFRET.
A time lapse should be set up to record dynamics.
Suggested controls
5. Image the recommended FRET positive control. From the
CFP-Venus tandem protein sample collect Idonor, Iacceptor, and
IFRET.
6. Image the recommended FRET negative control. Imaging the
cytosolic CFP and Venus sample, collect Idonor, Iacceptor, and
IFRET.
3.3 Image All image processing steps can be performed with Fiji (see Note 8).
Processing and The menu navigation in Fiji for the required functions is in Table 3.
Analysis Images are referred to by the sample and the imaging condition, for
example CFPIdonor is the CFP sample acquired with donor imaging
conditions.
1. Calculate background from the medium only Idonor, Iacceptor,
and IFRET images. Background is a 20 frame average of the
medium only sample:
FRET Microscopy 337
Table 3
ImageJ/Fiji operations
VenusI FRET
α¼
VenusI acceptor
CFPI FRET
β ¼
CFPI donor
Select regions of the image where there was CFP fluores-
cence and measure the average β. Measure for each field of view
and average all β to obtain the final bleed-through correction
value β.
338 Alexa L. Mattheyses and Adam I. Marcus
4 Notes
1. This protocol can be adapted for fixed cell samples that do not
require labeling with antibodies. Transfect samples with donor
and acceptor fusion proteins as described. Following adequate
expression time, fix with 4 % paraformaldehyde and mount the
cells on slides for microscopy.
2. The donor and acceptor only controls are needed to measure the
bleed-through of direct excitation and emission into IFRET. Cyto-
solic expression of CFP and Venus will simplify accurate measure-
ments of these bleed-through contributions; however, protein-
A-CFP and protein-B-Venus fusion proteins can be utilized.
3. The placement of the fluorescent tags in the fusion protein is
critical for FRET. A negative “no FRET” result does not rule
out protein interaction. If the distance between the donor and
acceptor is too great (opposite sides of a complex) or if the
orientation is not optimal, there may be no FRET even when
protein-A and protein-B are in fact interacting. The labeling
strategy should be carefully considered and different tag loca-
tions explored [13].
4. Sensitized emission FRET measurements can be conducted on
a widefield or epi-fluorescence microscope. The widefield
microscope should be equipped with a fluorescence light source
(Hg lamp or LED), a scientific CCD or CMOS camera, and
three filter cubes corresponding to the three imaging channels
(Table 2). For CFP-Venus FRET the three filter cubes should
be: Donor Cube: 430/20 ex 480/30 em; Acceptor Cube:
500/20 ex 535/30 em; and FRET Cube: 430/20 ex 535/
30 em. The protocol can be followed substituting the appro-
priate filter cube for Idonor, Iacceptor, and IFRET.
5. To reduce background, the cell imaging medium should be
phenol-red free. When observed with widefield fluorescence,
the sample is photobleaching. Therefore it is important to
minimize the time the fluorescence lamp shutter is open and
to use neutral density (ND) filters to attenuate the lamp illumi-
nation intensity.
6. The Nyquist sampling theorem is commonly utilized to deter-
mine the ideal pixel size for optimal high-resolution imaging.
7. To focus in the medium it is easiest to focus on some dust or
particles on the coverslip and then up into the medium. Make
sure the areas imaged do not contain any non-uniformities.
FRET Microscopy 339
Acknowledgements
References
1. Lakowicz JR (1988) Principles of frequency- 8. Piston DW, Rizzo MA (2008) FRET by fluo-
domain fluorescence spectroscopy and applica- rescence polarization microscopy. Methods
tions to cell membranes. Subcell Biochem Cell Biol 85:415–430
13:89–126 9. Hoppe A, Christensen K, Swanson JA (2002)
2. Stryer L (1978) Fluorescence energy transfer as Fluorescence resonance energy transfer-based
a spectroscopic ruler. Annu Rev Biochem stoichiometry in living cells. Biophys J
47:819–846 83:3652–3664
3. Lam AJ, St-Pierre F, Gong Y et al (2012) 10. Gordon GW, Berry G, Liang XH et al (1998)
Improving FRET dynamic range with bright Quantitative fluorescence resonance energy
green and red fluorescent proteins. Nat Meth- transfer measurements using fluorescence
ods 9:1005–1012 microscopy. Biophys J 74:2702–2713
4. Gautier I, Tramier M, Durieux C et al (2001) 11. Goedhart J, Hink MA, Jalink K (2014) An
Homo-FRET microscopy in living cells to introduction to fluorescence imaging techni-
measure monomer-dimer transition of GFP- ques geared towards biosensor applications.
tagged proteins. Biophys J 80:3000–3008 Methods Mol Biol 1071:17–28
5. Mattheyses AL, Hoppe AD, Axelrod D (2004) 12. North AJ (2006) Seeing is believing? A begin-
Polarized fluorescence resonance energy trans- ners’ guide to practical pitfalls in image acqui-
fer microscopy. Biophys J 87:2787–2797 sition. J Cell Biol 172:9–18
6. Padilla-Parra S, Tramier M (2012) FRET micros- 13. Miyawaki A, Tsien RY (2000) Monitoring
copy in the living cell: different approaches, protein conformations and interactions by
strengths and weaknesses. Bioessays 34:369–376 fluorescence resonance energy transfer
7. Piston DW, Kremers GJ (2007) Fluorescent between mutants of green fluorescent protein.
protein FRET: the good, the bad and the Methods Enzymol 327:472–500
ugly. Trends Biochem Sci 32:407–414
Chapter 21
Abstract
Enzyme-linked immunosorbent assay (ELISA) is a commonly used method in analyzing biomolecular
interactions. As a rapid, specific, and easy-to-operate method, ELISA has been used as a research tool as well
as a widely adopted diagnostic method in clinical settings and for microbial testing in various industries.
Inhibition ELISA is a one-site binding analysis method, which can monitor protein-protein interactions in
solution as opposed to more commonly used sandwich ELISA in which the analyte capture step is required
on a solid surface either through specific capture or through passive adsorption. Here, we introduce
inhibition ELISA procedures, using a recombinant viral protein as an example, with emphasis on how
inhibition ELISA could be used to probe subtle protein conformational changes in solution impacting
protein–protein binding affinity. Inhibition ELISA is used to probe one binding site at a time for binding
partners in solution with unrestricted conformation. The assay can be performed in a quantitative manner
with a serially diluted analyte in solution for solution antigenicity or binding activity assessment.
Key words Inhibition ELISA, Monoclonal antibody, Antigen–antibody interaction, The half-
maximal inhibitory concentration (IC50), Conformational change, Disulfide bond, Binding affinity,
Binding analysis in solution
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_21, © Springer Science+Business Media New York 2015
341
342 Zusen Weng and Qinjian Zhao
2 Materials
2.2 Antigens and 1. Purified HBsAg: The HBsAg (NIDVD, Xiamen, China) was
Conjugates expressed in CHO (Chinese hamster ovary) cells and purified
to >95 % purity. The concentration of HBsAg was 1.0 mg/mL.
2. Purified HBsAg conformational monoclonal antibody IgG1
5F11 (see Note 5).
3. Horseradish peroxidase (HRP), DTT, NaIO4, glycol, and
NaBH4 were all bought from Sigma.
4. TMB (Tetramethylbenzidine) Reagent A and TMB Reagent B
(INNOVAX, Beijing, China).
5. TMB Stop Solution: 2 M H2SO4 (Beijing Wantai Biopharma-
ceuticals, Beijing, China).
3 Methods
3.2 Determination of 1. Dilute the HBsAg stock to 1.0 μg/mL with PBS for plate
the Proper Working coating.
Concentration of 5F11- 2. Add 100 μL to each well on 96-well plates. Seal the plates and
HRP incubate at 37 C for 6 h.
3. Wash the 96-well plates with 300 μL of PBS-T per well. Tap the
plates on bibulous paper when the wells point down until the
wells are dry.
4. Add 200 μL of assay diluent to each well on 96-well plates (see
Note 12). Seal the plates with plate sealers and incubate at
37 C for 2 h.
5. Wash the 96-well plates with PBS-T and tap again. Seal the
plates and store the plates at 4 C.
6. Dilute 1 μL of 1.0 mg/mL 5F11-HRP solution in 1 mL of
PBS. Then dilute the diluent at serial twofold dilutions (see
Note 13).
7. Transfer 100 μL of the diluents in step 6 to each well on the
plates in step 5. Seal plates and incubate at 37 C for 1 h.
8. Wash the 96-well plates with 300 μL of PBS-T per well five
times. Then tap the plates until the wells are dry as mentioned
before.
346 Zusen Weng and Qinjian Zhao
3.3 Inhibition ELISA The flowchart of solution inhibition ELISA is shown in Fig. 2. The
protocol for inhibition ELISA below should correspond to the
flowchart shown (Fig. 2).
1. Dilute the 5F11-HRP with assay diluent to 10 ng/mL.
2. Dilute the HBsAg samples used to detect twofold serial
dilutions—starting with 4 105 ng/mL (see Note 16). Each
sample should be more than 60 μL.
3. Mix 60 μL of diluted HBsAg sample with 60 μL of diluted
5F11-HRP in each well on plates (see Note 17). Seal the plates
and incubate at 37 C for 30 min. Then centrifuge the plates
for 5 min at 1,500 g.
4. Transfer 100 μL of the supernatant in step 3 to each well of
plates from step 5 of Subheading 3.2 (see Note 18). Seal the
plates and incubate at 37 C for 1 h.
5. Wash the 96-well plates with 300 μL of PBS-T per well five
times. Then tap the plates until no residual liquid remains, as
mentioned before.
Protein-Protein Interaction by ELISA 347
Fig. 2 Flowchart of a solution inhibition ELISA. The reagent HBsAg passively adsorbed on the surface in the
microwells is shown black. The serially diluted analyte in solution, the HBsAg reference or test sample, are
shown in gray. The IC50 range of analyte to reference indicates binding affinity changes to antibody 5F11 (see
Note 20)
4 Notes
Fig. 3 Inhibition ELISA curves for different HBsAg samples (i.e., HBsAg refer-
ence, 0.5 mM DTT treated HBsAg, Post DTT removal HBsAg with oxidative
maturation). Fitted curves and the IC50 values were obtained using “Prism 5.”
The IC50 values for HBsAg reference, 0.5 mM DTT treated HBsAg, Post DTT
removal HBsAg with oxidative maturation are 97, 6,572, and 810 ng/mL,
respectively (or relative IC50 of 1, 0.015, and 0.12, respectively). Nearly tenfold
increase in solution antigenicity was observed as a results of HBsAg spontane-
ous maturation
Acknowledgement
References
1. Leng SX, McElhaney JE, Walston JD et al to the standardization process. Biologicals
(2008) ELISA and multiplex technologies for 32:171–176
cytokine measurement in inflammation and 11. Li H, Shen DT, Jessup DA et al (1996) Preva-
aging research. J Gerontol A Biol Sci Med Sci lence of antibody to malignant catarrhal fever
63:879–884 virus in wild and domestic ruminants by
2. Catt K, Tregear G (1967) Solid-phase radioim- competitive-inhibition ELISA. J Wildl Dis
munoassay in antibody-coated tubes. Science 32:437–443
158:1570 12. Tai HC, Campanile N, Ezzelarab M et al
3. Aviñó A, Gómara M, Malakoutikhah M et al (2006) Measurement of anti-CD154 mono-
(2012) Oligonucleotide-peptide conjugates: clonal antibody in primate sera by competitive
solid-phase synthesis under acidic conditions inhibition ELISA. Xenotransplantation
and use in ELISA assays. Molecules 13:566–570
17:13825–13843 13. Raj GD, Rajanathan TM, Kumar CS et al
4. Yalow RS, Berson SA (1996) Immunoassay of (2008) Detection of peste des petits ruminants
endogenous plasma insulin in man. Obes Res virus antigen using immunofiltration and
4:583–600 antigen-competition ELISA methods. Vet
5. Smitsaart EN, Fernandez E, Maradei E et al Microbiol 129:246–251
(1994) Validation of an inhibition ELISA 14. Dessy FJ, Giannini SL, Bougelet CA et al
using a monoclonal antibody for foot-and- (2008) Correlation between direct ELISA, sin-
mouth disease (FMD) primary diagnosis. Zen- gle epitope-based inhibition ELISA and
tralbl Veterinarmed B 41:313–319 pseudovirion-based neutralization assay for
6. Aleshukina A, Iagovkin É, Bondarenko V measuring anti-HPV-16 and anti-HPV-18
(2011) Change of proinflammatory cytokine antibody response after vaccination with the
profile in human intestine in dysbacteriosis AS04-adjuvanted HPV-16/18 cervical cancer
caused by the antibiotics therapy. Zh Mikrobiol vaccine. Hum Vaccin 4:425–434
Epidemiol Immunobiol: 81–85 15. Giffroy D, Mazy C, Duchene M (2006) Vali-
7. Meng Y, High K, Antonello J et al (2005) dation of a new ELISA method for in vitro
Enhanced sensitivity and precision in an potency assay of hepatitis B-containing vac-
enzyme-linked immunosorbent assay with cines. Pharmeuropa Bio 2006:7
fluorogenic substrates compared with com- 16. Chovel Cuervo ML, Sterling AL, Nicot AI et al
monly used chromogenic substrates. Anal Bio- (2008) Validation of a new alternative for
chem 345:227–236 determining in vitro potency in vaccines con-
8. D€ahnrich C, Komorowski L, Probst C et al taining Hepatitis B from two different manu-
(2013) Development of a standardized ELISA facturers. Biologicals 36:375–382
for the determination of autoantibodies against 17. Friguet B, Chaffotte AF, Djavadi-Ohaniance L
human M-type phospholipase A2 receptor in et al (1985) Measurements of the true affinity
primary membranous nephropathy. Clin Chim constant in solution of antigen-antibody com-
Acta 421:213–218 plexes by enzyme-linked immunosorbent assay.
9. Crowther JR (2000) The ELISA guidebook, J Immunol Methods 77:305–319
vol 149. Humana press, Totowa, New Jersey 18. Zhao Q, Modis Y, High K et al (2012) Disas-
10. Cuervo MLC, de Castro Yanes AF (2004) sembly and reassembly of human papillomavi-
Comparison between in vitro potency tests rus virus-like particles produces more virion-
for Cuban Hepatitis B vaccine: contribution like antibody reactivity. Virol J 9:52
352 Zusen Weng and Qinjian Zhao
19. Harrison RO, Goodrow MH, Hammock BD treatment: Multifaceted biochemical and
(1991) Competitive inhibition ELISA for the immunochemical characterization. Vaccine
s-triazine herbicides: assay optimization and 29:7936–7941
antibody characterization. J Agric Food Chem 22. Mulder AM, Carragher B, Towne V et al
39:122–128 (2012) Toolbox for non-intrusive structural
20. Zhao Q, Wang Y, Abraham D et al (2011) Real and functional analysis of recombinant VLP
time monitoring of antigenicity development based vaccines: a case study with hepatitis B
of HBsAg virus-like particles (VLPs) during vaccine. PLoS One 7:e33235
heat- and redox-treatment. Biochem Biophys 23. High K, Meng Y, Washabaugh MW et al
Res Commun 408:447–453 (2005) Determination of picomolar equilib-
21. Zhao Q, Towne V, Brown M et al (2011) rium dissociation constants in solution by
In-depth process understanding of enzyme-linked immunosorbent assay with
RECOMBIVAX HB® maturation and fluorescence detection. Anal Biochem
potential epitope improvements with redox 347:159–161
Chapter 22
Abstract
Glutathione-S-transferase (GST)-fusion proteins have become an effective reagent to use in the study of
protein-protein interactions. GST-fusion proteins can be produced in bacterial and mammalian cells in large
quantities and purified rapidly. GST can be coupled to a glutathione matrix, which permits its use as an
effective affinity column to study interactions in vitro or to purify protein complexes in cells expressing the
GST-fusion protein. Here, we provide a technical description of the utilization of GST-fusion proteins as
both a tool to study protein-protein interactions and also as a means to purify interacting proteins.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_22, © Springer Science+Business Media New York 2015
353
354 Haris G. Vikis and Kun-Liang Guan
Protein mixture
GSH
GSH
Agarose
Bound protein
GST X
GSH
GSH
GST-X agarose affinity column
Unbound Protein
2 Materials
3 Methods
3.1 Expression Vectors that direct expression of GST fusion proteins in E. coli are
Plasmids widely available commercially. Furthermore, a large collection of
empty vectors and vectors with cDNA inserts are also available
3.1.1 pGEX-KG E. coli through resources such as Addgene (addgene.org) and PlasmID
Expression Vector (plasmid.med.harvard.edu/PLASMID). We commonly use the
pGEX-KG expression vector for production of GST-fusion proteins
in E. coli [1] (Fig. 2). This vector is based on a set of vectors initially
constructed by Pharmacia Biotech. It is typical of many prokaryotic
expression vectors in that it contains an origin of replication
(pBR322 ori), an ampicillin resistance gene (Ampr), and a strong
356 Haris G. Vikis and Kun-Liang Guan
MCS:
5’ CTG GTT CCG CGT GGA TCC CCG GGA ATT TCC GGT GGT GGT GGT
BamHI SmaI
GGAATT CTA GAC TCC ATG GGT CGA CTC GAG CTC AAG CTT AAT TCA 3’
EcoRI XbaI NcoI SalI XhoI SacI HindIII
L MCS
ST Stop codons
G
Am
pr
P tac
pGEX-KG
lac
lq
pBR322 ori
Fig. 2 pGEX-KG E. coli expression vector. pGEX-KG vector shown with essential components: GST gene,
L (linker region), MCS (multiple cloning site), Ampr (ampicillin resistance gene), pBR322 ori (origin of
replication), lacIq (lac repressor), and Ptac (promoter). The MCS is shown with restriction sites
promoter (Ptac) which is kept in the “off” state by the lac repressor
(lacIq). Addition of the non-hydrolyzable lactose analog, isopropyl
β-D-1-thiogalactopyranoside (IPTG) inactivates the lac repressor
and permits activation of the promoter. The GST gene lies down-
stream of the promoter and is followed by a linker region which is a
stretch of amino acids predicted to be very flexible and with no
apparent structure. This is then followed by the multiple cloning
site (MCS) which contains a number of commonly used restriction
sites for insertion of the cDNA of interest.
GST-Fusion Proteins 357
3.1.2 Cloning Strategy Insertion of the cDNA of interest into the pGEX-KG expression
vector MCS is performed by conventional molecular biology clon-
ing manipulations. It is worth noting that the cDNA need not
contain an initiator methionine codon; however, it must be inserted
in the same frame as the GST gene. DNA sequencing may be a
necessary diagnostic method to confirm the construct.
3.2.1 E. coli 1. Transform plasmid expression vector into E. coli strain using
Transformation and standard heat shock methods and plate on an LB/Amp agar
Induction of Protein plate overnight at 37 C.
Expression 2. Pick an individual colony the next day (after 12–18 h of
growth), inoculate 10 mL of LB/Amp (50–100 μg/mL) liquid
media, and grow overnight in a 37 C shaker (see Note 2).
3. The next morning pour the 10 mL culture of E. coli into 1 L of
LB/Amp and grow for 2–5 h until an OD600 of 0.5–0.8 has
been reached (log phase growth).
4. Add IPTG to 0.1 mM and shake cells for 3–4 h at 37 C or
overnight at room temperature (see Note 3).
3.2.3 Elution and Dialysis 1. Elute the protein twice with 0.5 mL of elution buffer (10 min
of GST-Fusion Protein each).
2. To remove the glutathione from the buffer, the eluted protein
can be dialyzed against a buffer of choice at 4 C. We typically
dialyze 1 mL of eluted protein against 1 L of elution buffer
(without glutathione) and replace 1 mM DTT with 0.1 %
β-Me. However, we recommend choosing whichever dialysis
buffer system you deem most appropriate for your protein.
3. Quantify the amount of protein by running an SDS-PAGE gel
against known protein quantities (e.g., BSA). Electrophoresis/
Coomassie staining is preferable to using the Bradford assay
because of the ability to assess protein purity as well (Fig. 3).
Performing a Western blot with α-GST antibody will also allow
you to assess purity and the presence of degradation products.
GST-Ras
Coomassie
1 2 3
Fig. 3 IPTG induction and purity of GST-Ras protein. Samples were run on a
12.5 % SDS-PAGE gel and stained with Coomassie blue. Lane 1, E. coli lysate
prior to IPTG induction; lane 2, E. coli lysate after 3 h of IPTG induction; lane 3,
purified GST-Ras protein. The arrow indicates the location of the GST-Ras protein
in the induced lysate and after purification by glutathione–agarose
GST-Fusion Proteins 359
3.2.4 Storage of Various methods of storage are used and each should be tested to
GST-Fusion Proteins see whether it affects protein stability, activity, etc. GST-fusions can
be stored on beads or in solution. Common storage methods
include:
1. Addition of glycerol to 50 % and storage at 20 C (prevents
sample from freezing).
2. Addition of glycerol to 5–10 % and storage at 80 C.
3. Addition of glycerol to 5–10 % and snap-freeze in liquid N2,
prior to storage at 80 C.
After thawing and prior to use in an experiment, the beads
should be washed to remove glycerol.
3.3 Using GST-Ras to To illustrate the use of a GST-fusion protein in a binding reaction,
Analyze the Interaction we analyzed the interaction of the H-Ras GTPase with the Raf
with Raf kinase. We constructed pGEX-KG-H-RasV12 and -H-RasN17
expression vectors which express two mutants of the human
H-Ras protein. The mutation G12V locks Ras in the GTP nucleo-
tide bound conformation, while the T17N mutation locks the
protein in the GDP nucleotide bound conformation [2, 3]. GTP
bound Ras is in the “on” conformation and is able to interact with
numerous effector molecules such as the protein kinase, Raf,
through the Raf N-terminal domain. GDP bound Ras is in the
“off” conformation and unable to interact with Raf.
We purified GST-RasV12 and GST-RasN17 using the protocol
described in Subheading 3.2. It is important to note that these
small GTPases are required to be purified in the presence of 5 mM
MgCl2 to prevent the loss of bound nucleotide. The following
procedure describes the analysis of the interaction between E. coli-
expressed GST-Ras mutants and mammalian expressed N-terminal
domain (amino acids 1–269) of Raf. Raf 1–269 expression was
directed from the plasmid pCDNA3-cRaf-1-269, which was trans-
fected into mammalian HEK293 cells according to standard lipo-
fection protocols.
1. Transfect a 10 cm dish of mammalian HEK293 cells (using
Lipofectamine (Invitrogen)) with 10 μg of the pCDNA3-cRaf
(1–269) plasmid and grow for a further 48 h.
2. Wash cells once with 10 mL of phosphate buffered saline (PBS)
and lyse with 1 mL of lysis buffer B.
3. Place cells on ice for 10 min, scrape and collect into an eppen-
dorf tube.
4. Centrifuge the lysate at 15,000 g for 15 min at 4 C and
collect the supernatant.
5. Add 10 μg GST, GST-RasV12, and GST-RasN17 into three
separate tubes and to these tubes add the 293 cell lysate col-
lected in step 4. Mix by incubating at 4 C for 1–2 h on a
rocking/rotating platform.
360 Haris G. Vikis and Kun-Liang Guan
GST-RasN17
GST-RasV12
GST
Pulldown:
WB: α -cRaf
GST-RasV12
GST-RasN17
coomassie
GST
Fig. 4 Interaction of Raf and Ras. Top panel, Raf (1–269) binds GST-RasV12 as
detected by α-Raf Western blot. Bottom panel, Coomassie blue stained gel of the
GST-fusions used in the assay
3.4 Production of One benefit of expression in mammalian cells is that many eukary-
GST-Fusion Proteins otic proteins undergo modifications that do not occur in E. coli. For
in Mammalian Cell example, the Ras protein is C-terminal prenylated when expressed
Culture in mammalian cells, but does not undergo this modification in
E. coli because the appropriate modification enzymes are not pres-
ent in bacteria. Furthermore, mammalian proteins that typically
cannot be expressed in E. coli or are easily degraded in E. coli can
often be expressed in mammalian cells where conditions (tRNA,
folding machinery, etc.) are more suitable. However, the fact that
expression in mammalian cells results in less protein compared to E.
coli makes it less economical.
Using glutathione–agarose to purify GST-fusion proteins from
mammalian cells bypasses the need for using antibodies and thus
immunoprecipitation methods. This is particularly useful because
immunopurified proteins cannot easily be eluted from the antibody
and must be eluted by boiling which releases the antibody into the
GST-Fusion Proteins 361
HA- HA-
BRaf AKT
GST-B-Raf
GST-AKT
GST
GST
glutathione-agarose:
HA-B-Raf
pulldown,
WB: α-HA
HA-AKT
GST-B-Raf
* GST-AKT
pulldown,
WB: α-GST
GST
lysate, HA-B-Raf
WB: α-HA *
HA-AKT
1 2 3 4
Fig. 6 Interaction of B-Raf with AKT. HEK293 cells were transfected with the
plasmid combinations indicated and grown for 48 h. Cells were lysed and GST-
fusion proteins were purified by addition of glutathione–agarose. To detect the
presence of co-purified proteins, SDS-PAGE was performed followed by an α-HA
Western blot. Asterisk nonspecific band
3.7 Different Lysis Reagents in the binding buffer may influence protein-protein inter-
Buffers to Use actions. It is often useful to try different buffer systems when
analyzing binding. Three commonly used buffer systems are
provided in order of increasing stringency:
1. NP-40 buffer: 20 mM Tris–HCl, pH 7.5, 100 mM NaCl, 1 %
Nonidet P-40
2. Triton buffer: 20 mM Tris–HCl, pH 7.5, 100 mM NaCl, 1 %
TX-100
3. RIPA buffer: 0.1 % SDS, 1 % Triton X-100, 0.5 % deoxycholate,
50 mM Tris–HCl, pH 7.5, 150 mM NaCl.
4 Notes
Acknowledgments
The authors would like to thank Huira Chong and Jennifer Aur-
andt for critical review of the manuscript. This work was supported
by grants from National Institutes of Health and Walther Cancer
Institute (K.L.G.). K.L.G. is a MacArthur Fellow. H.V. is sup-
ported by a Rackham Predoctoral Fellowship.
References
1. Guan KL, Dixon JE (1991) Eukaryotic proteins 3. Bourne HR, Sanders DA, McCormick F (1991)
expressed in Escherichia coli: an improved The GTPase superfamily: conserved structure and
thrombin cleavage and purification procedure molecular mechanism. Nature 349:117–127
of fusion proteins with glutathione S-transferase. 4. Dyson MR, Shadbolt SP, Vincent KJ, Perera RL,
Anal Biochem 192:262–267 McCafferty J (2004) Production of soluble
2. Katz ME, McCormick F (1997) Signal transduc- mammalian proteins in Escherichia coli: identifi-
tion from multiple Ras effectors. Curr Opin cation of protein features that correlate with
Genet Dev 7:75–79 successful expression. BMC Biotechnol 4:32
Chapter 23
Abstract
Fusion-protein tags provide a useful method to study protein-protein interactions. One widely used fusion
tag is hexahistidine (6xHis). This tag has unique advantages over others due to its small size and the
relatively low abundance of naturally occurring consecutive histidine repeats. 6xHis tags can interact with
immobilized metal cations to provide for the capture of proteins and protein complexes of interest. In this
chapter, a description of the benefits and uses of 6xHis-fusion proteins as well as a detailed method for
performing a 6xHis-pulldown assay are described.
Key words Hexahistidine tag, 6xHis pull down, Affinity chromatography, Protein-protein
interactions
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_23, © Springer Science+Business Media New York 2015
365
366 Mary C. Puckett
2 Materials
3 Methods
3.1 Expression Many commercial plasmids are available for the production of
Plasmids 6xHis fusion proteins. Two commonly used vectors are pDEST17
for bacterial expression and pDEST26 for mammalian expression
(Invitrogen). These plasmids can be used within the gateway clon-
ing system to easily shuttle genes of interest into various vectors.
However, other 6xHis vectors are commercially available.
H2O2 (1mM) - +
HA-ASK1
6xHis
Pull-Down 6xHis-14-3-3
HA-ASK1
Cell Lysate
6xHis-14-3-3
Fig. 3 Example of a 6xHis pulldown assay. Cells were transfected with HA-ASK1
and 6xHis-14-3-3γ. 48 h after transfection, cells were treated with H2O2 and
harvested. Lysate samples were analyzed by performing a 6xHis pulldown
assay, SDS-PAGE, and Western blot
3.3 Performing a His 1. Add 160 μL of the lysate to 25 μL of charged his resin beads (see
Pulldown Subheading 3.4) in a microcentrifuge tube.
2. Rotate samples slowly at 4 C for 2 h (see Note 3).
3. Quickly centrifuge beads and discard the supernatant.
4. Wash the beads twice by adding 200 μL of washing buffer and
once with 200 μL of binding buffer. For each wash, gently
agitate the beads by turning the tube up and down four to
five times, quickly centrifuge the beads, and discard the super-
natant (see Note 4).
5. Recover bound proteins from the beads by boiling in 20 μL of
2 SDS sample loading buffer for 5 min.
6. Analyze proteins by SDS-PAGE and Western blotting. Anti-
6xHis antibodies are available from commercial sources (see
Note 5). A couple commonly used antibodies are listed in
Subheading 2.
3.4 Charging 6xHis 1. Add 3 mL his resin to a chromatography column, such as the
Resin Beads Bio-Rad Poly-Prep column. (Volumes can be scaled up or
down depending on need.)
2. Add 10 mL of nanopure water to the column, and allow to flow
through by gravity. Repeat a total of three times. Allow the
water to flow through completely before each repetition.
3. Add 15 mL of charging buffer to the column.
4. Add 20 mL of binding buffer to the column.
370 Mary C. Puckett
4 Notes
References
Abstract
Blot overlay is a useful method for studying protein-protein interactions. This technique involves
fractionating proteins on SDS-PAGE, blotting to nitrocellulose or PVDF membrane, and then incubating
with a probe of interest. The probe is typically a protein that is radiolabeled, biotinylated, or simply
visualized with a specific antibody. When the probe is visualized via antibody detection, this technique is
often referred to as “Far Western blot.” Many different kinds of protein-protein interactions can be studied
via blot overlay, and the method is applicable to screens for unknown protein-protein interactions as well as
to the detailed characterization of known interactions.
Key words Protein-protein interactions, Blot overlay, Far Western blot, Protein, Receptor,
Association, Nitrocellulose, SDS-PAGE, Binding
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_24, © Springer Science+Business Media New York 2015
371
372 Randy A. Hall
often a fusion protein that is easy to detect. The overlaid probe can
be detected either via incubation with an antibody (this method is
often referred to as a “Far Western blot”), via incubation with
streptavidin (if the probe is biotinylated), or via autoradiography
if the overlaid probe is radiolabeled with 32P. The specific method
that will be described here is a Far Western blot overlay that was
used to detect the binding of blotted hexahistidine-tagged PDZ
domain fusion proteins to soluble GST fusion proteins
corresponding to adrenergic receptor carboxyl-termini [2].
However, this method may be adapted to a wide variety of
applications.
2 Materials
3 Methods
3.1 SDS-PAGE and The purpose of this step is to immobilize the samples of interest on
Blotting nitrocellulose or an equivalent matrix, such as PVDF. It is very
important to keep the blot clean during the handling steps involved
in the transfer procedure, since contaminants can contribute to
increased background problems later on during detection of the
overlaid probe.
1. Place gel in SDS-PAGE apparatus and fill chamber with
running buffer.
2. Mix purified hexahistidine-tagged fusion proteins with SDS-
PAGE sample buffer to a final concentration of approximately
0.1 μg/μl of fusion protein (see Note 1).
3. Load 20 μl of fusion protein (2 μg total) in each lane of the gel.
If there are more lanes than samples, load 20 μl of sample buffer
in the extra lanes (see Note 2).
4. In at least one lane of the gel, load 20 μl of SDS-PAGE
molecular weight markers.
5. Run gel for approximately 1 h at 150 V using the power supply.
6. Stop gel, turn off the power supply, remove the gel from its
protective casing, and place in transfer buffer.
7. Place pre-cut nitrocellulose in transfer buffer to wet it.
8. Put nitrocellulose and gel together in transfer apparatus, and
transfer proteins from gel to nitrocellulose for 90 min at 200 V
using a power supply.
3.2 Overlay During the overlay step, the probe is incubated with the blot and
unbound probe is then washed away. The potential success of the
overlay depends heavily on the purity of the overlaid probe. GST
and hexahistidine-tagged fusion proteins should be purified as
extensively as possible. If the probe has many contaminants, this
may contribute to increasing the background during the detection
step, making visualization of the specifically bound probe more
difficult.
1. Block blot in blocking buffer for at least 30 min (see Note 3).
2. Add GST fusion proteins to a concentration of 25 nM in 10 ml
of blocking buffer.
3. Incubate GST fusion proteins with blot for 1 h at room tem-
perature while rocking slowly.
4. Discard GST fusion protein solution and wash blot three times
for 5 min each with 10 ml of blocking buffer while rocking
slowly.
374 Randy A. Hall
3.3 Detection of The final step of the overlay is to detect the probe that is bound
Overlaid Proteins specifically to proteins immobilized on the blot. In viewing differ-
ent exposures of the visualized probe, an effort should be made to
obtain the best possible signal-to-noise ratio. Nonspecific back-
ground binding will increase linearly with time of exposure. Thus,
shorter exposures may have more favorable signal-to-noise ratios.
1. Incubate blot with enhanced chemiluminescence solution for
60 s (see Note 5).
2. Remove excess ECL solution from blot and place blot in a clear
plastic sheet protector.
3. Tape sheet protector into an autoradiography cassette.
4. Move to the darkroom and place one sheet of film into the
autoradiography cassette with the blot.
5. Expose film for 5–2,000 s, depending on the intensity of the
signal.
6. Develop the film in standard film developer.
4 Notes
Fig. 2 Overlay of hexahistidine-tagged MAGI-2 PDZ1 onto GST-tagged adrenergic receptor carboxyl-termini.
(a) In the reverse of the overlay experiments illustrated in Fig. 1, equal amounts (2 μg) of purified GST fusion
proteins corresponding to the carboxyl-termini of various adrenergic receptor subtypes were immobilized on
nitrocellulose. Overlay with His/S-tagged MAGI-2 PDZ1 (20 nM) revealed strong binding to β1AR-CT-GST but
no detectable binding to control GST, β2AR-CT-GST or α1AR-CT-GST. These data demonstrate that the
interaction between the β1AR-CT and MAGI-2 PDZ1 can be visualized via overlay in either direction. (b)
Estimate of the affinity of the interaction between β1AR-CT and MAGI-2 PDZ1. Nitrocellulose strips containing
2 μg β1AR-CT-GST (equivalent to lane 2 in the preceding panel) were incubated with His/S-tagged MAGI-
2 PDZ1 at six concentrations between 1 and 300 nM. Specific binding of MAGI-2 PDZ1 did not increase
between 100 and 300 nM, and thus the binding observed at 300 nM was defined as “maximal” binding. The
binding observed at the other concentrations was expressed as a percentage of maximal binding within each
experiment. The bars and error bars shown on this graph indicate mean SEM (n ¼ 3). The KD for MAGI-
2 PDZ1 binding to β1AR-CT was estimated at 10 nM (see Note 6)
Blot Overlay 377
Acknowledgments
References
Abstract
Co-immunoprecipitation (Co-IP) is one of the most widely used methods to identify novel proteins that
associate with a protein of interest or to determine complex formation between known proteins. For this
technique, a protein of interest is captured using a specific antibody. The antibody-bound protein, as well as
any proteins bound to the protein of interest, is then precipitated using a resin (immunoprecipitation, IP).
Proteins that are not bound to the protein of interest are then removed from the sample with a series of
washes. The resulting immunocomplexes are then analyzed by immunoblot. As the requirements for
protein-protein interactions vary, optimal experimental conditions for examining the interacting partners
of different proteins of interest must be determined empirically. Once appropriate experimental conditions
have been established, the IP/Co-IP procedure is simple and straightforward. In this chapter, a standard
protocol for IP/co-IP, with several key factors for the success of IP/co-IP analyses, is discussed.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_25, © Springer Science+Business Media New York 2015
381
382 Yoshinori Takahashi
X Y
Z
5. Analyze by immunoblotting
2 Materials
3 Methods
4 Notes
References
1. Fu H (2004) Protein-Protein Interactions. endogenous proteins. Anal Bioanal Chem
Methods and Applications. Methods in Molecu- 389:461–473
lar Biology 261. 5. Berggard T, Linse S, James P (2007) Methods
2. Braun P, Gingras AC (2012) History of protein- for the detection and analysis of protein-protein
protein interactions: from egg-white to complex interactions. Proteomics 7:2833–2842
networks. Proteomics 12:1478–1498 6. Takahashi Y, Coppola D, Matsushita N et al
3. Dwane S, Kiely PA (2011) Tools used to study (2007) Bif-1 interacts with Beclin 1 through
how protein complexes are assembled in signal- UVRAG and regulates autophagy and tumori-
ing cascades. Bioeng Bugs 2:247–259 genesis. Nat Cell Biol 9:1142–1151
4. Markham K, Bai Y, Schmitt-Ulms G (2007) Co- 7. Hsu YT, Youle RJ (1997) Nonionic detergents
immunoprecipitations revisited: an update on induce dimerization among members of the Bcl-
experimental concepts and their implementation 2 family. J Biol Chem 272:13829–13834
for sensitive interactome investigations of
Chapter 26
Abstract
In the cell, homo- and hetero-associations of polypeptide chains evolve and take place within subcellular
compartments that are crowded with many other cellular macromolecules. In vivo chemical cross-linking of
proteins is a powerful method to examine changes in protein oligomerization and protein-protein interac-
tions upon cellular events such as signal transduction. This chapter is intended to provide a guide for the
selection of cell membrane permeable cross-linkers, the optimization of in vivo cross-linking conditions,
and the identification of specific cross-links in a cellular context where the frequency of random collisions is
high. By combining the chemoselectivity of the homo-bifunctional cross-linker and the length of its spacer
arm with knowledge on the protein structure, we show that selective cross-links can be introduced
specifically on either the dimer or the hexamer form of the same polypeptide in vitro as well as in vivo,
using the human type B nucleoside diphosphate kinase as a protein model.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_26, © Springer Science+Business Media New York 2015
391
392 Fabrice Agou and Michel Véron
2 Materials
2.1 Pure The recombinant wild type hexamer was purified as described in
Recombinant Proteins ref. [16]. The recombinant dimeric mutant P96S-A146 stop was
constructed by the overlap extension method [17]. The point
mutation P96S and the removal of the 7 C-terminal amino acids
in the crystallographic hexamer interface has been shown to greatly
affect hexamerization of NDPK [14]. The double mutant was
purified according to a procedure similar to the one used for the
wild type protein except that all purification buffers contained
0.05 mM of dodecyl-β-D-maltoside (DDM) detergent. Proteins
were stored at 20 C in 20 mM potassium phosphate pH 7.0
buffer containing 1 mM DTE and 50 % (v/v) glycerol.
2.5 Cell Culture 1. Cell culture equipment (plastic culture vessels, laminar flow
hood, CO2 incubator, water bath).
2. Complete culture medium (DMEM, fetal calf serum (FCS),
antibiotics).
3. HeLa cells (ATCC CCL-2) in serum supplemented DMEM.
4. Ca2+ and Mg2+-free phosphate-buffered saline (CMF-PBS).
5. Trypsin–EDTA solution in CMF-PBS.
2.7 Whole Cell Urea lysis buffer (SBLU): 62.5 mM Tris–HCl, pH 7.8, 2 % SDS,
Extracts 6 M urea, 100 mM DTE, and 0.05 % bromophenol blue.
3 Methods
3.1 General Cross-links can be more efficiently introduced in a protein when its
Considerations: In Vivo three-dimensional structure is known. The criteria to be considered
Selectivity in Cross- for optimal selectivity in in vivo protein cross-linking (hetero- or
Linking Based on homo-bifunctional reagent; type of reactive group; length and
Structural Design rigidity of the linker structure) are almost the same as those previ-
ously described in vitro [1, 18] except for two:
1. The cross-linker must diffuse across the membrane, and there-
fore its linker structure should be mostly hydrophobic.
2. Individual amino acids likely to react with cross-linker should be
limited in the 3D space to minimize nonspecific intermolecular
bridges in vivo (see Note 1). The use of a low cells–cross-linker
ratio is also advised to limit the extent of protein modifications,
thereby enhancing the relative differences in chemical reactivity
between the amino acids.
We used a non-cleavable, membrane-permeable, homo-
bifunctional reagent containing maleimide groups at both ends
for introducing selective cysteine cross-links in vivo. In addition
to the advantages of cysteine-based cross-linking, the extent of
protein modification through cysteine residues is usually reduced
due to the low content of cysteine in protein, minimizing nonspe-
cific intermolecular cross-linking in vivo.
Protein Cross-Linking in Living Cells 395
3.2.1 In Vitro Cross- 1. Dialyze extensively or desalt the protein with a small G25 col-
Linking of the NDPK-B umn into 50 mM sodium phosphate pH 7.2 reaction buffer
Hexamer (See Fig. 2) containing 150 mM sodium chloride to remove any trace of
reducing agents.
Fig. 2 In vitro cross-linking of the NDPK-B hexamer with short and long cysteine specific cross-linkers. NDPK-
B, 0.02 mg/ml (1.2 μM, subunit concentration), was reacted with either the short (BMOE) (a) or the long cross-
linker (BM[PEO]3) (b) using a concentration of 0.1 mM following the experimental procedure described in
Subheading 3.2.1. At the time indicated, the reaction was quenched by addition of SDS-PAGE loading buffer
and the samples were subjected to 15 % SDS-PAGE followed by Coomassie staining. M, D*, H*, and NS* refer
to the positions of monomers, cross-linked dimers, cross-linked hexamers, and nonspecific higher order
cross-linked species of NDPK-B subunits, respectively. (c): Scans of the SDS-PAGE gel shown in (a) in order to
quantify cross-linked species. The hexameric cross-linked species was very low as compared to the cross-
linked dimer even though the protein at 0.02 mg/ml was a hexamer as judged by the equilibrium sedimenta-
tion [16]. This reflects the very low efficiency of these short and long cross-linkers to cross-bridge all six
subunits together. Patterns were similar using a higher concentration of cross-linker or using different protein
concentrations (0.002 mg/ml or 0.2 mg/ml) confirming the low cross-linking efficiency in the formation of the
hexameric cross-linked species
Protein Cross-Linking in Living Cells 397
3.2.2 In Vitro Cross- Gel filtration analyses showed that the purified P96S-A146stop
Linking of the NDPK-B mutant formed a stable dimer in the protein concentration range
Dimeric Mutant (P96S- of 0.5–50 μM (subunit concentration). Experimental procedures to
A146stop) (See Fig. 3) cross-link the NDPK-B dimeric mutant with the long and short
cross-linkers were similar to those used for the wild type hexamer
except that the reaction buffer contained 0.05 mM of dodecyl-β-D-
maltoside (DDM) detergent, to stabilize the dimer.
Obviously the in vitro characterization of protein cross-linking
with pure components (as illustrated in Fig. 4) is not a prerequisite
step to carry out in vivo cross-linking. This was shown here for a
direct comparison of the selectivity of protein cross-linking in vitro
and in a cellular context.
Fig. 3 In vitro cross-linking of the dimeric mutant of NDPK-B with short and long cysteine specific cross-
linkers. Pure dimeric mutant, 0.02 mg/ml 1.2 μM (subunit concentration) was treated with either the short
(BMOE) (a), or the long cross-linker (BM[PEO]3) (b), according to the instructions described in Subheading 3.2.2
using 0.1 mM of the cross-linker. The reaction was quenched by addition of SDS-PAGE loading buffer at the
time indicated. Cross-linked species were then analyzed by SDS-PAGE and Coomassie blue staining. M and D*
denote the monomer and the cross-linked dimer. (c) Scans of the SDS-PAGE gel shown in (a). The graph
represents the percentage of each species with time and each curve was fitted with a monoexponential (solid
line) (see also Note 3). Scans of the SDS-PAGE gel shown in (b) are not represented under these experimental
conditions because the reaction was too fast
3.3.1 Transient 1. The day before the transfections, trypsinize and count the
Transfections of HeLa Cells HeLa cells, plate them at the appropriate plating density in a
to Overexpress the Dimeric complete medium so that they are 50–60 % confluent on the
Mutant and the Wild Type day of transfection. For cationic lipid-mediated transfections a
Hexamer 6-well plate was used, with each well containing 0.2 106
HeLa cells in a total complete media volume of 2 ml
(DMEM, 10 % FCS).
2. On the day of transfection, add the requisite volume of serum-
free medium as diluent to a total volume of 100 μl in a sterile
polystyrene or polypropylene tube, and then the cationic
lipid transfection reagent such as FuGENE 6 (Roche). Tap
gently to mix.
Protein Cross-Linking in Living Cells 399
Fig. 4 Diagram of in vitro cross-linking efficiencies of NDPK-B with short and long cysteine-specific cross-
linkers as a function of the oligomeric states. Bimolecular rate constants relative to short (BMOE) and long (BM
[PEO]3) cross-linkers with the dimer and the hexamer were determined using three independent cross-linker
concentrations as described in Subheading 3.2. (see Note 3). The arrow surfaces relative to cross-linking
reactions with short (in black) and long cross-linkers (in gray) are proportional to the magnitude of each rate
constant. The reaction D ! D* is the formation of the dimeric mutant into the cross-linked dimer; H ! D* is
the formation of the hexamer into two cross-linked subunits; D* ! H* is the formation of the cross-linked
dimer into the cross-linked hexamer and H ! H* is the direct conversion of the wild type into the cross-linked
hexamer. The rate constants k1long D ! D* and k1short D ! D* were 900 50 M1.s1 and 9.6
0.5 M1.s1, respectively, meaning that the cross-linking efficiency on the dimer is enhanced about
100-fold with the long cross-linker compared to the short cross-linker. The cross-linking efficiency
corresponding to the formation of two cross-linked subunits within the hexamer is lower with both reagents.
Bimolecular rate constants, k1long H ! D* and k1short H ! D*, were 0.9 M1.s1 and 3 M1.s1, respec-
tively, indicating that the short cross-linker is slightly more effective as compared to the long cross-linker in
hexamer cross-linking. Non-measurable rates of the cross-linked hexamer or the intermediate cross-linked
species (trimer, tetramer, or pentamer) are shown as dotted arrows
3.3.2 In Vivo Protein On the day of the treatment with cross-linkers, HeLa cells should
Cross-Linking (See Fig. 5) be 80 % confluent.
1. Dissolve the long and short cross-linkers in DMSO to a final
concentration of 20 mM.
2. Trypsinize each well with 100 μl of Trypsin–EDTA solution
and incubate for 5 min at 37 C. Stop the reaction by adding
400 μl of complete medium and count the cells.
3. Immediately after, transfer the cells still in suspension into a
sterile polystyrene or polypropylene tube. Wash the cells twice
with 500 μl of complete medium. Incubate cells for 30 min on
ice.
4. Distribute HeLa cells in sterile polystyrene or polypropylene
tubes with a cell density of 0.8 106 cells in 20 μl of com-
plete medium.
5. Add the cross-linker directly to the cell suspension at a final
concentration of 1 mM (1 μl of 20 mM stock in DMSO in a
total medium volume of 20 μl). Immediately mix gently. Take
one half of the cells without cross-linker treatment and add the
same volume of DMSO as a control. To check the quenching
efficiency, add the cross-linker in a tube already containing
30 mM of DTE. Incubate samples for 1 h at 37 C in 5 %
CO2. Swirl the suspension occasionally during the 37 C incu-
bation (see Notes 4 and 5).
6. Stop the cross-linking reaction by adding 4 μl of 6 quench-
ing buffer (for a final concentration of 30 mM). Let the cells
incubate for 10 min at 37 C to allow the quencher to fully
diffuse into the cells.
7. Visualize and compare cells which were treated and mock
treated by light microscopy, in order to check for cellular death.
8. Pellet cells by centrifugation at 4 C and wash twice with cold
CMF-PBS buffer at 4 C.
3.4 Concluding In vivo protein cross-linking based on structural design can greatly
Comments facilitate the introduction of selective cross-links on proteins to
study protein oligomerization or protein-protein interactions.
The combination of the chemoselectivity and regioselectivity of
the reagent with the protein structure can even allow examination
of changes in protein oligomerization upon cellular events and
specific protein-protein interactions upon ligand binding. By con-
sidering the specificity of the maleimide group for cysteine residues
and the spatial arrangement of these residues, selective cross-linking
can be introduced specifically on either the dimer or the hexamer
in vitro as well as in intracellular environments that are crowded
with many other cellular macromolecules. The method described
here allows investigation into the oligomerization of the NDPK-B
protein in metastatic, tumor, and normal cells (unpublished
results). This approach can be extended to signaling proteins,
such as the NEMO protein, an essential modulator involved in
the NF-κB pathway, which are usually less abundant in cells. We
have successfully used these methods to show that NEMO forms a
protein complex in cells, termed IκB kinase (IKK) complex, with
two IKKα/β kinases [20]. Strikingly, the molecular mass of the
cross-linked IKK complex perfectly matched the subunit composi-
tion of the IKK complex, which was accurately determined in vitro
more than one decade after the in vivo cross-linking experiments
[21]. Given the high sensitivity of this method to detect unstable
homo- and hetero-associations of proteins, the experimenter will
also have to address the question of whether the cross-links corre-
spond to a mature protein association or to an assembly intermedi-
ate. This is particularly true when in vivo cross-linking results are
compared in different cells at variable stages of proliferation.
4 Notes
Acknowledgments
The authors are very grateful to Samuel Levy, MD for his critical
reading of the manuscript. This work was supported, in whole or in
part, by the Fondation ARC pour la recherche sur le cancer and La
Ligue contre le cancer.
References
1. Ji TH (1983) Bifunctional reagents. Methods 5. Pornillos O, Ganser-Pornillos BK, Kelly BN
Enzymol 91:580–609 et al (2009) X-ray structures of the hexameric
2. Kluger R, Alagic A (2004) Chemical cross- building block of the HIV capsid. Cell
linking and protein-protein interactions-a 137:1282–1292
review with illustrative protocols. Bioorg 6. Mika JT, Poolman B (2011) Macromolecule
Chem 32:451–472 diffusion and confinement in prokaryotic
3. Petrotchenko EV, Borchers CH (2010) Cross- cells. Curr Opin Biotechnol 22:117–126
linking combined with mass spectrometry for 7. Wang K, Richards FM (1975) Reaction of
structural proteomics. Mass Spectrom Rev dimethyl-3,3’-dithiobispropionimidate with
29:862–876 intact human erythrocytes. Cross-linking of
4. Stengel F, Aebersold R, Robinson CV (2012) membrane proteins and hemoglobin. J Biol
Joining forces: integrating proteomics and Chem 250:6622–6626
cross-linking with the mass spectrometry of 8. Bruce JE (2012) In vivo protein complex
intact complexes. Mol Cell Proteomics 11 topologies: sights through a cross-linking
(R111):014027 lens. Proteomics 12:1565–1575
Protein Cross-Linking in Living Cells 405
9. Boissan M, Lacombe ML (2011) Learning 15. Mesnildrey S, Agou F, Veron M (1997) The
about the functions of NME/NM23: lessons in vitro DNA-binding properties of NDP
from knockout mice to silencing strategies. kinase are related to its oligomeric state. FEBS
Naunyn Schmiedebergs Arch Pharmacol Lett 418:53–57
384:421–431 16. Agou F, Raveh S, Mesnildrey S et al (1999)
10. Lascu I, Gonin P (2000) The catalytic mecha- Single strand DNA specificity analysis of
nism of nucleoside diphosphate kinases. J Bioe- human nucleoside diphosphate kinase B.
nerg Biomembr 32:237–246 J Biol Chem 274:19630–19638
11. Dexheimer TS, Carey SS, Zuohe S et al (2009) 17. Pogulis RJ, Vallejo AN, Pease LR (1996) In
NM23-H2 may play an indirect role in tran- vitro recombination and mutagenesis by over-
scriptional activation of c-myc gene expression lap extension PCR. Methods Mol Biol
but does not cleave the nuclease hypersensitive 57:167–176
element III(1). Mol Cancer Ther 18. Kluger R (1997) Chemical cross-linking and
8:1363–1377 protein function. In: Creighton TE (ed) Pro-
12. Postel EH, Berberich SJ, Flint SJ et al (1993) tein function a practical approach. IRL Press,
Human c-myc transcription factor PuF identi- Oxford, p 185
fied as nm23-H2 nucleoside diphosphate 19. Morera S, Lacombe M-L, Xu Y et al (1995)
kinase, a candidate suppressor of tumor metas- X-Ray structure of nm23 Human Nucleoside
tasis. Science 261:478–480 Diphophate Kinase B complexed with GDP at
13. Mesnildrey S, Agou F, Karlsson A et al (1998) 2A resolution. Structure 3:1307–1314
Coupling between catalysis and oligomeric 20. Agou F, Ye F, Goffinont S et al (2002)
structure in NDP kinase. J Biol Chem NEMO trimerizes through its coiled-coil C-
273:4436–4442 terminal domain. J Biol Chem 277:
14. Karlsson A, Mesnildrey S, Xu Y et al (1996) 17464–17475
Nucleoside diphosphate kinase. Investigation 21. Napetschnig J, Wu H (2013) Molecular Basis
of the intersubunit contacts by site-directed of NF-kappaB Signaling. Annu Rev Biophys
mutagenesis and crystallography. J Biol Chem 42:443–468
271:19928–19934
Part IV
Abstract
Yeast two-hybrid (Y2H) screening permits identification of completely new protein interaction partners for
a protein of interest, in addition to confirming binary protein-protein interactions. After discussing the
general advantages and drawbacks of Y2H and existing alternatives, this chapter provides a detailed protocol
for traditional Gal4p-based Y2H library screens in Saccharomyces cerevisiae AH109. This includes bait
transformation, bait auto-activation testing, prey library transformation, Y2H evaluation, and subsequent
identification of the prey plasmids. Moreover, a one-on-one mating protocol to confirm interactions
between suspected partners is given. Finally, a quantitative α-galactosidase assay protocol to compare
interaction strengths is provided.
1 Introduction
1.1 Gal4p-Based The standard Y2H system exploits the modular nature of eukary-
Yeast Two-Hybrid otic transcription factors like Gal4p of Saccharomyces cerevisiae. This
transcriptional activator has two functional domains: an amino
terminal DNA binding domain and a carboxy terminal activation
domain. While the former binds to the upstream activating
sequence (UAS) of the galactose metabolism genes in S. cerevisiae,
the latter activates their transcription by recruiting the RNA poly-
merase to the promoter region. Moreover, Keegan et al. [2]
demonstrated that Gal4p remains functional even if the two
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_27, © Springer Science+Business Media New York 2015
409
410 Jeroen Wagemans and Rob Lavigne
Fig. 1 The Y2H system. (a) Transcriptional activator Gal4p of S. cerevisiae is composed of two functional
domains: the DNA binding domain (DBD) that binds to the upstream activating sequence (UAS) and the
activation domain (AD) that recruits the RNA polymerase and activates transcription of a reporter gene. Both
components are required to produce a selectable phenotype. Y2H uses the modularity of Gal4p. (b) The bait
protein X is fused to the Gal4p DBD, which cannot activate transcription on its own. (c) The prey protein Y is
fused to the Gal4p AD. Since this fusion protein is not recruited to the promoter, it is also unable to activate
transcription on its own. (d) Only when X and Y interact, the DBD and AD are both present at the promoter site,
restoring Gal4p function resulting in a detectable phenotype
domains are not covalently attached as long as they are both present
at the promoter site. Fields [1] picked up this idea to fuse interact-
ing proteins to the two Gal4p domains, which physically brings the
two domains together and restores Gal4p function.
In Gal4p-based Y2H, the protein fused to the C-terminus of
the Gal4p DNA binding domain is known as the bait, while the
other interaction partner, fused to the C-terminus of the Gal4p
activation domain, is called the prey. These constructs are both
transformed into the same yeast cell. Finally, a third component
of the Y2H system (Fig. 1) is the reporter construct in this bait- and
prey-expressing yeast cell. This reporter gene is located down-
stream from a promoter with a UAS recognized by the Gal4p
DNA binding domain. If bait and prey interact, the Gal4p activa-
tion domain is tethered to the promoter region and transcription of
the reporter gene is activated. This results in a specific, selectable
phenotype.
While Fields originally designed the system to investigate
known interactions, Chien et al. [3] applied the technique 2 years
later to unravel completely new interactions for a particular protein
of interest. The known protein, fused to the Gal4p DNA binding
domain, was used as a bait to screen for interaction partners in a
whole library of potential interaction partners, fused to the activa-
tion domain. After Y2H interaction analysis, the prey plasmid was
harvested from those yeast colonies able to activate all reporter
genes. Finally, by sequencing the prey plasmid inserts, the interac-
tion partner of the protein of interest was identified.
Gal4p-Based Yeast Two-Hybrid 411
1.2 Drawbacks of As for any other protein-protein interaction analysis technique, one
the Yeast Two-Hybrid should realize it is almost impossible to detect all interacting pro-
System and Possible teins using Y2H. In contrast, Y2H can also identify interactions
Solutions that actually do not occur in vivo. These are called false negative and
false positive results, respectively. These often arise due to the use of
artificial fusion proteins in combination with an artificial reporter
construct, the removal of the proteins from their natural biological
context, their translocation to the yeast nucleus, or the production
of the hybrid protein at a different level than its natural concentra-
tion in the cell [4].
One potential problem is the bait protein, which could inde-
pendently lead to reporter gene activation (e.g., if the protein has
transcription activation activity or interacts nonspecifically with the
Gal4p activation domain). This is called auto-activation or self-
activation and can lead to false positive results. While this activity
can be related to the biological function of the protein, it can also
be a consequence of the fusion with the DNA binding domain or
the removal of the protein from its natural context [4]. Because of
this phenomenon, auto-activation of the bait protein should always
be checked before performing the YH2 screen. If this auto-
activation test is positive, the bait as such is not suited for the
screen. To resolve this problem, the transcription activation domain
could be removed and the screen performed with the other
domain, or another Y2H system could be used.
While only the lacZ reporter gene, encoding β-galactosidase,
was originally used in a classical blue/white screening assay [1],
current Y2H reporter yeast strains have several secondary auxotro-
phic reporter genes, which permit selection for interaction by
monitoring yeast cell growth on selective media. For instance,
Y2H strain S. cerevisiae AH109 [5] has the HIS3 and ADE2
reporter genes, encoding imidazole glycerol phosphate dehydratase
and phosphoribosylaminoimidazole carboxylase, respectively.
These enzymes are crucial in the biosynthesis of histidine and
adenine. By plating on medium lacking histidine and/or adenine,
interacting baits and preys are selected. In addition, S. cerevisiae
AH109 contains the MEL1 reporter gene, encoding α-galactosi-
dase, a secreted enzyme enabling direct blue/white screening on
X-α-gal indicator plates. The use of more than one reporter gene
reduces the number of false positive results [5].
Moreover, when using HIS3 as a reporter gene, as is the case
with strain S. cerevisiae AH109, 3-amino-1,2,4-triazole (3-AT)
should be added to the medium. HIS3 is the most leaky construct
of the three reporter genes in AH109, so there is always a low
background transcription activation of this gene. 3-AT is a compet-
itive inhibitor of the HIS3 product and will reduce false positive
results due to the background activation [6]. At the same time, the
3-AT concentration should not be too high to avoid losing identi-
fication of weaker interactions. The optimal concentration that
412 Jeroen Wagemans and Rob Lavigne
Table 1
Summary of applied yeast two-hybrid vectors
Characteristics Selection
1.3 Advantages of Despite its drawbacks, the Y2H system has some clear advantages
the Yeast Two-Hybrid over other methods. Firstly, it is clear that an in vivo assay more
Approach closely resembles cellular conditions. Furthermore, Y2H is able to
detect weak and transient interactions since a continuous activation
of the reporter genes is not necessary to see a signal. The fact that
Y2H only detects binary interactions can also be an advantage.
In contrast to methods such as affinity purification, which detect
whole complexes, Y2H directly identifies the two interaction part-
ners. Moreover, there is no need for recombinant proteins, which is
a huge advantage. Finally, although it is labor intensive, Y2H has a
relatively low investment cost. Only minimal laboratory equipment
and low-cost microbial growth media are needed [4].
This chapter provides a detailed protocol for Gal4p-based Y2H
screens using one protein as bait against a prey library. Assuming a
bait and prey library has been constructed, this protocol will first
describe small-scale yeast transformation of the bait plasmid.
In addition, this chapter includes a bait auto-activation test, large-
scale yeast transformation of the prey library, evaluation of the Y2H
results, yeast plasmid isolation, and identification of the inserts by
DNA sequencing. Since Y2H is prone to false positive results, it is
important to confirm the potential interactions. A one-on-one
protocol using mating between the two different S. cerevisiae strains
AH109 (mating type a) [5] and Y187 (mating type α) [18] will also
be discussed. This protocol can also be applied to directly confirm
interaction between two suspected interaction partners. Finally, a
quantitative α-galactosidase assay to compare different interaction
strengths will be provided.
414 Jeroen Wagemans and Rob Lavigne
2 Materials
2.1 Bait Plasmid 1. Yeast strain: S. cerevisiae AH109 with genotype MATa,
Transformation trp1-901, leu2-3,112, ura3-52, his3-200, gal4Δ, gal80Δ, LYS2::
GAL1UAS-GAL1TATA-HIS3,GAL2UAS-GAL2TATA-ADE2,
URA3::MEL1UAS-MEL1TATA-lacZ (Clontech) [5].
2. Bait plasmids: Gene of interest cloned in bait vector (e.g.,
pGBKT7g [17] or pDEST32 (Life Technologies)).
3. YPDA liquid medium: 10 g yeast extract, 20 g peptone. Bring
up to 880 ml with deionized water and autoclave. After sterili-
zation, let the medium cool to room temperature. Add 100 ml
of 20 % glucose stock solution and 20 ml of 2 mg/ml adenine
stock solution for a final volume of 1 L. Store at room
temperature.
4. YPDA solid medium: 10 g yeast extract, 20 g peptone, 20 g
agar. Bring up to 880 ml with deionized water and autoclave.
After sterilization, let the medium cool to 60 C. Add 100 ml of
20 % glucose stock solution and 20 ml of 2 mg/ml adenine
stock solution for a final volume of 1 L. Pour plates in a sterile
hood and let the medium solidify. Store plates at 4 C for 2–3
weeks.
5. 2 mg/ml Adenine stock solution: 200 mg adenine hemisulfate
salt. Bring up to 100 ml with deionized water and sterilize
through a 0.22 μm filter.
6. 20 % Glucose stock solution: Put 800 ml of ultrapure water in a
beaker and stir. Weigh 200 g α-D (+)-glucose (anhydrous) and
add it to the ultrapure water in small amounts to ensure that it is
completely dissolved. Autoclave.
7. Sterile water: autoclave ultrapure water.
8. 1 M LiOAc: 5.1 g lithium acetate dihydrate. Bring up to 50 ml
with ultrapure water and autoclave. Store at room temperature.
9. 100 mM LiOAc: 0.51 g lithium acetate dihydrate. Bring up to
50 ml with ultrapure water and autoclave.
10. 50 % (w/v) PEG3350: Add 50 g polyethylene glycol (average
molecular weight 3,350) to 50 ml of ultrapure water. Stir until
completely dissolved. Filter sterilize and store at room temper-
ature, securely capped to prevent evaporation. Water loss will
increase the PEG concentration and severely reduce the yield of
transformants. To avoid this, make fresh PEG3350 solution
every few months.
11. ssDNA: 10 mg/ml single-stranded fish sperm DNA, MB
grade. Store at 20 C in aliquots.
12. Selective liquid yeast medium: 6.9 g yeast nitrogen base without
amino acids. Bring up to 700 ml with ultrapure water and
Gal4p-Based Yeast Two-Hybrid 415
2.2 Bait Auto- 1. Yeast strain: S. cerevisiae AH109 containing the bait plasmid.
activation Test 2. Prey plasmids: empty prey vectors or prey vectors with an
independent protein inserted (e.g., if the bait is constructed
in pGBKT7g, use the empty pGADT7g vector as a control; if
the bait is constructed in pDEST32, use the empty pDEST22
as a control).
3. Solid media needed (25 ml medium/90 mm petri dish):
2 SD-Trp-Leu plates
1 SD-Trp-Leu-His plate
1 SD-Trp-Leu-His +1 mM 3-AT plate (25 μl 1 M 3-AT/25 ml
medium)
1 SD-Trp-Leu-His +3 mM 3-AT plate (75 μl 1 M 3-AT/25 ml
medium)
1 SD-Trp-Leu-His +5 mM 3-AT plate (125 μl 1 M 3-AT/
25 ml medium)
1 SD-Trp-Leu-His +10 mM 3-AT plate (250 μl 1 M 3-AT/
25 ml medium)
1 SD-Trp-Leu-His-Ade plate
2.3 Prey Library 1. Yeast strain: S. cerevisiae AH109 containing the bait plasmid.
Transformation 2. Prey library: library cloned in prey vector (e.g., pGADT7g [17]
or pDEST22 (Life Technologies)).
3. 2 YPDA liquid medium: 20 g yeast extract, 40 g peptone.
Bring up to 760 ml with deionized water and autoclave. After
sterilization, let the medium cool down. Add 200 ml of 20 %
glucose stock solution and 40 ml of 2 mg/ml adenine stock
solution for a final volume of 1 L.
4. Solid media needed (25 ml medium/90 mm petri dish; 80 ml
medium/150 mm petri dish):
12 SD-Trp-Leu 90 mm plates
30 SD-Trp-Leu-His 150 mm plates + x mM 3-AT (concentra-
tion determined from auto-activation test)
2.6 Identification 1. Competent Escherichia coli cells: such as One Shot TOP10
of Prey Inserts chemically competent E. coli (Life Technologies).
2. LB medium: 10 g tryptone, 5 g yeast extract, 10 g NaCl (15 g
agar). Bring up to 1 L with deionized water and autoclave. Let
cool before adding antibiotics.
3. Resuspension buffer: 6.06 g tris base, 3.72 g ethylenediamine-
tetraacetic acid disodium salt dihydrate (Na2EDTA 2H2O).
Add approximately 800 ml of deionized water and adjust pH
to 8. Bring up to 1 L with deionized water and autoclave. After
autoclaving, add DNase- and proteinase-free RNase A to a final
concentration of 100 μg/ml and store at 4 C.
4. Lysis buffer: 8 g NaOH, 10 g sodium dodecyl sulfate (SDS).
Bring up to 1 L with demineralized water. Filter sterilize and
store in a plastic bottle at room temperature. When stored too
cold, a white precipitate will appear. The precipitated SDS can
be dissolved again by microwaving.
5. Neutralization buffer: 294.5 g potassium acetate. Dissolve in
500 ml of deionized water and adjust pH to 5.5 with glacial
acetic acid (more than 100 ml will be needed). Bring up to 1 L
and autoclave. Store at room temperature.
6. 70 % ice cold ethanol: 70 ml ethanol, 30 ml deionized water.
Store at 20 C.
7. Sequencing primers: specific for the prey plasmids. E.g:
pDEST22_F: 50 -TATAACGCGTTTGGAATCACT-30
pDEST22_R: 50 -AGCCGACAACCTTGATTGGAGAC-30
pGADT7g_F: 50 -CTATTCGATGATGAAGATACCCCAC-
CAAACCC-30
pGADT7g_R: 50 -GTGAACTTGCGGGGTTTTTCAGTATC-
TACGATT-30
3 Methods
3.1 Bait Plasmid This protocol is adapted from the lithium acetate/single-stranded
Transformation carrier DNA/polyethylene glycol method by Gietz and Woods
[19], which permits high-efficiency transformation of yeast. This
method generally yields 105 transformants per μg of DNA.
1. Streak S. cerevisiae strain AH109 on a fresh YPDA agar plate
starting from a 25 % glycerol cell stock (stored at 80 C).
Incubate for 2–3 nights at 30 C (see Note 1).
2. Inoculate two 5 ml YPDA cultures (in 50 ml flasks) with one
big (3 mm) or two to four small (1–2 mm) colonies. Inoculate
different numbers of colonies for the two cultures to ensure
different cell densities the next day. Incubate overnight in a
shaking incubator at 30 C. The next morning, measure the
optical density at 600 nm (OD600). A preculture with a value
below 1 is preferred as these cells are still not stationary and will
have a shorter lag phase. Use this overnight culture to inoculate
a fresh yeast culture.
3. Inoculate 50 ml of YPDA (in a 500 ml flask) until an OD600
value of approximately 0.120–0.140 is achieved (see Note 2).
Incubate at 30 C until the OD600 reaches 0.480–0.560 (this
usually takes 4–5 h). At least two cell divisions are needed for a
good transformation efficiency. This efficiency remains con-
stant during the next three to four cell divisions.
Gal4p-Based Yeast Two-Hybrid 419
3.2 Bait Auto- This protocol also uses the LiOAc/ssDNA/PEG method. The
activation Test important differences are highlighted in bold.
1. Streak S. cerevisiae AH109 containing the bait plasmid
(obtained in Subheading 3.1) on a fresh selective agar plate.
Incubate for 2–3 nights at 30 C (see Note 8).
2. Inoculate two 5 ml SD medium cultures (in 50 ml flasks) with
one big (3 mm) or two to four small (1–2 mm) colonies.
Incubate overnight in a shaking incubator at 30 C. The next
morning, determine the OD600 and use the overnight culture
to inoculate a fresh yeast culture.
420 Jeroen Wagemans and Rob Lavigne
3. Steps 3–10 are exactly the same (see Note 9). In step 8, use
1 μg of empty prey vector or an independent prey construct to
transform the S. cerevisiae AH109 containing the bait plasmid.
4. After resuspension in 1 ml of sterile water, 10 μl and 100 μl are
plated on SD-Trp-Leu to check the transformation efficiency.
The remaining volume (890 μl) is divided between SD-Trp-
Leu-His +0, 1, 3, 5 and 10 mM 3-AT (100 μl on every plate)
and SD-Trp-Leu-His-Ade (the remaining volume) plates.
All plates are incubated at 30 C for 5 days.
5. Evaluation: a background of very small colonies, due to leaky
expression of the HIS3 reporter, can usually be observed. Only
large colonies on the SD-Trp-Leu-His plates (or maybe even
on the SD-Trp-Leu-His-Ade plate) are an indication of auto-
activation. Out of the different concentrations of 3-AT, choose
one concentration that will be used in the Y2H screen. This
concentration should not be too low to reduce the number of
false positive “interactions” but also not too high, to reduce the
chance of missing weaker interactions (see Note 10).
3.3 Prey Library 1. Streak S. cerevisiae AH109 containing the bait plasmid
Transformation (obtained in Subheading 3.1) on a fresh selective agar plate.
Incubate for 2–3 nights at 30 C (see Note 11).
2. Day 1: Inoculate the freshly streaked strain in 5 ml of selective
SD medium in a 50 ml flask (use several small colonies). Incu-
bate overnight at 30 C.
3. Day 2: Inoculate two 100 ml cultures of selective SD medium
in 1 L baffled flasks with the overnight culture (use two differ-
ent volumes: 1 and 2 ml) (see Note 12). The next day, one of
the 100 ml cultures should have an OD600 below 1 (still
exponentially growing, but preferably also close to 1 to have
enough inoculum for a 600 ml culture). Prewarm 600 ml
2 YPDA in a 2 L baffled flask at 30 C and autoclave two
additional empty 2 L baffled flasks and a 250 ml graduated
cylinder.
4. Day 3: Measure the OD600. Centrifuge the culture with the
value the closest to, but still below, 1 (divide between two
50 mL tubes) (3,500 g, 5 min). Discard the supernatant
and resuspend the cell pellets in 1 ml of 2 YPDA. Use this
concentrated cell solution to inoculate the 600 ml 2 YPDA
culture to an OD600 of approximately 0.120–0.140. Divide the
600 ml culture between the three 2 L baffled flasks using the
sterile graduated cylinder. Incubate the three cultures at 30 C
in a shaking incubator until the OD600 reaches 0.480–0.560.
This will take 4–5 h.
5. Divide the culture in twelve 50 ml sterile tubes. Centrifuge
(3,500 g, 5 min).
Gal4p-Based Yeast Two-Hybrid 421
3.4 Evaluation of 1. Check all 30 plates and select a maximum of 96 yeast colonies
Library Transformation to analyze (see Note 19).
2. Pick the selected colonies with a toothpick and transfer them to
a microtiter plate containing 100 μl of sterile water.
3. Shake for 15 min on a shaker to resuspend all cells.
422 Jeroen Wagemans and Rob Lavigne
3.6 Identification Because of its low yield and impurity, the isolated yeast prey plasmid
of Prey Inserts cannot be directly used as a template in DNA sequencing analysis.
To identify the prey inserts, the plasmid should first be transformed
back to E. coli. A direct PCR is not advisable, as there is still some
genomic yeast DNA present that may cause nonspecific products
and there can be more than one type of prey plasmid present.
1. For each blue colony, transform 5 μl of yeast plasmid DNA to
competent E. coli cells. Plate cells on LB medium selecting for
the prey plasmid.
2. For each transformation, four colonies (derived from one yeast
colony) are selected for further analysis (see Note 23). Grow
the yeast overnight in a 4 ml selective LB culture and isolate the
prey plasmid using a standard miniprep kit. In the case of a
larger number of yeast colonies, it is better to use a microtiter
plate scaled plasmid isolation method. Pick four colonies per
Gal4p-Based Yeast Two-Hybrid 423
3.7 One-on-One Y2H All the observed interactions could be false positive results and
Confirmation Test should be confirmed using an independent Y2H experiment.
Choose one prey construct (the plasmid isolated from E. coli in
Subheading 3.6) for each different potential interaction partner.
These prey constructs will be transformed to the α-mating type
yeast strain Y187 [18] and will be mated with an AH109 strain
containing the bait construct and another strain containing an
empty bait vector as a negative control. An independent protein
inserted in the bait vector could also be used. If there are not too
many different prey plasmids, the same protocol provided in Sub-
heading 3.1 can easily be followed to transform the plasmids into
S. cerevisiae Y187. After transformation, one can proceed with
step 15 from this protocol. In the case of a higher number of
different preys or different Y2H screens confirmation tests com-
bined in one experiment, the protocol given in this section, which is
adapted for up to 96 yeast transformations in parallel, can be
followed.
1. Streak S. cerevisiae Y187 on a fresh YPDA agar plate starting
from a 25 % glycerol cell stock (stored at 80 C). Incubate for
2–3 nights at 30 C.
2. Inoculate two tubes of 4 ml of YPDA with several small colo-
nies. Shake overnight at 30 C.
3. Dilute the overnight culture with an OD600 value below 1 in
100 ml of YPDA (in a 1 L baffled flask) until an OD600 of
0.120–0.140. Incubate at 30 C until the OD600 reaches
0.480–0.560.
4. Divide the culture between two conical tubes and centrifuge
(3,500 g, 5 min).
5. Discard the medium and resuspend the cells in 2 25 ml
sterile water. Centrifuge again.
6. Discard the supernatant and resuspend the cells in 2 1 ml of
100 mM LiOAc. Transfer to 1.5 ml microcentrifuge tubes.
7. Pellet the cells (14,000 g, 15 s) and remove the LiOAc with a
micropipette. Resuspend each cell pellet in 400 μl of 100 mM
LiOAc. The final volume is now around 2 500 μl. This is
enough for 96 small-scale transformations. Centrifuge
(14,000 g, 15 s) and remove the LiOAc.
8. Resuspend the two pellets in 1,480 μl of sterile water and
transfer to a 15 ml conical tube. The transformation mixture
is now added in the following order to the cells:
4,800 μl PEG3350 (50 % w/v)
Gal4p-Based Yeast Two-Hybrid 425
720 μl 1 M LiOAc
200 μl ssDNA (10 mg/ml)
9. Vortex for 10 min. Prepare a PCR plate by adding to each well
100 ng of plasmid DNA (a different plasmid per well).
10. Divide the transformation mixture in the PCR plate by adding
25 μl to each well. Pipette up and down a few times to mix.
Place a lid on the plate during incubation (see Note 26).
11. Incubate for 10 min at 30 C followed by 30 min at 42 C to
heat shock the cells. This step can be done on a PCR block.
12. Centrifuge the plate at 3,500 g for 1 min and discard the
supernatant by pipetting. Resuspend the cells in 100 μl of
sterile water.
13. Plate the entire cell mixture on selective SD medium and
incubate at 30 C for 3 nights (see Note 27).
14. On the same day as step 13, freshly streak the different AH109
strains on selective agar plates. Incubate for 3 nights at 30 C.
15. Fill a microtiter plate with 50 μl of sterile water and resuspend
the different S. cerevisiae Y187 and AH109 strains in different
wells (pick several colonies).
16. Mating: spot 2 μl a-yeast on a YPDA agar plate. Let dry. Spot
2 μl α-yeast on the same position as the a-yeast. Do this for all
different combinations (e.g., each prey in S. cerevisiae Y187 is
mated with S. cerevisiae AH109 containing the bait construct
and S. cerevisiae AH109 containing the empty bait vector).
There will be two positions for each prey which has to be
tested. Incubate overnight at 30 C.
17. The next day, mating has occurred. To select for diploid cells,
pick some cell material from every position in 50 μl of sterile
water and spot the different wells (2 μl) on SD-Trp-Leu. Incu-
bate for two nights at 30 C.
18. Resuspend the diploid cells again in 50 μl of sterile water and
spot 2 μl on SD-Trp-Leu-His + x mM 3-AT. Incubate for 5–7
nights at 30 C.
19. Pick the different cells in 50 μl of sterile water and spot 2 μl on
SD-Trp-Leu-His-Ade +40 mg/L X-α-gal. Incubate for 5–7
nights at 30 C.
20. Evaluation: those prey plasmids that only generate colonies
with a halo for the bait-prey combination but not when com-
bined with the empty bait vector are confirmed interactions.
4 Notes
Acknowledgements
References
1. Fields S, Song OK (1989) A novel genetic 8. Buchert M, Schneider S, Adams MT et al
system to detect protein protein interactions. (1997) Useful vectors for the two-hybrid sys-
Nature 340:245–246 tem in mammalian cells. Biotechniques
2. Keegan L, Gill G, Ptashne M (1986) Separa- 23:396–398, 400, 402
tion of DNA binding from the transcription- 9. Johnsson N, Varshavsky A (1994) Split ubiqui-
activating function of a eukaryotic regulatory tin as a sensor of protein interactions in vivo.
protein. Science 231:699–704 Proc Natl Acad Sci U S A 91:10340–10344
3. Chien CT, Bartel PL, Sternglanz R et al (1991) 10. Brachmann RK, Boeke JD (1997) Tag games
The two-hybrid system: a method to identify in yeast: the two-hybrid system and beyond.
and clone genes for proteins that interact with a Curr Opin Biotechnol 8:561–568
protein of interest. Proc Natl Acad Sci U S A 11. Uetz P (2002) Two-hybrid arrays. Curr Opin
88:9578–9582 Chem Biol 6:57–62
4. Van Criekinge W, Beyaert R (1999) Yeast two- 12. Golemis EA, Khazak V (1997) Alternative
hybrid: state of the art. Biol Proced Online yeast two-hybrid systems. The interaction trap
2:1–38 and interaction mating. Methods Mol Biol
5. James P, Halladay J, Craig EA (1996) Genomic 63:197–218
libraries and a host strain designed for highly 13. Serebriiskii I, Khazak V, Golemis EA (1999) A
efficient two-hybrid selection in yeast. Genetics two-hybrid dual bait system to discriminate
144:1425–1436 specificity of protein interactions. J Biol Chem
6. Durfee T, Becherer K, Chen PL et al (1993) 274:17080–17087
The retinoblastoma protein associates with the 14. Le Douarin B, Pierrat B, vom Baur E et al
protein phosphatase type 1 catalytic subunit. (1995) A new version of the two-hybrid assay
Genes Dev 7:555–569 for detection of protein-protein interactions.
7. Hu JC, Kornacker MG, Hochschild A (2000) Nucleic Acids Res 23:876–878
Escherichia coli one- and two-hybrid systems 15. Caufield JH, Sakhawalkar N, Uetz P (2012) A
for the analysis and identification of protein- comparison and optimization of yeast two-
protein interactions. Methods 20:80–94 hybrid systems. Methods 58:317–324
Gal4p-Based Yeast Two-Hybrid 431
16. Chen YC, Rajagopala SV, Stellberger T et al 20. Lazo PS, Ochoa AG, Gascon S (1978) alpha-
(2010) Exhaustive benchmarking of the yeast Galactosidase (melibiase) from Saccharomyces
two-hybrid system. Nat Methods 7:667–668, carlsbergensis: structural and kinetic properties.
author reply 668 Arch Biochem Biophys 191:316–324
17. Uetz P, Dong YA, Zeretzke C et al (2006) 21. Clarke L, Carbon J (1976) A colony bank con-
Herpesviral protein networks and their interac- taining synthetic Col El hybrid plasmids repre-
tion with the human proteome. Science sentative of the entire E. coli genome. Cell
311:239–242 9:91–99
18. Harper JW, Adami GR, Wei N et al (1993) The 22. Blattner FR, Plunkett G 3rd, Bloch CA et al
p21 Cdk-interacting protein Cip1 is a potent (1997) The complete genome sequence of
inhibitor of G1 cyclin-dependent kinases. Cell Escherichia coli K-12. Science 277:1453–1462
75:805–816 23. Clontech Yeast Protocols Handbook. Clontech
19. Gietz RD, Woods RA (2002) Transformation Laboratories, Inc. www.clontech.com. Proto-
of yeast by lithium acetate/single-stranded car- col No. PT3024-1. 2. Version No. PR973283
rier DNA/polyethylene glycol method. Meth-
ods Enzymol 350:87–96
Chapter 28
Abstract
Use of the yeast two-hybrid system has provided definition to many previously uncharacterized pathways
through the identification and characterization of novel protein-protein interactions. The two-hybrid
system uses the bifunctional nature of transcription factors, such as the yeast enhancer Gal4, to allow
protein-protein interactions to be monitored through changes in transcription of reporter genes. Once a
positive interaction has been identified, either of the interacting proteins can be mutated by site-specific or
randomly introduced changes, to produce proteins with a decreased ability to interact. Mutants generated
using this strategy are very powerful reagents in tests of the biological significance of the interaction and in
defining the residues involved in the interaction. Such techniques are termed reverse two-hybrid methods.
We describe a reverse two-hybrid method that generates loss-of-interaction mutations of the catalytic
subunit of the Escherichia coli heat-labile toxin (LTA1) with decreased binding to the active
(GTP-bound) form of human ARF3, its protein cofactor. While newer methods are emerging for
performing interaction screens in mammalian cells, instead of yeast, the use of reverse two-hybrid in yeast
remains a robust and powerful means of identifying loss-of-interaction point mutants and compensating
changes that remain among the most powerful tools of testing the biological significance of a protein-
protein interaction.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_28, © Springer Science+Business Media New York 2015
433
434 Matthew A. Bennett et al.
Fig. 1 Expression of reporter genes (e.g., LacZ) in two-hybrid systems is dependent upon the interaction of two
fusion proteins that can be lost as a result of a point mutation. The Gal4 protein, two interacting proteins of
interest, and resulting fusion proteins are shown pictorially on the left. An “x” in a protein represents a point
mutation. Interaction between the two fusion proteins generates a transactivator capable of promoting
transcription of reporter genes (LacZ) downstream of the Gal4 upstream activating sequence (UAS). Point
mutations in either fusion protein that decrease binding result in decreases in or elimination of reporter gene
expression. The level of expression is shown on the right as the amount of β-galactosidase (β-gal) activity
hybrid have emerged over the years that offer greater versatility,
depending on needs [19–21]. These approaches are particularly
attractive when one of the proteins under study binds multiple
protein partners as mutagenesis and two-hybrid screens can identify
specific loss of binding mutants that retain binding to other part-
ners. In this chapter, we describe a reverse two-hybrid method for
generating mutations of the catalytic subunit of the Escherichia coli
heat-labile toxin (LTA1) with reduced binding to the activated
(GTP bound) form of human ARF3, [Q71L]ARF3 [22, 23].
As seen here and in other studies (e.g., Das et al. [24]), the use of
reverse two-hybrid is an attractive means of testing the relevance of
protein interactions in pathogen biology.
The main features of this approach include PCR-based random
mutagenesis over a defined region of any coding region, and iden-
tification of full-length mutants, expressed to the same levels as
wild-type protein by immunoblot analysis using an epitope tag on
the fusion proteins. The conditions used here have yielded approxi-
mately one change per 250 bp amplified. PCR allows the extent of
mutagenesis to be controlled by modifying the concentration of
nucleotides or manganese in the PCR reaction [25]. Although the
use of selectable markers fused to the C-terminus of the mutated
protein allows for genetic selection of full-length proteins [26–28],
which is important to high-throughput screens and can speed up
this rate limiting step in reverse two-hybrid screens, we prefer the
use of immunoblots to allow estimates of protein expression as well
as confirmation that the protein is full-length. Using the methods
described below we have been able to generate hundreds of colo-
nies with reduced β-galactosidase activity in less than a week and
typically 25–50 % of those were found to result from point muta-
tions in full-length proteins, suitable for further analysis. Thus, tens
of mutants can be obtained, their interactions with multiple effec-
tors characterized, and sequences determined in less than 1 month.
The loss of an interaction is limited in interpretations due to the
negative nature of the data. However, loss of one binding partner
with retention of others provides a powerful probe of activities in
live cells. In addition, after the generation of loss-of-interaction
mutants in one protein it is even easier to then mutate the other
binding partner and screen for mutants that regain the interaction.
Such pairs of mutants can provide a formal proof of specific func-
tionalities in live cells. Such mutants may have the added benefit of
not binding the wild-type protein and can allow cell studies to be
carried out independently of interference or complications pre-
sented by the presence of endogenous proteins. As with any tech-
nique, the data and conclusions from the use of reverse two-hybrid
techniques are strengthened by confirmation using independent
methods.
436 Matthew A. Bennett et al.
2 Materials
Fig. 2 Reverse two-hybrid screens use random PCR mutagenesis and gap repair
in yeast to generate a large library of mutants, ready for screening. Amplification
of the coding region of LTA1 in pXJ12 is performed with primers that anneal
50 bp outside of the region targeted for mutagenesis to yield a product with
ends homologous to those in the gapped plasmid, shown below. The error rate of
the polymerase is increased by carrying out the PCR under conditions of reduced
stringency. Note that any region of DNA can be targeted for mutagenesis. Co-
transformation of the mutated PCR product with gapped pACT2 plasmid,
prepared by restriction digestion, into yeast allows for repair of the plasmid by
homologous recombination. When transformed into a yeast strain expressing the
[Q71L]ARF3-BD fusion proteins, the resulting transformants can be assayed
directly for loss of interaction using the colony β-galactosidase assay
3 Methods
3.1 Generation This method exploits the lack of proofreading by Taq polymerase
of Mutant Substrates and ability to replicate DNA with low fidelity and generate random
for Plasmids changes into any region of DNA, in this case the LTA1-encoding
portion of the AD-LTA1 plasmid (see Fig. 2 and Note 2).
3.1.2 Gapped Plasmid Create the gapped plasmid by digesting pXJ12 with NcoI and
(pACT2) Production XhoI (see Note 3). Purify the resulting linearized, 8 kb vector back-
bone from a 1 % agarose gel using a commercially available gel
extraction kit.
3.1.3 Estimating Vector Estimate the concentration of the purified, gapped (pACT2-
and PCR Product derived) plasmid and PCR products by comparing the ethidium
Concentration bromide fluorescence intensity from each sample in a 1 % agarose
gel to that of a known amount of a λ HindIII DNA marker of
similar size.
3.2 Co- The insertion of the mutagenized open reading frame into the
transformation of gapped plasmid can be accomplished by taking advantage of the
Gapped Plasmid and high level of homologous recombination in yeast cells.
PCR Product Co-transformation of yeast with gapped plasmid and PCR products
that contain regions of identity of 50 bp at each end is approxi-
mately 50 % as efficient as transformation with the circular plasmid
alone (see Fig. 2 and Note 4).
1. Inoculate 50 mL of SD trp with YAB457 and grow at 30 C
to OD600 ~ 0.6.
440 Matthew A. Bennett et al.
3.3 Assaying The X-gal filter assay is used to measure levels of β-galactosidase
Transformants for activity in yeast colonies as an indicator of protein interactions [33].
Loss-of-Interaction The two fusion proteins (often referred to as bait and prey) interact,
thereby activating transcription of the lacZ gene and production of
β-galactosidase, which catalyzes the hydrolysis of X-gal to yield a
blue product. Both the time of incubation and the darkness of the
blue product formed on the filter should be monitored and com-
pared to the parent plasmid to allow initial determination of loss-of-
interaction mutations.
1. Place a nitrocellulose filter on each plate for 30 s (or until wet)
to replica the yeast colonies onto the filter.
2. Drop the filter into liquid nitrogen and carefully remove after
20 s. Avoid breaking the filters that will be brittle while frozen.
3. Transfer the frozen nitrocellulose filter, with the yeast side
facing up, to the lid of a 100 mm dish on which has been placed
Whatman filter paper wetted with X-gal in Z buffer (1.5 mL of
Z-buffer with 15 μL of 100 mg/mL X-gal solution to yield a
final X-gal concentration of 1 mg/mL).
Reverse Two-Hybrid 441
3.4 Recovering Loss- Recover plasmids from yeast using a modification of the standard
of-Interaction Mutant glass bead method [34], also known as a “smash and grab” prepa-
Plasmids and ration. The modification simply adds the use of commercial plasmid
Retesting purification columns to further remove contaminants that inhibit
transformation. The use of plasmid purification columns is not
required or used in all laboratories but we have found it increases
success rates, sometimes markedly.
1. Streak colonies with diminished β-galactosidase activity onto SD
leu cycloheximide plates to select for loss of the [Q71L]ARF3-
BD expressing plasmid. This step is not required but often facil-
itates the isolation of the desired (AD-LTA1 mutant) plasmid
from strains carrying two different plasmids (see Note 6).
2. Incubate the plates for 3 days at 28 C.
3. Inoculate 20 mL of SD leu media with colonies from the SD
leu cycloheximide plates and grow each culture to an OD600
nm ~ 1.
4. Collect cells by centrifugation at 1,000 g for 10 min.
5. Wash each pellet in 1 mL of STES buffer and collect again.
6. Resuspend in 100 μL of STES and add an equal volume of glass
beads (see Note 7).
7. Vortex each tube for 2 min.
8. Add 100 μL of STES and 200 μL phenol–chloroform and
vortex for 5–30 min. Times will vary with the vortex used.
9. Pellet cells and debris and separate phases in a microfuge at
maximal speed (approximately 14,000 g) for 10 min, and
transfer the aqueous phase to a new tube.
442 Matthew A. Bennett et al.
3.5 Characterizing Multiple factors can be responsible for a loss of interaction, some of
the Noninteracting which are far less informative than a point mutation in a critical
Proteins amino acid residue. Two types of uninteresting changes, premature
stop codons and decreased levels of protein expression, can be
screened for by immunoblotting cell lysates with an HA antibody
to determine the presence and relative abundance of the epitope tag
in the LTA1-AD fusion proteins. The levels of protein expressed in
each strain expressing mutant LTA1 should be compared to wild-
type LTA1 (see Note 9). Protein preparations from total yeast cell
lysates can be obtained using a modification of Horvath et al. [35].
1. Grow five OD of each yeast strain in selective liquid medium.
2. Collect cells by centrifugation at 1,000 g for 5 min.
3. Wash cells with water and collect again at 1,000 g for 5 min.
4. Resuspend each pellet in 20 μL of 1 SDS sample buffer and
boil at 95 C for 5 min.
5. Add one cell volume of acid-washed glass beads and vortex
3 min at maximum speed.
6. Add 120 μL sample buffer and boil for 5 min.
7. Pellet cells and debris by centrifugation in a microfuge for
15 min at maximal setting. Transfer supernatant to another
tube and boil for 5 min.
8. Load 7.5 μL of the sample on a 12 % SDS gel and resolve
proteins using standard methods.
9. Transfer the resolved proteins to nitrocellulose at 60 V for 2 h
and immunoblot with the 12CA5 (HA) antibody using stan-
dard methods.
Reverse Two-Hybrid 443
4 Notes
References
1. Fields S, Song O (1989) A novel genetic system 9. Maier RH, Maier CJ, Hintner H et al (2012)
to detect protein-protein interactions. Nature Quantitative real-time PCR as a sensitive
340:245–246 protein-protein interaction quantification
2. Chien CT, Bartel PL, Sternglanz R et al (1991) method and a partial solution for non-
The two-hybrid system: a method to identify accessible autoactivator and false-negative mol-
and clone genes for proteins that interact with a ecule analysis in the yeast two-hybrid system.
protein of interest. Proc Natl Acad Sci U S A Methods 58:376–384
88:9578–9582 10. Pellet J, Meyniel L, Vidalain PO et al (2009)
3. Boman AL, Zhang C, Zhu X et al (2000) A pISTil: a pipeline for yeast two-hybrid Interac-
family of ADP-ribosylation factor effectors that tion Sequence Tags identification and analysis.
can alter membrane transport through the BMC Res Notes 2:220
trans-Golgi. Mol Biol Cell 11:1241–1255 11. Rajagopala SV, Uetz P (2009) Analysis of
4. Van Valkenburgh H, Shern JF, Sharer JD et al protein-protein interactions using array-based
(2001) ADP-ribosylation factors (ARFs) and yeast two-hybrid screens. Methods Mol Biol
ARF-like 1 (ARL1) have both specific and 548:223–245
shared effectors: characterizing ARL1-binding 12. Ratushny V, Golemis E (2008) Resolving the
proteins. J Biol Chem 276:22826–22837 network of cell signaling pathways using the
5. Ito T, Chiba T, Ozawa R et al (2001) A com- evolving yeast two-hybrid system. Biotechni-
prehensive two-hybrid analysis to explore the ques 44:655–662
yeast protein interactome. Proc Natl Acad Sci 13. Cagney G, Uetz P (2001) High-throughput
U S A 98:4569–4574 screening for protein-protein interactions
6. Ito T, Tashiro K, Muta S et al (2000) Toward a using yeast two-hybrid arrays. Curr Protoc
protein-protein interaction map of the bud- Protein Sci Chapter 19:Unit 19 16.
ding yeast: a comprehensive system to examine 14. Parrish JR, Gulyas KD, Finley RL Jr (2006)
two-hybrid interactions in all possible combi- Yeast two-hybrid contributions to interactome
nations between the yeast proteins. Proc Natl mapping. Curr Opin Biotechnol 17:387–393
Acad Sci U S A 97:1143–1147 15. Vidalain PO, Boxem M, Ge H et al (2004)
7. Uetz P (2002) Two-hybrid arrays. Curr Opin Increasing specificity in high-throughput yeast
Chem Biol 6:57–62 two-hybrid experiments. Methods
8. Uetz P, Giot L, Cagney G et al (2000) A com- 32:363–370
prehensive analysis of protein-protein interac- 16. Legrain P, Selig L (2000) Genome-wide pro-
tions in Saccharomyces cerevisiae [see tein interaction maps using two-hybrid sys-
comments]. Nature 403:623–627 tems. FEBS Lett 480:32–36
446 Matthew A. Bennett et al.
17. Walhout AJ, Boulton SJ, Vidal M (2000) Yeast 29. Durfee T, Becherer K, Chen PL et al (1993)
two-hybrid systems and protein interaction The retinoblastoma protein associates with the
mapping projects for yeast and worm. Yeast protein phosphatase type 1 catalytic subunit.
17:88–94 Genes Dev 7:555–569
18. Chappell TG, Gray PN (2008) Protein interac- 30. Boman AL, Kuai J, Zhu X et al (1999) Arf
tions: analysis using allele libraries. Adv Bio- proteins bind to mitotic kinesin-like protein 1
chem Eng Biotechnol 110:47–66 (MKLP1) in a GTP-dependent fashion. Cell
19. Endoh H, Vincent S, Jacob Y et al (2002) Motil Cytoskeleton 44:119–132
Integrated version of reverse two-hybrid sys- 31. Kuai J, Kahn RA (2000) Residues forming a
tem for the postproteomic era. Methods Enzy- hydrophobic pocket in ARF3 are determinants
mol 350:525–545 of GDP dissociation and effector interactions.
20. Endoh H, Walhout AJ, Vidal M (2000) A FEBS Lett 487:252–256
green fluorescent protein-based reverse two- 32. Muhlrad D, Hunter R, Parker R (1992) A
hybrid system: application to the characteriza- rapid method for localized mutagenesis of
tion of large numbers of potential protein- yeast genes. Yeast 8:79–82
protein interactions. Methods Enzymol 33. Bai C, Elledge SJ (1996) Gene identification
328:74–88 using the yeast two-hybrid system. Methods
21. Vidal M, Endoh H (1999) Prospects for drug Enzymol 273:331–347
screening using the reverse two-hybrid system. 34. Rose MD, Winston F, Hieter P (1990) In:
Trends Biotechnol 17:374–381 Rose MD, Winston F, Hieter P (eds) Labora-
22. Zhu X, Kahn RA (2001) The Escherichia coli tory course manual for methods in yeast genet-
heat labile toxin binds to Golgi membranes and ics. Cold Spring Harbor Laboratory, Cold
alters Golgi and cell morphologies using ADP- Spring Harbor, NY
ribosylation factor-dependent processes. J Biol 35. Horvath A, Riezman H (1994) Rapid protein
Chem 276:25014–25021 extraction from Saccharomyces cerevisiae. Yeast
23. Zhu X, Kim E, Boman AL et al (2001) ARF 10:1305–1310
binds the C-terminal region of the Escherichia 36. Bartel P, Chien CT, Sternglanz R et al (1993)
coli heat-labile toxin (LTA1) and competes for Elimination of false positives that arise in using
the binding of LTA2. Biochemistry the two-hybrid system. Biotechniques
40:4560–4568 14:920–924
24. Das S, Kalpana GV (2009) Reverse two-hybrid 37. James P, Halladay J, Craig EA (1996) Genomic
screening to analyze protein-protein interac- libraries and a host strain designed for highly
tion of HIV-1 viral and cellular proteins. Meth- efficient two-hybrid selection in yeast. Genetics
ods Mol Biol 485:271–293 144:1425–1436
25. Cadwell RC, Joyce GF (1992) Randomization 38. Brent R, Finley RL Jr (1997) Understanding
of genes by PCR mutagenesis. PCR Methods gene and allele function with two-hybrid meth-
Appl 2:28–33 ods. Annu Rev Genet 31:663–704
26. Leanna CA, Hannink M (1996) The reverse 39. Vidal M, Legrain P (1999) Yeast forward and
two-hybrid system: a genetic scheme for selec- reverse ‘n’-hybrid systems. Nucleic Acids Res
tion against specific protein/protein interac- 27:919–929
tions. Nucleic Acids Res 24:3341–3347 40. Shafikhani S, Siegel RA, Ferrari E et al (1997)
27. Puthalakath H, Strasser A, Huang DC (2001) Generation of large libraries of random
Rapid selection against truncation mutants in mutants in Bacillus subtilis by PCR-based plas-
yeast reverse two-hybrid screens. Biotechni- mid multimerization. Biotechniques
ques 30:984–988 23:304–310
28. Vidal M, Brachmann RK, Fattaey A et al 41. Guarente L (1983) Yeast promoters and lacZ
(1996) Reverse two-hybrid and one-hybrid fusions designed to study expression of cloned
systems to detect dissociation of protein- genes in yeast. Methods Enzymol
protein and DNA-protein interactions. Proc 101:181–191
Natl Acad Sci U S A 93:10315–10320
Chapter 29
Abstract
MAPPIT (MAmmalian Protein-Protein Interaction Trap) is a two-hybrid technology that facilitates the
detection and analysis of interactions between proteins in living mammalian cells. The system is based on
type 1 cytokine receptor signaling. The bait protein of interest is fused to a chimeric signaling-deficient
cytokine receptor, the signaling competence of which is restored upon recruitment of a prey protein that is
coupled to a functional cytokine receptor domain. MAPPIT exhibits an excellent signal-to-noise ratio,
detects a wide variety of protein-protein interactions (PPIs) including transient and indirect interactions,
and has been shown to be highly complementary to other two-hybrid methods with respect to the
interactions it can detect. Variants of the method were developed to allow large-scale PPI screening,
mapping of protein interaction interfaces, PPI inhibitor screening and drug profiling. This chapter
describes a basic 4-day MAPPIT protocol for the analysis of interaction between two designated proteins.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_29, © Springer Science+Business Media New York 2015
447
448 Irma Lemmens et al.
EpoR
Cellular membrane
JAK JAK
LepR F F
F F
F F
P
P B
B B
B P STAT3
Nuclear membrane
induction of STAT3
responsive reporter gene
2 Materials
2.2 Cell Culture 1. Human embryonic kidney (Hek) 293 T cell line.
2. Growth medium: Dulbecco’s modified Eagle’s medium
(DMEM) supplemented with 10 % Fetal Calf Serum and anti-
biotics like gentamycin or penicillin/streptomycin.
3. Human Epo.
3 Methods
3.1 Cloning Bait and 1. Design primers flanking your gene of interest that
Prey of Interest incorporate relevant restriction sites to clone the desired bait
in the pSEL(+2L) plasmid and the prey in the pMG1 construct.
Suitable restriction sites to clone the bait in the pSEL(+2L)
vector are SalI or SacI in combination with NotI or XbaI. The
prey can be cloned in the pMG1 vector using EcoRI combined
with NotI or XbaI (see Notes 1 and 2). Both plasmids also exist
in a gateway recombinatorial cloning compatible version,
allowing the transfer of bait and prey using an LR clonase
reaction (Invitrogen).
2. Perform a Polymerase Chain Reaction using a polymerase with
proofreading activity (e.g., Pfu DNA polymerase).
3. Use traditional cloning methods or gateway recombination to
insert the genes of interest into the appropriate vectors (Fig. 2).
4. Transform the resulting plasmids into a suitable bacterial line.
5. Prepare plasmid DNA of transfection-suitable quality, and vali-
date the plasmids by restriction digest or sequencing.
Fig. 2 Overview of the MAPPIT bait and prey plasmids. Plasmid pSEL(+2L) is
used for cloning the bait; plasmid pMG1 to fuse the prey C-terminally to the
gp130 moiety. A hinge (amino acids Gly–Gly–Ser) is placed between the fusions
for extra flexibility. EpoR, erythropoietin receptor; +2L, two extra leucines were
added in the transmembrane domain to optimize the fusion between the
extracellular domain of the EpoR and the cytoplasmic tail of the LepR. LepRF3,
leptin receptor variant with 3 tyrosines mutated to phenylalanines that lacks a
functional STAT3 recruitment site; gp130, glycoprotein 130; SV40, simian virus
40 early promoter; SRα, promoter comprising SV40 and the R-U5 segment of
human T-cell leukemia virus type 1 long terminal repeat; GGS, glycine–glyci-
ne–serine; pA, SV40 polyadenylation signal
452 Irma Lemmens et al.
3.2 Seeding Cells 1. For each bait–prey combination to be tested seed 1 104
subconfluent Hek293T cells in six wells of a black 96-well
plate in 100 μl growth medium per well. Cell lines other than
Hek293T cells can be used (see Notes 3 and 4).
Besides testing the bait–prey interaction the following con-
trols should be included: a bait–irrelevant prey and irrelevant
bait–prey combination to control for a possible background
caused by the bait or prey, respectively. Additionally, a prey (like
SH2beta) that can interact with the bait receptor independent
of the bait can serve as an additional control to check the
signaling capacity of the bait receptor.
2. Grow overnight in a humidified atmosphere at 37 C and 5–8 %
CO2.
3.3 Transient 1. For each bait–prey combination, make a DNA and CaCl2
Ca3(PO4)2 Transfection mixture containing 250 ng of the bait plasmid, 500 ng of the
prey plasmid, and 50 ng of the reporter (pXP2d2-rPAP1-luci)
with 5 μl of sterile 2.5 M CaCl2 in a total volume of 50 μl of
distilled water in a 96-well plate (see Notes 5 and 6).
2. Add 50 μl of 2 HeBS to each well containing the DNA and
CaCl2 mixtures.
3. Shake the plate for 1 min at 800 rpm.
4. Resuspend the DNA, CaCl2 and 2 HeBS mixture.
5. Add 10 μl to the cells in the 96-well plate in sextuple for every
bait–prey combination.
6. Incubate the cells overnight in a humidified atmosphere at
37 C and 5–8 % CO2.
The precipitate can be checked under a microscope: the parti-
cles should look like small speckles to obtain optimal transfection
efficiencies (see Note 7). Alternative commercially available trans-
fection reagents like lipofectamine can be used as well.
3.4 Stimulation of 1. For each bait–prey combination, stimulate three of the six wells
the Cells with 50 μl Epo at a final concentration of 5 ng/μl. To the
remaining three wells 50 μl of growth medium (without cyto-
kine) should be added.
2. Incubate the cells overnight in a humidified atmosphere at
37 C and 5–8 % CO2.
3.5 Measurement of 1. Remove growth medium from the black 96-well plates.
the Luciferase Activity 2. Add 50 μl of luciferase lysis buffer to each well and incubate for
10 min at room temperature.
3. Add 35 μl of luciferase substrate buffer to each well and imme-
diately measure the luminescent signals using a chemilumines-
cence reader suitable for 96-well format.
MAPPIT, In-Cell PPI Detection 453
3.6 Data Analysis 1. Calculate the fold induction value for each bait–prey
combination by dividing the average of the values of the sti-
mulated wells by the average of the values of the non-
stimulated wells.
2. The bait–prey interaction is scored positive if the fold induction
value obtained by the bait–prey combination is at least three
times higher than both the fold induction value of the
bait–irrelevant prey and irrelevant bait–prey interactions.
4 Notes
Acknowledgements
References
1. Eyckerman S, Verhee A, Van der Heyden J et al 9. Pattyn E, Lavens D, Van der Heyden J et al
(2001) Design and application of a cytokine- (2008) MAPPIT (MAmmalian Protein-
receptor-based interaction trap. Nat Cell Biol Protein Interaction Trap) as a tool to study
3:1114–1119 HIV reverse transcriptase dimerization in intact
2. Montoye T, Lemmens I, Catteeuw D et al human cells. J Virol Methods 153:7–15
(2005) A systematic scan of interactions with 10. Ulrichts P, Peelman F, Beyaert R et al (2007)
tyrosine motifs in the erythropoietin receptor MAPPIT analysis of TLR adaptor complexes.
using a mammalian 2-hybrid approach. Blood FEBS Lett 581:629–636
105:4264–4271 11. Braun P, Tasan M, Dreze M et al (2009) An
3. Uyttendaele I, Lemmens I, Verhee A et al experimentally derived confidence score
(2007) Mammalian protein-protein interaction for binary protein-protein interactions. Nat
trap (MAPPIT) analysis of STAT5, CIS, Methods 6:91–97
and SOCS2 interactions with the growth 12. Boxem M, Maliga Z, Klitgord N et al (2008) A
hormone receptor. Mol Endocrinol 21: protein domain-based interactome network for
2821–2831 C. elegans early embryogenesis. Cell
4. Lavens D, Montoye T, Piessevaux J et al (2006) 134:534–545
A complex interaction pattern of CIS and 13. Simonis N, Rual JF, Carvunis AR et al (2009)
SOCS2 with the leptin receptor. J Cell Sci Empirically controlled mapping of the Caenor-
119:2214–2224 habditis elegans protein-protein interactome
5. Lavens D, Ulrichts P, Catteeuw D et al (2007) network. Nat Methods 6:47–54
The C-terminus of CIS defines its interaction 14. Simonis N, Rual JF, Lemmens I et al (2012)
pattern. Biochem J 401:257–267 Host-pathogen interactome mapping for
6. Piessevaux J, Lavens D, Montoye T et al (2006) HTLV-1 and -2 retroviruses. Retrovirology
Functional cross-modulation between SOCS 9:26
proteins can stimulate cytokine signaling. 15. Venkatesan K, Rual JF, Vazquez A et al (2009)
J Biol Chem 281:32953–32966 An empirical framework for binary interactome
7. Piessevaux J, De Ceuninck L, Catteeuw D et al mapping. Nat Methods 6:83–90
(2008) Elongin B/C recruitment regulates 16. Yu H, Braun P, Yildirim MA et al (2008) High-
substrate binding by CIS. J Biol Chem quality binary protein interaction map of the
283:21334–21346 yeast interactome network. Science
8. Ulrichts P, Tavernier J (2008) MAPPIT analy- 322:104–110
sis of early Toll-like receptor signalling events. 17. Bovijn C, Ulrichts P, De Smet AS et al (2012)
Immunol Lett 116:141–148 Identification of interaction sites for
MAPPIT, In-Cell PPI Detection 455
Abstract
Bioluminescence resonance energy transfer (BRET) is a valuable tool to detect protein-protein interactions.
BRET utilizes bioluminescent and fluorescent protein tags with compatible emission and excitation proper-
ties, making it possible to examine resonance energy transfer when the tags are in close proximity (<10 nm)
as a typical result of protein-protein interactions. Here we describe a protocol for detecting BRET from two
known protein binding partners (Gαi1 and RGS14) in HEK 293 cells using Renilla luciferase and yellow
fluorescent protein tags. We discuss the calculation of the acceptor/donor ratio as well as net BRET and
demonstrate that BRET can be used as a platform to investigate the regulation of protein-protein interac-
tions in live cells in real time.
Key words Bioluminescence resonance energy transfer (BRET), Renilla luciferase (RLuc), Yellow
fluorescent protein (YFP), RGS14, G protein regulatory (GPR) motif, G protein
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_30, © Springer Science+Business Media New York 2015
457
458 Nicole E. Brown et al.
Fig. 1 BRET is dependent on the distance between the donor luciferase and the
acceptor fluorophore. Addition of the cell permeant Renilla luciferase substrate
coelenterazine (ctz) results in oxidation of the substrate to coelenteramide,
which produces blue light at 482 nm. When protein-protein interactions between
Protein X and Protein Y bring the donor luciferase (RLuc) and acceptor fluor-
ophore (YFP) in close proximity (<10 nm), the energy from the donor can be
transferred to the acceptor and light is produced at 527 nm. When the BRET tags
are not in close enough proximity, light is only emitted at 482 nm
Fig. 2 The energy transfer between BRET pairs depends on the overlap of the
donor emission spectrum with the excitation spectrum of the acceptor. For
Renilla luciferase, oxidation of coelenterazine results in an emission peak at
482 nm. This emission overlaps well with the excitation spectrum of yellow
fluorescent protein (excitation peak: 514 nm). The resulting energy transfer
yields yellow light with an emission peak of 527 nm
2 Materials
2.1 Cell Lines 1. Maintain HEK 293 cells in 1 Dulbecco’s Modified Eagle
Medium (DMEM) without phenol red indicator, supplemented
with 2 mM L-glutamine, 100 U/mL penicillin, 100 mg/mL
streptomycin, and 10 % fetal bovine serum (5 % for transfection).
Grow cells in a humidified incubator with 5 % CO2 at 37 C.
460 Nicole E. Brown et al.
2.3 Instrumentation 1. Microplate reader with 485 nm emission and excitation filters,
530 nm emission filters, and compatible white-bottomed
96-well plates (see Note 3).
2. Compatible microplate reader, spreadsheet, and graphing
software (see Note 3).
3 Methods
3.2 Transient 1. Seed 8 105 cells per well in six-well plates in 2 mL medium
Transfection with per well, grow in a humidified incubator at 37 C overnight
Polyethylenimine (PEI) with 5 % CO2.
(See Note 5) 2. Prior to transfection, change medium to 1 DMEM contain-
ing 2 mM L-glutamine, 100 U/mL penicillin, 100 mg/mL
streptomycin, and 5 % fetal bovine serum, 2 mL per well.
3. Generate solution A by adding 8 μL of 1 mg/mL PEI from
stock to 92 μL of serum-free medium for each well, allow this
solution to incubate for 3 min.
4. Generate solution B by adding up to 1.5 μg of DNA to 100 μL
of serum-free medium for each condition in 1.5 mL microcen-
trifuge tubes. DNA amount is adjusted to a final concentration
of 1.5 μg by adding empty pcDNA3.1 plasmid.
5. Add 100 μL of solution A to microcentrifuge tubes containing
solution B to create solution C.
6. Cap the 1.5 mL microcentrifuge tube and immediately vortex
for 3 s.
7. Incubate solution C at room temperature for 15 min.
8. Add solution C (~200 μL) dropwise to the appropriate well of
cells in the six-well plates.
9. Allow cells to grow for 1–2 days (the medium does not need to
be changed for PEI transfection).
3.4 Analysis 1. Export fluorescence and BRET data into spreadsheet software.
2. The BRET ratio can be determined by dividing fluorescence by
luminescence (BRET readings at 530/485 nm).
3. Calculate net BRET by subtracting out background lumines-
cence (BRET readings in cells expressing the donor without
any acceptor).
4. Calculate the acceptor/donor ratio by dividing the initial fluo-
rescence measurements (530 nm) by the luminescence mea-
surements (485 nm).
5. Using graphing software, plot the acceptor/donor ratio against
the net BRET as in Fig. 3. The data can then be fit using a
nonlinear regression, (typically a one-site binding [hyperbola]
is the most appropriate) to observe BRET saturation as a key
indicator of signal specificity.
4 Notes
Fig. 3 HEK 293 cells were transfected with increasing amounts of Gαi1-YFP (0,
10, 50, 100, 250, and 500 ng) and either 5 ng RGS14-WT-Luc or RGS14-Q515A/
R516A-Luc. Wild type RGS14 shows a robust BRET signal with Gαi1. Conversely,
the RGS14 mutant (Q515A/R516A) that can no longer bind Gαi1 shows a
drastically reduced maximal BRET signal indicating a disruption in the protein-
protein interaction. The above data is representative of three independent
experiments. Curves were generated with GraphPad Prism 5 using the one-
site binding curve fitting function. Additionally, Gαi1-YFP expression levels were
verified by immunoblot analysis
References
1. Xu Y, Piston DW, Johnson CH (1999) A inhibitors of cholinesterase-8A (Ric-8A) both
bioluminescence resonance energy transfer regulate the regulator of g protein signaling 14
(BRET) system: application to interacting cir- RGS14.Galphai1 complex in live cells. J Biol
cadian clock proteins. Proc Natl Acad Sci U S A Chem 286:38659–38669
96:151–156 11. Oner SS, Maher EM, Breton B et al (2010)
2. Wu P, Brand L (1994) Resonance energy trans- Receptor-regulated interaction of activator of
fer: methods and applications. Anal Biochem G-protein signaling-4 and Galphai. J Biol
218:1–13 Chem 285:20588–20594
3. Xu Y, Kanauchi A, von Arnim AG et al (2003) 12. Oner SS, An N, Vural A et al (2010) Regula-
Bioluminescence resonance energy transfer: tion of the AGS3.G{alpha}i signaling complex
monitoring protein-protein interactions in liv- by a seven-transmembrane span receptor. J Biol
ing cells. Methods Enzymol 360:289–301 Chem 285:33949–33958
4. Lohse MJ, Nuber S, Hoffmann C (2012) Fluo- 13. Jiang LI, Collins J, Davis R et al (2007) Use of
rescence/bioluminescence resonance energy a cAMP BRET sensor to characterize a novel
transfer techniques to study G-protein-coupled regulation of cAMP by the sphingosine 1-
receptor activation and signaling. Pharmacol phosphate/G13 pathway. J Biol Chem
Rev 64:299–336 282:10576–10584
5. Nagai T, Ibata K, Park ES et al (2002) A variant 14. Lan TH, Liu Q, Li C et al (2012) Sensitive and
of yellow fluorescent protein with fast and effi- high resolution localization and tracking of
cient maturation for cell-biological applica- membrane proteins in live cells with BRET.
tions. Nat Biotechnol 20:87–90 Traffic 13:1450–1456
6. Romero-Fernandez W, Borroto-Escuela DO, 15. Couturier C, Deprez B (2012) Setting Up a
Tarakanov AO et al (2011) Agonist-induced Bioluminescence Resonance Energy Transfer
formation of FGFR1 homodimers and signal- High throughput Screening Assay to Search
ing differ among members of the FGF family. for Protein/Protein Interaction Inhibitors in
Biochem Biophys Res Commun 409:764–768 Mammalian Cells. Front Endocrinol 3:100
7. Gales C, Rebois RV, Hogue M et al (2005) 16. Hollinger S, Taylor JB, Goldman EH et al
Real-time monitoring of receptor and G- (2001) RGS14 is a bifunctional regulator of
protein interactions in living cells. Nat Meth- Galphai/o activity that exists in multiple
ods 2:177–184 populations in brain. J Neurochem
8. Angers S, Salahpour A, Joly E et al (2000) 79:941–949
Detection of beta 2-adrenergic receptor dimer- 17. Kimple RJ, De Vries L, Tronchere H et al
ization in living cells using bioluminescence (2001) RGS12 and RGS14 GoLoco motifs
resonance energy transfer (BRET). Proc Natl are G alpha(i) interaction sites with guanine
Acad Sci U S A 97:3684–3689 nucleotide dissociation inhibitor Activity. J
9. Hamdan FF, Audet M, Garneau P et al (2005) Biol Chem 276:29275–29281
High-throughput screening of G protein- 18. Gibson SK, Gilman AG (2006) Gialpha and
coupled receptor antagonists using a biolumi- Gbeta subunits both define selectivity of G
nescence resonance energy transfer 1-based protein activation by alpha2-adrenergic recep-
beta-arrestin2 recruitment assay. J Biomol tors. Proc Natl Acad Sci U S A 103:212–217
Screen 10:463–475 19. Bacart J, Corbel C, Jockers R et al (2008) The
10. Vellano CP, Maher EM, Hepler JR et al (2011) BRET technology and its application to screen-
G protein-coupled receptors and resistance to ing assays. Biotechnol J 3:311–324
Chapter 31
Abstract
Cellular biochemical machineries, what we call pathways, consist of dynamically assembling and
disassembling macromolecular complexes. Although our models for the organization of biochemical
machines are derived largely from in vitro experiments, do they reflect their organization in intact, living
cells? We have developed a general experimental strategy that addresses this question by allowing the
quantitative probing of molecular interactions in intact, living cells. The experimental strategy is based on
Protein fragment Complementation Assays (PCA), a method whereby protein interactions are coupled to
refolding of enzymes from cognate fragments where reconstitution of enzyme activity acts as the detector of
a protein interaction. A biochemical machine or pathway is defined by grouping interacting proteins into
those that are perturbed in the same way by common factors (hormones, metabolites, enzyme inhibitors,
etc.). In this chapter we review some of the essential principles of PCA and provide details and protocols for
applications of PCA, particularly in mammalian cells, based on three PCA reporters, dihydrofolate reduc-
tase, green fluorescent protein, and β-lactamase.
Key words Protein fragment Complementation Assays, Dihydrofolate reductase, Green fluorescent
protein, TEM β-lactamase, Two-hybrid, Protein-protein interactions, Methotrexate, CCF2/AM,
Nitrocefin, Fluorescein, Flow cytometry, CHO, COS, HEK 293 cells
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_31, © Springer Science+Business Media New York 2015
467
468 Ingrid Remy and Stephen W. Michnick
a b c
d e f
g h
Relative intensity
NO
Number of cells
1.0
EPO
EMP1
0.5
0.0
0
10 101 10 2
10 3 10-13 10-11 10-9 10-7
FITC units Concentration EPO
a
48.18
Hydrolysis of CCF2/AM ate 460nm
b
50.00
(fluorescence units/min)
40.00
30.00
c
20.00
11.59
9.44 8.48
10.00
0.00
FRB+ZIP pMT3 CCF2AM ZIP+ZIP
Fig. 3 ß-Lactamase PCA using the fluorescent substrate CCF2/AM. (a) ZIP (GCN4 leucine zipper-forming
sequences) are tested in HEK 293 cells as described in the text. FRB (rapamycin-FKBP binding domain of
FRAP) is used as a negative control. pMT3 is the expression vector alone and ZIP + ZIP is the positive control.
Data recorded in white microtiter plates on a Perkin Elmer HTS 7000 plate reader. (b, c) Fluorescent
micrographs of cells expressing ß-lactamase PCA showing negative (b, FRB + ZIP) or positive (c, ZIP + ZIP)
response
2 Materials
2.1 DHFR PCA 1. 12-well plates, tissue culture treated; six-well plates, tissue
Survival Assay culture treated.
2. Minimum essential medium: alpha medium without ribonu-
cleosides and deoxyribonucleosides (α-MEM).
3. Dialyzed fetal bovine serum (Hyclone, Cat. no: SH30079-03).
4. Adenosine; deoxyadenosine; thymidine.
5. Transfection reagent (e.g., Lipofectamine Plus reagent, Life
Technologies).
6. Trypsin–EDTA.
7. Cloning cylinders.
Mapping Biochemical Networks with PCA 473
3 Methods
3.1 DHFR PCA 1. Twenty-four hours before transfection, plate 1 105 CHO
Survival Assay DUKX-B11 cells (DHFR-negative; could also be done in
other cells lines, see Note 1) in 12-well plates in α-MEM
medium enriched with 10 % dialyzed fetal bovine serum and
supplemented with 10 μg/ml of adenosine, deoxyadenosine,
and thymidine.
2. Co-transfect cells with the PCA fusion partners (see Note 2)
using a transfection reagent (e.g., Lipofectamine Plus reagent)
according to the manufacturer’s instructions.
3. Forty-eight hours after the beginning of the transfection, split
cells at approximately 5 104 in 6-well plates in selective
medium consisting of α-MEM enriched with dialyzed FBS
but without addition of nucleotides (see Notes 3 and 4).
4. Change medium every 3 days. The appearance of distinct colo-
nies usually occurs after 4–10 days of incubation in selective
medium. Colonies are observed only for clones that simulta-
neously express both interacting proteins fused to one or the
other complementary DHFR fragments. Only interacting pro-
teins will be able to achieve normal cell division and colony
formation.
For further analysis of the interacting protein pair:
5. Isolate three to five colonies per interacting partners by trypsi-
nization (trypsin–EDTA) using cloning cylinders and grow
them separately.
Mapping Biochemical Networks with PCA 475
3.2 DHFR PCA 1. Twenty-four hours before transfection, plate 1 105 COS
Fluorescence Assay cells (this assay can be performed using any other cell line, see
Note 5) on 18 mm circular glass coverslips in 12-well plates in
3.2.1 Fluorescence
DMEM medium enriched with 10 % Cosmic calf serum.
Microscopy
2. Transiently co-transfect cells with the PCA fusion partners (see
Note 2) using a transfection reagent (e.g., Lipofectamine Plus
reagent) according to the manufacturer’s instructions.
3. The next day, change medium and add fluorescein-conjugated
methotrexate (fMTX) to the cells at a final concentration of
10 μM (see Note 6).
For stable cell lines:
For CHO DUKX-B11 cells (or other cell line) stably expres-
sing PCA fusion partners, seed cells to approximately 2 105
on 18 mm glass coverslips in 12-well plates in α-MEM medium
enriched with 10 % dialyzed FBS. The next day, fMTX is added
to the cells at a final concentration of 10 μM.
4. After incubation with fMTX for 22 h at 37 C, remove the
medium and wash the cells with PBS and re-incubate for
15–20 min at 37 C in the culture medium to allow for efflux
of unbound fMTX (see Note 7). Remove medium and wash the
cells four times with cold PBS on ice and finally mount the
coverslips on microscope glass slides with an aqueous mount-
ing medium.
5. Perform fluorescence microscopy on live cells (see Note 8).
These experiments must be performed within 30 min of the
wash procedure. If the negative control (untransfected cells
treated with fMTX) is too fluorescent, the wash procedure
must be modified (see Note 9).
3.2.2 Flow Cytometry Preparation of cells for fluorescence-activated cell sorting (FACS)
Analysis analysis is the same as described for fluorescence microscopy, except
that following the PBS wash (twice in this case), cells are gently
trypsinized (trypsin–EDTA), suspended in 500 μl of cold PBS and
kept on ice prior to flow cytometric analysis within 30 min. Data are
collected on a FACS analyzer with stimulation with an argon laser
tuned to 488 nm with emission recorded through a 525 nm band-
width filter.
476 Ingrid Remy and Stephen W. Michnick
3.2.3 Fluorometric Preparation of cells for fluorometric analysis is the same as described
Analysis for fluorescence microscopy, except that following the PBS wash
(twice in this case), cells are gently trypsinized (trypsin–EDTA).
Plates are put on ice and 100 μl of cold PBS is added to the cells.
The total cell suspensions are transferred to 96-well white microti-
ter plates (Dynex) and keep on ice prior to fluorometric analysis.
The assay can be performed on any microtiter plate reader; we use a
Perkin-Elmer HTS 7000 Series Bio Assay Reader in the fluores-
cence mode. The excitation and emission wavelengths for the
fMTX are 497 nm and 516 nm, respectively. Afterward, the data
are normalized to total protein concentration in cell lysates.
3.3 GFP PCA All procedures describe for the DHFR PCA fluorescence assays are
Fluorescence Assay the same for GFP PCA, except that there are no use of fMTX and
no washing steps. The wash procedure is obviously irrelevant in the
case of the GFP PCA where the folded/reassembled protein is a
fluorophore itself.
4 Notes
References
1. Drees BL (1999) Progress and variations in 9. Hu CD, Kerppola TK (2003) Simultaneous
two-hybrid and three-hybrid technologies. visualization of multiple protein interactions
Curr Opin Chem Biol 3:64–70 in living cells using multicolor fluorescence
2. Evangelista C, Lockshon D, Fields S (1996) complementation analysis. Nat Biotechnol
The yeast two-hybrid system: prospects for 21:539–545
protein linkage maps. Trends Cell Biol 10. Paulmurugan R, Gambhir SS (2003) Monitor-
6:196–199 ing protein-protein interactions using split syn-
3. Fields S, Song O (1989) A novel genetic system thetic renilla luciferase protein-fragment-
to detect protein-protein interactions. Nature assisted complementation. Anal Chem
340:245–246 75:1584–1589
4. Vidal M, Legrain P (1999) Yeast forward and 11. Luker KE, Smith MC, Luker GD et al (2004)
reverse ‘n’-hybrid systems. Nucleic Acids Res Kinetics of regulated protein-protein interac-
27:919–929 tions revealed with firefly luciferase comple-
5. Walhout AJ, Sordella R, Lu X et al (2000) mentation imaging in cells and living animals.
Protein interaction mapping in C. elegans Proc Natl Acad Sci U S A 101:12288–12293
using proteins involved in vulval development. 12. Magliery TJ, Wilson CG, Pan W et al (2005)
Science 287:116–122 Detecting protein-protein interactions with a
6. Uetz P, Giot L, Cagney G et al (2000) A com- green fluorescent protein fragment reassembly
prehensive analysis of protein-protein interac- trap: scope and mechanism. J Am Chem Soc
tions in Saccharomyces cerevisiae. Nature 127:146–157
403:623–627 13. Remy I, Michnick SW (2006) A highly sensi-
7. Michnick SW, Remy I, Campbell-Valois FX tive protein-protein interaction assay based on
et al (2000) Detection of protein-protein inter- Gaussia luciferase. Nat Methods 3:977–979
actions by protein fragment complementation 14. Wehr MC, Laage R, Bolz U et al (2006) Mon-
strategies. Methods Enzymol 328:208–230 itoring regulated protein-protein interactions
8. Paulmurugan R, Umezawa Y, Gambhir SS using split TEV. Nat Methods 3:985–993
(2002) Noninvasive imaging of protein- 15. Michnick SW, Ear PH, Manderson EN et al
protein interactions in living subjects by using (2007) Universal strategies in research and
reporter protein complementation and recon- drug discovery based on protein-fragment
stitution strategies. Proc Natl Acad Sci U S A complementation assays. Nat Rev Drug Discov
99:15608–15613 6:569–582
Mapping Biochemical Networks with PCA 481
16. Remy I, Michnick SW (2007) Application of 29. Remy I, Michnick SW (1999) Clonal selection
protein-fragment complementation assays in and in vivo quantitation of protein interactions
cell biology. Biotechniques 42:137, 139, 141 with protein-fragment complementation
passim assays. Proc Natl Acad Sci U S A
17. Stefan E, Aquin S, Berger N et al (2007) Quan- 96:5394–5399
tification of dynamic protein complexes using 30. Remy I, Wilson IA, Michnick SW (1999)
Renilla luciferase fragment complementation Erythropoietin receptor activation by a
applied to protein kinase A activities in vivo. ligand-induced conformation change. Science
Proc Natl Acad Sci U S A 104:16916–16921 283:990–993
18. Michnick SW, Ear PH, Landry C et al (2010) A 31. Remy I, Michnick SW (2001) Visualization of
toolkit of protein-fragment complementation biochemical networks in living cells. Proc Natl
assays for studying and dissecting large-scale Acad Sci U S A 98:7678–7683
and dynamic protein-protein interactions in liv- 32. Remy I, Campbell-Valois FX, Michnick SW
ing cells. Methods Enzymol 470:335–368 (2007) Detection of protein-protein interac-
19. Michnick SW, Ear PH, Landry C et al (2011) tions using a simple survival protein-fragment
Protein-fragment complementation assays for complementation assay based on the enzyme
large-scale analysis, functional dissection and dihydrofolate reductase. Nat Protoc
dynamic studies of protein-protein interactions 2:2120–2125
in living cells. Methods Mol Biol 756:395–425 33. Tarassov K, Messier V, Landry CR et al (2008)
20. Anfinsen CB, Haber E, Sela M et al (1961) The An in vivo map of the yeast protein interac-
kinetics of formation of native ribonuclease tome. Science 320:1465–1470
during oxidation of the reduced polypeptide 34. Galarneau A, Primeau M, Trudeau LE et al
chain. Proc Natl Acad Sci U S A 47:1309–1314 (2002) Beta-lactamase protein fragment com-
21. Anfinsen CB (1973) Principles that govern the plementation assays as in vivo and in vitro sen-
folding of protein chains. Science sors of protein protein interactions. Nat
181:223–230 Biotechnol 20:619–622
22. Gutte B, Merrifield RB (1971) The synthesis of 35. Remy I, Ghaddar G, Michnick SW (2007)
ribonuclease A. J Biol Chem 246:1922–1941 Using the beta-lactamase protein-fragment
23. Richards FM (1958) On the Enzymic Activity complementation assay to probe dynamic
of Subtilisin-Modified Ribonuclease. Proc Natl protein-protein interactions. Nat Protoc
Acad Sci U S A 44:162–166 2:2302–2306
24. Taniuchi H, Anfinsen CB (1971) Simultaneous 36. Israel DI, Kaufman RJ (1993) Dexamethasone
formation of two alternative enzymology active negatively regulates the activity of a chimeric
structures by complementation of two overlap- dihydrofolate reductase/glucocorticoid recep-
ping fragments of staphylococcal nuclease. J tor protein. Proc Natl Acad Sci U S A
Biol Chem 246:2291–2301 90:4290–4294
25. Pelletier JN, Campbell-Valois FX, Michnick 37. Kaufman RJ, Bertino JR, Schimke RT (1978)
SW (1998) Oligomerization domain-directed Quantitation of dihydrofolate reductase in
reassembly of active dihydrofolate reductase individual parental and methotrexate-
from rationally designed fragments. Proc Natl resistant murine cells. Use of a fluorescence
Acad Sci U S A 95:12141–12146 activated cell sorter. J Biol Chem
26. Pelletier JN, Michnick SW (1997) A protein 253:5852–5860
complementation assay for detection of 38. Kaufman RJ (1990) Selection and coamplifica-
protein-protein interactions in vivo. Protein tion of heterologous genes in mammalian cells.
Eng 10:89 Methods Enzymol 185:537–566
27. Johnsson N, Varshavsky A (1994) Split ubiqui- 39. Zlokarnik G, Negulescu PA, Knapp TE et al
tin as a sensor of protein interactions in vivo. (1998) Quantitation of transcription and
Proc Natl Acad Sci U S A 91:10340–10344 clonal selection of single living cells with beta-
28. Rossi F, Charlton CA, Blau HM (1997) Moni- lactamase as reporter. Science 279:84–88
toring protein-protein interactions in intact 40. Zlokarnik G (2000) Fusions to beta-
eukaryotic cells by beta-galactosidase comple- lactamase as a reporter for gene expression
mentation. Proc Natl Acad Sci U S A in live mammalian cells. Methods Enzymol
94:8405–8410 326:221–244
Chapter 32
Abstract
The bimolecular fluorescence complementation (BiFC) assay is a versatile technique for investigating
protein-protein interaction (PPI) in living systems. The BiFC assay exploits the color-emitting moiety
and the modular structure of fluorescent proteins to provide both temporal and spatial information of the
PPI. The modular property of fluorescent proteins enables researchers to strategically partition a fluorescent
protein into two nonfluorescent units, which can be independently fused to other proteins. When the
fusion proteins interact with each other, the nonfluorescent fragments reconstitute to generate a fluores-
cence signal. PPI can then be detected by capturing the fluorescence signal with a fluorescence microscope.
In this chapter, the Venus fluorescent protein is employed to demonstrate the application of the BiFC assay.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_32, © Springer Science+Business Media New York 2015
483
484 Cau D. Pham
Fig. 1 The principle of Venus BiFC technology. The Venus fluorescent protein is strategically split into two
nonfluorescent fragments. The reconstitution of Venus protein and signal from the nonfluorescent constituents
can be mediated by two interacting proteins. Venus protein structure was drawn using the I-TASSER program [8]
Table 1
Fluorescent proteins commonly utilized for BiFC technology
Brightness Oligomerization
Color Percentage half-lifeb (in of fully mature
Protein Organism Ex/Em class of brightnessa seconds) protein BiFC split position Reference
CFP Aequorea 433/475 Cyan 19 64 Monomer 154–155;172–173 [2, 6]
victoria
Cerulean Aequorea 433/475 Cyan 39 36 Weak dimer 154–155;172–173 [2, 6]
victoria
GFP Aequorea 485/500 Green 49 174 Weak dimer 157–158 [2, 6]
victoria
YFP Aequorea 514/527 Yellow 74 60 Weak dimer 154–155;172–173 [2, 6]
victoria
Venus Aequorea 515/528 Yellow 76 15 Weak dimer 154–155;172–173 [2, 6]
victoria
Citrine Aequorea 516/529 Yellow 85 49 Monomer 154–155;172–173 [2, 6]
victoria
mRFP1-Q66T Discosoma sp. 549/570 Red ND ND ND 154–155;168–169 [2, 6]
485
486 Cau D. Pham
as in live cells or animals [11, 12]. Secondly, the BiFC assay can
provide valuable information regarding the sub-cellular location of
the PPI [12]. Such information has good predictive value about the
functional roles of the interaction. These and other aforementioned
attributes are the reason why BiFC is a valuable tool to have in one’s
molecular cabinet. In this chapter, one will learn how to detect PPI
in mammalian cells using Venus BiFC.
2 Materials
1. Thermocycler.
2. DNA polymerase and buffer (e.g., Phusion, New England
BioLabs).
3. P38 and MKK3 cDNAs (Open Biosystems).
4. Gateway® entry vector (Life Technologies).
5. TE (10 mM Tris–HCl, 1 mM EDTA) pH 8.0.
6. BP Clonase™ II enzyme mix (Life Technologies).
7. Shaker-incubator and water bath.
8. Competent bacteria (e.g., NEB10β).
9. Bacterial growth media including S.O.C and LB. LB medium is
supplemented with the appropriate antibiotic (100 μg/mL
Ampicillin, 50 μg/mL Kanamycin).
10. Mini-prep/midi-prep kits.
11. BsrGI restriction endonuclease and buffer.
12. Agarose gel electrophoresis reagents (molecular grade agarose,
Tris-Borate-EDTA buffer, ethidium bromide).
13. Gene-specific oligonucleotide primers for cloning and
sequencing.
14. pSCM167-NV and pDEST26-CV mammalian expression vec-
tors (developed in the lab of Dr. Haian Fu, Emory University).
15. LR Clonase™ II enzyme mix (Life Technologies).
16. A nucleic acid spectrophotometer (e.g., NanoDrop).
17. Cell growth medium for the specific cell line (e.g., DMEM
supplemented with 5 % FBS and 1 % Penicillin–Streptomycin).
18. Cell line that can be transfected (e.g., the LN229 cell line).
19. Cell culture incubator with CO2 supply.
20. 0.05 % Trypsin/0.53 mM EDTA.
21. Opti-MEM.
22. Transfection reagent (e.g., FuGENE® HD transfection
reagent, Promega).
23. Fluorescence microscope and filters.
BiFC Assay for PPI Detection 487
3 Methods
Described below are (1) the steps to create BiFC vectors and (2)
how to examine PPI in mammalian cells (see Note 1). A previously
known interacting protein pair MKK3 and P38 [13] will be used to
demonstrate the use of a BiFC assay to monitor PPI. The BiFC
reporter used in this example is the Venus fluorescent protein, a
variant of EYFP (see Note 2). Venus fluoresces brighter, matures
rapidly at 37 C, and is less sensitive to environmental conditions
[3, 14, 15]. Both Venus fragments are in the Gateway® vector
system (Life Technologies) and will generate N-terminal fusion
proteins (the Venus fragments fuse to the N-terminus of the pro-
tein of interest) (Fig. 2). The N-terminal fragment of Venus (NV),
Fig. 2 Gateway® cloning technology and a schematic of the Venus BiFC constructs. (a) Gateway® vectors
allow the shuttling of DNA fragments from one vector to another compatible vector via site-specific homolo-
gous recombination catalyzed by recombinase enzymes. (b) N-terminal fusion constructs regulated by the
CMV promoter. For these vectors, the Venus fragments are attached to the protein of interest through a
(2xGGGS) linker sequence
488 Cau D. Pham
Fig. 3 The interaction between P38 and MKK3 was used to demonstrate the Venus BiFC assay. Cells that
expressed both P38 and MKK3 proteins fused to compatible Venus fragments exhibit strong Venus signals.
The Venus signal in cells that expressed only one fusion protein along with its complementary Venus fragment
was more than two times lower
residues 1–173, along with a linker (see Note 3), was inserted into
the pSCM167 vector (Haian Fu lab) to generate the pSCM167-NV
expression vector (see Note 4). The C-terminal fragment of Venus
(CV), residues 156–239, along with a linker, was inserted into the
pDEST26 vector (Life Technologies) to make the pDEST26-CV
expression vector (see Note 4).
3.1 Creating BiFC Gateway® cloning technology can be used to clone DNA fragments
Constructs for PPI into the Venus BiFC expression vectors (as an example, P38 and
Detection MKK3 were cloned into pSCM167-NV and pDEST26-CV, respec-
tively) (Figs. 2 and 3). Gateway® technology employs a two-vector
system, an entry and destination vector, to shuttle DNA fragments
between vectors. To utilize this technology, a DNA fragment is first
inserted into an entry vector. From the entry vector, the DNA
fragment can be transferred to Gateway® destination/expression
vectors via site-specific homologous recombination.
3.1.1 Cloning of DNA 1. Use a polymerase chain reaction (PCR) to add attB sites to the
Fragments into a 50 and 30 ends of the DNA fragment as described by the
Gateway® Entry Vector manufacturer (Life Technologies) (see Note 5). For example,
attB sequences {50 -GGG GAC AAG TTT GTA CAA AAA
AGC AGG CTT C-30 and 50 -GGG GAC CAC TTT GTA
CAA GAA AGC TGG GTC TTA CTA-30 } were added to the
50 and 30 , respectively, of both P38 and MKK3. The attB
sequences facilitate the insertion of DNA fragments into the
Gateway® entry vectors via site-specific homologous
recombination.
2. Perform the cloning of the DNA fragment, flanked by the attB
sites, into the Gateway® entry vector (in this case pDONR201)
using BP clonase enzyme and bacterial transformation as sug-
gested by the manufacturer’s instruction (with minor
BiFC Assay for PPI Detection 489
3.1.2 Cloning of the DNA Both the pSCM167-NV and the pDEST26-CV are Gateway®-
Fragment into the based expression vectors. This enables the transfer of DNA frag-
pSCM167-NV and ments from entry vectors (such as pDONR201) directly into the
pDEST26-CV Vectors pSCM167-NV and pDEST26-CV vectors via site-specific homolo-
gous recombination. The transferring of a DNA fragment from
Gateway® entry vectors into Gateway® destination vectors is cata-
lyzed by the LR Clonase™ enzyme and is performed as instructed
by the manufacturer (with minor modifications).
1. First, combine the purified entry vector (15–150 ng) and
150 ng of the destination vector (pSCM167-NV or
pDEST26-CV) in a 1.5 mL microcentrifuge tube. Next, if
necessary, adjust the reaction volume to 8 μL with TE buffer.
Then, add 2 μL of LR Clonase™ II enzyme mix to the reaction,
490 Cau D. Pham
3.2 Performing In order to perform the BiFC assay, cells must uptake the expres-
the BiFC Assay sion vectors. Prior knowledge about the model system is essential
(see Note 1). Prior to perform BiFC assay, one needs to confirm the
expression level of the fusion proteins. This can be determined by
Western blot (see Note 8). One also needs to consider checking the
folding of the proteins of interest. Incorrect folding of the proteins
of interest caused by the fused fluorescent protein fragments could
affect the stability and localization of the protein [11] (see Note 9).
Localization of fusion proteins and untagged wild-type proteins
can be assessed by indirect immunofluorescence assay [11].
3.2.2 Cell Growth In this example, low passage LN229 cells were cultured in DMEM
Conditions, Cell Seeding, supplemented with 5 % FBS and 1 % Penicillin/Streptomycin
and Cell Transfection (please note that the cell culture and transfection conditions will
need to be adjusted for the cell lines being used). Cells were kept in
a water-jacketed incubator at 37 C and supplemented with 5 %
CO2 humidity. For transfection, the following steps were taken.
BiFC Assay for PPI Detection 491
3.2.3 Detecting PPI with Proteins that show strong interaction can be detected 24 h post-
a Fluorescence Microscope transfection (see Note 15). Once the cells have been transfected
with the appropriate plasmids, PPI can be observed using a fluores-
cence microscope with the correct filter. The YFP filter set can be
used to detect Venus signal. As demonstrated in Fig. 3, strong
Venus signal was observed in cells that were co-transfected with
the pSCM167-NV-P38 and the pDEST26-CV-MKK3 vectors.
Noticeably, a low Venus signal was also observed in cells that were
co-transfected with control vectors; the pSCM167-NV-P38 and
pDEST26-CV pair as well as pDEST26-CV-MKK3 and
492 Cau D. Pham
4 Notes
11. Good quality DNA (260/280 ratio close to 2.0) is essential for
a successful assay. Poor quality DNA can reduce transfection
efficiency or can be detrimental to the cell.
12. Make sure that FBS is present in the growth medium. FBS
inhibits trypsin activity [16].
13. Too much DNA can be toxic to some cell lines. Moreover, too
much DNA could lead to high background noise [15].
14. Various transfection reagents may be toxic to some cell lines.
If using FuGENE® HD reagent, minimize contact of the
reagent with plastic surface (i.e., microcentrifuge tube’s wall)
because this could lead to a decrease in transfection efficiency.
15. Strong interacting pairs of proteins can be detected as early as
24 h post-transfection. Long incubation times can lead to high
background signal.
16. Self-assembly of the Venus BiFC fragments as well as of other
BiFC fragments is often a drawback for this assay. Self-assembly
of the BiFC fragments is a common contributor of false-
positive results. Self-assembly signals are typically lower than
protein-interaction assisted reconstitution signals for many
PPIs. However, self-assembly signals may pose a problem for
detecting weak PPIs. Replacing certain amino acids in BiFC
systems (i.e., Venus) can reduce self-assembly [14].
Acknowledgements
I would like to thank Drs. Haian Fu and Jonathan Havel for their
generous gifts of the pSCM167-NV and pDEST26-CV Gateway®
destination vectors as well as constructive input to make this assay
work.
References
1. Shimomura O, Johnson FH, Saiga Y (1962) 5. Hu CD, Chinenov Y, Kerppola TK (2002)
Extraction, purification, and properties of Visualization of interactions among bZIP and
aequorin, a bioluminescent protein from the Rel family proteins in living cells using bimo-
luminous hydromedusan, Aequorea. J Cell lecular fluorescence complementation. Mol
Comp Physiol 59:223–239 Cell 9:789–798
2. Shaner NC, Steinbach PA, Tsien RY (2005) A 6. Vidi PA, Watts VJ (2009) Fluorescent and bio-
guide to choosing fluorescent proteins. Nat luminescent protein-fragment complementa-
Methods 2:905–909 tion assays in the study of G protein-coupled
3. Kremers G-J, Gilbert SG, Cranfill PJ, Davidson receptor oligomerization and signaling. Mol
MW, Piston DW (2011) Fluorescent proteins Pharmacol 75:733–739
at a glance. J Cell Sci 124:157–160 7. Ozawa T (2006) Designing split reporter pro-
4. Remington SJ (2006) Fluorescent proteins: teins for analytical tools. Anal Chim Acta
maturation, photochemistry, and photophy- 556:58–68
sics. Curr Opin Struct Biol 16:714–721
BiFC Assay for PPI Detection 495
8. Zhang Y (2008) I-TASSER server for protein 3D network mapping. Curr Opin Biotechnol
structure prediction. BMC Bioinformatics 9:40 14:610–6177
9. Remy I, Michnick SW (2007) Application of 13. Raingeau J, Whitmarsh AJ, Barrett T, Derijard
protein-fragment complementation assays in B, Davis RJ (1996) MKK3- and MKK6-
cell biology. Biotechniques 42:137–145 regulated gene expression is mediated by the
10. Gandhi TK, Zhong J, Mathivanan S, Karthick p38 mitogen-activated protein kinase signal
L, Chandrika KN, Mohan SS, Sharma S, Pin- transduction pathway. Mol Cell Biol
kert S, Nagaraju S, Periaswamy B, Mishra G, 16:1247–1255
Nandakumar K, Shen B, Deshpande N, Nayak 14. Kodama Y, Hu CD (2010) An improved bimo-
R, Sarker M, Boeke JD, Parmigiani G, Schultz lecular fluorescence complementation assay
J, Bader JS, Pandey A (2006) Analysis of the with a high signal-to-noise ratio. Biotechni-
human protein interactome and comparison ques 49:793–805
with yeast, worm and fly interaction datasets. 15. Shyu YJ, Liu H, Deng X, Hu CD (2006) Iden-
Nat Genet 38:285–293 tification of new fluorescent protein fragments
11. Kerppola TK (2006) Design and implementation for bimolecular fluorescence complementation
of bimolecular fluorescence complementation analysis under physiological condition. Bio-
(BiFC) assays for the visualization of protein techniques 40:61–66
interactions in living cells. Nat Protoc 16. Rogers M, Dani JA (1995) Comparison of
1:1278–1286 quantitative calcium flux through NMDA,
12. Michnick SW (2003) Protein fragment ATP, and ACh receptor channels. Biophys J
complementation strategies for biochemical 68:501–506
Chapter 33
Abstract
The understanding of ion channel function continues to be a significant driver in molecular pharmacology.
In this field of study, protein-protein interactions are emerging as fundamental molecular determinants of
ion channel function and as such are becoming an attractive source of highly specific targets for drug
development. The investigation of ion channel macromolecular complexes, however, still relies on conven-
tional methods that are usually technically challenging and time-consuming, significantly hampering our
ability to identify, characterize and modify ion channel function through targeted molecular approaches. As
a response to the urgent need of developing rapid and albeit accurate technologies to survey ion channel
molecular complexes, we describe a new application of the split-luciferase complementation assay to study
the interaction of the voltage-gated Na + channel with the intracellular fibroblast growth factor 14 and its
dynamic regulation in live cells. We envision that the flexibility and accessibility of this assay will have a broad
impact in the ion channel field complementing structural and functional studies, enabling the interrogation
of protein–channel dynamic interactions in complex cellular contexts and laying the basis for new frame-
works in drug discovery campaigns.
Key words Protein-protein interactions, Protein fragment complementation, Split luciferase comple-
mentation, Firefly luciferase, Bioluminescence, Two-way protein fragment complementation assay,
Application of LCA, Sub-cloning of ion channel
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_33, © Springer Science+Business Media New York 2015
497
498 Alexander S. Shavkunov et al.
express the fusion proteins. The cells are then re-plated onto
multi-well plates, substrate is added, and luminescence resulting
from complementation of luciferase fragments is measured as a
function of the protein-protein interaction of interest. At this step,
additional experimental manipulations (e.g., pharmacological) can
be introduced to test the effect on the protein-protein interaction
of interest in live cells. Additional control experiments are utilized to
rule out possible artifacts that may arise from changes in the reporter
construct abundance (expression and/or degradation levels), viabil-
ity and metabolic state of transfected cells, or direct impact on the
reporter enzymatic activity non-related to the efficiency of the
complex assembly. In this chapter, we describe a new application
of LCA for the study of transmembrane ion channels.
Recent advancements in biology and pharmacology highlight
the crucial role of protein-protein interactions for ion channel
proper functioning and regulation [12–14]. The protein interfaces
through which these interactions are mediated present a novel class
of promising drug targets with high potential for improved speci-
ficity of therapeutic effects [15–19]. However, characterization of
the interactions of ion channels with their regulatory proteins, as
well as high-throughput screening for potential modulators of
these interactions, proves to be quite challenging with conventional
techniques [20, 21]. Direct analysis of protein-protein interactions
by traditional biochemical methods, like enzyme-linked immuno-
sorbent assays (ELISA), surface plasmon resonance (SPR), and
fluorescence polarization (FP), is complicated by the nature of ion
channels, which are difficult to express, purify, and reconstitute
in vitro. The functional effect of protein binding to ion channels
has been also studied using manual and/or automated patch-clamp
electrophysiology [22], fluorescence-based methods [23], or ion
flux assays [24]. Yet, despite certain advantages of these methods
over biochemical assays (higher cost efficiency, no need for protein
purification, measurements done in the cellular context), they pos-
sess inherent shortages and limitations of their own, such as lower
processivity, complicated manual operation, etc.
These considerations stimulated us to seek alternative strategies
to characterize the interaction between the voltage-gated sodium
channel Nav1.6 and the intracellular fibroblast growth factor
14 (FGF14), a biologically relevant regulatory protein which con-
trols gating, stability, and targeting of Nav channel α subunits
(Nav1.1–Nav1.9) through a high affinity interaction with the intra-
cellular C-terminal tail [25–30]. Our approach relied on the
bioluminescence-based split luciferase complementation assay
(LCA) introduced by Luker et al. [31]. This method is based on
functional complementation between two separated fragments of
the Photinus pyralis firefly luciferase which are fused to the pair of
interacting proteins of interest. The interaction of the respective
binding partners drives the complementation of the luciferase
LCA to Detect Channel–Protein Interactions 499
2 Materials
2.1 Cell Lines and 1. HEK-293 cells (or another cell line that is easily transfected).
Bacterial Strains 2. Chemically competent E. coli (such as One Shot TOP10
Chemically Competent E. coli, Invitrogen).
500 Alexander S. Shavkunov et al.
2.3 Cloning and DNA 1. FRB-NLuc (Gift from Dr. Piwnica-Worms, Washington Uni-
Preparation versity, St. Louis, MO [1]).
2. CLuc-FKBP (Gift from Dr. Piwnica-Worms, Washington Uni-
versity, St. Louis, MO [1]).
3. CD4-Nav1.6Ctail (Gift of Dr. Benedict Dargent, INSERM,
France [34]).
4. FGF14-1b-GFP [28].
5. pGL3 firefly luciferase plasmid (Promega, Madison, WI).
6. Phusion High-Fidelity DNA Polymerase.
7. DNA restriction enzymes (see Note 1).
8. Primers.
9. T4 DNA ligase.
10. Gel Extraction Kit.
11. LB broth: 10 g of Bacto-Tryptone, 5 g of yeast extract, 10 g of
NaCl dissolved in 1 l of distilled or deionized H2O and ster-
ilized by autoclaving.
12. LB agar: 10 g of Bacto-Tryptone, 5 g of yeast extract, 10 g of
NaCl, 15 g of agar dissolved in 1 l of distilled or deionized H2O
with heating and sterilized by autoclaving.
13. Ampicillin.
14. Selection media: LB agar plates supplemented with 100 μg/ml
ampicillin.
15. Miniprep Kit.
16. EndoFree Plasmid Maxi Kit.
LCA to Detect Channel–Protein Interactions 501
2.5 Cell Viability 1. Cell Proliferation Assay Kit (such as CyQUANT, Invitrogen).
Assay
3 Methods
3.1 DNA One of the major advantages of LCA is the possibility to monitor
Manipulation protein-protein interactions in live cells. This usually requires trans-
fection of mammalian cells with DNA constructs expressing hybrid
proteins bearing complementary fragments of the luciferase
reporter. As an example, we describe the sub-cloning of the DNA
coding sequences of our proteins of interest into the expression
vectors encoding complementary Photinus pyralis luciferase frag-
ments which were designed and constructed by Luker et al. [31].
The resulting plasmids were used to express hybrid protein con-
structs CLuc-FGF14, containing the C-terminus fragment (CLuc,
aa. 398–550) of the luciferase and the full-length human FGF14
isoform 1b, and CD4-Nav1.6-NLuc, containing a chimera of the
CD4ΔCtail (aa. 1–395) and the Nav1.6 C-tail (aa. 1763–1976)
fused to the N-terminus fragment (NLuc, aa. 2–416) of the lucif-
erase (Figs. 1 and 2). Using the CD4-Nav1.6 chimera has a number
advantages, as it is expressed more efficiently than the full-length
Nav1.6 in a heterologous system, ensures proper membrane target-
ing [34] and exposes the Nav channel C-tail on the plasma mem-
brane in the correct orientation. This also allows for the isolation of
the C-tail from the rest of the Nav channel, limiting any potential
indirect modulatory effects on the protein–channel complex
induced by other intracellular domains of the Nav channel. The
design of fusions between the reporter fragments and the proteins
of interest may vary depending on the particular pair of interaction
partners. The N-Luc or the C-Luc fragment of the reporter may be
placed at either terminus of the polypeptide chain of each of the
proteins of interest. Important factors to consider are the spatial
orientation of the interaction partners, in case structural
Fig. 2 Map of the expression vectors used for of the LCA. The detailed
description of the cloning procedure can be found in Subheading 3.1
LCA to Detect Channel–Protein Interactions 503
2. Plate 4.5 105 HEK293 cells per each well of a 24-well tissue
culture plate and incubate overnight to give monolayers at
90–100 % confluency (see Note 3).
3. The next day, pre-warm sterile PBS and DMEM/F12 without
serum and antibiotics at 37 C.
4. Prepare two Eppendorf tubes with equal volumes of DMEM/
F12 without serum or antibiotics (50 μl of medium per each
well of cells in the 24-well plate).
5. Add 1 μg of CD4-Nav1.6-NLuc and 1 μg of CLuc-FGF14
plasmid DNA per each well to be transfected into tube 1.
6. Add 2 μl of transfection reagent (e.g., Lipofectamine 2000) per
each well to be transfected (1 μl of the transfection reagent per
1 μg of plasmid DNA) into tube 2. Incubate for 5 min at room
temperature (see Note 4).
7. Transfer the medium containing DNA from tube 1 into tube
2 with the medium containing the transfection reagent. Mix by
gentle tapping (do not vortex). Incubate the transfection mix-
ture for 20 min at room temperature.
8. During incubation, aspirate the medium from the wells of the
24-well plate with HEK293 cells, wash briefly with PBS and
replace with 100 μl per well of DMEM/F12 without serum or
antibiotics.
9. Carefully dispense 100 μl of the transfection mixture to the
respective well of the 24-well plate containing cells (see Note
5).
10. Incubate the plate with the cells for 6 h at 37 C with 5 % CO2.
11. Add 800 μl of supplemented DMEM/F12 per well and let the
cells grow for 24 h (see Note 6).
3.3 Preparation 1. 24 h post transfection aspirate the culture medium from the 24-
of Transiently well plate with transfected HEK293 cells (see Note 7).
Transfected 2. Wash each well briefly with 400 μl of PBS and dispense 100 μl
HEK293 Cells of 0.04 % trypsin solution (0.25 % trypsin diluted with PBS) per
for Luminescence well (see Note 8).
Measurement 3. Place the plate in the CO2 incubator and monitor the cells
periodically under a microscope (see Note 9).
4. As soon as the cells detach from the plastic (typically 3–5 min)
add 800 μl of supplemented DMEM/F12 per each well to stop
trypsinization.
5. Wash the cells off the plastic by careful pipetting and transfer
the cell suspension from individual wells into a 15-ml tube with
a conical bottom.
6. Centrifuge for 5 min at 800 g.
506 Alexander S. Shavkunov et al.
3.4 Luminescence 1. Set up the experimental protocol for the Synergy H4 Multi-
Reader Method Setup Mode Microplate Reader with the following parameters
and Data Collection l Maintain temperature at 37 C.
l Dispense 100 μl of substrate solution per well prior to
measurement.
l Shake the plate for 3 s.
l Perform luminescence readings for 30 min at 2 min intervals
(open hole; integration time 0.5 s, sensitivity 245).
2. Replace the culture medium in the wells of the 96-well plate
with HEK293 cells (prepared as described in Subheadings 3.2
and 3.3) with 100 μl of DMEM/F12 without Phenol Red (see
Note 11).
3. Prepare working solution of 1.5 mg/ml of D-luciferin in PBS
(see Note 12).
4. Prime the substrate dispenser of the Synergy H4 Multi-Mode
Microplate Reader with 1,200 μl of D-luciferin working
solution.
5. Load the 96-well plate with cells into the Synergy H4 and
initiate the experimental protocol sequence.
3.5 Identification The LCA provides opportunities to test the effects of various
of Compounds pharmacological agents on the stability of a protein complex [1].
Modulating We have successfully applied this methodology to identify protein
the Protein-Protein kinase pathways which modulate the interaction between FGF14
Interaction Using and the Nav channel (Fig. 3a, b) [10, 11]. It is important to exclude
the LCA false positive hits due to direct influence of the compound on the
luciferase enzymatic activity; this type of artifact has been reported
previously with conventional luciferase reporter systems [5, 35].
We address this issue by parallel testing of candidate inhibitors on
transfected cells expressing full length Photinus pyralis luciferase
(from which the NLuc and CLuc fragments of the reporter have
been engineered) treated under the same conditions as in the LCA
experiment (Fig. 3c).
LCA to Detect Channel–Protein Interactions 507
Fig. 3 The FGF14-Nav1.6Ctail complex formation is regulated by specific kinase inhibitors. (a) HEK293 cells
were transiently transfected with CLuc-FGF14 and CD4-Nav1.6-NLuc and treated with the c-JNK inhibitor
SP600125 (50 μM; closed triangle), the IKK inhibitor, BAY 11-7082 (10 μM; closed diamond) or dimethylsulf-
oxide (DMSO; 0.5 % control; open circle); control without treatment is also shown (open square). Assembly of
the LCA pair is detected as luminescence upon the addition of the D-luciferin substrate at time zero and
normalized to % maximal luminescence signal in the untreated control; data are mean SEM, representing
quadruplicates from one representative experiment. (b) Bar graph represents mean SEM expressed as %
maximal luminescence of control (0.5 % DMSO) from at least three independent experiments. The graphs
illustrates the effect of 50 μM SP600125 and 10 μM BAY 11-7082 on the FGF14-Nav1.6 channel C-tail
assembly. BAY 11-7082 causes a twofold reduction of the LCA reporter assembly and luminescence output,
while SP600125 has no significant impact. (c) HEK293 cells were transiently transfected pGL3 expressing
508 Alexander S. Shavkunov et al.
3.6 Data Analysis 1. Export the relative luminescence values measured by Synergy
H4 and organized by well position and time point into Micro-
soft Excel.
2. Plot the luminescence values of the corresponding experimen-
tal wells as a function of time.
3. To compare the luminescence levels across experimental con-
ditions, calculate maximal signal intensity for each well as the
ä
Fig. 3 (Continued) full-length Photinus pyralis firefly luciferase holoenzyme and treated for 1 h prior to the
assay with the indicated compounds. Bar graph expressed as % maximal luminescence illustrates the effect
of the indicated compounds on the intrinsic enzymatic activity of luciferase; no statistically significant change
in full-length lucidferase activity was observed. (d) HEK293 cells were transiently transfected with CLuc-
FGF14 and CD4-Nav1.6-NLuc and treated with the indicated compounds. The effect of compounds on cell
viability was determined using a CyQUANT Cell Proliferation Assay Kit (Promega). Bar graph expressed as %
control fluorescence illustrates the effect of the indicated compounds on cell viability. Data are mean SEM;
***p < 0.001, Student’s t-test. (e) Representative example of Western blots of lysates (equal amount of
protein per lane) from cells transfected with CLuc-FGF14 + CD4-Nav1.6-NLuc and treated for 1 h with 0.5 %
DMSO (2, 4), 50 μM SP600125 (3), and 10 μM BAY 11-7082 (5). Untreated control is also shown (1). Western
blots were probed with a polyclonal anti-luciferase antibody; immunodetection of calnexin was used as
loading control
LCA to Detect Channel–Protein Interactions 509
3.7 Cell Viability Cell toxicity of test compounds is one of the factors which may
Testing affect the LCA output and produce false-positive results. To
account for this type of artifact, positive identification of com-
pounds by the LCA should be followed by tests on cell viability
under conditions identical to the LCA (Fig. 3d).
1. Transfect the cells with the LCA reporter constructs as
described in Subheading 3.2 and re-plate them onto a 96-
well plate as in Subheading 3.3.
2. Treat the cells with the test compounds as described in Sub-
heading 3.4, under the same conditions as in the LCA
experiment.
3. After incubation with the test compounds carefully aspirate the
culture medium and wash the cells with PBS.
4. Aspirate the PBS, freeze the cells, and store the plate at 80 C
until the samples are to be assayed (see Note 16).
5. Prepare the working solution for the cell proliferation assay
(the procedure below is for the CyQUANT assay).
l Dilute Component B (Cell lysis buffer) 1:20 in nuclease-free
distilled H2O (1 ml +19 ml).
l Thaw Component A (CyQUANT GR Dye) at room tem-
perature; add 250 μl (1:80) to the solution of Component B
prepared as above (see Note 17).
6. Thaw the cells at room temperature and add 200 μl of the
working solution to each well, including empty wells as blanks.
7. Incubate for 2–5 min at room temperature protected from light.
8. Measure fluorescence using a Synergy™ H4 Multi-Mode
Microplate Reader (excitation λ ¼ 485 nm, emission λ ¼
528 nm) or any suitable microplate reader with filters appro-
priate for ~480 nm excitation and ~520 nm emission maxima.
9. Subtract the mean fluorescence value of the blank from fluores-
cence values of the wells containing cells; calculate relative cell
viability as percent mean fluorescent signal intensity in the mock-
treated control samples from the same experimental plate.
6 Notes
References
1. Luker KE, Smith MC, Luker GD et al (2004) 6. Herbst KJ, Allen MD, Zhang J (2011) Lumi-
Kinetics of regulated protein–protein interac- nescent kinase activity biosensors based on a
tions revealed with firefly luciferase comple- versatile bimolecular switch. J Am Chem Soc
mentation imaging in cells and living animals. 133:5676–5679
Proc Natl Acad Sci U S A 101:12288–12293 7. Stefan E, Aquin S, Berger N et al (2007) Quan-
2. Misawa N, Kafi AK, Hattori M et al (2010) tification of dynamic protein complexes using
Rapid and high-sensitivity cell-based assays of Renilla luciferase fragment complementation
protein–protein interactions using split click applied to protein kinase A activities in vivo.
beetle luciferase complementation: an Proc Natl Acad Sci U S A 104:16916–16921
approach to the study of G-protein-coupled 8. Thorne N, Inglese J, Auld DS (2010) Illumi-
receptors. Anal Chem 82:2552–2560 nating insights into firefly luciferase and other
3. Paulmurugan R, Gambhir SS (2003) Monitor- bioluminescent reporters used in chemical
ing protein–protein interactions using split biology. Chem Biol 17:646–657
synthetic renilla luciferase protein-fragment- 9. Auld DS, Thorne N, Nguyen DT et al (2008)
assisted complementation. Anal Chem A specific mechanism for nonspecific activation
75:1584–1589 in reporter-gene assays. ACS Chem Biol
4. Paulmurugan R, Umezawa Y, Gambhir SS 3:463–470
(2002) Noninvasive imaging of protein–pro- 10. Shavkunov A, Panova N, Prasai A et al (2012)
tein interactions in living subjects by using Bioluminescence methodology for the detec-
reporter protein complementation and recon- tion of protein–protein interactions within the
stitution strategies. Proc Natl Acad Sci voltage-gated sodium channel macromolecular
99:15608–15613 complex. Assay Drug Dev Technol
5. Herbst KJ, Allen MD, Zhang J (2009) The 10:148–160
cAMP-dependent protein kinase inhibitor H- 11. Shavkunov AS, Wildburger NC, Nenov MN
89 attenuates the bioluminescence signal pro- et al (2013) The fibroblast growth factor 14
duced by Renilla luciferase. PLoS One 4: (FGF14)/voltage-gated sodium channel com-
e5642 plex is a new target of glycogen synthase kinase
514 Alexander S. Shavkunov et al.
3 (GSK3). J Biol Chem 288 26. Goetz R, Dover K, Laezza F et al (2009) Crys-
(27):19370–19385 tal structure of a fibroblast growth factor
12. Thayer DA, Jan LY (2010) Mechanisms of dis- homologous factor (FHF) defines a conserved
tribution and targeting of neuronal ion chan- surface on FHFs for binding and modulation of
nels. Curr Opin Drug Discov Devel voltage-gated sodium channels. J Biol Chem
13:559–567 284:17883–17896
13. Leterrier C, Brachet A, Fache MP et al (2010) 27. Laezza F, Gerber BR, Lou JY et al (2007) The
Voltage-gated sodium channel organization in FGF14(F145S) mutation disrupts the interac-
neurons:protein interactions and trafficking tion of FGF14 with voltage-gated Na + chan-
pathways. Neurosci Lett 486:92–100 nels and impairs neuronal excitability. J
14. Catterall WA (2010) Signaling complexes of Neurosci 27:12033–12044
voltage-gated sodium and calcium channels. 28. Lou JY, Laezza F, Gerber BR et al (2005)
Neurosci Lett 486:107–116 Fibroblast growth factor 14 is an intracellular
15. Jubb H, Higueruelo AP, Winter A et al (2012) modulator of voltage-gated sodium channels. J
Structural biology and drug discovery for pro- Physiol 569:179–193
tein–protein interactions. Trends Pharmacol 29. Goldfarb M, Schoorlemmer J, Williams A et al
Sci 33:241–248 (2007) Fibroblast growth factor homologous
16. Smith MC, Gestwicki JE (2012) Features of factors control neuronal excitability through
protein–protein interactions that translate into modulation of voltage-gated sodium channels.
potent inhibitors: topology, surface area and Neuron 55:449–463
affinity. Expert Rev Mol Med 14:e16 30. Dover K, Solinas S, D’Angelo E et al (2010)
17. Wells JA, McClendon CL (2007) Reaching for Long-term inactivation particle for voltage-
high-hanging fruit in drug discovery at pro- gated sodium channels. J Physiol 589(Pt
tein–protein interfaces. Nature 6):1505
450:1001–1009 31. Luker KE, Piwnica-Worms D (2004) Optimiz-
18. Buchwald P (2010) Small-molecule protein–- ing luciferase protein fragment complementa-
protein interaction inhibitors: therapeutic tion for bioluminescent imaging of
potential in light of molecular size, chemical protein–protein interactions in live cells and
space, and ligand binding efficiency considera- animals. Methods Enzymol 385:349–360
tions. IUBMB Life 62:724–731 32. Yang KS, Ilagan MX, Piwnica-Worms D et al
19. Zinzalla G, Thurston DE (2009) Targeting (2009) Luciferase fragment complementation
protein–protein interactions for therapeutic imaging of conformational changes in the epi-
intervention: a challenge for the future. Future dermal growth factor receptor. J Biol Chem
Med Chem 1:65–93 284:7474–7482
20. Clare JJ (2010) Targeting ion channels for 33. Ilagan MX, Lim S, Fulbright M et al (2011)
drug discovery. Discov Med 9:253–260 Real-time imaging of notch activation with a
luciferase complementation-based reporter. Sci
21. Wickenden A, Priest B, Erdemli G (2012) Ion Signal 4:rs7
channel drug discovery: challenges and future
directions. Future Med Chem 4:661–679 34. Garrido JJ, Fernandes F, Giraud P et al (2001)
Identification of an axonal determinant in the
22. Kiss L, Bennett PB, Uebele VN et al (2003) C-terminus of the sodium channel Na(v)1.2.
High throughput ion-channel pharmacology: EMBO J 20:5950–5961
planar-array-based voltage clamp. Assay Drug
Dev Technol 1:127–135 35. Thorne N, Auld DS, Inglese J (2010) Apparent
activity in high-throughput screening: origins
23. Masi A, Cicchi R, Carloni A et al (2010) Opti- of compound-dependent assay interference.
cal methods in the study of protein–protein Curr Opin Chem Biol 14:315–324
interactions. Adv Exp Med Biol 674:33–42
36. Villalobos V, Naik S, Bruinsma M et al (2010)
24. Terstappen GC (2004) Nonradioactive rubid- Dual-color click beetle luciferase heteroprotein
ium ion efflux assay and its applications in drug fragment complementation assays. Chem Biol
discovery and development. Assay Drug Dev 17:1018–1029
Technol 2:553–559
37. Lazo JS, Brady LS, Dingledine R (2007)
25. Laezza F, Lampert A, Kozel MA et al (2009) Building a pharmacological lexicon: small
FGF14 N-terminal splice variants differentially molecule discovery in academia. Mol Pharma-
modulate Nav1.2 and Nav1.6-encoded sodium col 72:1–7
channels. Mol Cell Neurosci 42:90–101
Chapter 34
Abstract
Confocal laser scanning microscopy is the best method to visualize intracellular co-localization of proteins
in intact cells. Because of the point scan/pinhole detection system, light contribution from the neighbor-
hood of the scanning spot in the specimen can be eliminated, allowing high Z-axis resolution. Fluorescence
detection by sensitive photomultiplier tubes allows the usage of filters with a narrow bandpath, resulting in
minimal cross-talk (overlap) between two spectra. This is particularly important in demonstrating co-
localization of proteins with multicolor labeling. Here, the methods outlining the detection of transiently
expressed tagged proteins and the detection of endogenous proteins are described. Ideally, the intracellular
co-localization of two endogenous proteins should be demonstrated. However, when antibodies raised
against the protein of interest are unavailable for immunofluorescence or the available cell lines do not
express the protein of interest sufficiently enough for immunofluorescence, an alternative method is to
transfect cells with expression plasmids that encode tagged proteins and stain the cells with anti-tag
antibodies. However, it should be noted that the tagging of proteins of interest or their overexpression
could potentially alter the intracellular localization or the function of the target protein.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_34, © Springer Science+Business Media New York 2015
515
516 Toshiyuki Miyashita
Photomultiplier
Pinhole
Pinhole
Pinhole
Laser Beam Splitter
(Dichroic Mirror)
Scanner
Objective Lens
2 Materials
3 Methods
3.1 Detection of When antibodies raised against a protein of interest are unavailable
Transiently Expressed for immunofluorescence or the available cell lines do not express
GFP- and HA-Tagged the protein of interest sufficiently enough for immunofluorescence,
Proteins an alternative method is to transfect cells with expression plasmids
that encode tagged proteins and stain the cells with anti-tag
antibodies. In addition, the protein of interest can be fused with a
fluorophore for direct detection. Currently, a wide variety of
fluorescent proteins have been developed for generating fusion
proteins. By selecting a proper combination (e.g., EGFP and
DsRed2-Monomer), it is possible to investigate the subcellular
518 Toshiyuki Miyashita
Table 1
Approximate spectral properties of representative fluorophores used for multicolor labeling
Absorption Emission
Fluorophore (nm) (nm) Laser Notes
Group 1 (green)
FITC 490 512 Ar.488 Most widely used green fluorescent dye,
easy to photobleach
EGFP 488 507 Ar.488 Brighter than wild-type GFP, for
generating fusion proteins
Alexa Fluor 488 495 519 Ar.488 Brighter and more photostable than FITC
Group 2 (red)
TRITC 541 572 HeNe543 or Commonly used red fluorescent dye in
Ar.Kr.568 combination with FITC
Texas red 596 620 HeNe543 or Good spectral separation from FITC,
Ar.Kr.568 slightly higher background staining [4]
DsRed2-monomer 556 586 HeNe543 or For generating fusion proteins
Ar.Kr.568
Cy3 552 565 HeNe543 or Brighter than TRITC
Ar.Kr.568
Alexa Fluor 546 556 573 HeNe543 or Brighter than TRITC or Cy3
Ar.Kr.568
Propidium iodide 530 615 HeNe543 or For DNA/RNA staining
Ar.Kr.568
Group 3 (blue)
DAPI 345 425 UV.Ar.364 For nuclear staining
Hoechst33342 355 465 UV.Ar.364 For nuclear staining
For multicolor labeling, choose one fluorophore from each group depending on the laser sources equipped in the CLSM,
thus allowing up to three-color labeling
3.1.1 Transfection 1. The day before transfection, seed HeLa cells at a density of
7 104 cells/well (when two well chamber slides are used).
2. Incubate the cells at 37 C and 5 % CO2 in DMEM medium.
3. Prepare 0.5 μg of each plasmid that encodes GFP- or HA-
tagged protein. The minimum DNA concentration should be
0.1 μg/μl.
4. Dilute the DNA in 60 μl of Buffer EC (Effectene transfection
reagent kit). Add 6 μl of Enhancer and mix by vortexing for 1 s
(see Note 2).
5. Incubate at room temperature for 2–5 min and spin down the
mixture for a few seconds.
6. Add 10 μl of Effectene Transfection Reagent to the mixture.
Mix by vortexing for 10 s.
7. Incubate the samples for 5–10 min at room temperature to
allow complex formation.
8. During step 7, aspirate the medium from the slides and wash
the cells once with PBS. Add 1 ml of fresh growth medium to
each well.
9. Add the transfection complexes drop-wise onto the cells.
Gently swirl the plates to ensure uniform distribution of the
complexes.
10. Incubate the cells with the complexes at 37 C and 5 % CO2 for
24 h to allow for gene expression.
3.1.2 Immunofluore- 1. Carefully aspirate the medium and add 0.5 ml/well of 4 %
scence paraformaldehyde in PBS drop-wise onto the cells (see Note 3).
2. To fix the cells, incubate the samples for 1 h at 4 C.
3. Carefully replace the solution with 0.5 ml/well of permeabili-
zation solution.
4. Incubate the samples for 5 min at room temperature.
5. Carefully replace the solution with 0.5 ml/well of preblock
solution and incubate the samples for 1 h at room temperature.
6. Carefully aspirate the preblock solution and add 0.5 ml/well of
anti-HA mouse monoclonal antibody diluted with preblock
solution 1:200.
7. Incubate the samples for 1 h at room temperature (see Note 4).
8. Carefully aspirate the antibody solution and add 1 ml/well of
PBS.
520 Toshiyuki Miyashita
3.1.3 Confocal Laser Since the operation procedure of a CLSM depends on manufac-
Scanning Microscopy turer and systems, it is not possible to generalize this procedure.
This chapter describes techniques for the Olympus CLSM. The
manufacturer’s manual should be consulted for details.
1. Choose the correct combination of laser, barrier filters, and
excitation dichroic mirrors for the dyes in use according to
the manufacturer’s recommendation (for GFP/TRITC, the
following combination is appropriate. Laser combination;
Ar488nm + HeNe543nm, Barrier filters; BA510IF +
BA530RIF for channel 1, BA565IF + BA590 for channel 2,
dichroic mirror; DM488/543).
2. Set the power switch of each unit to ON. To stabilize the laser
beam output, allow the system to warm up for at least 10 min
after turning the laser power ON.
3. Start the FLUOVIEW software by double-clicking the FLUO-
VIEW icon on the desktop.
4. Place the specimen in an inverted position (in combination
with an inverted microscope) on the microscope stage and
turn the light path selector to the “Binocular” section.
5. Engage the optimum cube for specimen dye by operating the
cube turret and focus on the specimen initially using transmit-
ted light and then quickly observe the cells with fluorescence.
6. When the area to be observed with confocal microscopy is
determined, turn the light path selector to the “Side port”
position and rotate the cube turret so that no cube is engaged.
Confocal Microscopy 521
7. Choose the highest scan speed and set the zoom ratio to “X1.”
Set the channel to be acquired (brightness should be adjusted
one channel at a time).
8. Click the “Focus” button to acquire repeated images at a high
speed.
9. Focus on the cells of interest and adjust the image brightness.
Parameters to be considered at this step are “PMT voltage,”
“Offset,” and “Gain,” each of which can be adjusted indepen-
dently. The ND filters can also be selected using the LASER
INTENSITY turret. Select the optimum ND filters according
to the brightness of the specimen. Please note that a strong
laser intensity will make the fluorescence fade quickly.
10. When it is necessary to observe the detail of a specific area, the
image of a limited area can be acquired by using the “Zoom”
scale and the “Pan” buttons.
11. Once the appropriate parameters have been determined, stop
repeated scanning, set a lower scan speed and acquire the image
by selecting the “Once” button (see Notes 6 and 7).
12. After the desired image has been acquired, save it to a disk. It is
possible to save images acquired with more than one channel at
a time. The user can select the file types used for saving an
image in a file. Fluoview Multi Tiff format is used for image
analysis and processing on FLUOVIEW. Single TIF is a format
for maximum portability between Macintosh and IBM PCs and
programs such as Adobe Photoshop. To demonstrate the co-
localization of two proteins, the merged image of the two
proteins in a single view should be shown as well as displaying
images of the two channels side by side. For example, when the
expression of a GFP-fused protein is displayed in green and that
of an HA-tagged protein is shown in red, then co-localization
of the two proteins is visualized in yellow in the merged image
(Fig. 2a–c).
Fluorophore α
Fluorophore β
(1)
(2)
X Y X Y
antibodies should (1) be derived from the same host species so that
they do not recognize one another (Fig. 2 (1)), (2) not cross-react
with other primary antibodies used in the assay system (Fig. 2 (2)),
(3) not cross-react with endogenous proteins present in the cell
lines under investigation. Secondary antibodies absorbed against
the sera of a number of species to minimize cross-reactivity are
commercially available and are usually marked as “highly cross-
absorbed” or “for multiple labeling.” When cells growing in sus-
pension are to be stained, they should be cytocentrifuged prior to
the fixation using a Cytospin or equivalent that allows low-speed
centrifugal force to separate the cells on slides while maintaining
cellular integrity.
1. Carefully aspirate the medium and add 0.5 ml/well of 4 %
paraformaldehyde in PBS drop-wise onto the cells (see Note 3).
2. To fix the cells, incubate the samples for 1 h at 4 C.
3. Carefully replace the solution with 0.5 ml/well of permeabili-
zation solution.
4. Incubate the samples for 5 min at room temperature.
5. Carefully replace the solution with 0.5 ml/well of preblock
solution and incubate the samples for 1 h at room temperature.
6. Carefully aspirate the preblock solution and add 0.5 ml/well of
anti-protein X mouse monoclonal antibody diluted with
Confocal Microscopy 523
Fig. 3 Multicolor labeling of HeLa cells. (a–c) HeLa cells were transfected with pSDHIp-EGFP and pHA-Bak
that encode the EGFP-tagged iron sulfur subunit of human succinate dehydrogenase [5] and HA-tagged
proapoptotic Bcl-2 family member, Bak [6, 7], respectively. At 24 h after transfection, cells were stained with
anti-HA antibody followed by TRITC-labeled secondary antibody. Panels (a) and (b) show the subcellular
localizations of SDHIp and Bak, respectively. A merged picture is shown in panel (c). SDHIp is a mitochondrial
protein and Bak is shown to co-localize with SDHIp. Arrows indicate cells transfected with only pSDHIp-EGFP.
(d–f) HeLa cells were incubated with mouse anti-GM130 monoclonal antibody (BD transduction Laboratories)
and rabbit anti-DRPLA antibody [8]. The primary antibodies were detected by secondary antibodies conjugated
to Alexa Fluor 488 or 546. DRPLA protein, a product of the gene responsible for a genetic neurodegenerative
disease, dentatorubral-pallidoluysian atrophy (DRPLA) [9], is localized in the nucleus [10]. However, a closer
observation reveals that it is also localized in the juxtanuclear region (arrowhead) where it co-localizes with
GM130 (f), which is a cis-Golgi network marker [11]. (g–i) HeLa cells were treated similarly as in (d–f) except
that anti-PML mouse monoclonal antibody (Santa Cruz) was used as one of the primary antibodies. The
promyelocytic leukemia (PML) protein is a major component of nuclear dot-like structures known as PML
nuclear bodies (NBs) or PML oncogenic domains (POD) [12, 13] (g). These panels show that the nuclear DRPLA
protein is not recruited to NBs or POD
Confocal Microscopy 525
4 Notes
Acknowledgments
References
1. Abe Y, Oka A, Mizuguchi M, Igarashi T, Modulation of apoptosis by the widely
Ishikawa S, Aburatani H, Yokoyama S, Asahara distributed Bcl-2 homologue Bak. Nature
H, Nagao K, Yamada M, Miyashita T (2009) 374:736–739
EYA4, deleted in a case with middle interhemi- 8. Miyashita T, Okamura-Oho Y, Mito Y, Naga-
spheric variant of holoprosencephaly, interacts fuchi S, Yamada M (1997) Dentatorubral pal-
with SIX3 both physically and functionally. lidoluysian atrophy (DRPLA) protein is cleaved
Hum Mutat 30:E946–E955 by caspase-3 during apoptosis. J Biol Chem
2. Yamagata K, Suetsugu R, Wakayama T (2009) 272:29238–29242
Long-term, six-dimensional live-cell imaging 9. Nagafuchi S, Yanagisawa H, Ohsaki E, Shir-
for the mouse preimplantation embryo ayama T, Tadokoro K, Inoue T, Yamada M
that does not affect full-term development. (1994) Structure and expression of the gene
J Reprod Dev 55:343–350 responsible for the triplet repeat disorder, den-
3. Sambrook J, Russell DW (2001) Molecular tatorubral and pallidoluysian atrophy
cloning, a laboratory manual, 3rd edn. Cold (DRPLA). Nat Genet 8:177–182
Spring Harbor Laboratory Press, Cold Spring 10. Miyashita T, Nagao K, Ohmi K, Yanagisawa H,
Harbor, NY Okamura-Oho Y, Yamada M (1998) Intracel-
4. Wessendorf MW, Brelje TC (1992) Which lular aggregate formation of dentatorubral-
fluorophore is brightest? A comparison of the pallidoluysian atrophy (DRPLA) protein with
staining obtained using fluorescein, tetra- the extended polyglutamine. Biochem Biophys
methylrhodamine, lissamine rhodamine, Texas Res Commun 249:96–102
red, and cyanine 3.18. Histochemistry 11. Nakamura N, Rabouille C, Watson R, Nilsson
98:81–85 T, Hui N, Slusarewicz P, Kreis TE, Warren G
5. Kita K, Oya H, Gennis RB, Ackrell BC, Kasa- (1995) Characterization of a cis-Golgi matrix
hara M (1990) Human complex II (succinate- protein, GM130. J Cell Biol 131:1715–1726
ubiquinone oxidoreductase): cDNA cloning 12. Dyck JA, Maul GG, Miller WHJ, Chen JD,
of iron sulfur (Ip) subunit of liver mitochon- Kakizuka A, Evans RM (1994) A novel macro-
dria. Biochem Biophys Res Commun molecular structure is a target of the
166:101–108 promyelocyte-retinoic acid receptor oncopro-
6. Chittenden T, Harrington EA, O’Connor R, tein. Cell 76:333–343
Flemington C, Lutz RJ, Evan GI, Guild BC 13. Weis K, Rambaud S, Lavau C, Jansen J, Carvalho
(1995) Induction of apoptosis by the Bcl-2- T, Carmo-Fonseca M, Lamond A, Dejean A
homologue Bak. Nature 374:733–736 (1994) Retinoic acid regulates aberrant nuclear
7. Kiefer MC, Brauer MJ, Powers VC, Wu JJ, localization of PML-RAR alpha in acute pro-
Umansky SR, Tomei LD, Barr PJ (1995) myelocytic leukemia cells. Cell 76:345–356
Part V
Abstract
Fluorescence polarization (FP) technology is based on the measurement of molecule rotation, and has been
widely used to study molecular interactions in solution. This method can be used to measure binding and
dissociation between two molecules if one of the binding molecules is relatively small and fluorescent. The
fluorescently labeled small molecule (such as a small peptide) rotates rapidly in the solution. Upon
excitation by polarized light, the emitted light remains depolarized and gives rise to a low FP signal.
When the fluorescent small molecules in solution are bound to bigger molecules (such as a protein), the
movement of the complex becomes slower. When such a complex is excited with polarized light, much of
the emitted light is polarized because of the slow movement of the complex. Thus, the binding of a
fluorescently labeled small molecule to a bigger molecule can be monitored by the change in polarization
and measured by the generation of an increased FP signal. This chapter aims to provide a step-by-step
practical procedure for developing an FP assay in a multi-well plate format to monitor protein-protein
interaction (PPI) in a homogenous format.
Key words Fluorescence polarization (FP), Protein-protein interaction (PPI), Small molecule, Probe,
Multi-well plate, Plate reader, Dissociation constant (Kd), Competition assay
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_35, © Springer Science+Business Media New York 2015
529
530 Yuhong Du
Polarized
Polarized bigger molecules
excitation light
excitation light (protein, > 10, 000 Da)
Probe
(fluorescent
small molecules,
< 1500 Da)
Low FP FP signal
The tracer alone without protein should give rise to only minimal
FP signal for the free tracer. Increasing FP signal with increasing
protein concentrations in the presence of tracer should be observed
if the protein binds to the tracer. At higher protein concentrations,
the FP signal should rise to a plateau that corresponds to complete
binding of all tracers. A binding curve can be generated by plotting
FP signal against protein concentration, and used for dissociation
constant (Kd) calculation.
The following protocol describes a general step-by-step proce-
dure of developing an FP binding assay to monitor the interaction
of two biomolecules. The results of FP assay development for
monitoring the interaction of 14-3-3 protein with a fluorescently
labeled phosphopeptide binding partner are presented as a case
study [2]. The 14-3-3 proteins mediate phosphorylation-
dependent protein-protein interactions. Through binding to
numerous client proteins, 14-3-3 controls a wide range of physio-
logical processes and has been implicated in a variety of diseases,
including cancer and neurodegenerative disorders [3]. In order to
better understand the structure and function of 14-3-3 proteins
and to develop small molecule modulators of 14-3-3 proteins for
physiological studies and potential therapeutic interventions, a
highly sensitive FP-based 14-3-3 assay was designed and optimized.
Using the interaction of 14-3-3 with a fluorescently labeled phos-
phopeptide derived from Raf-1 as a model system (see Fig. 3a), the
detailed procedure to develop an FP assay for monitoring the
interaction of 14-3-3 protein with its binding partners is described.
This 14-3-3 FP binding assay is a simple one-step “mix-and-read”
method for analyzing 14-3-3 proteins. This solution based, versa-
tile method can be used to monitor the binding of 14-3-3 with a
variety of client proteins. While this chapter uses 14-3-3 protein
interactions as an example, the procedure described in this chapter
can be adapted and applied to any FP-suited system for monitoring
the interaction of two molecules.
2 Materials
5. Glutathione-Sepharose beads.
6. Elution buffer: PBS, pH 9, containing 20 mM reduced gluta-
thione, 0.25 mM PMSF, 5 mM dithiothreitol (DTT), and
0.1 % Triton X-100.
7. De-salt gel filtration column (such as a PD-10 gel filtration
column from GE Healthcare).
8. Fluorescent Probe (TMR-pS259-Raf): a phosphopeptide
derived from a well-studied 14-3-3 binding protein, Raf-1,
was synthesized and labeled with 5/6 carboxytetramethylrho-
damine (TMR). The TMR-pS259-Raf contains 15 residues:
5/6-TMR-LSQRQRST[pS]TPNVHM, and was dissolved in
HEPES buffer as 200 μM stock and stored at 20 C
before use.
9. Unlabeled antagonist or control peptides: R18
(PHCVPKNLSWLNLEAN MCLP) [4] and mutated R18,
R18Lys, where D12 and E14 were changed to K [5], were
dissolved in HEPES buffer as 200 μM stock and stored at
20 C before use.
10. Plate: 384-well solid bottom black plate (see Note 2).
11. Plate reader for FP signal measurements (see Note 3): For 14-
3-3 FP assay development, FP measurements were performed
on an Analyst HT plate reader (Molecular Devices) using an FP
protocol. For the tetramethylrhodamine (TMR)-labeled probe
(Ex: 545 nm; Em: 610 nm), a dichroic mirror of 565 nm was
used.
3 Methods
3.1 Protein E. coli BL21 (DE3) strains were grown in LB medium with ampi-
Purification cillin (100 μg/ml) for protein auto-induction. After overnight
incubation with shaking at 37 C, the cells were harvested and the
pellet was resuspended in ice-cold resuspension buffer. The GST-
14-3-3 γ protein was then purified by using Glutathione-Sepharose
beads, and the bound protein was eluted in elution buffer. Salts
were removed from the pooled elution fractions containing the
GST-14-3-3γ fusion protein using a PD-10 gel filtration column
equilibrated in assay buffer without DTT. The protein concentra-
tion was measured and GST-14-3-3γ protein was stored at 20 C
before use.
Fig. 2 An example plate format for probe (a) and protein (b) titrations for FP assay development in a 384-well
plate format. The FP binding assay is then preformed by adding 15 μl of probe at a selected concentration to
the serially diluted protein. The final volume for each well of the 384-well plate is 30 μl
8. Centrifuge the plate at 1,000 rpm for 2 min to get rid of any
bubbles in the wells. This step is important especially for
handling low volumes, as the bubbles will affect the reading.
9. Load the plate on the plate reader.
10. Set up the proper settings for the instrument software for the
FP measurement.
For 14-3-3 FP assay development, an Analyst HT plate
reader was used. An integration time of 100 ms was used, and
the Z height was set at 2.15 mm (middle). The excitation
polarization was set at “static,” and emission polarization was
set at “dynamic.” For the TMR-labeled probe, an excitation
filter at 545 nm and an emission filter at 610–675 nm were used
with a dichroic mirror of 565 nm. Three panels of the data were
obtained from the FP measurement (see Note 3).
11. Data analysis for peptide titration:
Caluculate the average fluorescence intensity (FI) in parallel
channel for each sample. The concentrations of peptide that
exhibit about ten times or more FI signal compared to buffer-
only background controls should be selected for the following
FP binding assay development (see Note 4).
For the 14-3-3 FP binding assay, 1 nM TMR-pS259-Raf
peptide was chosen based on the observation that this concen-
tration of the TMR-labeled peptide exhibited about ten times
more FI signal compared to buffer-only background controls.
3.3 Development of After selecting the probe concentration, the binding of 14-3-3
the FP Binding Assay protein to TMR-pS259-Raf peptide was performed by titrating
(See Note 5) 14-3-3 protein using a fixed concentration of TMR-pS259-Raf
peptide (1 nM) (see Note 6). Serial dilution of protein was carried
out using a similar procedure as the peptide serial dilution except
the initial volume for each well was different. An example plate
format for protein dilution is shown in Fig. 2b, the final volume for
each well was 15 μl. The FP binding reaction was then performed
by adding 15 μl of peptide at the selected final concentration to all
wells containing increasing concentrations of protein.
1. Add 15 μl of assay buffer to columns 1–11, rows D, E, and F, as
triplicates, in the same plate as the peptide titration.
2. In a 1.5 ml microcentrifuge tube, make 100 μl of 2 of the
highest concentration of protein tested (e.g., 5 μM of GST-14-
3-3γ protein) in assay buffer.
3. Pipette 30 μl of 5 μM GST-14-3-3γ protein to column 12, rows
D, E, and F as triplicates.
4. Load three tips using a multichannel pipette for 384-well
plates, transfer 15 μl of the highest concentration of protein
Fluorescence Polarization for Bimolecular Interactions 535
3.4 Data Analysis for The dynamic range of the FP binding assay (i.e., the FP assay
the FP Binding Assay window) is defined by the difference of the measured FP signal
between bound fluorescent probe in the presence of protein and
unbound (free) probe in the absence of protein.
1. Calculate the FP assay window for each protein concentration
using the following equation:
Fig. 3 Development of an FP binding assay for monitoring 14-3-3 protein interactions. (a) 14-3-3 binding FP
assay design. The 14-3-3 binding peptide, pS259-Raf, is labeled with 5/6 carboxytetramethylrhodamine
(TMR). The unbound peptide (fluorescent small molecule) rotates fast in solution and gives a low FP signal
when excited with polarized light. The association of 14-3-3 protein (large molecule) slows down the rotation
of the peptide, leading to an increased FP signal. (b) Interaction of 14-3-3 TMR-pS259-Raf peptide induces the
generation of an FP signal. TMR-pS259-Raf peptide (1 nM, final) was incubated with increasing concentra-
tions of GST-14-3-3γ or GST proteins in a 384-well black microplate. The FP signals were recorded after 1 h
of incubation using an Analyst HT. (c) The assay window was calculated by subtracting free peptide
polarization values from values of bound peptide recorded in the presence of specified protein concentrations
of GST-14-3-3. The data were fit to the model FP = ð½14-3-3 BmaxÞ=ð½14-3-3 þ KdÞ using nonlinear
regression analysis, where Bmax is the maximal binding (Prism 4.0; Graphpad). Data shown are the average
values from triplicate samples with standard deviation (SD)
3.5 Development of Once the FP binding assay has been developed with a reasonable
an FP Competition maximum FP signal window (above ~60 mP), the specificity of the
Assay to Validate the FP signal should be evaluated in a competition FP binding assay
Specificity of the FP format. Such an assay works by measurement of the decrease in FP
Binding Signal signal caused by a competitor, e.g., an un-labeled or antagonist
ligand that displaces the binding of the labeled probe to the pro-
tein. To establish the competition assay, titration of a competitor to
a reaction mixture containing fixed concentrations of protein and
probe is performed as follows.
1. Select the protein concentration for the competition assay.
A concentration of the protein should be chosen that gen-
erates a signal between the EC50 to EC80 of the maximum FP
signal window based on the established FP binding curve.
Higher concentrations of the protein will lie in the plateau
region of the binding curve, which may lead to the competition
assay being insensitive to competitors. Lower concentrations of
the protein will not provide a good assay window for measure-
ment of the decreased signal by displacement of the probe.
Based on the 14-3-3 FP binding curve (Fig. 3b), 0.5 μM
GST-14-3-3γ was selected for the competition assay. This con-
centration of the protein produced ~70 mP FP assay window
and was close to its Kd or EC50 concentration (0.4 μM).
2. Prepare serially diluted competitors in a separate plate (see
Note 8).
In a 384-well round bottom plate, make 5 serially diluted
antagonist or control peptide in assay buffer using the
procedures described in Subheadings 3.2 and 3.3. An example
plate format is given in Fig. 4a.
3. Prepare two mixtures containing 1.25 the selected concen-
tration of peptide and protein in two 1.5 ml microcentrifuge
tubes. Mixture 1 should contain 1.25 nM TMR-pS259-Raf
peptide only and mixture 2 should contain both the peptide
and protein at 1.25 the selected concentrations (1.25 nM of
TMR-pS259-Raf and 0.63 μM of GST-14-3-3γ).
538 Yuhong Du
Fig. 4 An example plate format and procedure for developing a competition FP assay. (1) Make serial dilutions
of competitors in a 384-well round-bottom plate. (2) Prepare an assay plate in a 384-well black plate
containing selected concentrations of probe and protein. Probe only without protein is included as a
background control which gives rise to minimal FP signals. (3) Perform the competition assay by transferring
the competitor to assay plate with triplicate samples
Fig. 5 Competition FP assay. (a) Competitive inhibition of 14-3-3 antagonist peptide, R18, on the 14-3-3/Raf
peptide interaction FP assay. A mutant derivative of R18, R18Lys, which cannot bind 14-3-3, was used as a
negative control. Increasing concentrations of R18 or R18Lys were added to the reaction containing 1 nM
TMR-pS259-Raf peptide and 0.5 μM GST-14-3-3γ. After incubation for 1 h at room temperature, the FP
signals were recorded. Percent of control was calculated as described in Subheading 3.5 and plotted against
peptide concentration. (b) Competition of R18 peptide for the binding of TMR-pS259-Raf peptide to increasing
concentrations of GST-14-3-3γ. Increasing concentrations of R18 peptide were added to four panels of
reactions containing 0.25, 0.5, 1, and 2.5 μM of GST-14-3-3γ and 1 nM of TMR-pS259-Raf peptide. The FP
signals were recorded after 1 h incubation at RT. Percent of control were calculated and plotted against R18
peptide concentrations. Data shown are the average signal from triplicates with SD
4 Notes
5 Summary
Acknowledgments
References
1. Perrin F (1926) Polarization of light of fluores- regulation. Annu Rev Pharmacol Toxicol
cence, average life of molecules. J Phys Radium 40:617–647
7:390–401 4. Wang B, Yang H, Liu YC et al (1999) Isolation
2. Du Y, Masters SC, Khuri FR et al (2006) Moni- of high-affinity peptide antagonists of 14-3-3
toring 14-3-3 protein interactions with a homo- proteins by phage display. Biochemistry
geneous fluorescence polarization assay. J 38:12499–12504
Biomol Screen 11:269–276 5. Masters SC, Fu H (2001) 14-3-3 proteins medi-
3. Fu H, Subramanian RR, Masters SC (2000) ate an essential anti-apoptotic signal. J Biol
14-3-3 proteins: structure, function, and Chem 276:45193–45200
544 Yuhong Du
6. Jameson DM, Croney JC (2003) Fluo- perspectives and primer. J Biomol Screen
rescence polarization: past, present and future. 5:297–306
Comb Chem High Throughput Screen 8. Burke TJ, Loniello KR, Beebe JA et al (2003)
6:167–173 Development and application of fluorescence
7. Owicki JC (2000) Fluorescence polarization and polarization assays in drug discovery. Comb
anisotropy in high throughput screening: Chem High Throughput Screen 6:183–194
Chapter 36
Abstract
Time-resolved fluorescence resonance energy transfer, TR-FRET, is a time-gated fluorescence intensity
measurement which defines the relative proximity of two biomolecules (e.g., proteins, peptides, or DNA)
based on the extent of non-radiative energy transfer between two fluorophores with overlapping emission/
excitation spectra. In these assays, an excited lanthanide ion acts as a “donor” that transfers energy to an
“acceptor” fluorophore through dipole–dipole interactions. A FRET signal is reported as the ratio of
acceptor to donor emission following donor excitation. When a donor-conjugated protein interacts with
an acceptor-conjugated protein, the donor and acceptor fluorophores are brought in close proximity
allowing energy transfer from the donor to the acceptor resulting in a FRET signal. Because the lanthanide
donors have a long emission half-life, the energy transfer measurement can be time-gated, which dramati-
cally reduces assay interference (due to background autofluorescence and direct acceptor excitation) and
thereby increases data quality. Here, we describe a TR-FRET assay that monitors the interaction of the
estrogen receptor (ER) α ligand binding domain (labeled with a terbium chelate via a streptavidin–biotin
interaction) with a sequence of coactivator protein SRC3 (labeled directly with fluorescein) and the
disruption of this interaction with a peptide and a small molecule inhibitor.
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_36, © Springer Science+Business Media New York 2015
545
546 Terry W. Moore et al.
340 nm
(D λex) 520 nm
FRET(i.e.A λex) (A λem)
Tb
Fl
ER
SRC
SA B E2
495 nm
340 nm
(D λem)
(D λex)
Tb
Fl
ER
SA CBI SRC
B
E2
2 Materials
3 Methods
Fig. 2 Representative data from a fluorescence resonance energy transfer (FRET) assay. By plotting the ratio of
the emission intensities of the acceptor to the donor (A/D 1,000) against the log of ligand molar
concentration, dose–response curves for displacement of SRC-3-NRD-fluorescein by the steroid receptor
coactivator peptide (left ) and pyrimidine coactivator binding inhibitor 1 (right) can be generated. Varying the
concentration of the agonist 17β-estradiol (E2; 500 nM (filled square) and 50 μM (filled triangle)) has no
substantial effect on the IC50 values of the compounds, implying that these positive control compounds do not
compete with E2 for the ligand-binding pocket but act by direct displacement of the SRC-3-NRD-fluorescein
4 Notes
HN
N NH
pyrimidine CBI 1
14. This stock solution can be divided into eight wells of a dilution
plate so that it can be dispensed into the final black plate using
an autopipettor.
15. This solution should be kept in a cooling block on ice. Any
extra should be discarded at the end of the day.
16. We prefer eight concentrations for dose–response assays using
1:10 and 1:3 serial dilutions. For example, to make approxi-
mately 100 μL of each concentration point, put 100 μL of
20 mM solution in the first well. Make the second well by
adding 30 μL of 20 mM stock to 70 μL of DMF. Then serially
dilute each of these two wells 1:10 into DMF. In this case, the
DMF concentrations would be 20, 6, 2, 0.6, 0.2, 0.06, 0.02,
and 0.006 mM.
17. We often use DMF to prepare stock solutions because DMF
does not freeze and is not as hygroscopic as DMSO. DMF is,
however, less inert than DMSO, and, in other systems, it may
denature proteins. We have found that, in this assay, either
DMSO or DMF can be used with no noticeable effect on
assay performance.
18. For example, add 10 μL of DMF solution to 90 μL TR-FRET
buffer. At this point, the compound concentrations are 2,000,
600, 200, 60, 20, 6, 2, and 0.6 μM, with 10 % DMF.
19. It is not recommended to keep the diluted compounds either in
solvent or in buffer. Even with the most careful covering with
acetate covers and foil wrap, some evaporation does occur,
changing the concentration of the compounds. If the dilution
plates are to be used for several hours, keep covered using a
solvent-resistant plate cover and wrap the plate in foil. Keep the
plate on a chilled block.
20. The 200 nM fluorescein-SRC-3-NRD stock solution can be
divided into eight wells of a dilution plate so that it can be
dispensed into the final black plate using an autopipettor.
21. At this final point, the compound concentrations are 1,000,
300, 100, 30, 10, 3, 1, and 0.3 μM, with 5 % DMF. The
concentrations of each assay component are, for ERα-417,
2 nM; for 17β-estradiol, 1 μM; and for LanthaScreen™
Streptavidin-Terbium, 0.5 nM.
22. Although we recommend waiting for an hour for incubation of
the assay components before reading, the final dose–response
curve and, therefore, Ki value are nearly identical after incuba-
tion for only 5 min. Taking a measurement at an earlier time-
point can be helpful if protein viability needs to be checked or
preliminary results are needed extremely quickly.
552 Terry W. Moore et al.
References
1. Tamrazi A, Carlson KE, Rodriguez AL et al estrogen receptor with coactivator peptides.
(2005) Coactivator proteins as determinants Angew Chem Int Ed 46:4471–4473
of estrogen receptor structure and function: 6. Ozers MS, Ervin KM, Steffen CL et al (2005)
spectroscopic evidence for a novel coactivator- Analysis of ligand-dependent recruitment of
stabilized receptor conformation. Mol Endo- coactivator peptides to estrogen receptor
crinol 19:1516–1528 using fluorescence polarization. Mol Endocri-
2. Bramlett KS, Wu YF, Burris TP (2001) Ligands nol 19:25–34
specify coactivator nuclear receptor (NR) box 7. Gowda K, Marks BD, Zielinski TK et al (2006)
affinity for estrogen receptor subtypes. Mol Development of a coactivator displacement
Endocrinol 15:909–922 assay for the orphan receptor estrogen-related
3. Mueller SO (2004) Xenoestrogens: mechan- receptor-gamma using time-resolved fluores-
isms of action and detection methods. Anal cence resonance energy transfer. Anal Biochem
Bioanal Chem 378:582–587 357:105–115
4. Rodriguez AL, Tamrazi A, Collins ML et al 8. Liu JW, Knappenberger KS, Kack H et al
(2004) Design, synthesis, and in vitro (2003) A homogeneous in vitro functional
biological evaluation of small molecule inhibi- assay for estrogen receptors: coactivator
tors of estrogen receptor a coactivator binding. recruitment. Mol Endocrinol 17:346–355
J Med Chem 47:600–611 9. Zhou GC, Cummings R, Li Y et al (1998)
5. Becerril J, Hamilton AD (2007) Helix Nuclear receptors have distinct affinities for
mimetics as inhibitors of the interaction of the coactivators: characterization by fluorescence
Estrogen Receptor/Coactivator TR-FRET Assay 553
resonance energy transfer. Mol Endocrinol 16. Parent AA, Gunther JR, Katzenellenbogen JA
12:1594–1604 (2008) Blocking estrogen signaling after the
10. Gunther JR, Moore TW, Collins ML et al hormone: pyrimidine-core inhibitors of estro-
(2008) Amphipathic benzenes are designed gen receptor-coactivator binding. J Med Chem
inhibitors of the estrogen receptor α/steroid 51:6512–6530
receptor coactivator interaction. ACS Chem 17. Shukla SJ, Nguyen DT, MacArthur R et al
Biol 3:282–286 (2009) Identification of pregnane X receptor
11. Gunther JR, Du Y, Rhoden E et al (2009) A set ligands using time-resolved fluorescence reso-
of time-resolved fluorescence resonance energy nance energy transfer and quantitative high-
transfer assays for the discovery of inhibitors of throughput screening. Assay Drug Dev Tech-
estrogen receptor-coactivator binding. J Bio- nol 7:143–169
mol Screen 14:181–193 18. Wang Y, Yang DZ, Chang A et al (2012) Syn-
12. Mathis G (1995) Probing molecular- thesis of a ligand-quencher conjugate for the
interactions with homogeneous techniques ligand binding study of the aryl hydrocarbon
based on rare-earth cryptates and fluorescence receptor using a FRET assay. Med Chem Res
energy-transfer. Clin Chem 41:1391–1397 21:711–721
13. Kim SH, Gunther JR, Katzenellenbogen JA 19. Schaufele F (2011) FRET analysis of androgen
(2010) Monitoring a coordinated exchange pro- receptor structure and biochemistry in living
cess in a four-component biological interaction cells. Methods Mol Biol 776:147–166
system: development of a time-resolved terbium- 20. Vogel KW, Marks BD, Kupcho KR et al (2010)
based one-donor/three-acceptor multicolor Improved nuclear receptor binding assays for
FRET system. J Am Chem Soc 132:4685–4692 HTS by conversion from FP to TR-FRET read-
14. Jeyakunnar M, Carlson KE, Gunther JR et al out. Endocr Rev 31
(2011) Exploration of dimensions of estrogen 21. Hilal T, Puetter V, Otto C et al (2010) A dual
potency parsing ligand binding and coactivator estrogen receptor TR-FRET assay for simulta-
binding affinities. J Biol Chem 286: neous measurement of steroid site binding and
12971–12982 coactivator recruitment. J Biomol Screen
15. Gunther JR, Parent AA, Katzenellenbogen JA 15:268–278
(2009) Alternative inhibition of androgen 22. Tamrazi A, Carlson KE, Daniels JR et al (2002)
receptor signaling: peptidomimetic pyrimi- Estrogen receptor dimerization: ligand binding
dines as direct androgen receptor/coactivator regulates dimer affinity and dimer dissociation
disruptors. ACS Chem Biol 4:435–440 rate. Mol Endocrinol 16:2706–2719
Chapter 37
Abstract
This chapter describes the implementation of the p53–hDM2 protein-protein interaction (PPI) biosensor
(PPIB) HCS assay to identify disruptors of p53–hDM2 PPIs. Recombinant adenovirus expression
constructs were generated bearing the individual p53–GFP and hDM2–RFP PPI partners. The N-terminal
p53 transactivating domain that contains the binding site for hDM2 is expressed as a GFP fusion protein
that is targeted and anchored in the nucleolus of infected cells by a nuclear localization (NLS) sequence.
The p53–GFP biosensor is localized to the nucleolus to enhance and facilitate the image acquisition and
analysis of the PPIs. The N-terminus of hDM2 encodes the domain for binding to the transactivating
domain of p53, and is expressed as a RFP fusion protein that includes both an NLS and a nuclear export
sequence (NES). In U-2 OS cells co-infected with both adenovirus constructs, the binding interactions
between hDM2 and p53 result in both biosensors becoming co-localized within the nucleolus. Upon
disruption of the p53–hDM2 PPIs, the p53–GFP biosensor remains in the nucleolus while the shuttling
hDM2–RFP biosensor redistributes into the cytoplasm. p53–hDM2 PPIs are measured by acquiring
fluorescent images of cells co-infected with both adenovirus biosensors on an automated HCS
imaging platform and using an image analysis algorithm to quantify the relative distribution of the
hDM2–RFP shuttling component of the biosensor between the cytoplasm and nuclear regions of
compound treated cells.
Key words Protein-protein interaction biosensors, High content screening, Imaging, Image analysis
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_37, © Springer Science+Business Media New York 2015
555
556 Yun Hua et al.
Fig. 1 p53–hDM2 protein-protein interaction biosensor design and distribution phenotypes. Recombinant
adenovirus expression constructs were generated bearing the individual p53–GFP and hDM2–RFP PPI
partners. The N-terminal residues 1–131 of p53 include the transactivating domain that contains the binding
site for hDM2, and this protein fragment is expressed as a GFP fusion protein that is targeted and anchored in
the nucleolus of infected cells by the inclusion of nuclear localization (NLS) and nucleolus localization (NLOS)
sequences. The p53–GFP component is directed to the nucleolus to enhance and facilitate the image
acquisition and analysis of the PPIs. Route 1: In cells infected with the p53–GFP “bait” biosensor adenovirus
alone, the p53–GFP remains localized to bright fluorescent puncta in the nucleolus and its distribution does not
alter upon exposure to Nutlin-3 or other p53–hDM2 disruptors. The N-terminal residues 1–118 of hDM2
encode the domain for binding to the p53 transactivating domain, and this fragment is expressed as a RFP
fusion protein that includes both an NLS and a nuclear export sequence (NES). Route 2: In cells infected with
the hDM2–RFP adenovirus “prey” biosensor alone, hDM2–RFP expression is localized only in the cytoplasm of
cells and does not change upon exposure to Nutlin-3 or other p53–hDM2 disruptors. Route 3: In U-2 OS cells
that are co-infected with both adenovirus constructs, however, the binding interactions between the hDM2
and p53 components of the biosensors result in both proteins becoming localized to the nucleolus. Route 4:
Upon disruption of the p53–hDM2 protein-protein interaction with a compound like Nutlin-3, the p53–GFP
interaction partner remains nucleolar, while the shuttling hDM2–RFP interaction partner redistributes into the
cytoplasm. Route 5: Upon removal of a disrupting agent like Nutlin 3, the shuttling hDM2–RFP interaction
partner redistributes back into the p53–GFP containing nucleolus
Fig. 2 Color composite images of U-2 OS cells infected with (a) the p53–GFP biosensor alone, (b) the
hDM2–RFP biosensor alone, or (c) co-infected with both biosensors and then treated 10 μM Nutlin-3 for
90 min. U-2 OS cells were infected with (a) the p53–GFP biosensor alone, (b) the hDM2 biosensor alone, or (c)
co-infected with both biosensors and 2,500 cells were seeded into the wells of 384-well assay plates,
cultured overnight at 37 C, 5 % CO2, and 95 % humidity, and then treated 10 μM Nutlin-3 for 90 min. Cells
were then fixed and stained with Hoechst 33342 and 20 0.4 NA images of three fluorescent channels
(Ch1—Hoechst blue, Ch2—p53–GFP green, and Ch3—hDM2–RFP red) were acquired on the ArrayScan VTI
automated imaging platform as described above. Representative color composite images of control and
Nutlin-3 treated U-2 OS cells individually infected with or co-infected with both the p53–GFP and hDM2–RFP
biosensors are presented. In cells infected with each of the biosensors alone, the p53–GFP biosensor remains
localized to bright fluorescent puncta within the nucleoli of Hoechst stained nuclei and its distribution does not
alter upon exposure to Nutlin-3, while the hDM2–RFP biosensor remains localized in the cytoplasm of cells
and does not change upon exposure to Nutlin-3. In U-2 OS cells co-infected with both biosensors, however,
the two biosensors become co-localized to the nucleolus, and upon exposure to Nutlin-3 the hDM2–RFP
interaction partner redistributes into the cytoplasm while the p53–GFP interaction partner remains nucleolar
the nucleolus and its distribution does not alter upon exposure to
Nutlin-3 or when co-infected with the hDM2–RFP biosensor
(Figs. 1, route 1 and 2a). The N-terminal residues 1–118 of
hDM2 encode the domain for binding to the N-terminal transacti-
vating domain of p53, and this fragment is expressed as a RFP
fusion protein that includes both an NLS and a nuclear export
sequence (NES) (Fig. 1) [1, 2]. In cells infected with the
hDM2–RFP adenovirus “prey” biosensor alone, hDM2–RFP
p53–hDM2 Protein-Protein Interaction Biosensor HCS Assay 559
Fig. 3 Molecular translocation image analysis algorithm. U-2 OS cells co-infected with the p53–hDM2 PPIB
adenoviruses were seeded at 2,500 cells per well in 384-well Greiner collagen-coated assay plates, cultured
overnight at 37 C, 5 % CO2, and 95 % humidity, and were then exposed to the indicated concentrations of
Nutlin-3 in 0.5 % DMSO for 90 min prior to fixation with 3.7 % formaldehyde containing 2 μg/mL Hoechst
33342. Images in three fluorescent channels were sequentially acquired on the ArrayScan VTI platform using a
20 0.4 NA objective with the XF93 excitation and emission filter set and were analyzed with the molecular
translocation (MT) image analysis algorithm. (a) Image segmentation. In Ch1, Hoechst stained objects that
exhibited the appropriate morphological characteristics (width, length, and area) with fluorescent intensities
sufficiently above a background threshold were identified and classified by the image segmentation as nuclei.
The nuclear mask derived from Ch1 (dark blue ring) was then used to segment the images from Ch2 and Ch3
p53–hDM2 Protein-Protein Interaction Biosensor HCS Assay 561
2 Materials
1. Nutlin-3.
2. Formaldehyde.
3. Dimethyl sulfoxide (DMSO) (99.9 % high performance liquid
chromatography-grade, under argon).
4. Hoechst 33342.
5. U-2 OS osteosarcoma cell line.
6. Culture medium: McCoy’s 5A medium, 2 mM L-glutamine,
10 % fetal bovine serum, and 100 U/mL penicillin and
streptomycin.
7. Humidified incubator at 37 C, 5 % CO2, and 95 % humidity.
8. Dulbecco’s Mg2+ and Ca2+ free phosphate buffered saline
(PBS).
Fig. 3 (continued) into nuclear (Circ) and cytoplasmic (Ring) regions. To reduce cytoplasm contamination
within the nuclear area the nuclear mask was eroded, and the reduced mask (yellow ring) was used to
quantify the amount of target channel, p53–GFP in Ch2 and hDM2–RFP in Ch3, fluorescence within the
nuclear region. To measure fluorescence in a region of the cytoplasm in Ch2 and Ch3, the nuclear mask
derived from Ch1 was dilated to cover as much of the cytoplasm region as possible without going outside the
cell boundary. Removal of the original nuclear region from this dilated mask creates a ring mask that covers
the cytoplasm region outside the nuclear envelope. The number of pixels away from the nuclear mask and the
number of pixels (width) between the inner and outer ring masks were selectable within the MT bio-
application software. The ring masks were then used to quantify the amount of target channel, p53–GFP
(Ch2, mauve rings) or hDM2–RFP (Ch3, light blue rings), fluorescence within the cytoplasm region. (b)
Chemical structure of Nutlin-3 and concentration dependent disruption of p53–hDM2 PPIs. The molecular
translocation image analysis algorithm produces a mean average intensity difference in Ch3 (MCRAID-Ch3)
parameter calculated by subtracting the average hDM2–RFP intensity in the Ring (Cytoplasm) region from the
average hDM2–RFP intensity in the Circ (Nuclear) region of Ch3. High MCRAID-Ch3 values indicate that the
hDM2–RFP biosensor is predominantly localized within the nuclear region, while low MCRAID-Ch3 values
indicate a more prominent localization within the cytoplasm. Nutlin-3 treatment induced a concentration
dependent decrease in the hDM2–RFP MCRAID-Ch3 signal that was consistent with the redistribution of the
hDM2–RFP from the nucleus to the cytoplasm. Nutlin-3 consistently exhibited an IC50 of 0.608 0.382 μM
(n ¼ 5) for the disruption of the p53–hDM2 PPIs
562 Yun Hua et al.
3 Methods
4 Notes
References
1. Dudgeon D, Shinde SN, Shun TY, Lazo JS, HIV-1 rev mutants in living cells. Virology
Strock CJ, Giuliano KA, Taylor DL, Johnston 251:38–48
PA, Johnston PA (2010) Characterization and 8. Trask O, Nickischer D, Burton A, Williams
optimization of a novel protein-protein inter- RG, Kandasamy RA, Johnston PA, Johnston
action biosensor HCS assay to identify disrup- PA (2009) High-throughput automated con-
tors of the interactions between p53 and focal microscopy imaging screen of a kinase-
hDM2. Assay Drug Dev Technol 8:437–458 focused library to identify p38 mitogen-
2. Dudgeon D, Shinde SN, Hua Y, Shun TY, activated protein kinase inhibitors using the
Lazo JS, Strock CJ, Giuliano KA, Taylor DL, GE InCell 3000 analyzer. Methods Mol Biol
Johnston PA, Johnston PA (2010) Implemen- 565:159–186
tation of a 220,000 compound HCS campaign 9. Trask OJ Jr, Baker A, Williams RG et al (2006)
to identify disruptors of the interaction Assay development and case history of a 32 K-
between p53 and hDM2, and characterization biased library high-content MK2-EGFP trans-
of the confirmed hits. J Biomol Screen location screen to identify p38 mitogen-
15:152–174 activated protein kinase inhibitors on the
3. Johnston PA, Shinde SN, Hua Y, Shun TY, ArrayScan 3.1 imaging platform. Methods
Lazo JS, Day BW (2012) Development and Enzymol 414:419–439
validation of a high-content screening assay to 10. Kumar S, Saradhi M, Chaturvedi NK, Tyagi
identify inhibitors of cytoplasmic dynein- RK (2006) Intracellular localization and
mediated transport of glucocorticoid receptor nucleocytoplasmic trafficking of steroid recep-
to the nucleus. Assay Drug Dev Technol tors: an overview. Mol Cell Endocrinol
10:432–456 246:147–156
4. Knauer S, Moodt S, Berg T, Liebel U, Pepper- 11. Stauber R (2002) Analysis of nucleocytoplas-
kok R, Stauber RH (2005) Translocation bio- mic transport using green fluorescent protein.
sensors to study signal-specific nucleo- Methods Mol Biol 183:181–198
cytoplasmic transport, protease activity and 12. Nickischer D, Laethem C, Trask OJ Jr et al
protein-protein interactions. Traffic (2006) Development and implementation of
6:594–606 three mitogen-activated protein kinase
5. Knauer S, Stauber RH (2005) Development of (MAPK) signaling pathway imaging assays to
an autofluorescent translocation biosensor sys- provide MAPK module selectivity profiling for
tem to investigate protein-protein interactions kinase inhibitors: MK2-EGFP translocation, c-
in living cells. Anal Chem 77:4815–4820 Jun, and ERK activation. Methods Enzymol
6. Lundholt B, Heydorn A, Bjorn SP, Praeste- 414:389–418
gaard M (2006) A simple cell-based HTS 13. Williams RG, Kandasamy R, Nickischer D et al
assay system to screen for inhibitors of p53- (2006) Generation and characterization of a
Hdm2 protein-protein interactions. Assay stable MK2-EGFP cell line and subsequent
Drug Dev Technol 4:679–688 development of a high-content imaging assay
7. Stauber R, Afonina E, Gulnik S, Erickson J, on the Cellomics ArrayScan platform to screen
Pavlakis GN (1998) Analysis of intracellular for p38 mitogen-activated protein kinase inhi-
trafficking and interactions of cytoplasmic bitors. Methods Enzymol 414:364–389
Chapter 38
Abstract
The p53 protein, a tumor suppressor, is inactivated in many human cancers through mutations or by its
interaction with an oncoprotein, MDM2. Blocking the MDM2–p53 protein-protein interaction has the
effect of activating wild-type p53 and has been pursued as a novel anticancer strategy. Small-molecule
inhibitors of the MDM2–p53 interaction have been discovered through various approaches, and a number
of them have progressed into clinical trials for cancer treatment. Here, we describe the methods and
techniques used in the discovery of small-molecule inhibitors of the MDM2–p53 interaction.
Key words p53, MDM2, Protein-protein interaction, Cancer therapy, High-throughput screening,
Virtual screening
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_38, © Springer Science+Business Media New York 2015
567
568 Liu Liu et al.
1.1 Regulation of p53 The interaction between MDM2 and p53 regulates the cellular
and MDM2 basal levels and activity of p53 through an autoregulatory feedback
loop (Fig. 1). The expression of MDM2 protein is induced by p53
which binds to the P2 promoter of the MDM2 gene leading to its
transcriptional activation. MDM2 binds to p53 protein and inhibits
its activity by various mechanisms such as inhibition of the transac-
tivation function of p53, export of p53 out of the nucleus, or direct
ubiquitination of p53 leading to its proteasomal degradation
[26–28]. Thus inhibition of the MDM2–p53 interaction can lead
to activation of p53 and control of its tumor suppressing capacity.
1.2 Structural Basis In 1996, the crystal structure of HDM2 in a complex with an
of the MDM2–p53 N-terminal fragment of the p53 protein was published [29], reveal-
Interaction ing the structural basis for the MDM2–p53 protein-protein
interaction. On the MDM2 protein surface, there was observed a
well-defined binding pocket involving mostly hydrophobic resi-
dues, including Met50, Leu54, Leu57, Gly58, Ile61, Met62, Tyr67,
His73, Val75, Phe91, Val93, His96, Ile99, and Tyr100, that comple-
mented the hydrophobic surface of the α-helical p53 peptide.
Interestingly, three residues, Phe19, Trp23, and Leu26, from the
p53 peptide were found to occupy this hydrophobic pocket, with
the formation of a hydrogen bond between the backbone carbonyl
of Leu54 in MDM2 and the indole nitrogen of Trp23 in p53
(Fig. 2). The compact binding pocket on the MDM2 protein
surface is suitable for the development of small-molecule com-
pounds blocking the MDM2 interaction with p53, and a variety
of such inhibitors have been reported in the literature and in
patents [30–32].
Fig. 2 Crystal structure of MDM2 protein in complex with p53 peptide (PDB ID:
1YCR). The MDM2 protein surface is shown with the p53 peptide in a green
cartoon representation. The p53 residues Phe19, Trp23, and Leu26 are shown in
stick form. The hydrogen bond between Leu54 of MDM2 and Trp23 of p53 is
indicated by a dashed line
2.1.1 Surface Plasmon Surface plasmon resonance (SPR) currently is one of the most
Resonance (SPR) widely used label-free methods to evaluate protein–protein and
protein–small molecule interactions. In addition to affinities, it
can determine kinetic parameters, including kon and koff rates,
which provide additional useful information for protein-protein
interaction studies and drug discovery.
1. SPR as a primary assay
At Hoffmann-La Roche, lead compounds of the first class of potent
and selective small-molecule inhibitors of the MDM2–p53 interac-
tion, named Nutlins, were discovered by HTS of a diverse library of
synthetic compounds using SPR [47]. Although the throughput of
SPR is typically not high enough to be compatible with HTS,
researchers of Roche successfully integrated a competitive interac-
tion strategy into the SPR assay to increase its throughput. Instead
of directly evaluating the interaction between compounds and the
MDM2 protein, the interaction between MDM2 and p53 in the
presence of test compounds was evaluated to determine the inhibi-
tory abilities of those compounds. No details of this work were
provided however.
Materials The popular Biacore series of SPR instruments was initially intro-
duced by Sweden’s Biacore AB Corporation, which was purchased
by GE Healthcare in 2006. Currently, SPR instruments are avail-
able from a variety of manufacturers, such as Biosensing Instru-
ment, Sensia, Thermo, and Bio-Rad. Most of these are much less
expensive than those from GE/Biacore [48]. In the Roche study,
competitive assays were performed on a Biacore S51 SPR instru-
ment. CM5 sensor chips were used for protein immobilization.
Reagents included an EDC/NHS activation kit from GE Health-
care, PentaHis antibody from Qiagen, His-tagged p53 protein, and
MDM2 protein. The running buffer was 10 mM HEPES with
0.15 % NaCl and 2 % DMSO. The Evaluation software provided
with the instrument was used to fit the kinetic sensorgrams and
Microsoft Excel was used to calculate the IC50 values of the com-
pounds tested.
Case Study: Discovery of Inhibitors of the MDM2–p53 Protein-Protein Interaction 571
Methods In this study, the capture approach rather than the commonly used
direct immobilization approach was used to load p53 protein onto
the sensor chip. PentaHis antibody (Qiagen) was pre-immobilized
on the Biacore CM5 sensor chip by amine coupling chemistry,
which subsequently was able to capture the His-tagged p53 protein.
Since the location of the HisTag can be precisely controlled on
either the N- or C-terminus of the p53 protein, the protein orienta-
tion in the immobilization/capture process can be controlled and
the influence on the p53–MDM2 interacting model is minimized.
To assure data consistency, the level of p53 captured was controlled
at around 200 response units (RU; one RU corresponds to 1 pg of
protein per mm2). In all the assays, 300 nM of MDM2 protein was
included and 10 mM DMSO stock solutions of test compounds
were further serially diluted. The binding of immobilized p53 pro-
tein and 300 nM of MDM2 protein in the presence of test com-
pounds was determined as a percentage of binding in the absence of
inhibitor. IC50 values were calculated using Microsoft Excel.
Materials Assays were performed on the Biacore S51 SPR instrument and
CM5 sensor chips used just as in the Roche competitive assay. As
mentioned above, the major difference of materials used here was
that p53 protein was no longer needed. Both GST and AviTAG
labeled MDM2 proteins, goat anti-GST antibody, and Streptavidin
(SA) were used to prepare MDM2 surface on the sensor chips
through the capture approach. Other common reagents for SPR
included an EDC/NHS amine activation kit, acetate buffer for
Anti-GST antibody or SA immobilization, MDM2 protein capture,
and assay running buffer (25 mM Tris–HCl, pH 7.5, 150 mM
NaCl, 0.2 mM TCEP, and 0.005 % Tween 20). Instead of the
Evaluation software, Scrubber2 software (BioLogic Software Pty
Ltd.) was used to fit the kinetic data.
Methods CM5 chips were activated with the EDC/NHS activation kit fol-
lowing protocols provided by Biacore. Immediately after activation,
Anti-GST antibody or SA was immobilized on the CM5 chips
which would capture GST or AviTAG labeled MDM2 proteins to
prepare a stable MDM2 surface where interactions with small
molecules would take place. Serial dilutions of p53 peptide, as the
positive control, and compound stock solutions in DMSO were
further diluted to the desired concentration range in the assay
buffer. Compound working solutions were then injected over cap-
tured MDM2 surfaces. Two minutes of association and 3–4 min of
dissociation were monitored and raw data collected were fit, using
Scrubber2, to a typical 1:1 binding model from which Kd, kon, and
koff were obtained.
Materials The essential materials for an HTRF assay are interacting partners
labeled with fluorescent donor and acceptor, respectively. In this
case, GST-MDM2 protein which would be recognized by anti-GST
antibody labeled with europium cryptate and Avi-p53 protein teth-
ered to SA-Xlent were designated as the donor–acceptor pair.
Besides these carefully designed fluorescently labeled proteins,
other materials included phosphate assay buffer, an Envision plate
reader, Perkin Elmer white 384 Opti plates, and some typical data
processing software. All materials are commonly needed for typical
in vitro biochemical assays and excellent evidence of the simplicity
of HTRF method.
Notes Although ThermoFluor is a quick and easy method and feasible for
most laboratories, one must understand that the correlation
between melting temperature change obtained from ThermoFluor
and affinity is qualitative, and an increase in melting temperature
does not necessarily correspond to a specific interaction of interest.
A secondary specific and quantitative assay is required to obtain
accurate affinity information. In these studies, MDM2 binders were
tested with an FP-based peptide displacement binding assay (see
below) to identify bona fide inhibitors of the MDM2–p53 interac-
tion. This led to the identification of some benzodiazepinedione-
based MDM2 inhibitors: compounds 7 (IC50 ¼ 420 nM) and
8 (IC50 ¼ 490 nM) which disrupt the MDM2–p53 interaction,
and the optimized compound 9 whose Kd value is 80 nM (Fig. 3).
Crystal structures of 9 in a complex with MDM2 revealed that 9 fits
into the same pockets in the MDM2 binding cleft used by the p53
peptide side chains Phe19, Trp23, and Leu26. As with the Nutlins,
the interactions of these inhibitors with the MDM2 protein are
primarily through van der Waals contacts. Another series of benzo-
diazepinedione compounds identified by the same ThermoFluor
method followed by optimization led to TDP521252 (10) and
TDP665759 (11) (Fig. 3) which bind to MDM2 with IC50 values
of 708 nM and 704 nM, respectively [38].
2.1.4 Fluorescence The FP assay, which can involve a direct binding or a competitive
Polarization (FP) approach, is probably the most widely used method for screening
small molecules targeting protein-protein interactions. Through the
direct binding approach, Kd values can be easily determined accu-
rately by monitoring the change in FP of the fluorescently labeled
small molecule or peptide while binding to different concentrations
of protein. This approach is not suitable for HTS since it is not
practical to fluorescently label each compound tested. However, it
can be easily adapted to HTS by employing a competitive approach
in which compounds compete with a fluorescently labeled small-
molecule compound or peptide, typically termed a tracer, for bind-
ing to the target protein. In most cases, the Ki values of compounds
based on the experimental IC50 values obtained can be accurately
determined, as can the Kd value of the tracer to the target protein, the
latter being determined with the direct binding approach.
There are many successful cases reported of discovery and
development of MDM2–p53 interaction inhibitors using the FP
method. Among them, the most successful one probably is the
spiro-oxindole class of compounds developed at the University of
Michigan [34, 35, 53, 54]. Starting from the crystal structure of
the MDM2 protein complexed with the p53 peptide, a structure-
based de novo rational strategy was employed to design new classes
of small-molecule inhibitors targeting the MDM2–p53 interaction.
With the assistance of computational screening, a spiro-oxindole
578 Liu Liu et al.
2.1.5 ELISA, Isothermal Other methods to study protein–protein and protein–small mole-
Titration Calorimetry (ITC), cule interactions were used successfully on the MDM2–p53 inter-
and NMR action. These included the ELISA, ITC, and NMR methods.
Compounds 16 and 17 (Fig. 3), which inhibit the MDM2–p53
interaction with IC50 values of 10 μM and 15 μM, respectively, are
terphenyl compounds which were identified using an in vitro,
quantitative, ELISA-based assay as a screen [44]. The screening of
a small library of 16 chalcones, compounds known to have antican-
cer properties [63], with a similar ELISA-based assay monitoring
disruption of the MDM2–p53 interaction resulted in the identifi-
cation of two compounds 18 and 19 (Fig. 3) [64] which have IC50
values of 49 μM and 117 μM, respectively. NMR titration experi-
ments were used to confirm the binding, and the compounds were
also found to inhibit the MDM2–p53 interaction in an in vitro
DNA binding electrophoretic mobility super-shift assay (EMSA).
Recently, researchers from Daiichi Sankyo developed an imida-
zothiazole class of potent MDM2 inhibitors starting from the
core structure of the Nutlins. Based on extensive SAR information
obtained from ELISA, the most potent compound 20 (Fig. 3) had
an IC50 value of 1.2 nM in their ELISA assay [65].
ITC, another quite popular and unique method for protein–
protein and protein–small molecule interaction studies, is the only
100 % real homogeneous and label-free method available. In addi-
tion to the binding affinities, it can also produce very useful infor-
mation on the stoichiometry and ΔH of the interaction of interest,
data which are obtained only with great difficulty by any other
method. However, it also has huge disadvantages over other meth-
ods. The most important of these are the significantly large amount
of protein that is consumed and the very low throughput that is
obtained. Thus the most suitable application for ITC is its use as a
confirmatory assay for fully optimized compounds that are ready to
enter the next research stage. Researchers from Amgen designed
and developed a robust ITC assay and confirmed their new class of
MDM2 inhibitors that were identified by HTRF, as discussed
above. In this way, novel structural features were discovered of
the MDM2–small molecule interaction model involving the
MDM2 N-terminal residues that had previously been regarded as
“structureless” [66].
Fig. 4 Small-molecule inhibitors of the MDM2–p53 interaction discovered by virtual database screening
582 Liu Liu et al.
3 Concluding Remarks
Acknowledgements
Disclosure statement
References
1. Fry DC, Vassilev LT (2005) Targeting protein- 2. Murray JK, Gellman SH (2007) Targeting
protein interactions for cancer therapy. J Mol protein-protein interactions: lessons from
Med 83:955–963 p53/MDM2. Biopolymers 88:657–686
Case Study: Discovery of Inhibitors of the MDM2–p53 Protein-Protein Interaction 583
3. Fridman JS, Lowe SW (2003) Control of apo- implications for chemotherapy. Curr Cancer
ptosis by p53. Oncogene 22:9030–9040 Drug Targets 5:27–41
4. Hainaut P, Hollstein M (2000) p53 and human 18. Momand J, Jung D, Wilczynski S et al (1998)
cancer: the first ten thousand mutations. Adv The MDM2 gene amplification database.
Cancer Res 77:81–137 Nucleic Acids Res 26:3453–3459
5. Vogelstein B, Lane D, Levine AJ (2000) Surf- 19. Gunther T, Schneider-Stock R, Hackel C et al
ing the p53 network. Nature 408:307–310 (2000) Mdm2 gene amplification in gastric
6. Vousden KH, Lu X (2002) Live or let die: the cancer correlation with expression of Mdm2
cell’s response to p53. Nat Rev Cancer protein and p53 alterations. Mod Pathol
2:594–604 13:621–626
7. Feki A, Irminger-Finger I (2004) Mutational 20. Bond GL, Hu W, Levine AJ (2005) MDM2 is a
spectrum of p53 mutations in primary breast central node in the p53 pathway: 12 years and
and ovarian tumors. Crit Rev Oncol Hematol counting. Curr Cancer Drug Targets 5:3–8
52:103–116 21. Jones SN, Roe AE, Donehower LA et al (1995)
8. Momand J, Zambetti GP, Olson DC et al Rescue of embryonic lethality in Mdm2-
(1992) The mdm-2 oncogene product forms a deficient mice by absence of p53. Nature
complex with the p53 protein and inhibits p53- 378:206–208
mediated transactivation. Cell 69:1237–1245 22. de Oca M, Luna R, Wagner DS, Lozano G
9. Fakharzadeh SS, Trusko SP, George DL (1995) Rescue of early embryonic lethality in
(1991) Tumorigenic potential associated with mdm2-deficient mice by deletion of p53.
enhanced expression of a gene that is amplified Nature 378:203–206
in a mouse tumor cell line. EMBO J 23. Vassilev LT (2007) MDM2 inhibitors for can-
10:1565–1569 cer therapy. Trends Mol Med 13:23–31
10. Fakharzadeh SS, Rosenblum-Vos L, Murphy M 24. Chene P (2003) Inhibiting the p53-MDM2
et al (1993) Structure and organization of interaction: an important target for cancer
amplified DNA on double minutes containing therapy. Nat Rev Cancer 3:102–109
the mdm2 oncogene. Genomics 15:283–290 25. Shangary S, Wang S (2008) Targeting the
11. Lane DP, Crawford LV (1979) T antigen is MDM2-p53 interaction for cancer therapy.
bound to a host protein in SV40-transformed Clin Cancer Res 14:5318–5324
cells. Nature 278:261–263 26. Freedman DA, Wu L, Levine AJ (1999) Func-
12. Linzer DI, Levine AJ (1979) Characterization of tions of the MDM2 oncoprotein. Cell Mol Life
a 54 K dalton cellular SV40 tumor antigen pres- Sci 55:96–107
ent in SV40-transformed cells and uninfected 27. Juven-Gershon T, Oren M (1999) Mdm2: the
embryonal carcinoma cells. Cell 17:43–52 ups and downs. Mol Med 5:71–83
13. DeLeo AB, Jay G, Appella E et al (1979) 28. Wu X, Bayle JH, Olson D et al (1993) The
Detection of a transformation-related antigen p53-mdm-2 autoregulatory feedback loop.
in chemically induced sarcomas and other Genes Dev 7:1126–1132
transformed cells of the mouse. Proc Natl 29. Kussie PH, Gorina S, Marechal V et al (1996)
Acad Sci U S A 76:2420–2424 Structure of the MDM2 oncoprotein bound to
14. Bond GL, Hu W, Bond EE et al (2004) A the p53 tumor suppressor transactivation
single nucleotide polymorphism in the domain. Science 274:948–953
MDM2 promoter attenuates the p53 tumor 30. Millard M, Pathania D, Grande F et al (2011)
suppressor pathway and accelerates tumor for- Small-molecule inhibitors of p53-MDM2
mation in humans. Cell 119:591–602 interaction: the 2006-2010 update. Curr
15. Oliner JD, Kinzler KW, Meltzer PS et al (1992) Pharm Des 17:536–559
Amplification of a gene encoding a p53- 31. Zhan C, Lu W (2011) Peptide activators of the
associated protein in human sarcomas. Nature p53 tumor suppressor. Curr Pharm Des
358:80–83 17:603–609
16. Zhou M, Gu L, Abshire TC et al (2000) Inci- 32. Zak K, Pecak A, Rys B et al (2013) Mdm2 and
dence and prognostic significance of MDM2 MdmX inhibitors for the treatment of cancer: a
oncoprotein overexpression in relapsed child- patent review (2011–present). Expert Opin
hood acute lymphoblastic leukemia. Leukemia Ther Pat 23:425–448
14:61–67
33. Vassilev LT (2004) Small-molecule antagonists
17. Rayburn E, Zhang R, He J et al (2005) MDM2 of p53-MDM2 binding: research tools and
and human malignancies: expression, clinical potential therapeutics. Cell Cycle 3:419–421
pathology, prognostic markers, and
584 Liu Liu et al.
59. Munson PJ, Rodbard D (1988) An exact cor- 67. Lin J, Chen J, Elenbaas B et al (1994) Several
rection to the “Cheng-Prusoff” correction. J hydrophobic amino acids in the p53 amino-
Recept Res 8:533–546 terminal domain are required for transcrip-
60. Zhang R, Mayhood T, Lipari P et al (2004) tional activation, binding to mdm-2 and the
Fluorescence polarization assay and inhibitor adenovirus 5 E1B 55-kD protein. Genes Dev
design for MDM2/p53 interaction. Anal Bio- 8:1235–1246
chem 331:138–146 68. Bottger V, Bottger A, Howard SF et al (1996)
61. Kutzki O, Park HS, Ernst JT et al (2002) Identification of novel mdm2 binding peptides
Development of a potent Bcl-x(L) antagonist by phage display. Oncogene 13:2141–2147
based on alpha-helix mimicry. J Am Chem Soc 69. Bowman AL, Nikolovska-Coleska Z, Zhong H
124:11838–11839 et al (2007) Small molecule inhibitors of the
62. Boettcher A, Buschmann N, Furet P, et al. MDM2-p53 interaction discovered by
(2008) 3-imidazolyl-indoles for the treatment ensemble-based receptor models. J Am Chem
of proliferative diseases. US Patent WO Soc 129:12809–12814
2008119741 70. Jones G, Willett P, Glen RC et al (1997) Devel-
63. Go ML, Wu X, Liu XL (2005) Chalcones: an opment and validation of a genetic algorithm
update on cytotoxic and chemoprotective for flexible docking. J Mol Biol 267:727–748
properties. Curr Med Chem 12:481–499 71. Verdonk ML, Cole JC, Hartshorn MJ et al
64. Stoll R, Renner C, Hansen S et al (2001) Chal- (2003) Improved protein-ligand docking
cone derivatives antagonize interactions using GOLD. Proteins 52:609–623
between the human oncoprotein MDM2 and 72. Eldridge MD, Murray CW, Auton TR et al
p53. Biochemistry 40:336–344 (1997) Empirical scoring functions: I. The
65. Uoto K, Kawato H, Sugimoto Y, et al. (2009) development of a fast empirical scoring func-
Imidazothiazole derivative having 4,7-diazas- tion to estimate the binding affinity of ligands
piro[2.5]octane ring structure. US Patent in receptor complexes. J Comput Aided Mol
WO/2009/151069 Des 11:425–445
66. Michelsen K, Jordan JB, Lewis J et al (2012) 73. Wang R, Lai L, Wang W (2002) Further devel-
Ordering of the N-terminus of human MDM2 opment and validation of empirical scoring
by small molecule inhibitors. J Am Chem Soc functions for structure-based binding affinity
134:17059–17067 prediction. J Comput Aided Mol Des 16:11–26
Chapter 39
Abstract
Fragment-based lead discovery complements high-throughput screening and computer-aided drug design
for the discovery of small-molecule inhibitors of protein-protein interactions. Fragments are molecules with
molecular masses ca 280 Da or smaller, and are generally screened using structural or biophysical
approaches. Several methods of fragment-based screening are feasible for any soluble protein that can be
expressed and purified; specific techniques also have size limitations and/or require multiple milligrams of
protein. This chapter describes some of the most common fragment-discovery methods, including surface
plasmon resonance, nuclear magnetic resonance, differential scanning fluorimetry, and X-ray
crystallography.
Key words Fragment, Ligand, Discovery, Biophysics, SPR, NMR, DSF, Crystallography, Protein-
protein, Interaction
1 Introduction
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7_39, © Springer Science+Business Media New York 2015
587
588 Samuel J. Pfaff et al.
b c 80
80
60
Response (RU)
60
40 40
RU
20
20
0
0
0 50 100 0 250 500 750 1000 1250
Fig. 1 Fragment screening by SPR. (a) Primary screening data (courtesy of Stacie Bulfer). Green circles are
positive controls and black squares are fragments screened at 250 μM. The red line represents zero response
and the blue dashed line represents the hit cutoff of three standard deviations. (b) Fragment hit follow-up:
duplicate dose–response data for a fragment hit. This compound shows canonical fragment binding kinetics
with rapid approaches to equilibrium at the beginning and end of each injection. The injection start is indicated
by a closed arrow and the injection end (dissociation phase) is indicated by an open arrow. The data used to
generate equilibrium binding values are the averages of the values between the dashed lines. (c) Equilibrium
binding analysis of the compound used in (b). Data were fit to a simple 1:1 hyperbolic binding isotherm
∆Tm
{
Normalized Fluorescence
1.0
0.5
0.0
40 50 60 70 80 90 100
Temperature (°C)
[16]; the initial concentrations of protein and dye are the only
parameters that typically require optimization. This allows diver-
gent targets to be screened under very similar conditions. Buffer
and salt conditions can have significant effects on protein melting
curves, and can be optimized as well. However, it is usually desir-
able to employ a simple buffer system that best reflects the physio-
logical environment of the target protein. Assay throughput is high,
since 384 wells can be read simultaneously in 45–90 min, depend-
ing on assay parameters. Thus, an average-sized fragment library
can be screened in a day with plate-handling automation. Protein
consumption during screening is also quite low (typically 5 mg), as
very small volumes (<5 μL/well) can be used during the assay. For
targets that are challenging to isolate, this can be critical. Finally,
the reagents and instrumentation are relatively inexpensive. RT-
PCR instruments have become commonplace at research centers
in the last decade, and they are far less complex to maintain than
SPR, NMR, or in-house X-ray equipment. The dye necessary for a
screen is very inexpensive, and 384 well PCR plates are comparable
in price to most assay plates. On the other hand, For fragments,
thermal shifts can be within 1 C, and binding stoichiometry is not
directly measured. Taking advantage of its speed, DSF screening is
often used as a primary screen to identify binders, followed by
higher resolution methods.
DSF is also valuable as a first step in structural biology experi-
ments. Proteins can show wide ranges of thermal stabilities as a
function of buffer pH, salt identity and concentration, reducing
agents, and other additives. Using DSF, one can rapidly screen a
592 Samuel J. Pfaff et al.
A31 0.2
128 1 128
2
5
C184
129 129
I182
130 130
V176
131 V181 131
132 132
8.8 8.6 8.4 8.2 8.0 7.8 7.6
1H
2- (ppm)
Fig. 3 NMR experiments for fragment screening. (a) Saturation transfer difference. 1H NMR spectrum of a
fragment (blue) is reduced when it binds to protein (red). Measuring the difference spectrum then gives a
negative signal with the same chemical shifts as the fragment (not shown). (b) 1H-15N HSQC. Each backbone
NH has a characteristic chemical shift. When fragments bind, they induce changes to these shifts in dose-
dependent manner (see legend) (Data courtesy of Mark Kelly, Michael Chimenti, and Rich Tjhen)
1.4 X-Ray The ideal situation for most fragment screening efforts is to obtain
Crystallography atomic-resolution structures of fragment hits bound to the target
(Fig. 4). High-resolution structures allow medicinal chemists to
improve compound potency by rationally altering chemical struc-
ture to better complement the target site. For small fragments,
whose binding modes are difficult to model precisely or predict a
Fig. 4 High-resolution X-ray structure of a PPI inhibitor fragment hit. RadA bound
to fragment hit indazole at 1.3 Å resolution (PDB: 4B2I) [3]. 2Fo-Fc electron
density map is shown for the compound contoured at 1.5σ. The electrostatic
surface and figure were generated using PyMOL [42]
596 Samuel J. Pfaff et al.
1.5 Summary The techniques overviewed above are the most commonly used
techniques in FBLD and each technique has a proven record of
success in identifying promising chemical matter against many
targets. Given below are detailed experimental protocols for devel-
oping these assays and employing them in an FBLD campaign
aimed at a protein or PPI target.
2 Materials
2.2 Differential 1. PCR plates suitable for the instrument being used.
Scanning Fluorimetry 2. Fluorescent dye.
3. Protein of interest.
4. Fragment library.
5. Assay Buffer: 20 mM HEPES, pH 7.4, 150 mM NaCl,
0.25 mM TCEP.
6. Optically clear sealing tape.
3 Methods
3.1 Surface Plasmon These methods assume that the user is familiar with the operation
Resonance of the SPR instrumentation in his/her laboratory.
3.1.1 Select SPR Chip The first step in performing any SPR experiment is selecting a strategy
Surface for immobilizing the target protein to the sensor surface. The two
most common immobilization strategies are (a) covalent coupling
through lysine side chains and N-termini using EDC/NHS-activated
carboxyl groups on the sensor surface (random-amine coupling) and
598 Samuel J. Pfaff et al.
The mass ratio above becomes very small for fragments against
average-sized protein targets (0.01 or less), necessitating a high
immobilization density to compensate. This is one negative aspect
of screening by SPR. High-density surfaces have often been asso-
ciated with artifactual protein behavior, due primarily to the high
concentration of protein at the surface. Additionally, some immo-
bilized protein might be inactive towards small-molecule binding
due to denaturation or orientation/immobilization to the SPR
surface. For these reasons, it is critical to optimize the protein-
immobilization procedure using a well-behaved control com-
pound, peptide, or protein binding partner as a control. This
positive control will also be used to monitor the stability of the
protein surface during screening.
Our preferred method of immobilization is biotin–avidin cap-
ture using site-specifically biotinylated proteins [27]. The AviTag
sequence is a short peptide that is a substrate for E. coli biotin ligase
BirA. If protein expression is performed in E. coli, the AviTagged
target protein can be coexpressed with BirA to yield protein that is
singly biotinylated at a specific lysine in the AviTag sequence.
Vectors for appending an AviTag to the N or C termini of targets
are commercially available. We prefer this method to random
biotinylation (coupling to lysine or cysteine residues) or covalent
coupling, as it is a consistent, predictable method that requires no
extra processing steps, and it does not compromise potentially
relevant protein amines.
A generic protocol for immobilization using biotinylated
protein follows. Streptavidin and neutravidin-coated SPR surfaces
(chips) can be purchased from many instrument vendors or can be
prepared from carboxymethyl chips, such as Biacore CM5 or CM7
(these two chips differ in density of carboxyl groups, see Chapter 7
for more details). We prefer neutravidin, a mutant form of strepta-
vidin, as it contains fewer potential small-molecule binding sites,
and is more amenable to covalent coupling due to its increased pI.
Steps 2–5 describe the protocol for immobilizing neutravidin; this
protocol is similar for covalent coupling of any target to a carbox-
ymethyl chip surface by EDC/NHS chemistry.
Fragment-Based PPI Inhibitor Discovery 599
3.1.2 Prepare SPR Chip 1. Install SPR chip into instrument per vendor recommendation.
Surface
Covalently modify SPR chip with neutravidin:
2. Dilute neutravidin to 0.25 mg/mL into 10 mM Na-Acetate,
pH 4.5. Buffer should first be filtered through 0.2 μm bottle-
top filter (see Note 1).
3. Inject a 1:1 mixture of 0.1 M NHS and 0.4 M EDC for 5 min
to activate the chip surfaces.
4. Inject 0.25 mg/mL neutravidin for 4–7 min across all surfaces.
Using a Biacore CM5 chip this should yield 8,000–12,000 RU
of neutravidin density on all surfaces. Leave one surface as a
neutravidin-only reference.
5. Inject 1 M ethanolamine, pH 8.5, for 5 min across all surfaces
to block all remaining reactive sites.
Immobilize biotinylated protein
6. Dilute the biotinylated target protein to low concentration
(5–50 μg/mL) in immobilization buffer.
7. Filter the target protein solution using a 0.22 μm, 0.5 mL spin
filter.
8. Inject the target protein on an avidin-labeled flow cell for
1–5 min, or until the desired amount has been captured.
A surface of ~10,000 RUs for a protein of average size
(20–50 kDa) is a reasonable goal (see Note 2).
9. Inject 0.2 mg/mL Amine-PEG-Biotin (or a similar, soluble
biotin analog) for 4 min across all surfaces to block any
remaining biotin binding sites.
3.1.3 Establish the It is essential to determine the quality of the immobilized protein
Activity of the SPR Surfaces before proceeding with a fragment screening effort.
1. Select a running buffer. A typical buffer could contain 10 mM
Tris–HCl, pH 8.0, 150 mM NaCl, 0.25 mM TCEP, 0.05 %
(v/v) Tween 20, 5 % (v/v) DMSO (see Note 3).
2. Select a small molecule, peptide, or protein that binds to the
site of interest to be used as a positive control.
3. Serially dilute the control in DMSO and transfer to a running
buffer without DMSO, so that the % DMSO is identical in the
sample well and running buffer. For example, for a final DMSO
concentration of 5 % in 100 μL sample volume, dilute 5 μL of
compound into 95 μL of running buffer without DMSO.
Select a wide concentration range to span the expected binding
constant. Inject samples over the chip surface and measure the
SPR signal.
600 Samuel J. Pfaff et al.
3.1.4 Run Fragment 1. Set up assay plates using a standardized format based on the
Screen capabilities of your instrument. As with the control com-
pounds, the DMSO concentration and buffer should exactly
match the running buffer. Typical concentrations for fragments
are 100–500 μM in 2–5 % DMSO.
2. Run the screen using injection times between 10 and 30 s,
followed by dissociation monitoring for 30 s. Because frag-
ments should bind weakly and therefore reach equilibrium
quickly, these short injections provide a significant amount of
data with which to classify compounds.
3. Inject buffer blanks (containing DMSO) and positive control
molecules regularly throughout the screen (e.g., at the end of
a column or each plate), to evaluate assay stability. Blank
injections should also be used during data analysis to “dou-
ble-reference,” which is especially critical in fragment screening
where the signal is expected to be low [28]. The controls
should also be used to calculate z0 or other statistical factors
to assess the quality of the screen [29].
3.1.5 Identify Screening SPR data allows for hit selection based on both the % fractional
Hits occupancy the compound achieves, and also the kinetic profile it
produces.
1. Reference SPR data (“sensorgrams”) to the unmodified flow cell
and double-reference to blank injections using the instrument’s
software or third-party software such as Scrubber (Biologic
Software, http://www.biologic.com.au/). Additional data
smoothing and scaling can reduce complexity arising from
variation in positive control binding and experiments run on
separate surfaces [28].
2. Prepare a scatterplot (Fig. 1a) of the control and test
compounds. Select a time-range near the end of the sample
injection, where the positive control has achieved equilibrium.
Use this time range to determine the RU for each control and
test compound, and plot on the y-axis. Scatterplots can also be
scaled to binding stoichiometry:
Fragment-Based PPI Inhibitor Discovery 601
3.1.6 Initiate Secondary At this stage, repurchase or synthesize the most potent compounds
Assays with low stoichiometry and move forward to secondary screening
(such as functional activity), chemical optimization, and structure
determination by NMR or X-ray crystallography (see below).
3.2 Differential 1. Select reagents. The choice of plates will depend on the model
Scanning Fluorimetry of RT-PCR instrument to be used; in general, use 384-well
PCR plates with optically clear sealing tape. There are also
3.2.1 Develop DSF Assay
many potential dyes. No published study has systematically
compared the appropriate dyes for overall performance, and
target-specific effects of particular dyes may be possible.
SYPRO Orange is cost-effective and widely used. It is provided
as a 5,000 stock in DMSO, and our experience has shown it
to work well at 1 final concentration across a range of targets
and buffers.
2. Prepare a matrix containing several concentrations of protein
and several concentrations of dye, (e.g., SYPRO Orange); each
combination should be tested in triplicate. Typical starting
parameters are 2 μM protein with 5,000 diluted SYPRO
Orange [30]. Assay volumes can be as low as 5 μL with appro-
priate 384-well plates.
3. Measure the melting temperature of each dye–protein combi-
nation using a qRT-PCR instrument. Record fluorescence data
at the appropriate dye wavelengths, using a temperature range
602 Samuel J. Pfaff et al.
3.2.2 Run Fragment The following procedure is a general method for fragment screen-
Screen ing by DSF in 384-well plates with a final well volume of 20 μL and
a final compound concentration of 1 mM.
1. Prepare reagents for screening as follows: dilute protein from a
concentrated stock to 1.33 the predetermined optimal assay
concentration (usually between 0.5 and 5 μM) into assay buffer
without DMSO. Dilute dye to 5 the predetermined optimal
concentration into assay buffer without DMSO. Prepare or
store compounds at 20 final concentration in 100 % DMSO
(20 mM for a final assay volume of 1 mM).
2. Add 15 μL diluted protein to each well of a 384-well PCR
plate.
3. Add 1 μL DMSO to one column of the plate. These wells serve
as the negative controls.
4. If a positive control compound is available, add 1 μL of 20
positive control in DMSO to one column of the plate.
5. Add 1 μL of test compound to each non-control well using a
multichannel pipette or an automated liquid handler.
6. Add 4 μL of diluted dye to each well using a multichannel
pipette or an automated liquid handler. Seal the plate with
optically clear sealing tape, cover with foil, and mix by gentle
shaking at room temperature for 10–20 min.
7. Remove foil and centrifuge plate for 2 min at 1,000–2,000 g
to remove bubbles. The plate is now ready to be assayed.
8. Set up the RT-PCR instrument to record fluorescence data at
the appropriate dye wavelengths, for a temperature range of
25–95 C in 1 steps with 30 s per step. Load the plate into the
thermocycler and run the experiment. With these parameters,
each plate should take between 35 and 45 min.
9. Determine the Tm for fragments. Plot the fluorescence vs.
temperature data and fit to the Boltzmann equation:
F ¼ min þ ðmax minÞ=ð1 þ expððT m x Þ=slopeÞÞ
Fragment-Based PPI Inhibitor Discovery 603
3.2.3 Identify Screening To select screening hits, prepare a scatter plot of Tm value vs.
Hits compound and control wells. The control wells should produce a
precise Tm with low standard deviation (<1 ). Fragments that
produce a Tm increase greater than two times the standard devia-
tion of control wells are typically considered hits. This threshold
can be modified if the number of hits above 2 standard deviations is
thought to be too high or too low. Setting too low of a hit
threshold is, however, likely to generate many false positive com-
pounds that cannot be confirmed. In this case it might be advanta-
geous to rescreen at a higher compound concentration.
3.2.4 Initiate Secondary If desired, affinities can be directly determined by DSF using serial
Assays dilutions of fragments. Thermal shift dose–response data is not
fitted to a standard hyperbolic curve, but rather to a more compli-
cated expression that represents the free energy change of folding as
well as that of compound binding. Matulis et al. have derived a
model to fit DSF titration curves [31]. Accurate binding affinities
may be more readily obtained from other biophysical methods,
such as SPR, NMR, or ITC, as there is no reliance upon an inde-
pendently determined or estimated parameter, as is the case with
DSF. Since every assay format has advantages and limitations, it is
generally advisable to follow up hits from any fragment screen using
an orthogonal method.
3.3 Nuclear The detailed operation of NMR instruments is beyond the scope of
Magnetic Resonance this chapter. Omitted are basic preparative setup steps for acquiring
NMR spectra, such as shimming and tuning, as it is assumed the user
is familiar with these or will have support from an NMR spectrosco-
pist. For further reading, see Till Maurer’s excellent introduction to
NMR methods in fragment-based ligand screening and Bertini for
more detailed protocols and troubleshooting [32, 33].
3.3.1 Develop a The Saturation Transfer Difference (STD) experiment relies on the
Saturation Transfer nuclear Overhauser effect (nOe) between nuclei in the protein and
Difference (STD) Assay in a small-molecule ligand that binds transiently. The compounds
that interact with the protein will show reduced intensity in their
NMR signals relative to non-binders, whose signals will be unaf-
fected by the presence of protein. STD is performed as a subtractive
measurement, where the 1-D ligand spectra are collected in the
presence and absence of protein irradiation (“on-resonance” and
604 Samuel J. Pfaff et al.
3.3.2 Run an STD The following is a general outline for a protocol when performing
Fragment Screen STD experiments in standard 5 mm diameter NMR tubes.
Typically ~100 samples (5–10 fragments each) or more can be
screened in a 24 h period with automated hardware.
1. Prepare the sample(s) without protein in NMR buffer (final
volume ~450 μL). Samples will include fragment(s), ~10 μM
DSS (for referencing), a deuterated buffer and D2O (99.96 %)
as solvent (see Note 8).
2. Acquire a simple 1D spectrum at the temperature to be used for
screening for each fragment to be used as a reference spectrum
during data analysis.
3. Add protein to each sample. Place samples in a rack. Make a
“master mix” containing protein (plus fresh DTT if indicated),
and NaN3 (to control bacterial growth) along with 10–20 μL
of NMR buffer per sample. Dispense the correct volume of
master mix (usually ~50 μL) into each tube with a pipette
(by placing NMR tubes into a pipette tip box, an 8-channel
pipette can be used for this). Carefully swing the rack up and
down to force the solution to the bottom of each tube (cover
the top of the rack with a piece of cardboard and grip it firmly).
Allow the tubes to sit for 1–2 h or overnight at 4 C to mix.
4. After tuning, shimming, etc., load parameters for the STD
NMR experiment. Be sure to optimize the 1H transmitter
frequency to obtain good suppression of any residual water.
The number of scans is typically set to 16 or 32, saturation time
to 1.3 s, recycle delay to 3.7 s with a spectral width of 16 ppm
and collecting 4,096 complex data points. Experiments can be
acquired in 10–15 min per sample.
5. The data are then apodized (1–3 Hz line-broadening), zero-
filled once, Fourier transformed, and phased. 1D on- and
off-resonance STD are extracted from the pseudo 2D spectrum
and overlaid for comparison. The “off-resonance” experiment
is scaled down to the “on-resonance” experiment and the “%
STD” reduction for each peak measured.
6. When working with mixtures of ligands, repeating the experi-
ment with the individual fragments from a pool that showed a
“hit” can confirm the identity of the binding fragment(s).
606 Samuel J. Pfaff et al.
3.3.5 HSQC and HMQC One of the first fragment screening methods, called “SAR by
NMR NMR,” was based on a protein-observed HSQC experiment
[36]. A key advantage of protein-observed experiments is that the
structure and state of the target protein are monitored in each
experiment.
HSQC and related chemical-shift perturbation methods rely on
the availability of labeled protein, either 15N-labeling or 13C-labeling
for larger systems. The collection of a 13C/15N 2D inverse-
correlation or HSQC/HMQC type experiment yields a 2D spec-
trum, where cross-peaks corresponding to the frequencies of two
directly bound nuclei (e.g., Hn–15N or 1H–13C in a methyl group)
Fragment-Based PPI Inhibitor Discovery 607
3.4 X-Ray 1. Select the appropriate protein construct (see Note 9).
Crystallography 2. Select a storage buffer that confers good protein stability.
3.4.1 Validate Crystal Prescreening for protein thermal stability in a range of possible
Forms for Fragment storage buffers can be easily accomplished using differential
Screening scanning fluorimetry (DSF; see above).
3. Screen crystallography conditions. Obtaining protein crystals
that diffract to better than 2.5 Å resolution is almost always the
result of brute force screening. In a typical case, 3–10 protein
expression constructs are screened against >1,000 initial
conditions from sparse-matrix, commercial crystal screens
(see Note 10).
4. Assess the stability of crystals in cryogenic conditions [26].
Nearly all protein crystallography is carried out under cryo-
genic conditions, requiring the optimization of freezing con-
ditions that prevent crystal damage. Typically, this involves
temporary soaking of each crystal in a condition containing a
high concentration (20–30 % (v/v)) of glycerol or high molec-
ular weight PEG. This should be performed with several test
solutions in a trial and error fashion.
5. Assess the stability of crystals in DMSO (~10–15 % (v/v)) to
simulate the conditions that will be used for soaking in test
compounds. Several crystals should be transferred to drops
containing varying concentrations of DMSO and observed
intermittently over the next 24 h. Some crystals cannot tolerate
DMSO and will become visibly damaged. X-ray data sets
should then be collected from crystals that remain intact in
DMSO to test for more subtle damage caused by the addition
of organic solvent.
6. Plate compounds for an X-ray based fragment screen.
To increase throughput and reduce the number of crystals
needed, compounds are typically pooled in sets of 5–10.
Premixed and plated compounds can be stored in DMSO or
as dried powder mixes. In either case, after the compounds are
solubilized in DMSO, crystallization buffer is added to each
well to a final volume of 2–5 μL. Crystals are then transferred
into each test well and incubated for a variable length of time.
The time necessary to soak compounds into a crystal is highly
protein dependent.
3.4.2 Collect and Process 1. Prepare crystals for data collection. Data collection and crystal
X-Ray Diffraction Data handling procedures are dictated by the X-ray generation site,
be it a home source or an external light source. For screening,
some measure of automation in crystal handling is critical, as it
can greatly increase efficiency and decrease the risk of manual
error and sample loss. Many beamlines are now outfitted with
frozen crystal handling robots that mostly automate the
Fragment-Based PPI Inhibitor Discovery 609
3.4.3 Identify Hits from 1. Search electron density maps for bound fragments (Fig. 4).
X-Ray Screens Fragment occupancy can be evaluated manually or by compu-
tational methods. Both Coot and Phenix contain modules for
locating large unfilled regions of electron density.
2. When fragment density is observed, attempt to identify its
structure from among the compounds in the screening pool
of 5–10 fragments. This analysis is easiest when the fragment
pools are designed to contain very dissimilar compounds, the
fractional occupancy of the site is high, and the resolution of
the crystal is good (~2 Å). When the identity is not obvious,
one can build separate models for each possible compound and
refine them; the refinement process should eliminate some
compounds from consideration. More automated approaches
to considering fragment electron density have been reported by
industrial labs, but are not publicly available [25]. In many
cases, it is best to recrystallize the fragment/protein complex
from a pure solution of the compound.
3.4.4 Characterize Hits Developing secondary assays for an X-ray fragment screen depends
from X-Ray Screens on many factors, including target type, chemistry resources, and
availability of secondary biophysical assays to measure binding
affinities. For a PPI target, it is ideal to assess whether or not the
fragments inhibit the interaction, using some of the techniques
described elsewhere in this book. Biophysical competition assays
by SPR, analytical ultracentrifugation, analytical gel filtration, or
static light scattering can also give a measure of functional potency.
610 Samuel J. Pfaff et al.
4 Conclusion
5 Notes
References
1. Rees DC, Congreve M, Murray CW et al protein-protein interface. Drug Discov Today
(2004) Fragment-based lead discovery. Nat 14:155–161
Rev Drug Discov 3:660–672 8. Lau WF, Withka JM, Hepworth D et al (2011)
2. Hajduk PJ, Greer J (2007) A decade of Design of a multi-purpose fragment screening
fragment-based drug design: Strategic library using molecular complexity and orthog-
advances and lessons learned. Nat Rev Drug onal diversity metrics. J Comput Aided Mol
Discov 6:211–219 Des 25:621–636
3. Scott DE, Ehebauer MT, Pukala T et al (2013) 9. Na J, Hu Q (2011) Design of screening collec-
Using a fragment-based approach to target tions for successful fragment-based lead discov-
protein-protein interactions. Chembiochem ery. Methods Mol Biol 685:219–240
14:332–342 10. Chen IJ, Hubbard RE (2009) Lessons for frag-
4. Braisted AC, Oslob JD, Delano WL et al ment library design: analysis of output from
(2003) Discovery of a potent small molecule multiple screening campaigns. J Comput
IL-2 inhibitor through fragment assembly. J Aided Mol Des 23:603–620
Am Chem Soc 125:3714–3715 11. Erlanson DA, Wells JA, Braisted AC (2004)
5. Arkin MR, Randal M, Delano WL et al (2003) Tethering: fragment-based drug discovery.
Binding of small molecules to an adaptive Annu Rev Biophys Biomol Struct 33:199–223
protein-protein interface. Proc Natl Acad Sci 12. Wilson CG, Arkin MR (2013) Probing struc-
U S A 100:1603–1608 tural adaptivity at PPI interfaces with small
6. Petros AM, Huth JR, Oost T et al (2010) molecules Drug Discovery Today: Technolo-
Discovery of a potent and selective bcl-2- gies 10 (4):e501–e508
inhibitor using SAR by NMR. Bioorg Med 13. Giannetti AM, Koch BD, Browner MF (2008)
Chem Lett 20:6587–6591 Surface plasmon resonance based assay for the
7. Fuller JC, Burgoyne NJ, Jackson RM (2009) detection and characterization of promiscuous
Predicting druggable binding sites at the inhibitors. J Med Chem 51:574–580
Fragment-Based PPI Inhibitor Discovery 613
14. Babaoglu K, Simeonov A, Irwin JJ et al (2008) through of fragment lead identification using
Comprehensive mechanistic analysis of hits surface plasmon resonance. Methods Enzymol
from high-throughput and docking screens 493:169–218
against beta-lactamase. J Med Chem 29. Zhang JH, Chung TD, Oldenburg KR (1999)
51:2502–2511 A simple statistical parameter for use in evalua-
15. Cimmperman P, Baranauskiene L, Jachimovi- tion and validation of high throughput screen-
ciute S et al (2008) A quantitative model of ing assays. J Biomol Screen 4:67–73
thermal stabilization and destabilization of 30. Niesen FH, Berglund H, Vedadi M (2007) The
proteins by ligands. Biophys J 95:3222–3231 use of differential scanning fluorimetry to
16. Kranz JK, Schalk-Hihi C (2011) Protein ther- detect ligand interactions that promote protein
mal shifts to identify low molecular weight stability. Nat Protoc 2:2212–2221
fragments. Methods Enzymol 493:277–298 31. Matulis D, Kranz JK, Salemme FR et al (2005)
17. Rizo J, Rosen MK, Gardner KH (2012) Thermodynamic stability of carbonic anhy-
Enlightening molecular mechanisms through drase: measurements of binding affinity and
study of protein interactions. J Mol Cell Biol stoichiometry using thermofluor. Biochemistry
4:270–283 44:5258–5266
18. Pellecchia M, Bertini I, Cowburn D et al 32. Maurer T (2011) Advancing fragment binders
(2008) Perspectives on NMR in drug discov- to lead-like compounds using ligand and
ery: a technique comes of age. Nat Rev Drug protein-based NMR spectroscopy. Methods
Discov 7:738–745 Enzymol 493:469–485
19. Ito Y, Selenko P (2010) Cellular structural 33. Bertini I, Molinari H, Niccolai N (1991) NMR
biology. Curr Opin Struct Biol 20:640–648 and biomolecular structure, vol xvii. VCH,
20. Dalvit C, Fagerness PE, Hadden DT et al Weinheim, 209 p
(2003) Fluorine-NMR experiments for high- 34. Dalvit C, Pevarello P, Tato M et al (2000)
throughput screening: theoretical aspects, Identification of compounds with binding
practical considerations, and range of applica- affinity to proteins via magnetization transfer
bility. J Am Chem Soc 125:7696–7703 from bulk water. J Biomol NMR 18:65–68
21. Dalvit C, Flocco M, Veronesi M et al (2002) 35. Gossert AD, Henry C, Blommers MJ et al
Fluorine-NMR competition binding experi- (2009) Time efficient detection of protein-
ments for high-throughput screening of large ligand interactions with the polarization opti-
compound mixtures. Comb Chem High mized PO-WaterLOGSY NMR experiment. J
Throughput Screen 5:605–611 Biomol NMR 43:211–217
22. Hajduk PJ, Meadows RP, Fesik SW (1999) 36. Shuker SB, Hajduk PJ, Meadows RP et al
NMR-based screening in drug discovery. Q (1996) Discovering high-affinity ligands for
Rev Biophys 32:211–240 proteins: SAR by NMR. Science
23. Hajduk PJ, Gerfin T, Boehlen JM et al (1999) 274:1531–1534
High-throughput nuclear magnetic resonance- 37. Kabsch W (2010) Xds. Acta Crystallogr D Biol
based screening. J Med Chem 42:2315–2317 Crystallogr 66:125–132
24. Murray CW, Blundell TL (2010) Structural 38. Holton J, Alber T (2004) Automated protein
biology in fragment-based drug design. Curr crystal structure determination using ELVES.
Opin Struct Biol 20:497–507 Proc Natl Acad Sci U S A 101:1537–1542
25. Spurlino JC (2011) Fragment screening purely 39. Adams PD, Afonine PV, Bunkoczi G et al (2010)
with protein crystallography. Methods Enzy- Phenix: a comprehensive python-based system
mol 493:321–356 for macromolecular structure solution. Acta
26. Bottcher J, Jestel A, Kiefersauer R et al (2011) Crystallogr D Biol Crystallogr 66:213–221
Key factors for successful generation of 40. Emsley P, Cowtan K (2004) Coot: model-
protein-fragment structures requirement on building tools for molecular graphics. Acta
protein, crystals, and technology. Methods Crystallogr D Biol Crystallogr 60:2126–2132
Enzymol 493:61–89 41. Hamalainen MD, Zhukov A, Ivarsson M et al
27. Prakash O, Eisenberg MA (1979) Biotinyl 5’- (2008) Label-free primary screening and affin-
adenylate: corepressor role in the regulation of ity ranking of fragment libraries using parallel
the biotin genes of Escherichia coli k-12. Proc analysis of protein panels. J Biomol Screen
Natl Acad Sci U S A 76:5592–5595 13:202–209
28. Giannetti AM (2011) From experimental 42. Schrodinger, Llc (2010) The PyMOL molecu-
design to validated hits a comprehensive walk- lar graphics system, version 1.3r1
INDEX
Cheryl L. Meyerkord and Haian Fu (eds.), Protein-Protein Interactions: Methods and Applications, Methods in Molecular Biology,
vol. 1278, DOI 10.1007/978-1-4939-2425-7, © Springer Science+Business Media New York 2015
615
616 Index
Computational prediction (cont.) E
BLAST ............................................. 65, 71, 78, 86, 89
docking Electrophoresis ....................... 29, 66, 73, 282, 286, 358,
AutoDock .........................................81, 82, 86, 87 423, 436, 486, 489, 501, 503, 504, 509, 510.
DOCK ................................................................. 81 See also SDS-PAGE
flexX ..................................................................... 81 Enhanced chemiluminescence (ECL).........................311,
glide ..................................................................... 81 315–317, 372, 374, 393, 402, 501
GOLD .........................................................81, 581 Enthalpy........................ 8, 184, 185, 187–192, 197–199,
motif prediction .................................................. 60–73 201, 256, 257, 261–263
Pfam .......................................................................3, 15 Entropy .................................. 8, 184, 185, 191, 198, 257
scansite Enzyme-linked immunosorbent assay (ELISA)
database search ..............................62, 66–68, 286, competitive/inhibition ................. 342, 343, 346–350
295–298, 300 direct binding .......................................................... 342
input motif, using .........................................62, 64 sandwich .................................................................. 342
protein sequence .............................57, 61, 62, 65, Equilibrium constants.........................133, 206, 207, 221
66, 69, 78–80, 86, 282, 310, 314
F
public database, from.......................................... 58
quick motifs method.....................................66, 72 Far Western blot. See Blot overlay
scansite motif, using............................... 69, 71, 72 Fast protein liquid chromatography
sequence input ..............................................62, 66 (FPLC) .................................. 226, 230. See also
virtual screening Gel filtration chromatography
bindingDB ........................................................... 83 Flow cytometry ............................................................. 475
ChEMBL ............................................................. 83 Fluorescein ................................323, 324, 456, 470, 473,
drugBank ............................................................. 83 475, 477–479, 530, 578
OEChem ............................................................. 82 Fluorescence
PubChem............................................................. 83 emission ...............................329, 331, 332, 334, 338,
ZINC ................................................................... 82 457, 462, 476, 477, 509, 518, 540, 547, 549
Computational prediction. ........................ 39, 46–48, 50, excitation ............................. 330–332, 334, 457, 458,
53, 57–73. See also Internet resources 462, 476–448, 509, 534, 546, 547, 549
Confocal microscopy Fluorescence-activated cell sorting .............................. 475
detection of endogenous proteins ....... 517, 521, 522 Fluorescence anisotropy. See Fluorescence polarization
detection of transiently expressed Fluorescence Polarization (FP)
proteins ............................................... 116, 517 adaptation for HTS .............................. 472, 476, 477,
Coomassie blue ................................. 286, 292, 301, 358, 529–543, 574, 576, 577
360, 397, 398 fluorophores .................................323, 329, 330, 470,
Crystallography ............................. 4, 5, 58, 80, 101, 102, 517, 518, 530, 546
246, 267, 308, 392, 588, 592, 595, Fluorescence resonance energy transfer (FRET)
596, 601, 608. acceptor fluorophore.....................329, 330, 458, 459
See also X-ray crystallography donor fluorophore .................................................. 329
Fluorescence lifetime imaging microscopy
D (FILM)......................................................... 331
Differential scanning fluorimetry Förster radius/distance.................................. 331, 457
for fragment-based screening ........................ 587–612 live cell imaging.............................................. 334, 335
Dihydrofolate reductase (DHFR) fluorescence assay Time-resolved FRET (TR-FRET) ................ 545–552
flow cytometry analysis ........................................... 475 lanthanide .......................................................... 546
fluorometric analysis ............................................... 476 long-lifetime donor........................................... 546
microscopic analysis ................................................ 512 Fluorescent proteins................................... 331, 483–485,
survival assay .......................................... 470, 472, 474 492, 517. See also DsRed2-Monomer;
Dissociation constant ................................. 24, 27, 30, 32, Fluorescein; Green fluorescent protein (GFP);
33, 126, 130, 136, 138, 162, 167, 184, Red fluorescent protein (RFP); Venus; Yellow
187–190, 199, 201, 240, 251–257, 323, 531, fluorescent protein (YFP)
537, 578, 601 Fluorophore ..............................323, 324, 329, 330, 350,
Druggability ...........................80, 84–86, 94, 95, 99, 104 367, 457, 459, 470, 471, 476, 479, 517, 518,
DsRed2-Monomer............................................... 517, 518 525, 530, 546, 552, 578
Index 617
Förster resonance energy transfer. See Fluorescence Immunoprecipitation
resonance energy transfer co-immunoprecipitation .......................308, 381–389,
Fragment-based drug discovery .............................93–105 484, 492, 516
Fragment-based library............................... 102, 591, 597 Interactomics ................................................................. 449
Internet resources
G bookshelf at NCBI ........................46, 61, 65, 71, 286
Gal4...................409–430, 433, 434, 436, 437, 443, 445 databases
Gateway vectors................................. 368, 412, 413, 451, BIND ...................................................... 45, 46, 51
BioGRID ............... 39, 41, 43, 45, 47, 50, 51, 80
486–489, 493, 494
Gel filtration chromatography CATH .....................................................................5
AKTApurifier FPLC system.................................... 226 DIP ...................................................43, 45–47, 51
MINT ...............................................44–47, 51, 80
size exclusion chromatography .................... 206, 217,
223, 233 Pathguide............................................................. 40
stokes radii ............................................................... 223 Protein Data Bank (PDB) ........................ 3, 8, 14,
15, 58, 78, 82–89, 250, 273–275, 569,
Glutathione matrix...................................... 353, 354, 358
Glutathione-S-transferase (GST) ........................ 353–364 571, 595
Green fluorescent protein (GFP) ....................... 323–326, Reactome .............................39, 42, 44–47, 50, 51
STRING .................................... 39, 41, 47, 50, 51
383, 397, 398, 469–471, 473, 476, 483–485,
493, 500, 516–521, 524, 556–559, 561, 563 Domain searches and interaction predictions
GST-fusion techniques GeneMANIA ....................................39, 47, 49–51
expression and purification interdom .............................................................. 44
InterPro Scan ................................................69, 71
in E. coli ....................................................353–364
in mammalian cells ..................353, 360, 361, 368 Protein-Protein Interaction Prediction
expression plasmids .......................355, 368, 383, 517 (PIPs) ..........................................47, 50, 57–73
ScanSite (see Computational prediction)
lysis buffers ..................................................... 363, 381
radiolabeled technique for identification of novel ExPASy Molecular Biology Server ................ 133, 221
interacting proteins ............................ 354, 362 Locating reagents and protocols
BioSupplyNet ...................................459, 462, 464
H protocol Online................................................. 395
PubMed ...............................................................46, 50
Hexahistidine (6X His)......................114, 116, 365–370, Isopropyl-β-D-1-thiogalactopyranoside
372, 373, 375, 376 (IPTG) ........................................355–358, 363
High content screening Isothermal Titration Calorimetry (ITC) ........... 184–187,
biosensor assay for detection of PPI ............. 555–564 189, 191, 192, 194, 195, 199, 580, 603, 606
High performance liquid chromatography
(HPLC)...................230, 234, 235, 237, 285, K
289, 301
Kinetics ............................... 32, 110, 113, 117, 128, 132,
High throughput screening................................ 139, 141,
143, 149, 151, 449, 459, 468, 498, 499, 542, 133, 137, 141, 165, 167, 168,
546, 569 171–176, 178, 180, 267, 588–590,
601, 611
Homogeneous time resolved fluorescence (HTRF).
See Time-resolved FRET (TR-FRET)
L
Horseradish peroxidase..................... 311, 315, 316, 342,
344, 402 Label-free technology ................................................... 165
LacZ reporter ................................................................ 411
I LexA............................................................. 412, 433, 443
Ligand...............................................7, 13–15, 23–36, 43,
IC50, half maximal inhibitory concentration................ 30,
343, 347–350, 539, 542, 549, 556, 561, 570, 78, 79, 81–85, 89, 98, 100, 110–120,
571, 574, 575, 577, 579–581 122–128, 130–137, 151, 155, 160–162,
165–170, 172–177, 179–181, 183–186, 189,
Immunoblot/immunodetection........................ 282, 292,
382–386, 388, 389, 393, 402, 435, 436, 200, 240, 249, 252, 267–269, 273, 274,
442, 443, 445, 463, 475, 508, 513. See also 277, 324, 350, 402, 447–449, 530,
Western blot 537, 545, 547, 549, 570, 592–595,
597, 603–609
Immunofluorescence .................490, 516, 517, 519, 521
618 Index
Light scattering (LC) Polymerase chain reaction (PCR) ...................... 422, 425,
interferometric refractometer ........................ 234, 236 430, 435, 437–440, 444, 451, 488, 489, 492,
stoichiometry.................................................. 233–238 503, 504, 576, 591, 597, 601, 602
Luciferase Protein
assay ................................................................ 450, 501 co-localization ...................... 3, 46, 51, 329, 515–525
firefly luciferase ...................................... 498, 500, 508 conformation ........................ 239, 243, 244, 249, 332
renilla luciferase .............................................. 457, 458 cross-linking
split-luciferase complementation assay (See Protein cross-linkers ........... 392, 396, 397, 399–401, 403
fragment complementation assay) quantification............................................ 393, 402
in vitro ..................................... 391–393, 396–398
M in vivo ........................................................391–404
digestion ................................................ 283, 285, 300
Mass spectrometry
iTRAQ/TMT....................... 283, 285, 293, 298, 299 domain
LC-MS/MS................. 282, 283, 285–295, 298, 300 interaction......................... 43, 44, 50, 60, 78, 375
search .............................................................44, 78
MALDI-TOF ................................................. 283, 284
shotgun proteomics ....................................... 281, 282 folding............................................288, 366, 469, 493
spectral counting ........................................... 283, 287, identification........................281, 285, 286, 290, 295,
299, 300, 303
291, 298
stable isotope labeling with amino acids in cell motif ..................................14, 60–69, 142, 169, 375,
culture (SILAC) ................................ 283, 285, 447, 448, 459
286, 292, 293, 298, 299, 301, 302 oligomerization ..................................... 205, 402, 485
phosphorylation ............................... 17, 18, 142, 143,
Microscopy ..................................4, 5, 19, 329, 331, 334,
338, 386, 400, 470, 471, 475–477, 479, 480, 290, 377, 387, 447, 448, 531
515, 520, 523 post-translational modification..........................44, 51,
58, 72, 285, 290–291, 299, 412, 459
N quantification................................285–287, 291, 296,
298, 300, 473
Nuclear magnetic resonance (NMR) secondary structure (see Protein Secondary structure)
chemical-shift perturbation .................................... 606 thermodynamics ............................240, 241, 253, 257
for fragment-based screening ...............588, 591–595, unfolding ........................................................ 575, 590
603, 605 Protein A/Protein G ................................. 115, 251–254,
Heteronuclear single quantum correlation 332, 334–336, 338, 382, 384, 385, 387, 388,
(HSQC) ................... 270–272, 274, 276, 277, 478, 479, 576
579, 593, 594, 606, 607 Protein fragment complementation assay (PCA)
molecular docking......................................78, 86, 268 applications ..................................................... 470, 471
paramagnetic relaxation enhancement.................. 268, general principle ............................................. 467–480
269, 273 PCA reporters
saturation transfer difference ......................... 593, 603 β-lactamase assays (see β-lactamase assays)
WaterLOGSY........................................................... 606 Dihydrofolate reductase (DHFR) fluorescence
assay (see Dihydrofolate reductase (DHFR)
O
fluorescence assay)
Overlay assay. See Blot overlay Green fluorescent protein (GFP) assay ............ 471
Split-luciferase complementation
P assay......................................................497–513
Venus bimolecular fluorescence complementation
Peptide arrays
assay.................................... 484–488, 490–494
alanine screening ....................................308, 315–317
Protein Secondary structure
fmoc-chemistry........................................................ 307
identifying pockets
ResPep SL SPOT ........................................... 308, 312
CavBase................................................................ 79
Peptide library ................................................................. 99
fpocket .............................................. 79–81, 84–86
Phage display ................................................................... 99
FTSite ...............................................79, 80, 84–86
Pharmacophore screen......................................... 580–581
SiteMap............................................. 79–81, 84–86
Phosphopeptide ......................................... 286, 291, 302,
Voronoi tessellation............................................. 79
531, 532
Proteomics.............................. 18, 48, 67, 109, 221, 224,
Photobleaching ........................................... 331, 336, 338
281–283, 285, 293, 295
Polyacrylamide gel. See Electrophoresis
Index 619
Q Binding kinetics...................113, 117, 128, 133, 137,
165, 167, 168, 174, 176, 180, 588, 590
QCM-D chip selection .........................................111–126, 128,
binding affinity ........................................................ 154 130, 133, 136, 597, 598
EDC/NHS crosslinking ......................................... 158 for fragment-based screening ........................ 102, 603
sauerbrey equation ........................154, 160, 161, 164 sensor surface immobilization ...................... 112, 114,
sensor chip ...................................................... 161, 163 597, 598
Quantitative analysis ...................... 23–36, 294, 302, 350
T
R
ThermoFluor microcalorimetry .......................... 276, 575
Red fluorescent protein (RFP) ........... 556–559, 561–563 Time-resolved FRET (TR-FRET) ...................... 545–552
Resonant waveguide grating Transfection...................... 292, 334, 355, 361, 368–370,
biosensor.................................................139–143, 150 383, 384, 387, 394, 397–399, 401, 438,
corning Epic ................................................... 139–151 450–452, 459–461, 464, 472–477, 486, 490,
evanescent wave.............................................. 140, 141 491, 493, 494, 500, 502, 504, 505, 508, 512,
513, 516, 517, 519, 524, 525, 556
S
Transformation............................ 27, 195, 357, 413–425,
Sacchromyces cerevisia strains 427–430, 438–441, 444, 488–490
AH109 ........................ 411, 413, 414, 416–420, 424, Two-hybrid systems
425, 427 mammalian two-hybrid assay
Y187...............................................413, 418, 424, 425 MAmmalian Protein-Protein Interaction Trap
Y190....................................................... 436, 437, 443 (MAPPIT) ...........................................447–454
Scansite. See Computational prediction Plasmids ............................................449–451, 453
Screening library ........................................................... 101 reverse two-hybrid ......................................... 433–445
SDS-PAGE ................................................. 355, 358, 360, Characterization of isolated mutants .......433–445
362, 369, 371–374, 376, 383, 385, 393, 396, loss-of-interaction mutation ................... 434, 435,
398, 400, 401, 403, 425, 430, 436, 501, 510. 438, 440
See also Western blot mutagenesis ......................................435, 437–439
Sedimentation equilibrium plasmids ................................... 438, 439, 441–445
equilibrium constants............................ 206, 207, 221 strains (see Sacchromyces cerevisia strains)
heterologous system ............................................... 219 Yeast two-hybrid system ................................ 409–429
monomeric species ......................................... 214–216 3-AT (3-amino-1,2,4-triazole)........411, 418, 425
Sepharose beads ............................................................ 532 Bait auto-activation test .................................... 413
Size exclusion chromatography........ 206, 217, 223, 233. confirmation test ...................................... 417, 424
See also Gel filtration chromatography mating ...............................................413, 424, 425
Split-luciferase complementation assay ............... 497–513 medium................................... 411, 414–422, 424,
Dual-color split luciferase complementation 425, 428, 430
assay.............................................................. 511 plasmid isolation......................413, 417, 422, 443
Stoichiometry ................................ 26, 27, 29, 30, 32, 33, plasmids ..................................412, 414, 416, 417,
42, 83, 103, 113, 177, 196, 201, 212, 423–425, 427, 429
233–238, 255, 290, 332, 580, 588, 589, quantitative α-galactosidase assay........... 413, 418,
591, 600, 601, 606 425–426
Structural basis screening libraries..................................... 102, 587
crystallography ................................. 4, 5, 58, 80, 101, strains (see Sacchromyces cerevisia strains)
102, 246, 267, 308, 392, 588, 592, 595, 596,
601, 608 U
Domain-domain interaction.................................7, 23 Ubiquitin .............................................290, 300, 412, 568
Domain-peptide interaction ............... 6, 15, 151, 540
"Hot spot," ................................... 7–9, 11–13, 81, 94 V
signaling complex........................................... 6, 12, 13
Surface plasmon resonance (SPR) Venus ..................................................331, 332, 334–338,
Biacore/GE Healthcare...............110, 121, 122, 211, 458, 460, 484–488, 490–494
393, 532, 570, 589 Virtual Screening. See Computational prediction
620 Index
W X
Websites ............ 16, 286, 552. See also Internet resources X-α-Gal (5-bromo-4-chloro-3-indolyl-α-D-galactop-
CD analysis ..................................................... 239–263 yranoside)............................................ 415, 436
Cross-linkers ......................................... 392, 396, 397, X-ray crystallography
399–401, 403 for fragment-based screening ....................... 588, 595,
pGEX vectors ................................ 355–357, 359, 361 596, 601, 608
Protein Data Bank (PDB) ........................3, 8, 14, 15,
58, 78, 83–89, 273–275, 569, 571, 595 Y
Protein domains ............................44, 50, 60, 78, 375 Yeast two-hybrid. See Two-hybrid systems
Scansite ................................................... 61–63, 65–73 Yeast two-hybrid system
Sedimentation equilibrium ............................ 205–222
Medium
Two-hybrid systems .............................. 433, 434, 445 LB .....................................................417, 422, 423
Western blot ............................................... 292, 300, 342, SD .........415, 416, 419, 420, 422, 425, 426, 428
358, 360, 362, 365, 368–377, 393, 400–402, YPDA ........................................................ 414, 416
475, 479, 490, 501, 508, 509, 518, 521.
Yellow fluorescent protein (YFP) .............. 336, 458–461,
See also Immunoblot/immunodetection 463, 484, 485, 491, 492
Far Western blot (see Blot overlay)