Functional Proteomics Methods and Protocols
Functional Proteomics Methods and Protocols
Xing Wang
Matthew Kuruc
Editors
Functional
Proteomics
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Xing Wang
Array Bridge Inc., St. Louis, MO, USA
Matthew Kuruc
Biotech Support Group LLC, Monmouth Junction, NJ, USA
Editors
Xing Wang Matthew Kuruc
Array Bridge Inc. Biotech Support Group LLC
St. Louis, MO, USA Monmouth Junction, NJ, USA
This Humana Press imprint is published by the registered company Springer Science+Business Media, LLC, part of
Springer Nature.
The registered company address is: 233 Spring Street, New York, NY 10013, U.S.A.
Preface
Over the past two decades, tremendous progress has been made in the field of proteomics,
the purpose of which is to find systemic differences in protein populations. Once established,
measurable protein markers can then help to define biological mechanism, disease, identify
therapeutic targets, and offer better precision for personalized medical interventions.
Proteomics, like other “omics” analyses, is data driven and can generate unbiased
protein profiles for a variety of end points that can contrast, for example, treated vs. untreated
cell models or healthy vs. diseased tissue; it has provided us with more in-depth understanding
of many biological systems and diseases. This progress in the field of proteomics parallels with
the advancement in many analytical technologies, especially in mass spectrometry, which has
been evolved from a less sensitive and qualitative tool to high sensitivity and quantitation system
for protein analysis and characterization. Currently, system biology and proteomics in particular
are advancing biology at two opposite but equally important polarities; one is the holistic
understanding of a biological system, be it an organism, organ, tissue, or the human circulation
system, and the other one is for single-cell analysis whereby biological heterogeneity can be
minimized and a more discrete picture of biological processes can be modeled within a more
homogeneous context. Having the tools and methods necessary to address these important
studies will promote the significant impact that is foreseen in precision medicine and other
biological fields.
In the most common view of proteomics, data is generally acquired after proteolytic
processing of the parent proteomes. The derived peptides are then analyzed on instruments
coupling Nano-Liquid Chromatography to Mass Spectrometry (LC-MS). Such instruments
generate mass spectra of peptides and the MS2 spectra through further fragmentation,
which can be compared to theoretical amino acid spectra definable through public gene
repositories. Peptide sequence matches are thus computationally derived, and from that
data, protein identifications are inferred. From such analyses, peptide markers can be used as
surrogates for the gene products from which they are derived. Through differential expres-
sion analysis of these peptide markers, proteomics can thus help identify those gene products
that define a phenotype. However the functions of the proteome, the driving force for
almost all biological actions, are not adequately annotated through the current infrastruc-
ture of methods surrounding LC-MS sequence annotation. This book is intended to fill in
this knowledge and technology gap with a specific collection of technologies that have been
developed for the study of protein function at a proteome scale. In organizing the content of
this book, the following points were taken into consideration: (1) It should bridge the
understanding of biology from protein functions to other aspects of protein analysis,
especially in post-translational modification, as most cellular proteins use this mechanism
to carry out their unique role in cellular regulation. (2) The book should also act as a bridge
to other levels of system biology research including genomics and metabolomics, so that the
readers will gain a relatively complete picture of how one might study the biological system
of their interest. (3) Technologies are categorized toward different aspects of protein
functional analysis, so that readers can understand what is available to them in functional
proteomics research. (4) Finally, the selection of technologies also takes into consideration
the impact on current and future research in a variety of disease areas.
v
vi Preface
It is hoped that by using these novel technologies, new frontiers in biological research
will be created, important drug targets can be identified, and clinically validated biomarkers
and diagnostic tests can be developed. The aim of the editors of this book is to provide the
most precise description of our technological capabilities in functional proteomics research
and give our readers the tools they will need to create the new functional domains of our
knowledge in the understanding of various biological systems.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
1 Making the Case for Functional Proteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
Ray C. Perkins
2 Methods to Monitor the Functional Subproteomes of SERPIN
Protease Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Swapan Roy and Matthew Kuruc
3 Two-Dimensional 16-BAC/SDS Polyacrylamide Gel Electrophoresis
of Mitochondrial Membrane Proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
Gary Smejkal and Srikanth Kakumanu
4 Systematic Glycolytic Enzyme Activity Analysis from Human
Serum with PEP Technology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 69
David Wang
5 A Protein Decomplexation Strategy in Snake Venom Proteomics . . . . . . . . . . . . . 83
Choo Hock Tan, Kae Yi Tan, and Nget Hong Tan
6 Fractionation Techniques to Increase Plant Proteome Coverage:
Combining Separation in Parallel at the Protein and the Peptide Level . . . . . . . . 93
Martin Černý, Miroslav Berka, and Hana Habánová
7 A Systematic Analysis Workflow for High-Density Customized
Protein Microarrays in Biomarker Screening. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 107
Rodrigo Garcı́a-Valiente, Jonatan Fernández-Garcı́a,
Javier Carabias-Sánchez, Alicia Landeira-Viñuela, Rafael Gongora,
Marı́a Gonzalez-Gonzalez, and Manuel Fuentes
8 Metaproteomics Study of the Gut Microbiome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
Lisa A. Lai, Zachary Tong, Ru Chen, and Sheng Pan
9 Double One-Dimensional Electrophoresis (D1-DE) Adapted
for Immunoproteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 133
Youcef Shahali, Hélène Sénéchal, and Pascal Poncet
10 BioID: A Proximity-Dependent Labeling Approach in Proteomics Study . . . . . . 143
Peipei Li, Yuan Meng, Li Wang, and Li-jun Di
11 Functional Application of Snake Venom Proteomics in In Vivo
Antivenom Assessment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 153
Choo Hock Tan and Kae Yi Tan
12 Proteomic Detection of Carbohydrate-Active Enzymes (CAZymes)
in Microbial Secretomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 159
Tina R. Tuveng, Vincent G. H. Eijsink, and Magnus Ø. Arntzen
13 An Overview of Mass Spectrometry-Based Methods
for Functional Proteomics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
J. Robert O’Neill
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 467
Contributors
JUHANI AAKKO Turku Centre for Biotechnology, University of Turku and Åbo Akademi
University, Turku, Finland
SHIVA AHMADI Institute for Biochemistry and Molecular Biology, University of Bonn, Bonn,
Germany
MAGNUS Ø. ARNTZEN Faculty of Chemistry, Biotechnology and Food Science, Norwegian
University of Life Sciences (NMBU), Ås, Norway
SANDRINE AROS-CALT Service de Pharmacologie et d’Immunoanalyse, Laboratoire d’Etude
du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB,
Gif-sur-Yvette, France; bioMérieux S.A., Marcy l’Etoile, France
JOHN R. BARR Division of Laboratory Sciences, Centers for Disease Control and Prevention,
Atlanta, GA, USA
MATTEO BATTARRA Diabetes Research Institute, University of Miami, Miami, FL, USA
MIROSLAV BERKA Faculty of AgriSciences, Department of Molecular Biology and
Radiobiology, CEITEC—Central European Institute of Technology, Phytophthora Research
Centre, Mendel University in Brno, Brno, Czech Republic
KEREN BYRNE CSIRO Agriculture and Food, St Lucia, QLD, Australia
ZHENG CAO Department of Laboratory Medicine, Beijing Obstetrics and Gynecology
Hospital, Capital Medical University, Beijing, China
JAVIER CARABIAS-SÁNCHEZ Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/
USAL/IBSAL), Salamanca, Spain
FLORENCE A. CASTELLI Service de Pharmacologie et d’Immunoanalyse, Laboratoire d’Etude
du Métabolisme des Médicaments, CEA, INRA, Université Paris Saclay, MetaboHUB,
Gif-sur-Yvette, France
MARTIN ČERNÝ Faculty of AgriSciences, Department of Molecular Biology and
Radiobiology, CEITEC—Central European Institute of Technology, Phytophthora Research
Centre, Mendel University in Brno, Brno, Czech Republic
RU CHEN Department of Medicine, University of Washington, Seattle, WA, USA
YUN CHEN School of Pharmacy, Nanjing Medical University, Nanjing, China
MICHELLE L. COLGRAVE CSIRO Agriculture and Food, St Lucia, QLD, Australia
LI-JUN DI Faculty of Health Sciences, Cancer Center, University of Macau, Macau, China
HU DUAN State Key Laboratory of Proteomics, Beijing Proteome Research Center, National
Center for Protein Sciences (PHOENIX Center, Beijing), Beijing Institute of Lifeomics,
Beijing, China
VINCENT G. H. EIJSINK Faculty of Chemistry, Biotechnology and Food Science, Norwegian
University of Life Sciences (NMBU), Ås, Norway
LAURA L. ELO Turku Centre for Biotechnology, University of Turku and Åbo Akademi
University, Turku, Finland
ANDREW EMILI Donnelly Centre for Cellular and Biomolecular Research, University of
Toronto, Toronto, ON, Canada; Department of Biology, Boston University, Boston, MA,
USA; Department of Biochemistry, Boston University, Boston, MA, USA; Center for
Network System Biology, Boston University, Boston, MA, USA
ix
x Contributors
HAILI LI CSIRO Agriculture and Food, St Lucia, QLD, Australia; Institute of Animal
Husbandry and Veterinary Science, Henan Academy of Agricultural Sciences, Zhengzhou,
Henan, China
PEIPEI LI Faculty of Health Sciences, Cancer Center, University of Macau, Macau, China
LIANG LIU School of Pharmacy, Nanjing Medical University, Nanjing, China
DANIEL MALZL Department für Chemie, Universit€ a t für Bodenkultur, Vienna, Austria
YUAN MENG Faculty of Health Sciences, Cancer Center, University of Macau, Macau,
China
BRUNO H. MULLER bioMérieux S.A., Marcy l’Etoile, France
SHENG PAN Institute of Molecular Medicine, University of Texas Health Science Center at
Houston, Houston, TX, USA
KATHARINA PASCHINGER Department für Chemie, Universit€ a t für Bodenkultur, Vienna,
Austria
RAY C. PERKINS New Liberty Proteomics Corporation, New Liberty, KY, USA
SAMI PIETIL€a Turku Centre for Biotechnology, University of Turku and Åbo Akademi
University, Turku, Finland
SNEHA M. PINTO Center for Systems Biology and Molecular Medicine, Yenepoya Research
Centre, Yenepoya (Deemed to be University), Mangalore, India
PASCAL PONCET Allergy and Environment Team, Biochemistry Department, Armand
Trousseau Children Hospital (AP-HP), Paris, France; Center for Innovation and
Technological Research, Institute Pasteur, Paris, France
REZA POURHAGHIGHI Donnelly Centre for Cellular and Biomolecular Research, University
of Toronto, Toronto, ON, Canada
J. ROBERT O’NEILL Cancer Research UK Edinburgh Centre, MRC Institute of Genetics
and Molecular Medicine, The University of Edinburgh, Edinburgh, UK; Department
of Clinical Surgery, Royal Infirmary of Edinburgh, Edinburgh, UK
SWAPAN ROY Biotech Support Group LLC, Monmouth Junction, NJ, USA
HÉLÈNE SÉNÉCHAL Allergy and Environment Team, Biochemistry Department, Armand
Trousseau Children Hospital (AP-HP), Paris, France
YOUCEF SHAHALI Razi Vaccine and Serum Research Institute, Agricultural Research,
Education and Extension Organization (AREEO), Karaj, Iran
GARY SMEJKAL Focus Proteomics, Hudson, NH, USA
YASHWANTH SUBBANNAYYA Center for Systems Biology and Molecular Medicine, Yenepoya
Research Centre, Yenepoya (Deemed to be University), Mangalore, India
TOMI SUOMI Turku Centre for Biotechnology, University of Turku and Åbo Akademi
University, Turku, Finland
CHOO HOCK TAN Venom Research and Toxicology Laboratory, Department of
Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
KAE YI TAN Protein and Interactomic Laboratory, Department of Molecular Medicine,
Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
NGET HONG TAN Protein and Interactomic Laboratory, Department of Molecular
Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
ZACHARY TONG Department of Medicine, University of Washington, Seattle, WA, USA
CHRISTOPHER A. TOTH Division of Laboratory Sciences, Centers for Disease Control and
Prevention, Atlanta, GA, USA
TINA R. TUVENG Faculty of Chemistry, Biotechnology and Food Science, Norwegian
University of Life Sciences (NMBU), Ås, Norway
xii Contributors
Abstract
“Making the Case for Functional Proteomics” first differentiates the Functional Proteome from the
products of genetic protein expression. Qualitatively, the prevalence of posttranslational modifications
(PTMs) virtually insures that individual, functional proteins do not equate to their genetic expression
counterparts. Quantitatively, considering the frequency of PTMs and a conservative estimate of the number
of functional entities arising from protein interactions, the size of the Functional Proteome exceeds that of
the human genome by at least two orders of magnitude. The human genome does not, cannot, map the
Functional Proteome. Further, the collective genome of the human microbiome dwarfs the human
genome. With these facts established, “Making the Case. . .” proceeds to examine Functional Proteomics
(of which both “gene expression” and “epigenetics” are but parts of a larger whole) within the context of
Systems Biology, concluding that functionally related networks comprise the dominant motif for biological
activity. Creating just such a network focus is essential in not only expanding basic knowledge but also in
applying that knowledge in the pragmatic efforts of drug and biomarker development. Outlines for
development of drugs and biomarkers, as well as the realization of precision medicine, within a functional
proteomics-based, network motif are provided. The chapter proceeds to asses both the knowledge base and
the tools to fully embrace Functional Proteomics. Given the decades-long infatuation with the reduction-
ism of genomics, it is not surprising that both the proteomics knowledge base and tools are assessed as poor
to fair. However, even a minor shift in research funding and a renewed challenge to methods developers will
rapidly improve the current situation. Adoption of the included “Roadmap” will realistically make the
twenty-first century the century of a long-awaited revolution in biology.
Key words Protein, Gene, Genome, Proteome, Functional Proteome, Proteomics, Functional prote-
omics, Microbiome, Posttranslational modifications, Protein interactions, Epigenetics, Gene expres-
sion, Biological networks, Systems biology, Drug development, Biomarker development, Precision
medicine
1 Introduction
The title of this chapter, “Making the Case for Functional Proteo-
mics,” can elicit a sense of the absurd. For example, it is difficult to
name a single biological property or process that does not rely on
the “function” of proteins. Basic properties such as size and shape
of the organism, its interaction with the environment, and the
existence of functionally specialized structures directly reflect the
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_1, © Springer Science+Business Media, LLC, part of Springer Nature 2019
1
2 Ray C. Perkins
Table 1
Posttranslational categories and incidence
a b
1,976,760
Functional Proteome Size Estimate Non-Reduntant
(Human) Genes: Human vs
Microbiota
329,460
64,600
19,000
Fig. 1 (a) The challenge of proteomics lies in its fundamental, irreducible complexity. Taking into account
alternative splicing and posttranslational modifications, a single human gene can produce an average of
12 unique proteins, all needing to be identified and catalogued. The true interest in proteins lies in their
functions, however, and for that we must discern, quantify, catalogue, and compare all of their many
interactions. With an average of five binary interactions for every protein (ignoring multi-center protein
complexes and interactions with other types of molecules), we are confronted with a staggering sum of
nearly two million to identify and study! Compare this to the measly 19,000 protein-coding genes in the human
genome, and it is clear why reductionist approaches will never be up to the task. (b) The impact of the
microbiome, based on its sheer size alone, must be significant. However, there is scarce data on the impact of
non-infectious bacterial proteins on human proteins. (The effect of some infectious bacterial proteins are
discussed in “Proteases: Pivot Points in Functional Proteomics”)
This section of “Making the Case” is the context for the remainder
of the chapter. The backdrop is, once again, decades of emphasis on
the genome. The relatively recent expectation of genomics is
expressed in the following attributed to Francis Collins (currently
director of the US National Institutes of Health) in 2006 [6]:
“Essentially, we are now able to read our own instruction books. It is also a
history book explaining how humans have evolved over time. It’s a shop manual
that describes with incredible precision how to build every cell in the human
body. And it’s a medical textbook containing insights that will help doctors
predict and, eventually, cure disease.”
8 Ray C. Perkins
Fig. 2 The human body is so complex that we have traditionally approached it as a multilayered hierarchy of
tissues, cells, and molecules. While this is a useful organizational tool, in reality these layers are not separate.
They inform and influence each other. Therefore, our approach to studying them must not treat them as
separate but as an integrated whole, Systems Biology. When investigating the detailed mechanisms of protein
function, an awareness must be maintained of their cellular environment, and any deviations from biological
conditions should be minimized and must be acknowledged. Testing on the organism level should be done
with an eye toward how the interconnected pathways of molecular networks may cause unintended effects.
More than anything else, a common vocabulary of materials identification and handling must be developed
and maintained between the “disciplines” so that apples-to-apples comparisons can be made between
studies
a b
Diagnostic Prognostic
(Disease Differentiation) (Track Disease Progress)
FDA-Approved, In
2,200
Biomarkers Vitro Diagnostics
Therapy Selection Predictive
(Match Disease & Patient) (Anticipate Disease)
"Companion"
c
“Pattern Diagnostics” Cancer Nucleotide
Diagnostic in Development Protein
9 66
Fig. 3 (a) Biomarkers are, in essence, anything that we can measure that informs us about a person’s health.
Historically, we have used them to diagnose disease and to track disease progress. Recently, there has been
an effort to use them as a means of determining which patients would be best served by which treatments,
which has met with some success. There has also been an effort to use them to predict which diseases a
patient is likely to be afflicted by, which has met with less success and more controversy. (b) In vitro
diagnostic tests are regulated by the FDA, and tests of protein quantities and/or activities outnumber tests for
gene variants 33 to 1. “Companion” diagnostics (of which there are only 9) are tests that select treatment-
suitable patients already diagnosed with a specific disease condition, the hopeful beginnings of the modern
Precision Medicine movement. (c) A recent study of cancer patients [13] detected a pattern of elevated serum
levels of multiple proteins associated with Hemostasis, Inflammation, and the Complement System. For more
information on the connections between these systems, see “Proteases: Pivot Points in Functional
Proteomics”
Differentiation Set V0
Diagnostic V0
for breast cancer, nine out of ten identified as at-risk will not have
the disease. Unfortunately, in addition to the considerable emo-
tional trauma induced by such errors, many women are unnecessar-
ily subject to both invasive biopsies and exposure to hard radiation
(ironically a procedure that increases risk of cancer). The bottom
line is that the bar for Predictive Diagnostics must be set orders of
magnitude higher than for Clinical Diagnostics. Unfortunately, in
today’s “wild west” practices, the reverse is true [16].
a b
100
New Pharmaceuticals
NMEs per Dollar (normalized to 1970-75)
Productivity Decline
Drug Discovery
“Chain of Translatability”
Assay
Biology Disease
0
1975 1980 1985 1990 1995 2000
Five-Year Rolling Average
Fig. 5 (a) The efficiency of drug discovery research has plummeted to less than 10% of what it had been in
1975 (data adapted from reference 19). While many factors are at play, a significant shift in approach
coincides with this period of decline: Target-based Drug Discovery (TDD). TDD is largely gene-centric,
selecting “target” proteins based on gene mutations—a model that has failed. A resurgence of more
phenotypic approaches is occurring, a resurgence highly intertwined with expanded characterization of the
Functional Proteome. (b) A succinct summary of Phenotypic Drug Discovery approaches that emphasizes the
fundamental importance of the Assay. The “Chain of Translatability” (reference 22) implies that the Assay
must reflect and inform Biology and Disease pathology
Making the Case for Functional Proteomics 19
and one not, exhibit selective affinity for different peptide oligo-
mers. Work is underway to reanalyze the library screening in accord
with this new knowledge. Had the original screening work begun at
the Cellular level, the Molecular activity would have been missed.
More to the point of PDD practices, any impact of test molecules
on the peptide screening readout would be misinterpreted and
assignment of mechanisms of action compromised.
This example is telling within both PDD and Functional Pro-
teomics contexts. Translatability to both biology and disease is
established through choice of test materials, the peptide and pro-
tein. In a direct sense, those crucial connections define the assay.
The assay itself directly tested for protein functionality, namely
interaction of a peptide and protein. Once executed, the assay
further expanded knowledge of the behavior of the peptide and
connected to disease via differential and entity-selective binding to
the protein. Finally as was mentioned above, the assay was immedi-
ately extended into assessments of modulation of protein networks
and ex vivo application of the selfsame assay is in planning.
What, then, is the future of phenotypic drug discovery within
the multiple contexts of Functional Proteomics, Systems Biology
and Translatability? First, starting in the middle of the Systems
Biology complexity hierarchy—Cells—may be a high-risk decision.
Until and unless cell systems are developed that mimic chronic
disease, the classic starting point for pathogenic PDD is problem-
atic. By contrast, most diseases have identifiable relationship to one
or more proteins so that a PDD development program can launch
with the least complexity but still in a disease-relevant mode. As a
bonus, molecular level screening is both faster and less expensive.
Next, as with the example provided, extending a molecular study
into network or pathway assays is, in principle if not in practice,
straightforward. The combination of molecular and network find-
ings will predictably inform both biology/disease and assay
planning for cells. Extension, then, into original source material
(patient-derived cells, fluids, extracts, etc.) is both abbreviated and
as fully informed as can be made possible. A realistic bonus is the
acquisition of knowledge that informs likely avenues for either a
disease or companion diagnostic. Finally, the example cited points
to yet another desirable outcome attainable through phenotypic
approaches—selection of test molecules for their multiple or “poly-
pharmacological” activities [26].
Within the complexity of any organism no molecule exhibits a
single activity at all time points. The very functional attribute of any
molecule that defines one activity, e.g., inhibition of a protein
interaction, statistically matches to multiple, comparable sites on
other proteins, cell, or nucleotides. Further, other attributes of the
same molecule enable its ability to engage in additional activities.
Thus multiple activities or functionalities are manifest by a single
test-library molecule (or any molecule). Aspirin is perhaps the
Making the Case for Functional Proteomics 23
a 42 Imprecise Medicine: b
Effectiveness of Common Traditional
Medications Precision Medicine Cycle
4 5 5
4
2
Aspirin (1200 mg) Aspirin (650 mg) Nexium Crestor Advair Diskus
Fig. 6 (a) The drive for precision (meaning individualized) medicine is clear when looking at “Number Needed
to Treat” figures for common medications. 1200 mg of Aspirin (equivalent to 2 extra-strength tablets or
capsules) is only effective for 42% of the American population due to variations in personal biochemistry. For
other popular medications a standard dose is effective for 5% or less. Knowing whether or not the patient is
one of the lucky 5% is critically important. (b) The cycle of precision medicine has been practiced for
millennia: Diagnose the patient. Determine what would be the most effective treatment and administer
it. (Hope fervently that the patient suffers no serious ill effects.) Check the patient’s progress and adjust
treatment, or even diagnosis, according to the results. With the development of Companion Diagnostics there
is the hope that the cycle can be made more efficient and minimize ill effects from inappropriate treatments
Table 2
Proteomic properties, methods, and proteome coverage
7.1 Proteomics Table 2 cross indexes key Functional Proteomics metrics to: Meth-
Methods Appraisal ods employed to assess the metric (Method Examples), the reliabil-
ity of a given measurement by the Method (Methods’ Reliability),
the potential for the Method to address the whole of the Functional
Proteome (Coverage Potential) for the Proteomic Property, and
the extent to which all existing knowledge addresses the whole of
the Functional Proteome for a given Proteomic Property. A couple
of examples are helpful. The structural Method, X-ray, provides
exquisitely detailed spatial information and is, therefore, a Reliable
method. However, its ability to Cover the entire proteome is
Limited by the inability to crystallize every protein in the proteome.
By comparison, the Activity method for protein interactions, Yeast
Two-Hybrid, is prone to a high incidence of false-positive and false-
Making the Case for Functional Proteomics 29
7.2 Identity The majority of “modern” efforts in proteomics has been and is
focused on protein identification, especially for mixtures of pro-
teins. Dominant in the effort is a variety of separation procedures
(“Sep-Sci,” gel electrophoresis, liquid chromatography, etc.) and
analysis by mass spectrometry (MS). Indeed the use of MS is often
(and wrongly) equated to proteomics and vice versa. Other identi-
fication methods include Edman sequencing, quantitative amino
acid analysis, and antibody-based analysis (e.g., enzyme-linked
immunosorbent assay or ELISA). Sep-Sci alone can be used for
rough identification though molecular weight resolution is typi-
cally insufficient to differentiate closely spaced isoforms or the
extent or kind of posttranslational modification. MS has somewhat
similar issues for some approaches though not for others. So-called,
“bottom-up” methods [33], typically applied to mixtures of pro-
teins, rely heavily on comparison of post-digestion sequence pat-
terns to databases of those sequences for individual proteins. Two
issues must be, and are, acknowledged: (1) Algorithm-driven ana-
lyses can only assess identification in accord with members of the
database (heavily weighted to proteins expressed by cDNA) and
(2) “Sequence coverage” for any given identified protein can range
from 30% to 99%, the range often reflecting relative concentrations
of proteins in the mixture. In contrast to bottom-up approaches,
several “top-down” MS approaches [34] are also available. As the
name implies, whole proteins comprise the test sample. Top-down
approaches can resolve closely spaced isoforms and can inform the
degree and kind of PTM. As with bottom-up, however, if algorith-
mic comparison to existing databases is employed, those analyses
are only trustworthy if the tested protein(s), including PTMs, is
already in the reference database. Further, Top-down methods are
typically applied to mixtures of fewer proteins. The bottom line for
Sep-Sci/MS is that identification results are reasonable within the
stated qualifiers. Confirmation, if needed, can be supplied by
“orthogonal” methods such as Edman Sequencing. Then and
only then can a non-qualified protein identification be reasonably
certain.
In sharp contrast to instrument-based methods such as MS,
antibody/antigen binding can be exploited to identify proteins
[35]. Generally, antibodies are prepared and isolated for single-
protein antigens. Subsequent binding to the antibody by a protein
from a mixture of proteins is taken as evidence that the protein is
30 Ray C. Perkins
7.3 Quantitation Hand in hand with the need for protein identification is determina-
tion of protein concentration. The list of applicable methods for
quantitation is comparable to those for identification, though exe-
cution differs. In the case of antibody-based work [35], addition of
a reporter group to the antibody, e.g., as is done in preparation of
ELISA assays, identification and quantitation occur in a single assay.
For MS-based work [36], a variety of labeling approaches provide
information on the relative concentrations of proteins in a mixture.
These methods coupled to carefully prepared control mixtures can
serve to provide some indication of absolute protein concentrations
in the original source material. However, for optimum determina-
tion of absolute protein concentration coupled to dramatically
improved detection of low-concentration proteins, application of
so-called “Multiple Reaction Monitoring (MRM)” [37] is the
approach of choice. It must be noted that MRM demands intensive
method development and absolute concentrations are available
only at the expense of “coverage.”
Making the Case for Functional Proteomics 31
7.4 Structure Structure and function are intimately related and only two methods
dominate determination of protein structures: X-ray crystallogra-
phy and “solutions” nuclear magnetic resonance spectroscopy
(NMR) [38]. Of these two, X-ray structures listed with the Protein
Data Bank (PDB) outnumber NMR structures listed by 10:1.
Combined, the two methods account for 98.4% of all structures
archived (cryo-Electron Microscopy, the object of a 2017 Nobel
prize, accounts for only 1.4% of PDB structures) [39]. The quality
of data forthcoming from these methods is outstanding—the issue
is coverage of the proteome.
As its name implies, X-ray crystallography demands samples
with significant, through-space uniformity. Coaxing a molecule
into such a form is a combination of science, method, and art,
and many proteins simply are not amenable to such forced unifor-
mity. This fact limits the range of proteins that can be studied by
X-ray. Coupled to this limitation is the inverse implication of cov-
erage: proteins forced into an extended, uniform state almost cer-
tainly do not represent the multiplicity of states of proteins in
solution. In the best of cases the crystallized structure may be
deemed a dominant or active conformer, in the worst the structure
may reflect no biologically active state. Solutions NMR largely
avoids this potential pitfall but has limitations of its own.
Samples for solutions NMR range in concentration from 0.1 to
5 mM [40] and, for the overwhelming majority of protein work,
contain proteins smaller than 50 kDa (slightly smaller than the
average for human proteins (53 kDa) and slightly larger than the
median (42 kDa)). For the most-practiced NMR methods as
applied to proteins, half the human proteome is not accessible
(NMR methods do exist to virtually eliminate the molecular weight
limitation but are limited by magnet technology—and cost). For
the 50% of human proteins accessible to solutions NMR, the infor-
mation content reflects not only secondary structures but also
partially inform variation in conformational states. Further, impacts
on structure by solution variation and small molecule effectors may
be assessed. Here, though, the second limitation of NMR, sensitiv-
ity/concentration, comes into play. With a preferred lower limit of
2 mM (1 mM ¼ 10 mg/mL for a 10 kDa protein), NMR sample
concentrations exceed the observed physiological concentrations of
nearly all proteins. Further, a typical sample size of slightly over
0.5 mL, samples can be costly, a consideration that is amplified by
32 Ray C. Perkins
7.5 Localization Two widely divergent methodologies must be considered for asses-
sing the status of protein localization: fluid-based and cell-based.
Biological systems naturally afford access to extracellular fluid,
examples being blood plasma, lymph fluid, cerebral spinal fluid,
and interstitial fluid. For these cases methods such as those dis-
cussed under Identification and Quantitation apply. Fluid-based
approaches can also be applied for selective isolation and lysis of
subcellular organelles. Thus, within the qualifiers noted above,
fluid-based localization studies are hypothetically amenable to
examination. Cell-based localization of proteins demands entirely
different approaches, exemplars being Immunofluorescence (IF)
and fluorescent-protein tagging (FP) [43].
Distinct proteins are selected for either direct modification
required for detection or as the antigen for protein-specific, fluo-
rescently labeled antibodies. Images of cells with incorporated
monitors reveal the localization of the labeled entities under a
variety of conditions of interest. For either labeling approach cer-
tain cautionary flags are raised. Extracellular introduction of labeled
proteins, whether FP or IF, cannot insure bio-relevant distribution,
especially to intracellular organelles. Next, as with all labeling
approaches the label itself may significantly alter behavior. In the
specific case of IF, lack of antibody selectivity produces false-
positive results. Given that they are by design active agents, anti-
bodies may also shift equilibria in the direction of the captured
antigen, thus perturbing the very system under observation. The
impact of these concerns is quantified in a side-by-side study of
506 target proteins [43].
Comparative results for the two labeling methods were cate-
gorized as identical, similar, or dissimilar among different subcellu-
lar locales. Identical results ranged from 15 to 70% among those
Making the Case for Functional Proteomics 33
7.6 Activity This segment primarily deals with the most prevalent protein activ-
ity, protein interactions—not simply binary protein-protein inter-
actions but the whole of protein interactions. In accord with the
analysis in this chapter, the dominant Functional Proteomics enti-
ties (83%) are bound proteins. Both Functional Proteomics and
Systems Biology demand assessment of protein interactions in
bio-relevant contexts. This is abundantly clear for translation into
essential arenas such as Biomarker and Drug Discovery, and Preci-
sion Medicine. Effectiveness in these arenas comprises the ability to
assess interactions among proteins (binary and groups), between
proteins and polynucleotides and between proteins and membrane
surfaces. Further, assessment must include the impact on those
interactions of variations in solution conditions, and the introduc-
tion of effector molecules such as cofactors, substrates or new drug
candidates. However, as two references attest, measurement even of
protein-protein interactions is the weakest link in the methodology
chain.
In 2002 a composite of existing data was analyzed [44]. High-
lights from that study concluded that only 3% of reported interac-
tions are supported by more than one method. That low “hit” ratio
continues in a 2009 study [45] in which multiple methods agreed
on only 8% of tested protein pairs—all of which were “known”
interacting pairs. Further, from the 2009 study the best method
test on known interacting pairs of proteins missed a full two-thirds
of the interactions—a false-negative value of 66%. Such outcomes
lead authors to conclude that, “. . . large datasets of protein-protein
interactions vary enormously in their error rates and there is no
34 Ray C. Perkins
7.7 Prioritized Table 2 assesses the overall knowledge base of the Functional
Roadmap Proteome as poor, and that Methods in most cases have limited
coverage potential. Any Roadmap must simultaneously focus on
accelerated data collection in areas of strength while improving
basic capabilities in areas of weakness. Areas of strength include
Making the Case for Functional Proteomics 35
the ability to Identify and Quantify proteins. This argues for aggres-
sive support for both updating reference materials to include
PTMs, etc., and widespread data collection. This work is founda-
tional and receives high priority. Structural determinations may
proceed in parallel along with the need for new methods develop-
ment to address know limitations. Localization methods must, for
now, be deemed developmental with Methods showing great
promise but lacking in robustness and reproducibility. Proteomic
Activity, as related to detection of protein interactions, is essentially
moribund. Wholly new approaches, especially approaches that per-
mit assessment in complex media, must be developed—now.
7.8 A Cautionary As was briefly mentioned at the beginning of this chapter segment,
Lesson on the Use of recombinant proteins (the major source of laboratory proteins) are
Recombinant Proteins modified in accord with the posttranslational product of the host
cell from which the protein is expressed. Bacterial cells will produce
bacterial PTMs, e.g., and human cells will produce human PTMs.
The degree and kind of PTMs are almost never specified or, as is
likely, known by the supplier. Therefore, almost the whole of pro-
tein research that employs recombinant proteins cannot be fully
specified. The content of those little vials opened thousands of
times a day is simply not known. Researchers must insist on full
protein specification, including PTMs (and protein content which
is typically only 50% of the total mass delivered). Finally, in-house
protein sequencing must become commonplace.
8 Closing Comments
References
1. Pundir S, Martin M, O’Donovan C (2017) 9. Weston AD, Hood L (2004) Systems biology,
UniProt protein knowledgebase. Methods proteomics, and the future of health care:
Mol Biol 1558:41–55. https://doi.org/10. toward predictive, preventative, and persona-
1007/978-1-4939-6783-4_2 lized medicine. J Proteome Res 3:179–196
2. Savage N (2015) Proteomics: high-protein 10. Butcher EC, Berg EL, Kunkel EJ (2004) Sys-
research. Nature 527:S6. https://doi.org/10. tems biology in drug discovery. Nat Biotechnol
1038/527S6a 22:1253–1259. https://doi.org/10.1038/
3. Pennisi E (2012) ENCODE project writes nbt1017
eulogy for junk DNA. Science 337 11. Perkins RC. Paul Kenis, Deborah Berthhold &
(6099):1159–1161. https://doi.org/10. Sarah-Ellen Leonard, University of Illinois,
1126/science.337.6099.1159 Urbana/Champaign; Jonathan Lee, recently
4. Ezkurdia I, Juan D, Rodriguez JM, Frankish A, of Eli Lily; and Ray Perkins, New Liberty
Diekhans M, Harrow J, Vazquez J, Valencia A, Proteomics
Tress ML (2014) Multiple evidence strands 12. Strimbu K, Tavel JA (2010) What are biomar-
suggest that there may be as few as 19 000 kers? Curr Opin HIV AIDS 5(6):463–466
human protein-coding genes. Hum Mol 13. Berger D (1999) A brief history of medical
Genet 23(22):5866–5878 diagnosis and the birth of the clinical labora-
5. Ponomarenko EA, Poverennaya EV, Ilgisonis tory. MLO Med Lab Obs 31(7). 28–30,
EV, Pyatnitskiy MA, Kopylov AT, Zgoda VG, 32, 34–40
Lisitsa AV, Archakov AI (2016) The size of the 14. FDA (2018.) In vitro diagnostics. https://
human proteome: the width and depth. Int J www.fda.gov/MedicalDevices/ProductsandM
Anal Chem 2016:7436849 edicalProcedures/InVitroDiagnostics/default.
6. Collins FS (2006) The language of god. Fran- htm
cis S. Collins on unveiling the human genome. 15. Kuruc M (2017) Stroma liquid biopsy—bio-
Free Press, New York, p 1–3 markers of the dysregulation of the serum pro-
7. Ball P (2010) Bursting the genomics bubble. teome in cancer First presented at NJ cancer
Nature. https://www.nature.com/news/2010/ Retreat, May 25, 2017 New Brunswick, NJ
100331/full/news.2010.145.html. https:// USA. https://www.biotechsupportgroup.com/
doi.org/10.1038/news.2010.145 v/vspfiles/templates/257/pdf/NJ%20Cancer
8. Gisler M (2010) The rise and fall of the human %20Retreat%20Stroma%20Liquid%20Biopsy%
genome project. MIT Technology Review 20Poster.pdf
Making the Case for Functional Proteomics 39
42. Yang Y, Ramelot TA, McCarrick RM, Ni S, 45. Braun P, Tasan M, Dreze M, Barrios-Rodiles-
Feldmann EA et al (2010) Combining NMR M, Lemmens I, Yu H, Sahalie JM, Murray RR,
and EPR methods for Homodimer protein Roncari L, A-Sd S, Venkatesan K, Rual J-F,
structure determination. J Am Chem Soc 132 Cusick ME, Pawson T, Hill DE, Tavernier J,
(34). https://doi.org/10.1021/ja105080h Wrana JL, Roth FP, Vidal M (2009) An experi-
43. Stadler C, Rexhepaj E, Singan VR, Murphy RF, mentally derived confidence score for binary
Pepperkok R, Uhlén M, Simpson JC, Lund- protein-protein interactions. Nat Methods 6
berg E (2013) Immunofluorescence and (1):91–97. https://doi.org/10.1038/nmeth.
fluorescent-protein tagging show high correla- 1281
tion for protein localization in mammalian 46. Rao VS, Srinivas K, Sujini GN, Kumar GNS
cells. Nat Methods 10:315–323. https://doi. (2014) Protein-protein interaction detection:
org/10.1038/nmeth.2377 methods and analysis. Int J Proteomics 2014:12.
44. von Mering C, Krause R, Snel B, Cornell M, https://doi.org/10.1155/2014/147648
Oliver SG, Fields S, Bork P (2002) Compara- 47. Klare J (2013) Site-directed spin labeling EPR
tive assessment of large-scale data sets of pro- spectroscopy in protein research. Biol Chem
tein–protein interactions. Nat Biotechnol 394(10):1281–1300. https://doi.org/10.
417:399–403 1515/hsz-2013-0155J
Chapter 2
Abstract
Conformational variants of the unique family of protease inhibitors annotated as SERPINs are most often
underrepresented in proteomic analyses. This limits understanding the complex regulation that this family
of proteins presents to the networks within the protease web of interactions. Using bead-based separation
provided by a family of proteomic enrichment products—notably AlbuVoid™ and AlbuSorb™, we
demonstrate their utility to satisfy investigations of serum SERPINs. We also suggest their use to develop
functional profiles of the SERPIN proteoforms, and how those can establish relationships to disease
phenotypes, gene mutations, and dysregulated mechanisms.
Key words SERPIN, SERPIN function, Functional proteomics, SERPIN mechanism, SERPIN
biomarkers, SERPIN proteoforms
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_2, © Springer Science+Business Media, LLC, part of Springer Nature 2019
41
42 Swapan Roy and Matthew Kuruc
1.1 The SERPIN The SERPIN family of suicidal serine protease inhibitors plays an
Superfamily of integral role in regulating a wide variety of biological activities, and
Suicidal Inhibitors represent 2–10% of circulating plasma proteins. SERPINs regulate
coagulation, hormone transport, complement and inflammation,
angiogenesis, and blood pressure along with many other pathways.
Among the key regulators in blood serum, SERPINA1 (also known
as ɑ1-antitrypsin) protects lung tissue from neutrophil elastase,
SERPINC1 (also known as antithrombin) controls coagulation
proteases, SERPING1 (also known as plasma C1 inhibitor) regu-
lates complement activation, and SERPINF2 (also known as ɑ-2-
antiplasmin) inhibits plasmin and regulates fibrinolysis [3, 4].
This unique family of protein inhibitors has been associated
with progression or remission of cancer and so they may become
valuable biomarkers for therapeutic or diagnostic use. Of clinical
utility, prostate-specific antigen (PSA), also known as kallikrein-3, is
commonly used as a biomarker for prostate cancer. However, the
kallikrein protease family of proteins is of very low abundance in
plasma, making observation and quantification difficult. Neverthe-
less, PSA is regulated by the SERPIN inhibitor family; in men with
prostate cancer the ratio of free (unbound) PSA to total PSA is
decreased, suggesting a greater role of inhibitory capacity in cancer.
By way of these examples, rather than focusing proteomic
discovery efforts on low-abundance proteins like the tissue kallik-
reins, it may be advantageous to profile much higher abundance
Tissue Kallikrein inhibitors like SERPINA5 (Protein C Inhibitor),
SERPINA3 (Antichymotrypsin), and SERPINA4 (Kallistatin), to
better understand underlying disease mechanisms and potentially
generate new biomarkers. However, the role of SERPINs in these
critical junctures is rarely straightforward as would be the case for
more simplistic binary binding inhibition. For functional interpre-
tation, reliance on strict abundance measurements, such as data that
might be derived by ELISA or quantitative LC-MS, does not
differentiate the subpopulations of the seemingly opposing out-
comes of the SERPIN interaction with its target protease.
This is because SERPINs differ from all other families of pro-
tease inhibitors in having a complex mechanism of action that
involves a drastic change in their shape, forming the basis of a
suicidal substrate inhibition mechanism [3, 4]. The reactive center
loop (RCL) extends out from the body of the protein and directs
binding to the target protease. The protease cleaves the SERPIN at
Methods to Monitor Functional SERPINs 43
6(53,1SURWHDVHLQKLELWRUVPXVWEHDFFRXQWHGIRU
GLIIHUHQWO\WKDQELQDU\ELQGHUV
%LQDU\ELQGLQJHYHQWSURWHDVHLQKLELWLRQLVUHJXODWHGE\UHODWLYH
FRQFHQWUDWLRQVRIWKHUHDFWDQWVದSURWHDVHDQGSURWHDVHLQKLELWRU
DQWLSURWHDVH
(TXLOLEULXP
6(53,1LQLWLDOLQWHUDFWLRQFDQSURGXFHWZR
IXQFWLRQDOO\RSSRVLQJRXWFRPHV
,QWHUPHGLDWH
&OHDYHG5&/
3URWHDVH3URWHDVH,QKLELWRU
3HUPDQHQWO\LQDFWLYH
FRPSOH[
6XLFLGDOWUDQVIRUPDWLRQRIWKHLQKLELWRUದ
5HDFWLYH&HQWHU/RRS FDQQRWEHUHJHQHUDWHGEDFNWRDFWLYHIRUP
5&/ UHJLRQZKHUH
WKHLPSRUWDQWELRORJ\
WDNHVSODFH
,UUHYHUVLEOHPRGLILFDWLRQDQG
VWDELOL]HGSURWHDVHFRPSOH[ERWK
SURWHDVHDQGLQKLELWRUDUH
SHUPDQHQWO\LQDFWLYH
Fig. 1 SERPIN protease inhibitors must be accounted for differently than binary binders
as the peptide RCL region is cleaved and can no longer bind target
substrates [4]. As a result, even minor changes in the structure due
to genetic variation and posttranslational modifications can modify
the function of SERPINs and give rise to a variety of clinical pre-
sentations. Some 200 different mutations in serpins are known to
result in disease [5]. In particular, mutations affecting antithrombin
confer a predisposition to thrombosis, those affecting C1 Inhibitor
confer a predisposition to angioedema, and those affecting anti-
plasmin confer a predisposition to hemorrhage. Interestingly, an
alternative function is made possible by a mutation in which the
methionine in the RCL region of Alpha-1-Antitrypsin is replaced
by an arginine converting its function as an inhibitor of neutrophil
elastase to a highly effective inhibitor of the coagulation proteases,
the consequence of which is life-threatening hemorrhagic
disease [6].
Mutations can affect function throughout the sequence. How-
ever, the most common loss of serpin function from mutation are
those affecting the mobile hinges of the molecule within or near the
RCL. These lead to spontaneous changes in conformation that
allow either the insertion of the intact reactive loop into the main
β sheet, resulting in the formation of an inactive “latent” form, or
the insertion of the loop of one molecule into the β sheet of the
next, resulting in the formation of polymers. Polymerization occurs
in Alpha-1-Antitrypsin with the common Z variant mutation, lead-
ing to decreased secretion from the liver into the circulation, result-
ing in emphysema and cirrhosis [7]. Amino acid substitution in the
RCL region is the likely event transforming the non-inhibitory
serpins. Posttranslational modifications at the RCL region such as
oxidation of methionine in Alpha-1-Antitrypsin have also been
proposed as a source of dysfunction [8].
So understanding the underlying mechanisms, contributions
from genetic wiring or environmental stresses, and their relation-
ships with aberrant proteolysis is necessary to characterize disease.
Functional proteomic analyses offer a new lens of observation to
examine the resulting conformational variants that can be reported
as potential biomarkers of disease phenotypes. As an example, one
such inhibitor SERPINA1, known more commonly as Alpha-1-
Antitrypsin (AAT), has several isoforms observed in plasma using
2-DE, and often serves as a model for conformational diseases
[5, 9]. Circulating levels of AAT are between 1.2 and 2 mg/mL
in healthy persons, but are known to increase during acute phases of
inflammation and infection. Its function and activity is controlled
by the many variants attributable to its conformational nexus of
features; the term “proteoform” is often used to describe such
conformational variability and we adopt that term here.
Other reports observe that the conformational properties of
AAT have multiple effects on tumor cell viability and diverse roles in
tumorigenesis, suggesting such isoforms may display a specific basis
Methods to Monitor Functional SERPINs 45
1.2 New Methods to By combining unique strategies of binding and voiding high-
Functionally Profile abundance proteins, we can observe different subpopulations with
SERPINs characteristic binding biases. We have previously reported for
Alpha-1-Antitrypsin that the resultant cleaved-RCL proteoform
and the uncleaved-RCL proteoform are very distinctive subpopula-
tions, separated by AlbuVoid™, and reported at the peptide fea-
ture level by LC-MS [12]. In this chapter, we consider how
Albumin Removal products—AlbuVoid™ and AlbuSorb™ (Bio-
tech Support Group LLC, Monmouth Junction, NJ, USA), can
help to functionally profile and unravel this complex biology of the
SERPIN superfamily of proteins.
Through a proprietary polymer coating, 50 μm porous silica
beads are crosslinked and passivated. This is the foundation of the
NuGel™ surface chemistry. Mixed-mode of binding interactions
form the basis of general nonspecific protein adsorbents or beads
with weak affinity or imperfect fit interactions. In this way, binding
behavior is very different from classical high affinity binding which
demands near perfect fits. Under protein saturation conditions,
progressive displacement provides a separation bias towards or
against select proteins. As a result, all derivative NuGel™ products
were empirically characterized to meet the needs of the application,
for example, AlbuVoid™ to selectively void (not bind) Albumin
with special bias toward the vast majority of the remaining
low-abundance serum proteome on the bead. Two NuGel™
based products support Albumin Removal:
1. AlbuSorb™ and AlbuSorb™ PLUS (also binds immunoglo-
bulins) for selective binding of Albumin.
46 Swapan Roy and Matthew Kuruc
1.5
1.
0.5
0.
0.5
AlbuSorb™
AlbuVoid™ bead flowthrough
Protein ID Also known as (conc.) Function bound S.Cts. (unbound) S.Cts. Reactive (RCL) bond site Notable variants
(continued)
47
48
Table 1
(continued)
AlbuSorb™
AlbuVoid™ bead flowthrough
Protein ID Also known as (conc.) Function bound S.Cts. (unbound) S.Cts. Reactive (RCL) bond site Notable variants
2 Materials
3 Methods
Serum
diluted
in
Binding
Buffer
added
Beads in to tube
Diluted serum
Spin-X
with beads
tube
Bead-based Protein Level Separation –
Analysis can be either or both bead-bound or flow-through
sub-populations
Bead-bound
proteome sub-
population
Trypsin Digest Options
- Elution from bead
In-gel
FASP
- On-Bead Digest
Flow-
through Serum
Bead bound (unbound) untreated
Cleaved RCL:
Reactive Center Loop
Permanently inactive
(RCL), region
4 Notes
l Bead-based proteomic enrichment methods as described can
support the functional and structural proteomic analyses neces-
sary to characterize these conformational subpopulations so that
they may become useful biomarkers for disease. It should none-
theless be recognized that the RCL reporting methods
described here only work for RCL regions where the cleavage
site is non-Tryptic, and these must be entered as special peptides
into LC-MS computational workflows. “neXtProt: a knowledge
platform for human proteins,” provides a useful web-based
resource for annotating RCL cleavage sites [13]. To distinguish
RCL regions where the cleavage site is Tryptic, it becomes
necessary to differentiate those sites that are cleaved in vivo by
those cleaved ex vivo. Several methods have been developed for
this purpose and generally fall under methods called
N-terminomics [14]. This is an area for future investigations.
l Classical high-abundance proteins like the SERPIN superfamily
(i.e., Alpha-1-Antitrypsin) are often overlooked as potential
biomarkers of disease. Yet discoveries certainly can rest in the
data-rich features of the diverse variety of conformational and
proteoform variants associated with many of the classical serum
proteins. When considering these mid- to high-abundance pro-
teins, disease differentiation can be obtained through the dis-
creet quantification of the multiple subpopulations available to
measure. The methods described in this chapter can begin to
unravel and sort these variant subpopulations so that LC-MS
peptide reporting features, and potentially other functional
reporting features (i.e., substrate turnover), can distinguish
these proteoforms with more functional details. It is our inten-
tion that these methods will lead to characteristic disease pro-
files, which can then be compared and evaluated for eventual
biomarker utility.
l Many trypsin digestion protocols have been developed to
improve the reproducibility and, in some cases, reduce the
digestion time necessary for LC-MS analysis [15]. While we
have shown methods that adapt AlbuVoid™ for on-bead diges-
tion, the bead-based enrichments described here are nonetheless
compatible, after elution from the beads, with other common
digestion methods, such as filter-aided (FASP) solution methods
and post-electrophoresis, in-gel methods.
l By using the peptide reporting features of the RCL peptide
regions within SERPIN inhibitors, both “potentially active”
and “permanently inactive” proteoforms are now distinguish-
able, adding a new level of proteomic characterization to the
underlying mechanisms of disease. As one example, hereditary
54 Swapan Roy and Matthew Kuruc
References
1. Fortelny N, Cox JH, Kappelhoff R, Starr AE from patients during an acute coronary syn-
et al (2014) Network analyses reveal pervasive drome. J Am Coll Cardiol 44(8):1578–1583
functional regulation between proteases in the 10. Wang Y, Kuramitsu Y, Yoshino S et al (2011)
human protease web. PLoS Biol 12(5): Screening for serological biomarkers of pancre-
e1001869 atic cancer by two-dimensional electrophoresis
2. Shamamian P, Schwartz JD, Pocock BJ et al and liquid chromatography-tandem mass spec-
(2001) Activation of progelatinase A trometry. Oncol Rep 26(1):287–292
(MMP-2) by neutrophil elastase, cathepsin G, 11. Zelvyte I, Sjögren HO, Janciauskiene S (2002)
and proteinase-3: a role for inflammatory cells Effects of native and cleaved forms of α1-anti-
in tumor invasion and angiogenesis. J Cell trypsin on ME 1477 tumor cell functional
Physiol 189(2):197–206 activity. Cancer Detect Prev 26(4):256–265
3. Law RH, Zhang Q, McGowan S et al (2006) 12. Zheng H, Zhao C, Roy S et al (2016) The
An overview of the serpin superfamily. Genome commonality of the cancer serum proteome
Biol 7(5):216 phenotype as analyzed by LC-MS/MS, and its
4. Khan MS, Singh P, Azhar A et al (2011) Serpin application to monitor dysregulated wellness.
inhibition mechanism: a delicate balance Poster presented at the AACR annual meeting
between native metastable state and polymeri- 2016 conference, New Orleans, LA, USA,
zation. J Amino Acids. https://doi.org/10. April 17–20 2016
4061/2011/606797 13. Lane L, Argoud-Puy G, Britan A et al (2011)
5. Carrell RW, Lomas DA (2002) Alpha1- neXtProt: a knowledge platform for human
antitrypsin deficiency—a model for conforma- proteins. Nucleic Acids Res 40(D1):D76–D83
tional diseases. N Engl J Med 346(1):45–53 14. Lai ZW, Petrera A, Schilling O (2015) Protein
6. Owen MC, Brennan SO, Lewis JH et al (1983) amino-terminal modifications and proteomic
Mutation of antitrypsin to antithrombin: α1- approaches for N-terminal profiling. Curr
antitrypsin Pittsburgh (358 Met!Arg), a fatal Opin Chem Biol 24:71–79
bleeding disorder. N Engl J Med 309 15. Zheng H, Zhao C, Qian M et al (2015) Albu-
(12):694–698 Void™ coupled to on-bead digestion-tackling
7. Sifers RN (1992) Z and the insoluble answer. the challenges of serum proteomics. J Proteom
Nature 357(6379):541 Bioinformatics 8(9):225
8. Janciauskiene S (2001) Conformational prop- 16. Sun Z, Yang P (2004) Role of imbalance
erties of serine proteinase inhibitors (serpins) between neutrophil elastase and α1-antitrypsin
confer multiple pathophysiological roles. Bio- in cancer development and progression. Lancet
chim Biophys Acta 1535(3):221–235 Oncol 5(3):182–190
9. Mateos-Cáceres PJ, Garcı́a-Méndez A, Farré
AL et al (2004) Proteomic analysis of plasma
Chapter 3
Abstract
The substitution of the reverse polarity benzyldimethyl-n-hexadecylammonium chloride (16-BAC) poly-
acrylamide gel electrophoresis (PAGE) for isoelectric focusing (IEF) in the first dimension of electrophore-
sis improves the solubility of extremely hydrophobic proteins and their recovery compared to conventional
2D IEF/SDS PAGE. The acidic environment of 16-BAC PAGE has also been shown to better preserve the
labile methylation of basic proteins such as the histones. Several improvements of the 2D 16-BAC/SDS
PAGE method are collectively described here with particular emphasis on the separation of mitochondrial
membrane proteins of low molecular mass. Lowering the 16-BAC concentration 50-fold in the gel and
buffers decreases the formation of mixed 16-BAC/SDS micelles, which otherwise interferes with the
separation of very low molecular mass proteins in second dimension SDS PAGE, and consequently
improved the resolution of mitochondrial membrane proteins in the 10–30 kDa range.
Abbreviations
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_3, © Springer Science+Business Media, LLC, part of Springer Nature 2019
55
56 Gary Smejkal and Srikanth Kakumanu
1 Introduction
2 Materials
3. Chloroform.
4. Methanol.
2.2 First Dimension Buffers and concentrated buffer stocks can be prepared in advance
16-BAC PAGE without the addition of 16-BAC. All buffers are filter sterilized
(e.g., Millipore Steriflip or similar) and can be stored up to
3 months at 4 C. The 16-BAC detergent is added to working
solutions immediately before use and used the same day.
1. Concentrated (4) resolving gel buffer: 300 mM KH2PO4
pH 2.1 (see Note 2).
2. Concentrated (4) stacking gel buffer: 500 mM KH2PO4
pH 4.1 (see Note 3).
3. 125 mM 16-BAC stock solution (see Note 4).
4. Concentrated (10) electrode buffer: 1.5 M glycine, 500 mM
ortho-phosphoric acid (see Note 5).
5. Working (1) electrode buffer: 150 mM glycine, 50 mM
ortho-phosphoric acid, 0.05 mM 16-BAC (see Note 6).
6. 9 M urea (see Note 7).
7. AG 501-X8 mixed bed ion exchange resin (see Notes 7 and 9).
8. Bond-Breaker™ 500 mM Tris (2-carboxyethyl) phosphine
(TCEP) solution, neutral pH (Thermo-Fisher Scientific,
77720).
9. 50% glycerol.
10. Concentrated (100) Pyronin Y tracking dye: 1 mg/mL Pyr-
onin Y in water.
11. 1 sample buffer: 4 M urea, 50 mM 16-BAC, 10 mM TCEP,
10.5% glycerol, 0.005% Pyronin Y (see Note 8).
12. 29.2% acrylamide, 0.8% methylene bisacrylamide solution (see
Note 9).
13. 80 mM ascorbic acid (see Note 10).
14. 5 mM ferrous sulfate (see Note 11).
15. 30% hydrogen peroxide.
16. 25% isopropanol.
17. Reflection™ Dual Vertical Electrophoresis System (Galileo
Biosciences, 85-1614).
18. Precision Plus Protein™ Standards (Biorad, 1610374).
19. Stainless steel tissue slicing blade, 22 cm length.
60 Gary Smejkal and Srikanth Kakumanu
3 Methods
3.1 Isolation of Mitochondria were isolated as described by Lee et al. [21]. Porcine
Mitochondrial heart was resected within minutes of euthanasia and placed imme-
Membrane Proteins diately on wet ice. The tissue was dissected within 1 h of collection.
All steps were performed at 4 C.
1. Remove connective tissue and fat and course grind the myo-
cardium in a food grinder.
2. Suspend the macerate in five volumes of MIB.
3. Blend the suspension in a food processor for three times 30 s.
4. Centrifuge the suspension at 650 RCF for 10 min and filter the
supernatant through multiple layers of cheese cloth.
5. Resuspend the remaining pellet in additional MIB and repeat
steps 3 and 4. Combine the two supernatants.
6. Centrifuge the pooled supernatants and centrifuge at 14,000
RCF for 20 min.
7. Homogenize the resulting pellet in a ground glass homoge-
nizer with Teflon plunger.
8. Centrifuge at 400 RCF for 8 min to pellet cellular debris.
Transfer the supernatant to a new tube and centrifuge for
14,000 RCF for 20 min.
Two-Dimensional 16-BAC/SDS Polyacrylamide Gel Electrophoresis. . . 61
3.2 16-BAC PAGE The Fenton reaction as modified by MacFarlane [13] is used to
catalyze the polymerization reaction. Generally, 16-BAC gels are
run on the same day they are cast. If necessary, the resolving gel can
be cast on the first day, then overlaid with 1 resolving buffer and
stored overnight at room temperature. The stacking gel should be
cast within a few hours of running the gel on the second day. All
steps are performed at room temperature.
1. Assemble two 16 14 cm glass plates and two 0.8 mm thick
spacers using the alignment pins of the upper buffer chamber
and clamp securely. The entire assembly is transferred to the
casting stand and cammed into place for leakproof casting
(Fig. 3).
2. Prepare the resolving gel as prescribed in Table 1. Combine all
components except for the catalysts in a 50 mL screw cap
centrifuge tube. Cap securely and mix by gentle inversion
taking care not to introduce bubbles.
3. Add sequentially the absorbic acid, FeSO4, and H2O2. Cap
securely and mix by gentle inversion following each addition.
4. Immediately pour the polymerization mixture into the gel
cassette to within 3 cm of the top of the notched glass plate.
Carefully overlay the gel with 400–600 μL of 25% isopropanol
(see Note 15).
5. Allow the gel to polymerize for 15–20 min (see Note 16).
62 Gary Smejkal and Srikanth Kakumanu
Fig. 3 Exploded view of the Galileo Biosciences large format electrophoresis system and leakproof gel casting
system. Glass plates and spacers are clamped to the upper buffer chamber during casting and are not
removed until electrophoresis is complete. This avoids the “flexing” of gels which can result in disadherence
of the gel from the glass surface and the formation of bubbles between the gel and glass
6. Decant the isopropyl overlay and wash the gel surface twice
with water. (If storing the gel overnight, overlay with 1X
resolving buffer.)
7. Prepare the stacking gel as prescribed in Table 2. Combine all
components except for the catalysts, cap securely, and mix by
gentle inversions. Add sequentially the absorbic acid, FeSO4,
and H2O2. Cap securely and mix by gentle inversion following
each addition.
8. Fill the remaining space in the gel cassette with stacking gel and
insert the 0.8 mm thick comb taking care not to trap air
bubbles. Allow to polymerize for at least 1 h.
9. Remove the comb and forcefully flush any unpolymerized
solution from the wells with water using a transfer pipette.
Two-Dimensional 16-BAC/SDS Polyacrylamide Gel Electrophoresis. . . 63
Table 1
16-BAC PAGE resolving gel composition
Table 2
16-BAC PAGE stacking gel composition
10. Fill the upper and lower buffer chambers with electrode buffer.
Flush the water from the wells with electrode buffer using a
transfer pipette.
3.3 Sample 16-BAC is dissolved in the sample buffer on the same day of the
Preparation analysis. Protein samples are prepared immediately before electro-
phoresis. Hartinger et al. [17] observed protein degradation and a
loss of resolution when samples were stored in the 16-BAC sample
buffer.
64 Gary Smejkal and Srikanth Kakumanu
3.5 Second 1. Flush the surface of the second dimension gel with water to
Dimension SDS PAGE remove residual storage buffer. Fill the upper buffer chamber
with SDS PAGE electrode buffer.
2. Soak a 100 20 mm filter paper in SDS PAGE electrode
buffer.
3. With a spatula, position the equilibrated gel strip on top of the
second dimensional gel. Place the saturated filter paper on top
of the equilibrated gel strip and gently press down to keep the
gel strip in intimate contact with the second dimension gel
(Fig. 4).
4. Connect to the power supply in normal polarity. Commence
electrophoresis at 40 mA constant current for 10 min. Turn the
power off and remove the filter papers. Continue electropho-
resis until the phenol red dye front has migrated to within a few
millimeters of the bottom of the gel.
3.6 KUMASI Staining 1. Fix the gel for at least 1 h in 100 mL of 30% ethanol, 10% acetic
acid (see Note 18).
Two-Dimensional 16-BAC/SDS Polyacrylamide Gel Electrophoresis. . . 65
Fig. 4 Second dimension SDS PAGE of excised lane from first dimension 16-BAC PAGE. SDS equilibrated gel
strips were held in intimate contact with the second dimension gel with a buffer-saturated filter paper wick
(arrow). This eliminates the need for an agarose gel overlay
4 Notes
15. The gel can be “misted” with 25% isopropanol from a spray
bottle.
16. Gel polymerization rates are affected by temperature and rela-
tive humidity. If the gel polymerizes in less than 10 min, discard
and prepare a new gel using 10% less of each catalyst.
17. Avoid heating the sample higher than 60 C. Asp-Pro linkages
are susceptible to hydrolysis at acidic pH and elevated
temperature.
18. For improved staining, fix overnight to completely remove
SDS from the gel.
19. The fixative and KUMASI staining solutions can be reused at
least two times.
20. Gels stored in enhancer solution with sodium azide added are
stable for years. We have observed improved contrast with no
significant loss of sensitivity in gels stored for 9 years.
References
1. Klose J, Kobalz U (1995) Two-dimensional preparation in biological mass spectrometry.
electrophoresis of proteins: an updated proto- Springer, Heidelberg, pp 411–434
col and implications for a functional analysis of 8. Rath A, Glibowicka M, Nadeau VG, Chen G,
the genome. Electrophoresis 16:1034–1059 Deber CM (2009) Detergent binding explains
2. Klose J, Nock C, Herrmann M, Stühler K, anomalous SDS-PAGE migration of mem-
Marcus K, Blüggel M, Krause E, Schalkwyk brane proteins. PNAS 106:1760–1765
LC, Rastan S, Brown SDM, Büssow K, 9. Smejkal GB, Bauer DJ (2012) High speed iso-
Himmelbauer H, Lehrach H (2002) Genetic electric focusing of proteins enabling rapid
analysis of the mouse brain proteome. Nat two-dimensional gel electrophoresis. Gel elec-
Genet 30:385–393 trophoresis: principles and basics. Intech,
3. Hamdan M, Righetti PG (2005) Proteomics Rijeka, pp 157–170
today: Protein assessment and biomarkers 10. Meisrimler CN, Lüthje S (2012) IPG-strips
using mass spectrometry, 2D electrophoresis, versus off-gel fractionation: advantages and
and microarray technology. Wiley & Sons, limits of two-dimensional PAGE in separation
Hoboken, NJ, pp 219–265 of microsomal fractions of frequently used
4. Klein C, Garcia-Rizo C, Bisle B, Scheffer B, plant species and tissues. J Proteome
Zischka H, Pfeiffer F, Siedler F, Oesterhelt D 75:2550–2562
(2005) The membrane proteome of Halobac- 11. Rabilloud T (2010) Variations on a theme:
terium salinarum. Proteomics 5:180–197 changes to electrophoretic separations that
5. Kalinowski J, Wolters D, Poetsch A (2008) can make a difference. J Proteome
Proteomics of Corynebacterium glutamicum 73:1562–1572
and other Corynebacteria. From Corynebac- 12. Miller M, Ivano Eberini I, Gianazza E (2010)
teria: genomics and molecular biology Other than IPG-DALT: 2-DE variants. Prote-
(Burkovski A, ed). Caister Academic Press, omics 10:586–610
Norfolk, pp 56–77 13. Macfarlane DE (1983) Use of benzyldimethyl-
6. Aivaliotis M, Corvey C, Tsirogianni I, Karas M, n-hexadecylammonium chloride (16-BAC), a
Tsiotis G (2004) Membrane proteome analysis cationic detergent, in an acidic polyacrylamide
of the green-sulfur bacterium Chlorobium gel electrophoresis system to detect base labile
tepidum. Electrophoresis 25:3468–3474 protein methylation in intact cells. Anal Bio-
7. Moller AJB, Witzel K, Vertommen A, chem 132:231–235
Barkholdt V, Svensson B, Carpentier S Mock 14. Macfarlane DE (1984) Inhibitors of cyclic
HP, Finne C (2011) Plant membrane proteo- nucleotide phosphodiesterases inhibit protein
mics: challenges and possibilities. Sample carboxyl methylation in intact blood platelets.
J Biol Chem 259:1357–1362
68 Gary Smejkal and Srikanth Kakumanu
15. Macfarlane DE (1986) Phorbol diester- 19. Zahedi RP, Moebius J, Sickmann A (2007)
induced phosphorylation of nuclear matrix Two-dimensional BAC/SDS-PAGE for mem-
proteins in HL60 promyelocytes. Possible role brane proteins. In: Bertrand E, Faupel M (eds)
in differentiation studied by cationic detergent Subcellular proteomics: from cell deconstruc-
gel electrophoresis systems. J Biol Chem tion to system reconstruction. Springer, Dor-
261:6947–6953 drecht, pp 13–20
16. Macfarlane DE (1989) Two dimensional 20. Kramer ML (2006) A new multiphasic buffer
benzyldimethyl-n-hexadecylammonium chlo- system for benzyldimethyl-n-hexadecylammo-
ride sodium dodecyl sulfate preparative poly- nium chloride polyacrylamide gel electropho-
acrylamide gel electrophoresis: a high capacity resis of proteins providing efficient stacking.
high resolution technique for the purification Electrophoresis 27:347–356
of proteins from complex mixtures. Anal Bio- 21. Lee I, Salomon AR, Yu K, Samavati L, Pecina P,
chem 176:457–463 Pecinova A, Huttemann M (2009) Isolation of
17. Hartinger J, Stenius K, Högemann D, Jahn R regulatory-competent, phosphorylated cyto-
(1996) 16-BAC/SDS-PAGE: a chrome c oxidase. Methods Enzymol
two-dimensional gel electrophoresis system 457:193–210
suitable for the separation of integral mem- 22. James GT (1978) Inactivation of the protease
brane proteins. Anal Biochem 240:126–133 inhibitor phenylmethylsulfonyl fluoride in buf-
18. Zahedi RP, Meisinger C, Sickmann A (2005) fers. Anal Biochem 86:574–579
Two-dimensional benzyldimethyl-nhexadecy- 23. Singh R, Whitesides GM (1995) Reagents for
lammonium chloride/SDS-PAGE for mem- raid reduction of disulfide bonds in proteins.
brane proteomics. Proteomics 2005 Techniq Protein Chem VI:259–266
(5):3581–3588
Chapter 4
Abstract
A functional proteomics technology was used to systematically monitor metabolic enzyme activities from
resolved serum proteins produced by a modified 2-D gel separation and subsequent Protein Elution Plate, a
method collectively called PEP. Both qualitative and quantitative differences in the metabolic enzyme
activity could be detected between cancer patient and control group, providing excellent biomarker
candidates for cancer diagnosis and drug development. This technology has a wide range of applications;
it can be used for rapid functional protein purification and characterization as well as drug target identifica-
tion and validation. The ability for the PEP technology to efficiently separate and recover functional
proteins makes it useful for the analysis of any proteins and its variants; this is especially advantageous for
enzyme families with large number of enzymes such as protein kinases, phosphatases, proteases, and
metabolic enzymes.
Key words Functional proteomics, 2-D gel electrophoresis, Protein purification, Biomarkers, Protein
elution plate (PEP), Cancer diagnosis, Drug target identification
1 Introduction
In the last decade, many new technologies have been utilized for
biomarker discovery with significant progress. Each of these tech-
nologies has focused on a different type of biological entity such as
circulating tumor cells (CTC), extracellular vesicles, micro-RNAs
and cancer-derived cell-free DNA or circulating tumor-derived
DNA (ctDNA) [1–9]. However, several fundamental issues such
as tumor heterogeneity, plasticity, and diversity of cancer stem cells
(CSC) make biomarker discovery and development a challenging
endeavor. The variation introduced during sample collection and
storage and the lack of robust validation approach once biomarker
leads are identified further complicate biomarker development
[10–19]. As a result of these hurdles, there are currently no United
States FDA-approved serum tests for early detection of the disease.
Given the considerable public health importance of breast cancer, it
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_4, © Springer Science+Business Media, LLC, part of Springer Nature 2019
69
70 David Wang
2 Materials
2.1 Chemicals All the chemicals were purchased from MilliporeSigma (St. Louis,
MO). Isoelectric focusing (IEF) unit that is capable of running IEF
at different length is from Bio-Rad (PROTEAN IEF Cell, Hercu-
les, CA). Spectrophotometer Plate Reader capable of reading
384-well plates with a wide wavelength selection and fluorescence
reading is the SPECTRAMax Plus from Molecular Devices (Sunny-
dale, CA). Semi-Blot unit for protein transfer was Bio-Rad’s Trans-
Blot SD Semi-Dry Transfer Cell. AlbuVoid™ serum protein enrich-
ment beads were from Biotech Support Group (Monmouth Junc-
tion, NJ). Protein Elution Plate (PEP) is a product of Array Bridge
(St. Louis, MO).
1. SDS-PAGE gels: Customer can choose any format SDS-PAGE
gel to run the sample. For 1-D gel, preferentially the loading
72 David Wang
Fig. 1 Diagram of the PEP Technology (adapted from Wang DL et al., PLoS One, 2015, 10(3) with permission)
2.2 Glycolytic The glycolytic enzymes from human serum were detected by mea-
Enzyme Activity Assay suring the first enzyme in the glycolytic pathway, hexokinase. By
using a beef liver extract to provide a low and basal level of glyco-
lytic enzymes, any additional enzymes from the PEP-eluted serum
sample can be detected by the increased hexokinase activity. There-
fore the measurement of the glycolytic enzyme activity from PEP
samples was calculated by the increased hexokinase activity from the
basal level of beef liver extract instead of the total hexokinase
activity.
Hexokinase activity can be monitored by a cascade reaction as
follows:
Hexokinase
Fig. 2 Measurement of hexokinase activity from normal and breast cancer serum (adapted from Wang DL
et al., BMC Biomarker Research, 2017, 5(11) with permission)
2.3 Supplied 1. 384-well PEP plate. A PEP plate is provided. The plate was
Components from the treated with a special solution to reduce the binding of the
PEP Universal Protein transferred protein and increase the recovery efficiency.
Purification Kit (Small 2. 384-well mater plate. A deep-well plate is provided to contain
Format PEP) from samples recovered from the PEP plate.
Array Bridge Inc. 3. 384-well enzyme assay plate. A standard 384-well polypropylene
(Catalog Number: plate is provided for enzyme assay to identify which wells
AB-000401) contain the protein of interest.
4. 10 protein transfer buffer (50 mL). Buffer used for running
the modified SDS-PAGE or the second dimension of 2-D gel,
Systematic Glycolytic Enzyme Activity Analysis from Human Serum with PEP. . . 75
and also used to wet the filter papers for the transfer of proteins
from the gel to the PEP plate.
5. 10 PBS (10 mL). Buffer used for the pretreatment of the
master plate and fill in each well of the Master Plate with
50 μL of PBS.
6. Standard SDS-PAGE sample buffer (0.5 mL). Solution used in
sample treatment for the standard SDS-PAGE to check enzyme
fraction purity.
7. PEP plate protein recovery buffer (25 mL). Solution used in the
PEP plate to recover proteins eluted from the gel and prevent
protein diffusion.
8. Plate sealer. For sealing the Master plate and the enzyme assay
plate during the purification process. Kit AB-00402 (two).
9. Filter papers. Used to form a sandwich in the protein transfer
process.
2.4 Instruments Gel electrophoresis unit includes power supply and gel unit.
Isoelectric focusing unit that is capable of running IEF at different
length, an example of such unit is the Bio-Rad PROTEAN IEF
Cell (Catalog Number: 165-4000).
Spectrophotometer Plate Reader capable of reading 384-well plates
with a wide wavelength selection and fluorescence reading.
Semi-Blot unit for protein transfer such as Bio-Rad’s Trans-Blot SD
Semi-Dry Transfer Cell (Catalog Number: 170-3940).
3 Methods
3.1 Sample High concentration of salt will interfere with the isoelectric focus-
Treatment ing step. If the protein concentration is less than 5 mg/mL and the
salt concentration is more than 100 mM, it is recommended to
dialyze the samples in 5 mM phosphate buffer, pH 7.2 before use or
using desalting column to reduce the salt concentration.
3.2 Running the IEF 1. It is suggested to use the 11 cm IPG strip (Bio-Rad, catalog
Gel First Followed by number: 163-2033) for the IEF. To rehydrate one IPG strip,
Running the Native or 225 μL of solution is needed. It is suggested to use 200 μL of
Modified SDS-PAGE samples with up to 200 μg total protein, add urea to a final
concentration of 8 M, add 2 μL of Ampholyte such as Bio-lyte
(Bio-Rad, catalog number: 163-2094). If the protein sample
can be lyophilized, then the lyophilized sample can be dissolved
into a sample solution with 8 M urea and 0.5% Bio-lyte.
2. The solution is first added to a rehydration tray, the IPG strip is
taken out from the storage, and the plastic cover is peeled off.
The side with the dried gel surface is facing down to make
76 David Wang
3.3 Protein Transfer 1. While the SDS-PAGE is still running, put the PEP plate in a
After the 2-D Gel tray and add 50 μL of the protein recovery solution to each well
of the plate with a multiple channel pipette, there will be some
overflow of the solution during this step, it is fine. If an eight
channel pipette is used, the solution could be dispensed every
other row. For example, in the first round, add solutions to
row A, C, E, and so on; in the second round, add solution in
the row B, D, F, and so on. Cover the tray to minimize
evaporation.
2. When the dye front is about 0.5 cm from the bottom of the gel,
stop the running and carefully take out the gel from the gel
cassette and rinse with Milli-Q water followed by adding
200 mL of the transfer buffer (supplied with the kit) in the
tray. Wet four pieces of the transfer filter paper (supplied with
the kit) completely in a different tray and lay down two pieces
on the metal plate of the Semi-Dry Trans-Blot (Bio-Rad or
similar Semi-Dry Trans-Blot from other manufacturers).
3. Lay the PEP plate on top of the filter papers followed by
carefully lay the gel on top of the PEP plate and make sure
the upper left corner of the gel align with the upper left corner
of the PEP plate.
4. Lay another two pieces of transfer filter paper on top of the gel
to form a sandwich (from the bottom it should be filter papers,
PEP plate, gel and filter papers again).
5. Cover the sandwich assembly with the other metal plate of the
Semi-Dry Trans-Blot, and transfer the proteins with constant
voltage at 20 voltages for 60 min. It is shown that under this
condition, the proteins in the gel will be efficiently transferred
into the PEP plate, longer protein transfer is not
recommended.
6. While the gel is transferring, condition the 384-well deep-well
plate by adding 100 μL PBS in each well (if protein kinase or
protein phosphatase assays are performed, a phosphate-free
buffer such as Tris–HCl should be used to minimize interfer-
ence from the buffer). This treatment will improve the protein
recovery in later steps for enzyme activity analysis and mass
spectrometry protein identification. After 30 min treatment,
completely empty the solution from each well and refill the well
with 50 μL PBS (for protein kinase assay or any other assay
where phosphate is interfering, Tris–HCl buffer or other buffer
of choice could be used).
7. When the protein transfer is completed, turn off the power,
take off the Semi-Dry Trans-Blot cover and release the top
metal plate. Wait for 10 s before lifting the top metal plate
(this is important to let some air in so that the solutions in the
PEP plate will not be sucked out to cause proteins in one well
78 David Wang
3.4 Glycolytic 1. After the transfer of samples from the PEP plate to the deep-
Enzyme Analysis well Master Plate, the Master Plate should be used immediately
(preferred) for glycolytic enzyme analysis. Multiple enzymes
can be analyzed from samples collected since the total volume
in each well of the Master Plate is about 90 μL (50 μL buffer
plus 40–45 μL sample transferred from the PEP plate). Refer to
Subheading 2.2 for details of the glycolytic enzyme assay pro-
tocol. Use a spectrophotometer to measure the glycolytic
enzyme activities at 340 nm. Before the assay readout, some
wells of the enzyme assay plate may contain bubbles because of
the SDS in the protein transfer buffer (one technique to avoid
bubble is to set the dispensing volume smaller than the aspirat-
ing volume so that the pipette will not cause bubbles when
dispensing). Use a transfer pipette tip to pinch the bubbles to
get rid of them before the reading; this will reduce the interfer-
ence from the bubble.
2. When reading the enzyme assay plate, use pipette to remove
the solutions from the well P24 (lower right corner well in the
384-well plate) and use this well as blank for the reading. It is
recommended to read at least 3 data points such as 0, 60, and
120 min and save the reading in separate files.
3.5 Data 1. Export the data set from the three readings (0, 60, and
Transformation and 120 min) to an Excel file (if not already in this format).
Analysis 2. In Microsoft Excel, subtract the 60 min readings from each
well from the corresponding readings at 0 min to obtain the
Systematic Glycolytic Enzyme Activity Analysis from Human Serum with PEP. . . 79
data set for the 340 nm absorbance difference which reflect the
glycolytic enzyme activities from the serum proteome. Use
Excel Heat Map to display the enzyme activity in a 384-well
table or use Insert function, and select the 3-D display to build
the graph of this data set in 3-D display.
3. Subtract the 120 min readings from each well from the
corresponding readings at 0 min to obtain the data set for the
340 nm absorbance difference which reflect the glycolytic
enzyme activities from the serum proteome for the second
data set.
3.6 Protein Purity 1. If the enzyme testing showed that some wells have the enzyme
Confirmation activity of interest, the next step is to test the purity of the
(Optional) protein in that well. Collect all the samples from the wells with
enzyme activity in a siliconized microcentrifuge tube, dry down
the solution, and resuspend into 20 μL of Milli-Q water. Take
10 μL and mixed with 10 μL of SDS sample buffer (this sample
buffer is a 2 SDS-PAGE sample buffer with 20 mM DTT),
incubate at 37 C for 60 min.
2. Load on a SDS-PAGE gel and run the gel as in Subheading 2 of
this protocol.
3. Fix the gel in a gel-fixing solution (10% each of ethanol and
acetic acid in Milli-Q water) for at least 2 h.
4. Rinse with distilled water and stain the gel in Sypro Ruby or
other fluorescence dye overnight.
5. The next day, remove the staining solution; wash the gel twice
with distilled water followed by incubation in the distilled
water for 5 min with moderate shaking.
6. Take the gel image with a CCD camera such as the Bio-Rad
ChemiDoc.
7. Save the image in tiff file for later image processing. The gel
image will tell whether the protein is pure or not.
3.7 Mass 1. If the gel staining in Subheading 3.6 shows that the fraction
Spectrometry to with enzyme activity is pure, the 10 μL Milli-Q water resus-
Identify the Protein of pended sample in Subheading 3.6, step 1 can be submitted for
Interest mass spectrometry analysis (sometimes fraction with more than
one protein bands can be submitted for MS analysis, and the
identity of the protein can be assigned by bioinformatics effort
based on protein homology, it is unlikely that more than one
protein from the preparation share the same type of enzyme
activity, for example, GAPDH).
2. Alternatively, if there is enough protein to be seen in Subhead-
ing 3.6, step 7 with the fluorescence staining, the protein band
can be excised and sent for MS analysis.
80 David Wang
4 Notes
Acknowledgments
I would like to thank Array Bridge Inc. for the supply of the PEP
Universal Protein Purification kits and the opportunity to carry out
this research in its laboratory. I would also like to thank Dr. Liang Li
for providing the breast cancer and normal people sera in this
research.
References
1. Dos Anjos Pultz B et al (2014) Far beyond the 5. Henderson MC et al (2016) Integration of
usual biomarkers in breast cancer: a review. J serum protein biomarker and tumor asso-
Cancer 5(7):13 ciated autoantibody expression data
2. Li J et al (2002) Proteomics and bioinformatics increases the ability of a blood-based proteo-
approaches for identification of serum biomar- mic assay to identify breast cancer. PLoS
kers to detect breast cancer. Clin Chem 48(8):9 One:11(8)
3. Chan MK, Cooper JD, Bahn S (2015) Com- 6. Ingvarsson J et al (2007) Design of recombi-
mercialisation of biomarker tests for mental ill- nant antibody microarrays for serum protein
nesses: advances and obstacles. Trends profiling: targeting of complement proteins. J
Biotechnol 33(12):12 Proteome Res 6:10
4. Chung L et al (2014) Novel serum protein 7. Lee JS, Magbanua MJM, Park JW (2016) Cir-
biomarker panel revealed by mass spectrometry culating tumor cells in breast cancer: applica-
and its prognostic value in breast cancer. Breast tions in personalized medicine. Breast Cancer
Cancer Res 16:R63 Res Treat 160:411–424
Systematic Glycolytic Enzyme Activity Analysis from Human Serum with PEP. . . 81
Abstract
Snake venoms are complex mixtures of proteins and peptides that play vital roles in the survival of venomous
snakes. As with their diverse pharmacological activities, snake venoms can be highly variable, hence the
importance of understanding the compositional details of different snake venoms. However, profiling
venom protein mixtures is challenging, in particular when dealing with the diversity of protein subtypes
and their abundances. Here we described an optimized strategy combining a protein decomplexation
method with in-solution trypsin digestion and mass spectrometry of snake venom proteins. The approach
involves the integrated use of C18 reverse-phase high-performance liquid chromatography (RP-HPLC),
sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), and nano-electrospray ionization
tandem mass spectrometry (nano-ESI-LC-MS/MS).
Key words Snake venom, Protein decomplexation, Venom separation, Reverse-phase high-
performance liquid chromatography, Tandem mass spectrometry, Venomics
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_5, © Springer Science+Business Media, LLC, part of Springer Nature 2019
83
84 Choo Hock Tan et al.
2 Materials
2.1 Snake Venom Freeze-dry snake venom samples and store at 20 C until use.
Samples
2.1.3 Protein Digestion Trypsin stock (0.1 μg/μL): Add 200 μL of ddH2O to 20 μg lyophi-
(In-Solution Tryptic lized trypsin with 1 mM HCl.
Digestion) Digestion buffer: 50 mM ammonium bicarbonate.
Reducing buffer: 100 mM dithiothreitol (DTT).
Alkylation buffer: 100 mM iodoacetamide (IAA).
Solution:
Wetting solution: 50% acetonitrile (ACN).
Equilibrium/wash solution: 0.1% formic acid (FA).
3 Methods
Fig. 1 Reverse-phase HPLC fractionation of snake venom using LiChrospher® WP 300 C18 column (upper
panel) following the chromatographic condition: 5% B for 10 min, 5–15% B for 20 min, followed by 15–45% B
for 120 min and 45–70% B for 20 min. The chromatographic fractions are collected manually at 215 nm
absorbance and the lyophilized fractions are further electrophoresed on SDS-PAGE (lower panel, under
reducing conditions). Protein marker is used for molecular weight calibration. The protein bands are visualized
by Coomassie blue staining
Fig. 2 Step-by-step workflows for protein digestion (upper panel) and peptides extraction and desalting (lower
panel) protocol
3.3.2 Nano-Electrospray 1. Perform the detection analysis using 1260 Infinity Nanoflow
Ionization-Liquid LC system (Agilent, Santa Clara, CA, USA) that is connected
Chromatography Tandem to Accurate-Mass Q-TOF 6550 series with a nano-electrospray
Mass Spectrometry ionization source.
(ESI-LC-MS/MS) and Data 2. Reconstitute the lyophilized peptide analytes in 7 μL of 0.1%
Mining formic acid in ddH2O. Subject the peptide analytes to HPLC
Large-Capacity Chip Column Zorbax 300-SB-C18 (160 nL
enrichment column, a 75 μm 150 mm analytical column with
5 μm particles) (Agilent, Santa Clara, CA, USA).
3. Adjust the injection volume to 1 μL per sample, using a flow
rate of 0.4 μL/min, with a linear gradient of 5–70% of solvent B
(0.1% formic acid in 100% acetonitrile).
4. Flow the drying gas at a rate of 11 L/min at temperature
290 C. Set the fragmentor voltage at 175 V and the capillary
voltage at 1800 V. Acquire the mass spectra using Mass Hunter
acquisition software (Agilent, Santa Clara, CA, USA) in a
MS/MS mode with an MS scan range of 200–3000 m/z and
MS/MS scan range of 50–3200 m/z.
5. Extract the data with MH+ mass range between 50 and
3200 Da and process with Agilent Spectrum Mill MS Proteo-
mics Workbench software packages version B.04.00 against
merged database incorporating both non-redundant NCBI
90 Choo Hock Tan et al.
4 Notes
References
1. Lomonte B, Fernández J, Sanz L, Angulo Y, kaouthia (monocled cobra) venoms from three
Sasa M, Gutiérrez JM, Calvete JJ (2014) Ven- different geographical regions of Southeast
omous snakes of Costa Rica: biological and Asia. J Proteome 120:105–125. https://doi.
medical implications of their venom proteomic org/10.1016/j.jprot.2015.02.012
profiles analyzed through the strategy of snake 6. Gutiérrez JM, Lomonte B, León G, Alape-
venomics. J Proteome 105(Supplement Girón A, Flores-Dı́az M, Sanz L, Angulo Y,
C):323–339. https://doi.org/10.1016/j. Calvete JJ (2009) Snake venomics and antive-
jprot.2014.02.020 nomics: proteomic tools in the design and con-
2. Calvete JJ, Sanz L, Angulo Y, Lomonte B, trol of antivenoms for the treatment of
Gutiérrez JM (2009) Venoms, venomics, anti- snakebite envenoming. J Proteome 72
venomics. FEBS Lett 583(11):1736–1743. (2):165–182. https://doi.org/10.1016/j.
https://doi.org/10.1016/j.febslet.2009.03. jprot.2009.01.008
029 7. Vetter I, Davis JL, Rash LD, Anangi R,
3. Tan CH, Tan KY, Lim SE, Tan NH (2015) Mobli M, Alewood PF, Lewis RJ, King GF
Venomics of the beaked sea snake, Hydrophis (2011) Venomics: a new paradigm for natural
schistosus: a minimalist toxin arsenal and its products-based drug discovery. Amino Acids
cross-neutralization by heterologous antive- 40(1):15–28. https://doi.org/10.1007/
noms. J Proteome 126:121–130. https://doi. s00726-010-0516-4
org/10.1016/j.jprot.2015.05.035 8. Tan KY, Tan CH, Chanhome L, Tan NH
4. Tan KY, Tan NH, Tan CH (2018) Venom (2017) Comparative venom gland transcrip-
proteomics and antivenom neutralization for tomics of Naja kaouthia (monocled cobra)
the Chinese eastern Russell’s viper, Daboia sia- from Malaysia and Thailand: elucidating geo-
mensis from Guangxi and Taiwan. Sci Rep graphical venom variation and insights into
8(1):8545. https://doi.org/10.1038/ sequence novelty. PeerJ 5:e3142. https://doi.
s41598-018-25955-y org/10.7717/peerj.3142
5. Tan KY, Tan CH, Fung SY, Tan NH (2015) 9. Augusto-de-Oliveira C, Stuginski DR, Kitano
Venomics, lethality and neutralization of Naja ES, Andrade-Silva D, Liberato T, Fukushima I,
92 Choo Hock Tan et al.
Serrano SM, Zelanis A (2016) Dynamic rear- 15. Petras D, Sanz L, Segura A, Herrera M,
rangement in snake venom gland proteome: Villalta M, Solano D, Vargas M, Leon G, War-
insights into Bothrops jararaca intraspecific rell DA, Theakston RD, Harrison RA,
venom variation. J Proteome Res 15 Durfa N, Nasidi A, Gutierrez JM, Calvete JJ
(10):3752–3762. https://doi.org/10.1021/ (2011) Snake venomics of African spitting
acs.jproteome.6b00561 cobras: toxin composition and assessment of
10. Calvete JJ (2014) Next-generation snake congeneric cross-reactivity of the pan-African
venomics: protein-locus resolution through EchiTAb-Plus-ICP antivenom by antivenomics
venom proteome decomplexation. Expert Rev and neutralization approaches. J Proteome Res
Proteomics 11(3):315–329. https://doi.org/ 10(3):1266–1280. https://doi.org/10.1021/
10.1586/14789450.2014.900447 pr101040f
11. Tan CH, Wong KY, Tan KY, Tan NH (2017) 16. Tan NH, Fung SY, Tan KY, Yap MKK, Gna-
Venom proteome of the yellow-lipped sea krait, nathasan CA, Tan CH (2015) Functional
Laticauda colubrina from Bali: insights into venomics of the Sri Lankan Russell’s viper
subvenomic diversity, venom antigenicity and (Daboia russelii) and its toxinological correla-
cross-neutralization by antivenom. J Proteome tions. J Proteome 128:403–423. https://doi.
166:48–58. https://doi.org/10.1016/j.jprot. org/10.1016/j.jprot.2015.08.017
2017.07.002 17. Tan CH, Fung SY, Yap MK, Leong PK, Liew
12. Alape-Giron A, Sanz L, Escolano J, Flores- JL, Tan NH (2016) Unveiling the elusive and
Diaz M, Madrigal M, Sasa M, Calvete JJ exotic: Venomics of the Malayan blue coral
(2008) Snake venomics of the lancehead pitvi- snake (Calliophis bivirgata flaviceps). J Prote-
per Bothrops asper: geographic, individual, and ome 132:1–12. https://doi.org/10.1016/j.
ontogenetic variations. J Proteome Res 7 jprot.2015.11.014
(8):3556–3571. https://doi.org/10.1021/ 18. Dutta S, Chanda A, Kalita B, Islam T, Patra A,
pr800332p Mukherjee AK (2017) Proteomic analysis to
13. Wong KY, Tan CH, Tan KY, Naeem QH, Tan unravel the complex venom proteome of east-
NH (2018) Elucidating the biogeographical ern India Naja naja: correlation of venom
variation of the venom of Naja naja (specta- composition with its biochemical and pharma-
cled cobra) from Pakistan through a venom- cological properties. J Proteome 156:29–39.
decomplexing proteomic study. J Proteome https://doi.org/10.1016/j.jprot.2016.12.
175:156–173. https://doi.org/10.1016/j. 018
jprot.2017.12.012 19. Tan CH, Tan KY, Yap MK, Tan NH (2017)
14. Faisal T, Tan KY, Sim SM, Quraishi N, Tan Venomics of Tropidolaemus wagleri, the sexu-
NH, Tan CH (2018) Proteomics, functional ally dimorphic temple pit viper: unveiling a
characterization and antivenom neutralization deeply conserved atypical toxin arsenal. Sci
of the venom of Pakistani Russell’s viper Rep 7:43237. https://doi.org/10.1038/
(Daboia russelii) from the wild. J Proteome srep43237
183:1–13. https://doi.org/10.1016/j.jprot.
2018.05.003
Chapter 6
Abstract
Peptide spectral libraries enable targeted identification and quantitation of low-abundance proteins in a
complex plant proteome. Here we describe parallel protein and peptide fractionation techniques to improve
plant proteome coverage and facilitate construction of spectral libraries.
Key words Plant proteomics, Protein fractionation, Peptide fractionation, C18, SCX, PEG
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_6, © Springer Science+Business Media, LLC, part of Springer Nature 2019
93
94 Martin Černý et al.
2 Materials
2.1 Homogenization 1. Mixer Mill MM 400 (Retsch), stainless steel grinding jars and
milling balls (see Note 1).
Plant Proteome Fractionation Protocol 95
Fig. 2 Denaturing protein extraction and protein digestion. (a–g) Protein extraction and purification. (h)
Determination of protein concentration, (i1–2) protein separation, and (i3, j) digestion
2. Liquid nitrogen.
3. 2.0 mL Eppendorf LoBind tubes or similarly coated low-
protein-binding microcentrifuge tubes.
96 Martin Černý et al.
2.7 Peptide 1. VersaPlate, collection plate, C18 (25 mg) and SCX (50 mg)
Fractionation tubes (Agilent), vacuum pump (see Note 10).
2. 1% TFA (prepared in Subheading 2.6).
3. Concentration series of acetonitrile in 0.1% (v/v) triethyla-
mine: Prepare 2 mL of 0.1% (v/v) triethylamine in water and
2 mL of 0.1% (v/v) triethylamine in acetonitrile. Mix 50, 75,
100, 125, 150, 175, 200, and 250 μL of triethylamine in
acetonitrile with the appropriate volume of triethylamine in
water to obtain 1 mL of each stock solution (5–50%).
4. 0.5% formic acid: Dilute formic acid (FA) with water to pro-
duce 15 mL of 0.5% (v/v) FA, store at room temperature.
5. Concentration series of ammonium acetate in 0.5% FA: Prepare
ca 1.5 mL of 500 mM ammonium acetate in 0.5% FA (dissolve
38.5 mg of ammonium acetate per 1 mL of 0.5% FA), and then
dilute this stock by mixing 50, 100, 250, 300, and 400 μL with
the necessary amounts of 0.5% FA to obtain 0.5 mL volumes of
50–400 mM stock solutions.
3 Methods
3.2 Native Extraction 1. Take a homogenized aliquot, place it on ice and add 1.0 mL of
and PEG Fractionation extraction buffer.
2. Sonicate at 4 C for 5 min and then incubate in the Thermo-
mixer at 4 C, 800 rpm for 10 min.
3. Centrifuge for 10 min (10,000 g, 4 C), transfer the super-
natant to a new 2.0 mL tube, and keep it on ice. Mix the pellet
with 10% (w/v) TCA in acetone and then follow steps 2–9 of
the denaturing extraction protocol (Subheading 3.3.).
4. Add 90 μL of 50% (w/v) PEG solution to give a final concen-
tration of ca 4% (w/v), incubate in the Thermomixer (4 C,
800 rpm, 20 min), then centrifuge for 10 min (10,000 g,
4 C), collect the supernatant, and process the pellet as in the
previous step.
5. Repeat the supernatant precipitation in a stepwise PEG con-
centration gradient, adding 100 and 125 μL of 50% (w/v) PEG
to give, respectively, a 8 and a 12% (w/v) mixture. Collect
pellets, mix with 10% (w/v) TCA in acetone, and transfer the
last supernatant into a 5.0 mL tube and precipitate it with
4.0 mL of 10% (w/v) TCA in acetone, then proceed to the
second step of the denaturing extraction (Subheading 3.3).
3.4 In-Solution 1. Dilute 300 μg of protein with an equal volume of water and
Digestion two volumes of ammonium bicarbonate buffer.
2. Add 50 μL of immobilized trypsin beads (Promega, see Note 7)
and incubate overnight on a rotator at 30 rpm in an incubator
at 30 C (see Note 8).
3.5 Protein 1. Dilute 300 μg of protein to a final volume of 260 μL with IEF
Separation and In-Gel solubilization solvent (if needed), add 1.3 μL of ampholytes
Digestion (pH 3–10), and load onto two 7 cm 3–10 NL IPG strips
(Bio-Rad) in a rehydration tray.
3.5.1 Isoelectric
Focusing
2. Overlay with mineral oil and rehydrate overnight at room
temperature.
3. Wet four paper wicks in water, transfer IPG strips into the
focusing tray, and put wet paper wicks between gel and elec-
trode to prevent their making direct contact. Overlay again
with mineral oil.
4. Isoelectrically focus proteins at 20 C in a PROTEAN IEF Cell
unit (Bio-Rad) in six steps: 150 V (20 min), 300 V (20 min),
600 V (20 min), 1500 V (20 min), 3000 V (20 min), and
4000 V up to 12,000 Vh.
5. Put the IPG strips onto a clean filter paper with the gel facing
up to dry off the mineral oil.
6. Align one IPG strip above the other, cut gels vertically into ten
equal fractions, and collect them in 1.5 mL LoBind tubes.
3.5.3 In-Gel Digestion 1. Wash gel pieces twice with 1.0 mL acetonitrile and dry samples
down in a SpeedVac Evaporator.
2. Use a Retsch mill and stainless steel milling balls to obtain a fine
powder (see Note 14). Place samples on ice, add 150 μL diges-
tion buffer, incubate for 15 min, then transfer tubes to 37 C
and incubate overnight.
3. Extract peptides twice with 150 μL acetonitrile, collect extracts
into 0.5 mL thin-walled PCR tubes, and dry down to ca 100 μL
in a SpeedVac Evaporator (see Note 15).
3.6 Peptide 1. Mix a peptide sample 1:1 with 1% TFA, shake, and clarify the
Desalting sample by centrifugation (10,000 g, 5 min).
2. Wash a C18 SPE column with 0.5 mL 100% acetonitrile,
2 1 mL of water, 1 1 mL of 1% TFA.
3. Load the acidified sample solution onto the SPE column (see
Note 16).
4. Wash the column twice with 0.5 mL 1% TFA.
5. Elute peptides in two steps: 100 μL 50% (v/v) acetonitrile in
water, followed by 300 μL of acetonitrile; collect the flow-
through into 0.5 mL thin-walled PCR tubes, dry samples
down in a SpeedVac Evaporator to ca 40 μL (see Note 15),
and estimate peptide concentration by Quantitative Colorimet-
ric Peptide Assay (Thermo Scientific).
3.7 Peptide 1. Mix the peptide sample from in-solution digestion 1:1 with 1%
Fractionation TFA, shake, and clarify the sample by centrifugation
(10,000 g, 5 min).
2. Wash two C18 SPE columns with 0.5 mL of 100% acetonitrile,
2 1.0 mL of water, 1 with 1.0 mL of 1% TFA.
3. Divide the acidified sample equally between two SPE columns,
wash once with 1% TFA and then proceed to high pH or SCX
fractionation.
3.7.1 High pH C18 1. Wash bound peptides with 1.0 mL of water (see Note 16).
Fractionation 2. Elute peptides with a stepwise gradient of acetonitrile in 0.1%
triethylamine. Load, successively, 200 μL each of 5%, 7.5%,
102 Martin Černý et al.
10%, 12.5%, 15%, 17.5%, 20%, 25%, and 50% acetonitrile, and
collect fractions into 0.5 mL thin-walled PCR tubes.
3. Dry down to ca 20–30 μL in a SpeedVac Evaporator (see Note
15) and estimate peptide concentration by Quantitative Col-
orimetric Peptide Assay (Thermo Scientific).
3.7.2 Peptide SCX 1. Elute bound peptides with 100 μL of 50% (v/v) acetonitrile in
Fractionation water followed by 300 μL of acetonitrile, collect fractions into
0.5 mL thin-walled PCR tubes.
2. Dry down to ca 100 μL in a SpeedVac Evaporator (see Note 15)
and dilute in 1.0 mL of 50 mM ammonium acetate in 0.5% FA.
3. Wash an SCX SPE column with, successively, 0.5 mL of
500 mM ammonium acetate in 0.5% FA and 2 1.0 mL of
50 mM ammonium acetate, then load the peptide sample and
collect the flow-through into a 0.5 mL thin-walled PCR tube
(the first fraction).
4. Elute peptides with a stepwise gradient of ammonium acetate
in 0.5% FA. Load, successively, 200 μL each of 100, 250,
300, 400, and 500 mM ammonium acetate, and collect frac-
tions into 1.5 mL tubes (see Note 17).
5. Dilute the collected eluates with 800 μL of 0.1% (v/v) FA in
water; desalt peptides on a C18 SPE column as described in
Subheading 3.6.
4 Notes
Acknowledgments
References
1. Milo R (2013) What is the total number of 9. Rappsilber J, Mann M, Ishihama Y (2007) Pro-
protein molecules per cell volume? A call to tocol for micro-purification, enrichment,
rethink some published values. BioEssays pre-fractionation and storage of peptides for
35:1050–1055 proteomics using StageTips. Nat Protoc
2. Picotti P, Bodenmiller B, Mueller LN, 2:1896–1906
Domon B, Aebersold R (2009) Full dynamic 10. Mostovenko E, Hassan C, Rattke J, Deelder
range proteome analysis of S. cerevisiae by tar- AM, van Veelen PA, Palmblad M (2013) Com-
geted proteomics. Cell 138:795–806 parison of peptide and protein fractionation
3. Schubert OT, Gillet LC, Collins BC, methods in proteomics. EuPA Open Proteom
Navarro P, Rosenberger G, Wolski WE et al 1:30–37
(2015) Building high-quality assay libraries 11. Černý M, Skalák J, Kurková B, Babuliaková E,
for targeted analysis of SWATH MS data. Nat Brzobohatý BB (2011) Using a commercial
Protoc 10(3):426–441 method for rubisco immunodepletion in anal-
4. Acquadro A, Flavo S, Mila S, Albo AG, ysis of plant proteome. Chemické listy
Comino C, Moglia A, Lanteri S (2009) Prote- 105:640–642
omics in globe artichoke: protein extraction 12. Černý M, Jedelský PL, Novák J, Schlosser A,
and sample complexity reduction by PEG frac- Brzobohatý B (2014) Cytokinin modulates
tionation. Electrophoresis 30(9):1594–1602 proteomic, transcriptomic and growth
5. Wang W-Q, Song B-Y, Deng Z-J, Wang Y, Liu responses to temperature shocks in Arabidop-
S-J, Møller IM, Song S-Q (2015) Proteomic sis. Plant Cell Environ 37:1641–1655
analysis of lettuce seed germination and ther- 13. Righetti PG, Boschetti E (2016) Global prote-
moinhibition by sampling of individual seeds at ome analysis in plants by means of peptide
germination and removal of storage proteins by libraries and applications. J Proteomics
polyethylene glycol fractionation. Plant Physiol 143:3–14
167(4):1332–1350 14. Stevens R, Stevens L, Price N (1983) The sta-
6. Cerna H, Černý M, Habánová H, Šafářová D, bilities of various thiol compounds used in pro-
Abushamsiya K, Navrátil M, Brzobohatý B tein purifications. Biochem Educ 11:70
(2017) Proteomics offers insight to the mech- 15. Berka M, Luklová M (2017) Limited drying
anism behind Pisum sativum L. response to Pea and its effect on peptide recovery rates. In:
seed-borne mosaic virus (PSbMV). J Proteo- Polak O et al (eds) MendelNet 2017 Proceed-
mics 153:78–88 ings of 24th International PhD Students Con-
7. Baldrianová J, Černý M, Novák J, Jedelský PL, ference. 24th International PhD Students
Divı́šková E, Brzobohatý B (2015) Arabidopsis Conference, Brno, November 2017. p 91
proteome responses to the smoke-derived 16. Nukarinen E, Tomanov K, Ziba I,
growth regulator karrikin. J Proteomics Weckwerth W, Bachmair A (2017) Protein
120:7–20 sumoylation and phosphorylation intersect in
8. Batth TS, Francavilla C, Olsen JV (2014) Arabidopsis signaling. Plant J 91:505–517
Off-line high-pH reversed-phase fractionation
for in-depth phosphoproteomics. J Proteome
Res 13:6176–6186
Chapter 7
Abstract
High-density protein microarrays constitute a promising high-throughput platform for the characterization
of protein expression patterns, biomarker discovery, and validation. Different types of protein microarrays
have been described according to several features (such as content, format, and detection system) present-
ing advantages and disadvantages which are relevant for the specific application and purposes. Therefore, an
experimental design is key for any screening based on protein microarrays assays; in fact, the data analysis
strategy is directly related to the experimental design, type of protein microarray and consequently the final
outcome, the data and results interpretation, is also directly linked. Here, it is proposed a systematic
workflow for biomarker discovery based on tailor-made protein microarrays platforms which obtain
comprehensively info for the functional protein characterization in high-throughput format.
Key words Protein microarray, Analysis, Proteome, Antibodies, Fluorescence, Proteomics, Normali-
zation, Biomarker, Protein microarray
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_7, © Springer Science+Business Media, LLC, part of Springer Nature 2019
107
108 Rodrigo Garcı́a-Valiente et al.
1.1 Microarray The majority of the protein microarrays are developed in two for-
Format mats (depending of the surface) [6, 9–11]:
1.1.1 Planar Arrays In this type of arrays, the content (protein, peptide, aptamer, tissue,
or cell lysates) is immobilized in microspots arranged in a
two-dimensions (2D) surface (around 250 μm of diameter and
separated ~300 μm) over a solid matrix. In this 2D organized
spatial distribution, spot density around 1000 spot per square
centimeter is normally reached in most of the protein microarrays
commercially available [4, 11].
In these arrays, several aspects are directly related with the
robustness and reproducibility of the assay performance [12],
such as spot (size, morphology, and reproducibility), the ligand
(binding capacity), the sample, surface, and method (background
signal), and the detection limits. In addition, other aspects are also
related to physicochemical properties of the surface [13] or bio-
molecules, which have a consequence in the assay development,
such as spotting buffer composition affecting the protein structure,
printing method (contact or noncontact) [14], hygroscopy, and
humidity. In addition, the ratio background/signal is important;
then, the unspecific binding onto the array surface has to be eval-
uated and controlled in order to detect correctly the ligand-agent
A Systematic Analysis Workflow for High-Density Customized Protein. . . 109
1.1.2 Bead Arrays In this format, the ligand is bound to addressable beads (color-
coded beads or quantum dots), whose diameter typically varies
from 0.02 to 0.1 μm (nanoparticles) or from 0.1 μm (micro-
spheres) [16]. Usually, to be able to distinguish them, beads with
different ligands are previously labeled with different combination
of fluorescent dyes.
Then, the color-coded beads are easily detected by flow cyto-
metry (in which one or several lasers excite the internal dye), and a
reporter fluorescence dye (which is directly linked to the identifica-
tion or quantification of the target protein, and excitation/emis-
sion are far different from the ones used for the internal color-
coded dyes). The detector captures the color profile and identifies
the ligand and therefore the target protein (by an assigned intensity
to the analyte).
1.1.3 Microarray Content Protein microarrays offer a wide diversity of subclasses according to
the biomolecules deposited or displayed onto the surface. To sim-
plify the classification, they can be categorized as assembled arrays
or self-assembled arrays [17].
Assembled Arrays Composed by, typically, antibodies, purified proteins or other enti-
ties which are immobilized onto a functionalized surface. Some
types of arrays included in this category are:
1. Capture arrays
They are generated by printing analyte-specific reagents
(ASRs), usually antibodies [18] (Fig. 1), but sometimes phages
[19] or others, over the array surface. These ASRs serve to
identify and quantify the presence of multiple entities simulta-
neously. Therefore, they are used to find biomarkers and detect
molecular signatures. The quality of the results depends on the
quality (specificity and affinity) of the ASRs, which, in the case
of antibodies, is related to them being poly- or monoclonal.
There are different detection methods for measuring the
analyte-reagent union, being direct (e.g., fluorescent, Cy3
and/or Cy5 antibodies) or indirect (biotinylated sample
revealed with streptavidine, or secondary antibodies marked
with HRP) [17].
2. Reverse-phase arrays
The concept is opposite to the capture arrays. On these arrays,
the samples are deposited onto the surface (Fig. 2). The
reverse-phase arrays are very useful to evaluate many samples
against a single ASR. It is critical to ensure the affinity of the
110 Rodrigo Garcı́a-Valiente et al.
2 Materials
2.1 Experimental Prepare all materials at room temperature unless indicated other-
Materials wise.
l Reactives
– Blocking buffer: PBS Na/K 1 + BSA 1% (w/v), 0.2% (w/v)
Tween 20.
– Streptavidine/fluor conjugate.
l Benchtop instruments
– Agitator.
– Orbital agitator.
– Array washer platform.
3 Methods
3.1 Customize Customize arrays (Fig. 4) will be designed according to the partic-
Design ular needs of the specific screening. Some aspects have to be taken
into account in an array-based assay.
1. According to the study, select appropriate Positive and Nega-
tive Controls and include them among the ligands. A standard
negative control is the cleaning buffer. Including internal con-
trols is an important tool to have an estimation of the behavior
of the array and/or the sample. “The more, the merrier.”
114 Rodrigo Garcı́a-Valiente et al.
Fig. 4 Scheme of an antibody microarray printing followed by the assay. In the first phase (1), each specific
antibody for each target protein is prepared in a plate, eluted in its corresponding master mix. The printing of
the array (2) is carried out in a chamber with controlled temperature and humidity. Between each spot, the pin
has to be cleaned with cleaning buffer to prevent cross contamination. After printing, the batch of arrays are
dried and can be conserved for posterior use. Before using the microarray, it has to be blocked to prevent
unspecific binding (3). After blocking and also after all the following steps (4 and 5) the microarray has to be
thoroughly rinsed with distilled water. The array is incubated with the sample (4) and developed (5). After the
last rinse, the array is dried and can be scanned. The microarrays can be preserved in darkness at r.t. in
controlled humidity
3.2 Experimental Carry out all procedures at room temperature unless otherwise is
Workflow specified on a protocol step.
In case of a high number of samples to be printed at once, then,
technical replicates and biological replicates must be randomly and
uniformly separated between the different experimental batches, to
decrease the so-called batch effect.
It is important to optimize the amount of sample and ligand.
For that, it is recommended to do previous smaller assays combin-
ing different dilutions of each, and to choose the combination that
offers a less background/signal ratio.
3.2.1 Array Printing 1. Create an Excel file with the future and random distribution of
the samples, negative controls and positive controls to print in
Sample Preparation
the microplate (see Note 1).
2. Clean the laboratory work bench with 70% ethanol. Prepare
required samples and reactives for the microplate preparation.
3. Protein samples shall be diluted in PBS Na/K 1. As cross-
linker BS3 at 50 mg/mL is used.
4. Microplates will be loaded according to the chosen distribu-
tion. Each sample will be loaded in a 1:1 dilution (v/v) with
47% glycerol (v/v).
5. After the microplates are set up, spin them in the centrifuge,
using a microtiter adaptor.
Array Printing 1. Turn on the injection array printer, the associated control
station, and linked computer.
2. Perform maintenance routine, specified by the manufacturer.
3. Introduce in the printer the microplate with the samples to
print, and the functionalized slides, the microarrays, using the
command to Load microplates and slides, ensuring that the
microplates and the slides are correctly positioned.
4. In the computer, create a folder for the experiment. In said
folder, save the execution parameters.
5. Start the printing, pressing the corresponding command. The
machine will start printing each array automatically.
6. Once the printing process is over, save the quality results in the
earlier mentioned folder.
7. Re-initialize the system (see Note 2).
8. Extract the microplate and store them in the conditions
required by the samples.
9. Extract the printed microarrays. Label them (see Note 3).
10. Dry the printed microarrays in the stove at 37 C along with an
absorbent agent, silica gel.
11. Store the microarrays at room temperature.
116 Rodrigo Garcı́a-Valiente et al.
3.2.2 Array Assays 1. Microarray blocking. Submerge the microarrays in the micro-
array washing chambers, in 6 mL of blocking solution on each
one of them, during 1 h, in orbital agitation (see Note 4).
2. Microarray washing. Wash intensely during 10 min using
milliQ water in the array washer platform. After this, wash
three times, placing each array in the array washing chambers,
filled with distilled water, in orbital agitation for 5 min each.
They will be kept in distilled water until their processing.
3. Biologic sample processing. In this case, sample must be bioti-
nilized, following the protocol described by Sierra A. et al.,
Journal Proteome Research 2016.
4. Microarray drying. Microarrays will be dried using centrifuga-
tion at 240 g during 3 min, using the adaptors for 50 mL
tubes. Once dried, they can be stored at 4 C during a maxi-
mum of 15 days.
5. Incubate biological samples at the chosen dilution (see Note 5)
overnight and at 4 C at orbital agitation.
6. Microarray washing (see Note 4). Wash for 7 min using milliQ
water in the array washer platform. After this, wash three times,
placing each array in the array washing chambers, filled with
distilled water, in orbital agitation for 5 min each. They will be
kept in distilled water until their processing.
7. Sample incubation by indirect method given marked -biotiny-
lated- samples.
(a) Preparation of the streptavidine. Dilute 1:200 (v/v) of
0.1 mg of streptavidine/fluor conjugate in milliQ water.
(b) Wet chamber preparation. Add distilled water to the wet
chamber, enough to create humidity, but not enough to
touch the arrays when they are added.
(c) Incubation. Put the cover slips over the arrays, and the
arrays in the wet chamber without touching the water.
Incubate 200 μL of the reveal sample over the full micro-
array for 20 min in the wet chamber, at no light
conditions.
8. Microarray washing (see Note 4). Wash for 7 min using milliQ
water in the array washer platform. After this, wash three times,
placing each array in the array washing chambers, filled with
distilled water, in orbital agitation for 5 min each. They will be
kept in distilled water until their processing.
9. Microarray drying. Microarrays will be dried using centrifuga-
tion at 240 g during 3 min, using the adaptors for 50 mL
tubes. Once dried, they have to be stored at no light exposure
until scanned.
A Systematic Analysis Workflow for High-Density Customized Protein. . . 117
Fig. 5 General scheme of the analysis process after the assay. After the assay, the image is scanned in a
fluorescent array imaging reader, which generates a file with the associated data for further statistical analysis
Fig. 6 Unacceptable rack configurations (left) vs. correct rack configurations (right)
3.3 Computational For this step, we recommend using the GenePix software, which
Analysis allows us to obtain a GenePix Results file (GPR). This image
intensity file is needed for ulterior steps.
3.3.1 Image Analysis
For each generated image:
1. Open image (Ctrl+O).
2. Choose correct wavelength.
3. Adjust brightness and contrast.
4. Open the gal file/Array List (Alt+Y).
5. Adjust gal (manually and/or pressing F5 for automatic mode).
6. Analyze (Ctrl+A).
7. Configure. Background subtraction method: local.
8. Save results as .gpr.
9. Explore .gpr files with Notepad++, focusing on spot IDs, and
checking they are correct.
3.3.2 Data Analysis In order to obtain reliable and meaningful biological information
from a microarray experiment, it has to be analyzed with a consis-
tent statistical method (see Note 7). There are different available
strategies to analyze microarrays, but not all of them, which were
developed to analyze other microarray technologies such as DNA
microarrays, are easily translatable to protein microarrays. The
common steps to perform an analysis for this kind of assay are the
following:
1. Import data sets. The Genepix output must be read into the
software that will be used for the analysis. Suitable software
choices for this task could be Python, R, or Matlab. GPR files
contain, among other parameters, the mean, median, and stan-
dard deviation of the pixel intensity, as well as the total intensity
for a spot at a given wavelength.
2. Background subtraction. A simple way to subtract the back-
ground for each intensity is using the default background
measure from Genepix. Working with median values is
recommended.
A Systematic Analysis Workflow for High-Density Customized Protein. . . 119
Fig. 7 Boxplots showing the distribution of the median intensities of the background (gray) and foreground
(yellow) of every spot after logarithmic transformation. Seven assays are shown
Fig. 8 Smooth histogram of the logarithmic transformed median intensity (after background is subtracted) of
all spots in a microarray. Different colors for each subarray, in case multiple assays are being carried over the
same slide. The cutoff point is plotted as the vertical blue dotted line. All points with less intensity than the
cutoff point will be considered negative and the ones with higher intensity will be further evaluated
Fig. 9 Smooth histogram (top) and Boxplots (bottom) showing the median intensities of the positive spots (after
logarithmic transformation) for every assay. Data not normalized (left), with standard scoring (center) and with
a quantile normalization (right) are shown
4 Notes
Acknowledgments
References
1. Sierra-Sánchez Á, Garrido-Martı́n D, 14. Glökler J, Angenendt P (2003) Protein and
Lourido L, González-González M, Dı́ez P, antibody microarray technology. J Chromatogr
Ruiz-Romero C et al (2017) Screening and B Anal Technol Biomed Life Sci 797:229–240
validation of novel biomarkers in osteoarticular 15. Kusnezow W, Jacob A, Walijew A, Diehl F,
pathologies by comprehensive combination of Hoheisel JD (2003) Antibody microarrays: an
protein array technologies. J Proteome Res 16 evaluation of production parameters. Proteo-
(5):1890–1899 mics 3(3):254–264
2. Zyuzin MV, Dı́ez P, Goldsmith M, Carregal- 16. Casado-Vela J, González-González M,
Romero S, Teodosio C, Rejman J et al (2017) Matarraz S, Martı́nez-Esteso MJ, Vilella M,
Comprehensive and systematic analysis of the Sayagués JM et al (2013) Protein arrays: recent
immunocompatibility of polyelectrolyte cap- achievements and their application to study the
sules. Bioconjug Chem 28(2):556–564 human proteome. Curr Proteomics 10
3. Dı́ez P, Ibarrola N, Dégano RM, Lécrevisse Q, (2):83–97. https://doi.org/10.2174/
Rodriguez-Caballero A, Criado I et al (2017) A 1570164611310020003
systematic approach for peptide characteriza- 17. Lourido L, Diez P, Dasilva N, Gonzalez-
tion of B-cell receptor in chronic lymphocytic Gonzalez M, Ruiz-Romero C, Blanco F, et al
leukemia cells. Oncotarget 8 (2014) Protein microarrays: overview, applica-
(26):42836–42846 tions and challenges. In: Genomics and prote-
4. Merbl Y, Kirschner MW (2011) Protein micro- omics for clinical discovery and development.
arrays for genome-wide posttranslational mod- Springer. p 147–173., https://doi.org/10.
ification analysis. Wiley Interdiscip Rev Syst 1007/978-94-017-9202-8_8
Biol Med 3(3):347–356 18. LaBaer J, Ramachandran N (2005) Protein
5. Dasgupta A (2008) Handbook of drug moni- microarrays as tools for functional proteomics.
toring methods: therapeutics and drugs of Curr Opin Chem Biol 9:14–19
abuse. Humana, Totowa, NJ, pp 1–445 19. Jara-Acevedo R, Dı́ez P, González-
6. Yu X, Schneiderhan-Marra N, Joos TO (2011) González M, Dégano RM, Ibarrola N, Gón-
Protein microarrays and personalized medi- gora R et al (2018) Screening phage-display
cine. Ann Biol Clin (Paris) 69(1):17–29 antibody libraries using protein arrays. In:
7. Yu X, Schneiderhan-Marra N, Joos TO (2010) Phage display. Methods Mol Biol
Protein microarrays for personalized medicine. 1701:365–380
Clin Chem 56:376–387 20. Spurrier B, Ramalingam S, Nishizuka S (2008)
8. Dı́ez P, Dasilva N, González-González M, Reverse-phase protein lysate microarrays for
Matarraz S, Casado-Vela J, Orfao A et al cell signaling analysis. Nat Protoc 3
(2012) Data analysis strategies for protein (11):1796–1808
microarrays. Microarrays 1(3):64–83 http:// 21. He M, Taussig MJ (2001) Single step genera-
www.mdpi.com/2076-3905/1/2/64/ tion of protein arrays from DNA by cell-free
9. Gonzalez-Gonzalez M, Jara-Acevedo R, expression and in situ immobilisation (PISA
Matarraz S, Jara-Acevedo M, Paradinas S, Saya- method). Nucleic Acids Res 29(15):E73–E73
gües JM et al (2012) Nanotechniques in prote- http://www.ncbi.nlm.nih.gov/entrez/query.
omics: protein microarrays and novel detection fcgi?cmd¼Retrieve&db¼PubMed&dopt¼Cita
platforms. Eur J Pharm Sci 45:499–506 tion&list_uids¼11470888
10. Dasilva N, Dı́ez P, Matarraz S, González- 22. He M, Stoevesandt O, Palmer EA, Khan F,
González M, Paradinas S, Orfao A et al Ericsson O, Taussig MJ (2008) Printing pro-
(2012) Biomarker discovery by novel sensors tein arrays from DNA arrays. Nat Methods 5
based on nanoproteomics approaches. Sensors (2):175–177
12:2284–2308 23. Tao SC, Zhu H (2006) Protein chip fabrication
11. Matarraz S, González-González M, Jara M, by capture of nascent polypeptides. Nat Bio-
Orfao A, Fuentes M (2011) New technologies technol 24(10):1253–1254
in cancer. Protein microarrays for biomarker 24. Ramachandran N, Raphael JV, Hainsworth E,
discovery. Clin Transl Oncol 13:156–161 Demirkan G, Fuentes MG, Rolfs A et al (2008)
12. Ellington AA, Kullo IJ, Bailey KR, Klee GG Next-generation high-density self-assembling
(2010) Antibody-based protein multiplex plat- functional protein arrays. Nat Methods 5
forms: technical and operational challenges. (6):535–538
Clin Chem 56:186–193 25. Hicks SC, Irizarry RA (2014) When to use quan-
13. Fuentes M, Dı́ez P, Casado-Vela J (2016) tile normalization? bioRxiv. doi: https://doi.
Nanotechnology in the fabrication of protein org/10.1101/012203. http://biorxiv.org/con
microarrays. Methods Mol Biol 1368:197–208 tent/early/2014/12/04/012203.abstract
Chapter 8
Abstract
Proteomics is a widely used method for defining the protein composition of a complex sample. As this
approach allows for identification and quantification of proteins across a broad dynamic range as well as
detection of post-translational modifications, proteomics is an ideal platform to investigate the gut micro-
biome at a functional level. The gut microbiome is a dynamic environment which is crucial for overall health
and fitness. Imbalances in the gut microbiome can influence nutrient absorption, pathogen resistance,
inflammation, and various human diseases. Metaproteomic analysis of the gut microbiome is currently
being performed on bacteria isolated from (1) fecal samples (2) colonic lavage, or (3) colon biopsies.
Investigation of the gut microbiome has demonstrated that within the colon, there are distinct commu-
nities based on spatial location, and separable from the gut microbiomes isolated from stool. In addition to
expanding our understanding of host–bacterial interactions for human health and disease, gut microbiome
analysis is being utilized for biomarker development to discriminate normal individuals and diseased (i.e.,
inflammatory bowel disease or colon cancer) patients as well as to monitor disease activity and prognosis.
The adult human gastrointestinal (GI) tract runs from the esopha-
gus through the stomach and colon to the rectum. These organs
are host to an enormous population of microorganisms, possibly
upward of 100 trillion representing between 15,000 and 36,000
different species of bacteria [1–3]. Through host interaction, these
bacteria, fungi, and viruses respond to stimuli within their micro-
environment and impact a broad spectrum of essential functions
including assisting with digestion of food, vitamin production/
absorption, metabolism, nutrient extraction, immune response,
and conferring resistance to pathogenic organisms [4, 5]. While
the numbers and density of bacteria within the human gut are
extremely high, the diversity is surprisingly low. Bacteria from
four phyla Bacteroides, Firmicutes, Proteobacteria, and
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_8, © Springer Science+Business Media, LLC, part of Springer Nature 2019
123
124 Lisa A. Lai et al.
Fig. 1 Pie chart illustrates the taxonomy distribution from metaproteomics analysis of a human fecal sample.
The center circle represents the organism with each concentric circle moving outward depicting taxonomies
(i.e., domain, kingdom, phylum, class, order, family, genus, and species)
130 Lisa A. Lai et al.
References
Abstract
The classical proteomics approach for the identification of allergen candidates consists on the separation of
proteins by high-resolution two-dimensional electrophoresis (2-DE) with subsequent IgE immunoblotting
and further analysis of IgE-reactive protein spots with mass spectrometry. In this approach at least two gels
most be run. One gel is used for staining and the other is for immunoblotting by antibodies labeled with
specific immunostains. Additional functional characterizations require either protein purification or 2-DE
replicates and appear to be time- and reagent-consuming. Here we described a modified double
one-dimensional electrophoresis (D1-DE) allowing the conversion of a protein spot previously visualized
by 2-DE into an extended protein band. In D1-DE, the purity of the protein of interest is similar to 2-DE
spots, but its abundance is many times higher than what can be found in a 2-DE single spot allowing many
other functional analyses from a single D1-DE separation.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_9, © Springer Science+Business Media, LLC, part of Springer Nature 2019
133
134 Youcef Shahali et al.
2 Materials
3 Methods
3.1 Principles Like standard 2-DE, the D1-DE reported here consists of IEF as a
of D1-DE first dimension followed by SDS-PAGE. The difference between
the two techniques mainly resides in the migration axis of the
second separation. This means that an acidic, neutral, or basic
horizontal band from the first dimension (IEF) is transferred to
SDS-PAGE (Fig. 1). Therefore, proteins are isolated as a long
continuous band with purity comparable to 2-DE spots. In the
present protocol, the sequential one-dimensional combination of
IEF and SDS-PAGE followed by immunoblotting is described for
the IgE screening of patients sera under equal conditions. For a
better correlation between 2-DE and D1-DE patterns, the same
IEF gel could be used as the first dimension of both 2-DE and
D1-DE immunoblotting.
IEF
Mpl Sample +
Orthogonal
90°
Coaxial
2-DE D1-DE
+ -
Mr Mr
kDa - kDa -
94 94
67 67 2
1 2
3
45 45
3
30 30
20 20
4 4
14.4 14.4
+ +
Fig. 1 Schematic illustration of a D1-DE adapted from Shahali et al. (2012) [10]. Both 2-DE and D1-DE consist
of the sequential combination of IEF and SDS-PAGE. The difference between the two techniques resides in the
axis of migration of the second separation
4. Place the CleanGel IEF precast gel into the pool for rehydra-
tion by laying the edge of the CleanGel, with the gel surface
facing down into the rehydration buffer, slowly lower it in
order to avoid air bubbles.
5. Using forceps, lift the film at the edges up to the middle and
lower it again without catching air bubbles to distribute evenly
the liquid. Very even rehydration is also obtained by shaking
the GelPool at a slow rotation rate.
6. Meanwhile, switch on the cooling system (thermostatic circu-
lator or Peltier cooling plate) of the horizontal flatbed
apparatus.
7. Remove the gel from the Gelpool after 1 h making sure the
rehydration has been evenly achieved.
8. Remove the excess buffer from the gel surface using the edges
of a Whatman filter paper N 1, until the gel surface is
completely dry (see Note 1).
9. The rehydrated gel is now ready for IEF run.
10. Pipet a low amount of kerosene (about 2.5 mL) onto the
horizontal cooling plate (e.g., Multhiphor II apparatus) to
improve the cooling contact.
Double One-Dimensional Electrophoresis (D1-DE) Adapted for Immunoproteomics 137
11. Place the gel (gel surface up/gel film or GelBond down) onto
the center of the cooling plate.
12. Cut the electrode strips at the gel length (26 cm).
13. Align strips on the cathodal and anodal edges of the gels, the
electrode strips overlapping the gel-edge with about 3 mm.
14. Apply evenly the cathode and anode buffers (Serva IEF buffers,
Heidelberg, Germany) on the cathode and anode strips,
respectively.
15. Cut two 10-cm-long and up to 0.7-cm-wide strips of a dry
Whatmans N 1 paper (GE Healthcare) for sample application
on anode side.
16. Cut a small piece of Whatman N 1 paper (0.5 0.5 cm) for
IEF protein standards (pI marker) and place it directly onto the
upper center of the gel (anode side).
17. Use forceps to align the strips on both sides of the pI marker at
the same distance from the paper piece (minimum 1 cm).
18. Pipet 2 μL of pI marker (Bio-Rad) for wide or short range IEF
as comparative references.
19. Load samples (55–60 μg of proteins from the allergen extract)
using a micropipette on each 10-cm-long sample paper piece.
20. Clean platinum electrode wires with a wet tissue paper.
21. Then, move electrodes on anode/cathode strips to ensure a
complete overlapping between buffer strips, gel, and electrode
wires.
22. For Multiphor, connect the cables of the electrodes to the
electrophoresis power supply apparatus (EPS 3501 XL, GE
Healthcare). Finally lower and close the safety lid.
23. Select the running program and start the IEF migration (see
Note 2).
24. Stop the migration and wash the cathode and anode borders of
the gel with PBS pH 7.5.
25. Lift the film at the edges up to the middle to form a U shape
and pour promptly PBS on the middle of the gel.
3.3 Preparation 1. After IEF running, cut out the gel at the pI marker along with a
of D1-DE small part of the sample separation for Coomassie blue or silver
staining.
2. Lay the gel face down on a clean plastic film or a glass plate.
Mark the area that should be cut out for transferring. The size
of this area should be 10 cm long and up to 7 mm wide.
3. Cut out horizontally the IEF strip in the selected narrow pI
range containing the allergenic fraction of interest by reference
to pI markers and sample stained.
138 Youcef Shahali et al.
Fig. 2 D1-DE combined with IgE immunoblots (adapted from Shahali et al. 2012) [10]. (a) Represents the initial
IEF separation. The silver staining was performed after excision of the basic and neutral bands. IgE
immunoblots of the basic (b) and neutral (c) proteins of cypress pollen (CP) extracts probed with sera of
30 CP allergic patients: lanes 1–30; lane 31: healthy donor serum; lane 32: no serum (negative control). D1-DE
allowed the MS/MS characterization of an allergenic polygalacturonase (PG) of 43 kDa which overlapped
(in previous 1-DE experiments) with another CP major allergen belonging to the pectate lyase (PL) family
referenced as Cup s 1 (see Note 6)
20. After staining, the protein band of interest could be cut out,
digested, and submitted to mass spectrometry analyses (see
Note 5) [10, 14].
4 Notes
References
5. Poncet P, Sénéchal H, Clement G, Purohit A, of cypress pollen allergens using double and
Sutra JP, Desvaux FX et al (2010) Evaluation of triple one-dimensional electrophoresis.
ash pollen sensitization pattern using proteo- Electrophoresis 33:462–469
mic approach with individual sera from allergic 11. Altland K, Silke R, Hackler R (1981) Demon-
patients. Allergy 65:571–580 station of human prealbumin by double
6. D’Amato A, Bachi A, Fasoli E, Boschetti E, one-dimensional slab gel electrophoresis. Elec-
Peltre G, Sénéchal H et al (2010) In-depth trophoresis 2:148–155
exploration of Hevea brasiliensis latex prote- 12. Blum H, Beier H, Gross HJ (1987) Improved
ome and “hidden allergens” via combinatorial silver staining of plant proteins, RNA and DNA
peptide ligand libraries. J Proteome in polyacrylamide gels. Electrophoresis
73:1368–1380 8:93–99
7. Shahali Y, Sutra JP, Fasoli E, D’Amato A, Righ- 13. Demeulemester C, Peltre G, Laurent M,
etti PG, Futamura N et al (2012) Allergomic Panheleux D, David B (1987) Cyanogen
study of cypress pollen via combinatorial pep- bromide-activated nitrocellulose membranes:
tide ligand libraries. J Proteome 77:101–110 a new tool for immunoprint techniques. Elec-
8. Shahali Y, Sutra JP, Peltre G, Charpin D, trophoresis 8:71–73
Sénéchal H, Poncet P (2010) IgE reactivity to 14. Shahali Y, Sutra JP, Hilger C, Swiontek K,
common cypress (C. Sempervirens) pollen Haddad I, Vinh J et al (2017) Identification
extracts: evidence for novel allergens. W Allergy of a polygalacturonase (Cup s 2) as the major
Organ J 3:229–234 CCD-bearing allergen in Cupressus sempervi-
9. Shahali Y, Nicaise P, Brazdova A, Charpin D, rens pollen. Allergy 72:1806–1810
Scala E, Mari A et al (2014) Complementarity 15. Sénéchal H, Šantrůček J, Melčová M,
between microarray and immunoblot for the Svoboda P, Zı́dková J, Charpin D et al (2018)
comparative evaluation of IgE repertoire of A new allergen family involved in pollen food-
French and Italian cypress pollen allergic associated syndrome: Snakin/gibberellin-
patients. Folia Biol 60:192 regulated proteins. J Allergy Clin Immunol
10. Shahali Y, Sutra JP, Haddad I, Vinh J, 141:411–414
Guilloux L, Peltre G et al (2012) Proteomics
Chapter 10
Abstract
Biological activities are mainly executed by proteins and in most of the occasions these activities are
accomplished by protein complexes or through protein–protein interactions (PPI). So it is critical to reveal
how the protein complexes are organized and demonstrate the PPIs involved in the biological processes. In
addition to the traditional biochemical approaches, proximity-dependent labeling (PDL) has recently been
proposed to identify the interacting partners of a given protein. PDL requires the fusion expression of the
target protein with an enzyme which catalyzes the attachment of a reactive molecule to the interacting
partners in a distance-dependent manner. Further analysis of all the proteins that are modified by the
reactive molecule discloses the identity of these proteins which are presumed to be interacting partners of
the target protein. BioID is one of those representative PDL methods with the most widely applications.
The enzyme used in BioID is a biotin ligase BirA which catalyzes the biotinylation of target protein with the
presence of biotin. Through streptavidin-mediated pull-down and mass spectrometry analysis, the inter-
acting protein candidates of a given protein can be obtained.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_10, © Springer Science+Business Media, LLC, part of Springer Nature 2019
143
144 Peipei Li et al.
2 Materials
2.1 BioID Vector 1. Expression vectors for BirA* can be obtained from Addgene
(pcDNA3.1 MCS-BirA(R118G)-HA #36047 or pcDNA3.1
mycBioID #35700).
2. Cloned coding sequence of the interested protein.
2.2 Validation of 1. 293T or other appropriate cell lines and the appropriate cell
BioID Fusion Protein culture medium.
2. 1 mM Biotin: 12.2 mg biotin (Sigma) dissolved in 50 mL
H2O, sterilize by passing through a 0.22-μm syringe-driven
filter, store at 4 C.
3. HRP-streptavidin (Sigma).
4. Primary antibodies for BioID fusion protein (e.g., anti-HA/
MYC, anti-interest protein).
5. Streptavidin-Alexa Fluor.
6. DNA labeling reagent (e.g., DAPI).
146 Peipei Li et al.
3 Methods
3.1 Generation of In order to study the interacting proteins of a give protein, this
BioID Construct protein needs to be expressed in fusion with BirA*. The expression
vectors of BirA* have provided multiple cloning site and the target
protein coding sequence can be cloned into this site. The gene
clone strategy is not given in this protocol. The fusion protein
(now named as BioID vector) shouldn’t affect the function and
localization of the original protein, gene fragment inserted into
either the N- or C-terminus of biotin ligase should be carefully
considered. Additionally, the fusion protein may be observed to
ensure the proper intracellular localization and showing the
expected function.
NOTE: The BioID vector should contain HA tag or Myc tag
for easy detection of the fusion protein. Also, it is important to
select N-terminal or C-terminal for insertion of the target protein
because the fusion protein may be influenced functionally if the
insertion site hasn’t been carefully tested.
Fig. 1 Detection of biotinylated proteins by BioID in HEK293 cells expressing CtBP2-BirA*. (a) Cells expressing
CtBP2-BirA* and control cells were incubated for 24 h with and without 50 μM biotin, the expression of CtBP2-
BirA* was detected by anti-HA, and β-actin served as loading control. (b) Biotinylated proteins were identified
by HRP-streptavidin in different conditions. (c) Fluorescence microscopy was used to observe the biotinylated
proteins in CtBP2-BirA* overexpressed cells incubating for 24 h with 50 μM biotin, biotinylated proteins were
detected by streptavidin-Alexa Fluor 594 (red), DNA was detected with DAPI (blue)
17. For immunofluorescence assay, fix the cell with 4% PFA for
10 min at room temperature, then permeate the cells with 0.2%
TritonX-100 for 10 min.
18. Blocking the cells with 1% BSA for 0.5 h, followed by incuba-
tion of streptavidin-Alexa Fluor (1:1000) and DAPI, observe
the fluorescence by microscopy (Fig. 1c).
NOTE: BSA has better effect in getting rid of free biotin than
milk.
3.3 BioID Pull-Down This protocol describes the cells transiently expressing BioID fusion
Assay protein to process large-scale (6 107 cells) BioID pull-down assay
and LC-mass (Liquid chromatograph mass spectrometry) analysis.
Cells stably expressing BioID fusion protein are also subjected to
BioID pull-down as well.
1. Plating two 15-cm dishes for each experimental condition (cells
expressing BioID construct and control cells).
BioID: A Proximity-Dependent Labeling Approach in Proteomics Study 149
4 Summary
Acknowledgments
References
Abstract
Reverse-phase high-performance liquid chromatography is commonly employed as a decomplexing strat-
egy in snake venom proteomics. The chromatographic fractions often contain relatively pure toxins that can
be assessed functionally for toxicity level through the determination of their median lethal doses (LD50).
Further, antivenom efficacy can be evaluated specifically against these venom fractions to understand the
limitation of the antivenom as the treatment for snake envenomation. However, methods of toxicity
assessment and antivenom evaluation vary across laboratories; hence there is a need to standardize the
protocols and parameters, in particular those related to the neutralizing efficacy of antivenom. This chapter
outlines the important in vivo techniques and data interpretation that can be applied in the functional study
of snake venom proteomes.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_11, © Springer Science+Business Media, LLC, part of Springer Nature 2019
153
154 Choo Hock Tan and Kae Yi Tan
2 Materials
2.1 Snake Venom/ Lyophilize the snake venom or its toxin fractions (obtained from
Toxin Fractions protein decomplexation approach, Chapter 5). Store at 20 C
until use.
3 Methods
3.1 Lethality 1. Divide ICR mice into four groups (n ¼ 5 per group, 20–25 g
Determination of body weight). Each group of mice receives venom or toxin
Venom/Toxin Fraction fraction at different concentrations (treated as “doses,” see
below). Estimate the median lethal dose using at least four
doses of the venom or individual toxin fraction (see Note 1).
2. Sample preparation: Estimate the protein concentration of
venom/toxin fraction using bicinchoninic acid assay or Lowry
assay. Dilute the venom/toxin fraction with normal saline to
serial concentrations. Fix the volume of injection at 100 μL per
mouse.
3. Hold the mice using a rodent restrainer. Inject the appropri-
ately diluted venom/toxin fraction intravenously into the mice
via caudal vein (see Note 2).
4. Allow the mice access to food and water ad libitum. Monitor
and record the survival ratio of the mice in each group for 48 h
(see Note 3).
5. Determine the median lethal dose (LD50) of the venom/toxin
fraction using Probit analysis (see Note 4).
*Figure 1 shows the schematic drawing of the assessment of
median lethal dose (LD50) of venom/toxin fraction.
3.2 Lethality 1. Divide ICR mice into four groups (n ¼ 5 per group, 20–25 g
Neutralization of body weight). Each group of mice receives venom or toxin
Venom/Toxin Fraction fraction that has been preincubated with varying doses of anti-
venom (see below). Estimate the antivenom efficacy and
Fig. 1 A schematic diagram shows the assessment of median lethal dose (LD50) of venom or toxin fraction
(flow following red arrows) and neutralization efficacy as well as potency of antivenom (flow following green
arrows). The results are determined using Probit analysis based on the recorded survival ratio throughout the
experiment. The test samples (venom/toxin fraction and antivenom mixtures) are intravenously injected into
mice to ensure a full systemic access under a control titration
156 Choo Hock Tan and Kae Yi Tan
4 Notes
1. The initial starting dose for median lethal dose (LD50) deter-
mination can be estimated based on reported values for venoms
or toxin derived from closely related species.
2. The route of administration is intravenous to ensure a full
systemic access of the venom or toxin into the animal. This
enables the assessment and interpretation of the systemic tox-
icity of venom/toxin that becomes fully bioavailable to the
animal.
3. The survival ratios obtained from the lethality assay should
contain an upper dose which shows 100% death of mice
(n ¼ 5), a lower dose with 100% survival (n ¼ 5), and interme-
diate doses with a mix of death and survival.
4. Median lethal dose (LD50) is determined with Probit analysis
applying the Finney method.
5. If 200 μL of reconstituted antivenom failed to provide full
protection to the mice, a lower challenge dose (2.5 or 1.5
LD50) can be used. All challenge doses should be proven to be
above the lethal dose 100% (LD100) when injected intrave-
nously into the mice. This can be assessed in an additional
group of mice constituting the control.
6. The survival ratios obtained from the neutralization assay
should contain an upper dose which shows 100% survival of
mice (n ¼ 5), a lower dose with 100% death (n ¼ 5), and
intermediate doses with a mix of death and survival. Median
effective dose (ED50) is determined using Probit analysis.
7. The neutralization potency (P) is an indicator of antivenom
neutralizing capacity and is theoretically independent of the
dosage of challenge dose. This is because it takes into consider-
ation the dose of antivenom that is able to completely neutral-
ize the lethal effect of venom/toxin by subtracting 1 LD50
158 Choo Hock Tan and Kae Yi Tan
References
1. Calvete JJ (2013) Snake venomics: from the Laticauda colubrina from Bali: insights into
inventory of toxins to biology. Toxicon 75 subvenomic diversity, venom antigenicity and
(Suppl C):44–62 https://doi.org/10.1016/j. cross-neutralization by antivenom. J Proteome
toxicon.2013.03.020 166:48–58. https://doi.org/10.1016/j.jprot.
2. Tan KY, Tan CH, Fung SY, Tan NH (2015) 2017.07.002
Venomics, lethality and neutralization of Naja 8. Tan KY, Tan CH, Sim SM, Fung SY, Tan NH
kaouthia (monocled cobra) venoms from three (2016) Geographical venom variations of the
different geographical regions of Southeast Southeast Asian monocled cobra (Naja
Asia. J Proteome 120:105–125. https://doi. kaouthia): venom-induced neuromuscular
org/10.1016/j.jprot.2015.02.012 depression and antivenom neutralization.
3. Wong KY, Tan CH, Tan KY, Naeem QH, Tan Comp Biochem Physiol C Toxicol Pharmacol
NH (2018) Elucidating the biogeographical 185–186:77–86 https://doi.org/10.1016/j.
variation of the venom of Naja naja (specta- cbpc.2016.03.005
cled cobra) from Pakistan through a venom- 9. World Health Organization (2010) WHO
decomplexing proteomic study. J Proteome Guidelines for the production control and reg-
175:156–173. https://doi.org/10.1016/j. ulation of snake antivenom immunoglobulins.
jprot.2017.12.012 WHO publication, 1–141
4. Tan CH, Tan KY, Lim SE, Tan NH (2015) 10. Faisal T, Tan KY, Sim SM, Quraishi N, Tan
Venomics of the beaked sea snake, Hydrophis NH, Tan CH (2018) Proteomics, functional
schistosus: a minimalist toxin arsenal and its characterization and antivenom neutralization
cross-neutralization by heterologous antive- of the venom of Pakistani Russell’s viper
noms. J Proteome 126:121–130. https://doi. (Daboia russelii) from the wild. J Proteome
org/10.1016/j.jprot.2015.05.035 183:1–13. https://doi.org/10.1016/j.jprot.
5. Oh AMF, Tan CH, Ariaranee GC, Quraishi N, 2018.05.003
Tan NH (2017) Venomics of Bungarus caeru- 11. Tan KY, Tan CH, Fung SY, Tan NH (2016)
leus (Indian krait): comparable venom profiles, Neutralization of the principal toxins from the
variable immunoreactivities among specimens venoms of Thai Naja kaouthia and Malaysian
from Sri Lanka, India and Pakistan. J Proteome Hydrophis schistosus: insights into toxin-specific
164:1–18. https://doi.org/10.1016/j.jprot. neutralization by two different antivenoms.
2017.04.018 Toxins 8(4):86. https://doi.org/10.3390/
6. Tan CH, Tan KY, Yap MK, Tan NH (2017) toxins8040086
Venomics of Tropidolaemus wagleri, the sexu- 12. Wong KY, Tan CH, Tan NH (2016) Venom
ally dimorphic temple pit viper: unveiling a and purified toxins of the spectacled cobra
deeply conserved atypical toxin arsenal. Sci (Naja naja) from Pakistan: insights into toxic-
Rep 7:43237. https://doi.org/10.1038/ ity and antivenom neutralization. Am J Trop
srep43237 Med Hyg 94(6):1392–1399. https://doi.org/
7. Tan CH, Wong KY, Tan KY, Tan NH (2017) 10.4269/ajtmh.15-0871
Venom proteome of the yellow-lipped sea krait,
Chapter 12
Abstract
Secretomes from microorganisms growing on biomass contain carbohydrate-active enzymes (CAZymes) of
potential biotechnological interest. By analyzing such secretomes, we may discover key enzymes involved in
degradation processes and potentially infer the mode-of-action of biomass conversion. Some of these
enzymes may have predicted functions in carbohydrate degradation, while others may not, while yet
exhibiting a similar expression pattern; these latter enzymes constitute potential novel enzymes involved
in the degradation process and provide a basis for further biochemical exploration. Hence, secretomes
represent an important source for the study of both predicted and novel CAZymes. Here we describe a
plate-based culturing technique that allows for collection of protein fractions that are highly enriched for
secreted proteins, bound or unbound to the substrate, and which minimizes contamination by intracellular
proteins trough unwanted cell lysis.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_12, © Springer Science+Business Media, LLC, part of Springer Nature 2019
159
160 Tina R. Tuveng et al.
2 Materials
2.4 Data Integration 1. Computer with a spreadsheet application (e.g., Excel from
and Heat Map Microsoft, Washington, USA).
Generation 2. An installation of Perseus. Perseus is a free software package for
the analysis of (prote)omics data and can be downloaded from
http://www.coxdocs.org/doku.php?id¼perseus:start. The ver-
sion used in this chapter is 1.6.0.7.
3 Methods
Fig. 1 The figure shows an example of CAZyme prediction using the dbCAN web server. The FASTA-formatted
sequence of the chitin-binding protein (UniProt: B3PDT6) from Cellvibrio japonicus Ueda107 was used as
input. A table shows the HMM hits to different parts of the protein sequence and a graphical representation
shows the domain architecture within the protein
4. Click submit, and after some computation time, the results will
be presented as a table and as a graphical representation of the
domain architecture (Fig. 1). Note that several CAZy modules
may be found within one protein sequence, leading to multiple
rows in the table per protein.
5. Click “Download parsed output” to download a tab-separated
file containing the results. This can be used for integration with
proteomic expression values in a spreadsheet application, see
Subheading 3.4.
3.2 Predicting Several algorithms have been developed for in silico prediction of
Protein Secretion different N-terminal signal peptides and, consequently, the subcel-
lular locations of their cognate proteins. These prediction tools
have since the 1980s developed from signal peptide prediction
based on weight matrices and the presence of specific amino acid
motifs to more sophisticated machine learning approaches [14],
including also so-called leaderless proteins, i.e., secreted proteins
without a typical signal peptide. Today, a combination of several
prediction servers is often used to obtain the most reliable predic-
tions possible [15]. It is important to remember that in silico
prediction of the subcellular location of proteins is not trivial, and
that the presence of a signal peptide is not necessarily a guarantee
for secretion. There is a plethora of prediction servers available
[16–18], some applicable to all types of organisms, while others
are more “specialized” for, e.g., bacteria. In any case, the SignalP
server (http://www.cbs.dtu.dk/services/SignalP/ [19]) offers a
good starting point for prediction of signal peptides and is
164 Tina R. Tuveng et al.
3.2.4 Nonclassically Although most secreted proteins have a signal peptide that guides
Secreted Proteins them to a secretory pathway, a limited number of proteins without
such a signal peptide (also called leaderless proteins) are secreted
166 Tina R. Tuveng et al.
3.3 Culture Plates for Carry out all procedures at room temperature unless otherwise
Secretome Enrichment specified.
3.3.1 Casting of Plates 1. The weights and volumes used below allow for preparation of
with Membrane (Work in 250 mL M9 agarose medium, which will give approx. 15 plates.
Sterile Hood) 2. (a) Using an insoluble carbon source: To achieve a 1% (w/v)
concentration of carbon source in the final plates (see Note
4): Weigh in 2.5 g insoluble carbon source and mix with
2.5 g agarose in 199.5 mL ultrapure water. Include a
magnet for later homogenization (see step 9 and Note 5).
(b) Using a soluble carbon source: Weigh in 2.5 g agarose and
mix with 187 mL ultrapure water. The soluble carbon
source will be added to the medium in step 7.
3. Autoclave at 121 C for 20 min and cool down to approx.
70 C.
4. Add 250 μL 0.1 M CaCl2 (to achieve a final concentration of
0.1 mM).
5. Add 250 μL 1 M MgSO4 (to achieve a final concentration of
1 mM).
6. Add 50 mL 5 M9 salt (to achieve a final concentration of 1).
7. If using a soluble carbon source: To achieve 1% (w/v) concen-
tration of carbon source in the final plates (see Note 4): Add
12.5 mL of the sterile carbon source stock solution (given that
the concentration of the stock is 20% (w/v), see Subheading
2.2).
8. Mix gently to ensure homogenization.
9. Poor 8 mL M9 minimal agarose medium (see Note 5) in an
80 mm glass Petri dish and let it solidify for 5–10 min. While
waiting for the agarose to solidify, store the bottle with melted
agarose at 60 C.
CAZymes in Microbial Secretomes 167
10. Gently place a sterile QM-A Quartz Filter in the middle of the
dish and poor another 8 mL (see Note 5) of the M9 minimal
agarose medium over the filter. Let it solidify for 5–10 min.
11. Store plates at 4 C until use. Always let the plates equilibrate to
the temperature of interest before use.
3.3.3 Protein Extraction As with all proteomics work, it is important to work clean to avoid
and Sample Preparation contamination from fingers, hairs, and similar.
1. Weigh an empty 50 mL Falcon tube and note the weight.
2. Flip the agar disc and punch out the agar with the 50 mL
Falcon tube directly under the filter (Fig. 2, step 3). Transfer
the plug to the Falcon tube, weigh again, and calculate the
sample net weight by subtracting the weight of the empty
Falcon tube in step 1.
3. Add 10 μL of 400 mM DTT stock solution per g sample to
achieve a final concentration of 4 mM DTT (by assuming 1 g
sample ¼ 1 mL sample).
4. Heat the sample until agar melts (i.e., by placing the tube in an
80–100 C water bath) and vortex, then boil the sample for
30 min. In this step, proteins bound to the solid substrate are
likely to be released.
5. Cool the sample to room temperature (the agar resolidifies).
6. Use a 2 mL disposable syringe (without needle), remove the
plunger, and transfer the solid sample into the syringe from the
top. Insert the plunger and crush the agar by pressing it
through the syringe, back into the Falcon tube.
7. Add 1 mL of 100 mM NH4HCO3 stock solution per g sample
to achieve a final concentration of 50 mM NH4HCO3. Mix by
gentle vortexing.
168 Tina R. Tuveng et al.
Fig. 2 Growth of microbes on membrane-containing plates and secretome sampling. Step 1: An agar plug
containing fungus grown on a normal plate is collected using the back of a sterile pipette tip (1A) and
transferred to a membrane-containing plate with the same carbon source and positioned in the center of the
plate, i.e., over the membrane (1B). In the case of bacteria, one applies a cell suspension instead of an agar
plug. Step 2: Incubate plates for the desired time. The pictures show growth of the fungus Hypocrea jecorina.
Step 3: After incubation, the agar is flipped out of the Petri dish, thus exposing the cell free agar between the
bottom of the dish and the agar-embedded membrane (3A). Use a sterile Falcon tube (or the back of a sterile
pipette tip if a smaller sample is desirable) to punch out an agar disc containing secreted proteins that have
passed through the filter (3B–C). This figure is reproduced from [8] with permission from Elsevier
3.4.1 Building a Heat Perseus is a free software package for analyzing quantitative (prote)
Map Using Perseus omics data and can be used with many different quantification
techniques [33], see Note 11.
1. In Perseus, click on Generic matrix upload, a small green
arrow in the upper-left corner.
2. Select the tabular file containing the quantitative proteomics
data and the predicted functional data, similar to Table 1.
Perseus supports tab- or comma-delimited text files.
3. Columns containing quantitative values should be selected as
“Main” columns.
4. Select dbCAN and secretion predictions as “Categorical”
columns.
5. Select accession number and protein name as “Text” columns.
Click OK.
6. Click Annotation of rows, and select Categorical annotation
of rows. Give biological replicates the same name, and keep
default settings. Click OK.
7. If you have quantification values ¼ 0, meaning that the protein
was not detected in quantifiable amounts, click Quality and
170
Table 1
An example of how to structure quantitative proteomics data in a spreadsheet application
B3PDT6 Chitin-binding protein, Y SpI 0.90 Y AA10 32.9 32.8 33.3 33.7 33.2 33.6 28.2 27.5 28.3
putative, cbp33/10B CBM10
P14768 Endo-1,4-beta-xylanase A Y SpI 0.96 Y CBM2 26.8 26.8 27.5 33.4 32.6 34.0 25.8 ND 25.5
CBM10
GH10
B3PDV8 Pullulanase, putative, pul13B Y SpII 0.94 Y CBM48 29.4 30.3 31.5 29.6 29.3 28.7 25.3 26.0 25.7
GH13
B3PK74 Alpha-glucosidase, putative, Y SpI 0.58 Y GH97 25.6 24.0 24.5 23.9 23.9 24.0 ND ND ND
adg97B
B3PBG2 Pilin N TMH 0.93 N 31.0 30.5 30.9 30.2 30.2 30.7 30.6 30.4 30.8
B3PF53 Putative lipoprotein N SpII 0.90 Y 28.8 26.7 27.3 28.3 28.6 27.9 26.2 ND 25.8
B3PI93 SrpA-related protein N CYT 0.18 N 30.6 30.8 31.2 27.8 27.4 28.8 ND ND 24.0
The data describes selected proteins detected in the secretome of Cellvibrio japonicus Ueda107 growing on α- or β-chitin or on glucose. The quantitative values are log2-
transformed LFQ-values from the MaxQuant [37] software. The complete proteome FASTA file was used for predicting secreted proteins using different prediction servers (see
Subheading 3.2) and for predicting CAZymes (see Subheading 3.1). The table shows results for four expressed CAZymes with varying expression levels on the different substrates
and three non-CAZymes for comparison; the data is adapted from [9]. The column labeled “Secreted?” is based on the use of three prediction servers SignalP, LipoP, and
SecretomeP and was marked with Y when at least two algorithms predicted secretion. ND not detected, R1–3 replicate 1–3
CAZymes in Microbial Secretomes 171
Fig. 3 A heat map representation of quantitative proteomics data where every protein (row) has a CAZyme
annotation (see Subheading 3.1) and predicted cellular location (from LipoP, see Subheading 3.2). The figure is
a filtered subset of the complete data set, showing only proteins with a CAZy annotation. The CAZy annotations
are colored as indicated below the figure; GH glycoside hydrolase, CE carbohydrate esterase, PL polysaccha-
ride lyase, AA auxiliary activity, CBM carbohydrate-binding module. The heat map generated by Perseus was
further manually sectioned into six clusters based on similar expression patterns. The colors in the heat map
indicate protein abundance, ranging from high (red color, MaxQuant LFQ 5 1010) to low (green color,
MaxQuant LFQ 7 106). Table 1 shows an example of the data used for generating this figure. The figure is
reproduced from [9] with permission from John Wiley & Sons
CAZymes in Microbial Secretomes 173
Table 2
Table for calculating the secretome enrichment
4 Notes
References
1. Himmel ME, Xu Q, Luo Y, Ding S-Y, 2. Payne CM, Knott BC, Mayes HB, Hansson H,
Lamed R, Bayer EA (2010) Microbial enzyme Himmel ME, Sandgren M, Ståhlberg J, Beck-
systems for biomass conversion: emerging ham GT (2015) Fungal cellulases. Chem Rev
paradigms. Biofuels 1(2):323–341. https:// 115(3):1308–1448. https://doi.org/10.
doi.org/10.4155/bfs.09.25 1021/cr500351c
176 Tina R. Tuveng et al.
3. Benz JP, Chau BH, Zheng D, Bauer S, Glass 12. Yin Y, Mao X, Yang J, Chen X, Mao F, Xu Y
NL, Somerville CR (2014) A comparative sys- (2012) dbCAN: a web resource for automated
tems analysis of polysaccharide-elicited carbohydrate-active enzyme annotation.
responses in Neurospora crassa reveals carbon Nucleic Acids Res 40(Web Server issue):
source-specific cellular adaptations. Mol W445–W451. https://doi.org/10.1093/
Microbiol 91(2):275–299. https://doi.org/ nar/gks479
10.1111/mmi.12459 13. Park BH, Karpinets TV, Syed MH, Leuze MR,
4. Suzuki K, Suzuki M, Taiyoji M, Nikaidou N, Uberbacher EC (2010) CAZymes Analysis
Watanabe T (1998) Chitin binding protein Toolkit (CAT): web service for searching and
(CBP21) in the culture supernatant of Serratia analyzing carbohydrate-active enzymes in a
marcescens 2170. Biosci Biotechnol Biochem newly sequenced organism using CAZy data-
62(1):128–135. https://doi.org/10.1271/ base. Glycobiology 20(12):1574–1584.
bbb.62.128 https://doi.org/10.1093/glycob/cwq106
5. Takasuka TE, Book AJ, Lewin GR, Currie CR, 14. Caccia D, Dugo M, Callari M, Bongarzone I
Fox BG (2013) Aerobic deconstruction of cel- (2013) Bioinformatics tools for secretome
lulosic biomass by an insect-associated Strepto- analysis. Biochim Biophys Acta Proteins Pro-
myces. Sci Rep 3:1030. https://doi.org/10. teom 1834(11):2442–2453. https://doi.org/
1038/srep01030 10.1016/j.bbapap.2013.01.039
6. Siljam€aki P, Varmanen P, Kankainen M, 15. Desvaux M, Hebraud M, Talon R, Henderson
Sukura A, Savijoki K, Nyman TA (2014) Com- IR (2009) Secretion and subcellular localiza-
parative exoprotein profiling of different Staph- tions of bacterial proteins: a semantic awareness
ylococcus epidermidis strains reveals potential issue. Trends Microbiol 17(4):139–145.
link between nonclassical protein export and https://doi.org/10.1016/j.tim.2009.01.004
virulence. J Proteome Res 13(7):3249–3261. 16. Nielsen H (2017) Predicting secretory proteins
https://doi.org/10.1021/pr500075j with SignalP. In: Kihara D (ed) Protein func-
7. Adav SS, Cheow ESH, Ravindran A, Dutta B, tion prediction: methods and protocols.
Sze SK (2012) Label free quantitative proteo- Springer, New York, pp 59–73. https://doi.
mic analysis of secretome by Thermobifida fusca org/10.1007/978-1-4939-7015-5_6
on different lignocellulosic biomass. J Prote- 17. Nielsen H (2017) Protein sorting prediction.
ome 75(12):3694–3706. https://doi.org/10. In: Journet L, Cascales E (eds) Bacterial pro-
1016/j.jprot.2012.04.031 tein secretion systems: methods and protocols.
8. Bengtsson O, Arntzen MØ, Mathiesen G, Springer, New York, pp 23–57. https://doi.
Skaugen M, Eijsink VGH (2016) A novel pro- org/10.1007/978-1-4939-7033-9_2
teomics sample preparation method for secre- 18. Nielsen H (2016) Predicting subcellular locali-
tome analysis of Hypocrea jecorina growing on zation of proteins by Bioinformatic algorithms.
insoluble substrates. J Proteome In: Bagnoli F, Rappuoli R (eds) Protein and
131:104–112. https://doi.org/10.1016/j. sugar export and assembly in gram-positive
jprot.2015.10.017 bacteria. Springer International Publishing,
9. Tuveng TR, Arntzen MØ, Bengtsson O, Gard- Cham, pp 129–158. https://doi.org/10.
ner JG, Vaaje-Kolstad G, Eijsink VGH (2016) 1007/82_2015_5006
Proteomic investigation of the secretome of 19. Petersen TN, Brunak S, Heijne G, Nielsen H
Cellvibrio japonicus during growth on chitin. (2011) SignalP 4.0: discriminating signal pep-
Proteomics 16(13):1904–1914. https://doi. tides from transmembrane regions. Nat Meth-
org/10.1002/pmic.201500419 ods 8:785. https://doi.org/10.1038/nmeth.
10. Lombard V, Golaconda Ramulu H, Drula E, 1701
Coutinho PM, Henrissat B (2014) The 20. Juncker AS, Willenbrock H, Von Heijne G,
carbohydrate-active enzymes database (CAZy) Brunak S, Nielsen H, Krogh A (2003) Predic-
in 2013. Nucleic Acids Res 42(D1): tion of lipoprotein signal peptides in Gram-
D490–D495. https://doi.org/10.1093/nar/ negative bacteria. Protein Sci 12
gkt1178 (8):1652–1662. https://doi.org/10.1110/
11. Cantarel BL, Coutinho PM, Rancurel C, ps.0303703
Bernard T, Lombard V, Henrissat B (2009) 21. Rahman O, Cummings SP, Harrington DJ,
The Carbohydrate-Active EnZymes database Sutcliffe IC (2008) Methods for the bioinfor-
(CAZy): an expert resource for Glycoge- matic identification of bacterial lipoproteins
nomics. Nucleic Acids Res 37(Database): encoded in the genomes of Gram-positive bac-
D233–D238. https://doi.org/10.1093/nar/ teria. World J Microbiol Biotechnol 24
gkn663 (11):2377. https://doi.org/10.1007/
s11274-008-9795-2
CAZymes in Microbial Secretomes 177
Abstract
The mechanism underlying many biological phenotypes remains unknown despite the increasing availabil-
ity of whole genome and transcriptome sequencing. Direct measurement of changes in protein expression is
an attractive alternative and has the potential to reveal novel processes. Mass spectrometry has become the
standard method for proteomics, allowing both the confident identification and quantification of thousands
of proteins from biological samples. In this review, mass spectrometry-based proteomic methods and their
applications are described.
Key words Mass spectrometry, Proteomics, Quantitation, Label-free, Selective reaction monitoring,
MALDI
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_13, © Springer Science+Business Media, LLC, part of Springer Nature 2019
179
180 J. Robert O’Neill
Tissue Proteins
Proteins A BC D A BC Identifications
enzymatic
digestion
protein
Computational
Experimental
inference
Peptides
fractionation
Peptide IDs
*
tandem mass database
spectrometry search
7 Quantitative Proteomics
7.3 Quantitative Chemical labeling can take place at the protein or peptide level. The
Shotgun Proteomics use of stable carbon, hydrogen, and nitrogen isotopes allows differ-
Using Labeling ential labeling of amino acids such as Leucine, Lysine, and Arginine
that will remain biochemically identical but through their mass
7.3.1 Stable Isotope differences are resolvable as discrete spectral peaks. This approach,
Labeling of Amino Acids in termed Stable Isotopic Labeling of Amino Acids in Culture
Culture (SILAC) (SILAC), allows the proteins in mammalian cells in culture to be
isotopically labeled by the use of medium containing only “heavy”
amino acids [63]. A typical experiment would comprise one trea-
ted, “heavy”-labeled cell line and a control unlabeled, “light”, cell
line. Cell lysates are mixed in a 1:1 ratio and then subjected to
standard LC-MS/MS workflow. Peptides are identified in the usual
fashion and the relative expression between cell line conditions
identified at the MS1 level by the ratio of heavy to light peptide
ion intensities. This approach has been shown to be reproducible
across the proteome with a coefficient of variation of ~30% [62]. By
using both heavy lysine and heavy arginine combinations, three
conditions can be compared simultaneously.
A disadvantage of SILAC approaches is the requirement for
complete label uptake by cultured cells, which limits the application
to cells which express stable phenotypes of interest across several
passages. The requirement for prior labeling in the conventional
SILAC method also precludes the study of human tissues samples
although fully isotopically labeled organisms have been described
which may have application in disease models [64–66].
7.3.2 Super-SILAC A variation of the SILAC method, termed super-SILAC, has been
applied to quantify the proteome of human cancer samples [67]. In
this procedure a mixture of cell lines derived from the cancer tissue
of interest and approximately covering the expression profile of the
tissue of interest are heavy-labeled using the SILAC method. A
mixture of lysates from these cell lines with a defined protein mass
is spiked-in to each tissue lysate in a 1:1 ratio before digestion,
fractionation, and LC-MS/MS using standard procedures. Peptide
identification and quantitation then proceeds as for a standard
SILAC experiment. The ratio of expression between heavy and
light peptides is calculated for each tissue sample. The constant
SILAC spike-in mass provides a method of normalizing between
experimental runs and also, by calculating the ratio of ratios, allows
the relative expression between tissue types to be calculated [68].
MS-Based Proteomics 187
7.3.3 Isotope-Coded In this method, the cysteine residues of reduced proteins are labeled
Affinity Tags (ICAT) with tags comprising a composite of a sulfhydryl reacting group, a
deuterated linker, and a biotin affinity tag [69]. Proteins from
discrete samples can be differentially labeled as both “light” and
“heavy” isotopes of the linker are available. Labeled samples are
then pooled and digested together. Cysteine-containing peptides
are then enriched by avidin-affinity chromatography. Peptides can
then be further fractionated or directly subjected to LC-MS/MS.
The different isotopes of the deuterated linker provide discrete
mass peaks during MS1 analysis to allow differential expression
analysis.
Unfortunately only cysteine-containing proteins can be stud-
ied, limiting proteome-wide efforts and the bulky affinity group,
biotin, introduces significant background into the MS/MS spectra
[70]. Furthermore, deuterated labels are more hydrophobic and
therefore are differentially eluted during reverse phase LC, compli-
cating the MS analysis [71]. This technique still has a role, however,
as the affinity enrichment step allows the study of low-abundance
proteins, not easily accessible by other methods.
18
7.3.4 O Labeling An advantage of this strategy is that it can be applied to almost any
sample. In an approach that predates the SILAC method, samples
for comparison are either proteolytically digested in 18O-contain-
ing water for the “heavy” sample or standard “light” water [72]. As
the protease, in most cases trypsin, cleaves the peptide bonds, the
heavy isotope is incorporated, so all tryptic peptides will be labeled.
The subsequent data analysis is identical to SILAC methods. A
disadvantage of this approach is the relative expense of H218O.
7.3.5 Dimethyl Isotopic A further method uses standard and deuterium isotopes of formal-
Labeling dehyde to label the amino-terminus of peptides or the amino group
of Lysine residues [73]. The isotopes are subsequently resolved by
their mass differences allowing peptide-level quantitation from the
MS1 scan. A further limitation common to SILAC, 18O, and
Dimethyl labeling is that maximum of three samples can be com-
pared per mass spectrometry analysis.
188 J. Robert O’Neill
O
N N
N O
O
Mass 114-117 Da Mass 28-31 Da
m/z )
m/z
m/z
m/z N (+0)
Mass 145 Da
Fig. 2 Schematic of the four-plex iTRAQ peptide label. A reporter group with a
defined mass between 114 and 117 Da is connected to a balancing linker.
Together the reporter and linkers have a fixed mass of 145 Da and they are
connected to an amine-reactive group which binds peptide amino-termini and
lysine residues. The label is cleaved at the balancing linker during MS2 fragment
ion generation to allow reporter ion detection. Figure adapted from [74]
7.3.6 Isobaric Peptide Isobaric peptide labels offer greater multiplexing capabilities with
Labeling 4-plex or 8-plex (Isobaric Tag for Relative and Absolute Quantifi-
cation; iTRAQ) [74] or 6-plex, 10-plex or 11-plex (Tandem Mass
Tags; TMT) commercial kits available [75]. These kits all rely on
the same underlying principle.
Each label consists of an amine-reactive ester, a balancing car-
bonyl linker, and a reporter ion (Fig. 2). Tryptic peptides form
amide linkages with the labels via N-termini or lysine residues. A
label with a different reporter is used for each different sample and
all the samples are mixed prior to fractionation and LC-MS/MS.
Each label has the same total mass and chromatographic properties
and therefore the LC retention time and mass/charge (m/z) sepa-
ration of each sample are not differentially affected during the MS1
scan [74].
Precursor ions are then sampled for MS/MS analysis and the
ionized-labeled peptides are fragmented with dissociation of the
reporter ions from the balancing carbonyl linker. The peptide frag-
ments are detected generating mass spectra in the usual manner.
The reporter ions are also detected as peaks at a predefined m/z.
For a four-plex iTRAQ experiment the reporters are detected at
114.1, 115.1, 116.1, and 117.1 m/z [74]. For a six-plex TMT
MS-Based Proteomics 189
7.4 Label-Free All sample manipulation steps during a proteomic workflow reduce
Quantitative Shotgun the data yield due to loss of proteins [77] and are additional sources
Proteomics of variation [78]. Eliminating the sample processing steps to incor-
porate labels for quantitation is clearly an advantage and underlies
the rationale to develop label-free methods of quantitation.
The total number of spectra matched to each peptide contri-
buting to a protein identification, termed the spectral count, has
been reported to correlate with absolute protein abundance
[79]. Various methods have been proposed to refine the spectral
count such as normalizing for protein length [80], or combination
scores including peptide count and fragment-ion intensity
[81]. For complex protein mixtures, spectral counts are still subject
to significant between-run variability and are highly dependent on
LC conditions and precursor ion selection. As a result, the quanti-
tative reproducibility of spectral counting is inferior to isobaric
labeling methods [82].
An alternative relies on the capture of precursor ion intensity as
a function of time to produce an ion chromatogram. The area
under the ion chromatogram curve is linearly proportional to the
peptide concentration [83]. Challenges exist in applying this
method across LC-MS/MS runs to allow differential analysis as
the same peptide ion must be identified and quantified despite
background noise, co-eluting peptides causing signal overlap, tech-
nical variations in retention time and total protein loading among
other factors [84].
A simultaneous advantage and disadvantage of both label-free
approaches is the requirement to analyze one sample per LC-MS/
MS run. This prevents sample dilution, provides maximum poten-
tial coverage, and prevents the potential failure to identify
190 J. Robert O’Neill
8 MALDI-Imaging MS (MALDI-IMS)
9 Conclusion
References
1. Wasinger VC, Cordwell SJ, Cerpa-Poljak A, 7. Gaudet P, Michel PA, Zahn-Zabal M, Britan A,
Yan JX, Gooley AA, Wilkins MR, Duncan Cusin I, Domagalski M, Duek PD, Gateau A,
MW, Harris R, Williams KL, Humphery- Gleizes A, Hinard V, Rech de Laval V, Lin J,
Smith I (1995) Progress with gene-product Nikitin F, Schaeffer M, Teixeira D, Lane L,
mapping of the Mollicutes: mycoplasma geni- Bairoch A (2017) The neXtProt knowledge-
talium. Electrophoresis 16(7):1090–1094 base on human proteins: 2017 update. Nucleic
2. Aebersold R, Mann M (2003) Mass Acids Res 45(D1):D177–D182. https://doi.
spectrometry-based proteomics. Nature 422 org/10.1093/nar/gkw1062
(6928):198–207. https://doi.org/10.1038/ 8. Saiki RK, Gelfand DH, Stoffel S, Scharf SJ,
nature01511 Higuchi R, Horn GT, Mullis KB, Erlich HA
3. de Godoy LM, Olsen JV, Cox J, Nielsen ML, (1988) Primer-directed enzymatic amplifica-
Hubner NC, Fröhlich F, Walther TC, Mann M tion of DNA with a thermostable DNA poly-
(2008) Comprehensive mass-spectrometry- merase. Science 239(4839):487–491
based proteome quantification of haploid ver- 9. Liu L, Li Y, Li S, Hu N, He Y, Pong R, Lin D,
sus diploid yeast. Nature 455 Lu L, Law M (2012) Comparison of next-
(7217):1251–1254. https://doi.org/10. generation sequencing systems. J Biomed Bio-
1038/nature07341 technol 2012:251364. https://doi.org/10.
4. Consortium IHGS (2004) Finishing the 1155/2012/251364
euchromatic sequence of the human genome. 10. Pareek CS, Smoczynski R, Tretyn A (2011)
Nature 431(7011):931–945. https://doi.org/ Sequencing technologies and genome
10.1038/nature03001 sequencing. J Appl Genet 52(4):413–435.
5. Legrain P, Aebersold R, Archakov A, https://doi.org/10.1007/s13353-011-0057-
Bairoch A, Bala K, Beretta L, Bergeron J, x
Borchers CH, Corthals GL, Costello CE, 11. Niall HD (1973) Automated Edman degrada-
Deutsch EW, Domon B, Hancock W, He F, tion: the protein sequenator. Methods Enzy-
Hochstrasser D, Marko-Varga G, Salekdeh mol 27:942–1010
GH, Sechi S, Snyder M, Srivastava S, 12. Sperling J, Azubel M, Sperling R (2008) Struc-
Uhlén M, Wu CH, Yamamoto T, Paik YK, ture and function of the Pre-mRNA splicing
Omenn GS (2011) The human proteome proj- machine. Structure 16(11):1605–1615.
ect: current state and future direction. Mol Cell https://doi.org/10.1016/j.str.2008.08.011
Proteomics 10(7):M111.009993. https://doi. 13. Ermini L, Olivieri C, Rizzi E, Corti G,
org/10.1074/mcp.M111.009993 Bonnal R, Soares P, Luciani S, Marota I, De
6. Omenn GS, Lane L, Lundberg EK, Overall Bellis G, Richards MB, Rollo F (2008) Com-
CM, Deutsch EW (2017) Progress on the plete mitochondrial genome sequence of the
HUPO Draft Human Proteome: 2017 Metrics Tyrolean iceman. Curr Biol 18
of the Human Proteome Project. J Proteome (21):1687–1693. https://doi.org/10.1016/j.
Res 16:4281. https://doi.org/10.1021/acs. cub.2008.09.028
jproteome.7b00375
192 J. Robert O’Neill
14. Elguoshy A, Hirao Y, Xu B, Saito S, Quadery top down LC MS/MS and versatile peptide
AF, Yamamoto K, Mitsui T, Yamamoto T, fragmentation modes. Mol Cell Proteomics
JProS CXPTo (2017) Identification and valida- 11(3):O111.013698. https://doi.org/10.
tion of human missing proteins and peptides in 1074/mcp.O111.013698
public proteome databases: data mining strat- 21. Gillet LC, Navarro P, Tate S, Röst H,
egy. J Proteome Res 16:4403. https://doi. Selevsek N, Reiter L, Bonner R, Aebersold R
org/10.1021/acs.jproteome.7b00423 (2012) Targeted data extraction of the
15. Thul PJ, Åkesson L, Wiking M, Mahdessian D, MS/MS spectra generated by data-
Geladaki A, Ait Blal H, Alm T, Asplund A, independent acquisition: a new concept for
Björk L, Breckels LM, B€ackström A, consistent and accurate proteome analysis.
Danielsson F, Fagerberg L, Fall J, Gatto L, Mol Cell Proteomics 11(6):O111.016717.
Gnann C, Hober S, Hjelmare M, https://doi.org/10.1074/mcp.O111.016717
Johansson F, Lee S, Lindskog C, Mulder J, 22. Souza GH, Guest PC, Martins-de-Souza D
Mulvey CM, Nilsson P, Oksvold P, (2017) LC-MS(E), multiplex MS/MS, ion
Rockberg J, Schutten R, Schwenk JM, Siverts- mobility, and label-free quantitation in clinical
son Å, Sjöstedt E, Skogs M, Stadler C, Sullivan proteomics. Methods Mol Biol 1546:57–73.
DP, Tegel H, Winsnes C, Zhang C, https://doi.org/10.1007/978-1-4939-6730-
Zwahlen M, Mardinoglu A, Pontén F, von 8_4
Feilitzen K, Lilley KS, Uhlén M, Lundberg E 23. Hardman M, Makarov AA (2003) Interfacing
(2017) A subcellular map of the human prote- the orbitrap mass analyzer to an electrospray
ome. Science 356(6340):eaal3321. https:// ion source. Anal Chem 75(7):1699–1705
doi.org/10.1126/science.aal3321
24. Krutchinsky AN, Kalkum M, Chait BT (2001)
16. Espina V, Edmiston KH, Heiby M, Automatic identification of proteins with a
Pierobon M, Sciro M, Merritt B, Banks S, MALDI-quadrupole ion trap mass spectrome-
Deng J, VanMeter AJ, Geho DH, Pastore L, ter. Anal Chem 73(21):5066–5077
Sennesh J, Petricoin EF, Liotta LA (2008) A
portrait of tissue phosphoprotein stability in 25. Johnson RS, Martin SA, Biemann K, Stults JT,
the clinical tissue procurement process. Mol Watson JT (1987) Novel fragmentation pro-
Cell Proteomics 7(10):1998–2018. https:// cess of peptides by collision-induced decompo-
doi.org/10.1074/mcp.M700596-MCP200 sition in a tandem mass spectrometer:
differentiation of leucine and isoleucine. Anal
17. Aebersold R, Mann M (2016) Mass- Chem 59(21):2621–2625
spectrometric exploration of proteome struc-
ture and function. Nature 537 26. Hughes C, Ma B, Lajoie GA (2010) De novo
(7620):347–355. https://doi.org/10.1038/ sequencing methods in proteomics. Methods
nature19949 Mol Biol 604:105–121. https://doi.org/10.
1007/978-1-60761-444-9_8
18. Hu Q, Noll RJ, Li H, Makarov A, Hardman M,
Graham Cooks R (2005) The Orbitrap: a new 27. Johnson RS, Taylor JA (2002) Searching
mass spectrometer. J Mass Spectrom 40 sequence databases via de novo peptide
(4):430–443. https://doi.org/10.1002/jms. sequencing by tandem mass spectrometry.
856 Mol Biotechnol 22(3):301–315. https://doi.
org/10.1385/MB:22:3:301
19. Collins BC, Hunter CL, Liu Y, Schilling B,
Rosenberger G, Bader SL, Chan DW, Gibson 28. Nesvizhskii AI, Vitek O, Aebersold R (2007)
BW, Gingras AC, Held JM, Hirayama-Kurogi- Analysis and validation of proteomic data gen-
M, Hou G, Krisp C, Larsen B, Lin L, Liu S, erated by tandem mass spectrometry. Nat
Molloy MP, Moritz RL, Ohtsuki S, Methods 4(10):787–797. https://doi.org/
Schlapbach R, Selevsek N, Thomas SN, Tzeng 10.1038/nmeth1088
SC, Zhang H, Aebersold R (2017) Multi- 29. Sadygov RG, Cociorva D, Yates JR (2004)
laboratory assessment of reproducibility, quali- Large-scale database searching using tandem
tative and quantitative performance of mass spectra: looking up the answer in the
SWATH-mass spectrometry. Nat Commun 8 back of the book. Nat Methods 1
(1):291. https://doi.org/10.1038/s41467- (3):195–202. https://doi.org/10.1038/
017-00249-5 nmeth725
20. Michalski A, Damoc E, Lange O, Denisov E, 30. Kusebauch U, Campbell DS, Deutsch EW,
Nolting D, Müller M, Viner R, Schwartz J, Chu CS, Spicer DA, Brusniak MY, Slagel J,
Remes P, Belford M, Dunyach JJ, Cox J, Sun Z, Stevens J, Grimes B, Shteynberg D,
Horning S, Mann M, Makarov A (2012) Hoopmann MR, Blattmann P, Ratushny AV,
Ultra high resolution linear ion trap Orbitrap Rinner O, Picotti P, Carapito C, Huang CY,
mass spectrometer (Orbitrap Elite) facilitates Kapousouz M, Lam H, Tran T, Demir E,
MS-Based Proteomics 193
Aitchison JD, Sander C, Hood L, Aebersold R, an LTQ Orbitrap mass spectrometer. Mol Cell
Moritz RL (2016) Human SRMAtlas: a Proteomics 7(9):1702–1713. https://doi.
resource of targeted assays to quantify the com- org/10.1074/mcp.M800029-MCP200
plete human proteome. Cell 166(3):766–778. 40. Garbis SD, Roumeliotis TI, Tyritzis SI, Zorpas
https://doi.org/10.1016/j.cell.2016.06.041 KM, Pavlakis K, Constantinides CA (2011) A
31. Picotti P, Clément-Ziza M, Lam H, Campbell novel multidimensional protein identification
DS, Schmidt A, Deutsch EW, Röst H, Sun Z, technology approach combining protein size
Rinner O, Reiter L, Shen Q, Michaelson JJ, exclusion prefractionation, peptide zwitterion-
Frei A, Alberti S, Kusebauch U, Wollscheid B, ion hydrophilic interaction chromatography,
Moritz RL, Beyer A, Aebersold R (2013) A and nano-ultraperformance RP
complete mass-spectrometric map of the yeast chromatography/nESI-MS2 for the in-depth
proteome applied to quantitative trait analysis. analysis of the serum proteome and phospho-
Nature 494(7436):266–270. https://doi.org/ proteome: application to clinical sera derived
10.1038/nature11835 from humans with benign prostate hyperplasia.
32. Dasari S, Chambers MC, Martinez MA, Car- Anal Chem 83(3):708–718. https://doi.org/
penter KL, Ham AJ, Vega-Montoto LJ, Tabb 10.1021/ac102075d
DL (2012) Pepitome: evaluating improved 41. Michalski A, Cox J, Mann M (2011) More than
spectral library search for identification com- 100,000 detectable peptide species elute in sin-
plementarity and quality assessment. J Prote- gle shotgun proteomics runs but the majority is
ome Res 11(3):1686–1695. https://doi.org/ inaccessible to data-dependent LC-MS/MS. J
10.1021/pr200874e Proteome Res 10(4):1785–1793. https://doi.
33. Lam H (2011) Building and searching tandem org/10.1021/pr101060v
mass spectral libraries for peptide identifica- 42. Panchaud A, Scherl A, Shaffer S, Haller P,
tion. Mol Cell Proteomics 10(12): Kulasekara H, Miller SI, Goodlett DR (2009)
R111.008565. https://doi.org/10.1074/ Precursor acquisition independent from ion
mcp.R111.008565 count: how to dive deeper into the proteomics
34. Jmeian Y, El Rassi Z (2009) Liquid-phase- ocean. Anal Chem 81:6481–6488
based separation systems for depletion, prefrac- 43. Scherl A, Shaffer SA, Taylor GK, Kulasekara
tionation and enrichment of proteins in HD, Miller SI, Goodlett DR (2008)
biological fluids for in-depth proteomics analy- Genome-specific gas-phase fractionation strat-
sis. Electrophoresis 30(1):249–261. https:// egy for improved shotgun proteomic profiling
doi.org/10.1002/elps.200800639 of proteotypic peptides. Anal Chem 80
35. Boisvert FM, Lam YW, Lamont D, Lamond AI (4):1182–1191. https://doi.org/10.1021/
(2010) A quantitative proteomics analysis of ac701680f
subcellular proteome localization and changes 44. Panchaud A, Jung S, Shaffer SA, Aitchison JD,
induced by DNA damage. Mol Cell Proteomics Goodlett DR (2011) Faster, quantitative, and
9(3):457–470. https://doi.org/10.1074/ accurate precursor acquisition independent
mcp.M900429-MCP200 from ion count. Anal Chem 83
36. Chenau J, Michelland S, Sidibe J, Seve M (6):2250–2257. https://doi.org/10.1021/
(2008) Peptides OFFGEL electrophoresis: a ac103079q
suitable pre-analytical step for complex eukary- 45. Savaryn JP, Catherman AD, Thomas PM, Abe-
otic samples fractionation compatible with cassis MM, Kelleher NL (2013) The emer-
quantitative iTRAQ labeling. Proteome Sci gence of top-down proteomics in clinical
6:9. https://doi.org/10.1186/1477-5956-6- research. Genome Med 5(6):53. https://doi.
9 org/10.1186/gm457
37. Batth TS, Olsen JV (2016) Offline high pH 46. Tran JC, Zamdborg L, Ahlf DR, Lee JE,
reversed-phase peptide fractionation for deep Catherman AD, Durbin KR, Tipton JD,
phosphoproteome coverage. Methods Mol Vellaichamy A, Kellie JF, Li M, Wu C, Sweet
Biol 1355:179–192. https://doi.org/10. SM, Early BP, Siuti N, LeDuc RD, Compton
1007/978-1-4939-3049-4_12 PD, Thomas PM, Kelleher NL (2011)
38. Boersema PJ, Mohammed S, Heck AJ (2008) Mapping intact protein isoforms in discovery
Hydrophilic interaction liquid chromatogra- mode using top-down proteomics. Nature 480
phy (HILIC) in proteomics. Anal Bioanal (7376):254–258. https://doi.org/10.1038/
Chem 391(1):151–159. https://doi.org/10. nature10575
1007/s00216-008-1865-7 47. Catherman AD, Durbin KR, Ahlf DR, Early
39. Bantscheff M, Boesche M, Eberhard D, BP, Fellers RT, Tran JC, Thomas PM, Kelleher
Matthieson T, Sweetman G, Kuster B (2008) NL (2013) Large-scale top-down proteomics
Robust and sensitive iTRAQ quantification on of the human proteome: membrane proteins,
194 J. Robert O’Neill
using liquid chromatography and mass spec- of peptides and proteins using MALDI-TOF
trometry. J Proteome Res 1(4):317–323 MS. Anal Chem 69(23):4751–4760
84. Listgarten J, Emili A (2005) Statistical and 88. Aichler M, Walch A (2015) MALDI Imaging
computational methods for comparative pro- mass spectrometry: current frontiers and per-
teomic profiling using liquid chromatography- spectives in pathology research and practice.
tandem mass spectrometry. Mol Cell Proteo- Lab Investig 95(4):422–431. https://doi.
mics 4(4):419–434. https://doi.org/10. org/10.1038/labinvest.2014.156
1074/mcp.R500005-MCP200 89. Minerva L, Clerens S, Baggerman G, Arckens L
85. Hanahan D, Weinberg RA (2011) Hallmarks (2008) Direct profiling and identification of
of cancer: the next generation. Cell 144 peptide expression differences in the pancreas
(5):646–674. https://doi.org/10.1016/j.cell. of control and ob/ob mice by imaging mass
2011.02.013 spectrometry. Proteomics 8(18):3763–3774.
86. Gerlinger M, Rowan AJ, Horswell S, Larkin J, https://doi.org/10.1002/pmic.200800237
Endesfelder D, Gronroos E, Martinez P, 90. Balluff B, Schöne C, Höfler H, Walch A (2011)
Matthews N, Stewart A, Tarpey P, Varela I, MALDI imaging mass spectrometry for direct
Phillimore B, Begum S, McDonald NQ, tissue analysis: technological advancements and
Butler A, Jones D, Raine K, Latimer C, Santos recent applications. Histochem Cell Biol 136
CR, Nohadani M, Eklund AC, Spencer- (3):227–244. https://doi.org/10.1007/
Dene B, Clark G, Pickering L, Stamp G, s00418-011-0843-x
Gore M, Szallasi Z, Downward J, Futreal PA, 91. Perkins DN, Pappin DJ, Creasy DM, Cottrell
Swanton C (2012) Intratumor heterogeneity JS (1999) Probability-based protein identifica-
and branched evolution revealed by multire- tion by searching sequence databases using
gion sequencing. N Engl J Med 366 mass spectrometry data. Electrophoresis 20
(10):883–892. https://doi.org/10.1056/ (18):3551–3567. https://doi.org/10.1002/(
NEJMoa1113205 SICI)1522-2683(19991201)20:18<3551::
87. Caprioli RM, Farmer TB, Gile J (1997) Molec- AID-ELPS3551>3.0.CO;2-2
ular imaging of biological samples: localization
Chapter 14
Abstract
The biological activities of a cell are determined by its response to external stimuli. The signals are
transduced from either intracellular or extracellular milieu through networks of multi-protein complexes
and post-translational modifications of proteins (PTMs). Most PTMs including phosphorylation, acetyla-
tion, ubiquitination, and SUMOylation, among others, modulate activities of proteins and regulate
biological processes such as proliferation, differentiation, as well as host pathogen interaction. Convention-
ally, reverse genetics analysis and single molecule-based studies were employed to identify and characterize
the function of PTMs and enzyme-substrate networks regulated by them. With the advent of high-
throughput technologies, it is now possible to identify and quantify thousands of PTM sites in a single
experiment. Here, we discuss recent advances in enrichment strategies of various PTMs. We also describe a
method for the identification and relative quantitation of proteins using a tandem mass tag labeling
approach combined with serial enrichment of phosphorylation, acetylation and succinylation using anti-
body enrichment strategy.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_14, © Springer Science+Business Media, LLC, part of Springer Nature 2019
197
198 Sneha M. Pinto et al.
1.1.1 Antibody-Based One of the widely employed methodology to assay PTMs of inter-
Enrichment methods est is the use of antibody-based enrichment strategies coupled with
LC-MS/MS analysis. The most common as well as the most popu-
lar PTM assayed includes the use of antibodies that specifically
enrich for tyrosine [12–14] or serine/threonine phosphorylated
proteins [15]. A detailed review of this topic has been provided
by Harsha et al. [16]. Antibody-based enrichment strategies have
now been extended to profile and quantify other PTMs including
ubiquitination, SUMOylation, lysine modifications such as acetyla-
tion, succinylation, and malonlylation, among others. Further-
more, Cell Signaling Technologies and PTM Biolabs have also
developed antibodies targeting methylation, crotonylation [17],
and modifications on lysine and arginine residues [18]. In addition
to pan-PTM-specific antibodies, antibodies specific to PTM motifs
have also been employed to identify and quantify substrates [19].
1.1.2 Metal-Ion-Based The alternative approach for enrichment of PTMs especially phos-
Enrichment Strategies phorylation moieties are chromatographic methods based on metal
affinity enrichment referred as immobilized metal affinity chroma-
tography (IMAC) and titanium dioxide (TiO2) based enrichment
[20]. Metal ions such as Fe(3+), Zr(4+), and Ga(3+) are employed
in IMAC, which is immobilized on beads and covalently bound to
negatively charged phosphopeptides [21–23]. TiO2 also works on a
similar principle and both approaches are employed in combination
with chromatography-based separation techniques such as strong
cation exchange chromatography and high pH reverse phase chro-
matography [14, 16, 24]. Sequential enrichment using IMAC
followed by TiO2 has also been reported to predominantly enrich
for monophosphorylated peptides [25].
1.1.3 Chemical With advances in chemical biology, PTM enrichment using chemi-
Proteomics cal tagging or derivatization has enabled detection and enrichment
of several PTMs including N-linked and O-linked glycosylation
[26, 27], acylation [28], palmitoylation [29, 30], and S-nitrosyla-
tion. The tagging methods include both in vitro and in vivo label-
ing of small chemical tag at the site of the modification and
subsequent enrichment using a capture reagent such as biotin. In
case of enrichment and separation of N-linked glycopeptides,
boronic acid chemistry, hydrazide chemistry, and alkyne click
chemistry, among others, have been employed [31]. A detailed
description of advances in chemical proteomics to study the
PTMome has been described by Tate [32].
1.1.4 Serial Enrichment It is known that proteins can be simultaneously modified by multi-
of PTMs ple PTMs and these PTMs may form the basis of regulatory cross
talk events [33]. Therefore, it is ideal to simultaneously study
numerous PTMs in a single experiment. This can be achieved by
carrying out PTM enrichment in a serial manner. Serial enrichment
200 Sneha M. Pinto et al.
1.2 Mass Over the years, mass spectrometry-based proteomic analysis has
Spectrometry to considerably increased our understanding of the occurrence and
Characterize Multiple dynamics of protein PTMs. Following PTM enrichment, sequence
Post-translational identification and confident site localization is vital. In case of
Modifications bottom-up approach which involves proteolytic digestion of pro-
teins followed by enrichment of PTMs, information on MS2 frag-
mentation provides data that is being used to deduce the sequence
as well as identify the site of modification. The fragmentation
approaches include collision-induced dissociation (CID) and
higher energy collisional dissociation (HCD), both of which gen-
erate predominantly b- and y-type ions and are universally
employed for global protein profiling as well as for the analyses of
stable PTMs. In the CID mode, fragmentation and data collection
occurs in ion trap at a faster acquisition speed resulting in higher
number of data points. On the contrary, HCD mode of fragmenta-
tion results in Fourier transform detection of the MS2 fragments in
the Orbitrap mass analyzer with data being acquired at a higher
resolution albeit lower scan rate. In case of labile PTMs such as
acetylation and glycosylation, electron transfer dissociation (ETD)
has been described as a preferred method for data acquisition
[36]. The mass accuracy of mass spectrometers can affect the iden-
tification and accurate site localization. Mass accuracy can also
significantly impact distinction of isobaric/indistinguishable mass
PTMs when mass analyzers such as ion trap are employed rather
than high-resolution mass analyzers such as Orbitrap. It is therefore
essential to choose the right mass spectrometry method for data
acquisition. To maximize the detection of PTM-modified peptides,
a combination of fragmentation methods can also be employed.
1.4 Studies on Mass A partial list of studies on mass spectrometry-based PTM analysis is
Spectrometry-Based provided in Table 1. For the purpose of this chapter, we have briefly
PTM Analysis described studies on phosphorylation, succinylation, acetylation,
ubiquitination, glycosylation, and palmitoylation.
Reversible protein phosphorylation at serine, threonine, and
tyrosine residues is one of the most common and widely studied
PTM of proteins. Other residues such as histidine, asparagine,
arginine, cysteine, and lysine are also known to be phosphorylated.
However, the extent of phosphorylation is relatively less known
owing to its occurrence in a low frequency or challenges in efficient
enrichment and identification. A previous study on human renal
cancer used a Ni-NTA-based enrichment method and identified
44,728 phosphosites on 6415 proteins [46]. Another study
expanded the understanding of the IL-33 signaling pathway by
identifying 7191 phosphorylation sites on 2746 proteins
[14]. Phosphorylation events have also been shown to be wide-
spread in prokaryotes using metal affinity-based enrichment
techniques [24].
Succinylation of lysine was identified as a new posttranslational
modification a few years ago [47]. Succinylation has been observed
to occur widely across both prokaryotes and eukaryotes
[48]. Enrichment of succinylated peptides has been typically carried
out using antibody-based affinity enrichment [49]. These enriched
peptides are then subjected to strong cation exchange chromatog-
raphy and subjected to LC-MS/MS.
Acetylation is known to regulate diverse cellular processes and
has several implications in the disease pathophysiology [50]. One of
the early studies surveyed acetylation sites in HeLa cells and mouse
liver mitochondria using an antibody-based enrichment approach
identified 388 sites [51]. Lysine acetylation has also been found to
be conserved in prokaryotes such as Escherichia coli [52] and Sac-
charomyces cerevisiae [53] and rat models [54] using antibody-
based enrichment methods. Quantitative proteomic techniques
such as TMT labeling are amenable to antibody-based acetyl lysine
peptide enrichment and have been applied to study lysine acetyla-
tion sites on perturbation [55]. In addition, technologies such as
SWATH are being increasingly used for quantitation of acetylated
proteins [56].
The ubiquitination PTM drives the localization, stability, and
activity of the protein by covalent attachment of the ubiquitin
moiety to cellular proteins [57]. Protein ubiquitination usually is
Table 1
202
Mass
Quantification Number of protein and spectrometer
PTM type Enrichment method method sites Model used type Reference
Phosphorylation Ni-NTA 44,728 phosphosites on Human renal cancer LTQ Orbitrap Peng et al. [46]
6415 proteins Velos
TiO2-based phosphopeptide TMT 512 phosphorylation sites Mycobacterium Orbitrap Verma et al.
enrichment derived from tuberculosis Fusion [24]
Sneha M. Pinto et al.
Table 2
Databases pertaining to posttranslational modifications
Table 2
(continued)
1.6 PTM Cross Talk PTM profiling studies have shown that numerous proteins can be
in Biological Systems modified by multiple PTMs, suggesting the possibility of PTM
crosstalk. Several examples of complex PTM crosstalk events reg-
ulating biological processes exist in the literature. An example of a
complex crosstalk involving PKCδ, Caspase-3, and p53 through
multiple PTMs is known to regulate various biological functions
Functional Proteomic Analysis to Characterize Signaling Crosstalk 207
2 Materials
2.3 Tandem Mass 1. TMT 6plex or 10plex isobaric label reagent set.
Tag Labeling of 2. Anhydrous acetonitrile.
Peptides
3. 5% hydroxylamine (Quenching Reagent).
4. Ultrasonic water bath.
5. Benchtop centrifuge and vortex mixer.
Experiment
LC-MS/MS analysis
50
m/z
Chemotactic factor
LPS INS Chemotactic factor
F2 GF FN1
ERK PIP3
Actin polymerization
Vav/Tiam1 Drf3 IRSp53 Mena F-actin
Adherens junction IQGAP Cdc42 Arp2/3
+p Filopodia
NWASP
RhoGEF GRLF1
Asef APC
Rho Rac
TMSB4
PIR121
PXN GIT1 PIX HSPC300
Nap125
+p +p WAVE1
Focal adhesion +p PAK Abi2 PFN Actin
ERM NHE1 ROCK +p polymerization
assembly +p +p
VCL +p +p IRSp53 WAVE2
PI4P5K MLCP MLCK PI4P5K LIMK F-actin
ACTN PIP2 +p Arp2/3
-p +p
Focal adhesion CFN Stabilization
MLC
mDia -p of actin Lamellipodia
PIP2
SSH
PFN
?
Actin VCL GSN
polymerization
stress fiber
Actomyosin
Focal adhesion Focal adhesion
assembly contraction
assembly
F-actin MyosinII
Stress fibers
Fig. 1 Workflow for the identification and quantification of PTM-modified peptides using LC-MS/MS analysis.
The cells are cultured in appropriate growth media and stimulated with growth factors/cytokines or treated
with inhibitors to study their effect. Proteins from each condition are harvested by cell lysis, enzymatically
Functional Proteomic Analysis to Characterize Signaling Crosstalk 211
3. Syringes.
4. Blunt end needle.
5. Benchtop centrifuge for microfuge tubes.
6. Vacuum concentrator.
7. Solvents.
(a) Acetonitrile (LC-MS grade).
(b) 0.1% formic acid.
(c) 80% acetonitrile, 0.5% acetic acid.
(d) 50% acetonitrile, 0.1% formic acid.
3 Methods
Fig. 1 (continued) digested and subsequently labeled with Tandem mass tags (TMT). The TMT-labeled
samples are pooled, fractionated using high pH reverse phase chromatography. The fractionated samples
are dried and subjected to the serial enrichment of posttranslational modifications using metal affinity/
immunoaffinity methods. It is preferable to first enrich for phosphorylated residues using metal affinity
chromatography as the solvents used are volatile and the flowthrough can be then subjected to immunoaffinity
purification. The enriched peptides as well as a fraction of the total peptides are analyzed using high-
resolution nano-LC-MS/MS. The raw data derived from the data acquisition are processed using software
suites to identify and quantify PTM-modified peptides and proteins. The reporter ions are used for the relative
quantitation of the abundance of peptides
212 Sneha M. Pinto et al.
3.2 Protein Digestion 1. Transfer 600–800 μg per condition into a new tube. Adjust the
final volume such that it is equal in all tubes.
2. Add reducing agent to a final concentration of 10 mM and
incubate at 60 C for 20 min. Cool the tubes to room
temperature.
3. Add alkylating agent to the final concentration of 20 mM and
incubate for 10 min in dark at room temperature.
4. Add six volumes of ice-cold acetone to each tube and incubate
at 20 C overnight (see Note 6).
5. Centrifuge the samples at 14,000 g for 10 min at 4 C.
Discard the supernatant and air-dry the pellet. Resuspend the
protein pellet in 200 μL of 50 mM TEABC buffer (see Note 7).
6. Prepare trypsin solution at a concentration of 1 mg/mL in
50 mM TEABC buffer. Add Trypsin solution at 1:20 (Enzyme
to substrate ratio) to the peptide digest and incubate the
microfuge tubes overnight at 37 C (see Note 8).
7. After the confirmation of digestion efficiency evaporate the
sample to complete dryness.
3.3 Tandem Mass 1. Bring down the TMT reagents to room temperature and add
Tag (TMT) Labeling of 41 μL of anhydrous acetonitrile to each of the TMT reagent
Peptides (0.8 mg) tube.
2. Briefly vortex the tubes to ensure complete dissolution.
3. Centrifuge the tubes and keep for 5 min at room temperature.
Check the pH (7.5–8).
4. Dissolve the vacuum-dried sample in 100 μL of 50 mM
TEABC (see Note 9). Centrifuge the tubes at 12,000 g for
10 min.
Functional Proteomic Analysis to Characterize Signaling Crosstalk 213
3.4 Fractionation 1. Fractionate the peptide digest using basic reverse phase chro-
matography. Reconstitute the TMT-labeled pooled peptide
digest in 1 mL Solvent A (10 mM TEABC buffer, pH 9.5).
2. Load the sample onto XBridge C18 column. Resolve the pep-
tides using a gradient of solvent A and B for 120 min with a
flow rate of 1 mL/min.
3. Collect the fractions in 96-well plate using a fraction collector
programmed to collect fractions from the start of the gradient
(see Note 10).
4. Reduce the volume of fractions at freezing temperature. Con-
catenate the fractions to obtain a total of 6 fractions. Dry the
samples using vacuum concentrator equipped with refrigerated
vapor trap.
4 Notes
Acknowledgements
References
1. UniProt C (2015) UniProt: a hub for protein kinome profiling by rational selection of rele-
information. Nucleic Acids Res 43(Database vant spots. Sci Rep 6:26695. https://doi.org/
issue):D204–D212. https://doi.org/10. 10.1038/srep26695
1093/nar/gku989 8. Baharani A, Trost B, Kusalik A, Napper S
2. Creasy DM, Cottrell JS (2004) Unimod: pro- (2017) Technological advances for interrogat-
tein modifications for mass spectrometry. Pro- ing the human kinome. Biochem Soc Trans 45
teomics 4(6):1534–1536. https://doi.org/10. (1):65–77. https://doi.org/10.1042/
1002/pmic.200300744 BST20160163
3. Zhang H, Shi X, Pelech S (2016) Monitoring 9. Kim MS, Pinto SM, Getnet D, Nirujogi RS,
protein kinase expression and phosphorylation Manda SS, Chaerkady R, Madugundu AK,
in cell lysates with antibody microarrays. Meth- Kelkar DS, Isserlin R, Jain S, Thomas JK,
ods Mol Biol 1360:107–122. https://doi.org/ Muthusamy B, Leal-Rojas P, Kumar P, Sahas-
10.1007/978-1-4939-3073-9_9 rabuddhe NA, Balakrishnan L, Advani J,
4. Shi J, Sharif S, Ruijtenbeek R, Pieters RJ George B, Renuse S, Selvan LD, Patil AH,
(2016) Activity based high-throughput screen- Nanjappa V, Radhakrishnan A, Prasad S,
ing for novel O-GlcNAc transferase substrates Subbannayya T, Raju R, Kumar M, Sreenivasa-
using a dynamic peptide microarray. PLoS One murthy SK, Marimuthu A, Sathe GJ, Chavan S,
11(3):e0151085. https://doi.org/10.1371/ Datta KK, Subbannayya Y, Sahu A, Yelamanchi
journal.pone.0151085 SD, Jayaram S, Rajagopalan P, Sharma J,
5. Zhu B, Farris TR, Milligan SL, Chen H, Murthy KR, Syed N, Goel R, Khan AA,
Zhu R, Hong A, Zhou X, Gao X, McBride Ahmad S, Dey G, Mudgal K, Chatterjee A,
JW (2016) Rapid identification of ubiquitina- Huang TC, Zhong J, Wu X, Shaw PG,
tion and SUMOylation target sites by micro- Freed D, Zahari MS, Mukherjee KK,
fluidic peptide array. Biochem Biophys Rep Shankar S, Mahadevan A, Lam H, Mitchell
5:430–438. https://doi.org/10.1016/j. CJ, Shankar SK, Satishchandra P, Schroeder
bbrep.2016.02.003 JT, Sirdeshmukh R, Maitra A, Leach SD,
Drake CG, Halushka MK, Prasad TS, Hruban
6. Al-Ejeh F, Miranda M, Shi W, Simpson PT, RH, Kerr CL, Bader GD, Iacobuzio-Donahue
Song S, Vargas AC, Saunus JM, Smart CE, CA, Gowda H, Pandey A (2014) A draft map
Mariasegaram M, Wiegmans AP, Chenevix- of the human proteome. Nature 509
Trench G, Lakhani SR, Khanna KK (2014) (7502):575–581. https://doi.org/10.1038/
Kinome profiling reveals breast cancer hetero- nature13302
geneity and identifies targeted therapeutic
opportunities for triple negative breast cancer. 10. Wilhelm M, Schlegl J, Hahne H, Gholami AM,
Oncotarget 5(10):3145–3158. https://doi. Lieberenz M, Savitski MM, Ziegler E,
org/10.18632/oncotarget.1865 Butzmann L, Gessulat S, Marx H,
Mathieson T, Lemeer S, Schnatbaum K,
7. Scholma J, Fuhler GM, Joore J, Hulsman M, Reimer U, Wenschuh H, Mollenhauer M,
Schivo S, List AF, Reinders MJ, Peppelenbosch Slotta-Huspenina J, Boese JH, Bantscheff M,
MP, Post JN (2016) Improved intra-array and Gerstmair A, Faerber F, Kuster B (2014) Mass-
interarray normalization of peptide microarray spectrometry-based draft of the human
phosphorylation for phosphorylome and
220 Sneha M. Pinto et al.
47. Zhang Z, Tan M, Xie Z, Dai L, Chen Y, Zhao Y Biochim Biophys Acta 1866:464. https://doi.
(2011) Identification of lysine succinylation as org/10.1016/j.bbapap.2017.11.011
a new post-translational modification. Nat 56. Meyer JG, D’Souza AK, Sorensen DJ, Rardin
Chem Biol 7(1):58–63. https://doi.org/10. MJ, Wolfe AJ, Gibson BW, Schilling B (2016)
1038/nchembio.495 Quantification of lysine acetylation and Succi-
48. Weinert BT, Scholz C, Wagner SA, nylation stoichiometry in proteins using mass
Iesmantavicius V, Su D, Daniel JA, Choudhary spectrometric data-independent acquisitions
C (2013) Lysine succinylation is a frequently (SWATH). J Am Soc Mass Spectrom 27
occurring modification in prokaryotes and (11):1758–1771. https://doi.org/10.1007/
eukaryotes and extensively overlaps with acety- s13361-016-1476-z
lation. Cell Rep 4(4):842–851. https://doi. 57. Pickart CM, Eddins MJ (2004) Ubiquitin:
org/10.1016/j.celrep.2013.07.024 structures, functions, mechanisms. Biochim
49. Xie Z, Dai J, Dai L, Tan M, Cheng Z, Wu Y, Biophys Acta 1695(1–3):55–72. https://doi.
Boeke JD, Zhao Y (2012) Lysine succinylation org/10.1016/j.bbamcr.2004.09.019
and lysine malonylation in histones. Mol Cell 58. Wu Q, Cheng Z, Zhu J, Xu W, Peng X,
Proteomics 11(5):100–107. https://doi.org/ Chen C, Li W, Wang F, Cao L, Yi X, Wu Z,
10.1074/mcp.M111.015875 Li J, Fan P (2015) Suberoylanilide hydroxamic
50. Drazic A, Myklebust LM, Ree R, Arnesen T acid treatment reveals crosstalks among prote-
(2016) The world of protein acetylation. Bio- ome, ubiquitylome and acetylome in non-small
chim Biophys Acta 1864(10):1372–1401. cell lung cancer A549 cell line. Sci Rep 5:9520.
https://doi.org/10.1016/j.bbapap.2016.06. https://doi.org/10.1038/srep09520
007 59. Iesmantavicius V, Weinert BT, Choudhary C
51. Kim SC, Sprung R, Chen Y, Xu Y, Ball H, Pei J, (2014) Convergence of ubiquitylation and
Cheng T, Kho Y, Xiao H, Xiao L, Grishin NV, phosphorylation signaling in rapamycin-
White M, Yang XJ, Zhao Y (2006) Substrate treated yeast cells. Mol Cell Proteomics 13
and functional diversity of lysine acetylation (8):1979–1992. https://doi.org/10.1074/
revealed by a proteomics survey. Mol Cell 23 mcp.O113.035683
(4):607–618. https://doi.org/10.1016/j. 60. Yang X, Liu F, Yan Y, Zhou T, Guo Y, Sun G,
molcel.2006.06.026 Zhou Z, Zhang W, Guo X, Sha J (2015) Pro-
52. Zhang J, Sprung R, Pei J, Tan X, Kim S, teomic analysis of N-glycosylation of human
Zhu H, Liu CF, Grishin NV, Zhao Y (2009) seminal plasma. Proteomics 15
Lysine acetylation is a highly abundant and (7):1255–1258. https://doi.org/10.1002/
evolutionarily conserved modification in pmic.201400203
Escherichia coli. Mol Cell Proteomics 8 61. Sudhir PR, Chen CH, Pavana Kumari M, Wang
(2):215–225. https://doi.org/10.1074/mcp. MJ, Tsou CC, Sung TY, Chen JY, Chen CH
M800187-MCP200 (2012) Label-free quantitative proteomics and
53. Henriksen P, Wagner SA, Weinert BT, N-glycoproteomics analysis of KRAS-activated
Sharma S, Bacinskaja G, Rehman M, Juffer human bronchial epithelial cells. Mol Cell Pro-
AH, Walther TC, Lisby M, Choudhary C teomics 11(10):901–915. https://doi.org/10.
(2012) Proteome-wide analysis of lysine acety- 1074/mcp.M112.020875
lation suggests its broad regulatory scope in 62. Chen W, Smeekens JM, Wu R (2014) A uni-
Saccharomyces cerevisiae. Mol Cell Proteomics versal chemical enrichment method for
11(11):1510–1522. https://doi.org/10. mapping the yeast N-glycoproteome by mass
1074/mcp.M112.017251 spectrometry (MS). Mol Cell Proteomics 13
54. Lundby A, Lage K, Weinert BT, Bekker-Jensen (6):1563–1572. https://doi.org/10.1074/
DB, Secher A, Skovgaard T, Kelstrup CD, mcp.M113.036251
Dmytriyev A, Choudhary C, Lundby C, 63. Guan X, Fierke CA (2011) Understanding pro-
Olsen JV (2012) Proteomic analysis of lysine tein palmitoylation: biological significance and
acetylation sites in rat tissues reveals organ enzymology. Sci China Chem 54
specificity and subcellular patterns. Cell Rep 2 (12):1888–1897. https://doi.org/10.1007/
(2):419–431. https://doi.org/10.1016/j.cel s11426-011-4428-2
rep.2012.07.006 64. Martin BR (2013) Nonradioactive analysis of
55. Xie C, Shen H, Zhang H, Yan J, Liu Y, Yao F, dynamic protein palmitoylation. Curr Protoc
Wang X, Cheng Z, Tang TS, Guo C (2017) Protein Sci 73(Unit 14):15. https://doi.org/
Quantitative proteomics analysis reveals altera- 10.1002/0471140864.ps1415s73
tions of lysine acetylation in mouse testis in 65. Martin BR, Wang C, Adibekian A, Tully SE,
response to heat shock and X-ray exposure. Cravatt BF (2011) Global profiling of dynamic
Functional Proteomic Analysis to Characterize Signaling Crosstalk 223
protein palmitoylation. Nat Methods 9 73. Minguez P, Parca L, Diella F, Mende DR,
(1):84–89. https://doi.org/10.1038/nmeth. Kumar R, Helmer-Citterich M, Gavin AC, van
1769 Noort V, Bork P (2012) Deciphering a global
66. Hornbeck PV, Zhang B, Murray B, Kornhauser network of functionally associated post-
JM, Latham V, Skrzypek E (2015) PhosphoSi- translational modifications. Mol Syst Biol
tePlus, 2014: mutations, PTMs and recalibra- 8:599. https://doi.org/10.1038/msb.2012.31
tions. Nucleic Acids Res 43(Database issue): 74. Trinidad JC, Barkan DT, Gulledge BF,
D512–D520. https://doi.org/10.1093/nar/ Thalhammer A, Sali A, Schoepfer R, Burlin-
gku1267 game AL (2012) Global identification and
67. Keshava Prasad TS, Goel R, Kandasamy K, characterization of both O-GlcNAcylation
Keerthikumar S, Kumar S, Mathivanan S, and phosphorylation at the murine synapse.
Telikicherla D, Raju R, Shafreen B, Mol Cell Proteomics 11(8):215–229. https://
Venugopal A, Balakrishnan L, Marimuthu A, doi.org/10.1074/mcp.O112.018366
Banerjee S, Somanathan DS, Sebastian A, 75. Kim MS, Zhong J, Pandey A (2016) Common
Rani S, Ray S, Harrys Kishore CJ, Kanth S, errors in mass spectrometry-based analysis of
Ahmed M, Kashyap MK, Mohmood R, Rama- post-translational modifications. Proteomics
chandra YL, Krishna V, Rahiman BA, Mohan S, 16(5):700–714. https://doi.org/10.1002/
Ranganathan P, Ramabadran S, Chaerkady R, pmic.201500355
Pandey A (2009) Human protein reference 76. Hughes CS, Spicer V, Krokhin OV, Morin GB
database—2009 update. Nucleic Acids Res 37 (2017) Investigating acquisition performance
(Database):D767–D772. https://doi.org/10. on the Orbitrap fusion when using tandem
1093/nar/gkn892 MS/MS/MS scanning with isobaric tags. J
68. Huang KY, Su MG, Kao HJ, Hsieh YC, Jhong Proteome Res 16(5):1839–1846. https://doi.
JH, Cheng KH, Huang HD, Lee TY (2016) org/10.1021/acs.jproteome.7b00091
dbPTM 2016: 10-year anniversary of a 77. Pan J, Chen R, Li C, Li W, Ye Z (2015) Global
resource for post-translational modification of analysis of protein lysine succinylation profiles
proteins. Nucleic Acids Res 44(D1): and their overlap with lysine acetylation in the
D435–D446. https://doi.org/10.1093/nar/ marine bacterium vibrio parahemolyticus. J
gkv1240 Proteome Res 14(10):4309–4318. https://
69. Liu Z, Wang Y, Gao T, Pan Z, Cheng H, doi.org/10.1021/acs.jproteome.5b00485
Yang Q, Cheng Z, Guo A, Ren J, Xue Y 78. Cheng Y, Hou T, Ping J, Chen G, Chen J
(2014) CPLM: a database of protein lysine (2016) Quantitative succinylome analysis in
modifications. Nucleic Acids Res 42(Database the liver of non-alcoholic fatty liver disease rat
issue):D531–D536. https://doi.org/10. model. Proteome Sci 14:3. https://doi.org/
1093/nar/gkt1093 10.1186/s12953-016-0092-y
70. Kennedy JJ, Yan P, Zhao L, Ivey RG, Voytovich 79. Colak G, Xie Z, Zhu AY, Dai L, Lu Z, Zhang Y,
UJ, Moore HD, Lin C, Pogosova-Agadjanyan Wan X, Chen Y, Cha YH, Lin H, Zhao Y, Tan
EL, Stirewalt DL, Reding KW, Whiteaker JR, M (2013) Identification of lysine succinylation
Paulovich AG (2016) Immobilized metal affin- substrates and the succinylation regulatory
ity chromatography coupled to multiple reac- enzyme CobB in Escherichia coli. Mol Cell
tion monitoring enables reproducible Proteomics 12(12):3509–3520. https://doi.
quantification of phospho-signaling. Mol Cell org/10.1074/mcp.M113.031567
Proteomics 15(2):726–739. https://doi.org/ 80. Sun G, Jiang M, Zhou T, Guo Y, Cui Y, Guo X,
10.1074/mcp.O115.054940 Sha J (2014) Insights into the lysine acetylpro-
71. Chick JM, Kolippakkam D, Nusinow DP, teome of human sperm. J Proteome
Zhai B, Rad R, Huttlin EL, Gygi SP (2015) A 109:199–211. https://doi.org/10.1016/j.
mass-tolerant database search identifies a large jprot.2014.07.002
proportion of unassigned spectra in shotgun 81. Xie L, Wang X, Zeng J, Zhou M, Duan X, Li Q,
proteomics as modified peptides. Nat Biotech- Zhang Z, Luo H, Pang L, Li W, Liao G, Yu X,
nol 33(7):743–749. https://doi.org/10. Li Y, Huang H, Xie J (2015) Proteome-wide
1038/nbt.3267 lysine acetylation profiling of the human path-
72. Pan Z, Liu Z, Cheng H, Wang Y, Gao T, ogen Mycobacterium tuberculosis. Int J Bio-
Ullah S, Ren J, Xue Y (2014) Systematic analy- chem Cell Biol 59:193–202. https://doi.org/
sis of the in situ crosstalk of tyrosine modifica- 10.1016/j.biocel.2014.11.010
tions reveals no additional natural selection on 82. Karg E, Smets M, Ryan J, Forne I, Qin W,
multiply modified residues. Sci Rep 4:7331. Mulholland CB, Kalideris G, Imhof A,
https://doi.org/10.1038/srep07331 Bultmann S, Leonhardt H (2017) Ubiquitome
224 Sneha M. Pinto et al.
Abstract
Peptide identification relies in the majority of mass spectrometry-based proteomics experiments on match-
ing of experimental data against peptide and fragment ion masses derived from in silico digests of protein
databases. One of the main drawbacks of this approach is that modifications have to be defined for database
searching and therefore no unexpected modifications can be identified in a standard setup. Consequently, in
many bottom-up proteomics experiments, unexpected modifications are not identified, even if high-quality
fragment ion spectra of the modified peptides were acquired. It is therefore often not straightforward to
identify unexpected modifications. In this protocol, we describe a stepwise procedure to identify unex-
pected modifications at peptides using the database search algorithm Mascot. The workflow includes
parallel searches for the identification of known modifications at unexpected amino acids, error tolerant
searches for modifications unexpected in the sample but known to the community, and mass tolerant
searches for entirely unknown modifications. Furthermore, we suggest a follow-up strategy consisting of
(1) verification of identified modifications in the initial dataset and (2) targeted experiments using synthetic
peptides.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_15, © Springer Science+Business Media, LLC, part of Springer Nature 2019
225
226 Shiva Ahmadi and Dominic Winter
2 Materials
2.1 Cell Culture Cell culture medium: Dulbecco’s modified Eagle medium
(DMEM) supplemented with 10% FCS, 100 U/mL penicillin,
and 100 μg/mL streptomycin.
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 227
2.5 LC-MS/MS 1. C18 analytical column: ESI spray tip produced in house with a
Measurements Sutter Instruments P2000 laser puller from a 360 μm outer
diameter, 100 μm inner diameter fused silica capillary and
packed with 5 μm particles [Dr. Maisch, Reprosil C-18 AQ].
Alternatively, any type of commercially available nanoflow C18
column in combination with an appropriate emitter can
be used.
2. Thermo Scientific EASY-nLC 1000 or similar nanoflow high/
or ultrahigh-performance liquid chromatography system.
3. Thermo Scientific Orbitrap Velos Mass Spectrometer or any
other high-resolution / high accuracy mass spectrometer.
4. Solvent A: Water with 0.1% FA.
5. Solvent B: ACN with 0.1% FA.
3 Methods
3.1 Sample If the choice of the specific subcellular fraction is not influenced by
Preparation: In-Gel a biological question and it serves only as a source of proteins, the
Digestion of a cytosolic fraction is the easiest to generate, as the other parts of the
Cytosolic HeLa cell can be removed by centrifugation. Furthermore, the cytosolic
Fraction fraction is of adequate complexity in order to allow a sufficient
coverage of the proteins present in the sample and to provide a
reasonable number of proteins in order to resemble the complexity
observed in medium to large scale proteomics studies. When
planning the experiment, care should be taken that the differential
treatment of the samples is not influencing the subsequent steps,
possibly resulting in secondary effects. For this reason, we decided
to perform reduction and alkylation after performing SDS-PAGE as
differential modification of the proteins may influence their run-
ning behavior in the gel electrophoresis.
1. Place the cell culture dishes on ice, aspirate the medium, and
wash the cells 3 with 10 mL ice-cold PBS (see Note 3).
2. Add 1 mL sucrose buffer, detach the cells using a cell scraper,
and transfer the cell suspension to a dounce homogenizer (see
Note 4).
3. Homogenize cells on ice using the dounce homogenizer
(30 strokes) and transfer the suspension to a 2 mL microtube
(for higher volumes 15 or 50 mL conical tubes can be used).
4. Centrifuge at 1000 g, 4 C for 10 min in order to pellet
intact cells and nuclei. Transfer the supernatant to a new 2 mL
microtube.
230 Shiva Ahmadi and Dominic Winter
4. Set the repeat count to one and the dynamic exclusion window
to a value larger than the average chromatographic peak width
at the basis in order to prevent repetitive fragmentation of
highly abundant ions.
5. If certain neutral losses are expected (e.g., 98 for phosphory-
lation) define them in the MSA scan setup (in case an Orbitrap
mass spectrometer is used).
6. If available, activate the lock mass option in order to achieve
maximal mass accuracy.
7. If possible, use either small RP particles or avoid the use of a
trap column to achieve maximal chromatographic resolution.
This results in increased signal intensities due to narrower peak
shapes aiding the identification of low-abundant species.
8. Perform nano(U)HPLC-MS/MS analysis of the sample.
3.3 Data Analysis Dependent on the availability of sample material and controls, differ-
ent strategies can be taken. Figure 1 shows the workflow for identifi-
cation of unexpected modifications, including possible steps and the
order of their execution. There are several algorithms available to
perform such analyses; we used Mascot (www.matrixscience.org)
either in combination with Proteome Discoverer (Thermo Scientific)
or individually by manual submission of mascot generic files (mgf).
Alternatively, dependent on their features other programs like Max-
Quant, SEQUEST, PEAKS DB, ProteinPilot, pFind, Byonic, or X!
Tandem can be used [23].
3.3.1 Initial Analyses In case of modifications due to a chemical treatment (in our case
Defining Known unspecific alkylation of amino acid side chains), whose composition
Modifications at All Amino is known but not the amino acid residue(s) modified, the possible
Acids modification(s) should initially be searched at all possible amino
acids. As the number of variable modifications which can be defined
in a Mascot search is limited to 9 at a time, three individual searches
have to be performed. This results in redundant assignments of
MS/MS spectra for the unmodified peptides in the single searches.
Furthermore, if the fragment ions are not conclusive for assigning
the modification to a distinct amino acid, the same spectrum may be
annotated with different modification positions in the separate
searches. Proteome discoverer allows dealing with these issues by
combining the results of the individual searches in one FDR analysis.
1. Extend the specificity definition of the modification in the
Mascot configuration editor to all possible amino acids and
define them as active so they can be selected for searching. In
our study, we specified carbamidomethyl (for IAA and CAA),
carboxymethyl (for IAC), and propionamide (for AA) at all
amino acids.
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 233
Fig. 2 Amino acids found to be modified by the reduction and alkylation procedure. Shown are the combined
results from six replicates. (a, b) Investigation for off-site alkylation at all possible amino acids in samples
reduced with DTT and alkylated with (a) AA (DA) and (b) IAA (DIA). Manual analysis of (b) revealed frequent
alkylation of seven amino acids and the peptide N-terminus. (c, d) PSMs annotated to be off-site (c) mono-
alkylated and (d) di-alkylated at Y, S, D, T, H, E, K, and the peptide N terminus (reproduced from [11]). DI
DTT/IAA, DIA DTT/IAC, DA DTT/AA, DC DTT/CAA, TI TCEP/IAA, TIA TCEP/IAC, TA TCEP/AA, TC TCEP/CAA, MI
BME/IAA, MIA BME/IAC, MA BME/AA, MC BME/CAA, N-term peptide N terminus
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 235
3.3.2 Estimating the If the database searches with known modifications do not yield any
Abundance of Unexpected conclusive results, a reasonable initial step is to investigate which
Modifications unmodified peptides are depleted from the sample as a consequence
of the investigated treatment. Therefore, if possible, the sample
exposed to the biological stimulus or chemical treatment should
be compared to an untreated control sample (see Note 10). This
allows estimating the extent of the modification(s) either due to the
lack of identification of unmodified peptides or their reduction in
signal intensity. In our example study, we compared the samples
treated with different alkylation reagents to an untreated control.
Each sample was generated in two independent replicates and then
each replicate measured three times by LC-MS/MS in order to
exclude false conclusions due to DDA-dependent variation in pep-
tide identification. This high degree of redundancy allowed us to
assess if the observed differences are random or follow a systematic
pattern.
1. If needed, convert MS raw files to an appropriate format (see
Note 11).
2. Define parameters for the initial database search.
(a) The precursor and fragment ion tolerance should be as
narrow as possible, dependent on the performance of the
mass spectrometer used. For most high-resolution mass
spectrometers, a precursor mass tolerance of 10 ppm is
reasonable (also applied for our dataset). For fragment
ions, 50 mmU should be selected for high-resolution
MS/MS spectra (Orbitrap and QTOF instruments) and
0.6–0.8 Da for low resolution fragment ion spectra
acquired in the LTQ part of the instrument (see Note 12).
(b) Choose a database and define the taxonomy based on the
organism used. When searching for unexpected modifica-
tions, it is advisable to use rather small high confidence
databases like, e.g., SwissProt (see Note 13).
(c) Define a way of false discovery rate (FDR) estimation; we
usually use Percolator [24] (which is included in Prote-
ome Discoverer) and set the FDR to 1% on the peptide
level.
(d) Based on the alkylation reagent, define the respective fixed
modification at cysteine, as well as the expected variable
modifications (e.g., oxidation of methionine) (see Note 14).
236 Shiva Ahmadi and Dominic Winter
a) b)
10000
8000
40
% Identification
# of PSMs
6000
4000 20
2000
0 0
I TI I I TI I
D IA DA DC A A
TI T TC M IA A C N D IA DA D
C
TI
A TA TC M IA MA MC ON
D M M M CO D M C
conditions conditions
Fig. 3 Results of the analyses of six replicates of in-gel digested cytosolic fractions of HeLa cells reduced and
alkylated with different combinations of reagents. (a) Number of peptide spectral matches (PSMs); (b)
percentage of identified spectra (reproduced from [11]). DI DTT/IAA, DIA DTT/IAC, DA DTT/AA, DC DTT/CAA,
TI TCEP/IAA, TIA TCEP/IAC, TA TCEP/AA, TC TCEP/CAA, MI BME/IAA, MIA BME/IAC, MA BME/AA, MC BME/CAA,
Con control sample
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 237
3.3.3 Relative On the PSM level, the influence of a modification can only be
Quantification of Signal identified if the investigated treatment results in a strong reduction
Reduction of the peptide. If it, however, only results in the modification of a
small percentage of the peptides, it may be difficult to realize the
effect based on the PSM number; as also ions reduced in abundance
are usually sufficient to result in MS/MS spectra allowing for
peptide assignment. In such cases, relative quantification can be
used in order to identify changes and to estimate the extent of
modification. While the easiest approach is a label-free quantifica-
tion by calculation of the area under the curve, a variety of stable
isotope labeling-based methods is available which allows for more
accurate quantification. They can be grouped in methods inducing
stable isotope labels through metabolic incorporation (15N/14N
metabolic labeling and SILAC), chemical derivatization (ICAT,
iTRAQ, TMT), or during proteolytic processing (18O labeling)
[25]. As the labels are introduced at different steps of the protocol
by the varying methods, the method of choice is dependent on the
experiment. In our example study, we applied dimethyl labeling as it
allowed us to use the same starting material for all conditions and to
label the peptides after reduction and alkylation with the different
reagents.
1. After sample treatment and proteolytic digestion, desalt the
peptides, dry them using a vacuum centrifuge, and resuspend
them at 1 μg/μL in 100 mM HEPES (pH 5–8.5) (see Note
17).
2. For 100 μL of sample volume, add 16 μL of light, medium, or
heavy dimethylation solution 1 to the respective samples.
3. Add 16 μL of light, medium, or heavy dimethylation solution
2 to the samples followed by incubation in the fume hood for
1 h at RT while mixing at 800 rpm.
4. Quench the labeling with 64 μL of dimethyl quenching solu-
tion/100 μg sample.
5. Add 32 μL of FA for acidification of the sample.
6. Combine the different labeled samples, desalt them using
STAGE tips [22], and proceed to MS analysis (see Note 18).
7. Process the raw files using the relative quantification by
dimethyl labeling option in Proteome Discoverer. Exclude
peptides containing the presumably modified amino acids
(e.g., cysteine in this study) from the list of the results as they
exhibit different masses and chromatographic retention times
due to the different modified groups. Also exclude samples
with missing values in one of the channels (see Note 19).
238 Shiva Ahmadi and Dominic Winter
a) b)
8 IAA / Control 8 AA / Control
6 6
4 4
Log2 fold change
2 2
0 0
-2 -2
-4 -4
-6 -6
-8 -8
500 1000 1500 500 1000 1500
Pepde # Pepde #
Fig. 4 Quantification of changes in peptide abundance due to differential alkylation. Shown are the log2 values
of the average fold-change from two independent experiments. Proteins were reduced and alkylated in the
gel, digested by trypsin, the individual peptide samples labeled by the dimethylation reagents (IAA: light, AA:
medium, control: heavy) and combined. PSMs with a negative fold change between 1 and 2 are indicated in
light red and those with >twofold in dark red. PSMs with a positive fold change between 1 and 2 are colored
light green and >twofold dark green (reproduced from [11])
3.3.4 Identification of The MS/MS identification rate data (Subheading 3.3.2) or the
Affected Amino Acids or relative peptide abundances (Subheading 3.3.3) can now be used
Functional Groups to determine if the affected amino acids follow a certain pattern.
(at Peptide Termini or For this purpose, all peptides which were either absent or signifi-
Specific Side Chains) cantly downregulated as a consequence of the treatment are ana-
lyzed for their amino acid composition. If the unknown
modification is specific for a certain amino acid or functional
group of the peptide/protein, as it usually is the case, the affected
amino acid is expected to be overrepresented in the regulated
population of peptides. This analysis can easily be performed in
MS Excel:
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 239
Error Tolerant Search Error tolerant searching can either be executed in the initial search
by selecting the respective option in the MS/MS Ions Search
interface or on the results page of any MS/MS ion search. The
240 Shiva Ahmadi and Dominic Winter
a)
2
-1
-2
A C D E F G H I K L M N P Q R S T V W Y
b)
2
normalized log2 abundance
-1
-2
A C D E F G H I K L M N P Q R S T V W Y
DI DA TI TA MI MA C ON
D IA DC T IA TC M IA MC
Mass Tolerant Search Mass tolerant searches have to be performed on the whole dataset.
If they result in too much server occupancy, the *.mgf files can be
generated from fewer *.raw files, or if the *.raw files are too large,
they can be divided in smaller packages. This has to be determined
based on the capacity of the server used. The wide-tolerance win-
dow for the precursor mass defined in the mass tolerant search
(250 Da in our example study) cannot be defined in the Mascot
MS/MS Ions Search interface. Therefore, the *.mgf file has to be
modified manually.
1. Create the *.mgf file and open it using an appropriate software
(e.g., Wordpad or Notepad++).
2. Modify the header of the *.mgf file to include the wide mass
tolerance of your choice. In our example study, we changed the
header to: peptide mass tolerance: 250 Da; fragment ion toler-
ance: 0.8 Da; search type: SQ; fixed modifications: carbamido-
methyl, carboxymethyl, or propionamide, respectively, at
cysteine residues depending on the alkylation reagent.
3. Perform Mascot MS/MS ion searches using the modified *.
mgf file. The mass tolerance settings in the *.mgf file overwrite
the settings selected in the MS/MS Ions Search interface;
therefore the default settings can be kept.
Interpretation of Error Both the error tolerant and the mass tolerant searches result in high
Tolerant and Mass Tolerant numbers of false positive identifications and are not compatible
Search Results with decoy database searching. Therefore, great care has to be
taken which peptide matches are accepted. In such case, an arbitrary
cutoff can be selected; in our experience an ion score of 30 is a
reasonable compromise between selectivity and sensitivity. Alterna-
tively, a cutoff to achieve 1% FDR at the peptide level can be
determined in a first target/decoy database search. One should
be, however, careful as the value determined by this approach will
definitely be too low. Therefore, it should only be used as a ten-
dency rather than a strict cutoff.
242 Shiva Ahmadi and Dominic Winter
For both the error and mass tolerant searches, the amino acids
found to be affected in Subheading 3.3.4 are expected to be present
in the identified modified peptides. If this is not the case, alterna-
tively the identification/quantification information for these pep-
tides can be extracted from the previous datasets to assess if they
were regulated. While the error tolerant search provides a sugges-
tion for the observed modification, the mass tolerant search only
gives a mass value which does not provide any explanation. In this
case, it would be ideal if it is possible to suggest modifications based
on the treatment investigated. If this is not applicable, a reasonable
first step is the determination of the modifications’ elemental com-
position. This can be achieved by calculation of its accurate mass
through the mean value of the mass error for all peptides found with
the respective modification. This then allows to suggest possible
sum formulas using the involved atoms (usually C, H, O, N, and S).
In our example study, error tolerant searches did not result in
the identification of any unusual modifications. In mass tolerant
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 243
a)
2000
1500
1000
IAA/IAC
# PSMs
500
0
-500
AA/CAA
-1000
-1500
-2000
b)
2000 oxM
1500 -64
1000
IAA/IAC
# PSMs
500
0
-500
AA/CAA
-1000
-1500
-2000
Fig. 6 Combined results of the mass tolerant searches for two independent replicates of all reduction and
alkylation reagents. Shown are the summed abundances for peptide mass errors binned in groups of 5 Da for
all reduction reagents in combination with the iodine-containing alkylation reagents (IAA/IAC, upper panel), as
well as the non-iodine-containing reagents (AA/CAA, lower panel). (a) Searches performed without variable
modifications did not identify any differences between the treatments except of the already identified offside
alkylation (+57 Da); (b) searches including oxidation at Methionine (oxM) as variable modification resulted in
the assignment of ~1000 PSMs with a mass error of 64 Da. All of these PSMs contained methionine
(reproduced from [11])
3.3.6 Manual Data The identified modifications have to be validated in order to pre-
Validation for Identified vent false conclusions. Due to the lack of an FDR analysis, this is
Modifications especially of importance in the case of error and mass tolerant
searches. Therefore, the initial database search should be repeated
with the newly discovered modification(s) as variable modification
at the respective amino acid(s). The modification can be defined in
the Mascot configuration editor. If the modifications’ elemental
composition could not be determined, one can alternatively use a
sum formula matching the detected mass error and increase the
search tolerance window to compensate for possible minor differ-
ences in the molecular weight of the modification. The amino acids
to be specified as modified are indicated in case of the error tolerant
searches; for the mass tolerant searches, however, as the mass errors
are only determined from the precursor ion, no suggestions can be
provided by Mascot. If no amino acid abundance analysis (see
Subheading 3.3.4) was performed or if it was inconclusive, another
set of parallel searches (see Subheading 3.3.1) can be performed in
order to narrow down the list of possibly affected amino acids.
Subsequently, several spectra with high ion scores should be eval-
uated manually by comparison of the fragment ion masses
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 245
Fig. 7 Carbamylated methionine results in a neutral loss of the amino acid side chain due to in-source
fragmentation. (a) Survey spectrum showing the neutral loss of the side chain from the peptide with alkylated
methionine; (b) extracted ion chromatogram (XICs) of the peptide with alkylated methionine, and its version
which lost the side chain due to in-source fragmentation, showing perfect co-elution (reproduced from [11])
246 Shiva Ahmadi and Dominic Winter
Fig. 8 (a) MALDI-MS spectra of the synthetic peptide APEIMLNSK, reduced with DTT and alkylated with IAA,
IAC, AA, and CAA, respectively, showing a loss of the methionine side chain (48 Da) in peptides alkylated
using iodine-containing reagents. (b) MALDI-MS/MS spectra of the unmodified peptide (upper panel) and the
signal at M-48 Da (lower panel). The fragment ion series confirms that the mass difference of 48 Da results
from a loss of the methionine side chain (reproduced from [11])
3.3.7 Validation of To provide final proof that the identified spectra indeed originate
Identified Modification from the proposed modification(s), experiments with synthetic
(s) Using Synthetic peptides should be performed. In the case of chemical artifacts,
Peptides this procedure is straightforward as any synthetic peptide contain-
ing the amino acid(s) susceptible to the modification can be used
(in our example study methionine). In case of biological modifica-
tions, ideally one or several of the peptides found to be modified
should be chemically synthesized and should be measured using the
same mass spectrometer in order to generate a reference spectrum,
to which the initial samples can be compared to. If the identified
modification is indeed correct, the fragment ion patterns are
expected to match perfectly. In our example study, we used a
peptide containing methionine, incubated it with the different
alkylation reagents and measured the sample by MALDI-MS/MS
(Fig. 8). This showed clearly that only iodine-containing reagents
are resulting in an unspecific alkylation of methionine followed by
the loss of its side chain (see Note 23).
4 Notes
References
1. Aebersold R, Mann M (2016) Mass- 9:4632–4641. https://doi.org/10.1002/
spectrometric exploration of proteome struc- pmic.200900398
ture and function. Nature 537:347–355. 11. Müller T, Winter D (2017) Systematic evalua-
https://doi.org/10.1038/nature19949 tion of protein reduction and alkylation reveals
2. Kalli A, Smith GT, Sweredoski MJ et al (2013) massive unspecific side effects by iodine-
Evaluation and optimization of mass spectro- containing reagents. Mol Cell Proteomics
metric settings during data-dependent acquisi- 16:1173–1187. https://doi.org/10.1074/
tion mode: focus on LTQ-orbitrap mass mcp.M116.064048
analyzers. J Proteome Res 12:3071–3086. 12. Laemmli UK (1970) Cleavage of structural
https://doi.org/10.1021/pr3011588 proteins during the assembly of the head of
3. Eng JK, McCormack AL, Yates JR (1994) An bacteriophage T4. Nature 227:680–685.
approach to correlate tandem mass spectral https://doi.org/10.1038/227680a0
data of peptides with amino acid sequences in 13. Boersema PJ, Raijmakers R, Lemeer S et al
a protein database. J Am Soc Mass Spectrom (2009) Multiplex peptide stable isotope
5:976–989. https://doi.org/10.1016/1044- dimethyl labeling for quantitative proteomics.
0305(94)80016-2 Nat Protoc 4:484–494. https://doi.org/10.
4. Griss J, Perez-Riverol Y, Lewis S et al (2016) 1038/nprot.2009.21
Recognizing millions of consistently unidenti- 14. Shevchenko A, Wilm M, Vorm O et al (1996)
fied spectra across hundreds of shotgun prote- Mass spectrometric sequencing of proteins
omics datasets. Nat Methods 13:651–656. from silver-stained polyacrylamide gels. Anal
https://doi.org/10.1038/nmeth.3902 Chem 68:850–858. https://doi.org/10.
5. Nielsen ML, Savitski MM, Ra Z (2006) Extent 1021/ac950914h
of modifications in human proteome samples 15. Chin Y, Aiken GR, O’Loughlin E (1994)
and their effect on dynamic range of analysis in Molecular weight, polydispersity, and spectro-
shotgun proteomics. Mol Cell Proteomics scopic properties of aquatic humic substances.
5:2384–2391. https://doi.org/10.1074/ Environ Sci 28:1853–1858. https://doi.org/
mcp.M600248-MCP200 10.1021/es00060a015
6. Nesvizhskii AI, Roos FF, Grossmann J et al 16. Williams A, Frasca V (2001) Ion-exchange
(2006) Dynamic spectrum quality assessment chromatography. Curr Protoc Protein Sci
and iterative computational analysis of shotgun 15:8.2.1–8.2.30
proteomic data: toward more efficient identifi- 17. Chen J, Lee CS, Shen Y et al (2002) Integra-
cation of post-translational modifications, tion of capillary isoelectric focusing with capil-
sequence polymorphisms, and novel peptides. lary reversed-phase liquid chromatography for
Mol Cell Proteomics 5:652–670. https://doi. two-dimensional proteomics separation. Elec-
org/10.1074/mcp.M500319-MCP200 trophoresis 23:3143–3148. https://doi.org/
7. Chick JM, Kolippakkam D, Nusinow DP et al 10.1002/1522-2683(200209)23:18<3143::
(2015) A mass-tolerant database search identi- AID-ELPS3143>3.0.CO;2-7
fies a large proportion of unassigned spectra in 18. Nühse TS, Stensballe A, Jensen ON et al
shotgun proteomics as modified peptides. Nat (2003) Large-scale analysis of in vivo phos-
Biotechnol 33:743–749. https://doi.org/10. phorylated membrane proteins by immobilized
1038/nbt.3267 metal ion affinity chromatography and mass
8. Tanner S, Shu H, Frank A et al (2005) spectrometry. Mol Cell Proteomics
InsPecT: identification of posttranslationally 2:1234–1243. https://doi.org/10.1074/
modified peptides from tandem mass spectra. mcp.T300006-MCP200
Anal Chem 77:4626–4639. https://doi.org/ 19. Beausoleil SA, Jedrychowski M, Schwartz D
10.1021/ac050102d et al (2004) Large-scale characterization of
9. Jensen ON (2004) Modification-specific pro- HeLa cell nuclear phosphoproteins. Proc Natl
teomics: characterization of post-translational Acad Sci 101:12130–12135. https://doi.org/
modifications by mass spectrometry. Curr Opin 10.1073/pnas.0404720101
Chem Biol 8:33–41. https://doi.org/10. 20. Michel PE, Reymond F, Arnaud IL et al (2003)
1016/j.cbpa.2003.12.009 Protein fractionation in a multicompartment
10. Zhao Y, Jensen ON (2009) Modification- device using Off-GelTM isoelectric focusing.
specific proteomics: strategies for characteriza- Electrophoresis 24:3–11. https://doi.org/10.
tion of post-translational modifications using 1002/elps.200390030
enrichment techniques. Proteomics
Identification of Unexpected Protein Modifications by Mass Spectrometry. . . 251
Abstract
Diabetes mellitus is caused by either loss of pancreatic islets β-cells (Type 1 Diabetes, T1D), insufficient
insulin release in the islet β-cells coupled with insulin resistance in target tissues (Type 2 Diabetes, T2D), or
impaired insulin release (genetic forms of diabetes and, possibly, T1D subtypes). The investigation of the
islet proteome could elucidate facets of the pathogenesis of diabetes. Enzymatically isolated and cultured
(EIC) islets are frequently used to investigate biochemical signaling pathways that could trigger β-cell
changes and death in diabetes. However, they cannot fully reflect the natural protein composition and
disease process of in vivo islets due to the stress from isolation procedures and in vitro culture. The laser
capture microdissection method employs a high-energy laser source to separate the desired cells from the
remaining tissue section in an environment which is well conserved and close to the natural condition.
Here, we describe a label-free proteomic workflow of laser capture microdissected (LCM) human islets
from fresh-frozen pancreas sections of cadaveric donors to obtain an accurate and unbiased profile of the
pancreatic islet proteome. The workflow includes preparation of frozen tissue section, staining and dehy-
dration, LCM islets collection, islet protein digestion, label-free Liquid Chromatography-Tandem Mass
Spectrometry (LC-MS/MS), database search, and statistical analysis.
Key words LCM human pancreatic islets, Label-free proteomics, LC-MS/MS, MaxQuant, Perseus
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_16, © Springer Science+Business Media, LLC, part of Springer Nature 2019
253
254 Lina Zhang et al.
models have some limitations: they do not fully reflect what hap-
pens in vivo due to a lack of the natural environment where islets
exist and due to the changes in cell physiology induced by isolation
and culture. The procedure of enzymatic isolation of pancreatic
islets causes major structural changes and induces upregulation of
stress-related genes in islets [4]. Furthermore, EIC islets frequently
contain a significant percentage of contaminating acinar cells and
duct cells [5]. Alternatively, human pancreatic tissue can be col-
lected from cadaveric individuals and preserved frozen for further
laser-capture microdissected (LCM) isolation. LCM employs a
high-energy laser source to separate the desired cells from the
remaining tissue section [6], a strategy that can minimize the
contamination of surrounding tissue. LCM isolation also enables
the extraction of samples from an environment which is well con-
served and close to the natural condition, to better investigate cell
physiology [7], cell biology [8], cell transcriptome [4], and prote-
ome [9]. The exploration of the proteome signature of LCM islets
with an unbiased method may provide information on the changes
of protein composition occurring in dysfunctional islets, even with
limited sample amounts, which may facilitate understanding of the
pathogenesis of diabetes.
Here we describe a workflow for label-free proteomic analysis
of LCM islets obtained from sections of fresh-frozen human pan-
creas. This method enables accurate and unbiased profiling of the
pancreatic islet proteome. The strategy avoids enzymatic treatment
for cell dissociation and in vitro culture, and is designed to maintain
protein composition close to that of the original tissue. The
method can be easily adapted to other tissues, organs, and species.
The workflow covers preparation of frozen tissue sections, immu-
nohistochemical staining of reference sections, staining and dehy-
dration for LCM, LCM of pancreatic islets and acinar tissue,
preparation of samples for proteomic analysis, label-free Liquid
Chromatography-Tandem Mass Spectrometry (LC-MS/MS) data
acquisition, database search for protein identification, quantifica-
tion, and statistical analysis for the determination of the proteins
differentially expressed between LCM islets and LCM acinar tissue.
2 Materials
3 Methods
3.1 Preparation of 1. Exercise care while processing pancreatic tissue: avoid squeez-
Frozen Tissue Sections ing or stretching the tissue, use a scalpel to obtain blunt cuts.
2. Resect pancreatic tissue blocks from the neck region of cadav-
eric pancreata from organ donors.
3. Collect tissue fragments of approximately 1 cm 0.5 cm and
position fragments in the center of cryomolds.
4. Embed tissue in Tissue-Tek O.C.T. compound and immedi-
ately freeze at 80 C by placing the mold holding the tissue
on top of dry ice (see Note 6).
5. Cut the blocks into sections of 10 μm thickness with a cryo-
tome, with temperature set at 20 C (see Note 7).
5. Transfer three pancreas sections onto each of 10 Leica PEN
Membrane slides.
6. Prepare extra reference sections on a regular glass slide for
insulin immunohistochemistry and mapping (see Note 8).
3.2 Immuno- 1. Fix the reference sections in 10% formalin for 15 min (see Note 9).
histochemical Staining 2. Wash four times in PBS.
of Reference Sections
3. Leave a drop of PBS on each tissue section and draw a circle
with a PAP pen to surround each section.
4. Remove the PBS.
5. Add Peroxo-Block™ for 45 s. Wash immediately.
6. Add 100 μL of anti-insulin antibody clone K36aC10 solution
(dilution 1:300) to each section to completely cover tissue.
7. Incubate in a humidified chamber at room temperature for
60 min.
8. Rinse with PBS for 5 min, three times.
9. Add 100 μL of secondary antibody to each section to
completely cover tissue and incubate for 10 min.
10. Rinse with PBS for 5 min, three times.
11. Add enough enzyme conjugate solution (from the Histostain
Plus Broadspectrum AEC kit) to each section to completely
cover tissue and incubate for 10 min.
12. Rinse with PBS for 2 min, three times.
13. Add chromogen AEC Single Solution and incubate 5–10 min.
14. Scan reference sections with a PathScan Enabler IV instrument
to obtain maps of the entire sections and identify stained
insulin-containing islets.
3.3 Staining and 1. The staining and dehydration protocol is performed with
Dehydration for LCM 8 clean Coplin jars, prefilled with 50 mL of 70% (jar # 1–5),
90% (jar #6), and 100% (jar #7 and 8) ethanol.
258 Lina Zhang et al.
2. Jars # 1–5 are maintained chilled on ice during the staining, jars
# 6–8 are maintained at room temperature to avoid condensa-
tion after dehydration.
3. The PEN membrane slides with tissue sections are dipped for
30 s in each jar from #1 to #3 of the ethanol series.
4. After jar #3, each slide is drained by gently placing the side edge
of the glass on a Kimwipe, then placed horizontally, with the
tissue sections on top.
5. The sections are stained for 90 s by adding 200 μL of Toluidine
blue O staining solution, then drained and transferred to jar #
4, to continue dehydration (see Note 10).
6. The slides are dipped for 30 s in each jar following the numeri-
cal order (jar # 4–8), to obtain dehydration.
7. The stained and dehydrated slide is drained with a Kimwipe and
placed under a laminar flow hood for 4 min to enable ethanol
evaporation.
8. The slide is placed in a slide box containing the desiccant
Drierite wrapped in Kimwipes and closed with tape (see
Note 11).
3.4 LCM of 1. The stage of the Leica Microscope LS LMD system is posi-
Pancreatic Islets and tioned in a clear acrylic box (microdissection chamber) where
Acinar Tissue (see the atmosphere can be controlled.
Note 12) 2. 1 h before microdissection, the workplace is cleaned and the
microdissection chamber is dehydrated using 2 kg of fresh
Drierite to minimize the humidity and enable membrane
microdissection.
3. The Leica LMD software is used to set up the laser (see
Table 1), to initialize the instrument and to control the move-
ments of the laser on the tissue section.
4. The PEN membrane slide with the stained tissue is positioned
on the stage, with the tissue facing down.
5. Empty sterile collection tubes are placed under the cutting area
(RNAse-free Eppendorf tubes, 500 μL volume, flat cap).
6. The scans of the reference sections are used to map the insulin-
containing islets.
7. Pancreatic islets are identified in the toluidine-stained tissue by
visualizing in bright field and in phase-contrast with 10 mag-
nification. In bright field, pancreatic islets appear as clusters of
cells with lightly colored cytoplasm, whereas the surrounding
acinar tissue is composed by cells with darker cytoplasm (see
Fig. 1). In phase-contrast, islet cells appear finely granulated.
Visualize islet borders at 20 magnification (see Note 13).
Label Free Proteomics of LCM Collected Human Pancreatic Islets 259
Table 1
System configuration for laser capture microdissection with the Leica LMD
instrument
3.5 Conversion of The total volume of isolated islets can be expressed as number of
LCM Areas to Volumes islet equivalents (IEQ) [10]. An IEQ corresponds to the volume of
and to IEQ a “standard” islet, a sphere with a diameter d ¼ 150 μm and a
volume of VIEQ ¼ 1.77 106 μm3. 1 IEQ contains approximately
1560 islet cells [11]. The area of laser-captured tissue is recorded,
and the volume is calculated by multiplying the total area collected
for the thickness of the tissue section (10 μm). The target total
volume of each microdissected sample is 1.06 107 μm3,
260 Lina Zhang et al.
3.6 Protein Digestion 1. Add 6 μL of 1% pps silent surfactant (PPS) to extract and
solubilize hydrophobic proteins.
2. Add 1.5 μL of 50 mM DTT and incubate at 95 C for 6 min.
3. Sonicate sample for 3 min.
4. Alkylate with 7.5 μL 50 mM iodoacetamide for 25 min at 45 C
in the dark.
5. Add 1 μg stock trypsin at 37 C overnight.
6. Hydrolyze PPS by adding 12 μL 2 M HCl at room temperature
for 2 h.
7. Centrifuge samples at 16,000g for 12 min and separate the
supernatant for LC-MS/MS analysis (see Note 16).
3.7 LC-MS/MS Protocols for LC-MS/MS analysis can vary because of diversity of
Analysis LC systems (manufacturer, column, solvent composition, gradient,
flow rate, etc.) and MS instruments (manufacturer, electrospray
condition, fragmentation, MS parameters, analyzer, etc.). The fol-
lowing is the practice routinely used in our laboratory.
1. The LC-MS/MS platform consists of an UltiMate 3000
RSLCnano system and a Q Exactive HF mass spectrometer
coupled with an EASY-Spray ion source (ThermoFisher
Scientific).
2. Peptide separation is performed on a PepMap C18 analytical
column (2 μm particle, 50 cm 75 μm, ThermoFisher Scien-
tific). Injection volume is 2.5 μL (0.5 μg peptide amount
loaded into column) per sample (see Note 17).
3. A binary solvent system consisting of 0.1% FA in water (solvent
A) and 0.1% FA in CH3CN (solvent B) is used at a flow rate of
250 nL min1 (see Note 18).
4. LC separation is performed using the following gradient
setting: hold at 4% B for 3 min (for desalting), from 4 to 8%
B in 0.1 min, 8 to 40% B in 90 min (effective gradient), 40 to
90% B in 0.1% min, hold at 90% B for 10 min (for washing
column), 90% to 4% B in 0.1 min, and hold at 4% B for 17 min
for re-equilibrating column (see Note 19).
5. MS data are acquired in profile mode and resolution for full
scan (400–2000 m/z) is set to 120,000 (at m/z 200) with
maximum ion injection time of 50 ms, and automatic gain
control (AGC) target of 1e6.
Label Free Proteomics of LCM Collected Human Pancreatic Islets 261
3.8 Database Search Many database search software packages are available for this pur-
for Protein pose. MaxQuant is demonstrated here [12].
Identification and
1. The acquired datasets (.raw files) are analyzed using MaxQuant
Quantification and the built-in Andromeda search engine against a UniProt
human database (see Note 20).
2. Variable modifications include protein N-terminal acetylation
and methionine oxidation.
3. Fixed modifications contain cysteine carbamidomethylation.
4. A maximum of two missed cleavages are allowed for the search.
5. Trypsin/P is selected as the specific proteolytic enzyme (see
Note 21).
6. For label-free quantification, “match between runs” is selected
(see Note 22).
7. The false discovery rate (FDR) cutoff used for both peptides
and proteins is 0.01 (1%) using decoy database.
8. Only the razor/unique peptides are used for quantitative
calculations.
9. The other parameters are the default settings in MaxQuant
software for processing orbitrap-type data.
4 Notes
21. The selection of enzyme used for search is based on the enzyme
that is chosen for protein digestion in Subheading 3.6.
22. “Match between runs” should be selected because it can
improve the search results for less missing values.
Acknowledgements
References
Targeted Proteomics
Yun Chen and Liang Liu
Abstract
Targeted proteomics detects proteins of interest with high sensitivity, quantitative accuracy, and reproduc-
ibility. In a targeted proteomics assay, surrogate peptides are generated by proteolytic digestion of target
proteins and selected reaction monitoring (SRM) assays are developed to quantify these peptides using
liquid chromatography-tandem mass spectrometry (LC-MS/MS). In this report, we describe the details of
quantitative analysis of target protein in cells and tissue samples.
Key words Targeted proteomics, Liquid chromatography-tandem mass spectrometry, Protein quan-
tification, Cells and tissue samples
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_17, © Springer Science+Business Media, LLC, part of Springer Nature 2019
265
266 Yun Chen and Liang Liu
Intensity
ESI Retention time
Source Q1 Q2 Q3 Detector
Fig. 1 A schematic of targeted proteomics strategy using a triple quadrupole mass spectrometer operating in
SRM mode
2 Materials
2.5 LC-MS/MS An Agilent Series 1200 HPLC system and a 6410 Triple Quad
Instrument LC/MS mass spectrometer (see Note 3).
3 Methods
3.1.2 Tissue 1. Thaw tissue samples to room temperature and rinse thoroughly
Homogenization with deionized water.
2. Remove fat tissue and cut the remaining tissue into small pieces
and transfer them to tubes.
3. Weigh approximately 50 mg of tissue and suspend it in tissue
homogenization buffer (see Note 4).
4. Homogenize the tissue suspension on ice using a Bio-Gen
PRO200 homogenizer.
3.2 Protein 1. Add cold RIPA buffer to sample. Keep the sample on ice for
Extraction 45 min and vortex every 15 min.
3.2.1 Cytosolic Proteins 2. Centrifuge the sample at 14,000 g for 15 min.
3. Transfer the supernatant to a new tube and determine protein
concentration using a BCA protein assay kit.
3.2.2 Membrane Proteins 1. Centrifuge the sample at 10,000 g for 10 min, and resuspend
the pellet in 500 μL of membrane protein extraction buffer.
2. Incubate the sample on ice for 30 min, and vortex every 10 min,
followed by incubation at 37 C for 10 min (see Note 5).
3. Centrifuge the mixture at 10,000 g for 3 min to separate
detergent and aqueous phases.
4. Add 500 μL of 1% extraction buffer and 500 μL of wash buffer
to the aqueous and detergent phases, respectively. Then repeat
the above incubation and centrifugation steps.
5. Combine the detergent phases and precipitate proteins using
cold acetone (pre-frozen at 20 C for 1 h before use).
6. Allow acetone to evaporate at room temperature.
7. Dissolve the protein pellet in 1% SDS solution.
8. Determine protein concentration of the obtained sample using
a BCA protein assay kit.
3.5 Surrogate 1. The most critical step in the establishment of a targeted prote-
Peptide Selection omics assay is the selection of proteolytic peptides that (1) are
unique to a candidate protein, (2) can provide an adequate
response, (3) are completely digested, and (4) can generate
high-quality SRM [16, 17] (see Note 7).
2. The uniqueness of selected surrogate peptides is normally
checked using a BLAST search. For example, the peptides of
434STTVQLMQR442, 674GSQAQDR680, and 368IIDNKP-
SIDSYSK380 were found to be unique to P-gp (accession
no. P08183 (MDR1 HUMAN), gi: 2506118) [8].
3. A LC-MS/MS analysis with a list of SRM transition pairs based
on either in silico prediction or spectral evidence from public
repositories is usually performed to identify the peptide with
the greatest abundance [18] (see Note 8). Synthetic references
peptides are usually employed for confirmation (Fig. 3).
4. The digestion efficiency is evaluated using the substrate peptide
containing the same peptide sequence (to mimic a piece of the
target protein). The digestion efficiency was calculated by com-
paring the response ratios of the tryptic peptide after digestion
and the equimolar synthetic peptide standard in the digestion
(Fig. 4) [19].
5. Optimize SRM transitions (see Notes 9–11].
a 100
y8 15eV
y1
146 b2 y2 b8
226233 681
b4 y4 y5
b3 598 y7
455483
341
798
300
Intensity, cps
200
100
0
1 2 3 4 5 6 7
Time, min
Fig. 3 The product ion spectrum and LC-MS/MS chromatogram of 368IIDNKPSIDSYSK380, a surrogate peptide
of P-gp. The characteristic sequence-specific b ions and y ions, and retention time are indicative of this
peptide (reproduced from ref. 9 with permission from Elsevier)
3.7 Immuno- 1. Add BioMagPlus IgG beads that are pre-incubated with anti-
Depleted Matrix target-protein antibody (see Note 12).
Preparation 2. Incubate the mixture at 25 C for 2 h with shaking.
3. Magnetically separate the beads and collect the supernatant.
4. Rinse the beads with 1% SDS solution and combine the eluate
with the above supernatant.
5. Examine this synthetic matrix using Western blotting and
LC-MS/MS-based targeted proteomics assay (Fig. 5).
a b
6x103 6x103 Undigested peptide GKSTTVQLMQRLY
Undigested peptide GKSTTVQLMQRLY
5 5
Intensity, cps
Intensity, cps
4 4
3 3
2 2
1 1
0 0
1 2 3 4 5 6 7 1 2 3 4 5 6 7
Time,min Time,min
3 3
6x10 6x10
Digested peptide STTVQLMQR Digested peptide STTVQLMQR
5 5
Intensity, cps
Intensity, cps
4 4
3 3
2 2
1 1
0 0
1 2 3 4 5 6 7 1 2 3 4 5 6 7
Time,min Time,min
Fig. 4 LC-MS/MS chromatograms of STTVQLMQ and its substrate peptide GKSTTVQLMQRLY (a) before and (b)
after tryptic digestion (reproduced from ref. 8 with permission from Elsevier)
Fig. 5 The Western blotting image and LC-MS/MS chromatogram for P-gp depleted tissue extract (reproduced
from ref. 9 with permission from Elsevier)
10, 25, 50, 100, 250, 400, 700, and 1000 ng/mL. The QC
standards for the lower limit of quantification (LLOQ), low
QC, mid QC, and high QC were prepared at 10, 30, 200, and
800 ng/mL, respectively, and frozen prior to use.
3. Construct the calibration curve using a weighted linear regres-
sion model with a weighting factor of 1/x2. The relative peak
area ratio of the analyte and the stable isotope-labeled internal
standard was plotted as a function of concentration (Fig. 6).
4. The precision and accuracy of the assay were assessed by
observing the response of the QC samples with four different
272 Yun Chen and Liang Liu
3.5
3 r2 = 0.9974
2.5
Area Ratio
2
1.5
0.5
0
0 200 400 600 800 1000
Concentration (ng/mL)
Table 1
Accuracy and precision for the QC samples using P-gp depleted matrix (reproduced from ref. 8 with
permission from Elsevier)
5x104
MCF-7/WT
4
Intensity, cps
3
0
1 2 3 4 5 6 7
Time, min
5x104
MCF-7/ADR
4
Intensity, cps
0
1 2 3 4 5 6 7
Time, min
4 Notes
1. DTT may not be stable and should not be stored for long
periods of time, so make up fresh before use [20].
2. If necessary, add protease and phosphatase inhibitors to RIPA
buffer immediately before use.
3. It should be noted that other instrument platforms (e.g., ion
trap, Q-TOF) are also capable of performing SRM like experi-
ments; however, quadrupole mass spectrometer is the preferred
technology for quantification and the most accessible instru-
ment in routine research and clinical lab [21].
4. Tissue collection must be approved by the Medical Ethics
Review.
274 Yun Chen and Liang Liu
2.0
1.5
ng/mg*
1.0
0.5
0.0
Normal Tumor
3.0x104
a
2.5
2.0
Intensity, cps
1.5
1.0 P3
P2
0.5 P1
P0
0.0
2 4 6 8 10
Time, min
1.5x104
b
1.2
Intensity, cps
0.9
P3
0.6
P2
0.3
P1
P0
0.0
2 4 6 8 10
Time, min
Fig. 9 The LC/MS-MS chromatograms of (a) HSP27 and (b) P-gp in freshly prepared (P0) and passage 1, 2,
3 (P1, P2, P3) MCF-7/WT cells after the treatment of DOX. The amounts are also listed in the table (reproduced
from ref. 15 with permission from Elsevier)
References
1. Yocum AK, Chinnaiyan AM (2009) Current 7. Doerr A (2013) Mass spectrometry-based tar-
affairs in quantitative targeted proteomics: geted proteomics. Nat Methods 10(1):23–23
multiple reaction monitoring–mass spectrome- 8. Yang T, Xu F, Xu J, Fang D, Yu Y, Chen Y
try. Brief Funct Genomics Proteomics 8 (2013) Comparison of liquid chromatogra-
(2):145–157 phy–tandem mass spectrometry-based targeted
2. Pan S, Aebersold R, Chen R, Rush J, Goodlett proteomics and conventional analytical meth-
DR, McIntosh MW, Zhang J, Brentnall TA ods for the determination of P-glycoprotein in
(2008) Mass spectrometry based targeted pro- human breast cancer cells. J Chromatogr B
tein quantification: methods and applications. J 936:18–24
Proteome Res 8(2):787–797 9. Yang T, Chen F, Xu F, Wang F, Xu Q, Chen Y
3. Parker CE, Pearson TW, Anderson NL, Borch- (2014) A liquid chromatography–tandem mass
ers CH (2010) Mass-spectrometry-based clini- spectrometry-based targeted proteomics assay
cal proteomics–a review and prospective. for monitoring P-glycoprotein levels in human
Analyst 135(8):1830–1838 breast tissue. Clin Chim Acta 436:283–289
4. Domon B, Aebersold R (2006) Mass spec- 10. Yu Y, Xu J, Liu Y, Chen Y (2012) Quantifica-
trometry and protein analysis. Science 312 tion of human serum transferrin using liquid
(5771):212–217 chromatography–tandem mass spectrometry
5. Marx V (2013) Targeted proteomics. Nat based targeted proteomics. J Chromatogr B
Methods 10(1):19–22 902:10–15
6. Doerr A (2010) Targeted proteomics. Nat 11. Yang T, Xu F, Zhao Y, Wang S, Yang M, Chen
Methods 7(1):34–34 Y (2014) A liquid chromatography-tandem
Targeted Proteomics 277
Abstract
Staphylococcus aureus is a major human pathogen that can readily acquire antibiotic resistance. For instance,
methicillin-resistant S. aureus represents a major cause of hospital- and community-acquired bacterial
infections. In this chapter, we first provide a detailed protocol for obtaining unbiased and reproducible
S. aureus metabolic profiles. The resulting intracellular metabolome is then analyzed in an untargeted
manner by using both hydrophilic interaction liquid chromatography and pentafluorophenyl-propyl col-
umns coupled to high-resolution mass spectrometry. Such analyses are done in conjunction with our
in-house spectral database to identify with high confidence as many meaningful S. aureus metabolites as
possible. Under these conditions, we can routinely monitor more than 200 annotated S. aureus metabo-
lites. We also indicate how this protocol can be used to investigate the metabolic differences between
methicillin-resistant and susceptible strains.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_18, © Springer Science+Business Media, LLC, part of Springer Nature 2019
279
280 Sandrine Aros-Calt et al.
poor clinical outcomes (~30% mortality rate) [4, 5]. Although the
level of MRSA isolates has decreased over time in Europe, some
European countries still report 25% or more of invasive S. aureus
isolates as MRSA [5]. Acquisition of antibiotic resistance can par-
tially be explained by the misuse and mishandling of drugs to treat
infections. Unfortunately, first antibiotic resistances often appear
very soon after the introduction of an antibiotic. When combined
with drug companies’ reluctance to develop novel antibiotics, this
has led to serious health concerns [6]. Therefore, numerous efforts
have been made to better understand antibiotic-resistance mechan-
isms in order to potentially find new therapeutic strategies [7–10].
In that objective, metabolomics can greatly help digging deeper
into the pathogenicity and biochemical mechanisms behind the anti-
biotic resistance of MRSA strains. Indeed, metabolomics provides the
most direct assessment of cellular phenotype and is well suited to the
quantitative and dynamic monitoring of bacterial metabolism varia-
tions in response to particular environmental conditions. Recently
published papers clearly support this statement regarding the study
MRSA strains [11–15]. Just to cite a few, the pioneering work of
Liebeke et al. regarding S. aureus metabolomics demonstrated the
possibility to simultaneously analyze about 80 S. aureus metabolites
by using liquid chromatography coupled to mass spectrometry
(LC-MS). They reported that the S. aureus central carbon metabo-
lism, which notably includes energy transfer molecules like nucleo-
tides, sugar mono- and bi-phosphates, and cofactors, can be used to
monitor metabolic disturbances in response to genetic deletions of
serine/threonine kinase and phosphatase [11].
Keaton et al. investigated the changes in metabolic pathways
when treating MRSA strains with subinhibitory concentrations of
β-lactam antibiotics. They used a combination of LC-MS and gas
chromatography coupled to mass spectrometry (GC-MS) techni-
ques to highlight significant increases in tricarboxylic acid (TCA)
cycle intermediates under the studied conditions, which tended to
demonstrate that the energy production of MRSA strains was
redirected to supply the cell wall synthesis/metabolism and so,
contributed to their survival in the presence of β-lactam antibiotics
[16]. In a recent study, Dorries et al. investigated the impact of five
antibiotics with different cellular targets on S. aureus metabolism
by studying its global intra- and extracellular metabolic profiles
thanks to LC-MS, GC-MS, and NMR techniques [13]. Their ana-
lytical platforms largely covering primary metabolism allowed them
to highlight accumulation as well as depletion of metabolites from
various biosynthetic pathways, such as central carbon and amino
acid metabolism; peptidoglycan, purine, and pyrimidine synthesis
[13]. Also, Ammons et al. successfully implemented an
NMR-based strategy to distinguish MRSA from MSSA strains
based on the quantitative monitoring of 40 intracellular metabo-
lites (essentially amino acids and TCA-cycle intermediates) [12].
S. aureus Metabolomics 281
2 Materials
2.2 Bacterial Culture 1. Mueller Hinton II broth (MHII, cation-adjusted) from Becton
Dickinson (product reference 212322, Franklin Lakes, NJ).
Dissolve 22 g of MHII medium in 1 L of ultrapure water,
autoclave the mixture at 120 C for 20 min, and store at 4 C
until use.
2. Colombia (COS) agar containing 5% sheep blood plates
(bioMérieux).
3. Cefoxitin (Sigma-Aldrich, Saint Quentin Fallavier, France).
4. Petri dishes (Sigma-Aldrich).
5. Erlenmeyer baffled cell culture flasks (Sigma-Aldrich).
6. Sterile inoculating loops and needles (VWR, Fontenay-sous-
Bois, France).
7. Serological pipettes (5 mL, Sigma-Aldrich).
8. Minitron II rotary shaker (Infors HT, Bottmingen-Basel,
Switzerland).
9. Eppendorf BioPhotometer (Eppendorf, Montesson, France)
or equivalent spectrophotometer.
2.4 LC–MS Analysis 1. Sequant ZIC-pHILIC column, 2.1150 mm, 5 μm, HPLC
of Intracellular PEEK (Merck, Darmstadt, Germany).
Metabolites 2. Discovery HSF5 Pentafluorophenylpropyl (PFPP) column,
2.4.1 Chromatographic 2.1 150 mm, 5 μm (Sigma-Aldrich).
Columns
2.4.4 Internal Solutions of internal standards are prepared in pure water (Table 1).
Standards (IS)
284 Sandrine Aros-Calt et al.
Table 1
List of internal standards used for LC-MS analysis
Concentration of Compound
individual stock concentration in the
solutions (mg/mL 20 standard mixture
Compound name water) (μg/mL)
13
C1-alanine 4 200
Ethylmalonic acid 4 30
15
N1-aspartic acid 4 200
13
C1-glucose 4 200
Ampicillin 2 50
Prednisone 2 10
Dihydrostreptomycin 4 200
Roxithromycin 2 200
15
N5-AMP 1 100
15
N5-ADP 1 50
15
N5-ATP 1 50
All compounds except 15N-labeled AMP and ATP (Euriso-Top, Saint-Aubin, France)
were from Sigma-Aldrich
3 Methods
3.1 Bacterial 1. Preheat liquid and solid cultivation media a few hours at 37 C
Pre-culture and to exclude any bacterial contamination.
Culture 2. S. aureus strains are first isolated from an overnight culture at
37 C on COS plates.
3. Bacterial pre-culture is performed under aerobic conditions by
inoculating few bacterial colonies in 12.5 mL of preheated
MHII medium within a 125 mL Erlenmeyer flask (10% of
total volume to ensure sufficient aeration). Incubate the result-
ing culture medium at 37 C for 12–18 h with vigorous shak-
ing (200 rpm).
4. Withdraw an aliquot and dilute to an optical density at 600 nm
(OD600) of ~0.1 in a fresh MHII medium (in a 2 L Erlenmeyer
flask). Allow bacteria growth at 37 C with vigorous shaking
(200 rpm). For all the strains studied, the early-exponential
phase corresponded to an OD600 of 1, which was equivalent to
5 108 CFU/mL (see Notes 1 and 2).
S. aureus Metabolomics 285
3.2 Bacteria 1. Prepare the filtration device, run the vacuum pump, and con-
Sampling and dition the filter by passing through 5 mL of MHII medium.
Metabolism Quenching 2. Withdraw a 5 mL aliquot from the main culture broth (at the
targeted OD600) and fill it into the filter system to rapidly
separate bacteria from culture medium.
3. Wash the bacteria retained on the filter with 5 mL of 0.6% NaCl
to remove culture medium (see Note 3).
4. The filter is then rapidly transferred to a 50 mL Falcon tube
containing 5 mL of ice-cold 60% ethanol (see Note 4). The
tube is subsequently quickly immersed in liquid nitrogen to
quench bacterial metabolism (see Note 5) before mechanical
cell disruption. At this step, samples can be conserved several
months at 80 C.
3.3 Mechanical Cell 1. Following quenching, tubes containing bacteria on filter in the
Disruption and extraction solution are vortexed 10 times (10 s at 4 C) to
Metabolite Extraction remove cells from the filter.
2. Transfer 1 mL of the bacterial suspension into the Precellys
tubes, while the remaining 4 mL are kept at 80 C.
3. Bacterial lysis is accomplished by performing three cycles in a
Precellys 24 homogenizer for 30 s at 3800 rpm and at ~4 C.
4. Centrifuge the tubes during 5 min at 4 C and 10,000 g to
remove glass beads and cell debris.
5. Withdraw 400 μL of the supernatant and transfer into a 1.5 mL
Eppendorf tube.
6. Evaporate under a stream of nitrogen.
7. 200 μL of each sample can also be withdrawn and pooled to
obtain a Quality Control (QC) sample. 400-μL aliquots of the
resulting mixture can then be evaporated under a stream of
nitrogen.
8. Store samples and QCs at 80 C until LC-MS analyses.
3.4 LC-MS Analysis A complete detailed protocol for running LC-MS experiments will
of Intracellular not be provided hereafter. Below, we only list the main highlights of
Metabolites our approach involving a combination of two complementary
ZIC-pHILIC and PFPP columns coupled to an Orbitrap instru-
ment to obtain an optimal coverage of the S. aureus metabolome.
For additional details, the reader can refer to Boudah et al. and
Aros-Calt et al. [14, 19]. In principle, most of routine methods
used in metabolomics laboratories are expected to be used for
S. aureus metabolite analysis with limited or no modifications,
even if they make use of another type of high-resolution mass
spectrometer (e.g., Q-TOF).
286 Sandrine Aros-Calt et al.
3.4.1 LC-MS Analysis 1. Dilute eight times the 20 internal standard mixture (Table 1)
Using a ZIC-pHILIC Column in 10 mM ammonium carbonate pH 10.5.
2. Estimate the number of CFU in each dried sample tube and
adjust the resuspension volume to obtain a concentration of
1.25 107 CFU per 10 μL. The protocol provided hereafter is
provided for a final volume of 80 μL.
3. Solubilize the dried bacterial extract and QC samples in 32 μL
of the diluted internal standard mixture (step 1). After vigor-
ous mixing, incubate the resulting mixture in an ultrasonic bath
for 5 min.
4. Centrifuge at 10,000 g for 5 min at 4 C.
5. Transfer the resulting supernatant in an injection vial and add
48 μL of acetonitrile.
6. Inject 10 μL into the LC-MS system.
7. Metabolites are eluted from the column (maintained at 15 C)
at a flow rate of 200 μL/min using the gradient reported in
Table 2.
8. The Exactive Orbitrap mass spectrometer is operated in the
negative ion mode at a resolution of 50,000 at m/z 200 (full
width at half-maximum), using the following source para-
meters: Capillary voltage, 3 kV; capillary temperature,
280 C; sheath gas pressure, 60 arbitrary units; auxiliary gas
pressure, 10 arbitrary units. The detection is performed from
m/z 75 to 1000, using an injection time set at 100 ms and an
AGC target value of 3 106.
3.4.2 LC–MS Analysis 1. Dilute 20 times the 20 internal standard mixture (Table 1) in
Using a PFPP Column water containing 0.1% formic acid.
2. Estimate the number of CFU in each dried sample tube and
adjust the resuspension volume to obtain a concentration of
Table 2
Gradient conditions used for metabolite LC-MS analysis using the
ZIC-pHILIC column
Table 3
Gradient conditions used for metabolite LC-MS analysis using the PFPP
column
3.5 Analysis of 1. AMP, ADP, and ATP were quantified by the isotope dilution
LC–MS Data method using their 15N-labeled homologues, with a procedure
similar to that described by Martano et al. [20]. Under our
3.5.1 Determination of
conditions, a mass accuracy better than 3 ppm for the endoge-
the Adenylate Energy
nous nucleotides and a perfect coelution with their labeled
Charge (AEC)
homologues ensure compound identification and accurate
quantification (see Note 6).
2. Calculate the AEC by using molar concentrations with the
following formula (see Note 7):
3.5.2 Well-Designed Obtaining reliable metabolic profiling data is a complex task and
Sample Dataset and Quality particular attention should be paid to the experimental design in
Control Samples (QCs) for order to avoid any instrumental or analytical bias. Well-designed,
Reliable Untargeted robust, and reproducible metabolomics workflows commonly
Metabolomics involve column conditioning, sample randomization, and use of
QC samples. QCs are made from a pooled sample obtained by
mixing an equal volume (10–100 μL) of each sample to be studied,
and therefore constitute a representative bulk control sample so as
the signal variations of any metabolite could be reflected in the QCs
[21]. QC samples proved really useful to correct for drifts in MS
response, mass measurement accuracy, as well as chromatographic
retention time between analytical runs or batches [22, 23]. Another
particularly relevant feature of QCs implies dilution of QCs to
evaluate the corresponding MS response linearity. Under these
conditions, the metabolites showing a linear trend can be consid-
ered as analytically relevant. Figure 2 depicts a typical sample run
order used for LC-HRMS-based metabolomics.
Fig. 2 Typical sample run order for LC-HRMS. If needed, additional inter-batches
QCs can be added
S. aureus Metabolomics 289
3.5.3 Data Processing Data processing as well as statistical analysis can be performed
automatically and reproducibly online using the Workflow4meta-
bolomics (W4M) platform for computational metabolomics
[24, 25]. W4M is built on the Galaxy environment providing
intuitive and powerful features that allow the analyst to set up and
run complex workflows (Fig. 3). Four main processing steps can be
considered.
1. Preprocessing: Automatic peak detection, alignment, and
extraction using the XCMS software.
2. Normalization thanks to QCs: Correction of signal variation,
selection of analytically pertinent metabolites.
3. Statistical analyses (univariate and multivariate statistical tools).
Other statistical software such as SIMCA-P (Umetrics, Umea,
Sweden) can be used to perform multivariate data analyses such
as principal component analysis (PCA) and partial least squares
discriminant analysis (PLS-DA) or Prism (GraphPad, La Jolla,
USA) to perform univariate data analyzes (e.g., t-test).
4. Compound annotation is commonly performed considering a
10 ppm mass tolerance using either our in-house database
(including ~1000 metabolites) [14, 19, 26] or publically avail-
able databases such as KEGG [27], HMDB [28], or
METLIN [29].
5. Confirmation of metabolite annotation and identification of
other statistically relevant unknown metabolites is accomplished
by MS/MS experiments. Resulting MS/MS spectra are com-
pared to those included in our in-house database or in public
databases such as METLIN. Figure 4 summarizes our workflow
for formal metabolite identification (see Notes 8 and 9).
Fig. 3 Data treatment tools available in the W4M infrastructure. The left and right panels give the tools and
parameters that can be used to build the workflow in the central panel
290 Sandrine Aros-Calt et al.
4 Notes
Fig. 5 PLS-DA score plot of features from ZIC-pHILIC/MS fingerprints of MRSA and MSSA harvested at the
mid-exponential growth phase and then exposed to cefoxitin. The PLS-DA model was validated by a
permutation test (100 times)
292 Sandrine Aros-Calt et al.
Acknowledgements
References
1. Lowy FD (1998) Staphylococcus aureus infec- 6. Boucher HW, Corey GR (2008) Epidemiology
tions. N Engl J Med 339:520–532 of methicillin-resistant Staphylococcus aureus.
2. Chambers HF, Deleo FR (2009) Waves of Clin Infect Dis Off Publ Infect Dis Soc Am
resistance: Staphylococcus aureus in the antibi- 46(Suppl 5):S344–S349
otic era. Nat Rev Microbiol 7:629–641 7. Sommer MOA, Dantas G (2011) Antibiotics
3. Gardete S, Tomasz A (2014) Mechanisms of and the resistant microbiome. Curr Opin
vancomycin resistance in Staphylococcus aureus. Microbiol 14:556–563
J Clin Invest 124:2836–2840 8. Munck C, Gumpert HK, Wallin AIN et al
4. Chastre J, Blasi F, Masterton RG et al (2014) (2014) Prediction of resistance development
European perspective and update on the man- against drug combinations by collateral
agement of nosocomial pneumonia due to responses to component drugs. Sci Transl
methicillin-resistant Staphylococcus aureus after Med 6:262ra156
more than 10 years of experience with linezo- 9. Belenky P, Ye JD, Porter CBM et al (2015)
lid. Clin Microbiol Infect Off Publ Eur Soc Bactericidal antibiotics induce toxic metabolic
Clin Microbiol Infect Dis 20(Suppl 4):19–36 perturbations that lead to cellular damage. Cell
5. Hassoun A, Linden PK, Friedman B (2017) Rep 13:968–980
Incidence, prevalence, and management of 10. Ling LL, Schneider T, Peoples AJ et al (2015)
MRSA bacteremia across patient populations- A new antibiotic kills pathogens without
a review of recent developments in MRSA detectable resistance. Nature 517:455–459
management and treatment. Crit Care Lond 11. Liebeke M, Meyer H, Donat S et al (2010) A
Engl 21:211 metabolomic view of Staphylococcus aureus and
its ser/thr kinase and phosphatase deletion
S. aureus Metabolomics 293
mutants: involvement in cell wall biosynthesis. of serum and plasma using gas chromatography
Chem Biol 17:820–830 and liquid chromatography coupled to mass
12. Ammons MCB, Tripet BP, Carlson RP et al spectrometry. Nat Protoc 6:1060–1083
(2014) Quantitative NMR metabolite profiling 23. Dunn WB, Wilson ID, Nicholls AW et al
of methicillin-resistant and methicillin- (2012) The importance of experimental design
susceptible Staphylococcus aureus discriminates and QC samples in large-scale and MS-driven
between biofilm and planktonic phenotypes. J untargeted metabolomic studies of humans.
Proteome Res 13:2973–2985 Bioanalysis 4:2249–2264
13. Dörries K, Schlueter R, Lalk M (2014) Impact 24. Giacomoni F, Le Corguillé G, Monsoor M et al
of antibiotics with various target sites on the (2015) Workflow4Metabolomics: a collabora-
metabolome of Staphylococcus aureus. Antimi- tive research infrastructure for computational
crob Agents Chemother 58:7151–7163 metabolomics. Bioinformatics 31:1493–1495
14. Aros-Calt S, Muller BH, Boudah S et al (2015) 25. Guitton Y, Tremblay-Franco M, Le Corguillé
Annotation of the Staphylococcus aureus Meta- G et al (2017) Create, run, share, publish, and
bolome using liquid chromatography coupled reference your LC-MS, FIA-MS, GC-MS, and
to high-resolution mass spectrometry and NMR data analysis workflows with the Work-
application to the study of methicillin resis- flow4Metabolomics 3.0 Galaxy online infra-
tance. J Proteome Res 14:4863–4875 structure for metabolomics. Int J Biochem
15. Schelli K, Zhong F, Zhu J (2017) Comparative Cell Biol 93:89–101
metabolomics revealing Staphylococcus aureus 26. Roux A, Xu Y, Heilier J-F et al (2012) Annota-
metabolic response to different antibiotics. tion of the human adult urinary metabolome
Microb Biotechnol 10:1764–1774 and metabolite identification using ultra high
16. Keaton MA, Rosato RR, Plata KB et al (2013) performance liquid chromatography coupled
Exposure of clinical MRSA heterogeneous to a linear quadrupole ion trap-Orbitrap mass
strains to β-lactams redirects metabolism to spectrometer. Anal Chem 84:6429–6437
optimize energy production through the TCA 27. Kanehisa M, Goto S (2000) KEGG: Kyoto
cycle. PLoS One 8:e71025 encyclopedia of genes and genomes. Nucleic
17. Sumner LW, Amberg A, Barrett D et al (2007) Acids Res 28:27–30
Proposed minimum reporting standards for 28. Wishart DS, Tzur D, Knox C et al (2007)
chemical analysis chemical analysis working HMDB: the human Metabolome database.
group (CAWG) metabolomics standards initia- Nucleic Acids Res 35:D521–D526
tive (MSI). Metabolomics 3:211–221 29. Smith CA, O’Maille G, Want EJ et al (2005)
18. Liebeke M, Lalk M (2014) Staphylococcus METLIN: a metabolite mass spectral database.
aureus metabolic response to changing envi- Ther Drug Monit 27:747–751
ronmental conditions—a metabolomics per- 30. Meyer H, Liebeke M, Lalk M (2010) A proto-
spective. Int J Med Microbiol 304:222–229 col for the investigation of the intracellular
19. Boudah S, Olivier M-F, Aros-Calt S et al Staphylococcus aureus metabolome. Anal Bio-
(2014) Annotation of the human serum meta- chem 401:250–259
bolome by coupling three liquid chromatogra- 31. Chapman AG, Fall L, DE A (1971) Adenylate
phy methods to high-resolution mass energy charge in Escherichia coli during growth
spectrometry. J Chromatogr B Analyt Technol and starvation. J Bacteriol 108:1072–1086
Biomed Life Sci 966:34–47 32. van der Werf MJ, Overkamp KM, Muilwijk B
20. Martano G, Delmotte N, Kiefer P et al (2015) et al (2008) Comprehensive analysis of the
Fast sampling method for mammalian cell met- metabolome of Pseudomonas putida S12
abolic analyses using liquid chromatography- grown on different carbon sources. Mol Bio-
mass spectrometry. Nat Protoc 10:1–11 Syst 4:315–327
21. Naz S, Vallejo M, Garcı́a A et al (2014) Method 33. Stuani L, Lechaplais C, Salminen AV et al
validation strategies involved in non-targeted (2014) Novel metabolic features in Acinetobac-
metabolomics. J Chromatogr A 1353:99–105 ter baylyi ADP1 revealed by a multiomics
22. Dunn WB, Broadhurst D, Begley P et al (2011) approach. Metabolomics 10:1223–1238
Procedures for large-scale metabolic profiling
Chapter 19
Abstract
Protein digestion coupled to liquid chromatography and tandem mass spectrometry (LC-MS/MS) detec-
tion enables multiplexed quantification of proteins in complex biological matrices. However, the reproduc-
ibility of enzymatic digestion of proteins to produce proteotypic target peptides is a major limiting factor of
assay precision. Online digestion using immobilized trypsin addresses this problem through precise control
of digestion conditions and time. Because online digestion is typically for a short time, the potential for
peptide degradation, a major source of measurement bias, is significantly reduced. Online proteolysis
requires minimal sample preparation and is easily coupled to LC-MS/MS systems, further reducing
potential method variability. We describe herein a method optimized for the multiplexed quantification
of several apolipoproteins in human serum using on-column digestion. We highlight key features of the
method that enhance assay accuracy and precision. These include the use of value-assigned serum as
calibrators and stable isotope-labeled (SIL) peptide analogs as internal standards. We also comment on
practical aspects of column switching valve design, instrument maintenance, tandem mass spectrometry
data acquisition, and data processing.
Key words Online digestion, Immobilized enzyme reactor, IMER, Apolipoproteins, Quantitative,
IDMS, IMER-LC-MS/MS
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_19, © Springer Science+Business Media, LLC, part of Springer Nature 2019
295
296 Christopher A. Toth et al.
2 Materials
2.6 Human Serum Frozen human serum from individual donors was purchased from a
specimen depository (Bioreclamation IVT, Westbury, NY).
3 Methods
3.2 Preparation of 1. Thaw one aliquot of each SIL stock solution on the bench,
Working SIL vortex-mix, and spin down.
Peptide Mix 2. To a 15 mL Falcon™ conical centrifuge tube, add 20–450 μL of
each individual SIL peptide stock solution (see Notes 11 and 22).
3. Add 100 μL of a 0.1% Zwittergent 3–12/0.1% formic acid in
water solution, cap, and vortex-mix (see Note 12).
4. Bring to 10 mL volume with 0.1% formic acid in water.
5. Store the SIL peptide mix for maximum 4 weeks at 4 C (see
Note 13).
6. Before analysis dispense a 1.5 mL aliquot into a standard
polypropylene autosampler vial and place into the reagent
tray of the autosampler.
Nuts and Bolts of Protein Quantification by Online Trypsin Digestion. . . 299
3.3 Preparation of 1. Allow frozen serum from individual donors to thaw on ice.
Calibrator Pool and 2. Merge equal volumes from each donor (see Note 14).
Storage
3. Vacuum filter the pool using a Nalgene™ Rapid-Flow™ filter
(0.45 μm pore size) or equivalent.
4. Continuously stir the pool at 250 rpm in an ice bath on a
magnetic stir plate while distributing a series of 100, 50, 20,
10, and 5 μL aliquots using a positive displacement repeater
pipette into 2 mL storage vials.
5. Verify the volume of each aliquot by mass using an analytical
balance and record.
6. Store the aliquots at 80 C until use.
7. For the value assignment of the calibrator pool by standard
addition methodology, see Note 15.
3.4 Preparation of 1. Pull a set of stored Calibrator Pool Aliquots (100, 50, 20,
Standard Series 10 and 5 μL) from the freezer and allow to thaw on ice.
2. Bring to volume with diluting buffer (Subheading 2.1, step 3)
following an appropriate dilution level (see Note 16).
3. Gently invert each tube 10 times to mix and spin down.
4. Use immediately or store at 4 C for no more than 4 days (see
Note 17).
3.7 Data Processing 1. Pull the raw mass spectrometry data into MultiQuant (SCIEX)
for the calculation of [native cleavage peptide peak area]/[SIL
peptide peak area] ratios.
2. Examine the automated peak integrations.
3. Before reporting concentrations review the data based on
acceptable range of SIL peak intensities, product ion ratios,
and deviations between technical repeats.
4 Notes
Fig. 1 The effect of Zwittergent 3-12 concentration on the digestion efficacy is demonstrated with bovine
serum albumin as a model protein. As the final detergent concentration in the sample well increases, the
peptide peak area/protein concentration increases for all tryptic peptides. This effect levels off just beyond the
critical micelle concentration (CMC) of Zwittergent 3-12 (4 mM, 0.15% w/v)
1
5 ApoA-2 EQLTPLIK +2y5 ApoA-4 LEPYADQLR +2y6 ApoC-1 TPDVSSALDK +2y8
2
0.8
4
1.5
0.6
3
1
0.4
2
0.2 0.5
1 y = 3.052x + 1.830 R² = 0.998 y = 0.687x + 0.166 R² = 0.999 y = 1.280x + 0.562 R² = 0.995
y = 3.954x R² = 0.997 y = 0.634x R² = 0.997 y = 1.248x R² = 0.997
0 0 0
0 0.5 1 0 0.5 1 0 0.5 1
4
ApoC-2 TYLPAVDEK +2y7 ApoC-3 GWVTDGFSSLK +2y7 1.25 ApoE SELEEQLTPVAEETR +2y7
0.4
3 1
0.3
0.75
2
0.2
0.5
1
0.1 0.25
y = 0.315x + 0.089 R² = 0.998 y = 3.113x + 0.477 R² = 0.999 y = 3.113x + 0.477 R² = 0.998
y = 3.132x R² = 0.998 y = 3.132x R² = 0.994
y = 0.302x R² = 0.999
0 0 0
0 0.5 1 0 0.5 1 0 0.5 1
Fig. 2 Area ratio (Y-axis) versus concentration ratio (X-axis) for standard addition of purified recombinant
proteins to a spiked matrix (square) and solvent blank (circle) for slope comparison. Error bars indicate
standard deviation (N ¼ 3). The comparable slope indicates that spiked recombinant proteins digest in an
analogous way to endogenous protein in diluted matrix, a critical assumption for this method
304 Christopher A. Toth et al.
ApoA1.THLAPYSDELR+2y8 ApoB.TGISPLALIK+2y6
R2 = 0.9996 R2 = 0.9897
Fig. 3 Demonstration of the linear dynamic range for two selected peptide transitions, apoA1: THLAPYSDELR
2y8 and apoB-100: TGISPLALIK 2y6 in diluted serum calibrator. The nine point calibration curve represents
1:15 and 1:1500 fold dilutions at the ULOQ and LLOQ, respectively
Table 1
Labsolutions pretreatment program
Line # Value
1 a1¼1001
2 vial a1
3 n.strk ns
4 aspir 5,5
5 air.a 1,5
6 rinse 200,50
7 vial sn
8 n.strk ns
9 aspir iv,ss
10 air.a 1,5
11 rinse 200,50
12 inj.p
13 s.inj
14 end
Fig. 4 Simplified diagram of the IMER-LC-MS/MS system showing sample flow path and valve positions
306 Christopher A. Toth et al.
Table 2
LC instrument parameters
21. The modified valve design shown in Fig. 6a, b allows simulta-
neous digestion and LC separation. This is accomplished by
employing a 10-port switching valve and two identical trapping
columns. While the cleavage products and SIL peptides are
collected on one column, the cleavage products and SIL pep-
tide from the previous sample eluted to the analytical column
and to the LC-MS interface. This alternating setup effectively
reduces total analysis time to the length of the LC gradient.
Additionally, the valve design reverses the direction of the flow
on the trapping column during desalting and elution, refocus-
ing analytes onto the analytical column and reducing peak
broadening.
22. See Fig. 7 and Table 3.
Fig. 6 Dual trapping valve design and valve positions corresponding to LC methods A and B
1
3.0e6
3 15
2 4
2.0e6
Intensity, cps
11 13 6
8 10 9
1.0e6
18
14 12
17
16 7
0.0
1.0 2.0 3.0 4.0 5.0 6.0 7.0 8.0 9.0
Time, min
Fig. 7 Representative chromatogram: 50 μL of 1:100 diluted serum calibrator (gray) and 5 μL SIL mix (black)
injected into system. Corresponding peak IDs are listed in Table 3
Table 3
MRM instrument parameters for native and labeled peptides
Native Label
Fragmention Fragmention
ID Protein Peptide sequence Precursor ion m/z 1 2 DP CE Peptide sequence Precursor ion m/z 1 2
*
1 apoA-I AKPALEDLR 506.8 716.4 813.4 68.1 27.1 AKPALED(L+7)R 510.3 723.4 820.5
*
2 ATEHLSTLSEK 608.3 664.4 777.4 75.5 30.8 ATEHLST(L+7)SEK 611.8 671.4 784.5
*
3 THLAPYSDELR 651.3 950.5 1063.5 78.6 32.3 THLAPYSDE(L+7)R 654.8 957.5 1070.6
*
4 apoA-II EQLTPLIK 471.3 470.3 571.4 65.5 25.8 EQLTPL(I+7)K 474.8 477.4 578.4
*
5 apoA-IV LEPYADQLR 552.8 765.4 862.4 71.4 28.8 LEPYADQ(L+7)R 556.3 772.4 869.5
*
6 LTPYADEFK 542.3 772.4 869.4 70.6 28.4 LTPYADEF(K+8) 546.3 780.4 877.4
*
7 apoB ATGVLYDYVNK 621.8 801.4 914.5 76.4 31.2 ATGVLYDY(V+6)NK 624.8 807.4 920.5
*
8 AAIQALR 371.7 487.3 600.4 58.2 22.2 AAIQA(L+7)R 375.2 494.3 607.4
*
9 TGISPLALIK 506.8 654.5 741.5 68.1 27.1 TGISPLAL(I+7)K 510.3 661.5 748.5
*
10 apoC-I EWFSETFQK 601.3 739.4 886.4 75 30.5 EWFSETFQ(K+8) 605.3 747.4 894.4
*
11 TPDVSSALDK 516.8 620.3 834.4 68.8 27.5 TPDVSSA(L+7)DK 520.3 627.3 841.4
*
12 apoC-II ESLSSYWESAK 643.8 870.4 957.4 78 32 ESLSSYWESA(K+8) 647.8 878.4 965.4
*
13 TYLPAVDEK 518.3 658.3 771.4 68.9 27.5 TYLPA(V+6)DEK 521.3 664.4 777.4
*
14 apoC-III DALSSVQESQVAQQAR 858.9 887.5 1016.5 93.7 39.8 DALSSVQESQVAQ(Q+7)AR 862.4 894.5 1023.5
*
15 GWVTDGFSSLK 598.8 753.4 854.4 74.8 30.4 GWVTDGFSS(L+7)K 602.3 760.4 861.4
*
16 apoE AATVGSLAGQPLQER 749.4 770.4 827.4 85.7 35.8 AATVGSLAGQPL(Q+7)ER 752.9 834.4 905.5
*
17 LGPLVEQGR 484.8 588.3 489.2 66.5 26.3 LGPLVE(Q+7)GR 488.3 496.3 595.3
*
Nuts and Bolts of Protein Quantification by Online Trypsin Digestion. . .
18 LQAEAFQAR 517.3 721.4 792.4 68.8 27.5 LQAEAF(Q+7)AR 520.8 728.4 799.4
Product ions marked with (*) were chosen for quantitation
309
310 Christopher A. Toth et al.
1.25e5
2.8e6
1.00e5
2.0e6
Intensity, cps
1st Blank
1.0e6
2nd Blank
THLAPYSDELR
TGISPLALIK
1.00e4
0.0 0.00
0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 5.5 6.0 6.5 7.0 7.5 8.0 8.5
Time, min
Fig. 8 Example of carry-over contribution to the native signal in one and two blank injections (50 μL of digest
buffer) immediately following a highly concentrated sample (50 μL of 1:15 diluted human serum on-column).
The intensities are plotted for two peptide transitions with contrasting native protein concentrations,
THLAPYSDELR (apoA-I; left axis) and TGISPLALIK (apoB; right axis)
Disclaimer
References
1. Picotti P, Aebersold R (2012) Selected reaction 1461:84–91. https://doi.org/10.1016/j.
monitoring-based proteomics: workflows, chroma.2016.07.058
potential, pitfalls and future directions. Nat 7. Toth CA, Kuklenyik Z, Jones JI, Parks BA,
Methods 9(6):555–566. https://doi.org/10. Gardner MS, Schieltz DM, Rees JC, Andrews
1038/nmeth.2015 ML, McWilliams LG, Pirkle JL, Barr JR (2017)
2. Shuford CM, Walters JJ, Holland PM, On-column trypsin digestion coupled with
Sreenivasan U, Askari N, Ray K, Grant RP LC-MS/MS for quantification of apolipopro-
(2017) Absolute protein quantification by mass teins. J Proteome 150:258–267. https://doi.
spectrometry: not as simple as advertised. Anal org/10.1016/j.jprot.2016.09.011
Chem 89(14):7406–7415. https://doi.org/10. 8. Hoofnagle AN, Whiteaker JR, Carr SA, Kuhn E,
1021/acs.analchem.7b00858 Liu T, Massoni SA, Thomas SN, Reid
3. Shuford CM, Sederoff RR, Chiang VL, Muddi- Townsend R, Zimmerman LJ, Boja E, Chen J,
man DC (2012) Peptide production and decay Crimmins DL, Davies SR, Gao Y, Hiltke TR,
rates affect the quantitative accuracy of protein Ketchum KA, Kinsinger CR, Mesri M, Meyer
cleavage isotope dilution mass spectrometry MR, Qian WJ, Schoenherr RM, Scott MG,
(PC-IDMS). Mol Cell Proteomics 11 Shi T, Whiteley GR, Wrobel JA, Wu C, Acker-
(9):814–823. https://doi.org/10.1074/mcp. mann BL, Aebersold R, Barnidge DR, Bunk
O112.017145 DM, Clarke N, Fishman JB, Grant RP,
4. Regnier FE, Kim JH (2014) Accelerating trypsin Kusebauch U, Kushnir MM, Lowenthal MS,
digestion: the immobilized enzyme reactor. Moritz RL, Neubert H, Patterson SD, Rock-
Bioanalysis 6(19):2685–2698. https://doi. wood AL, Rogers J, Singh RJ, Van Eyk JE,
org/10.4155/bio.14.216 Wong SH, Zhang S, Chan DW, Chen X, Ellis
5. Kuklenyik Z, Jones JI, Toth CA, Gardner MS, MJ, Liebler DC, Rodland KD, Rodriguez H,
Pirkle JL, Barr JR (2017) Optimization of the Smith RD, Zhang Z, Zhang H, Paulovich AG
linear quantification range of an online trypsin (2016) Recommendations for the generation,
digestion coupled liquid chromatography–tan- quantification, storage, and handling of peptides
dem mass spectrometry (LC–MS/MS) plat- used for mass spectrometry-based assays. Clin
form. Instrum Sci Technol 46:1–13. https:// Chem 62(1):48–69. https://doi.org/10.
doi.org/10.1080/10739149.2017.1311912 1373/clinchem.2015.250563
6. Bonichon M, Combès A, Desoubries C, 9. Grant RP, Hoofnagle AN (2014) From lost in
Bossée A, Pichon V (2016) Development of translation to paradise found: enabling protein
immobilized-pepsin microreactors coupled to biomarker method transfer by mass spectrome-
nano liquid chromatography and tandem mass try. Clin Chem 60(7):941–944. https://doi.
spectrometry for the quantitative analysis of org/10.1373/clinchem.2014.224840
human butyrylcholinesterase. J Chromatogr A
Chapter 20
Abstract
Proteases drive the life cycle of all proteins, ensuring the transportation and activation of newly minted,
would-be proteins into their functional form while recycling spent or unneeded proteins. Far from their
image as engines of protein digestion, proteases play fundamental roles in basic physiology and regulation at
multiple levels of systems biology. Proteases are intimately associated with disease and modulation of
proteolytic activity is the presumed target for successful therapeutics. “Proteases: Pivot Points in Functional
Proteomics” examines the crucial roles of proteolysis across a wide range of physiological processes and
diseases. The existing and potential impacts of proteolysis-related activity on drug and biomarker develop-
ment are presented in detail. All told the decisive roles of proteases in four major categories comprising
23 separate subcategories are addressed. Within this construct, 15 sets of subject-specific, tabulated data are
presented that include identification of proteases, protease inhibitors, substrates, and their actions. Said data
are derived from and confirmed by over 300 references. Cross comparison of datasets indicates that
proteases, their inhibitors/promoters and substrates intersect over a range of physiological processes and
diseases, both chronic and pathogenic. Indeed, “Proteases: Pivot Points . . .” closes by dramatizing this very
point through association of (pro)Thrombin and Fibrin(ogen) with: hemostasis, innate immunity, cardio-
vascular and metabolic disease, cancer, neurodegeneration, and bacterial self-defense.
Key words Protease, Peptidase, Proteolysis, Protease inhibitor, Protease promoter, Digestion,
Hemostasis, Complement system, Immune regulation, Signaling, Cell migration, Cell proliferation,
Programmed cell death, Protein secretion, DNA replication, DNA repair, DNA processing, Intra-
nuclear proteolysis, Transmembrane proteolysis, Intramembrane proteolysis, Cytosolic proteolysis,
Epigenetics, Inflammation, Cardiovascular disease, Metabolic disease, Stroke, Cancer, Neurodegener-
ative disease, Autoimmune disease, Infectious organisms, Drug target, Drug development, Biomarker
development, Precision medicine
1 Introduction
1.1 Proteases: More One or more proteolytic events initiate the active life for many
Than Just Protein proteins and proteolytic events terminate the relatively short life
Digestion of all proteins. Roughly a third of proteins, as translated, include
peptides that influence proteins’ life span (initiator methionine),
govern their transport (signal peptide), direct them to the appro-
priate organelle (transit peptide), and produce their active form
(propeptide). These processes are merely those proteolytic events
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_20, © Springer Science+Business Media, LLC, part of Springer Nature 2019
313
314 Ingrid M. Verhamme et al.
Translated Protein
that begin the active life of proteins. Over the course of lifetimes
that may be as brief as 11 min or as long as 4 months, additional
proteolytic events remake individual proteins—one example being
the much studied A4-Human amyloid precursor transmembrane
protein whose 13 proteolytic products include a family of
pathology-related, β amyloid peptides. Having served their tempo-
rary purposes, all proteins are once again substrates for proteases
with digestion proceeding to amino acids recycled as raw materials
for ongoing expression of new proteins. In the light of these varied
and continuous processes, proteolytic activity literally reshapes the
proteome. Of course, this brief discourse is but the tip of the
iceberg. Proteolytic enzymes—and their promoters and inhibi-
tors—are essential actors in nearly all biological processes. This
chapter addresses many of those processes especially in relation to
disease and development of new therapeutics. Truly, proteases give
definition to the Functional Proteome—one protein at a time
(Fig. 1).
In the 1830s, parallel studies in Europe and the USA demon-
strated that digestion of proteins in food required a compound in
the gastric mucosa, in addition to gastric juice hydrochloric acid
[1]. The discovery of pepsin triggered the formulation of popular
therapeutic preparations for dyspepsia, and intensified pepsin puri-
fication efforts. However, it would take nearly a century before
crystallization of pure pepsin was achieved. Northrop, Kunitz,
and Herriott published the earliest characterization of classical
digestive proteases such as pepsin, chymotrypsin, trypsin, pancreatic
carboxypeptidases, and their zymogens as pure compounds
Proteases: Pivot Points in Functional Proteomics 315
[2]. The protease field has come a long way since the discovery of
pepsin. Rapidly developing methods for protein purification,
sequencing, structure-function analysis, X-ray crystallography, and
synthetic substrate development have since then accelerated the
identification of countless mammalian, plant, fungal, and bacterial
proteolytic enzymes [3], and the number of identified sequences is
growing exponentially. As of September 2017, the MEROPS data-
base of proteolytic enzymes (https://www.ebi.ac.uk/merops/)
listed more than one million sequences [4].
The complete ensemble of human proteases, known as the
protease degradome, currently consists of 588 proteases, organized
into five classes: aspartic, cysteine, metallo-, serine, and threonine
proteases [5]. They represent >2% of the human genome, and a
database is maintained by the lab of López-Otı́n (http://
degradome.uniovi.es) [6]. Protease systems have gained consider-
able recognition since the realization that they are important reg-
ulators of countless biological mechanisms, and not simply part of a
machinery for nonspecific protein digestion. Blood coagulation,
fibrinolysis, complement activation, peptide hormone processing,
protein secretion and degradation, DNA replication and repair, cell
signaling and proliferation, and programmed cell death are just a
few of these processes in which physiological proteases target spe-
cific substrates. The peptidase field is rapidly evolving, and extensive
volumes have already been dedicated to cataloguing and docu-
menting its recently discovered and previously known members
[7]. Here we focus on novel aspects of the intimate connection
between disease and proteolysis, and the potential of mechanism-
based drug targeting.
2.1 Hemostasis If you are familiar with only one proteolytically regulated process in
the human body, it is likely hemostasis. Featured prominently in
documentaries about European royalty and in nightly medication
commercials, the formation and degradation of blood clots has
been intensely studied. It is a model system for illustrating the
complexity of proteolysis: multiple pathways acting both in concert
318 Ingrid M. Verhamme et al.
Table 1
Hemostasis clot formation: activities and Uniprot ID codes (where applicable) of proteases, protease
inhibitors, and cofactors associated with hemostasis, specific to clot formation
Table 2
Hemostasis clot degradation: activities and Uniprot ID codes (where applicable) of proteases,
protease inhibitors, and cofactors associated with hemostasis, specific to clot degradation
2.2 The Complement The plasma complement system regulates the innate immune
System and Immune defense by opsonization and elimination of pathogens, cell debris,
Regulation and host cells that have undergone alterations [23–25]. Activation
of the complement system occurs via three pathways: in the classical
pathway, the recognition protein C1q binds to antigen-antibody
complexes, the C1 complex is activated, and a series of serine
protease activation reactions leads to formation of C3- and ulti-
mately C5-convertase; in the lectin pathway, mannose-binding
lectin binds to mannose on the surface of pathogens as a trigger
Proteases: Pivot Points in Functional Proteomics 321
Table 3
Complement system and immune regulation: activities and Uniprot ID codes (where applicable) of
proteases, protease inhibitors, and cofactors associated with the complement system and immune
regulation
2.3 Proteolytic The lysosome and the ubiquitin-proteasome are the two major
Processing intracellular proteolytic systems keeping the protein pool in bal-
ance. Originally considered strictly degradative, these systems have
revealed regulatory functions beyond catabolism, and their molec-
ular defects are associated with various disease states. Lysosomes
contain cathepsins B, D, and L in addition to lipases, nucleases,
glycosidases, phospholipases, phosphatases, and sulfatases that are
active in an acidic milieu [32]. Lysosomes regulate autophagy
during nutrient starvation, and participate in development and
differentiation, induction of cathepsin-dependent cell death, and
degradation of apoptotic cells. Cancer cell lysosomes have a higher
membrane permeability (“leaky”) and express more cathepsin than
those of normal cells, and this property may be exploited in cancer
treatment. Agents such as tetrahydrocannabinol and chloroquine
may disrupt the lysosome and trigger killing of the cancer cells
(Table 4).
The ubiquitin-proteasome, an intracellular high molecular
weight protease complex predominantly located in the cytosol,
selectively degrades proteins tagged with ubiquitin at lysine resi-
dues [33]. Its “central pore” contains several inward facing protease
active sites, with caspase-, trypsin-, and chymotrypsin-like specific-
ity. This multi-protein construct is capped by one or two activator
complexes that conformationally regulate access of protein sub-
strates to the pore. Whereas the physiological function of the
proteasome was originally thought to be restricted to intracellular
protein catabolism, new functions have been discovered with
respect to regulation of the cell cycle progression, gene expression,
and responses to cellular stress [34]. Protein ubiquitination is
reversible, and more than 100 potentially regulatory deubiquitinase
(DUB) genes have been identified, mainly cysteine and metallopro-
teinases (MMPs). DUBs rescue proteins from degradation and
reverse ubiquitination-induced signaling. The immunoprotea-
some, containing specific subunits with increased chymotrypsin-
and trypsin-like activities, and decreased caspase-like activity, parti-
cipates in production of peptide epitopes for cytotoxic T lympho-
cytes. In the thymoproteasome, chymotryptic activity is attenuated
but the caspase- and trypsin-like activities are conserved. Its peptide
products are MHC class I ligands with moderate avidity, which
supports positive selection of CD8+ T cells [35]. Foreign peptides,
generated during the breakdown of virus and cancer cell proteins,
bind MHC class I molecules on the cell surface, and the cells are
recognized by cytotoxic T cells as potentially dangerous and are
destroyed. The proteasome discards misfolded proteins, and
324 Ingrid M. Verhamme et al.
Table 4
Proteolytic processing: activities and Uniprot ID codes (where applicable) of proteases, protease
inhibitors, and cofactors associated with proteolytic processing
Table 5
Tissue remodeling, signaling, cell migration, and proliferation: activities and Uniprot ID codes (where
applicable) of proteases, protease inhibitors, and cofactors associated with tissue remodeling, signaling,
cell migration, and proliferation
2.5 Programmed For obvious reasons, the process of apoptosis must be both highly
Cell Death regulated and a model of organized efficiency once initiated. It is
governed by a cascade of caspases which, though they sometimes
have other functions, thoroughly dismantle the innards of a cell so
that it may be phagocytosed by immune cells without releasing
cytosolic components into the extracellular space. Caspases are
cysteine aspartate proteases involved in cell death, cellular remodel-
ing, stem cell fate determination, spermatogenesis, and red blood
cell differentiation. Their sets of substrates with regard to apoptosis
are well defined, and cooperative cleavage of these substrate sets
triggers apoptosis. With regard to apoptosis, their functions fall
into one of two categories: Initiator (caspases 2, 8, 9, and 10) or
Executioner (caspases 3, 6, and 7). Caspases 1, 4, 5, and 12L are
considered inflammatory [47]. Activation of the Initiator caspases
Proteases: Pivot Points in Functional Proteomics 329
2.6 Protein Secretion Many current cardiovascular biomarkers are secreted proteins, gen-
erated by cleavage of their pro-proteins at the endoplasmic reticu-
lum (ER). Upon release of the mature protein, the signal peptide is
proteolytically separated from the ER by signal peptidase, an intra-
membrane aspartic protease. The long-held belief that signal pep-
tides are invariably recycled or degraded by ubiquitin-proteasome
related factors has been challenged during recent years, and several
were shown to remain intact after cleavage in the ER [64]. These
signal peptides have biological functions of their own, and play
roles in regulation of immunity, trafficking, and other processes.
In type I diabetes, a signal peptide fragment from pre-proinsulin,
presented at the surface of the pancreatic β-cells, acts as antigen and
flags the cell for destruction by cytotoxic T cells. Identifying agents
to control β-cell destruction may be a new therapeutic strategy.
Similarly, a signal peptide fragment from pre-procalcitonin, highly
abundant in several lung cancers and medullary thyroid cancers, is
an epitope for T-killer cells. This knowledge may aid in the devel-
opment of treatments of these cancers. Rapidly increasing plasma
levels of N-terminal adducted signal peptide fragments from A-, B-,
and C-type natriuretic peptides are characteristic of ST-elevated
myocardial infarction. The nature of the N-terminal adducts may
be useful in assay design and disease assessment, and development
of fast biomarker assays for these signal peptide fragments may
ultimately be beneficial in clinical decision-making (Table 7).
2.7 DNA Replication, DNA damage hinders replication, and may lead to strand breaks,
Repair, and Processing genomic instability, aging, and cancer [65]. DNA-topoisomerase
1 crosslinks (DPC) are bulky lesions that trap otherwise transient
covalent DNA-protein intermediates, and inhibit movement of
polymerases and helicase, causing stalling of the replication fork.
In yeast models, the protease Wss1 was identified as effector of
DPC repair. BLAST searches revealed a conserved family of DPC
proteases, with Spartan being the human member of this class
[66]. Spartan was recently characterized as a DNA replication-
Proteases: Pivot Points in Functional Proteomics 331
Table 6
Programmed cell death: activities and Uniprot ID codes (where applicable) of proteases, protease
inhibitors, and cofactors associated with programmed cell death
Table 7
Protein secretion: activities and Uniprot ID codes (where applicable) of proteases, protease inhibitors,
and cofactors associated with protein secretion
2.8 Intranuclear, In addition to Spartan and the proteasome involved in DNA pro-
Cytosolic, cessing, other proteases are known to have intranuclear functions.
Transmembrane, and Interleukin-1β-converting enzyme (ICE), also known as caspase-1,
Intramembrane plays a role in the inflammatory immune response. The N-terminal
Proteolysis prodomain of its precursor, procaspase-1, possesses a nuclear local-
ization signal. Tumor necrosis factor induces translocation of
procaspase-1 from the cytosol to the nucleus where it is activated
by proteolytic removal of the intact prodomain [73]. Cell transfec-
tion studies showed that the prodomain alone is capable of trigger-
ing apoptosis, which suggests separate nuclear roles for the
prodomain and active caspase-1. Caspase-3 also effects nuclear
changes in apoptotic cells. Its inactive precursor is cleaved in the
cytoplasm by initiator caspases in response to death signals, and also
by cytosolic MMP-2 and -9. The active caspase-3 dimerizes and
translocates to the nucleus by an active transport system. Caspase-7
is only found in the cytoplasm, suggesting that translocation is
caspase-3 specific and not the result of simple diffusion after the
nuclear-cytoplasmic barrier is disrupted [74]. The extracellular
properties of MMPs as tissue remodeling proteases are well docu-
mented but less is known about their intracellular functions. Vari-
ous MMPs have been detected in the intranuclear space, and they
are mostly associated with pathological processes [75, 76].
Oxygen-glucose deprivation in ischemic stroke induces an intrinsic,
caspase-independent apoptotic pathway in neurons, characterized
by elevated intranuclear MMP-2 and -9 activity that targets nuclear
DNA repair proteins. Similarly, MMP-2 in the nuclei of stressed
cardiac myocytes can induce apoptosis. Intranuclear MMP-3 in
osteoarthritis and certain cancers upregulates connective tissue
growth factor, a mediator of cell migration, proliferation, and
pathological fibrosis. During viral infections, macrophage-secreted
MMP-12 translocates to the nucleus of infected cells, and enhances
transcription of IκBα, a protein that promotes antiviral interferon-α
(IFN-α) secretion. Extracellular MMP-12 degrades excess secreted
IFN-α, thus limiting its systemic toxicity. It is becoming increas-
ingly clear that the same protease may exert significantly divergent
functions, depending on its microenvironment, substrates, and
effectors. Understanding these different mechanisms of interaction
on a molecular level is ultimately the key to successful design of
therapeutics (Table 8).
Promotion of angiogenesis is regulated by binding of single
chain urokinase-type plasminogen activator (scuPA) to its receptor
on the endothelial cell surface, and subsequent transport of the
334 Ingrid M. Verhamme et al.
Table 8
Intranuclear, transmembrane, intramembrane, and cytosolic proteolysis: activities and Uniprot ID
codes (where applicable) of proteases, protease inhibitors, and cofactors associated with intranuclear,
transmembrane, intramembrane, and cytosolic proteolysis
3.1 Epigenetics and Many inheritable diseases are directly related to DNA modification;
Disease however, epigenetic processes are equally prominent in the disease
state, and they are essential contributors to normal physiological
development. Environmental factors, diet, aging, and diseases such
as cancer may contribute to positive or negative changes in gene
expression that are passed onto daughter cells: DNA (hydroxyl)
methylation, covalent histone modification and chromatin
Proteases: Pivot Points in Functional Proteomics 337
Table 9
Inflammation as an over-arching symptom of disease: activities and Uniprot ID codes (where
applicable) of proteases, protease inhibitors, and cofactors associated with inflammation, specifically
as related to symptoms of disease
Table 9
(continued)
Table 10
Cardiovascular and metabolic diseases, and stroke: activities and Uniprot ID codes (where
applicable) of proteases, protease inhibitors, and cofactors associated with cardiovascular disease,
metabolic diseases, and stroke
Table 10
(continued)
Table 10
(continued)
3.4 Cancer The long-held concept that somatic mutations are the causal event
in the majority of cancers has recently come under scrutiny
[128, 129]. Priming the cellular microenvironment for develop-
ment of cancer is characterized by a sequence of events that precede
the transformation of a normal cell into a cancer cell, and somatic
mutations are actually later events in the development of many
cancers. Chronic inflammation and fibrosis have been identified as
two of these events. Hemostatic proteases have recently been
recognized to contribute to inflammatory processes in cancer. Dis-
ruption of the endothelial barrier during tissue damage allows
hemostatic zymogens to be activated. Not only do these proteases
contribute to extravascular coagulation and fibrinolysis, they also
trigger signaling through cell surface activation of PAR receptors,
binding to uPAR and LRP-1, and activation of MMPs
[130]. Inflammation also triggers the release of TGF-β that
potently induces MMP-2 and MMP-9 expression. In turn,
MMP-2, -9, and -14 proteolytically activate latent TGF-β in the
ECM. Transmembrane MMP-14 and several members of the
ADAM family are localized on invadopodia of migrating cells.
The involvement of MMPs in extracellular matrix remodeling facil-
itates tumor invasion, and MMPs also figure prominently in cancer-
related signaling. Whereas many MMPs are recognized as
pro-tumorigenic, some may negatively affect cancer progression,
depending on the microenvironment of the cell [131]. The MMP
and ADAM inhibitor Marimastat showed no broad therapeutic
anticancer potential due to lack of specificity; however, it inhibited
ADAM-17, highly expressed in renal cell carcinoma [132]. This
inhibition downregulated Notch pathway-mediated cell prolifera-
tion and invasion more effectively than γ-secretase inhibition.
Hence, Marimastat may have therapeutic potential in renal cell
cancer. Tissue inhibitors of metalloproteinases (TIMPs) are differ-
entially expressed in cancer: high TIMP1 expression is associated
with fibrotic processes and poor outcome, and TIMP3 silencing
indicates advanced disease [133]. TIMPs figure prominently in
other pathologies such as cardiovascular disease and sepsis, and
fibrosis as measured by TIMP1 levels was recently shown to predict
all-cause mortality in the AGES-Reykjavik Study [134] (Table 11).
Epigenetic processes are increasingly recognized as essential in
carcinogenesis. During epithelial-mesenchymal transition (EMT)
in cancer initiation, progression, and metastasis, epigenetic
Proteases: Pivot Points in Functional Proteomics 349
Table 11
Cancer: activities and Uniprot ID codes (where applicable) of proteases, protease inhibitors, and
cofactors associated with cancer
Table 11
(continued)
Table 13
Autoimmune diseases: activities and Uniprot ID codes (where applicable) of proteases, protease
inhibitors, and cofactors associated with autoimmune diseases
3.7 Proteases, Infectious organisms employ their own arsenal of proteases for
Inhibitors, and propagation and virulence, such as HIV protease [175], Trypano-
Cofactors in Infectious soma cruzi cruzipain [176], Porphyromonas gingivalis gingipains
Organisms [177], and Bacillus anthracis lethal factor [178]. Several bacterial,
viral, protozoan, and fungal proteases trigger inflammation by
activating the intrinsic coagulation pathway [102], or act as pro-
coagulants by non-canonical, direct activation of prothrombin
[179]. Large panels of small molecule inhibitors of the proteasome
360 Ingrid M. Verhamme et al.
5.1 Active Site Active site targeting is a main component in many established
Targeting, Exosite, and approaches of drug development for controlling protease activity.
Effector-Binding Sites However, small molecule inhibitors that are limited to interference
with the conserved catalytic machinery of an entire class of pro-
teases may have severe limitations due to their broad-spectrum
activity, resulting in off-target protease inhibition. This was illu-
strated by the failure of many small molecule and zinc-targeting
MMP inhibitors in clinical trials. Early irreversible inhibitors used
active site targeting of nucleophilic proteases with electrophilic
alkylating agents such as diazo- or halo-ketone warheads; however,
370 Ingrid M. Verhamme et al.
5.3 Drugging the Designing substrates, inhibitors, and activity-based probes with
Undruggable: A specificity toward a single protease is a major challenge when the
Familiar Theme, with protease shares a similar catalytic mechanism and substrate specific-
Some Fancy Targeting ity with other proteases in the same family, but is functionally
completely different [237]. The main approaches utilized to date
for optimizing complementarity with the protease specificity sites
include positional scanning synthetic combinatorial libraries with
coumarin-derived reporters (PS-SCL) [238], phage display, hybrid
combinatorial substrate libraries using unnatural amino acids that
allow more thorough scanning of the active site (HyCoSuL),
counter selection substrate libraries (CoSeSuL), internally
quenched fluorescent substrate or fluorescence resonance energy
transfer libraries (IQF or FRET), proteomics and exopeptidase
fingerprinting. HyCoSuL screening of the P3 and P4 positions,
respectively, with methionine sulfone and 2-amino-6-benzyloxy-
hexanoic acid allowed distinguishing between neutrophil elastase
and proteinase-3 for the first time, and tracking neutrophil elastase
activity in neutrophil traps [239]. Whereas PS-SCL, HyCoSuL, and
CoSeSuL allow determination of protease preferences only in prime
active site pockets, IQF can be used to refine complementarity to
both prime and non-prime pockets. The advantage of phage display
is its ability to generate large and diverse substrate arrays, up to 1010
peptides, and enrich for specificity after each cycle, which is not
feasible with chemical synthesis. The label-free nature of this
method requires individual kinetic analysis and reporter group
labeling of positive hits. Multiplex substrate profiling with liquid
chromatography–tandem mass spectrometry sequencing proved
successful for distinguishing neutral serine protease activity in
human neutrophil extracellular traps toward a large and diverse
tetradecapeptide panel, and ranking granzyme B substrate effi-
ciency, using label-free quantitation of precursor-ion abundance
[240]. The above techniques have also allowed differentiation
between various diverse members of the granzyme, kallikrein, cas-
pase, metalloproteinase, exopeptidase, deubiquitinating and desu-
moylating protease families. A novel approach of profiling protease
specificity is currently being developed, combining yeast endoplas-
mic reticulum (ER) sequestration with next-generation sequencing
Proteases: Pivot Points in Functional Proteomics 375
5.5 A Two-Protein This chapter presents, in text and tabulated formats, the diversity of
Wrap-Up proteases and protease-related activity. Perhaps the most compel-
ling overall observation is the degree of overlap among physiologi-
cal processes and disease states at the level of individual proteases.
This is most readily seen in the tabulated summary of proteases,
inhibitors and activity. A thumbnail analysis shows that half of all
listed proteases are active in multiple physiological processes
and/or associated with multiple disease states. Chief among the
multi-process activities are the cathepsins, followed by the caspases
and MMPs. The chapter closes with an examination of the multiple
associations with physiological processes and disease states of two
related proteins: the protease thrombin and the proteolysis product
fibrin (gene expression products, prothrombin and fibrinogen).
Textbooks cite the cleavage of fibrinogen by thrombin to form
fibrin as a central event in formation of blood clots formed in
response to injury. This common response is certainly beneficial.
However, other actions and associations of thrombin and fibrin are
of a darker or unexpected nature (Fig. 2). In addition to its partici-
pation in blood clot formation, thrombin also triggers proteases
that attenuate clot formation [20]. Thus thrombin (and
378 Ingrid M. Verhamme et al.
Cardiovascular Cancer
(pro)Thrombin Pathway Fiber
Metabolic
Thrombosis Bleeding Fibrin(ogen) Activation Component
Fig. 2 Multiple Physiological and Pathological Roles of the (pro)Thrombin - Fibrin(ogen) Interaction
Acknowledgements
References
vascular biology. J Thromb Haemost 3 52. Kuida K (2000) Caspase-9. Int J Biochem
(8):1800–1814. https://doi.org/10.1111/j. Cell Biol 32(2):121–124
1538-7836.2005.01377.x 53. Blasche S, Mortl M, Steuber H, Siszler G,
43. Fender AC, Rauch BH, Geisler T, Schror K Nisa S, Schwarz F, Lavrik I, Gronewold TM,
(2017) Protease-activated receptor par-4: an Maskos K, Donnenberg MS, Ullmann D,
inducible switch between thrombosis and vas- Uetz P, Kogl M (2013) The E. coli effector
cular inflammation? Thromb Haemost 117 protein NleF is a caspase inhibitor. PLoS One
(11):2013–2025. https://doi.org/10.1160/ 8(3):e58937. https://doi.org/10.1371/jour
TH17-03-0219 nal.pone.0058937
44. Takamori N, Azuma H, Kato M, 54. Li P, Nijhawan D, Wang X (2004) Mitochon-
Hashizume S, Aihara K, Akaike M, drial activation of apoptosis. Cell 116
Tamura K, Matsumoto T (2004) High plasma (2 Suppl):S57–59, 52 p following S59
heparin cofactor II activity is associated with 55. Denault JB, Eckelman BP, Shin H, Pop C,
reduced incidence of in-stent restenosis after Salvesen GS (2007) Caspase 3 attenuates
percutaneous coronary intervention. Circula- XIAP (X-linked inhibitor of apoptosis
tion 109(4):481–486. https://doi.org/10. protein)-mediated inhibition of caspase
1161/01.CIR.0000109695.39671.37 9. Biochem J 405(1):11–19. https://doi.
45. Nierodzik ML, Karpatkin S (2006) Thrombin org/10.1042/BJ20070288
induces tumor growth, metastasis, and angio- 56. Creagh EM (2014) Caspase crosstalk: integra-
genesis: Evidence for a thrombin-regulated tion of apoptotic and innate immune signal-
dormant tumor phenotype. Cancer Cell 10 ling pathways. Trends Immunol 35
(5):355–362. https://doi.org/10.1016/j. (12):631–640. https://doi.org/10.1016/j.
ccr.2006.10.002 it.2014.10.004
46. Asanuma K, Wakabayashi H, Okamoto T, 57. Wang XJ, Cao Q, Liu X, Wang KT, Mi W,
Asanuma Y, Akita N, Yoshikawa T, Zhang Y, Li LF, LeBlanc AC, Su XD (2010)
Hayashi T, Matsumine A, Uchida A, Sudo A Crystal structures of human caspase 6 reveal a
(2013) The thrombin inhibitor, argatroban, new mechanism for intramolecular cleavage
inhibits breast cancer metastasis to bone. self-activation. EMBO Rep 11(11):841–847.
Breast Cancer 20(3):241–246. https://doi. https://doi.org/10.1038/embor.2010.141
org/10.1007/s12282-012-0334-5 58. Graham RK, Ehrnhoefer DE, Hayden MR
47. McIlwain DR, Berger T, Mak TW (2013) (2011) Caspase-6 and neurodegeneration.
Caspase functions in cell death and disease. Trends Neurosci 34(12):646–656. https://
Cold Spring Harb Perspect Biol 5(4): doi.org/10.1016/j.tins.2011.09.001
a008656. https://doi.org/10.1101/ 59. Bartel A, Gohler A, Hopf V, Breitbach K
cshperspect.a008656 (2017) Caspase-6 mediates resistance against
48. Li P, Nijhawan D, Budihardjo I, Srinivasula Burkholderia pseudomallei infection and
SM, Ahmad M, Alnemri ES, Wang X (1997) influences the expression of detrimental cyto-
Cytochrome c and dATP-dependent forma- kines. PLoS One 12(7):e0180203. https://
tion of Apaf-1/caspase-9 complex initiates doi.org/10.1371/journal.pone.0180203
an apoptotic protease cascade. Cell 91 60. Sollberger G, Strittmatter GE,
(4):479–489 Garstkiewicz M, Sand J, Beer HD (2014)
49. Zhivotovsky B, Samali A, Gahm A, Orrenius S Caspase-1: the inflammasome and beyond.
(1999) Caspases: their intracellular localiza- Innate Immun 20(2):115–125. https://doi.
tion and translocation during apoptosis. Cell org/10.1177/1753425913484374
Death Differ 6(7):644–651. https://doi.org/ 61. Duclos C, Lavoie C, Denault JB (2017) Cas-
10.1038/sj.cdd.4400536 pases rule the intracellular trafficking cartel.
50. Li P, Zhou L, Zhao T, Liu X, Zhang P, Liu Y, FEBS J 284(10):1394–1420. https://doi.
Zheng X, Li Q (2017) Caspase-9: structure, org/10.1111/febs.14071
mechanisms and clinical application. Oncotar- 62. Julien O, Wells JA (2017) Caspases and their
get 8(14):23996–24008. https://doi.org/ substrates. Cell Death Differ 24
10.18632/oncotarget.15098 (8):1380–1389. https://doi.org/10.1038/
51. Vu NT, Park MA, Shultz JC, Goehe RW, Hoe- cdd.2017.44
ferlin LA, Shultz MD, Smith SA, Lynch KW, 63. Aziz M, Jacob A, Wang P (2014) Revisiting
Chalfant CE (2013) hnRNP U enhances caspases in sepsis. Cell Death Dis 5:e1526.
caspase-9 splicing and is modulated by https://doi.org/10.1038/cddis.2014.488
AKT-dependent phosphorylation of hnRNP
L. J Biol Chem 288(12):8575–8584. https:// 64. Pemberton CJ (2014) Signal peptides: new
doi.org/10.1074/jbc.M112.443333 markers in cardiovascular disease? Biomark
382 Ingrid M. Verhamme et al.
dependent coagulation initiation to epithelial antitrypsin deficiency and lung cancer risk: a
proteolysis and signaling. Blood 127 case-control study in never-smokers. J Thorac
(25):3260–3269. https://doi.org/10.1182/ Oncol 10(9):1279–1284. https://doi.org/
blood-2015-11-683110 10.1097/JTO.0000000000000609
82. Verhelst SHL (2017) Intramembrane pro- 91. Soderberg D, Segelmark M (2016) Neutro-
teases as drug targets. FEBS J 284 phil extracellular traps in ANCA-associated
(10):1489–1502. https://doi.org/10.1111/ vasculitis. Front Immunol 7:256. https://
febs.13979 doi.org/10.3389/fimmu.2016.00256
83. Dusterhoft S, Kunzel U, Freeman M (2017) 92. Denadai-Souza A, Ribeiro CM, Rolland C,
Rhomboid proteases in human disease: Thouard A, Deraison C, Scavone C,
Mechanisms and future prospects. Biochim Gonzalez-Dunia D, Vergnolle N, Avellar
Biophys Acta 1864(11 Pt B):2200–2209. MCW (2017) Effect of tryptase inhibition
https://doi.org/10.1016/j.bbamcr.2017. on joint inflammation: a pharmacological
04.016 and lentivirus-mediated gene transfer study.
84. Saita S, Nolte H, Fiedler KU, Kashkar H, Arthritis Res Ther 19. https://doi.org/10.
Venne AS, Zahedi RP, Kruger M, Langer T 1186/s13075-017-1326-9
(2017) PARL mediates Smac proteolytic mat- 93. Leskinen MJ, Lindstedt KA, Wang Y, Kova-
uration in mitochondria to promote apopto- nen PT (2003) Mast cell chymase induces
sis. Nat Cell Biol 19(4):318–328. https:// smooth muscle cell apoptosis by a mechanism
doi.org/10.1038/ncb3488 involving fibronectin degradation and disrup-
85. Ranganathan P, Weaver KL, Capobianco AJ tion of focal adhesions. Arterioscler Thromb
(2011) Notch signalling in solid tumours: a Vasc Biol 23(2):238–243
little bit of everything but not all the time. 94. He A, Shi GP (2013) Mast cell chymase and
Nat Rev Cancer 11(5):338–351. https://doi. tryptase as targets for cardiovascular and met-
org/10.1038/nrc3035 abolic diseases. Curr Pharm Des 19
86. Chauhan S, Mandal P, Tomar RS (2016) Bio- (6):1114–1125
chemical analysis reveals the multifactorial 95. Shirai T, Hilhorst M, Harrison DG, Goronzy
mechanism of histone H3 clipping by chicken JJ, Weyand CM (2015) Macrophages in vas-
liver histone H3 protease. Biochemistry 55 cular inflammation—from atherosclerosis to
(38):5464–5482. https://doi.org/10.1021/ vasculitis. Autoimmunity 48(3):139–151.
acs.biochem.6b00625 https://doi.org/10.3109/08916934.2015.
87. Vossaert L, Meert P, Scheerlinck E, Glibert P, 1027815
Van Roy N, Heindryckx B, De Sutter P, 96. Sendler M, Maertin S, John D, Persike M,
Dhaenens M, Deforce D (2014) Identifica- Weiss FU, Kruger B, Wartmann T, Wagh P,
tion of histone H3 clipping activity in human Halangk W, Schaschke N, Mayerle J, Lerch
embryonic stem cells. Stem Cell Res 13 MM (2016) Cathepsin B activity initiates apo-
(1):123–134. https://doi.org/10.1016/j. ptosis via digestive protease activation in pan-
scr.2014.05.002 creatic acinar cells and experimental
88. Deraison C, Bonnart C, Vergnolle N (2018) pancreatitis. J Biol Chem 291
Proteases. In: Cavaillon J-M, Singer M (eds) (28):14717–14731. https://doi.org/10.
Inflammation: from molecular and cellular 1074/jbc.M116.718999
mechanisms to the clinic. Wiley-VCH, Wein- 97. Kayode O, Huang Z, Soares AS, Caulfield
heim, Germany, pp 727–766 TR, Dong Z, Bode AM, Radisky ES (2017)
89. Chotirmall SH, Al-Alawi M, McEnery T, Small molecule inhibitors of mesotrypsin
McElvaney NG (2015) Alpha-1 proteinase from a structure-based docking screen. PLoS
inhibitors for the treatment of alpha-1 anti- One 12(5):e0176694. https://doi.org/10.
trypsin deficiency: safety, tolerability, and 1371/journal.pone.0176694
patient outcomes. Ther Clin Risk Manag 98. Rolland-Fourcade C, Denadai-Souza A,
11:143–151. https://doi.org/10.2147/ Cirillo C, Lopez C, Jaramillo JO,
TCRM.S51474 Desormeaux C, Cenac N, Motta JP,
90. Torres-Duran M, Ruano-Ravina A, Parente- Larauche M, Tache Y, Berghe PV,
Lamelas I, Abal-Arca J, Leiro-Fernandez V, Neunlist M, Coron E, Kirzin S, Portier G,
Montero-Martinez C, Pena C, Castro- Bonnet D, Alric L, Vanner S, Deraison C,
Anon O, Golpe-Gomez A, Gonzalez-Barcala Vergnolle N (2017) Epithelial expression
FJ, Martinez C, Guzman-Taveras R, and function of trypsin-3 in irritable bowel
Provencio M, Mejuto-Marti MJ, Fernandez- syndrome. Gut 66(10):1767–1778. https://
Villar A, Barros-Dios JM (2015) Alpha-1 doi.org/10.1136/gutjnl-2016-312094
384 Ingrid M. Verhamme et al.
ZM, Renne T, Ruf W, Munzel T, Wenzel P 127. Crocker SJ, Pagenstecher A, Campbell IL
(2017) Platelet-localized FXI promotes a vas- (2004) The TIMPs tango with MMPs and
cular coagulation-inflammatory circuit in more in the central nervous system. J Neu-
arterial hypertension. Sci Transl Med 9 rosci Res 75(1):1–11. https://doi.org/10.
(375). https://doi.org/10.1126/ 1002/jnr.10836
scitranslmed.aah4923 128. Brucher BL, Jamall IS (2016) Somatic muta-
119. Camare C, Pucelle M, Negre-Salvayre A, Sal- tion theory—why it’s wrong for most cancers.
vayre R (2017) Angiogenesis in the athero- Cell Physiol Biochem 38(5):1663–1680.
sclerotic plaque. Redox Biol 12:18–34. https://doi.org/10.1159/000443106
https://doi.org/10.1016/j.redox.2017.01. 129. Brucher BL, Jamall IS (2014) Epistemology
007 of the origin of cancer: a new paradigm. BMC
120. Wilson WRW, Anderton M, Choke EC, Cancer 14:331. https://doi.org/10.1186/
Dawson J, Loftus IM, Thompson MM 1471-2407-14-331
(2008) Elevated plasma MMP1 and MMP9 130. Schuliga M (2015) The inflammatory actions
are associated with abdominal aortic aneu- of coagulant and fibrinolytic proteases in dis-
rysm rupture. Eur J Vasc Endovasc 35 ease. Mediators Inflamm 2015:437695.
(5):580–584. https://doi.org/10.1016/j. https://doi.org/10.1155/2015/437695
ejvs.2007.12.004 131. Fan J, Ning B, Lyon CJ, Hu TY (2017) Cir-
121. Xue L, Borne Y, Mattisson IY, Wigren M, culating peptidome and tumor-resident pro-
Melander O, Ohro-Melander M, teolysis. Enzyme 42:1–25. https://doi.org/
Bengtsson E, Fredrikson GN, Nilsson J, 10.1016/bs.enz.2017.08.001
Engstrom G (2017) FADD, caspase-3, and 132. Guo Z, Jin X, Jia H (2013) Inhibition of
caspase-8 and incidence of coronary events. ADAM-17 more effectively down-regulates
Arterioscler Thromb Vasc Biol 37 the Notch pathway than that of gamma-
(5):983–989. https://doi.org/10.1161/ secretase in renal carcinoma. J Exp Clin Can-
ATVBAHA.117.308995 cer Res 32:26. https://doi.org/10.1186/
122. Musante L, Tataruch D, Gu D, Liu X, 1756-9966-32-26
Forsblom C, Groop PH, Holthofer H 133. Jackson HW, Defamie V, Waterhouse P, Kho-
(2015) Proteases and protease inhibitors of kha R (2017) TIMPs: versatile extracellular
urinary extracellular vesicles in diabetic regulators in cancer. Nat Rev Cancer 17
nephropathy. J Diabetes Res 2015:289734. (1):38–53. https://doi.org/10.1038/nrc.
https://doi.org/10.1155/2015/289734 2016.115
123. Zhao Z, Yang P, Eckert RL, Reece EA (2009) 134. LaRocca G, Aspelund T, Greve AM,
Caspase-8: a key role in the pathogenesis of Eiriksdottir G, Acharya T, Thorgeirsson G,
diabetic embryopathy. Birth Defects Res B Harris TB, Launer LJ, Gudnason V, Arai AE
Dev Reprod Toxicol 86(1):72–77. https:// (2017) Fibrosis as measured by the bio-
doi.org/10.1002/bdrb.20185 marker, tissue inhibitor metalloproteinase-1,
124. Augstein P, Bahr J, Wachlin G, Heinke P, predicts mortality in Age Gene Environment
Berg S, Salzsieder E, Harrison LC (2004) Susceptibility-Reykjavik (AGES-Reykjavik)
Cytokines activate caspase-3 in insulinoma Study. Eur Heart J 38(46):3423–3430.
cells of diabetes-prone NOD mice directly https://doi.org/10.1093/eurheartj/ehx510
and via upregulation of Fas. J Autoimmun 135. Lee JY, Kong G (2016) Roles and epigenetic
23(4):301–309. https://doi.org/10.1016/j. regulation of epithelial-mesenchymal transi-
jaut.2004.09.006 tion and its transcription factors in cancer
125. Trompet S, Pons D, Kanse SM, de Craen AJ, initiation and progression. Cell Mol Life Sci
Ikram MA, Verschuren JJ, Zwinderman AH, 73(24):4643–4660. https://doi.org/10.
Doevendans PA, Tio RA, de Winter RJ, Slag- 1007/s00018-016-2313-z
boom PE, Westendorp RG, Jukema JW 136. Otsuki T, Fujimoto D, Hirono Y, Goi T,
(2011) Factor VII activating protease poly- Yamaguchi A (2014) Thrombin conducts
morphism (G534E) is associated with epithelialmesenchymal transition via protea-
increased risk for stroke and mortality. Stroke seactivated receptor1 in human gastric cancer.
Res Treat 2011:424759. https://doi.org/10. Int J Oncol 45(6):2287–2294. https://doi.
4061/2011/424759 org/10.3892/ijo.2014.2651
126. Turner RJ, Sharp FR (2016) Implications of 137. Bawa-Khalfe T, Lu LS, Zuo Y, Huang C,
MMP9 for blood brain barrier disruption and Dere R, Lin FM, Yeh ET (2012) Differential
hemorrhagic transformation following ische- expression of SUMO-specific protease 7 var-
mic stroke. Front Cell Neurosci 10:56. iants regulates epithelial-mesenchymal
https://doi.org/10.3389/fncel.2016.00056
386 Ingrid M. Verhamme et al.
Song L, Hanlon D, Tan Hehir CA, Baker D, 165. Siklos M, BenAissa M, Thatcher GR (2015)
Blennow K, Hansson O (2016) Plasma beta- Cysteine proteases as therapeutic targets: does
amyloid in Alzheimer’s disease and vascular selectivity matter? A systematic review of cal-
disease. Sci Rep 6:26801. https://doi.org/ pain and cathepsin inhibitors. Acta Pharm Sin
10.1038/srep26801 B 5(6):506–519. https://doi.org/10.1016/
157. Roher AE, Kokjohn TA, Clarke SG, Sierks j.apsb.2015.08.001
MR, Maarouf CL, Serrano GE, Sabbagh 166. Martinelli P, Rugarli EI (2010) Emerging
MS, Beach TG (2017) APP/Abeta structural roles of mitochondrial proteases in neurode-
diversity and Alzheimer’s disease pathogene- generation. Biochim Biophys Acta 1797
sis. Neurochem Int 110:1–13. https://doi. (1):1–10. https://doi.org/10.1016/j.
org/10.1016/j.neuint.2017.08.007 bbabio.2009.07.013
158. Evin G, Li QX (2012) Platelets and Alzhei- 167. Konig T, Troder SE, Bakka K, Korwitz A,
mer’s disease: potential of APP as a bio- Richter-Dennerlein R, Lampe PA, Patron M,
marker. World J Psychiatry 2(6):102–113. Muhlmeister M, Guerrero-Castillo S,
https://doi.org/10.5498/wjp.v2.i6.102 Brandt U, Decker T, Lauria I, Paggio A,
159. Wetzel S, Seipold L, Saftig P (2017) The Rizzuto R, Rugarli EI, De Stefani D, Langer
metalloproteinase ADAM10: A useful thera- T (2016) The m-AAA protease associated
peutic target? Biochim Biophys Acta 1864 with neurodegeneration limits MCU activity
(11 Pt B):2071–2081. https://doi.org/10. in mitochondria. Mol Cell 64(1):148–162.
1016/j.bbamcr.2017.06.005 https://doi.org/10.1016/j.molcel.2016.08.
160. Bu XL, Xiang Y, Jin WS, Wang J, Shen LL, 020
Huang ZL, Zhang K, Liu YH, Zeng F, Liu 168. Strauss KM, Martins LM, Plun-Favreau H,
JH, Sun HL, Zhuang ZQ, Chen SH, Yao XQ, Marx FP, Kautzmann S, Berg D, Gasser T,
Giunta B, Shan YC, Tan J, Chen XW, Dong Wszolek Z, Muller T, Bornemann A,
ZF, Zhou HD, Zhou XF, Song W, Wang YJ Wolburg H, Downward J, Riess O, Schulz
(2017) Blood-derived amyloid-beta protein JB, Kruger R (2005) Loss of function muta-
induces Alzheimer’s disease pathologies. Mol tions in the gene encoding Omi/HtrA2 in
Psychiatry. https://doi.org/10.1038/mp. Parkinson’s disease. Hum Mol Genet 14
2017.204 (15):2099–2111. https://doi.org/10.1093/
161. Budd Haeberlein S, O’Gorman J, Chiao P, hmg/ddi215
Bussiere T, von Rosenstiel P, Tian Y, Zhu Y, 169. Fu J, Yu HM, Chiu SY, Mirando AJ, Mar-
von Hehn C, Gheuens S, Skordos L, Chen T, uyama EO, Cheng JG, Hsu W (2014) Dis-
Sandrock A (2017) Clinical development of ruption of SUMO-specific protease 2 induces
aducanumab, an anti-abeta human monoclo- mitochondria mediated neurodegeneration.
nal antibody being investigated for the treat- PLoS Genet 10(10):e1004579. https://doi.
ment of early Alzheimer’s disease. J Prev org/10.1371/journal.pgen.1004579
Alzheimers Dis 4(4):255–263. https://doi. 170. Orsini F, De Blasio D, Zangari R, Zanier ER,
org/10.14283/jpad.2017.39 De Simoni MG (2014) Versatility of the com-
162. Honig LS, Vellas B, Woodward M, Boada M, plement system in neuroinflammation, neuro-
Bullock R, Borrie M, Hager K, Andreasen N, degeneration and brain homeostasis. Front
Scarpini E, Liu-Seifert H, Case M, Dean RA, Cell Neurosci 8:380. https://doi.org/10.
Hake A, Sundell K, Poole Hoffmann V, 3389/fncel.2014.00380
Carlson C, Khanna R, Mintun M, 171. Descamps FJ, Van den Steen PE, Nelissen I,
DeMattos R, Selzler KJ, Siemers E (2018) Van Damme J, Opdenakker G (2003) Rem-
Trial of solanezumab for mild dementia due nant epitopes generate autoimmunity: from
to Alzheimer’s disease. N Engl J Med 378 rheumatoid arthritis and multiple sclerosis to
(4):321–330. https://doi.org/10.1056/ diabetes. Adv Exp Med Biol 535:69–77
NEJMoa1705971 172. Dinarello CA, Simon A, van der Meer JW
163. De Strooper B (2014) Lessons from a failed (2012) Treating inflammation by blocking
gamma-secretase Alzheimer trial. Cell 159 interleukin-1 in a broad spectrum of diseases.
(4):721–726. https://doi.org/10.1016/j. Nat Rev Drug Discov 11(8):633–652.
cell.2014.10.016 https://doi.org/10.1038/nrd3800
164. Lorenzl S, Albers DS, Relkin N, Ngyuen T, 173. Rupanagudi KV, Kulkarni OP, Lichtnekert J,
Hilgenberg SL, Chirichigno J, Cudkowicz Darisipudi MN, Mulay SR, Schott B,
ME, Beal MF (2003) Increased plasma levels Gruner S, Haap W, Hartmann G, Anders HJ
of matrix metalloproteinase-9 in patients with (2015) Cathepsin S inhibition suppresses sys-
Alzheimer’s disease. Neurochem Int 43 temic lupus erythematosus and lupus nephri-
(3):191–196 tis because cathepsin S is essential for MHC
388 Ingrid M. Verhamme et al.
class II-mediated CD4 T cell and B cell 182. Carroll IM, Maharshak N (2013) Enteric bac-
priming. Ann Rheum Dis 74(2):452–463. terial proteases in inflammatory bowel dis-
https://doi.org/10.1136/annrheumdis- ease- pathophysiology and clinical
2013-203717 implications. World J Gastroenterol 19
174. Schaller M, Vogel M, Kentouche K, (43):7531–7543. https://doi.org/10.3748/
Lammle B, Kremer Hovinga JA (2014) The wjg.v19.i43.7531
splenic autoimmune response to ADAMTS13 183. Boxrud PD, Verhamme IM, Bock PE (2004)
in thrombotic thrombocytopenic purpura Resolution of conformational activation in
contains recurrent antigen-binding CDR3 the kinetic mechanism of plasminogen activa-
motifs. Blood 124(23):3469–3479. https:// tion by streptokinase. J Biol Chem 279
doi.org/10.1182/blood-2014-04-561142 (35):36633–36641. https://doi.org/10.
175. Sadiq SK, Noe F, De Fabritiis G (2012) 1074/jbc.M405264200
Kinetic characterization of the critical step in 184. Chandrahas V, Glinton K, Liang Z, Donahue
HIV-1 protease maturation. Proc Natl Acad DL, Ploplis VA, Castellino FJ (2015) Direct
Sci U S A 109(50):20449–20454. https:// host plasminogen binding to bacterial surface
doi.org/10.1073/pnas.1210983109 M-protein in pattern D strains of streptococ-
176. Duschak VG, Couto AS (2009) Cruzipain, cus pyogenes is required for activation by its
the major cysteine protease of Trypanosoma natural coinherited SK2b protein. J Biol
cruzi: a sulfated glycoprotein antigen as rele- Chem 290(30):18833–18842. https://doi.
vant candidate for vaccine development and org/10.1074/jbc.M115.655365
drug target. A review. Curr Med Chem 16 185. Panizzi P, Friedrich R, Fuentes-Prior P,
(24):3174–3202 Bode W, Bock PE (2004) The staphylocoagu-
177. Plaza K, Kalinska M, Bochenska O, Meyer- lase family of zymogen activator and adhesion
Hoffert U, Wu Z, Fischer J, Falkowski K, proteins. Cell Mol Life Sci 61
Sasiadek L, Bielecka E, Potempa B, Kozik A, (22):2793–2798. https://doi.org/10.1007/
Potempa J, Kantyka T (2016) Gingipains of s00018-004-4285-7
porphyromonas gingivalis affect the stability 186. Parry MA, Zhang XC, Bode I (2000) Molec-
and function of serine protease inhibitor of ular mechanisms of plasminogen activation:
kazal-type 6 (SPINK6), a tissue inhibitor of bacterial cofactors provide clues. Trends Bio-
human kallikreins. J Biol Chem 291 chem Sci 25(2):53–59
(36):18753–18764. https://doi.org/10. 187. Wiles KG, Panizzi P, Kroh HK, Bock PE
1074/jbc.M116.722942 (2010) Skizzle is a novel plasminogen- and
178. Culp E, Wright GD (2017) Bacterial pro- plasmin-binding protein from Streptococcus
teases, untapped antimicrobial drug targets. agalactiae that targets proteins of human fibri-
J Antibiot (Tokyo) 70(4):366–377. https:// nolysis to promote plasmin generation. J Biol
doi.org/10.1038/ja.2016.138 Chem 285(27):21153–21164. https://doi.
179. Chang AK, Kim HY, Park JE, Acharya P, Park org/10.1074/jbc.M110.107730
IS, Yoon SM, You HJ, Hahm KS, Park JK, 188. Verhamme IM, Panizzi PR, Bock PE (2015)
Lee JS (2005) Vibrio vulnificus secretes a Pathogen activators of plasminogen. J
broad-specificity metalloprotease capable of Thromb Haemost 13(Suppl 1):S106–S114.
interfering with blood homeostasis through https://doi.org/10.1111/jth.12939
prothrombin activation and fibrinolysis. J 189. Meliopoulos VA, Andersen LE, Brooks P,
Bacteriol 187(20):6909–6916. https://doi. Yan X, Bakre A, Coleman JK, Tompkins SM,
org/10.1128/JB.187.20.6909-6916.2005 Tripp RA (2012) MicroRNA regulation of
180. Bibo-Verdugo B, Jiang Z, Caffrey CR, human protease genes essential for influenza
O’Donoghue AJ (2017) Targeting protea- virus replication. PLoS One 7(5):e37169.
somes in infectious organisms to combat dis- https://doi.org/10.1371/journal.pone.
ease. FEBS J 284(10):1503–1517. https:// 0037169
doi.org/10.1111/febs.14029 190. Homma T, Ishibashi D, Nakagaki T, Fuse T,
181. Pontarollo G, Acquasaliente L, Peterle D, Mori T, Satoh K, Atarashi R, Nishida N
Frasson R, Artusi I, De Filippis V (2017) (2015) Ubiquitin-specific protease 14 modu-
Non-canonical proteolytic activation of lates degradation of cellular prion protein. Sci
human prothrombin by subtilisin from Bacil- Rep 5:11028. https://doi.org/10.1038/
lus subtilis may shift the procoagulant- srep11028
anticoagulant equilibrium toward thrombo- 191. Michaud DS, Lu J, Peacock-Villada AY, Bar-
sis. J Biol Chem 292(37):15161–15179. ber JR, Joshu CE, Prizment AE, Beck JD,
https://doi.org/10.1074/jbc.M117. Offenbacher S, Platz EA (2018) Periodontal
795245 disease assessed using clinical dental
Proteases: Pivot Points in Functional Proteomics 389
measurements and cancer risk in the ARIC 202. Tallant C, Marrero A, Gomis-Ruth FX (2010)
study. J Natl Cancer Inst. https://doi.org/ Matrix metalloproteinases: fold and function
10.1093/jnci/djx278 of their catalytic domains. Biochim Biophys
192. Drag M, Salvesen GS (2010) Emerging prin- Acta 1803(1):20–28. https://doi.org/10.
ciples in protease-based drug discovery. Nat 1016/j.bbamcr.2009.04.003
Rev Drug Discov 9(9):690–701. https://doi. 203. Gomis-Ruth FX (2017) Third time lucky?
org/10.1038/nrd3053 Getting a grip on matrix metalloproteinases.
193. Herzog RW (2015) Hemophilia gene ther- J Biol Chem 292(43):17975–17976. https://
apy: caught between a cure and an immune doi.org/10.1074/jbc.H117.806075
response. Mol Ther 23(9):1411–1412. 204. Scannevin RH, Alexander R, Haarlander TM,
https://doi.org/10.1038/mt.2015.135 Burke SL, Singer M, Huo C, Zhang YM,
194. Rangarajan S, Walsh L, Lester W, Perry D, Maguire D, Spurlino J, Deckman I, Carroll
Madan B, Laffan M, Yu H, Vettermann C, KI, Lewandowski F, Devine E,
Pierce GF, Wong WY, Pasi KJ (2017) AAV5- Dzordzorme K, Tounge B, Milligan C,
factor VIII gene transfer in severe hemophilia Bayoumy S, Williams R, Schalk-Hihi C,
A. N Engl J Med. https://doi.org/10.1056/ Leonard K, Jackson P, Todd M, Kuo LC,
NEJMoa1708483 Rhodes KJ (2017) Discovery of a highly selec-
195. George LA, Sullivan SK, Giermasz A, Rasko tive chemical inhibitor of matrix
JEJ, Samelson-Jones BJ, Ducore J, Cuker A, metalloproteinase-9 (MMP-9) that allosteri-
Sullivan LM, Majumdar S, Teitel J, McGuinn cally inhibits zymogen activation. J Biol
CE, Ragni MV, Luk AY, Hui D, Wright JF, Chem 292(43):17963–17974. https://doi.
Chen Y, Liu Y, Wachtel K, Winters A, org/10.1074/jbc.M117.806075
Tiefenbacher S, Arruda VR, van der Loo 205. Grunwald B, Vandooren J, Gerg M,
JCM, Zelenaia O, Takefman D, Carr ME, Ahomaa K, Hunger A, Berchtold S,
Couto LB, Anguela XM, High KA (2017) Akbareian S, Schaten S, Knolle P, Edwards
Hemophilia B gene therapy with a high- DR, Opdenakker G, Kruger A (2016) Sys-
specific-activity factor IX variant. N Engl J temic ablation of MMP-9 triggers invasive
Med 377(23):2215–2227. https://doi.org/ growth and metastasis of pancreatic cancer
10.1056/NEJMoa1708538 via deregulation of IL6 expression in the
196. Lai PS, Thompson BT (2013) Why activated bone marrow. Mol Cancer Res 14
protein C was not successful in severe sepsis (11):1147–1158. https://doi.org/10.1158/
and septic shock: are we still tilting at wind- 1541-7786.MCR-16-0180
mills? Curr Infect Dis Rep 15(5):407–412. 206. Radisky ES, Raeeszadeh-Sarmazdeh M,
https://doi.org/10.1007/s11908-013- Radisky DC (2017) Therapeutic potential of
0358-9 matrix metalloproteinase inhibition in breast
197. Janciauskiene SM, Bals R, Koczulla R, cancer. J Cell Biochem 118(11):3531–3548.
Vogelmeier C, Kohnlein T, Welte T (2011) https://doi.org/10.1002/jcb.26185
The discovery of alpha1-antitrypsin and its 207. Xia D, Watanabe H, Wu B, Lee SH, Li Y,
role in health and disease. Respir Med 105 Tsvetkov E, Bolshakov VY, Shen J, Kelleher
(8):1129–1139. https://doi.org/10.1016/j. RJ 3rd (2015) Presenilin-1 knockin mice
rmed.2011.02.002 reveal loss-of-function mechanism for familial
198. Carugati A, Pappalardo E, Zingale LC, Alzheimer’s disease. Neuron 85(5):967–981.
Cicardi M (2001) C1-inhibitor deficiency https://doi.org/10.1016/j.neuron.2015.
and angioedema. Mol Immunol 38 02.010
(2-3):161–173 208. Ran Y, Hossain F, Pannuti A, Lessard CB,
199. Ricklin D, Lambris JD (2016) New mile- Ladd GZ, Jung JI, Minter LM, Osborne BA,
stones ahead in complement-targeted ther- Miele L, Golde TE (2017) gamma-Secretase
apy. Semin Immunol 28(3):208–222. inhibitors in cancer clinical trials are pharma-
https://doi.org/10.1016/j.smim.2016.06. cologically and functionally distinct. EMBO
001 Mol Med 9(7):950–966. https://doi.org/
10.15252/emmm.201607265
200. Towards better patient care: drugs to avoid in
2014 (2014). Prescrire Int 23(150):161–165 209. Duong le T, Leung AT, Langdahl B (2016)
Cathepsin K inhibition: a new mechanism for
201. Adcock DM, Gosselin R (2015) Direct Oral the treatment of osteoporosis. Calcif Tissue
Anticoagulants (DOACs) in the Laboratory: Int 98(4):381–397. https://doi.org/10.
2015 Review. Thromb Res 136(1):7–12. 1007/s00223-015-0051-0
https://doi.org/10.1016/j.thromres.2015.
05.001 210. Drake MT, Clarke BL, Oursler MJ, Khosla S
(2017) Cathepsin K inhibitors for
390 Ingrid M. Verhamme et al.
229. Vance NR, Gakhar L, Spies MA (2017) Allo- Drag M (2014) Design of ultrasensitive
steric tuning of caspase-7: a fragment-based probes for human neutrophil elastase through
drug discovery approach. Angew Chem Int hybrid combinatorial substrate library
Ed Engl 56(46):14443–14447. https://doi. profiling. Proc Natl Acad Sci U S A 111
org/10.1002/anie.201706959 (7):2518–2523. https://doi.org/10.1073/
230. Weiss-Sadan T, Gotsman I, Blum G (2017) pnas.1318548111
Cysteine proteases in atherosclerosis. FEBS J 240. O’Donoghue AJ, Eroy-Reveles AA, Knudsen
284(10):1455–1472. https://doi.org/10. GM, Ingram J, Zhou M, Statnekov JB, Gre-
1111/febs.14043 ninger AL, Hostetter DR, Qu G, Maltby DA,
231. Lee S, Xie J, Chen X (2010) Activatable Anderson MO, Derisi JL, McKerrow JH,
molecular probes for cancer imaging. Curr Burlingame AL, Craik CS (2012) Global
Top Med Chem 10(11):1135–1144 identification of peptidase specificity by mul-
232. Ren G, Blum G, Verdoes M, Liu H, Syed S, tiplex substrate profiling. Nat Methods 9
Edgington LE, Gheysens O, Miao Z, Jiang H, (11):1095–1100. https://doi.org/10.1038/
Gambhir SS, Bogyo M, Cheng Z (2011) nmeth.2182
Non-invasive imaging of cysteine cathepsin 241. Li Q, Yi L, Hoi KH, Marek P, Georgiou G,
activity in solid tumors using a 64Cu-labeled Iverson BL (2017) Profiling protease specific-
activity-based probe. PLoS One 6(11): ity: combining yeast ER sequestration screen-
e28029. https://doi.org/10.1371/journal. ing (YESS) with next generation sequencing.
pone.0028029 ACS Chem Biol 12(2):510–518. https://doi.
233. Indalao IL, Sawabuchi T, Takahashi E, Kido org/10.1021/acschembio.6b00547
H (2017) IL-1beta is a key cytokine that 242. Fulcher LJ, Hutchinson LD, Macartney TJ,
induces trypsin upregulation in the influenza Turnbull C, Sapkota GP (2017) Targeting
virus-cytokine-trypsin cycle. Arch Virol 162 endogenous proteins for degradation
(1):201–211. https://doi.org/10.1007/ through the affinity-directed protein missile
s00705-016-3093-3 system. Open Biol 7(5). https://doi.org/10.
234. Kelso EB, Lockhart JC, Hembrough T, 1098/rsob.170066
Dunning L, Plevin R, Hollenberg MD, Som- 243. Grossi G, Dalgaard Ebbesen Jepsen M,
merhoff CP, McLean JS, Ferrell WR (2006) Kjems J, Andersen ES (2017) Control of
Therapeutic promise of proteinase-activated enzyme reactions by a reconfigurable DNA
receptor-2 antagonism in joint inflammation. nanovault. Nat Commun 8(1):992. https://
J Pharmacol Exp Ther 316(3):1017–1024. doi.org/10.1038/s41467-017-01072-8
https://doi.org/10.1124/jpet.105.093807 244. Lange PF, Huesgen PF, Overall CM (2012)
235. Vergnolle N (2009) Protease-activated recep- TopFIND 2.0—linking protein termini with
tors as drug targets in inflammation and pain. proteolytic processing and modifications
Pharmacol Ther 123(3):292–309. https:// altering protein function. Nucleic Acids Res
doi.org/10.1016/j.pharmthera.2009.05. 40(Database issue):D351–D361. https://
004 doi.org/10.1093/nar/gkr1025
236. French SL, Hamilton JR (2016) Protease- 245. Wang X, Davies M, Roy S, Kuruc M (2015)
activated receptor 4: from structure to func- Bead based proteome enrichment enhances
tion and back again. Br J Pharmacol 173 features of the protein elution plate (PEP)
(20):2952–2965. https://doi.org/10.1111/ for functional proteomic profiling. Proteomes
bph.13455 3(4):454–466. https://doi.org/10.3390/
237. Kasperkiewicz P, Poreba M, Groborz K, Drag proteomes3040454
M (2017) Emerging challenges in the design 246. Zheng H, Roy S, Soherwardy A, Rahman S,
of selective substrates, inhibitors and activity- Kuruc M (2017) Stroma liquid biopsy—pro-
based probes for indistinguishable proteases. teomic profiles for cancer biomarkers. Poster
FEBS J 284(10):1518–1539. https://doi. reprint first presented at NJ Cancer Retreat,
org/10.1111/febs.14001 May 25, 2017 New Brunswick, NJ, USA
238. Harris JL, Backes BJ, Leonetti F, Mahrus S, 247. Rifai N, Gillette MA, Carr SA (2006) Protein
Ellman JA, Craik CS (2000) Rapid and gen- biomarker discovery and validation: the long
eral profiling of protease specificity by using and uncertain path to clinical utility. Nat Bio-
combinatorial fluorogenic substrate libraries. technol 24(8):971–983. https://doi.org/10.
Proc Natl Acad Sci U S A 97(14):7754–7759. 1038/nbt1235
https://doi.org/10.1073/pnas.140132697 248. Koenig W (2003) Fibrin(ogen) in cardiovas-
239. Kasperkiewicz P, Poreba M, Snipas SJ, cular disease: an update. Thromb Haemost 89
Parker H, Winterbourn CC, Salvesen GS, (4):601–609
392 Ingrid M. Verhamme et al.
249. Dunn EJ, Ariens RA, Grant PJ (2005) The (2010) Alzheimer’s disease peptide beta-
influence of type 2 diabetes on fibrin structure amyloid interacts with fibrinogen and induces
and function. Diabetologia 48 its oligomerization. Proc Natl Acad Sci U S A
(6):1198–1206. https://doi.org/10.1007/ 107(50):21812–21817. https://doi.org/10.
s00125-005-1742-2 1073/pnas.1010373107
250. Grammas P, Martinez JM (2014) Targeting 252. Amara U, Rittirsch D, Flierl M, Bruckner U,
thrombin: an inflammatory neurotoxin in Klos A, Gebhard F, Lambris JD, Huber-Lang
Alzheimer’s disease. J Alzheimers Dis 42 M (2008) Interaction between the coagula-
(Suppl 4):S537–S544. https://doi.org/10. tion and complement system. Adv Exp Med
3233/JAD-141557 Biol 632:71–79
251. Ahn HJ, Zamolodchikov D, Cortes-Canteli-
M, Norris EH, Glickman JF, Strickland S
Chapter 21
Abstract
The recent progress of proteomic protocols led to more efficient protein extraction and concentration
procedures to remove nonprotein interfering compounds present in the starting material and to increase
the concentration of underrepresented proteins. Combinatorial hexapeptide ligand libraries (CPLL) were
recently applied to both plant- and animal-derived tissues for capturing the low- and very low-abundance
allergens. Several IgE-binding proteins which were previously absent or poorly represented by using
conventional proteomics tools have been detected and characterized through a CPLL-based approach. In
the present chapter, a protocol based on improved protein extraction and enrichment by CPLL, allowing
the immunochemical characterization of several “hidden allergens” in cypress pollen, is described in detail.
Key words Hexapeptide ligand libraries, Low-abundance allergens, Pollen allergens, Mass spectrom-
etry, Proteomics
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_21, © Springer Science+Business Media, LLC, part of Springer Nature 2019
393
394 Youcef Shahali et al.
Fig. 1 Schematic illustration of the enrichment process of low-abundant protein species using ProteoMiner
beads. A high number of hexapeptide ligands are used in a single analysis which independently bind specific
proteins until they reach their maximum capacity. Hexapeptides having an affinity to high-abundant proteins
(e.g., P1 and P2) are rapidly saturate, whereas low-abundance proteins (e.g., P5 and P6) are quantitatively
trapped and concentrated. High-abundant proteins exceeding the capacity of the beads are removed
2 Materials
Cypress pollen
protein extraction
Fig. 2 Schematic experimental procedure of pollen extraction, CPLL treatment, and protein elution from the
ProteoMiner beads (adapted from Shahali et al.) [3]. Since the pH influences the protein capture by CPLL,
protein enrichment by CPLL was performed at three different pHs. In order to ensure complete protein
stripping from the beads, four elution steps were sequentially performed
Fig. 3 Overlapping Venn diagram of the total proteins detected by LC-MS/MS analysis. LC-MS analysis was
performed in control, untreated PUN and PBS extracts (blue circle) and in pooled eluates (yellow circle) from
hexapeptide ligand libraries (from PUN and PBS). The green represented common proteins found in the two
types of extraction products. Adapted from Shahali et al. [3]
The Use of Combinatorial Hexapeptide Ligand Library (CPLL) in Allergomics 397
Fig. 4 Two-dimensional IgE immunoblotting probed with the serum of a cypress pollen allergic patient
(adapted from Shahali et al.) [3]. PBS control extract: a; PBS eluate: c; PUN control extract: b; PUN eluate:
d. Novel IgE-binding were identified in CPLL eluates (outlined in boxes)
3 Method
3.1 Pollen Protein 1. Slurry ten times 5 g of pollen (total 50 g) in PUN buffer (see
Extraction Note 2).
2. Shake tubes containing the pollen suspension for 2–3 min to
wet the pollen grains.
3. Sonicate the suspension for 20 s at 4 C.
4. Place the tube on a rotator and gently shake the suspension
overnight at room temperature.
5. Centrifuge the suspension at 10,000 g and 4 C during
15 min.
6. Pipet the supernatant into a new tube, label PUN extract
and date.
7. Add 1 mL of PUN buffer on remaining pollen pellets.
8. Homogenize and centrifuge the suspension at 10,000 g and
4 C during 15 min.
9. Pool the supernatant with the PUN extract previously obtained
in step 6. The overall volume of the PUN extract was 100 mL.
10. Add 30 mL of PBS pH 7.5 to each pollen pellet.
11. Place the tube on a rotator and shake the suspension overnight
at room temperature.
12. Collect the supernatants by centrifugation at 10,000 g and
4 C during 15 min.
13. Pool all PBS extracts. The resulting total volume was 125 mL.
3.2 Removal of 1. Add ammonium sulfate to both PUN and PBS extracts under
Interfering Substances stirring to reach up to 90% saturation or, respectively, 66 g (for
Prior to CPLL Analysis 100 mL) and 82.5 g (for 125 mL).
2. Gently agitate the two mixtures at 4 C overnight.
The Use of Combinatorial Hexapeptide Ligand Library (CPLL) in Allergomics 399
14. Wash the beads with distilled water and then separate the bead
pellet by centrifugation at 1000 g for 30–60 s at 15 C.
15. Discard excess water and store the bead pellet at 4 C for
further use.
16. Mix the supernatant collected in the step 13 with a third
sample of 100 μL prewashed ProteoMiner beads.
17. Adjust the pH of the suspension to 9.25 by dropwise addition
of 4 M ammonia.
18. Gently shake the resulting suspension at room temperature for
2 h.
19. Separate beads by centrifugation at 1000 g for 30–60 s and
discard the supernatant.
20. Wash the beads once with distilled water and then separate the
bead pellet by centrifugation at 1000 g for 30–60 s at 15 C.
21. Discard excess water and store the bead pellet at 4 C for
further use.
3.4 Protein elution 1. Blend together the three bead aliquots of 100 μL each (col-
from CPLL Beads lected from steps 8, 15, and 21 of the CPLL treatment).
2. Rapidly wash once the beads with 600 μL of distilled water and
discard supernatants.
3. Add 500 μL of TUC elution solution to 300 μL bead pellets.
4. Gently shake the mixture for 2 h at room temperature.
5. Centrifuge the mixture at 1000 g for 30–60 s and collect the
supernatant comprising desorbed proteins (eluate “a”).
6. Wash the bead pellet once with 300 μL distilled water and
collect by centrifugation the supernatant (eluate “b”).
7. Submit the resulting bead pellet to the third elution by adding
500 μL of a solution composed of 6% SDS and 2%
2-Mercaptoethanol (SDS+ 2-Me).
8. Gently shake the mixture for 2 h at room temperature.
9. Centrifuge the mixture at 1000 g for 30–60 s and collect the
supernatant (eluate “c”).
10. In order to ensure complete protein stripping from the beads,
perform a fourth elution step by adding the UCA solution to
the bead pellet.
11. Gently shake the mixture for 2 h at room temperature.
12. Centrifuge the mixture at 1000 g for 30–60 s and collect the
supernatant (eluate “d”).
13. Pool the four eluates together (PUN eluate).
14. Adjust the pH to 7.0 and dialyze (cutoff 3500 Da) PUN eluate
against 3 M urea overnight (see Note 8).
The Use of Combinatorial Hexapeptide Ligand Library (CPLL) in Allergomics 401
4 Notes
Acknowledgments
References
(2012) DIGE enables the detection of a puta- between microarray and immunoblot for the
tive serum biomarker of fungal origin in a comparative evaluation of IgE repertoire of
mouse model of invasive aspergillosis. J Prote- French and Italian cypress pollen allergic
ome 75:2536–2549 patients. Folia Biologica (Prague) 60:192
11. Martos G, López-Fandiño R, Molina E (2013) 14. Danti R, Della Rocca G, Calamassi R, Mori B,
Immunoreactivity of hen egg allergens: influ- Mariotti Lippi M (2011) Insights into a hydra-
ence on in vitro gastrointestinal digestion of tion regulating system in Cupressus pollen
the presence of other egg white proteins and grains. Ann Bot 108:299–306
of egg yolk. Food Chem 136:775–781 15. Shahali Y (2011) Etude analytique de l’allergie
12. Hartwig S, Lehr S (2012) Combination of au pollen de cyprès: aspects moléculaires et
highly efficient hexapeptide ligand library- particulaires, Thesis Université Paris VI, Pierre
based sample preparation with 2D DIGE for et Marie Curie, Paris, France, p 220
the analysis of the hidden human serum/ 16. Fasoli E, Farinazzo A, Sun CJ, Kravchuk AV,
plasma proteome. Methods Mol Biol Guerrier L, Fortis F et al (2010) Interaction
854:169–180 among proteins and peptide libraries in prote-
13. Shahali Y, Nicaise P, Brazdova A, Charpin D, ome analysis: pH involvement for a larger cap-
Scala E, Mari A et al (2014) Complementarity ture of species. J Proteome 73:733–742
Chapter 22
Abstract
Coeliac disease (CD) is a T-cell mediated autoimmune disorder triggered by ingestion of cereal gluten
found in wheat (gliadins and glutenins), barley (hordeins), and rye (secalins). As the only treatment for CD
is a lifelong gluten-free diet, the measurement of gluten in raw ingredients and processed food products is
critical to protecting people with CD or gluten intolerance. The most commonly employed method is the
enzyme-linked immunosorbent assay (ELISA), but more recently mass spectrometry has been employed
wherein the extracted gluten proteins are digested to peptides that are then directly measured. To achieve
the goal of accurate gluten quantitation, gluten must be efficiently extracted from the ingredient or food
matrix and then digested to yield the peptides that are monitored by LC-MS. In this chapter, a rapid,
simple, and reproducible protocol for extraction and digestion of gluten proteins is described.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_22, © Springer Science+Business Media, LLC, part of Springer Nature 2019
405
406 Haili Li et al.
2 Materials
2.3 HPLC 1. HPLC buffer A: 0.1% formic acid, 99.9% water. Mix by inver-
Components sion (see Note 9).
2. HPLC buffer B: 0.1% formic acid, 90% acetonitrile, 9.9% water.
Mix by inversion.
3 Methods
3.1 Gluten Extraction 1. Weigh 20 mg of flour (or milled food product) into a 1.5 mL
eppendorf tube.
2. Add 200 μL of 55% IPA/2% DTT and vortex the tube until
flour is properly mixed with solution (see Note 10).
3. Place the tube in an ultrasonic bath for 5 min at room
temperature.
4. Put the tube in a dry block heater at 50 C for 30 min (see Note 11).
5. Centrifuge the suspension for 10 min at 20,800 g.
6. Transfer the supernatant, containing the gluten, into a fresh tube.
3.2 Protein Digestion 1. Transfer 100 μL of gluten extract to a 10 kDa MWCO filter,
add 100 μL of UA buffer, and centrifuge at 20,800 g for
15 min.
2. Wash the protein by addition of 200 μL of UA buffer to the
filter unit. Centrifuge at 20,800 g for 15 min.
3. Add 100 μL of IAM solution and incubate at room tempera-
ture for 20 min in the dark (see Notes 5 and 12).
4. Centrifuge the filter units at 20,800 g for 15 min.
5. Add 200 μL of UA to the filter unit and centrifuge at
20,800 g for 15 min to remove excess IAM. Discard the
flow-through from the collection tube.
6. Exchange the buffer (see Note 13) by adding 200 μL of 50 mM
ammonium bicarbonate to the filter unit and centrifuge at
20,800 g for 15 min. Repeat.
7. Transfer the filter units to new collection tubes. Digest the
protein by adding 200 μL of 0.25 mg/mL trypsin or chymo-
trypsin (in 50 mM ammonium bicarbonate, 1 mM CaCl2) and
mix briefly at low speed (400 rpm). Incubate the units in a wet
chamber at 37 C overnight (~18 h).
8. Centrifuge the filter units at 20,800 g for 15 min to collect
the digested peptides. Wash the filter by adding 200 μL of
50 mM ammonium bicarbonate and centrifuge the filter units
at 20,800 g for 15 min.
9. Lyophilize the filtrate in a vacuum centrifuge and store at
20 C until analysis.
408 Haili Li et al.
Table 1
Gluten peptide markers used in the assessment of digestion efficiency. All transitions for a given
peptide are summed to give the peak area
Uniprot RT Q3 m/z CE
# Peptide sequence accession (min) Q1 m/z (z) (fragment, z) (V)
G1 ELQESSLEAC(cam)R Barley: I6TRS8 3.15 661.296 (2+) 735.325 (y6, 1+) 33.1
Wheat: P10387 822.357 (y7, 1+)
Rye: Q94IL1 951.400 (y7, 1+)
G2 AQQLAAQLPAMC Barley: I6TRS8 4.90 729.361 (2+) 747.343 (y6, 1+) 36.5
(cam)R Wheat: P08489 946.439 (y8, 1+)
Rye: D3XQB7 1017.476 (y9, 1+)
G2* AQQLAAQLPAM(ox)C Barley: I6TRS8 4.00 737.361 (2+) 763.343 (y6, 1+) 36.9
(cam)R Wheat: P08489 962.439 (y8, 1+)
Rye: D3XQB7 1033.476 (y9, 1+)
RT retention time (min), Q1 precursor ion m/z with charge, z, Q3 product ion m/z with fragment ion assignment and
charge, z, CE collision energy (V)
Gluten Extraction and Digestion 409
4 Notes
Acknowledgments
References
1. Guandalini S, Assiri A (2014) Celiac disease: a 2. Colgrave ML, Byrne K, Blundell M et al
review. JAMA Pediatr 168(3):272–278 (2016) Comparing multiple reaction monitor-
ing and sequential window acquisition of all
412 Haili Li et al.
theoretical mass spectra for the relative quanti- 9. Colgrave ML, Byrne K, Blundell M et al
fication of barley gluten in selectively bred bar- (2016) Identification of barley-specific peptide
ley lines. Anal Chem 88(18):9127–9135 markers that persist in processed foods and are
3. Stamnaes J, Sollid LM (2015) Celiac disease: capable of detecting barley contamination by
autoimmunity in response to food antigen. LC-MS/MS. J Proteome 147:169–176
Semin Immunol 27(5):343–352 10. Colgrave ML, Goswami H, Byrne K et al
4. Fasano A, Berti I, Gerarduzzi T et al (2003) (2015) Proteomic profiling of 16 cereal grains
Prevalence of celiac disease in at-risk and not- and the application of targeted proteomics to
at-risk groups in the United States: a large detect wheat contamination. J Proteome Res
multicenter study. Arch Intern Med 163 14(6):2659–2668
(3):286–292 11. Fiedler KL, McGrath SC, Callahan JH et al
5. Lionetti E, Gatti S, Pulvirenti A et al (2015) (2014) Characterization of grain-specific pep-
Celiac disease from a global perspective. Best tide markers for the detection of gluten by mass
Pract Res Clin Gastroenterol 29(3):365–379 spectrometry. J Agric Food Chem 62
6. Catassi C, Bai JC, Bonaz B et al (2013) (25):5835–5844
Non-celiac gluten sensitivity: the new frontier 12. Gomaa A, Boye J (2015) Simultaneous detec-
of gluten related disorders. Nutrients 5 tion of multi-allergens in an incurred food
(10):3839–3853 matrix using ELISA, multiplex flow cytometry
7. Koerner TB, Abbott M, Godefroy SB et al and liquid chromatography mass spectrometry
(2013) Validation procedures for quantitative (LC-MS). Food Chem 175:585–592
gluten ELISA methods: AOAC allergen com- 13. Sealey-Voyksner JA, Khosla C, Voyksner RD
munity guidance and best practices. J AOAC et al (2010) Novel aspects of quantitation of
Int 96(5):1033–1040 immunogenic wheat gluten peptides by liquid
8. Thompson T, Mendez E (2008) Commercial chromatography-mass spectrometry/mass
assays to assess gluten content of gluten-free spectrometry. J Chromatogr A 1217
foods: why they are not created equal. J Am (25):4167–4183
Diet Assoc 108(10):1682–1687
Chapter 23
Abstract
As one of the most important posttranslational modifications, glycosylation plays critical roles in protein
folding, trafficking, cell differentiation, immune recognition, etc. The alteration of glycosylation is closely
associated with the pathological processes during and after caner development, and thus holds great value in
cancer detection. In this chapter, we describe a protocol on the glycosylation profiling of tumor marker in
plasma using bead-based immunoassay with CA125 as a model, including bead coupling, coupling control,
glycosylation assay, as well as the plasma screening for breast cancer patients. This protocol can be used to
profile the glycosylation of protein markers in clinical plasma or serum samples for different human cancers.
Key words Posttranslational modification, Glycosylation, Tumor marker, Plasma, Lectin, Bead-based
immunoassay
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_23, © Springer Science+Business Media, LLC, part of Springer Nature 2019
413
414 Hongye Wang et al.
2 Materials
2.1 Reagents 1. Beads: Luminex MegPlex beads (Luminex Inc., Austin, TX,
USA).
2. Activation buffer: 0.1 M NaH2PO4 (Sigma-Aldrich, St. Louis,
MO, USA), pH 6.2.
3. EDC solution: 50 mg/mL EDC (1-ethyl-3-[3-dimethylami-
nopropyl] carbodiimide hydrochloride, Thermo Fisher Scien-
tific, IL, USA).
4. Sulfo-NHS solution: 50 mg/mL sulfo-NHS (Thermo Fisher
Scientific, IL, USA).
5. Coupling buffer: 50 mM MES, pH 5.0 (Sigma-Aldrich,
St. Louis, MO, USA).
6. Assay/Washing buffer: 0.05% (v/v) Tween 20 in 0.01 M PBS.
7. Blocking buffer: PBS-TBN, 1% BSA in washing buffer.
8. Lectin: Biotinylated Aleuria Aurantia Lectin (AAL), Biotiny-
lated Phaseolus vulgaris Erythroagglutinin (PHA-E),
Glycosylation Profiling of Tumor Marker in Plasma Using Bead-Based Immunoassay 415
y
od
ra d
ib
ke te
nt
Quantification
ar la
Add Cy3 labeled
m tiny
Protein
streptavidin
or io
m b
tu d
ti- Ad
An
Add Cy3 labeled
1 streptavidin
d c tin
Ad d-le
l a te
Beads
t iny
coupling Plasma bio
Detections of Protein
bio
tiny Add
Glycosylation
Magnetic Anti-tumor late
beads marker d-le Add Cy3 labeled
ctin
2 streptavidin
Ad
capture
db
antibody
iot
iny
lat
ed
-le
Add Cy3 labeled
cti
n3
streptavidin
3 Methods
3.1 Beads Coupling 1. Resuspend the stock beads by vortex for 30 s and followed with
sonication for 20 s (see Note 1).
2. Take 100 μL of 1.25 107 beads into a microcentrifuge tube.
Put the tube into a magnetic separator and let it stay for 1 min
(see Note 2).
3. Remove the supernatant and resuspend the beads in 100 μL
dH2O by vortex for 30 s.
416 Hongye Wang et al.
4. Keep the tube in a magnetic separator for 1 min and remove the
supernatant without disturbing the beads.
5. Resuspend the beads in 80 μL activation buffer by vortex for
30 s.
6. Prepare 50 mg/mL Sulfo-NHS and EDC diluted in dH2O.
7. Add 10 μL 50 mg/mL Sulfo-NHS to the beads and mix gently
by vortex.
8. Add 10 μL 50 mg/mL EDC to the beads and mix gently by
vortex.
9. Incubate for 20 min at room temperature with gentle mixing.
10. Put the tube into a magnetic separator for 1 min and remove
the supernatant.
11. Wash the beads with 250 μL coupling buffer twice.
12. Resuspend the beads in 100 μL washing buffer and add 5 μg
anti-CA125 antibody. Incubate for 2 h at room temperature
with gentle mixing (see Note 3).
13. Wash the coupled beads with 500 μL blocking buffer, then
incubate for 30 min at room temperature.
14. Wash the blocked beads with 500 μL washing buffer and
resuspend the beads in 1000 μL washing buffer.
15. Count the number of beads using BD FACSVerse (see Note 4).
3.4 Glycosylation 1. The plasma samples are thawed at 4 C and centrifuged for
Profiling of CA125 in 10 min at 14,000 g (see Note 6).
Plasma for Breast 2. Dilute the coupled beads to a final concentration of 50 beads/μ
Cancer Patients L with assay buffer and add 50 μL to a 96-well plate
(pre-blocked with PBS-TBN buffer) in duplicate.
3. Add 50 μL plasma sample and mix with Eppendorf MixMate at
390 g. Incubate for 2 h at room temperature.
4. Wash the beads with 150 μL washing buffer three times.
5. Add 100 μL biotinylated-lectin(2 μg/ml) and incubate for 1 h
at room temperature.
6. Wash the beads with 150 μL washing buffer three times.
7. Add 100 μL Cy3-Streptavidin (4 μg/mL) to each well and
incubate at room temperature. For 1 h on an Eppendorf Mix-
Mate at 390 g.
8. Wash the beads with 150 μL washing buffer twice and resus-
pend the beads in 150 μL washing buffer.
9. Submit the resulting beads to a Luminex200 instrument for
the fluorescent measurement (see Note 7). The representative
results of plasma screening are shown in Fig. 4, in which
differential profiling of CA125 glycosylation can be observed
with the lectins of AAL, PHA-E, and VVA.
4 Notes
Fig. 4 Glycosylated profiling of CA125 in the plasma of breast cancer patients using biotinylated lectins. All
plasma samples were collected with the written informed consent under the approval of intuitional review
board (IRB) from Beijing Proteome Research Center
Acknowledgments
References
1. Chen W et al (2016) Cancer statistics in China, multi-society task force on colorectal cancer,
2015. CA Cancer J Clin 66:115–132 and the American College of Radiology. CA
2. Levin B et al (2008) Screening and surveillance Cancer J Clin 58:130–160
for the early detection of colorectal cancer and 3. Cuzick J et al (2014) Prevention and early
adenomatous polyps, 2008: a joint guideline detection of prostate cancer. Lancet Oncol
from the American Cancer Society, the US 15:e484–e492
420 Hongye Wang et al.
4. Wang D, Yang L, Zhang P, et al (2017) AAgA- 10. Hanash SM, Pitteri SJ, Faca VM (2008)
tlas 1.0: a human autoantigen database. Mining the plasma proteome for cancer bio-
Nucleic acids research 45 (D1): D769–D776 markers. Nature 452:571–579
5. Hanash SM, Baik CS, Kallioniemi O (2011) 11. Wu L, Qu X (2015) Cancer biomarker detec-
Emerging molecular biomarkers—blood- tion: recent achievements and challenges.
based strategies to detect and monitor cancer. Chem Soc Rev 44:2963–2997
Nat Rev Clin Oncol 8:142–150 12. Syed P et al (2016) Role of lectin microarrays in
6. Yu X, Schneiderhan-Marra N, Joos TO (2010) cancer diagnosis. Proteomics 16:1257–1265
Protein microarrays for personalized medicine. 13. Chen S et al (2007) Multiplexed analysis of
Clin Chem 56:376–387 glycan variation on native proteins captured by
7. Wang JR et al (2017) A method to identify antibody microarrays. Nat Methods 4:437–444
trace sulfated IgG N-glycans as biomarkers for 14. Li D, Chiu H, Chen J, Zhang H, Chan DW
rheumatoid arthritis. Nat Commun 8:631 (2013) Integrated analyses of proteins and
8. Li D, Mallory T, Satomura S (2001) AFP-L3: a their glycans in a magnetic bead-based multi-
new generation of tumor marker for hepatocel- plex assay format. Clin Chem 59:315–324
lular carcinoma. Clin Chim Acta 313:15–19 15. Yu X, Petritis B, Duan H, Xu D, LaBaer J
9. Kirwan A, Utratna M, O’Dwyer ME, Joshi L, (2018) Advances in cell-free protein array
Kilcoyne M (2015) Glycosylation-based serum methods. Expert Rev Proteomics 15:1–11
biomarkers for cancer diagnostics and prognos-
tics. Biomed Res Int 2015:490531
Chapter 24
Abstract
N-Glycans are posttranslational modifications of proteins attached to the amide side chains of asparagine
residues, with possible heterogeneity due to different structures being possible at the same glycosylation
site. In contrast to the mammalian systems, invertebrate N-glycosylation presents a challenge in analysis as
there exist unfamiliar epitopes and a high degree of structural and isomeric variation between different
species. A simple analytical approach to analyze N-glycans on specific glycoproteins is presented, which
involves a combination of tryptic peptide mass spectrometry and “off-line” RP-HPLC MALDI-TOF
MS/MS complemented by blotting to recognize specific epitopes. An additional N-glycan enrichment
and labeling step can facilitate the analysis of single structures and even provide isomeric separation of
N-glycans from specific proteins.
Abbreviations
DTT Dithiothreitol
HRP Horseradish peroxidase
MALDI-TOF MS matrix-assisted laser-desorption/ionization time-of-flight mass spectrometry
NPGC Nonporous graphitized carbon
PC Phosphorylcholine
PE Phosphoethanolamine
RP-HPLC Reversed phase high pressure liquid chromatography
SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel electrophoresis
Full data on the N-glycans of honeybee royal jelly will appear in a forthcoming paper: Hykollari et al. 2018.
Isomeric separation and recognition of anionic and zwitterionic N-glycans from royal jelly glycoproteins.
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_24, © Springer Science+Business Media, LLC, part of Springer Nature 2019
421
422 Alba Hykollari et al.
1 Introduction
2 Materials
2.2.2 SDS-PAGE and 1. 12% SDS-PAGE gel (using 40% acrylamide stock from
Western Blotting Bio-Rad, diluted with either stacking gel buffer with 0.5 M
Tris/HCl pH 6.8 or separation gel buffer with 1.5 M Tris/
HCl pH 8.8).
2. SDS-PAGE running buffer (25 mM Tris, 192 mM glycine,
0.1% SDS; components from either VWR or Roth).
424 Alba Hykollari et al.
2.2.3 Tryptic Peptide 1. Colloidal Coomassie Blue staining solution: 0.02% (w/v) Coo-
Mapping massie Brilliant Blue G-250 (Bio-Rad), 5% aluminum sulfate-
(14-18)-hydrate [e.g., Al2(SO4)3.16H2O (Roth)], ethanol
96% (VWR), phosphoric acid 85% (Roth). Weigh in 100 g of
aluminum sulfate and dissolve it in 1500 mL of water; add
200 mL ethanol and mix well; add 0.4 g of Coomassie Brilliant
Blue G-250 and mix well for at least 30 min; add slowly 47 mL
of phosphoric acid and mix well; make up to 2000 mL with
water (see Note 3).
2. Acetonitrile LC-MS grade (VWR), ammonium bicarbonate
(Roth), water HPLC super gradient grade (VWR), iodoaceta-
mide (Sigma), dithiothreitol (Roth), trifluoroacetic acid
(Fluka).
3. Sequencing grade modified trypsin dissolved to 0.1 mg/mL in
50 mM acetic acid (Promega); typically cleaves after Arg and
Lys residues.
Table 1
List of selected antibodies, lectins, and pentraxins for N-glycan epitope screening (see Note 4)
Fig. 1 A potential glycome and glycoproteomic workflow. Starting from biological material, proteins can be
separated by SDS-PAGE prior to Western blotting or peptide map fingerprinting. The peptides and glycopep-
tides are analyzed directly by mass spectrometry; the glycans are released by an N-glycanase such as PNGase
Ar and purified by two rounds of solid-phase extraction prior to mass spectrometry and/or HPLC. Glycans
(examples from honeybee royal jelly) are depicted according to the Symbol Nomenclature for
Glycans, whereby circles, squares, triangles and diamonds respectively represent hexose (here Man or
Gal), N-acetylhexosamine (GalNAc or GlcNAc), deoxyhexose (Fuc) or hexuronic acids (GlcA); S, sulfate; PE,
phosphoethanolamine
428 Alba Hykollari et al.
Fig. 2 Example of a glycoproteomic study of honeybee glycoprotein MRJP1 found in royal jelly. The flow chart
scheme shows the glycoproteomic workflow for the biological sample; the letters refer to steps exemplified by
the following data. (a) N-Glycan epitope detection by Western blotting of the royal jelly glycoproteins after
incubation with anti-HRP antibodies. (b) Tryptic peptide mapping of one of the major royal jelly glycoproteins
as measured with MALDI TOF MS in positive ion mode. (c) Free N-glycans from MRJP1 measured with MALDI-
TOF MS after deglycosylation with PNGase Ar with the [M + Na]+ ions annotated with abbreviations of the form
HxNy, where H is hexose and N is N-acetylhexosamine. (d) RP-HPLC chromatogram of reductively aminated
N-glycans from royal jelly glycoprotein MRJP1 with fractions annotated with the detected glycan m/z values
and calibrated in terms of glucose units. (e) MALDI-TOF MS/MS data of fractionated MRJP1 glycans, whereby
[M + H]+ ions were fragmented and key B and Y ions are annotated. Note that the precursor ions for the H6N4
and H4N5PE structures cannot be separated (Δm/z ¼ 2), but the zoom shows that Y ions derived from both
are present; the structure of the PA (pyridylamino) reducing-terminal label as well as the symbol nomenclature
are shown beneath the flowchart
3.1.2 SDS-PAGE and For initial screening of the N-glycan epitopes, approx. 2 μg of
Western Blotting proteins are subject to SDS-PAGE under reducing conditions,
followed by protein transfer to a nitrocellulose membrane (Western
blotting).
1. Check the quality of the successful transfer by incubating the
membrane with Ponceau S staining solution for 1 min. After
Invertebrate Glycoprotein Analysis 429
de-staining with water (protein bands will stain red), block the
membrane with Tris buffered saline containing 0.05% Tween
and 0.5% BSA for 1 h at room temperature under smooth
shaking.
2. Wash the membrane three times using Tris buffered saline with
0.05% Tween (washing buffer).
3. Incubate with biotinylated lectins, pentraxins or primary anti-
bodies in blocking/dilution buffer for 60 min (see Table 1 and
Note 4).
4. Wash the membrane again thrice as above and incubate with
the relevant peroxidase or alkaline phosphatase conjugated
secondary antibodies in blocking/dilution buffer for 60 min.
5. Again wash the membrane three times as above.
6. Develop the Western blots respectively for peroxidase or phos-
phatase conjugates with either SigmaFAST 3,30 -diaminobenzi-
dine tetrahydrochloride or SigmaFAST BCIP/NBT (dissolve
tablets first in water). Chemiluminescence or other detection
methods can also be used.
3.2 Tryptic Peptide 1. For the peptide mass fingerprinting identification of proteins
Mapping (See Note 5) with MALDI-TOF MS, apply 10 μg of protein to the
SDS-PAGE and stain with Coomassie Blue.
2. After de-staining the gel with water, excise the protein bands in
small pieces on glass plates using a clean scalpel.
3. Wash/destain the gel pieces twice with 50% acetonitrile in
water and successively once with 1:1 0.1 M ammonium bicar-
bonate/acetonitrile and 100% acetonitrile only, prior to drying
in a Speedvac.
4. In addition, reduce the gel pieces with 10 mM DTT for 1 h at
56 C and alkylate for 45 min at room temperature with
iodoacetamide (55 mM in 0.1 M ammonium bicarbonate) in
the dark. Subject the gel pieces to a second round of serial
washing (twice 50% acetonitrile, 1:1 0.1 M ammonium bicar-
bonate/acetonitrile and 100% acetonitrile only) and drying in a
Speedvac.
5. For proteolytic digestion, cover the gel pieces with a 1:2 mix-
ture of 0.1 M ammonium bicarbonate /trypsin (100 ng/μL)
and incubate overnight at 37 C.
6. Extract the peptides at room temperature three times using
acetonitrile/water/trifluoroacetic acid in a ratio of 660:330:1
(v/v/v). Dry the enriched glycopeptides using a vacuum cen-
trifuge and redissolve them in 5 μL water, prior to spotting on a
target plate for MALDI-TOF MS analysis.
7. Spot 0.5 μL of the peptides before applying the matrix (either
α-cyanocinnamic acid (ACH) or 6-aza thiothymine (ATT).
430 Alba Hykollari et al.
4 Notes.
Acknowledgments
This work was supported by the Austrian Fonds zur Förderung der
wissenschaftlichen Forschung (FWF; grants P26662, P25058, and
P23922 to A.H., K.P, and I.B.H.W.).
Invertebrate Glycoprotein Analysis 435
References
1. Spiro RG (2002) Protein glycosylation: nature, antibodies using remodeled glycoproteins.
distribution, enzymatic formation, and disease Anal Biochem 386:133–146
implications of glycopeptide bonds. Glycobiol- 14. Mikolajek H, Kolstoe SE, Pye VE, Mangione P,
ogy 12:43R–56R Pepys MB, Wood SP (2011) Structural basis of
2. Aebi M (2013) N-linked protein glycosylation ligand specificity in the human pentraxins,
in the ER. Biochim Biophys Acta C-reactive protein and serum amyloid P com-
1833:2430–2437 ponent. J Mol Recognit 24:371–377
3. Schiller B, Hykollari A, Yan S, Paschinger K, 15. Hykollari A, Paschinger K, Eckmair B, Wilson
Wilson IBH (2012) Complicated N-linked gly- IBH (2017) Analysis of invertebrate and pro-
cans in simple organisms. Biol Chem Hoppe tist N-glycans. Methods Mol Biol
Seyler 393:661–673 1503:167–184
4. Eckmair B, Jin C, Abed-Navandi D, Paschinger 16. Purohit S, Li T, Guan W, Song X, Song J,
K (2016) Multi-step fractionation and mass Tian Y, Li L, Sharma A, Dun B, Mysona D,
spectrometry reveals zwitterionic and anionic Ghamande S, Rungruang B, Cummings RD,
modifications of the N- and O-glycans of a Wang PG, She JX (2018) Multiplex glycan
marine snail. Mol Cell Proteomics 15:573–597 bead array for high throughput and high con-
5. Stanton R, Hykollari A, Eckmair B, Malzl D, tent analyses of glycan binding proteins. Nat
Dragosits M, Palmberger D, Wang P, Wilson Commun 9:258
IBH, Paschinger K (2017) The underestimated 17. Paschinger K, Rendić D, Wilson IBH (2009)
N-glycomes of lepidopteran species. Biochim Revealing the anti-HRP epitope in Drosoph-
Biophys Acta 1861:699–714 ila and Caenorhabditis. Glycoconj J 26:
6. Yan S, Vanbeselaere J, Jin C, Blaukopf M, 385–395
Wols F, Wilson IBH, Paschinger K (2018) 18. Tsai PL, Chen SF (2017) A brief review of
Core richness of N-glycans of Caenorhabditis bioinformatics tools for glycosylation analysis
elegans: a case study on chemical and enzy- by mass spectrometry. Mass Spectrom (Tokyo)
matic release. Anal Chem 90:928–935 6:S0064
7. Tretter V, Altmann F, Kubelka V, M€arz L, 19. York WS, Agravat S, Aoki-Kinoshita KF,
Becker WM (1993) Fucose α1,3-linked to the McBride R, Campbell MP, Costello CE,
core region of glycoprotein N-glycans creates Dell A, Feizi T, Haslam SM, Karlsson N,
an important epitope for IgE from honeybee Khoo KH, Kolarich D, Liu Y, Novotny M,
venom allergic individuals. Int Arch Allergy Packer NH, Paulson JC, Rapp E,
Immunol 102:259–266 Ranzinger R, Rudd PM, Smith DF, Struwe
8. Prasanphanich NS, Mickum ML, Heimburg- WB, Tiemeyer M, Wells L, Zaia J, Kettner C
Molinaro J, Cummings RD (2013) Glycocon- (2014) MIRAGE: the minimum information
jugates in host-helminth interactions. Front required for a glycomics experiment. Glyco-
Immunol 4:240 biology 24:402–406
9. Geisler C, Mabashi-Asazuma H, Jarvis DL 20. Varki A, Cummings RD, Aebi M, Packer NH,
(2015) An overview and history of glyco- Seeberger PH, Esko JD, Stanley P, Hart G,
engineering in insect expression systems. Darvill A, Kinoshita T, Prestegard JJ, Schnaar
Methods Mol Biol 1321:131–152 RL, Freeze HH, Marth JD, Bertozzi CR,
10. Hykollari A, Malzl D, Yan S, Wilson IBH, Etzler ME, Frank M, Vliegenthart JF,
Paschinger K (2017) Hydrophilic interaction Lutteke T, Perez S, Bolton E, Rudd P,
anion exchange for separation of multiply mod- Paulson J, Kanehisa M, Toukach P, Aoki-
ified neutral and anionic Dictyostelium Kinoshita KF, Dell A, Narimatsu H, York W,
N-glycans. Electrophoresis 38:2175–2183 Taniguchi N, Kornfeld S (2015) Symbol
nomenclature for graphical representations of
11. Paschinger K, Wilson IBH (2016) Analysis of glycans. Glycobiology 25:1323–1324
zwitterionic and anionic N-linked glycans from
invertebrates and protists by mass spectrome- 21. Aoki K, Perlman M, Lim JM, Cantu R, Wells L,
try. Glycoconj J 33:273–283 Tiemeyer M (2007) Dynamic developmental
elaboration of N-linked glycan complexity in
12. Paschinger K, Gonzalez-Sapienza GG, Wilson the Drosophila melanogaster embryo. J Biol
IBH (2012) Mass spectrometric analysis of the Chem 282:9127–9142
immunodominant glycan epitope of Echino-
coccus granulosus antigen Ag5. Int J Parasitol 22. Miyata S, Sato C, Kumita H, Toriyama M, Vac-
42:279–285 quier VD, Kitajima K (2006) Flagellasialin: a
novel sulfated α2,9-linked polysialic acid glyco-
13. Iskratsch T, Braun A, Paschinger K, Wilson protein of sea urchin sperm flagella. Glycobiol-
IBH (2009) Specificity analysis of lectins and ogy 16:1229–1241
Chapter 25
Abstract
Proteomics studies that characterize hundreds or thousands of proteins in parallel can play an important
part in the identification of moonlighting proteins, proteins that perform two or more distinct and
physiologically relevant biochemical or biophysical functions. Functional assays, including ligand-binding
assays, can find a surprising second function for a protein that was previously identified as performing a
different function, for example, a DNA-binding ability for an enzyme in amino acid metabolism. The results
of large-scale assays of protein–protein interactions, gene knockouts, or subcellular protein localizations, or
bioinformatics analysis of amino acid sequences and three-dimensional structures, can also be used to
predict that a protein has additional functions, but in these cases it is important to use biochemical and
biophysical methods to confirm the protein can perform each function.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_25, © Springer Science+Business Media, LLC, part of Springer Nature 2019
437
438 Constance Jeffery
2 Methods
2.1 Experimental The proteomics methods that have been the most useful in finding
Methods proteins with multiple functions include those that test binding to a
specific molecule, protein–protein interactions, the results of gene
knockout experiments, and cellular localization.
2.1.1 Binding Studies Proteomics studies that involve screening hundreds or thousands of
proteins to find those that bind to DNA, extracellular matrix, or
other macromolecules have identified several dozen proteins that
were already known to have a different function. This is not too
surprising because dozens of the known moonlighting proteins
have at least one function that involves binding to another mole-
cule—as a cell surface receptor for a soluble ligand or the extracel-
lular matrix, as a secreted ligand binding to a receptor on another
cell, or as a DNA- or RNA-binding protein.
The use of microarrays of proteins, DNA oligonucleotides, or
RNA oligonucleotides enables screening of vast numbers of pro-
teins to find those that bind to a chosen macromolecule. Hall and
coworkers screened a microarray of yeast proteins for binding to
DNA oligonucleotides and identified mitochondrial Arg5,6
(N-acetylglutamate kinase/N-acetylglutamyl-phosphate reduc-
tase), an enzyme in the arginine biosynthetic pathway, as having
DNA-binding activity. Complementary chromatin immunoprecip-
itation experiments and gene deletion experiments confirmed that
Moonlighting Proteins in Proteomics 439
2.1.3 Gene Knockouts When a single protein participates in several cellular processes,
deletion of the gene encoding the protein can result in phenotypes
that are more complex than can be explained by the loss of a single
function. Several labs made use of yeast genetics to look for
enzymes for which replacement of the wild-type enzyme with a
440 Constance Jeffery
3 Conclusions
References
1. Jeffery CJ (1999) Moonlighting proteins. Microbiol 8:1150. https://doi.org/10.3389/
Trends Biochem Sci 24(1):8–11. PMID: fmicb.2017.01150
10087914 13. Pancholi V, Fischetti VA (1992) A major sur-
2. Piatigorsky J, Wistow GJ (1989) Enzyme/ face protein on group a streptococci is a glycer-
crystallins: gene sharing as an evolutionary aldehyde-3-phosphate-dehydrogenase with
strategy. Cell 57:197–199 multiple binding activity. J Exp Med
3. Wistow GJ, Kim H (1991) Lens protein 176:415–426
expression in mammals: taxon specificity and 14. Olaya-Abril A, Jiménez-Munguı́a I, Gómez-
the recruitment of crystallins. J Mol Evol Gascón L, Rodrı́guez-Ortega MJ (2014) Sur-
32:262–269 fomics: shaving live organisms for a fast proteo-
4. Huang QL, Russell P, Stone SH, Zigler JS Jr mic identification of surface proteins. J
(1987) Zeta-crystallin, a novel lens protein Proteome 97:164–176. https://doi.org/10.
from the Guinea pig. Curr Eye Res 1016/j.jprot.2013.03.035
6:725–732. PMID: 3595182 15. Wang W, Jeffery CJ (2016) An analysis of sur-
5. Mani M, Chen C, Amblee V, Liu H, Mathur T, face proteomics results reveals novel candidates
Zwicke G, Zabad S, Patel B, Thakkar J, Jeffery for intracellular/surface moonlighting proteins
CJ (2015) MoonProt: a database for proteins in bacteria. Mol BioSyst 12:1420–1431
that are known to moonlight. Nucleic Acids 16. Thul PJ, Åkesson L, Wiking M, Mahdessian D,
Res 43:D277–D282 Geladaki A, Ait Blal H, Alm T, Asplund A,
6. Hall DA, Zhu H, Zhu X, Royce T, Gerstein M, Björk L, Breckels LM, B€ackström A,
Snyder M (2004) Regulation of gene expres- Danielsson F, Fagerberg L, Fall J, Gatto L,
sion by a metabolic enzyme. Science Gnann C, Hober S, Hjelmare M,
306:482–484 Johansson F, Lee S, Lindskog C, Mulder J,
7. Crowe JD, Sievwright IK, Auld GC, Moore Mulvey CM, Nilsson P, Oksvold P,
NR, Gow NA, Booth NA (2003) Candida Rockberg J, Schutten R, Schwenk JM, Siverts-
albicans binds human plasminogen: identifica- son Å, Sjöstedt E, Skogs M, Stadler C, Sullivan
tion of eight plasminogen-binding proteins. DP, Tegel H, Winsnes C, Zhang C,
Mol Microbiol 47:1637–1651. PMID: Zwahlen M, Mardinoglu A, Pontén F, von
12622818 Feilitzen K, Lilley KS, Uhlén M, Lundberg E
(2017) A subcellular map of the human prote-
8. Candela M, Bergmann S, Vici M, Vitali B, ome. Science 356:eaal3321. https://doi.org/
Turroni S, Eikmanns BJ, Hammerschmidt S, 10.1126/science.aal3321
Brigidi P (2007) Binding of human plasmino-
gen to Bifidobacterium. J Bacteriol 17. Consortium GO (2015) Gene ontology con-
189:5929–5936. https://doi.org/10.1128/ sortium: going forward. Nucleic Acids Res 43:
JB.00159-07 D1049–D1056
9. Gómez A, Hernández S, Amela I, Piñol J, 18. UniProt Consortium (2015) UniProt: a hub
Cedano J, Querol E (2011) Do protein- for protein information. Nucleic Acids Res 43:
protein interaction databases identify moon- D204–D212
lighting proteins? Mol BioSyst 7:2379–2382. 19. Khan IK, Bhuiyan M, Kihara D (2017)
https://doi.org/10.1039/c1mb05180f DextMP: deep dive into text for predicting
10. Chapple CE, Robisson B, Spinelli L, Guien C, moonlighting proteins. Bioinformatics 33:
Becker E, Brun C (2015) Extreme multifunc- i83–i91. https://doi.org/10.1093/bioinfor
tional proteins identified from a human protein matics/btx231
interaction network. Nat Commun 6:7412. 20. Pritykin Y, Ghersi D, Singh M (2015)
https://doi.org/10.1038/ncomms8412 Genome-wide detection and analysis of multi-
11. Espinosa-Cantú A, Ascencio D, Herrera- functional genes. PLoS Comput Biol 11:
Basurto S, Xu J, Roguev A, Krogan NJ, e1004467. https://doi.org/10.1371/journal.
DeLuna A (2018) Protein moonlighting pcbi.1004467
revealed by noncatalytic phenotypes of yeast 21. Zhao B, Lei L, Vassylyev DG, Lin X, Cane DE,
enzymes. Genetics 208:419–431. https://doi. Kelly SL, Yuan H, Lamb DC, Waterman MR
org/10.1534/genetics.117.300377 (2009) Crystal structure of albaflavenone
12. Escalera-Fanjul X, Campero-Basaldua C, monooxygenase containing a moonlighting
Colón M, González J, Márquez D, González terpene synthase active site. J Biol Chem
A (2017) Evolutionary diversification of ala- 284:36711–36719. https://doi.org/10.
nine transaminases in yeast: catabolic speciali- 1074/jbc.M109.064683
zation and biosynthetic redundancy. Front 22. Khan I, Chitale M, Rayon C, Kihara D (2012)
Evaluation of function predictions by PFP,
Moonlighting Proteins in Proteomics 443
ESG, and PSI-BLAST for moonlighting pro- 24. Khan IK, Kihara D (2016) Genome-scale pre-
teins. BMC Proc 6(Suppl 7):S5. https://doi. diction of moonlighting proteins using diverse
org/10.1186/1753-6561-6-S7-S5 protein association information. Bioinformat-
23. Hernández S, Franco L, Calvo A, Ferragut G, ics 32:2281–2288. https://doi.org/10.1093/
Hermoso A, Amela I, Gómez A, Querol E, bioinformatics/btw166
Cedano J (2015) Bioinformatics and moon- 25. Khan I, McGraw J, Kihara D (2017) MPFit:
lighting proteins. Front Bioeng Biotechnol computational tool for predicting moonlight-
3:90. https://doi.org/10.3389/fbioe.2015. ing proteins. Methods Mol Biol 1611:45–57.
00090 https://doi.org/10.1007/978-1-4939-7015-
5_5
Chapter 26
Abstract
A high-resolution two-dimensional (2-D) proteomic fractionation technique for the systematic purification
and subsequent mass spectrometry-based identification of endogenous protein macromolecular complexes
is described. The method hyphenates preparative isoelectric focusing (IEF) with mixed-bed ion exchange
chromatography (IEX) to efficiently separate cell- or tissue- derived soluble protein mixtures, allowing for
more effective and less biased physiochemical characterization of stable multiprotein assemblies. After
comprehensive 2D fractionation of cell-free lysates, each fraction is subjected to quantitative tandem
mass spectrometry (MS/MS) and subsequent computational analysis to map high-confidence protein–-
protein interactions (PPIs). Herein, the experimental component (workflow protocols) for this global
“interactome” network mapping platform is described.
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_26, © Springer Science+Business Media, LLC, part of Springer Nature 2019
445
446 Reza Pourhaghighi and Andrew Emili
2 Materials
3 Methods
3.1 Soluble Protein 1. Pellet E. coli cells by centrifugation at 5000 g for 10 min (see
Extraction Note 6).
2. Add 4 mL of modified B-PER protein extraction buffer per one
gram of cell pellet and mix gently until the pellet is fully
dissolved.
3. Lightly mix the lysate for 30 min at 4 C using a rotatory
shaker.
4. Centrifuge lysate at 15,000 g for 10 min at 4 C to separate
soluble proteins from insolubles and the cell debris.
5. Remove and filter the supernatant with 0.45 μm centrifugal
filters according to manufacturer’s recommended time and
speed.
6. Use Bradford protein assay to measure the protein concentra-
tion in lysate obtained.
that the needles could pass sealing tape and penetrate the
harvesting ports.
20. After the fractions are aspirated into the harvesting tray, turn
off the vacuum source and remove the harvesting tray.
21. Transfer the fractions to 1.5 mL tubes and store them at 4 C.
3.3 Ion Exchange NOTE: No sample clean-up and/or buffer exchange step is
Chromatography required prior to IEX-HPLC separation of IEF fractions collected.
(IEX-HPLC) Depending of the pH of the IEF fractions, Tris–HCl pH 8 (for
acidic IEF fractions with the pH up to 7) or MES pH 6 buffer (for
basic fractions with pHs above 7) are used for IEX separations of
IEF focused protein complexes. During IEX separation, proteins
can be eluted from the IEX-HPLC column with a salt (NaCl)
gradient and recovered in biologically active forms.
1. Equilibrate the IEX-HPLC column by running two blank
gradients using the buffer systems preferred before running
the protein sample. Likewise, re-equilibrate the column after
each gradient with at least 30 min running 100% buffer-A.
2. Set the injection volume of the HPLC method to inject the
entire sample collected from IEF chamber into the IEX column
(normally between 200 and 250 μL).
3. The recommended flowrate for a 2.1-mm i.d. column as
described here is 0.2 mL/min.
4. A typical gradient as below could be employed for IEX-HPLC
separation:
(a) 100% A from 0 to 3 min, followed by a shallow gradient to
10% B from 3 to 45 min, a linear gradient to 35% B from
45 to 65 min and then a gradient to 100% B from 65 to
80 min followed by an isocratic hold at 100% B until
90 min.
5. Monitor the protein elution by UV absorption signal at
280 nm.
6. Set the method to collect the fractions with 2-min intervals
during IEX separation run. Depending on the chromatogram,
the collected fractions could be further merged to reduce the
total number of fractions for subsequent analysis.
The HPLC-IEX parameters described are summarized in
Table 1. Figure 2 shows a representative IEX-HPLC chromato-
gram recorded after running combined IEF fractions 1 and
2 (pH 5–5.6) using a Tris pH 8 buffer system and using the
parameters described in Table 1.
Two-Dimensional Biochemical Purification for Global Proteomic Analysis. . . 451
Table 1
IEX-HPLC parameters
800
600
400
200
0
10
20
30
40
50
60
70
80
90
Retention Time(min)
Fig. 2 Representative chromatogram of IEX-HPLC fractionated E. coli protein extract, generated after
combining IEF pre-fractions 1 and 2 (pH 5–5.6). Detailed experimental parameters are listed in Table 1
3.6 LC-MS/MS 1. Set the method to inject about 1–2 μg for digested protein
samples into the nanoLC column for MS analysis.
2. A 60-min LC gradient as outlined below is generally appropri-
ate for nLC-MS/MS analysis of the fractions obtained: A linear
gradient from 5% to 30% B from 0 to 46 min and then a
gradient to 100% B from 46 to 50 min followed by an isocratic
hold at 100% B until 60 min.
3. The parameters for MS analysis of peptide fractions depend on
the type and performance specifications of MS instrument
used. As an illustrative example, the recommended MS para-
meters for a 60-min method on an Orbitrap Q-Exactive HF
instrument are listed in Table 2.
3.7 Computational Search all MS/MS spectra against an appropriate fasta file using a
Proteomics Analysis search engine (e.g., MaxQuant). The calculated related intensity of
proteins in each IEX fraction can then be used to calculate similarity
profiles, to predict protein associations and co-complex member-
ships, using computational algorithms (e.g., machine learning clas-
sifiers) and statistical filtering to identify high-confidence physical
interactions among the co-eluting proteins as described in [6].
Two-Dimensional Biochemical Purification for Global Proteomic Analysis. . . 453
Table 2
nLC-MS/MS parameters
4 Notes
References
1. Krogan NJ, Cagney G, Yu H et al (2006) Global 4. Kristensen AR, Gsponer J, Foster LJ (2012) A
landscape of protein complexes in the yeast Sac- high-throughput approach for measuring tem-
charomyces cerevisiae. Nature 440:637–643. poral changes in the interactome. Nat Methods
https://doi.org/10.1038/nature04670 9:907–909. https://doi.org/10.1038/nmeth.
2. Tarassov K, Messier V, Landry CR et al (2008) 2131
An in vivo map of the yeast protein interactome. 5. Havugimana PC, Hart GT, Nepusz T et al
Science 320:1465–1470. https://doi.org/10. (2012) A census of human soluble protein com-
1126/science.1153878 plexes. Cell 150:1068–1081. https://doi.org/
3. Uetz P, Giot L, Cagney G et al (2000) A com- 10.1016/j.cell.2012.08.011
prehensive analysis of protein-protein interac- 6. Zhong Ming Hu L, Goebels F, Wan C, et al
tions in Saccharomyces cerevisiae. Nature EPIC: elution profile-based inference of protein
403:623–627. https://doi.org/10.1038/ complex membership. Nat Methods
35001009
Chapter 27
Abstract
Data-independent acquisition (DIA) mode of mass spectrometry, such as the SWATH-MS technology,
enables accurate and consistent measurement of proteins, which is crucial for comparative proteomics
studies. However, there is lack of free and easy to implement data analysis protocols that can handle the
different data processing steps from raw spectrum files to peptide intensity matrix and its downstream
analysis. Here, we provide a data analysis protocol, named diatools, covering all these steps from spectral
library building to differential expression analysis of DIA proteomics data. The data analysis tools used in
this protocol are open source and the protocol is distributed at Docker Hub as a complete software
environment that supports Linux, Windows, and macOS operating systems.
Key words Proteomics, Mass spectrometry, DDA, DIA, SWATH-MS, Spectral library, Data analysis
1 Introduction
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3_27, © Springer Science+Business Media, LLC, part of Springer Nature 2019
455
456 € et al.
Sami Pietila
2 Materials
3 Methods
This chapter describes the steps of our diatools data analysis proto-
col, including data conversion, peptide identification, quantifica-
tion, and differential expression analysis for the acquired data. A
schematic illustration of the protocol is shown in Fig. 1.
optional
optional
Fig. 1 The diatools data analysis protocol for quantitative data-independent acquisition proteomics data.
Protocol steps are shown with their corresponding section numbers and their input/output files. Optional steps
are marked with dashed line
458 € et al.
Sami Pietila
3.1 Software Install the ProteoWizard software to the Windows machine that is
Installation and Data used to convert the raw data files to an open format. The Proteo-
Folder Structure Wizard software can be downloaded from http://proteowizard.
Preparation sourceforge.net/.
Install Docker on the machine on which the data analysis
protocol will be run. Docker installation package for Linux, Win-
dows, and macOS can be downloaded from www.docker.org. If the
Linux distribution is Ubuntu, RedHat, or CentOS Linux, the
Docker can be installed from their respective software repositories
(see Note 1). For Windows, the latest version 10 is recommended.
On Windows, allow Docker to access the needed drive (for exam-
ple, C:) from the Docker settings.
Once Docker is installed, download the data analysis environ-
ment with the command:
3.2 Conversion of Use the ProteoWizard tool to convert the raw data files to an open
Raw Data Files to Open format.
Format To convert the raw DDA files to mzXML format, open Win-
dows Command Prompt and go to the folder containing the DDA
raw data. Run qtofpeakpicker from ProteoWizard to pick peaks and
to convert the raw files to mzXML format:
3.3 Construction of Create a sequence database in FASTA format that consists of all
the Sequence proteins that may exist in the sample set under analysis. The FASTA
Database FASTA File file is used to construct the spectral library by searching the DDA
files against it. Create a FASTA file that contains the following
protein or peptide sequences:
l Proteins of interest (for example Swiss-Prot Human)
l IRT peptides1 (Biognosys|iRT-Kit_WR_fusion)
l Peptides related to lysis (Uniprot ID: Q7M135)
l Digestion enzyme (typically Trypsin (Uniprot ID: P00761))
l Possible contaminants
Do not generate decoy sequences to the FASTA file manually.
They are automatically generated by the protocol by reversing the
peptide/protein sequences. Copy the FASTA file to the dataset/ref
folder and name it as sequences.fasta.
3.4 Peptide Search The default parameters of the protocol are for the nanoflow HPLC
Parameters system (Easy-nLC1200, Thermo Fisher Scientific) coupled to the
Q Exactive HF mass spectrometer (Thermo Fisher Scientific)
equipped with a nano-electrospray ionization source. The device
and lab protocol specific default settings are listed below:
l Precursor mass tolerance: 10 ppm
l Fragment ion tolerance: 0.02 Da
l Cleavage site: Trypsin_P
l Fixed modification: Carbamidomethyl (C)
l Variable modification: Oxidation (M)
If another type of instrument is used, these settings need to be
customized (see Note 2).
1
https://biognosys.com/media.ashx/irtfusion.fasta
460 € et al.
Sami Pietila
3.5 Running the Data Open terminal prompt and set the working directory to dataset/out
Analysis Protocol folder, where the LOCALPATH refers to the path to the previously
created folder structure:
cd /LOCALPATH/dataset/out
--design-file <designFilename>
Table 1
Example design file
values for each sample. The files can be opened with MS Excel or
with LibreOffice Calc. The output folder contains also files of
intermediate results written by various external tools run by the
protocol as well as a log.txt file which includes details on the run.
The log can be used for troubleshooting if the run fails.
If the optional differential expression analysis is performed,
those results are stored as tab-separated files in the dataset/out
folder with the compared groups as filenames. For each identified
protein, the result files contain the protein name, the value of the
test statistic (t), the number of peptides per protein (n), the signifi-
cance p-value (p), and the estimated false discovery rate (p.fdr). In
addition to performing the differential expression analysis by run-
ning the diatools protocol, it is possible to perform the differential
expression analysis separately using the peptide intensity file pro-
duced by the protocol (see Note 5).
4 Notes
--comet-cfg-template config/comet.params.template
--xtandem-cfg-template config/xtandem_settings.xml
--library-FDR
--feature-alignment-FDR
--library-FDR 0.05
--threads 20
Quantitative DIA Data Analysis Protocol 463
source(“https://bioconductor.org/biocLite.R”)
biocLite("SWATH2stats")
library(data.table)
library(SWATH2stats)
data <- data.frame(fread(file=" peptide-intensity-
matrix.tsv",
sep=’\t’, header=TRUE))
Next, get rid of unneeded columns (line 2), remove the iRT
peptides that are used for retention time normalization (line 3),
filter out rows corresponding to multiple proteins (line 4), and
shorten the protein names (line 5):
source(“https://bioconductor.org/biocLite.R”)
biocLite("PECA")
library(PECA)
comb <- combn(unique(design$Condition),2)
for(i in 1:ncol(comb)) {
group1 <- paste(design$Condition,
design$BioReplicate,sep="_")
[design$Condition==comb[1,i]]
group2 <- paste(design$Condition,
design$BioReplicate,sep="_")
[design$Condition==comb[2,i]]
peca.out <- PECA_df(data, group1, group2,
id="ProteinName", normalize="median",
test="rots", progress=TRUE)
write.table(peca.out,
file=paste("PECA_",comb[1,i],
"-",comb[2,i],".txt",sep=""),
sep="\t", quote=FALSE, row.names=TRUE, col.names=NA)
}
References
1. Aebersold R, Mann M (2003) Mass targeted analysis of data-independent acquisi-
spectrometry-based proteomics. Nature tion MS data. Nat Biotechnol 32:219–223
422:198–207 6. Röst HL, Liu Y, D’Agostino G et al (2016)
2. Gillet LC, Navarro P, Tate S et al (2012) Tar- TRIC: an automated alignment strategy for
geted data extraction of the MS/MS spectra reproducible protein quantification in targeted
generated by data-independent acquisition: a proteomics. Nat Methods 13:777–783
new concept for consistent and accurate prote- 7. Merkel D (2014) Docker: lightweight Linux
ome analysis. Mol Cell Proteomics 11: containers for consistent development and
O111.016717 deployment. Linux J
3. Huang Q, Yang L, Luo J et al (2015) SWATH 8. Chambers MC, Maclean B, Burke R et al
enables precise label-free quantification on pro- (2012) A cross-platform toolkit for mass spec-
teome scale. Proteomics 15:1215–1223 trometry and proteomics. Nat Biotechnol
4. Schubert OT, Gillet LC, Collins BC et al 30:918–920
(2015) Building high-quality assay libraries 9. Sturm M, Bertsch A, Gröpl C et al (2008)
for targeted analysis of SWATH MS data. Nat OpenMS—an open-source software frame-
Protoc 10:426–441 work for mass spectrometry. BMC Bioinfor-
5. Röst HL, Rosenberger G, Navarro P et al matics 9:163
(2014) OpenSWATH enables automated,
Quantitative DIA Data Analysis Protocol 465
10. Deutsch EW, Mendoza L, Shteynberg D et al analysis with bioconductor. Nat Methods
(2010) A guided tour of the trans-proteomic 12:115–121
pipeline. Proteomics 10:1150–1159 13. Elo LL, Filén S, Lahesmaa R et al (2008)
11. Suomi T, Corthals GL, Nevalainen OS et al Reproducibility-optimized test statistic for rank-
(2015) Using peptide-level proteomics data ing genes in microarray studies. IEEE/ACM
for detecting differentially expressed proteins. Trans Comput Biol Bioinform 5:423–431
J Proteome Res 14:4564–4570 14. Suomi T, Elo LL (2017) Enhanced differential
12. Huber W, Carey VJ, Gentleman R et al (2015) expression statistics for data-independent
Orchestrating high-throughput genomic acquisition proteomics. Sci Rep 7:5869
INDEX
Xing Wang and Matthew Kuruc (eds.), Functional Proteomics: Methods and Protocols, Methods in Molecular Biology, vol. 1871,
https://doi.org/10.1007/978-1-4939-8814-3, © Springer Science+Business Media, LLC, part of Springer Nature 2019
467
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
468 Index
C CAZymes in microbial secretomes
biomass conversion ................................................. 160
C1 inhibitor........................................................ 42, 44, 47 biotechnological applications ................................. 159
C18 peptide fractionation .............................................. 94 culture plates
C3/C5-convertase ........................................................ 321 bacteria inoculation........................................... 167
CA125 .................................................................. 414–419 casting plate with membrane................... 166, 167
CA15-3 .......................................................................... 413 fungi inoculation ............................................... 167
CA19-9 .......................................................................... 413 membrane filter ........................................ 160, 161
Calpains ......................................330, 339, 347, 357, 368 protein extraction and sample
Calpastatins.................................................. 330, 355, 357 preparation.......................................... 167, 169
Cancer sampling............................................................. 168
angiogenesis and metastasis.................................... 363 data integration and heat map generation............. 162
atherosclerosis ......................................................... 346 plate-based method................................................. 160
biomarkers ............................................................... 353 polysaccharides ........................................................ 159
caspase-8 .................................................................. 351 prediction
cathepsins B ............................................................. 351 database ............................................................. 162
cell lysosomes .......................................................... 321 dbCAN web server................................... 162, 163
cell proliferation ............................................. 341, 352 protein secretion (see Protein secretion)
cellular micro-environment .................................... 341 protein extraction (see Protein extraction)
chronic inflammation .............................................. 341 quantitative proteomics data
colorectal ................................................................. 336 calculation, secretome enrichment................... 173
disease prognosis ..................................................... 353 Perseus, heat map.............................168, 171, 172
epigenetic................................................................. 341 research .............................................................. 168
epitopes.................................................................... 353 sample ................................................................ 173
extracellular matrix.................................................. 353 spreadsheet application ..................................... 170
hemostasis................................................................ 354 technique ........................................................... 168
inflammation ........................................................... 352 transparency....................................................... 168
inflammatory breast cancer (IBC).......................... 352 Cell migration ............................................ 324, 326–328,
lung ................................................321, 329, 337, 362 331, 336
mesotrypsin ............................................................. 351 Cell painting ..............................................................20, 21
MMPs ............................................................. 341, 366 Cell proliferation ........................................ 315, 324, 326,
ofatumumab ............................................................ 352 328, 331, 335, 336, 341, 345, 351–353
PC-3 cells................................................................. 335 Cells and tissue samples ................................................ 266
plasma ............................................................. 351, 353 Chain of translatability..............................................20, 38
procoagulant............................................................ 354 Chemical proteomics .................................................... 199
proliferation ............................................................. 353 Chromatin immunoprecipitation ................................. 438
protease inhibitors................................. 349, 350, 353 Chymotrypsin.............................................. 406, 407, 409
proteasome .............................................................. 352 Chymotrypsinogen .............................................. 324, 325
rituximab ................................................................. 352 Clinical Laboratory Improvement Amendments
signaling.......................................................... 341, 353 (CLIA) ........................................................... 13
thyroid ..................................................................... 329 Coeliac disease (CD)..................................................... 405
treatment ........................................................ 321, 330 Collagenases .................................................................. 326
zymogens ................................................................. 341 Collision induced dissociation (CID) .......................... 230
Cancer diagnosis .......................................................70, 71 Colonic biopsies ............................................................ 127
Candida albicans .......................................................... 439 Colonic lavage ............................................................... 127
Capture arrays ............................................................... 109 Combinatorial hexapeptide ligand library (CPLL)
Carbohydrate-Active Enzymes (CAZymes), see CAZymes allergomics............................................................... 394
in microbial secretomes IgE-binding proteins .............................................. 394
Cardiovascular disease.........................124, 339, 342–344 preparation protocol ............................................... 395
Caspase-8 .................................................... 314, 331, 344, protein elution................................................ 400, 401
346, 347, 351 protein enrichment ................................................. 394
Catalase .......................................................................... 439 treatment ............................................... 394, 396, 399
Cathepsins ..................................314, 321, 330, 337, 340 Companion diagnostic.................................13, 22–24, 36
Cathepsins B.................................................................. 351 Complement system ............................................ 318, 322
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
Index 469
Computational analysis DNA repair ..........................................315, 329, 331, 332
data......................................................... 118, 119, 121 DNA replication .......................................... 315, 329, 332
image........................................................................ 118 DNA/RNA binding protein. ....................................... 438
Computational proteomics analysis ............................. 452 Double-one dimensional electrophoresis (D1-DE)
Conformational variants ...........................................44, 45 allergomics application............................................ 134
Critical micelle concentration (CMC) ........................... 58 IEF and SDS-PAGE................................................ 136
Crosstalk ..............................................197, 200, 206, 207 IEF preparation .............................................. 136, 137
Cryomolds ............................................................ 255, 257 IgE immunoblots .................................................... 139
Cryotome .................................................... 255, 257, 262 materials.......................................................... 134, 135
Cytosolic proteins ................................................ 266, 268 preparation ............................................ 136, 138, 139
Cytosolic proteolysis ............................................ 331, 334 principles.................................................................. 135
screening .................................................................. 134
D DPP-4 (membrane metallo-endopeptidase and
Data analysis ........................................226, 229, 232, 233 dipeptidyl peptidase-IV).............................. 414
Drug development ..................... 363, 366–368, 370, 372
Data independent acquisition (DIA)
DDA ........................................................................ 458 Drug discovery
FDR ......................................................................... 462 binary interaction model .......................................... 21
cell-killers ................................................................... 19
General Public Licence (GPL) ............................... 456
mass spectrometry................................................... 457 FDA ........................................................................... 20
reproducibility-optimized test statistic isoforms ..................................................................... 21
(ROTS) ........................................................ 464 oligomers ................................................................... 21
PDD..................................................................... 19–22
spectral library ................................................ 455, 459
SWATH-MS ............................................................ 455 SNPs .......................................................................... 19
Data mining..................................................198, 202–206 TDD ....................................................................19, 20
Drug targets
Data-dependent acquisition (DDA) .......... 455, 458, 459
Data-independent acquisition (DIA) anti-AD .................................................................... 336
LC-MS/MS............................................................. 455 chymase and tryptase .............................................. 346
exogenous proteolytic activity .............. 366, 368, 369
Digestion .................................................... 313–316, 376,
406–409, 411 identification.............................................................. 71
Dimethyl isotopic labelling........................................... 187 non-protease components ...................................... 370
Dimethylation .....................................227, 236, 238, 240 protease inhibitors.......................................... 363–367
proteases ......................................................... 316, 341
Disease
Alzheimer’s ....................................336, 366, 376, 378
E
assessment................................................................ 329
autoimmune ......................................... 321, 337, 340, E. coli .................................................................... 446–448
356, 359, 366 Eculizumab........................................................... 321, 370
cardiovascular and metabolic.................339, 342–344 Effectors............................. 314, 316, 320, 329, 331, 347
chronic ..................................................................... 378 Electrolyte........................................................................ 72
degenerative............................................................. 340 Electron paramagnetic resonance
epigenetics ............................................................... 334 spectroscopy (EPR) ....................................... 32
Huntington’s........................................................... 366 Electrophoresis system.................................................... 62
immune.................................................................... 321 Electrospray ionization (ESI) ....................................... 181
inflammation .................................................. 338, 339 Enamelysin (MMP-20) ................................................. 326
inflammatory bowel ................................................ 321 Enrich post-translational modifications .............. 198–200
liver .......................................................................... 363 Enterokinase .................................................................. 325
lymphoproliferative ................................................. 364 Enzyme-linked immunosorbent assays
metabolic ................................................................. 340 (ELISAs) .........................................29, 30, 405
neurodegenerative................ 324, 329, 350, 355–357 Epidermal growth factor (EGF)................. 316, 324, 336
PAR4 signaling ........................................................ 328 Epigenetics ................................................. 8, 26, 35, 316,
Parkinson’s .............................................................. 336 332, 334, 337, 341, 351, 352
prognosis ................................................................. 353 Epilysin (MMP-28)....................................................... 326
proteases ......................................................... 316, 364 Epitopes ......................................321, 329, 339, 353, 356
thromboembolic ..................................................... 363 Error tolerant search ............................................ 239, 242
DNA processing ................................................... 329, 332 Exoglycosidase .............................................................. 422
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
470 Index
Extracellular matrix (ECM)................................ 324–327, MS........................................................................29, 30
334, 338, 339, 341, 343, 346, 349, 350, 353, NMR....................................................................31, 32
360, 363 PDD............................................................................. 3
Extracellular regulated protein kinase (ERK).............. 266 PM (see Precision Medicine (PM))
protein activity........................................................... 33
F protein localization ................................................... 32
Factor VII ................. 314, 318, 335, 342, 344, 347, 363 proteomics methods appraisal ............................28, 29
Factor Xa .................. 318, 319, 335, 342, 360, 363, 364 PTMs ..................................................... 27, 29, 35, 37
quantitation .........................................................30, 31
Factor XII ............................................................. 319, 364
Factor XIIIa ................................................................... 319 Sep-Sci/MS .........................................................29, 30
False discovery rate (FDR) .................................. 460, 462 specification ................................................................. 2
spin labeling............................................................... 34
Fecal samples ................................................126–128, 130
Fenton reaction ............................................................... 61 systems biology .................................................2, 7–10
Flour ..................................................................... 407, 411 Uniprot profile .......................................................... 27
X-ray crystallography ................................................ 31
Fluorescence resonance emission transfer
(FRET) technology ..................................... 144 Y2H............................................................................ 34
Fluorescent-protein tagging (FP) ..................... 28, 32, 33 Fungi.............................................................................. 123
Folate receptor (FR) ..................................................... 266
G
Fractionation ............................................... 446, 447, 454
Fructose bisphosphate aldolase .................................... 439 Galileo Biosciences.......................................................... 62
Functional proteomics ..............................................44, 45 Gas chromatography coupled to mass
biomarker spectrometry (GC-MS) ............................... 280
definition ............................................................. 11 Gastrointestinal (GI)..................................................... 123
FDA ..................................................................... 12 Gel electrophoresis........................................................ 394
function ............................................................... 11 Gelatinases ..................................................................... 326
functional networks and pathways ........................3 Gene expression ................................................. 14, 26, 35
nucleotides........................................................... 13 Gene knockouts ................................................... 438, 439
therapy selection.................................................. 11 Genome
urine analysis........................................................ 11 COL1A1 ...................................................................... 3
bottom-up approaches.............................................. 27 HBB, HBA1, and HBA2............................................ 3
diagnostic/prognostic discovery HGP............................................................................. 5
biomarker............................................................. 15 proteome ..................................................................... 5
Differentiation Set V0 ......................................... 16 PRSS1 .......................................................................... 3
LC-MS/MS......................................................... 14 PTM............................................................................. 5
pattern diagnostics .............................................. 14 Glucose-6-Phosphate Dehydrogenase
protein-based....................................................... 14 (G-6-PDH) .................................................... 72
“risk assessment” tests ........................................ 17 Glutamine synthetase.................................................... 439
serum proteome .................................................. 15 Gluten .......................................................... 405, 407, 409
systems biology ................................................... 17 Glyceraldehyde 3-phosphate dehydrogenase
drug discovery (GAPDH) .................................................... 440
binary interaction model .................................... 21 Glycoepitope recognition .................................... 426–429
cell-killers ............................................................. 19 Glycolytic enzyme activity
FDA ..................................................................... 20 data transformation and analysis .............................. 78
isoforms ............................................................... 21 electrolyte .................................................................. 72
oligomers ............................................................. 21 G-6-PDH .................................................................. 72
PDD............................................................... 19–22 hexokinase activity..................................................... 72
SNPs .................................................................... 19 IEF .......................................................................75, 76
TDD ..............................................................19, 20 IPG.......................................................................72, 76
EPR ......................................................................32, 34 MS.............................................................................. 79
FP ............................................................................... 32 NADP ........................................................................ 72
genome ................................................................ 3–5, 7 P24............................................................................. 78
HPP ........................................................................... 27 PEP ......................................................................72, 78
IF................................................................................ 32 PEP Universal Protein Purification kit .................... 74
“known” interacting pairs ........................................ 33 protein purity confirmation ...................................... 79
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
Index 471
protein staining ......................................................... 73 Immobilized enzyme reactor (IMER) ........................296,
protein transfer ....................................................77, 78 297, 304–306
sample treatment ....................................................... 75 Immobilized metal affinity chromatography
SDS-PAGE gel .......................................................... 71 (IMAC) ............................................... 199, 206
Glycopeptide .......................................422, 429, 430, 434 Immobilized pH gradient (IPG).................................... 72
Glycoproteomics .................................................. 422, 427 Immune regulation .............................................. 318, 322
Glycosidases................................................................... 321 Immunoassay........................................................ 414–417
Glycosylation .............................................. 199–201, 203, Immunofluorescence (IF).........................................28, 32
204, 206, 413, 414, 418, 421, 434 Immunohistochemical staining ........................... 254, 263
Guinea pig lens.............................................................. 437 Immunoproteomics ............................................. 133, 393
Gut microbiome Infectious organisms ............................................ 359–362
adult gastrointestinal tract ...................................... 123 Inflammation .............................................. 314, 316, 321,
challenges................................................................. 128 325, 328, 330, 335, 337–341, 345–347, 352,
diet ........................................................................... 124 356, 359, 362, 366, 370, 372, 377
early childhood........................................................ 124 Inflammatory bowel disease (IBD) ..................... 124, 125
HMP ........................................................................ 124 Inflammatory breast cancer (IBC) ............................... 352
human ...................................................................... 124 In-gel digestion ........................................... 228, 229, 247
inflammation, diseases ............................................ 125 In-solution tryptic digestion ........................................ 268
low abundance ........................................................ 124 Intramembrane proteolysis......................... 331, 334, 335
mass spectrometry instrumentation ....................... 130 Intranuclear proteolysis ....................................... 331, 334
metaproteomics, complex samples................ 125, 126 Iodoacetamide (IAA) ............................................. 70, 267
Pie chart................................................................... 129 Ion chromatogram curve.............................................. 189
sample collection ............................................ 127, 128 Ion exchange chromatography (IEX-HPLC) .... 449, 451
shotgun proteomics approach ................................ 126 Isobaric peptide labels.......................................... 188, 189
software tools ................................................. 128–130 Isobaric Tag for Relative and Absolute
Quantification (iTRAQ).............................. 188
H Isoelectric focusing (IEF) ....................................... 56, 57,
70, 71, 446, 448, 449, 454
Heat shock proteins 27 (HSP27) ................................ 266
Heavy stable isotopes .................................................... 266 Isoforms........................................................................... 21
Hemocytometer ................................................... 266, 267 Isotope dilution MS (IDMS) ....................................... 295
Hemostasis .................................316, 328, 339, 341, 354 Isotope-coded Affinity Tags (ICAT)............................ 187
Hexapeptide ligand libraries ................................ 396, 398
K
Hexokinase activity ...................................................72, 74
High performance liquid chromatography Kallikreins ..................................................................42, 48
(HPLC)..................................... 297, 298, 301, Kluyveromyces lactis (KlAlt1) ........................................ 440
408, 411, 446, 447, 449 KUMASI staining .....................................................65, 67
High-density microarray, see Protein microarray
Higher energy collisional dissociation (HCD)............ 230 L
High-performance liquid chromatography Label-free proteomics ................................................... 254
(HPLC)............................................... 406, 407
Label-free quantitative shotgun proteomics
High-resolution mass spectrometry............................. 285 LC-MS/MS............................................................. 190
Human Microbiome Project (HMP)........................... 124 processing ................................................................ 189
Human Proteome Project (HPP) .................................. 26 protein identification .............................................. 189
Human serum ...................................................... 298, 301
Lacchancea kluyveri (LkAlt1) ....................................... 440
Huntington’s disease .......................................... 324, 331, Laser microdissection system ....................................... 255
339, 344, 355, 357, 366 LCM human pancreatic islets....................................... 256
Hydrazide chemistry ................................... 199, 203, 204
LC-MSMS ..................................................................... 394
Hydrophobic proteins .................................................... 56 Leakproof gel casting system.......................................... 62
Lectin affinity chromatography ........................... 203, 204
I
Lectins................................................................... 414, 418
IgE ............................................................... 393, 394, 397 Ligands ....................................................... 314, 316, 320,
IgE binding proteins............................................ 133, 140 321, 324, 328, 329, 331, 332, 369
IMER-LC-MS/MS.............................296, 297, 299, 303 Lipids ................................................................................. 7
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
472 Index
Liquid chromatography, see Staphylococcus aureus MaxQuant ............................................................ 232, 261
Liquid chromatography-electrospray ionization- Mean spectral intensity (MSI) ........................................ 90
tandem mass spectrometry Median effective dose (ED50) ..................................... 156
(LC-ESI-MS/MS)....................................... 225 Median lethal dose (LD50) ................................. 155, 156
Liquid chromatography-tandem mass spectrometry Membrane proteins............................................ 56–58, 60
(LC-MS/MS) ...................................... 14, 254, Mesotrypsin ......................................................... 324, 325,
260, 265, 295, 452, 455 340, 350, 351
Low-abundance allergens ............................................. 393 Metabolic disease .........................................339, 342–344
Metabolic disorders....................................................... 124
M Metabolic enzyme ........................................................... 71
Magnetic beads ........................................... 414, 416, 417 Metabolites ..................................................................7, 12
Metabolomics, see Staphylococcus aureus
MALDI, see Matrix-assisted laser desorption and
ionization (MALDI) Metal-ion based enrichment......................................... 199
MALDI-Imaging MS (MALDI-IMS) ................ 190, 191 Metallo-elastase (MMP-12) ......................................... 326
Metaproteomics analysis, gut microbiome,
MALDI-MS.......................................................... 225, 246
Mascot ............................................... 226, 228, 232, 239, see Gut microbiome
241, 242, 244, 245 Methicillin resistance............................................ 279, 281
Microarray
Mass spectrometry (MS)...................................29, 30, 79,
198, 200–204, 216–217, 394, 405, 422, 430, protein (see Protein microarray)
455, 457 Microbiome .................. 6, 7, 14, 26 See Gut microbiome
biological samples ................................................... 190 Microbiota ..................................................................... 3, 7
Microdissected...................................................... 254, 259
challenges, proteome measures ..................... 179, 180
components ............................................................. 180 MicroRotofor ....................................................... 447–449
dimethylation .......................................................... 227 Mitochondria................................................................... 60
Mitochondria isolation buffer (MIB) ............................ 58
error tolerant searching .......................................... 239
ESI ........................................................................... 181 MMP-1 ................................................326, 346, 366, 371
fractionation ............................................................ 182 MMP-2 and -9 ...........................326, 327, 331, 341, 347
MMP-3, -10, and -11 .......................................... 326, 327
in-gel digestion........................................................ 229
MALDI .................................................................... 181 Moonlighting proteins, see Proteomics
MALDI-IMS .................................................. 190, 191 MoonProt ............................................................. 438, 441
Mascot ..................................................................... 226 MRM, see Multiple reaction monitoring (MRM)
MSConvert GUI ........................................................... 249
mass tolerant searches ............................................. 241
Orbitrap ................................................................... 228 Mucosal lavage ..................................................... 127, 128
protein identification Multifunctional proteins ...................................... 437, 439
Multiple post translational modifications .................... 200
bottom-up approach ................................ 181, 182
top-down approach.................................. 183, 184 Multiple reaction monitoring (MRM) ................. 30, 184
Proteome Discoverer .............................................. 236 Murine double minute.................................................. 266
proteotypic peptide ................................................. 184
N
PTMs ....................................................................... 230
QTOF ...................................................................... 233 Nano-electrospray ionization tandem mass
quantitative proteomics .......................................... 185 spectrometry (nano-ESI-LC-
selective reaction monitoring ................................. 184 MS/MS) ........................................................ 89
shotgun proteomics ................................................ 183 Nanoflow liquid chromatography tandem mass
(see also Shotgun proteomics) spectrometry (nLC-MS/MS) ....................448,
trypsin ...................................................................... 227 452, 453
Unimod ................................................................... 226 Neurodegenerative diseases ................................ 324, 329,
Mass tolerant search.....................................239, 241–243 350, 355–357
Matrilysin ....................................................................... 326 Neutralization potency (P) ........................................... 156
Matrilysin-2 (MMP-26)................................................ 326 New Liberty Proteomics (NLP)....................................... 9
Matrix metalloproteinases (MMPs) ............................314, N-glycans ............................................................. 421, 426,
321, 326, 331, 339–341, 346, 357, 363, 428, 430, 431, 434
366, 376 N-glycomes........................................................... 422, 433
Matrix-assisted laser desorption and ionization Normalization, data ...................................................... 119
(MALDI) ................................... 181, 190, 394 Normalized potency (n-P)................................... 156, 158
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
Index 473
Nuclear magnetic resonance spectroscopy separation......................................97–98, 100–101
(NMR) ....................................... 28, 31, 32, 36 SDS-PAGE .............................................................. 100
Nucleotides........................................................... 7, 13, 22 Plasma ...............................318, 321, 329, 335, 346, 347,
NuGel™ ........................................................................... 43 350, 351, 353, 356, 357, 359, 360, 363, 364,
414, 416, 418
O Plasminogen ...................................... 316, 319, 320, 331,
Obesity........................................................................... 124 338, 343, 345, 360, 361, 439–441
Ofatumumab ................................................................. 352 Pleiotropic effects.......................................................... 437
Pollen ...................................................394, 396, 398, 401
“Off-line” MALDI-TOF MS ....................................... 433
18
O labelling.................................................................. 187 Polyacrylamide gel electrophoresis
Oligomers ........................................................................ 21 (PAGE).................................56, 57, 59, 60, 64
Polyethylene glycol (PEG) ............................................. 94
Online digestion............................................................ 297
Orbitrap ....................................................... 228, 230, 233 Polymerase chain reaction (PCR) ................................ 179
Post-translational modifications
P (PTMs)............... 4–6, 28, 107, 226, 230, 413
PPI, see Protein-protein interactions (PPI)
Palmitoylation .....................................199, 201, 203, 204 Precision Medicine (PM)..........................................23–26
Parkinson’s disease ..............................324, 336, 355, 357 Pre-Human Genome Project (HGP)............................... 5
Pathogenic organisms ................................................... 123 Pre-procalcitonin........................................................... 329
PDL, see Proximity-dependent labeling (PDL) Procarboxypeptidases.................................................... 325
PEAKS ........................................................................... 232 Procoagulant .......................................316, 320, 354, 359
PEP Universal Protein Purification kit .......................... 74 Proelastases .................................................................... 325
Peptidase.............................................................. 315, 329, Programmed cell death................................315, 326–331
332, 352, 356, 368 Prolipases ....................................................................... 325
Peptide fractionation......................................98, 101–102 proMMP-2 ...................................................................... 42
Peptide spectral library ................................................... 94 Propeptide ...........................................313, 314, 326, 334
Peptides .............................................................. 3, 7, 9, 21 Prostate cancer ................................................................ 42
Peptide-spectrum matches............................................ 182 Prostate specific antigen (PSA) ...................................... 42
Perseus ........................................................................... 261 Protease degradome...................................................... 315
pFind.............................................................................. 232 Protease inhibitors ............................................ 16, 41–43,
p-Glycoprotein (P-gp) .................................................. 266 316, 319, 326, 341, 353, 360, 363, 366, 367,
Phenotypic drug discovery (PDD) ...........................3, 18, 369, 370, 377
19, 21, 22, 26 Protease promoter....................................... 314, 334, 351
assay ........................................................................... 22 Proteases
cellular level ............................................................... 20 diseases
to TDD................................................................19, 20 autoimmune ...................................................... 356
Phosphatases............................................ 71, 77, 321, 375 cancer (see Cancer)
Phosphoglycerate kinase ............................................... 439 cardiovascular and metabolic diseases .............. 339
Phosphoglycerate mutase ............................................. 439 epigenetics ................................................ 334, 337
Phospholipases .............................................................. 321 infectious organisms.........................359, 360, 362
Phosphorylation ......................... 197, 199–202, 207, 218 inflammation ............................................ 337, 340
Plant proteome neurodegenerative.....................................350–358
acetone/TCA/phenol extraction ......................95, 99 stroke ................................................................. 339
extraction .............................................................95, 99 physiological and regulatory roles................. 314, 316
homogenization ..................................................94, 98 protein digestion ............................................ 313–315
in-gel digestion...................................97–98, 100–101 regulation of physiological processes
in-solution digestion ........................................ 97, 100 cell migration and proliferation................324–328
PEG fractionation ........................................ 94, 95, 99 complement system and immune
peptide regulation.............................................318–323
desalting................................................ 96, 98, 101 DNA replication, repair and
fractionation ................................ 96, 98, 101–102 processing ........................................... 329, 333
protein hemostasis..................................................316–320
digestion .............................................................. 95 intramembrane proteolysis ............................... 335
extraction ............................................................. 95 intranuclear proteolysis ..................................... 331
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
474 Index
programmed cell death ..................................... 326 assembled arrays ...................................................... 109
protein secretion ............................................... 329 bead arrays ............................................................... 109
proteolytic processing ....................................... 321 computational analysis (see Computational
tissue remodeling .............................................. 324 analysis)
transmembrane proteolysis ...................... 334, 335 computational resources ......................................... 113
Proteasome ................................ 314, 317, 321, 329–331, customize design ............................................ 113, 114
350, 352, 359, 360, 362, 368, 369, 375 experimental workflow............................................ 116
Protein high-density............................................................. 108
Differentiation Set V0 ............................................... 16 image acquisition
digestion .................................................................... 89 experimental workflow...................................... 117
functionality............................................................... 21 materials............................................................. 113
HGP............................................................................. 4 planar arrays.................................................... 108, 109
identification........................................................27, 87 reverse-phase arrays................................................. 109
interaction............................................................22, 27 self-assembled arrays ............................................... 110
quantitation ............................................................... 90 Protein purification ......................................................... 74
separation.............................................................84, 86 Protein quantification .......................................... 266, 274
Uniprot ........................................................................ 4 Protein secretion ......................................... 315, 329, 332
visualization ............................................................... 86 fungi......................................................................... 165
Protein complexes ......................................................... 145 in silico prediction ................................................... 163
biochemical separation............................................ 445 lipoproteins..................................................... 164, 165
computational proteomics analysis ........................ 452 machine learning approaches.................................. 163
E. coli ....................................................................... 447 materials................................................................... 160
fractionation ............................................................ 447 nonclassical .............................................................. 166
HPLC ...................................................................... 446 SignalP ............................................................ 163–165
IEF ........................................................................... 448 WoLF PSORT ......................................................... 165
IEX-HPLC .............................................................. 449 Protein staining ............................................................... 73
LC-MS/MS............................................................. 452 ProteinPilot ................................................................... 232
nLC-MS/MS .......................................................... 448 Protein–protein interactions (PPIs) ................... 143–145,
non-denaturing isoelectric focusing....................... 447 150, 438, 439, 445
PPIs .......................................................................... 445 Proteoforms............................................. 44, 52, 183, 296
soluble protein extraction ....................................... 448 Proteolysis
TCA ......................................................................... 451 complexity ............................................................... 316
trypsin digestion...................................................... 452 cytosolic ................................................................... 331
2-D IEF-IEX ........................................................... 446 drug and diagnostic development
2-D proteomic fractionation .................................. 446 active site targeting ........................................... 368
Protein Data Bank (PDB) .............................................. 31 exosite and effector binding sites ..................... 371
Protein elution plate (PEP) ............................................ 71 inhibitor biomarkers .................................375–377
glycolytic enzyme activity ......................................... 72 mechanism-based targeting .............................. 370
protein purification ................................................... 74 drug targets (see Drug targets)
protein transfer .......................................................... 77 intramembrane ........................................................ 335
Protein fractionation, see Plant proteome organelle membranes .............................................. 329
Protein function prediction .......................................... 437 regulatory mechanisms ........................................... 316
Protein interactions............................................... 5, 9, 22, transmembrane............................................... 334, 335
28, 33, 34, 37 Proteolytic enzymes ............................................. 314, 315
Protein isoforms (proteoforms) ................................... 183 Proteolytic processing.......................................... 321–325
Protein kinases................................................................. 71 Proteome Discoverer .......................................... 228, 232,
Protein markers ........................................... 413, 414, 418 233, 236, 239, 248, 249
Protein microarray Proteomics................................................... 107, 108, 393
array assays bioinformatics analysis ............................................ 441
experimental workflow...................................... 116 D1-DE (see Double-one dimensional
materials.................................................... 112, 113 electrophoresis (D1-DE))
array printing expression patterns/cellular localization ............... 440
experimental workflow...................................... 114 gene knockouts ....................................................... 439
materials.................................................... 111, 112 MS (see Mass spectrometry (MS))
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
Index 475
protein–protein interactions ................................... 439 Serine suicidal protease inhibitors .................................. 42
(see also Targeted proteomics) SERPIN
Proteomics methods appraisal ..................................28, 29 AAT............................................................................ 44
Proteotypic peptide....................................................... 184 AlbuSorb™ ...........................................................43, 46
ProteoWizard .............................................. 249, 456, 458 AlbuSorb™ PLUS ..................................................... 43
Proximity-dependent labeling (PDL) .......................... 144 AlbuVoid™ ...........................................................43, 46
PTM crosstalk ............................................................... 206 biomarkers .................................................... 42, 44, 53
PTM sites.............................................................. 198, 207 function ........................................................ 42–44, 48
LC-MS .................................................................47, 52
Q mechanisms .................................................. 42, 44, 53
Q-Orbitrap mass spectrometer..................................... 283 NuGel™ ..................................................................... 43
protease...................................................................... 43
QTOF ................................................................... 230, 233
Quantitation ............................................... 184, 186, 187, proteoforms ............................................................... 53
189, 191, 200, 201, 211, 217 PSA ............................................................................ 42
RCL .....................................................................42, 44
Quantitative.......................................................... 295, 297
Quantitative proteomics SERPINA1 ................................................................ 42
gel-based methods .................................................. 185 SERPINC1 ................................................................ 42
SERPINF2................................................................. 42
protein expression ................................................... 185
Quantitative shotgun proteomics SERPING1................................................................ 42
label-free (see Label-free quantitative shotgun tissue kallikreins......................................................... 42
proteomics) Serpins................................ 316, 319, 337, 360, 363, 372
Serum.................................................................... 414, 418
labelling
18
O labelling...................................................... 187 Shotgun proteomics............................................. 126, 130
dimethyl isotopic labelling................................ 187 data-dependent and independent .......................... 183
protein identification .............................................. 182
ICAT .................................................................. 187
isobaric peptide ........................................ 188, 189 quantitative (see Quantitative shotgun proteomics)
SILAC ................................................................ 186 Signaling .................................... 316, 321, 324–328, 335,
339–341, 345, 351–353, 356, 363, 366, 367
super-SILAC...................................................... 186
Quinone oxidoreductase .............................................. 437 Signaling pathways ...................................... 198, 201, 216
Single-nucleotide polymorphisms (SNPs)..................... 19
R Snake venom
ESI-LC-MS/MS ....................................................... 89
Reactive center loop (RCL)............................... 42, 44–46 peptides extraction and desalting ............................. 86
Reversed-phase high performance liquid chromatography protein
(RP-HPLC) .......................................... 84, 229 digestion ........................................................85, 89
Reverse-phase arrays...................................................... 109 identification........................................................ 87
Rituximab ............................................................. 352, 356 and peptides......................................................... 84
RNA splice variants ....................................................... 437 quantitation ......................................................... 90
separation............................................................. 86
S
visualization ......................................................... 86
Saccharomyces cerevisiae................................200–204, 440 RP-HPLC .................................................................. 84
Secretomics, see CAZymes in microbial secretomes SDS-PAGE ................................................... 84, 85, 87
SELDI............................................................................ 394 separation................................................................... 91
Selected/multiple reaction monitoring Snake venom proteomes
(SRM/MRM)....................................... 93, 265 antivenom (see Antivenom)
Selective reaction monitoring (SRM) ................. 184, 455 complexity ............................................................... 153
Self-assembled arrays..................................................... 110 death and survival ................................................... 156
Sequential window acquisition of all theoretical evolution, clinical syndrome................................... 154
spectra (SWATH) .......................................... 94 lethality determination............................................ 155
Sequential windowed acquisition of all theoretical lethality neutralization ................................... 156, 157
fragment ion mass spectra materials................................................................... 154
(SWATH-MS) ........... 184, 455, 456, 458–460 median effective dose.............................................. 156
SEQUEST ..................................................................... 232 median lethal dose .................................................. 156
Serial enrichment of PTMs........................................... 199 toxicity ..................................................................... 154
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
476 Index
Sodium dodecyl sulphate polyacrylamide gel Three-dimensional structures....................................... 441
electrophoresis (SDS-PAGE) .....................423, Threonine proteases............................................. 315, 360
426, 427, 429–431 Thrombomodulin ...................................... 316, 318–320,
Sodium dodecylsulfate (SDS)............................ 56, 57, 60 336, 342, 349
Spectral counting .......................................................... 189 Tight-binding tissue inhibitors of metalloproteinases
Spectral library.....................................455, 456, 459, 462 (TIMPs) ....................................................... 326
Stable isotope-labeled (SIL) ............................... 295, 296, TIMP-1 (tissue inhibitor of metallopeptidase 1) ........ 414
298, 300, 302, 304, 307 TMT, see Tandem Mass Tags (TMT)
Stable isotopic labelling of amino acids in Toxin-based drug discovery ........................................... 84
culture (SILAC)........................................... 186 Transcription elongation factor.................................... 439
Staphylococcus aureus Transferrin (Tr) ............................................................. 266
bacteria sampling and metabolism quenching ...... 285 Transferrin receptor (TfR)............................................ 266
bacterial culture ....................................................... 282 Transmembrane domains (TMDs) ..........................56, 57
bacterial pre-culture and culture ............................ 284 Transmembrane proteolysis........................ 331, 334, 335
bacterial strains ........................................................ 281 Trichloroacetic acid (TCA)........................................... 451
GC-MS .................................................................... 280 Trypsin ........................................................ 227, 230, 238,
intracellular metabolites................................. 285–287 406, 407, 409
LC-MS analysis........................................................ 283 Trypsin digestion ................................................... 85, 452
metabolite extraction ..................................... 282–283 Tryptic peptide mapping ............................ 424, 428, 429
MRSA ...................................................................... 279 Tumor markers.............................................................. 414
Streptococci ................................................................... 440 Two dimensional differential gel electrophoresis
Streptomyces coelicolor .................................................... 441 (2D-DIGE).................................................. 126
Stroke.......................................... 331, 339–348, 363, 367 Two-dimensional electrophoresis
Stromelysins................................................................... 326 (2-DE).........................................133–135, 140
Strong anion exchange (SAX) ............................. 229, 249 2-D gel electrophoresis.............................................56, 70
Strong cation exchange (SCX) peptide 2-D gel separation........................................................... 72
fractionation.......................................... 94, 102 2D IEF/SDS PAGE ....................................................... 57
Succinylation .......................................199, 201, 202, 205 2-D IEF-IEX ................................................................. 446
Sulfatases........................................................................ 321 2-D proteomic fractionation ........................................ 446
Surrogate peptide selection .......................................... 269 2-D separation............................................................... 446
SwissProt ....................................................................... 233
Systems biology............................................................. 265 U
Drug discovery ............................................................ 8 Ubiquitination..............................................197, 199–201
molecular level........................................................... 10 Unexpected modifications .................................. 226, 228,
NLP ............................................................................. 9
230, 232, 233, 239
Unimod ....................................................... 226, 239, 245
T
Uniprot .............................................................................. 4
Tandem mass spectrometry (MS/MS) ................ 84, 126, Urokinase............................................................. 319, 320,
181, 186–188, 190 324, 339, 344, 347, 349
Tandem mass tag labeling ............................................ 208
Tandem Mass Tags (TMT)........................................... 188 V
Target-based drug discovery (TDD) .......................18, 19 Validation..................................................... 226, 242, 245
Targeted proteomics Venom proteome ................................................. 153, 154
cell viability test ....................................................... 266
Venom separation............................................................ 91
LC-MS/MS............................................................. 265 Venom toxicity .............................................................. 154
MS/MS instrument ................................................ 267 Venomics ......................................................................... 83
protein extraction.................................................... 268
protein extraction buffers ....................................... 266 W
SRM/MRM ............................................................ 265
surrogate peptide selection..................................... 269 Wash Buffer AVWB......................................................... 50
tissue homogenization ............................................ 266 Western blotting.................................................. 422, 423,
tryptic digestion buffer ........................................... 267 428, 430, 431, 434
Thioredoxin peroxidase ................................................ 439
FUNCTIONAL PROTEOMICS: METHODS AND PROTOCOLS
Index 477
X Z
X!Tandem ............................................................. 232, 462 Zeta-crystallin................................................................ 437
X-ray crystallography .....................................31, 315, 372 Zymogens ..................................................... 41, 314, 326,
341, 360, 371
Y
Yeast two-hybrid (Y2H) ..................................28, 34, 439