Proteomic Profiling - Methods and Protocols
Proteomic Profiling - Methods and Protocols
Proteomic
Profiling
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
Proteomic Profiling
Methods and Protocols
Edited by
Anton Posch
Bio-Rad Laboratories GmbH, Munich, Germany
Editor
Anton Posch
Bio-Rad Laboratories GmbH
Munich, Germany
ISSN 1064-3745
ISSN 1940-6029 (electronic)
Methods in Molecular Biology
ISBN 978-1-4939-2549-0
ISBN 978-1-4939-2550-6 (eBook)
DOI 10.1007/978-1-4939-2550-6
Library of Congress Control Number: 2015933382
Springer New York Heidelberg Dordrecht London
Springer Science+Business Media New York 2015
This work is subject to copyright. All rights are reserved by the Publisher, whether the whole or part of the material is
concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction
on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation,
computer software, or by similar or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not
imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and
regulations and therefore free for general use.
The publisher, the authors and the editors are safe to assume that the advice and information in this book are believed to
be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty,
express or implied, with respect to the material contained herein or for any errors or omissions that may have been made.
Printed on acid-free paper
Humana Press is a brand of Springer
Springer Science+Business Media LLC New York is part of Springer Science+Business Media (www.springer.com)
Preface
Sample Preparation Methods and Protein Techniques for Proteomic Profiling
This volume is a comprehensive continuation and extension of a book called 2D PAGE:
Sample Preparation and Fractionation which was published in 2008.
This book presents the latest developments of the main pillars of protein analysis,
namely sample preparation, separation, and characterization. Individual technologies of
each pillar combined into complementary and robust workflows render proteomic analysis
of complex biological samples even more powerful and are the prerequisite to gain maximum
value from biological samples in a single experiment.
In this volume, basic but important sample preparation protocols are described again,
followed by sophisticated procedures to enrich for specific protein classes and completed by
the detailed description of integrated workflows for comprehensive protein analysis and
characterization. The authors of the individual chapters are well-known protein biochemists, and all of them have set value to provide a detailed representation of their lab work and
to share important tips and tricks for a successful and reproducible employment of their
precious protocols in other laboratories.
This book is for students of Biochemistry, Biomedicine, Biology, and Genomics and
will be an invaluable source for the experienced, practicing scientist, too.
Munich, Germany
Anton Posch
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
1 Mechanical/Physical Methods of Cell Distribution
and Tissue Homogenization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Stanley Goldberg
2 Sample Preservation Through Heat Stabilization of Proteins:
Principles and Examples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Mats Born
3 Isolating Peripheral Lymphocytes by Density Gradient Centrifugation
and Magnetic Cell Sorting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Frederic Brosseron, Katrin Marcus, and Caroline May
4 Investigating the Adipose Tissue Secretome: A Protocol
to Generate High-Quality Samples Appropriate for Comprehensive
Proteomic Profiling. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Simon Gddeke, Jorg Kotzka, and Stefan Lehr
5 Methods for Proteomics-Based Analysis of the Human Muscle Secretome
Using an In Vitro Exercise Model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Mika Scheler, Martin Hrab de Angelis, Hadi Al-Hasani,
Hans-Ulrich Hring, Cora Weigert, and Stefan Lehr
6 Urinary Pellet Sample Preparation for Shotgun Proteomic Analysis
of Microbial Infection and HostPathogen Interactions . . . . . . . . . . . . . . . . .
Yanbao Yu and Rembert Pieper
7 A Protocol for the Parallel Isolation of Intact Mitochondria
from Rat Liver, Kidney, Heart, and Brain . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Sabine Schulz, Josef Lichtmannegger, Sabine Schmitt, Christin Leitzinger,
Carola Eberhagen, Claudia Einer, Julian Kerth, Michaela Aichler,
and Hans Zischka
8 Isolation of Mitochondria from Cultured Cells and Liver Tissue
Biopsies for Molecular and Biochemical Analyses. . . . . . . . . . . . . . . . . . . . . . .
Sabine Schmitt, Carola Eberhagen, Susanne Weber, Michaela Aichler,
and Hans Zischka
9 Dynamic Range Compression with ProteoMiner:
Principles and Examples . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Lei Li
10 Qualitative and Quantitative Proteomic Analysis of Formalin-Fixed
Paraffin-Embedded (FFPE) Tissue . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Omid Azimzadeh, Michael J. Atkinson, and Soile Tapio
vii
v
xi
1
21
33
43
55
65
75
87
99
109
viii
Contents
117
135
153
167
179
211
225
235
249
259
275
293
305
323
355
Contents
ix
369
381
393
415
427
441
455
465
479
497
Contributors
MARIA FILIPPA ADDIS Porto Conte Ricerche, Tramariglio, Alghero(SS), Italy
MICHAELA AICHLER Research Unit Analytical PathologyInstitute of Pathology,
Helmholtz Center Munich, German Research Center for Environmental Health,
Neuherberg, Germany
HADI AL-HASANI Institute of Clinical Biochemistry and Pathobiochemistry,
German Diabetes Center, Duesseldorf, Germany; German Center for Diabetes
Research (DZD), Duesseldorf, Germany
MARTIN HRAB DE ANGELIS Institute of Experimental Genetics, Helmholtz Zentrum
Mnchen, German Research Center for Environmental Health, Neuherberg, Germany;
Center of Life and Food Sciences Weihenstephan, Technische Universitt Mnchen,
Freising-Weihenstephan, Germany; German Center for Diabetes
Research (DZD), Duesseldorf, Germany
MICHAEL J. ATKINSON Institute of Radiation Biology, Helmholtz Zentrum Mnchen,
German Research Center for Environmental Health, Neuherberg, Germany; Technical
University of Munich, Munich, Germany
OMID AZIMZADEH Institute of Radiation Biology, Helmholtz Zentrum Mnchen,
German Research Center for Environmental Health, Neuherberg, Germany
SRICHARAN BANDHAKAVI diaDexus, South San Francisco, CA, USA
ROMAN G. BAYER Department of Ecogenomics and Systems Biology, University of Vienna,
Vienna, Austria
TOM BERKELMAN Bio-Rad Laboratories, Hercules, CA, USA
SETH BLACKSHAW Solomon H. Snyder Department of Neuroscience, Johns Hopkins
University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering,
Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department
of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA;
Center for High-Throughput Biology, Johns Hopkins University School of Medicine,
Baltimore, MD, USA
MATS BORN Denator AB, Gothenburg, Sweden
FREDERIC BROSSERON Deutsches Zentrum fr Neurodegenerative Erkrankungen (DZNE) e.V.,
Bonn, Germany
OLGA BUNEEVA Institute of Biomedical Chemistry, Moscow, Russia
DONG-SIC CHOI Department of Life Sciences, Pohang University of Science and Technology,
Pohang, Republic of Korea
BERTRAND COLIGNON Dpartement Sciences du Vivant, Centre wallon de Recherches
agronomiques, Gembloux, Belgium; URBC-NARILIS, Universit de Namur,
Namur, Belgium
STUART J. CORDWELL School of Molecular Bioscience, The University of Sidney, Sidney,
Australia; Discipline of Pathology, School of Medical Sciences, The University of Sidney,
Sidney, Australia; Charles Perkins Centre, The University of Sidney, Sidney, Australia
xi
xii
Contributors
ERIC COX Biochemistry, Cellular and Molecular Biology Graduate Program, Johns
Hopkins University School of Medicine, Baltimore, MD, USA; Solomon H. Snyder
Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore,
MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins
University School of Medicine, Baltimore, MD, USA
ALIREZA DEHGHANI Institute for Biochemistry and Molecular Biology, University of Bonn,
Bonn, Germany
EDOUARD DELAIVE URBC-NARILIS, Universit de Namur, Namur, Belgium
JINGJING DENG Department of Biochemistry and Molecular Pharmacology, Kimmel
Center for Biology and Medicine at the Skirball Institute, New York University School
of Medicine, New York, NY, USA
MARC DIEU URBC-NARILIS, Universit de Namur, Namur, Belgium
CAROLA EBERHAGEN Institute of Molecular Toxicology and Pharmacology, Helmholtz Center
Munich, German Research Center for Environmental Health, Neuherberg, Germany
LUTZ ANDREAS EICHACKER Center of Organelle Research, University of Stavanger,
Stavanger, Norway
CLAUDIA EINER Institute of Molecular Toxicology and Pharmacology, Helmholtz Center
Munich, German Research Center for Environmental Health, Neuherberg, Germany
MICHAELA D. FILIOU Max Planck Institute of Psychiatry, Munich, Germany
THOMAS FRANZ Max Planck Institute for Biology of Ageing, Cologne, Germany
YONG SONG GHO Department of Life Sciences, Pohang University of Science
and Technology, Pohang, Republic of Korea
JENNIFER E. GILDA Department of Neurobiology, Physiology, and Behavior, University
of California, Davis, CA, USA
OKSANA GNEDENKO Institute of Biomedical Chemistry, Moscow, Russia
SIMON GDDEKE Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes
Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Duesseldorf,
Germany; German Center for Diabetes Research (DZD), Duesseldorf, Germany
STANLEY GOLDBERG Glen Mills Inc., Clifton, NJ, USA
ALDRIN V. GOMES Department of Neurobiology, Physiology, and Behavior, University
of California, Davis, CA, USA; Department of Physiology and Membrane Biology,
University of California, Davis, CA, USA
DAVID W. GREENING Department of Biochemistry, La Trobe Institute for Molecular
Science, La Trobe University, Melbourne, Australia
ANJA GRIEBEL SERVA Electrophoresis GmbH, Heidelberg, Germany
NICOLA GROLL Department of Protein Analytics, NMI Natural and Medical Sciences
Institute at the University of Tuebingen, Reutlingen, Germany
RAVI GUPTA Department of Plant Bioscience, Pusan National University, Miryang,
Republic of Korea
CATHERINE GUZZO Department of Biochemistry and Molecular Biology, Bloomberg School
of Public Health, Johns Hopkins University, Baltimore, MD, USA
ADRIANA HARBERS Bio-Rad Laboratories, Hercules, CA, USA
HANS-ULRICH HRING Division of Endocrinology, Diabetology, Angiology, Nephrology,
Pathobiochemistry and Clinical Chemistry, Department of Internal Medicine, University
of Tbingen, Tbingen, Germany; Institute for Diabetes Research and Metabolic Diseases
of the Helmholtz Zentrum Mnchen at the University of Tbingen, Tbingen, Germany;
German Center for Diabetes Research (DZD), Duesseldorf, Germany
Contributors
xiii
xiv
Contributors
Contributors
xv
Chapter 1
Mechanical/Physical Methods of Cell Distribution
and Tissue Homogenization
Stanley Goldberg
Abstract
This chapter covers the various methods of Mechanical Cell Disruption and Tissue Homogenization that
are currently commercially available for processing minute samples (<1 mL) to larger production quantities.
These mechanical methods of lysing do not introduce chemicals or enzymes to the system. However,
the energies needed when using these harsh methods can be high and destroy the very proteins
being sought.
The destruction of cell membranes and walls by these harsh methods is effected by subjecting the
cells (1) to shearing by liquid flow, (2) to exploding by pressure differences between inside and outside of
cell, (3) to collision forces by impact of beads or paddles, or (4) a combination of these forces. Practical
suggestions to optimize each method, where to acquire such equipment, and links to reference sources
are included.
Key words Cell disruption, Bead mills, BioNeb cell disruption, Cell disruption vessel, Douce tissue
grinder, Dyno-Mill, French Press G-M, Gaulin high-pressure homogenizer, High-pressure homogenizers, Megatron, Microfluidics, Mixer-Mill, Mortar, Pestle, Nitrogen Parr vessel, Opposed jet
homogenization, Parr nitrogen vessel, Polytron, Potter-Elvehjem tissue grinders, Pressure vessel,
Sonicator, Sonitube, Tissue grinders, Tissue homogenization, Ultrasonic processor, Electro Water
Separation
Introduction
The need to release cell components without introducing encumbering
chemicals or enzymes suggests the use of mechanical methods of
lysing. The destruction of cell membranes and walls by these
harsh methods is effected by subjecting the cells (1) to shearing
by liquid flow, (2) to exploding by pressure differences between
inside and outside of cell, (3) to collision forces by impact of beads
or paddles, or (4) a combination of these forces.
Generally speaking, any of the techniques described here can,
to some degree, disrupt any cells or tissues. For more difficult
materials, just the increase of motivation force or time of exposure
will improve breakage. However, the use of excessive force is
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_1, Springer Science+Business Media New York 2015
Stanley Goldberg
Trade name(s)
Websites
www.RETSCH.de
www.BIOSPEC.com
DYNO-MILL [3]
www.WAB.ch
www.GLENMILLS.com
Rotor/statorshear
by spinning shaft
Polytron [4]
www.KINEMATICA.ch
Mortar/pestleshear by
mechanical pressure
Potter-Elvehjem tissue
grinders [5]
www.WHEATON.com
High-pressure batchliquid
expansion
www.GLENMILLS.com
High-pressure batchgas
expansion
www.PARRINST.com
High-pressure flowhigh
velocity liquid shear
www.SPX.com
High-pressureopposed
liquid streams
Microfluidizer [9]
www.MICROFLUIDICSCORP.com
Dropletlow pressure
flow droplet nebulizing
BioNeb [10]
www.GLASCOL.com
Ultrasonicshear by
collapsing bubbles
Sonicator [11]
Sonitube
www.SONICATOR.com
www.GLENMILLS.com
Electromotive force
Electro Water
Separation [12]
www.ORIGINOIL.com
Table 2
Suitable subjects and capacity for each method
Bacteria
Yeast,
Algae,
Fungus,
Spores
Seeds
Plants
Tissues
Capacity
S/M
M/L
S/M/L
Mortar/pestle shear
by mechanical pressure
S/M
S/M
S/M
S/M/L
S/M/L
S/M
S/M/L
M/L
Suitability: Ygeneral good practice; Nnot recommended; ?not known or marginal success
Quantities: S = small 0.125 mL; M = medium 10500 mL; L = 250 mL to many liters
2
2.1
Stanley Goldberg
Hints for successful temperature control include: (1) Prechilling of samples and containers; (2) Runs of short duration
with rest time to allow for re-chilling samples on ice; (3) Use
of fewer beads and/or extra buffer to act a heat sink; (4)
Reduced degree of shaking vigor.
3
3.1
Beads size: For small diameter cells (e.g. bacteria) use beads of
0.100.5 mm diameter. For larger cells (e.g. yeast, algae,
hyphae) use beads of 0.51.25 mm. Glass (sp. gr. 2.5) is a
good starting material due to low cost. For homogenizing
plant or animal tissues that have been previously chopped with
a razor, beads of 1.05.0 mm diameter are used.
Bead density: If additional energy is needed to improve breakage/homogenization of tough cells, then use higher density
materials. These material types include ceramic (zirconium
oxide family) with specific gravities from 3.8 to 6.0, stainless
steel of sp. gr. 7.0+, and tungsten carbide of sp. gr. 14.2+.
Also, the use of larger diameter beads from 2 to 20 mm can
improve breakage. Recently, success has been reported with
SiC grit with its sharp edges, and with stainless steel ballcones
that have a wedged edge at the equator.
For modest sample quantities of 50 mL and scaling up to industrial amounts of several thousand liters, the agitation of the beads
by a turning agitator within the vessel is the method of choice. In
this class of agitated bead mills, the beads and the cell suspension
are loaded into a chamber. Into the mix is placed one or more
spinning discs that accelerate the beads. The beads striking the
cells combined with the shearing by the moving liquid phase will
disrupt the cells.
Stanley Goldberg
These units are normally used for disruption of free microorganism cells (bacteria, yeasts, hyphae, mycelia) and not for tissue
samples. For lesser quantities, see previous section on shaking container method. Materials needed to operate the agitated bead mills
include the cells/tissue, grinding media (beads), liquid phase such
as buffer, cooling ice or jacket fluids, and the mill equipment.
Variables include the bead selection (density, diameter, and quantity), speed of agitation, cell concentration, and duration of run
(Figs. 3 and 4).
3.2 Practical
Aspects
4
4.1
RotorStator Homogenizer
Theory
Stanley Goldberg
Next choose the correct size generator for the sample volume
to be homogenized. Factory tables provide the volume ranges
suitable for each generator thus selecting the correct parts is
quite easy. For example, a sample of size from 0.5 mL to 5 mL
Interchangeable generators are designed to fit onto only certain motors. Therefore, one must decide which motor is the
most suitable for the particular range of applications and then
be sure to select only those generators designed to fit onto
that motor.
The homogenizers currently available can process sample volumes anywhere from less than 0.5 mL up to 150,000 L. Batch
equipment (e.g. Polytron) is used for small sample quantities
from less than 0.5 mL though larger units can handle several
hundred liters. For larger industrial in-line system for either
continuous or re-circulation flow, there are single-, double-, or
even triple-stage rotor/stator configurations (Megatron).
10
6
6.1
Stanley Goldberg
There are two widely used cell disruption methods that employ
rapidly expanding fluids from within the cell to explode the cell
membranes. The French Press G-M uses liquid under pressure,
and the Parr cell disruption vessel uses compressed gases. Since
these are bath operations, they are only suitable for small quantities
of less than about a liter.
The Parr cell disruption vessel is a pressure vessel into which the
sample to be disrupted is placed along with a dip tube fitted with
an exit valve. Suitable gas such as nitrogen at 2 kpsi is forced into
the vessel and dissolved into the cells. When the exit valve is opened
the gas pressure is suddenly released causing the nitrogen to come
out of the solution within the cells as expanding bubbles. This
action stretches the membranes of each cell until they rupture and
releases the contents of the cell. Although sometimes referred to as
explosive decompression, nitrogen decompression is actually a
gentle method.
This method is suited for treating mammalian and other
membrane-bound cells, for treating plant cells, for releasing virus
from fertilized eggs, and for treating fragile bacteria. It is not recommended for untreated bacterial cells, unless using various
pretreatment procedures to weaken the cell wall. Yeast, fungus,
spores, and other materials with tough walls do not respond well
to this method (Figs. 8 and 9).
6.2 Practical
Aspects
Removal of air prior to closing the exit valve will minimize the
amount of oxygen degradation of the released proteins.
11
12
Stanley Goldberg
7
7.1
13
7.1.1 High-Pressure
Valve with Impingement
Wall: Gaulin
7.1.2 High-Pressure
Flow Narrow Tubes
or Opposed Jets:
Microfluidics
14
Stanley Goldberg
15
stream of pressurized cell suspension fluid into two legs. These are
then directed at one another in an interaction chamber where they
collide, disrupting the cells.
This equipment is suitable for a variety of free cells (bacteria,
yeasts, mammalian cells), but not seeds, tissue samples, nor plant
materials. The design of identical fluid channels in both laboratoryscale and large-scale units allows for direct scale-up from the smallest laboratory unit (14 mL batch) directly to large production
units (tens of L/min) (Fig. 12).
7.1.3 Practical Aspects
Using High-Pressure Valve
with Impingement Wall:
Gaulin
16
Stanley Goldberg
Most models are autoclavable and air driven, though some are
electric-hydraulic systems. In many circumstances, especially when
samples are processed multiple times, the Microfluidizer processors require sample cooling before and/or after processing.
9
9.1
The use of sound waves in fluids can disrupt cells. The operation
starts with normal electrical current (50 Hz or 60 Hz) being transformed to 20,000 Hz. This electrical signal is fed to a piezo-electric
crystal causing it to oscillation at this high frequency. The vibrations move a titanium metal HORN about 515 m. The shape of
the horn amplifies this motion to 100150 m/cycle. By placing
the horns endthe tipinto fluid, the tip moves the liquid
17
forward (away) and then retracts (back) quicker than the liquid can
return. During the return stroke, the pressure in the system drops
below the vapor pressure of the liquid so boiling occurs (cavitation). As the liquid flows back, the bubbles collapse. This bubble
collapsing imparts the energy needed to disrupt the cells (Fig. 14).
9.2 Practical
Aspects
18
Stanley Goldberg
10
10.1
10.2 Practical
Applications
Acknowledgements
Superb assistance was rendered by the following people who are
most conversant in their noted equipment areas. Bead Milling
by Tim Hopkins (Biospec Products) and Harald Frommherz
19
Electrical Pulses
Contaminated
Water
Clear
Water
Electro-Coagulation
Electrical Pulses
20
Stanley Goldberg
Chapter 2
Sample Preservation Through Heat Stabilization
of Proteins: Principles and Examples
Mats Born
Abstract
Due to post-sampling changes, caused by residual enzyme activity in the sample, levels of analytes can
change from their in vivo levels so that they no longer accurately reflect conditions in the living system.
The Stabilizor system accomplishes elimination of enzyme activity through heat-induced denaturation of
enzymes by permanently altering the 3D protein structure of the enzymes. Heat stabilization can be introduced in the workflow either directly after sampling, with the instrument just next to where the sample is
taken, or prior to sample homogenization and extraction, when samples are heat denatured directly from
a frozen state. Initially, heat stabilization was developed to enable mass spectrometric analysis of neuropeptides. Heat stabilization has since been further developed and applied to a range of samples and downstream protein analysis techniques such as western blot, 2D gels and phosphorylation analysis with LC-MS.
Key words Heat inactivation, Post-sampling change, Phosphorylation, Post-translational modification,
Brain, Neuropeptides
Introduction
Biosampling for subsequent analysis of molecular biomarkers is a
corner stone in modern medicine and biological research.
Measured levels of various analytes are the basis for diagnosis and
the overall understanding of biology. It is thus of uttermost
importance that measured levels accurately reflect actual in vivo
levels as close as possible. Due to post-sampling changes, caused
by residual enzyme activity in the sample, levels of analytes can
change from their in vivo levels so that they no longer accurately
reflect conditions in the living system. In the living organism, levels of analytes are tightly controlled by cellular signaling and
homeostasis is maintained. When a sample is removed from its
natural environment, a dramatic signaling cascade is initiated. In the
case of a tissue biopsy the removal results in loss of blood flow, which
subsequently leads to low oxygen levels and a switch to anaerobic
metabolism. This results in low energy levels and a drop in sample pH.
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_2, Springer Science+Business Media New York 2015
21
22
Mats Born
All these changes, low oxygen, low ATP and low energy are signals
which are sensed on a cellular and protein level and results in an
active response from the still living cells in the sample. This results
in changes in protein levels as well as post-translational modifications (PTMs). This is a very fast process which initiates within
seconds of sampling and leads to measurable changes [1]. In addition
to the initial response, primarily an active regulated response,
which takes place from the moment of sampling until stabilization,
by freezing or heat inactivation, there is a second response, mainly
reactive in nature, which occurs during sample preparation due
to residual enzyme activity in the homogenate [2]. During this
phase, enzymes released from their cellular confinement and controls are free to interact with whatever substrates they encounter.
Both phases of enzyme driven change post-sampling must be
adequately addressed and minimized in order to successfully
measure in-vivo relevant levels of analytes.
The standard way of addressing the problem of enzymatically
driven post-sampling changes have been to add chemical inhibitors
and work fast while keep the samples cool during homogenization and
extraction. This is only partly successful as enzyme inhibitors are
selective and reduce enzyme activity rather than eliminate it [3].
A few years ago, an alternative approach, to preserving sample
quality in protein studies, was introduced, the Stabilizor system [3].
The Stabilizor system accomplishes elimination of enzyme activity
through heat-induced denaturation of enzymes by permanently
altering the 3D protein structure of the enzymes. In contrast to
chemical inhibitors, heat-induced enzyme denaturation is a general
principle affecting all protein-modifying enzymes, virtually reducing their activity to zero in the stabilized sample. Heat stabilization
can be introduced in the workflow either directly after sampling
with the instrument present just next to where the sample is taken
or just prior to sample homogenization and extraction when samples are heat denatured directly from a frozen state.
Heat stabilization has been applied to a range of samples and
downstream analysis techniques. It has proven itself as a valuable
tool in sample preparation for protein analysis. Initially, heat stabilization was developed to enable mass spectrometric analysis of
neuropeptides. The applicability has since been expanded and it
has been advantageously applied to a diverse array of protein analysis
techniques including western blot with phospho-specific antibodies,
top-down 2D PAGE, bottom-up phospho-shot gun, and MALDI
imaging [4]. As the Stabilizor system induces protein denaturation
and loss of protein 3D structure it is not suitable for sample preparation prior to analysis of enzyme activity, multi-protein complexes
or formalin-fixed epitopes, e.g. immuno-histochemistry. Using
western blots, stabilization of protein phosphorylation levels has
been shown. Flash-frozen and heat-stabilized samples either frozen
directly or left at room temperature for 30 min were compared.
23
Fig. 1 Quantitation of phospho-proteins in hippocampal lysates shows that heat-stabilized levels remain
unchanged while frozen tissue levels are decreased with 30 min room temperature incubation. Signals from
replicate Western blots were normalized to actin. Histograms represent average values for each group of
samples relative to 100 % for frozen tissues at 0 min RT (white bars); black bars, stabilized tissues 0 min at
RT; grey bars, frozen tissues 30 min at RT; striped bars, stabilized tissues 30 min at RT. Errors bars are the
standard error of the mean. Statistical significance was determined by the unpaired Students t-test. *p < 0.05;
**p < 0.001; ***p < 0.0001. Reprinted from J. Neurosci. Methods, 196(1), 99106. Ahmed, MM., and Gardiner,
KJ. Preserving protein profiles in tissue samples: differing outcomes with and without heat stabilization (2011),
with permission from Elsevier
24
Mats Born
Fig. 2 2-DE analysis of differentially expressed proteins from stabilized or snap-frozen mouse cortex. Spots
with differential intensity between the two samples are highlighted in color: higher intensity spots in heatstabilized samples (green), higher intensity spots in snap-frozen samples (red and blue). Numbers refer to
protein spots that were subjected to MS analysis. Reprinted from Proteomics, 9(19), 443344. Robinson, AA.,
Westbrook, JA., English, JA., et al., Assessing the use of thermal treatment to preserve the intact proteomes of
post-mortem heart and brain tissue. (2009), with permission from Wiley
Materials
2.1 The
Stabilizor System
25
Fig. 3 Phosphorylation states of murine PPIA isoforms in (a) stabilized and (b) unstabilized homogenates and
ATP synthase subunit a in stabilized (c) and unstabilized (d) homogenates, indicate a shift in phosphorylation
levels between isoforms between stabilized (a & c) and unstabilized (b & d) samples. In the unstabilized
sample (d), progressive phosphorylation of ATP synthase subunit a (21) resulted in a 550 % increase in the
higher phosphorylation state (40). The sum of mean spot volumes (62, 66, 79) were nearly identical in stabilized and unstabilized samples, respectively, as well as the sum of the mean volumes for spots 40, 53, and 21
were nearly identical in stabilized and unstabilized samples indicating that there were no preferential loss of
protein from either sample. Experimental pI values are provided on the abscissa. Reprinted from Electrophoresis,
32(16), 22062215, Smejkal, GB., Rivas-Morello, C., Chang, JH., et al., Thermal stabilization of tissues and the
preservation of protein phosphorylation states for two-dimensional gel electrophoresis (2011) with permission
from Wiley
26
Mats Born
Fig. 4 Percent distribution of ratios of median intensity of 1,107 peptides with tyrosine phosphorylation
between snap-frozen (SF) and heat-stabilized (Stabilized) samples isolated from mice brain tissue. 34 % of
peptides are detected at intensities at least 50 % higher or only in stabilized samples whereas only 4 % are
detected at intensities at least 50 % higher or only in snap-frozen samples, indicating a preservation of phosphorylation levels in stabilized samples. Mice brains were collected in triplicates and either just snap-frozen
or heat-stabilized using the Stabilizor system prior to freezing. Proteins were extracted using a urea-based
extraction buffer with protease and phosphatase inhibitors and turned into peptides using trypsin. Peptides
with tyrosine phosphorylations were affinity-enriched using a tyrosine-specific antibody (Cell Signaling,
#9411). Peptide intensities were analyzed using LC-MS/MS
Methods
Enzyme inactivation using heat-induced enzyme denaturation is a
preparative step prior to a large range of protein as well as small
molecule analysis techniques. Heat inactivation in the Stabilizor
T1 is performed using a number of predefined programs depending on the state of the sample, fresh or frozen, as well as the need
to preserve sample morphology. Once the sample has been heatstabilized, the use of correct extraction buffers and homogenization
is vital in order to resolubilize proteins and enable analysis. In this
section, the standard ways for using the Stabilizor T1 instrument
will be covered. Special considerations for homogenization and
resolubilization of heat-inactivated samples will be covered in
Subheading 3.3.
3.1 Heat
Stabilization of Fresh
Samples Directly
After Removal
from the Source
27
3.2 Heat
Stabilization
of Frozen Samples
28
Mats Born
3.3.1 Denaturing
Extraction Buffer
3.3.2 Buffer
to Sample Ratio
29
3.3.4 Homogenization
of Tissue in Hot 1 % SDS
Buffer Using Micro pestle
Grinding Followed
by Heating and Microtip
Rod Sonication
30
Mats Born
3.4 Analysis
of Extracted HeatStabilized Samples
Notes
1. Denaturing agents of buffers should be >1 % SDS or >8 M
urea. In addition to denaturing agents other components can
be added, e.g. detergents and salts, depending on the needs of
the specific work flow, the SDS and/or urea contents may
however not be lowered.
2. Use dust mask to avoid getting powdered SDS into the lungs
during weighing. It is also a good idea to make a 10 % SDS
stock solution to use for further dilutions to minimize the
number of times powdered SDS have to be handled.
3. As urea dissolves the solution becomes cold. Do not heat to
speed up solubilization as this may create isocyanic acid which
will cause protein carbamylation in later steps. Isocyanic acid
can also form naturally as the urea buffer ages. To avoid this,
always prepare urea containing extraction buffers fresh prior to
use.
4. Both Fresh methods, Quick and Structural preserve, will inactivate enzymes in the sample. The Quick method will compress
the sample to make it thinner, giving a faster heating throughout the sample. The Structure preserve on the other hand use
minimal compression to preserve sample structure. The sample
is thus thicker and stabilization takes longer time.
5. The Post mortem clock is basically a timer keeping track of the
time between sacrifice and stabilization. It is important to try
to keep this time as short and comparative between samples as
possible. The use of the timer helps to focus attention on the
dissection and avoids unnecessary delays. As dissection is an art
which needs practice it is quite common for the dissection time
to decrease by ~50 % between the first and the fifth animal
before leveling out. It is thus good practice to either start with
control animals not part of the study or a group where time is
not as important.
31
32
Mats Born
References
1. Skld K, Alm H, Scholz B (2013) The impact
of biosampling procedures on molecular data
interpretation. Mol Cell Proteomics 12(6):
14891501
2. Stingl C, Sderquist M, Karlsson O et al (2014)
Uncovering effects of ex vivo protease activity
during proteomics and peptidomics sample
extraction in rat brain tissue by oxygen-18
labeling. J Proteome Res 13(6):28072817
3. Svensson M, Born M, Skld K et al (2009)
Heat stabilization of the tissue proteome: a new
technology for improved proteomics. J Proteome
Res 8(2):974981
4. Kultima K, Skld K, Born M (2011) Biomarkers
of disease and post-mortem changesheat stabilization, a necessary tool for measurement of
protein regulation. J Proteomics 75(1):
145159
5. Ahmed MM, Gardiner KJ (2011) Preserving
protein profiles in tissue samples: differing out-
6.
7.
8.
9.
Chapter 3
Isolating Peripheral Lymphocytes by Density
Gradient Centrifugation and Magnetic Cell Sorting
Frederic Brosseron, Katrin Marcus, and Caroline May
Abstract
Combining density gradient centrifugation with magnetic cell sorting provides a powerful tool to isolate
blood cells with high reproducibility, yield, and purity. It also allows for subsequent separation of multiple
cell types, resulting in the possibility to analyze different purified fractions from one donors sample.
The centrifugation step divides whole blood into peripheral blood mononuclear cells (PBMC), erythrocytes, and platelet-rich plasma. In the following, lymphocyte subtypes can be consecutively isolated from
the PBMC fraction. This chapter describes enrichment of erythrocytes, CD14-positive monocytes and
CD3-positive T lymphocytes. Alternatively, other cell types can be targeted by using magnetic beads
specific for the desired subpopulation.
Key words Peripheral blood mononuclear cells (PBMC), Magnetic-activated cell sorting (MACS),
Fluorescence-activated cell sorting (FACS)
Introduction
Isolation of blood cells for research purposes should ensure optimal
experimental conditions by providing maximum specificity, purity,
yield, speed, and reproducibility [1, 2]. Today, numerous methods
for enrichment of different cell types exist, the most common
being density gradient centrifugation and several variants of antibody-based immunoaffinity methods [3, 4]. Density gradients are
typically used to fractionate full blood into platelet-rich plasma,
erythrocytes, and peripheral blood mononuclear cells (PBMCs)
composed of lymphocytes, monocytes, and macrophages [5, 6].
Cell (sub)types can be efficiently obtained by use of immunoaffinity purifications [4]: These methods make use of antibodies
targeting characteristic proteins like cell surface receptors, for
example to attach labels for instrumental supported isolations like
fluorescence-activated cell sorting (FACS) [7]. Likewise, antibodies are immobilized on supporting material with properties
suited for enrichment methods (e.g., magnetic cell sorting, MACS).
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_3, Springer Science+Business Media New York 2015
33
34
Density gradient
centrifugation
Platelet-rich plasma
PBMCs
Pancoll
Erythrocytes
PBMCs
MACS
CD14+ MACS
CD14+ Monocytes
Erythrocytes
CD3+ MACS
CD3+ T-Lymphocytes
FACS
Fig. 1 Overview of workflow. He isolation procedure begins with a density gradient centrifugation which fractionates whole blood into erythrocytes, PBMCs, and platelet-rich plasma. Fractions of intereste.g., erythrocytescan be preserved for analysis directly after the gradient. The PBMC fraction can be further subdivided
by subsequent cell type-specific magnetic cell sorting, as shown in this protocol for CD14-positive monocytes
and CD3-positive T lymphocytes
35
Materials
2.1 Technical
Equipment
2.1.1 General
1. 20 mL syringes.
2. Butterfly or similar needle.
3. Adapters to connect needle to syringes.
36
15. Micropipettes.
16. Sterile micropipette tips.
17. Automatic cell counter or microscope with Neumann chamber.
2.1.5 Washing of Purified
Monocytes and
T Lymphocytes
1. Centrifuge for 15 and 50 mL tubes, swing bucket rotor, adjustable acceleration and breaking, cooling option.
2. Sterile serological pipettes.
3. Pipetting device or Peleus ball for serological pipettes.
4. Sterile 15 and 50 mL conical centrifuge tubes.
5. Stand for centrifuge tubes.
6. Table centrifuge for 1.5 mL reaction tubes, cooling option.
7. 1.5 mL sterile polypropylene reaction tubes.
8. Stand for reaction tubes.
9. Micropipettes.
10. Sterile micropipette tips.
2.2 Buffers
and Solutions
2.2.1 General
2.2.2 Blood Sampling
1. Anti-CD14-FITC.
2. Anti-CD3-PE.
3. Anti-CD19 FITC.
4. Anti-CD56 PE.
3
3.1
37
Methods
Blood Sampling
3.2
Density Gradient
Diluted
blood
Pancoll
Fig. 2 Preparation of density gradient. To prepare the density gradient, begin by placing a small volume of
diluted blood as slow as possible on top of the Pancoll at the edge of the centrifuge tube. Stabilize the pipette
by pressing it at the edges of the centrifuge tube. Wait until the blood has distributed over the Pancoll, then
slowly move the pipette up while continuously letting the diluted blood flow out. Avoid too fast outlet of the
blood as this will cause disturbances in the gradient, resulting in poor performance
38
3.4 Washing
of PBMCs
39
3.6 Washing
of Purified Monocytes
and T Lymphocytes
40
1. For each isolated fraction (PBMCs, Monocytes, T lymphocytes), label 3 FACS vials as follows:
(a) Non-labeled control
(b) CD14 FITC + CD3 PE
(c) CD19 FITC + CD56 PE
2. Pipette 200,000 cells of the respective isolated fraction in
each vial.
3. Incubate cells with FACS antibodies according to the vial
labels and manufacturers instructions.
4. Measure on a flow cytometer according to the manufacturers
instructions (see Note 11).
5. Determine composition of fractions by comparing ratios of
CD14+ (monocytes), CD3+ (T lymphocytes), CD19+ (B lymphocytes), and CD56+ (NK cells). The isolation of monocytes
and T lymphocytes by MACS should provide purities > 85 %
(Fig. 3).
Notes
1. The MACS buffer is best prepared on the day before the cell
isolation.
2. The described protocol was originally designed for large yields
for proteomics experiments without further culture of the cells
and is therefore based on a large blood donation of 80 mL [9].
The procedure is adaptable to any other volume by proportionally adjusting volumes of Pancoll and MACS beads.
Magnetic cell sorting can also be performed directly from full
blood, but the density gradient will help to pre-purify and concentrate the PBMCs in a small volume for MACS isolation.
3. The appropriate blood sampling system/adapters should be
chosen by authorized and experienced medical personal.
4. Cold Pancoll has a different density than warm Pancoll
which can cause inefficient separation in density gradient.
Remove Pancoll from freezer before blood sampling is performed to ensure that there is no time delay between sampling
and gradient.
41
PBMC-Fraction
CD3+
FL2-Height
FL2-Height
CD3+
102
101
unlabeled
cells
0.2%
59.1%
103
28.3%
12.4%
CD14+
100
100
101
102
Unlabeled
cells
4.0%
0.4%
3.1%
92.5%
103
102
101
CD14+
100
100
103 104
101
102
103
104
FL1-Height
FL1-Height
MACS
CD3+ Fraction
FL2-Height
CD3+
94.9%
0.4%
2.4%
2.3%
103
102
101
CD14+
Unlabeled
cells
100
100
101
102
103 104
FL1-Height
Fig. 3 Purity control by FACS. Lymphocyte fractions analyzed by FACS. In fractions isolated from PBMCs by
MACS, proportions of the selected cells are drastically increased: In this example, the PBMC fractions consist
of 46 % of CD3+ cells (T lymphocytes) and 3 % of CD14+ cells (monocytes). By MACS sorting, these proportions are increased to 88 % (CD14+ fraction) and 95 % (CD3+ fraction)
5. This step is necessary to adjust the density of the blood for the
gradient.
6. Fixed angle rotors and strong acceleration or deceleration will
cause inefficient separation in gradient.
7. When collecting the PBMCs, include parts of the supernatant
and Pancoll to ensure complete harvest of the PBMCs. The
following washing and magnetic cell isolation will remove the
contaminants.
8. Erythrocytes will not form a pellet, but a concentrated
suspension.
9. This step can be performed together with step 2 in
Subheading 3.4.
10. The necessary amount of cells for FACS analysis depends on
the used instrument and measurement protocols.
11. FACS analysis should be performed in a lab with high experience
in analysis of mononuclear cells.
42
Acknowledgement
This work was supported by P.U.R.E. (Protein Unit for Research
in Europe), a project of Nordrhein-Westfalen, a federal state of
Germany, the Bundesministerium fr Bildung und Forschung
(NGFNplus, FZ 01GS08143), Germany, and the German Center
for Neurodegenerative Diseases (DZNE e.V.) within the Helmholtz
Association.
References
1. Blonder J, Issaq HJ, Veenstra TD (2011)
Proteomic biomarker discovery: its more than
just mass spectrometry. Electrophoresis 32:
15411548
2. Mallick P, Kuster B (2010) Proteomics: a pragmatic perspective. Nat Biotechnol 28:695709
3. Bennett S, Breit SN (1994) Variables in the
isolation and culture of human monocytes that
are of particular relevance to studies of HIV.
J Leukoc Biol 56:236240
4. Dainiak MB, Kumar A, Galaev IY et al (2007)
Methods in cell separations. Adv Biochem Eng
Biotechnol 106:118
5. Syrovets T, Tippler B, Rieks M et al (1997)
Plasmin is a potent and specific chemoattractant
for human peripheral monocytes acting via a
cyclic guanosine monophosphate-dependent
pathway. Blood 89:45744583
Chapter 4
Investigating the Adipose Tissue Secretome: A Protocol
to Generate High-Quality Samples Appropriate
for Comprehensive Proteomic Profiling
Simon Gddeke, Jorg Kotzka, and Stefan Lehr
Abstract
In this chapter, we describe in detail how to prepare a sample containing the complete entity of secretion
products from murine primary adipocytes, which are suitable for comprehensive and sensitive secretome
analysis. The underlying protocol should be seen as a starting point guiding through critical steps of the
complex workflow in order to approximate to the real secretome in the context of different sample types
used for the diverse research questions the protocol has to be carefully adjusted.
Key words Secretome analysis, Adipokines, Murine adipose tissue, Pre-adipocytes
Introduction
Obesity, which is favored by imbalanced energy supply and
energy consumption coupled with a sedentary lifestyle is considered as an epidemic disease and represents a burden for almost
all societies [1]. Therefore, overweight today is the major risk
factor for developing various metabolic complications such as
insulin resistance, type 2 diabetes, non-alcoholic liver disease,
and cardiovascular diseases [26].
Over the last decade we have learned that adipose tissue, besides
its predominant role within energy homeostasis also represents a
major endocrine organ, releasing a wide variety of signaling and
mediator proteins. These so-called adipokines seem to be causally
involved in the development of a wide variety of metabolic diseases.
Over the last years, several attempts have been made to characterize
the adipokines by utilizing diverse proteomic profiling approaches.
This has led to a catalog of adipokines comprising several hundred
potentially secreted peptides and proteins [7]. Nevertheless, the limited overlap of identified adipokines in published studies impressively
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_4, Springer Science+Business Media New York 2015
43
44
Materials
2.1 General
Hardware
and Consumables
2.2
Tissue Biopsies
2.3 Isolation
of Pre-adipocytes
45
3. Mesh, 75 m.
4. Syringe filters, 25 m.
5. Special accuracy scale.
6. Collagenase (NB8, Clostridium Histolyticum).
7. Phosphate buffered saline (PBS) w/o Calcium and Magnesium,
sterile.
8. Collagenase solution (750 U collagenase/g adipose tissue):
250 U/ml in HBSS (pH 7.4) (see Note 1) supplemented with
3 mM CaCl2 (see Note 2) and 4 mM glucose. Sterile filtration
(10 ml syringe), G26 cannula (1), sterile syringe filters
(0.2 m, 33 mm).
9. HBSS to dilute the collagenase: add CaCl2 (3 mM).
10. Erythrocyte lysis buffer: 8.29 g/l NH4Cl, 0.99 g/l K2HPO4,
0.04 g/l EDTA, pH 7.3 sterile filtration.
11. Basal medium: DMEM/F-12 (17.5 mM glucose), 1.25 g/l
NaHCO3, 16 mg/l Biotin, 8 mg/l Calcium-D-pantothenate,
pH 7.3, sterile filtration. Before use add 5 ml/l Gentamycin.
12. Basal medium w/o phenol-red: DMEM, w/o phenol red,
w/o FCS, 10 pmol/ml insulin. Before use add 5 ml/l
Gentamycin.
2.4 Differentiation
of Pre-adipocytes
46
1. Oil Red O stock solution: Mix stock solution in 15 ml PP centrifugation tubes with ddH2O (6:4, v/v) and incubate for at
least 2 h at room temperature in the dark. Filtrate the solution
before use and centrifuge for 10 min at 3,200 g.
2. Hemalaun solution: Dissolve Hematoxylin in ddH2O (1 mg/ml)
and add sodium iodate (0.2 mg/ml) and potassium sulfate
(50 mg/ml) while agitating (see Note 5). After mixing add
chloride-hydrate (50 mg/ml) and crystalline citric acid (1 mg/ml)
(see Note 6).
3. Fixation solution: Mix 15 ml Picric acid with 5 ml Formol
(37 %) and 1 ml acetic acid (100 %).
4. Oil Red O stock solution: 0.3 g Oil Red O solve in 100 ml
isoproanol (99 %) (see Note 7).
2.6 Collecting
Secreted Peptides/
Proteins
1. Mesh, 50 m.
2. Ethylendiaminetetraacetic acid (EDTA) solution pH 8.0
(1 mg/ml).
3. Phosphate buffered saline (PBS) w/o Calcium and Magnesium,
sterile.
2.7 Concentration
via Centrifugal Filter
Concentrator
Methods
In this chapter we describe in detail how to prepare a specimen,
containing the complete entity of secretion products from murine
primary adipocytes, which are suitable for comprehensive and sensitive secretome analysis. The underlying protocol should be seen
as a starting point guiding through critical steps of the complex
workflow in order to approximate to the real secretome in the context of different sample types used for the diverse research questions the protocol has to be carefully adjusted (Fig. 1).
3.1
Tissue Biopsies
47
Fig. 1 Workflow scheme to generate secretome sample from differentiated mouse adipocytes
3.2 Isolation
of Pre-adipocytes
(Figs. 2, 3, and 4)
shaking
48
Fig. 3 Adipocyte-free pellet before (left) and after erythrocyte lysis (right; the
pellet should be white and free of erythrocytes)
49
Oil Red O protocol is an assay performed to detect mature adipocytes in histological visualization of fat cells by staining neutral fat
1. Soak off the culture medium and wash the cells very gently
with 1 PBS two times.
2. Discard the supernatant by aspiration and add the fixationsolution.
3. Incubate at RT for 2 h (see Note 19).
4. Discard the fixation solution and wash with 1 PBS two times
(see Note 20).
5. Accordingly incubate the cells in 40 % isopropyl alcohol for
5 min.
6. Discard the isopropyl-solution and add the Oil Red O solution.
50
51
Fig. 6 Visualization and quality control of secretome samples using 2-dimensional gel electrophoresis. Aliquots
(40 g protein each) of (a) secretome of differentiated, primary adipocytes, (b) total lysate of differentiated
primary adipocytes and (c) Fetal Calf Serum (FCS). Samples were separated by 2-dimensional gel electrophoresis according to the 2D-ToGo workflow [9] standard operation procedure established in our group. Accordingly,
in the first dimension samples in ReadyPrep Sample Buffer (8 M urea, 3 % CHAPS, 50 mM DTT, 0.2 % (w/v)
Bio-Lyte 3/10 ampholytes) were separated by isoelectric focusing (IEF) using pH 310 linear ReadyStrip
IPGstrips (pH 310, 11 cm) performed on a Protean i12 electrophoresis unit (Bio-Rad) and in the second
dimension by SDS-PAGE using Criterion TGX Stainfree Any KD precast gels (Bio-Rad). After electrophoretic separation spot pattern were acquired and documented without further staining, using a ChemiDoc MP
(Bio-Rad) equipped with Image Lab Software
3.6 Concentration
via Centrifugal Filter
Concentrator
Notes
1. Consider the unique activity of each collagenase-charge: e.g.
PZU activity of 1.0 = 1,000 U/mg. When weighing the collagenase wearing a surgical mask is recommended.
2. The CaCl2 is crucial for activation of the collagenase.
3. Do not use this medium for more than a week.
4. TZO must be diluted in DMSO, not in aqueous solvent.
5. The solution should get a violet-blue staining.
52
6. The mixture should get a red-violet staining, filtrate the solution with a 150 m paper filter before use.
7. Only use the solution for 12 weeks until precipitation.
8. The buffer has to be freshly prepared, sterile filtered and stored
on ice.
9. For rough dissection the use of a forceps and medical scissors
is recommended. Therefore, you mince the adipose tissue, e.g.
in a weighing pan.
10. Collagenase mixture = tissue/collagenase solution (1:1, v/v).
11. In all of the following centrifugation steps, the use of a swingout rotor is necessary to ensure adequate separation of the
fractions.
12. At this point harvested adipocytes can be used for further analysis. On the one hand, it is possible to generate secretome from
the floating mature adipocytes and furthermore they can be
used for Western blot and qPCR analyses.
13. Do not use a solution of more than 0.5 M of EDTA, because
otherwise cells tend to clot. Using serum/FCS is not efficient
enough to stop the collagenase reaction.
14. This step is crucial to remove contaminating erythrocytes. It is
recommended to monitor the reaction, because ammonium
chloride might damage the cells when incubating too long.
15. Both filtering steps are needed to isolate the pre-adipocytes,
because using only a single step/mesh might end up with
plugging of the mesh and less yield of pre-adipocytes.
16. Never centrifuge for more than 10 min, because the cells are
destroyed otherwise.
17. If not planned to work on parallel with the mature adipocytes.
18. This medium was changed every 23 days, until full
differentiation.
19. This incubation step can be elongated up to 24 h if needed.
20. Wash until the yellowish dye of the washing buffer is extincted.
21. The incubation time depends upon the staining. Better monitor with microscope.
22. For visualizing of the nucleus cells can stained with hemalaun.
23. Use enough water to cover all the cells. Use tap water to ensure
presence of HO-ions which activate the metalhematoxylin
complex. The incubation will probably take longer than 10 min
until blue staining.
24. The addition of CaCl2 is crucial to wash away the FCS totally.
This enables a subsequent 2D gel analysis. Wash very carefully,
because attachment of cells is not very strong.
53
6.
7.
8.
9.
syndrome: contribution to global cardiometabolic risk. Arterioscler Thromb Vasc Biol 28:
10391049
Hauner H, Petruschke T, Russ M et al (1995)
Effects of tumour necrosis factor alpha (TNF
alpha) on glucose transport and lipid metabolism of newly-differentiated human fat cells in
cell culture. Diabetologia 38:764771
Lehr S, Hartwig S, Sell H (2012) Adipokines: a
treasure trove for the discovery of biomarkers
for metabolic disorders. Proteomics Clin Appl
6:111
Brown KJ, Formolo CA, Seol H et al (2012)
Advances in the proteomic investigation of the
cell secretome. Expert Rev Proteomics 9:
337345
Posch A, Franz T, Hartwig S et al (2013)
2D-ToGo workflow: increasing feasibility and
reproducibility of 2-dimensional gel electrophoresis. Arch Physiol Biochem 119:108113
Chapter 5
Methods for Proteomics-Based Analysis of the Human
Muscle Secretome Using an In Vitro Exercise Model
Mika Scheler, Martin Hrabe de Angelis, Hadi Al-Hasani,
Hans-Ulrich Hring, Cora Weigert, and Stefan Lehr
Abstract
Over the last decade, the skeletal muscle as a secretory organ gained in importance. A growing number of
peptides are described which are produced and released by the muscle fibers and work in an autocrine,
paracrine, and endocrine fashion. The contraction-induced secretion of these myokines is considered to
contribute to the health-promoting effects of exercise. To gain further insights into the molecular processes that occur during contraction an in vitro exercise model, electric pulse stimulation (EPS), was established. Recent publications show that this model is suitable to electro-stimulate human skeletal muscle cells
and thus mimic muscle contraction in vitro. Here, we provide a detailed protocol for the proteomics-based
analysis of the human muscle secretome, starting with the cultivation of human myotubes and their electric
pulse stimulation, ending with sample preparation for targeted and untargeted proteome analysis of the
cell culture supernatant. This whole workflow should allow deeper insights into the complex nature of the
muscle secretome and the identification of new myokines which might help to understand the crosstalk of
the working muscle with different organs and the beneficial effects of exercise.
Key words Secretome, Electric pulse stimulation, Myokine, Human myotubes, 2D-DIGE, LC-MS
Introduction
It is commonly accepted that regular physical activity entails
multiple health-promoting effects, plays a key role in the prevention and treatment of metabolic and cardiovascular diseases [1]
and reduces systemic inflammation [2]. For decades, it was hypothesized that circulating factors are produced and released from the
contracting muscle and enable the communication between the
energy-demanding muscle and the energy-supplying organs like
adipose tissue and liver during exercise [3]. Interleukin 6 (IL-6)
was first discovered as such an exercise factor when a higher
expression and release of this cytokine was detected in human
skeletal muscle after contraction [4] and the putative function of
IL-6 as enhancer of endogenous glucose production in exercising
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_5, Springer Science+Business Media New York 2015
55
56
57
Materials
2.1 General
Consumables
2.2
Cell Culture
2.4 Targeted
Proteomics
2.5 Untargeted
Proteomics
58
3
3.1
Methods
Cell Culture
Fig. 1 (continued) myotubes (immunostaining shows MyHC II, a fast-type skeletal muscle myosin in green;
nuclei are shown in blue (DAPI staining)). 2. After 67 days of differentiation myotubes are electro-stimulated. Therefore, the electrodes dip into the media in the cell culture dish and the whole sandwich is put into
the incubator. A cable connects the electrode (C-dish) with the power supply (C-Pace EP system) outside (after
IonOptix, Dublin, Ireland; detailed brochure: Culture Pacing System). 3. For targeted analysis of the muscle
secretome small amounts of media are sufficient whereas untargeted analysis needs larger amounts of
media which should be free of FBS and phenol red
59
1. Cell culture
Satellite cell
isolation
Myoblasts
Myotubes
Power supply
2. EPS
Incubator
3. Proteomics
Normal
Fusion media
Targeted immunoassay
Fusion media
phenolred
FCS
Untargeted approach
Separation
(gel-based vs. gel-free)
MS-based analysis
for protein identification
Fig. 1 Workflow showing secretome analysis of human myotubes. 1. Skeletal muscle cells are obtained from
percutaneous needle biopsies performed on the lateral portion of quadriceps femoris (vastus lateralis) of
human subjects. Satellite cells are prepared, myoblasts are cultivated and differentiated to multinucleated
Absorbance 450nm
(fold change)
b 250
1.2
CK activity (U/L)
60
1
0.8
0.6
0.4
0.2
200
15
10
5
0
0
con
con
EPS
EPS
SN
250
con
EPS
lysate
200
15
10
5
0
con
EPS
SN
con
EPS
lysate
Fig. 2 EPS does not induce cytotoxicity. Cell viability was measured by XTT cell viability assay (a). Measured
absorbance at 450 nm of supernatant is shown as fold change of EPS vs. control supernatant (means SEM;
n = 5). Creatine kinase (CK; b) and Lactate dehydrogenase (LDH; c) activities were measured in the supernatant (SN, filled bars) and in Triton X-100 lysates of the cells (striped bars). Values are shown in U/L (means SEM;
n = 58; p < 0.05) adapted from [11]
3.3 Collection
of Medium
for Targeted
Secretome Studies
Usually cell culture medium contains FBS to stimulate cell proliferation or in case of serum-reduced media like Fusion media to
maintain the viability of the cell. The high abundance of serum
proteins in the FBS requires additional enrichment of low-abundant
proteins, e.g. using Proteominer technology or metabolic labeling
with deuterated amino acids [16, 17]. An efficient and sensitive
way to analyze protein signatures in FBS containing supernatant is
via commercially available multiplex bead-based immunoassays
(see Notes 6 and 7). The following steps are necessary to collect
conditioned medium and prepare it for measuring with multiplex
bead-based immunoassays.
1. Directly after end of stimulation, take off medium and put on
ice until proceeding with the next step (see Note 8).
2. To remove detached cells, spin supernatant with 2,700 g for
4 min at 4 C.
3. Transfer supernatant in a new tube and store at 80 C until
used for further analysis.
61
62
Notes
1. Be sure to use 6-well dishes that fit with the C-dishes (electrodes).
The IonOptix handbook proposes 6-well dishes of several
companies. To avoid contaminations, heat the electrodes at
100 C for 3 h. After experiment soak electrodes in demineralized water for several days to neutralize from salts that might
have formed on the carbon electrodes and sterilize again.
2. Experiments are performed on the first and second passage of
subcultured cells.
3. We use 2 mL medium per well (6-well dish). Be sure that the
electrodes dip into the medium and do not contact the bottom
of the dish containing the cells.
4. Be careful that the cable is not kinked by the incubator door
since it can be damaged then.
5. Lactate dehydrogenase (LDH) is another marker for muscle
fiber damage [15]. After 24 h of EP-stimulation, LDH is significantly increased in the total cell lysates of EPS cells and also
in the supernatant. These data suggest that elevated levels of
cytosolic proteins in the secretome needs to be considered
carefully and do not necessarily indicate higher percentage of
damaged cells (Fig. 2c; adapted from [11]).
6. For the selection of candidate myokines it might be helpful to
do a whole genome transcriptome analysis or quantitative
Real-time PCR prior to secretome study to find EPS-regulated
proteins.
7. The multiplex assays have several advantages. Only a small
sample volume is necessary due to its high sensitivity.
Additionally, it is easy to handle, enables an exact quantification in a concerted system. The biggest advantage compared
to commercial available enzyme-linked immunosorbent assay
(ELISA) is the measurement of several proteins simultaneously
in one assay.
8. During immunoassay antibodies might cross-react with proteins
of FBS, hence a sample of unconditioned medium (reference
medium) is urgently needed for multiplex measurement. Spin
down the reference medium according to methods in step 2 in
Subheading 3.3 and store supernatant at 80 C until used for
further analysis.
9. When mixing individual singleplexes check x-Plex Bead
Regions and avoid using 2 singleplexes with similar x-Plex
Bead Regions in one assay.
10. For the conditioned medium of human myotubes, neat samples
were used (50 L sample/well). In our case, all measured ana-
63
lytes were in the assay working range. If you are above the
upper limit of quantification make sure to dilute the samples
properly.
11. For comprehensive analysis of the secretome via 2DE, a protein concentration up to mg/mL range is required. In the special case of DIGE technology at least 150 g protein is essential.
Thus, huge amounts of medium are necessary.
12. To get rid of the complex protein mixture included in FBS,
cells are washed 2 times with PBS containing Ca2+/Mg2+. This
is used to wash adherent cell cultures when one merely wishes
to wash and have the cells remain adherent because adhesion
proteins require divalent cations to function. The third washing step is done with PBS without Ca2+/Mg2+ to decrease salt
concentration in the medium.
13. For untargeted proteomics the medium should be free of phenol red because it might interfere with protein assays performed
to determine protein concentration for subsequent gels.
14. Since the native protein concentration is very low, it is essential
to concentrate the supernatant to achieve a working concentration for subsequent gels. It might be necessary to transfer
the supernatant during concentration process in an even
smaller protein concentrator tube to gain smaller volumes and
the required protein concentration. Be aware that with every
change of the device material gets lost. To minimize the loss,
rinse the membranes of the larger device with flow through
and add it to the eluent of the smaller device. Additionally, take
care of protein precipitations that might form.
Acknowledgements
This study was supported in part by a grant from the Leibniz
Gemeinschaft (SAW-FBN-2013-3) to C. Weigert and by a grant
from the German Federal Ministry of Education and Research
(BMBF) to the German Center for Diabetes research (DZD e.V.;
No. 01GI0925).
References
1. Pedersen BK (2009) The diseasome of physical
inactivityand the role of myokines in muscle
fat cross talk. J Physiol 587:55595568
2. Gleeson M (2007) Immune function in sport and
exercise. J Appl Physiol (1985) 103:693699
3. Goldstein MS (1961) Humoral nature of the
hypoglycemic factor of muscular work.
Diabetes 10:232234
64
13.
14.
15.
16.
17.
Chapter 6
Urinary Pellet Sample Preparation for Shotgun
Proteomic Analysis of Microbial Infection
and HostPathogen Interactions
Yanbao Yu and Rembert Pieper
Abstract
Urine is one of the most important biofluids in clinical proteomics, and in the past decades many potential
disease biomarkers have been identified using mass spectrometry-based proteomics. Current studies mainly
perform analyses of the urine supernatant devoid of cells and cell debris, and the pellet (or sediment) fraction is discarded. However, the pellet fraction is biologically of interest. It may contain whole human cells
shed into the urine from anatomically proximal tissues and organs (e.g., kidney, prostate, bladder, urothelium, and genitals), disintegrated cells and cell aggregates derived from such tissues, viruses and microbial
organisms which colonize or infect the urogenital tract. Knowledge of the function, abundance, and tissue
of origin of such proteins can explain a pathological process, identify a microbe as the cause of urinary tract
infection, and measure the human immune response to the infection-associated pathogen(s). Successful
detection of microbial species in the urinary pellet via proteomics can serve as a clinical diagnostic alternative to traditional cell culture-based laboratory tests. Filter-aided sample preparation (FASP) has been
widely used in shotgun proteomics. The methodology presented here implements an effective lysis of cells
present in urinary pellets, solubilizes the majority of the proteins derived from microbial and human cells,
and generates enzymatic digestion-compatible protein mixtures using FASP followed by optimized desalting procedures to provide a peptide fraction for sensitive and comprehensive LC-MS/MS analysis. A highly
parallel sample preparation method in 96-well plates to allow scaling up such experiments is discussed as
well. Separating peptides by nano-LC in one dimension followed by online MS/MS analysis on a
Q-Exactive mass spectrometer, we have shown that more than 1,000 distinct microbial proteins and 1,000
distinct human proteins can be identified from a single experiment.
Key words Urine proteomics, Urinary pellet, Microbial infection, Hostpathogen interaction,
Filter-aided sample preparation (FASP), 96FASP
Abbreviations
FASP
HCD
LC
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_6, Springer Science+Business Media New York 2015
65
66
MS
MWCO
TIC
UTI
Mass spectrometry
Molecular weight cut off
Total ion chromatogram
Urinary tract infection
Introduction
Urine is a sample source of high importance for biomarker discovery because it is easily available and collected non-invasively in
large quantities [1, 2]. The identity and quantity of proteins
excreted into urine may reflect pathological conditions that can be
traced to different organs in the body, particularly the kidneys,
prostate, and urogenital tract [3]. Currently, most urine proteomic
studies focus on the analysis of the urinary supernatant, that is the
soluble fraction of the collected urine sample following centrifugation at 1,5005,000 g for 515 min [4, 5]. The resulting urinary
pellet is frequently discarded. However, the urinary pellets, especially those from patients with urinary tract infection (UTI), which
is one of the most common conditions that lead to hospital visits
[6], contain not only pathogenic microbes, in most cases bacteria
that colonize the urinary tract of the patient, but also host proteins
associated with the inflammatory process following colonization
with the microbial pathogen. Inflammation may include activities
such as recognition of pathogen molecular patterns, cytokine
release, leukocyte recruitment, lymphocyte recruitment, complement activation, immunoglobulin secretion, fibrin deposition,
release of iron-sequestering proteins and direct microbial killing
via enzymatic activities and permeabilization or disintegration of
bacterial membranes [68]. The presence and relative quantity of
such proteins serve as a diagnostic indicator of infection and inflammation [7]. Non-pathogenic bacteria not inducing inflammatory
responses may also be identified from urinary pellets [9].
Our laboratory reported the first metaproteomic analysis of
urinary pellets derived from patients diagnosed with either asymptomatic bacteriuria or UTI, and identified the microbial causes of
bacteriuria [9]. Most recently, our laboratory developed a highthroughput urinary sample preparation approach, 96FASP (96well filter-aided sample preparation), for quantitative shotgun
proteomic analysis [10]. This method promises to be the prototype of an economical method for the diagnosis of urinary tract
infections and inflammation in the future. This article describes an
extensively used, robust step-by-step procedure pertaining to the
preparation of urinary pellet samples using the FASP approach.
The optional 96FASP method that is adapted to process multiple
samples simultaneously is described as well.
67
Materials
2.2 StageTip
Desalting
2.3
LC-MS/MS
68
Methods
An overview of the protein sample preparation for urinary pellets is
provided in Fig. 1, as explained in detail in the following procedures. The schematic also shows the downstream applications (e.g.,
LC-MS/MS and database search) for urinary proteome analysis.
69
Fig. 1 An overview of the urinary pellet sample preparation for shotgun proteomics. The procedures are explained
in detail in this chapter. Briefly, the urine samples are first spin down to collect pellet fraction. The pellets are
then lysed and digested following FASP protocol. A protocol of 96-well format FASP is also illustrated. Afterwards,
the protein digests are cleaned using StageTip and analyzed by nanoLC-MS/MS. The host and pathogen protein
identifications can be obtained by searching a metaproteome database which includes human proteins as well
as the proteins of common pathogens related to your study (in this case, the urinary tract infection)
70
3.4 Peptide
Desalting Using
StageTip Protocol
71
1. This method is adapted from a published protocol [11]; several changes have been made to optimally fit the preparation
of urinary pellet samples (see Note 4).
2. Prepare StageTip by punching out small discs (13 layers
depending on the sample amount) of C18 Empore filter using
a 22 G flat-tipped syringe and ejecting the discs into P200
pipette tips. Ensure that the disc is securely wedged in the bottom of the tip.
3. Activate a tip by forcing 200 l methanol through the tip. Use
this step to check if the StageTip is leaky or overtight.
4. Force 200 l Elution Solution-II through the tip.
5. Equilibrate tip by forcing 200 l Wash and equilibration buffer through the tip. The tip is now ready for sample loading.
6. Resuspend the dried peptides into 100 l of Wash and equilibration buffer, and vortex for 10 min.
7. Load the 100 l peptide solutionmade in step [5] above
by forcing them through the C18-StageTip. Do not discard
the flow-through, but collect it into the original tube.
8. Reload the peptide solution onto the tip. This step may be
repeated two or three times.
9. Wash the tip with 200 l Wash buffer, and repeat this step one
or two times. The flow-through during this step can be
discarded.
10. Elute the peptides with 200 l Elution Solution-I (once) and
200 l Elution Solution-II (twice). Collect all of the eluates
(~600 l) into one maximum recovery microtube.
11. Dry the peptide eluates in the Speed-Vac (this may take 12 h).
12. Store then at 80 C, or resuspend it with LC solvent A for
immediate LC-MS/MS analysis.
3.5 Nano-LC-MS/MS
and Computational
Analysis
72
3. The acquired raw files are then processed using the Proteome
Discoverer software. The protein database involved in this
study contains UniProt human protein sequences and common urinary tract pathogens [12]. They can be downloaded
from UniProt website (http://www.uniprot.org/). MS search
parameters are similar to published previously [10].
4. Figure 2 shows a typical nanoLC-MS/MS analysis of the urinary pellet samples. The base peak of the TIC shows the eluted
peptides from HPLC column are acquired by mass spectrometer in 120 min. Shown in the upper panel is a representative
full MS scan collected at retention time 75.28 min. The mass
of each peak (peptide) are accurately measured by the Orbitrapbased mass analyzer at high resolution (e.g., 70,000), and
then sequenced in the HCD-based collision cell. The resulting
Fig. 2 A typical LC-MS analysis of the urinary pellet sample. The full mass (MS) and fragmentation (MS/MS)
are recorded by high resolution and high accuracy mass spectrometer. A representative MS scan at a given
time (for example, t = 75.3 min, as indicated by the red bar and arrows in the lower panel) with mass and
charge state information (for example, m/z = 668.3510, z = 2) is shown in the middle panel. Most of the ions in
this spectrum will be isolated and fragmentized afterwards. Then, the bioinformatics tool (for example, Sequest
algorithm) will assign the most confident amino acid sequence to each peak based on the sophisticated scoring system. Detailed explanations of the figure are provided in this chapter
73
Notes
1. Usually 550 ml of urine is collected from human subjects. The
preferred approach is to process the urine sample immediately
after collection by centrifugation (see step 2). It is possible to
store the urine at 4 C for up to 6 h before centrifugation.
Finally, the entire urine sample may be stored at 80 C at the
clinical site, shipped to the site of proteomic analysis and thawed
prior to centrifugal separation of urinary supernatant and urinary pellet. The freezethaw step may alter the composition of
the urinary pellet. For a given project with multiple samples, a
distinct urine storage and processing method should be selected.
2. In the case that gram-positive bacteria with thick cell walls,
such as Streptococcus pneumoniae and Staphylococcus aureus,
are suspected to be present in urine samples, resuspend the
urinary pellet samples in a volume of TMN buffer to have an
approximate volume ratio of 1:10, usually less than 200 l.
Pipette the suspension up and down a few times in a 1.5 ml
tube. Add lysostaphin and mutanolysin to a final concentration of 20 g/ml. Mix gently to homogenize the enzymes and
suspended cells. Incubate the sample in the 37 C shaker-incubator. Take out the tube every 30 min and check if a pellet
collects at the bottom. If so, briefly vortex every 30 min.
Complete digestion after a 3 h incubation.
After pre-treatment with lysostaphin and mutanolysin,
add up to 600 l USED buffer into the lysate.
3. To estimate the protein concentration, take 10 l aliquot of
the supernatant to another new microtube, mix with SDS
loading buffer and run it in a SDS-PAGE gel. Load 2 and 5 g
BSA standards in the same gel. Coomassie Blue (CB)-G250
stain the gel followed by its destaining with standard procedures [13]. From the overall CB-G250 staining intensity of
urinary pellet lysate bands, estimate the total protein amount
74
in the lane based on BSA staining intensities. The protein concentration could also be measured by tryptophan fluorescence
as reported before [14].
4. Instead of using syringe to manually push solvents through
the StageTips, one can use pipette tip adaptors (commercially
available from The Nest Group, MA) which fit the 1.5- or 2.0mL microtubes well. This way, all the processing steps with
syringe can now be done using a bench-top centrifuge [15].
Acknowledgments
This work was supported in part by Grant NIH-1R01GM103598
(National Institute of General Medical Sciences).
References
1. Decramer S, de Peredo AG, Breuil B, Mischak
H, Monsarrat B, Bascands J-L, Schanstra JP
(2008) Urine in clinical proteomics. Mol Cell
Proteomics 7:18501862
2. Wood SL, Knowles MA, Thompson D, Selby
PJ, Banks RE (2013) Proteomic studies of urinary biomarkers for prostate, bladder and kidney cancers. Nat Rev Urol 10:206218
3. Barratt J, Topham P (2007) Urine proteomics:
the present and future of measuring urinary
protein components in disease. Can Med
Assoc J 177:361368
4. Adachi J, Kumar C, Zhang Y, Olsen J, Mann
M (2006) The human urinary proteome contains more than 1500 proteins, including a
large proportion of membrane proteins.
Genome Biol 7:R80
5. Rodrguez-Surez E, Siwy J, Zrbig P, Mischak
H (2014) Urine as a source for clinical proteome analysis: from discovery to clinical application. Biochim Biophys Acta 1844:884898
6. Nielubowicz GR, Mobley HLT (2010) Hostpathogen interactions in urinary tract infection. Nat Rev Urol 7:430441
7. Pieper R, Suh M, Fouts D, Nelson K (2012)
Metaproteomic method for diagnosis of bacteriuria, urogenital tract and kidney infections
from urinary pellet samples. US Patent App.
13/728,106
8. Weichhart T, Haidinger M, Hrl WH, Semann
MD (2008) Current concepts of molecular
defence mechanisms operative during urinary
tract infection. Eur J Clin Invest 38:2938
Chapter 7
A Protocol for the Parallel Isolation of Intact Mitochondria
from Rat Liver, Kidney, Heart, and Brain
Sabine Schulz, Josef Lichtmannegger, Sabine Schmitt,
Christin Leitzinger, Carola Eberhagen, Claudia Einer,
Julian Kerth, Michaela Aichler, and Hans Zischka
Abstract
Mitochondria are key organelles for cellular energy production and cell death decisions. Consequently, a
plethora of conditions which are toxic to cells are known to directly attack these organelles. However,
mitochondria originating from different tissues differ in their sensitivity to toxic insults. Thus, in order to
predict the potential organ-specific toxicity of a given drug or pathological condition at the mitochondrial
level, test settings are needed that directly compare the responses and vulnerabilities of mitochondria from
different organs. As a prerequisite for such test strategies, we provide here a robust, prompt, and easy-tofollow step-by-step protocol to simultaneously isolate functional and intact mitochondria from rat liver,
kidney, heart, and brain. This isolation procedure ensures mitochondrial preparations of comparable purity
and reproducible quantities which can be subsequently analyzed for organ-specific mitochondrial toxicity.
Key words Mitochondria, Liver, Heart, Brain, Kidney
Introduction
The prime role of mitochondria in cell metabolism reflects the fact
that these organelles are crucial in controlling the cells fate, i.e.
survival or death, most importantly by balancing the cellular energy
demands. However, metabolic differences and metabolite preferences exist in the different healthy tissues of our body [1]. Whereas
brain tissue relies on glucose as the major metabolite, liver, especially in the postresorption phase, relies on fatty acids [1].
Consistent with these metabolic preferences, marked differences in
the molecular composition of the respective mitochondrial populations are known [24]. Thus, mitochondria delicately adapt at the
molecular level to the metabolic state of their tissue origin.
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_7, Springer Science+Business Media New York 2015
75
76
Isolation of Intact Mitochondria from Rat Liver, Kidney, Heart, and Brain
77
Fig. 1 Electron micrographs of liver, kidney, heart, and brain mitochondria in situ (Scale bars equal 2 m for 1,600
magnification, and 500 nm for 10,000 and 20,000 magnifications, respectively). Hepatocyte mitochondria
appear round or elongated in shape, with an electron-dense matrix, mostly tubular cristae and dense mitochondrial granules. Kidney tubule cell mitochondria contain densely packed elongated mitochondria oriented in right
angles to the base of the cell. Myocardial muscle cell mitochondria are round or elongated in shape comprising
numerous, closely arranged cristae. Myelinated axon mitochondria appear with lamellar and tubular cristae
78
Fig. 2 Electron micrographs of liver, kidney, heart, and brain mitochondria isolated in parallel by the herein
presented protocol from the same animal (scale bars equal 5 m for 5,000, 1 m for 20,000, and 500 nm
for 50,000 magnifications). Homogeneous mitochondrial populations are obtained. However, as observed in
situ, depending on their tissue origin isolated mitochondria appear with significant structural differences
Materials
2.1 Isolation
Procedure
Components
Fig. 3 A time stable mitochondrial membrane potential in freshly isolated mitochondria confirms the functional integrity of their inner membranes. Assessment of the
mitochondrial membrane potential was done using the fluorescent dye Rhodamine
123 according to published protocols [13]. (a) Due to accumulation within mitochondria and fluorescence quenching of the positively charged dye upon an intact,
negative inside membrane potential, a low fluorescence is indicative for an existing
membrane potential. Upon addition of the protonophor FCCP as internal control,
dissipation of the mitochondrial membrane potential occurs, which can be followed
by a steep increase in fluorescence. (b) As a semi-quantitative measure for the time
stable mitochondrial integrity the fluorescence ratio before and immediately after
FCCP addition is calculated using the time points t = 0 (first enlarged light grey
diamond) and t = 60 (second enlarged grey diamond) minutes
Table 1
Average tissue-dependent yield of mitochondria given
as mg mitochondrial protein per g tissue wet weight [14]
Tissue
Animals
Liver
17.7 5.1
11
Kidney
3.9 1.5
11
Heart
1.2 0.6
11
Brain
0.4 0.3
11
80
Isolation of Intact Mitochondria from Rat Liver, Kidney, Heart, and Brain
81
Methods
3.1 Preparation
of Percoll Gradients
3.2 Tissue
Harvesting
82
3.3 Homogenization
Procedure (See Note 1)
3.3.1 Liver, Kidney,
and Brain
1. Cut the heart in ~500 mg pieces (23 pieces) and wash out
residual blood if necessary.
2. Remove the cardiac fibrous skeleton carefully.
3. Transfer 1 500 mg piece into a 60 mm Petri dish with 2 mL
of ice cold isolation buffer with 0.1 % BSA.
4. Mince 500 mg heart tissue with scissors to obtain ~1 mm tissue pieces.
5. Mince further using a blade to obtain 0.30.5 mm pieces.
6. Transfer 0.30.5 mm heart tissue pieces into a 5 mL rough
glass-in-glass homogenizer and fill up with ice cold isolation
buffer with 0.1 % BSA to 5 mL.
7. Homogenize heart tissue with five strokes by hand, carefully
rotating the pestle during each stroke. Transfer homogenized
heart tissue to a centrifuge tube.
8. Repeat steps 37 until heart tissue is homogenized.
9. Dilute with ice cold isolation buffer with 0.1 % BSA using a
total volume of ~30 mL (2 15 mL centrifugation tubes).
10. Seal the centrifugation tubes with Parafilm and invert.
3.4 Centrifugation
Steps (See Note 10)
Isolation of Intact Mitochondria from Rat Liver, Kidney, Heart, and Brain
83
Fig. 4 Density gradient purification of mitochondrial populations using 18/30/60 % Percoll steps according to
Subheading 3.4. The arrow indicates mitochondria gathering at the 30/60 % Percoll interphase. Liver and
kidney mitochondria appear as prominent bands despite turbid 18 and 30 % Percoll phases. Heart and brain
mitochondria gather in a faint but visible band. For the latter Percoll gradients appear with whitely clouded more
intense 18 and 30 % phases than the actual mitochondria band at the 30/60 % Percoll interphase
84
Notes
1. The isolation procedure is ideally performed by two persons,
particularly at the homogenization steps. Recommended
apportionment: person A homogenizes liver, kidney, and
brain, person B takes responsibility for the homogenization of
the heart. This is because heart muscle-tissue is homogenized
with one additional mincing step and thereafter manually
homogenized. The heart homogenization procedure takes
approximately the same amount of time as the homogenization of liver, kidney, and brain together.
2. Be aware that mitochondria isolation from pancreas or spleen
may require supplementation with protease inhibitors during
tissue homogenization in order to retain mitochondrial
functionality.
3. EGTA dissolves only slowly, causing the pH to fall to ~3. Since
the supplied amount of EGTA is relatively small, care must be
taken to ensure complete solubilization by visual inspection.
4. Do not re-adjust the pH once the intended pH is exceeded.
5. The indicated volumes are sufficient for up to seven Percoll
gradients. For the liver 46 Percoll gradients are needed (see
Note 12), and 1 each for kidney, heart, and brain. Prepare
additional gradients if necessary. For heart and brain Percoll
gradients prepare only a two-phase 30/60 % Percoll
gradient.
6. The use of a Peleus ball is critical as the flow rate of a commonuse electric pipettor causes the Percoll-layers to mix.
Alternatively, a syringe can be used. Do not aim to empty the
pipette (or syringe) when releasing 4 mL, as final emptying
may also destroy the Percoll layers.
7. Weighing of organs is recommended to estimate the required
number of Percoll gradients for the liver (see Note 12).
Further, the organ weight is needed to calculate the obtained
mitochondrial protein per mg organ wet weight (cf. Table 1).
8. Tissue homogenization is critical, in the sense of liberating a
maximal amount of mitochondria from the cells without
destroying them immediately thereafter due to excessive
homogenization. Therefore, perform a minimum of strokes,
Isolation of Intact Mitochondria from Rat Liver, Kidney, Heart, and Brain
85
86
Acknowledgements
We would like to acknowledge E.E. Rojo for critical reading of the
manuscript. This study was supported in parts by the Deutsche
Forschungsgemeinschaft (DFG) grant RU742/6-1 to H.Z.
References
1. Lffler G, Petrides PE (1990) Biochemie und
pathobiochemie, 5th edn. Springer, Berlin
2. Mootha VK et al (2003) Integrated analysis of
protein composition, tissue diversity, and gene
regulation in mouse mitochondria. Cell
115(5):629640
3. Veltri KL, Espiritu M, Singh G (1990) Distinct
genomic copy number in mitochondria of different mammalian organs. J Cell Physiol
143(1):160164
4. Vijayasarathy C et al (1998) Variations in the
subunit content and catalytic activity of the
cytochrome c oxidase complex from different
tissues and different cardiac compartments.
Biochim Biophys Acta 1371(1):7182
5. Tahara EB, Navarete FD, Kowaltowski AJ
(2009) Tissue-, substrate-, and site-specific
characteristics of mitochondrial reactive oxygen species generation. Free Radic Biol Med
46(9):12831297
6. Andreyev A, Fiskum G (1999) Calcium
induced release of mitochondrial cytochrome c by different mechanisms selective
for brain versus liver. Cell Death Differ 6(9):
825832
7. Berman SB, Watkins SC, Hastings TG (2000)
Quantitative biochemical and ultrastructural
comparison of mitochondrial permeability
transition in isolated brain and liver mitochondria: evidence for reduced sensitivity
8.
9.
10.
11.
12.
13.
Chapter 8
Isolation of Mitochondria from Cultured Cells and Liver
Tissue Biopsies for Molecular and Biochemical Analyses
Sabine Schmitt, Carola Eberhagen, Susanne Weber,
Michaela Aichler, and Hans Zischka
Abstract
We recently reported a new method to isolate functionally intact mitochondria from cell culture and small
tissue samples (Schmitt et al., Anal Biochem 443(1):6674, 2013). This method comprises a semiautomated cell rupture, termed pump controlled cell rupture system (PCC), which can be precisely adjusted
to the specific cellular source of isolation and which can be tightly controlled (Schmitt et al., Anal Biochem
443(1):6674, 2013). Here we provide a detailed hands-on protocol of this PCC method which results in
an efficient cell breakage but preserving the mitochondrial integrity. Upon subsequent purification steps,
the obtained mitochondrial fraction meets the quality and purity required for molecular analyses, e.g. proteomic comparisons, as well as for biochemical analyses, e.g. determination of diverse enzymatic activities.
Key words Mitochondria, Cell culture, Biopsies, Balch homogenizer
Introduction
Mitochondria are the cellular powerhouses and key integrators of
cell death decisions [1]. Consequently, conditions that impair mitochondria can lead to severe human diseases. A prime example may
be the intoxication of hepatocyte mitochondria by genetically caused
excessive copper burdens in Wilson disease [2, 3]. While augmented
mitochondria-dependent cell death is a major obstacle in neurodegenerative disorders [4, 5], avoidance of cell death is a hallmark of
cancer [6]. Consequently, the identification of specific mitochondrial targets to either stabilize or impair their structure or function is
a central aim in biomedical research [7]. Typically, the identification
of such targets is achieved by comparing mitochondria isolated from
healthy controls to mitochondria from diseased tissues, by proteomics, immuno-blotting or enzymatic measurements, etc. Such
analyses may identify molecular and functional mitochondrial alterations that can assist in understanding disease progression [8], or in
determining new targets for specific therapeutic intervention [9], or
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_8, Springer Science+Business Media New York 2015
87
88
even provide important insights why conventional therapeutic interventions may help in some patients but fail in others [10].
Such comparative mitochondrial investigations of normal versus pathological situations require suspensions of isolated mitochondria with high and comparable purity. A most critical step in
the procedure to isolate mitochondria is the initial step of cell rupture. Especially if the starting material is limited, e.g. in cases of
cultured cells or tissue biopsies, an efficient cell membrane breakage is needed to obtain enough mitochondria for subsequent purification steps. However, if this is forcibly approached, a damage of
the mitochondrial membranes could occur. Thus, cell rupture or
tissue homogenization conditions have to be balanced between
efficient plasma membrane rupture and consequent mitochondrial
yield and mitochondrial integrity. To this end, means are needed
that can be precisely adjusted to the respective source of isolation.
We recently reported [11] that this can be accomplished using the
pump controlled cell rupture system (PCC). PCC consists of a
current version of the Balch-homogenizer (Isobiotec, Germany)
[12] coupled to a high precision pump. Within the Balchhomogenizer, the cells or the tissue have to pass an exactly adjustable clearance (Fig. 1). This allows for defining and controlling the
shear forces, which affect the cells and the mitochondria in the
course of the homogenization process. The high precision pump
further increases the reproducibility and controllability of the overall homogenization process.
In order to further extend the practical value of this method,
with a special focus on potential experimental pitfalls, we provide
here an easy-to-follow protocol for the use of PCC and downstream centrifugation steps to isolate mitochondria from cultured
cells and minute tissue samples (Fig. 2). Taken together, the herein
described protocol reproducibly yields isolated mitochondria from
cultured cells or minute tissue samples that meet the quality and
purity required for molecular as well as for functional analyses.
Materials
Prepare all solutions using ultrapure water (Prepared by purifying
deionized water to attain a resistivity of 18.2 M-cm at 25 C).
2.1 Isolation
of Mitochondria
from Cell Culture
or Tissue Biopsies
89
Fig. 1 Cartoon of the pump controlled cell rupture system (PCC). A high precision
pump (1) ensures, via gastight syringes (2), the continuous sample delivery (3) in a
constant rate to the Balch-homogenizer (4). Cell breakage occurs upon passage
through a defined clearance (square), which is adjusted by selecting tungsten carbide balls of different diameters (5). The cell homogenate is collected in a second
syringe (6) and can be re-subjected to the homogenizer, which is thermally equilibrated by a cooling plate (7). In this protocol, the syringe that initially contains the
sample (cell suspension or tissue pieces) is referred to as syringe B and the second
syringe is referred to as syringe A. Reproduced with permission from: Schmitt S,
Saathoff F, Meissner L, Schropp EM, Lichtmannegger J, Schulz S, Eberhagen C,
Borchard S, Aichler M, Adamski J, Plesnila N, Rothenfusser S, Kroemer G, Zischka
H (2013) A semi-automated method for isolating functionally intact mitochondria
from cultured cells and tissue biopsies. Anal Biochem 443:6674
90
Fig. 2 Flow chart for the isolation of mitochondria from cultured cells and liver tissue biopsies for molecular
and biochemical analyses
2.2 Purification
of the Isolated
Mitochondria
91
Methods
3.1 Isolation
of a Crude
Mitochondrial Fraction
from Cultured Cells
and Liver Tissue
92
Table 1
Experimental settings and mitochondrial yield (9,000 g pellet) for the isolation of mitochondria from
diverse cell types (in parts reproduced with permission from Schmitt S, Saathoff F, Meissner L,
Schropp EM, Lichtmannegger J, Schulz S, Eberhagen C, Borchard S, Aichler M, Adamski J, Plesnila N,
Rothenfusser S, Kroemer G, Zischka H (2013) A semi-automated method for isolating functionally
intact mitochondria from cultured cells and tissue biopsies. Anal Biochem 443:6674)
Concentration
[cells/ml]
Clearance
[m]
Number
of strokes
Yield of crude
mitochondria
[g/5 106 cells]
[ratio
RFU FCCP]
3 106
10
1,230
1.8
McA 7777
5 106
10
146
1.6
5 10
82
1.6
H4IIE
7 10
70
1.9
HepT1
5 106
114
1.5
Huh6
5 106
10
60
1.6
HepG2
5 10
128
1.6
HEK 293
5 106
70
1.7
HeLa
5 106
107
1.7
BeWo
5 10
10
90
1.7
1205Lu
5 10
10
110
1.6
Panc02
5 106
162
1.5
MEF
5 106
90
1.6
Fao
All isolated mitochondrial suspensions efficiently built up an inner transmembrane potential (m) which remained
stable for at least 1 h
93
94
Fig. 3 Monitoring of the isolation and purification of mitochondria from H4IIE cells via a Nycodenz density
gradient (a) by immunoblot analysis (b) and electron microscopy (c)
Notes
1. We do not recommend using plastic syringes due to their lack
of stability. In our hands, best results were obtained with
syringes from the given supplier.
2. This syringe pump model can be precisely adjusted and withstands the back-pressure that emerges during the homogenization process.
3. The manual metal-lift table can thereby assist the cooling of
the cell homogenizer during the homogenization process.
Two manual lift tables should be at hand if more than 25 106
cells will be homogenized. Exchanging against a pre-cooled
lift table every 30 min assists in effective cooling throughout
the homogenization process.
95
96
avoid residual contaminants in the cell homogenizer. If subsequently, mitochondria from other sources shall be isolated, we
recommend at least 34 cleaning passages of fresh isolation
buffer each. Alternatively, the whole homogenization unit
may be disassembled, cleaned, and reassembled again before
continuing.
12. Some small pieces of the liver tissue may enter the cell homogenizer, but not pass the clearance. We therefore recommend
that the second stroke starts with the syringe used for the first
stroke (syringe B). Tissue pieces that remained within the cell
homogenizer can now pass the clearance with this second
stroke, thereby increasing the yield without re-stressing already
isolated mitochondria.
13. In order to control the number of passages for the single
pieces of tissue, the system unit needs to be cleared from
remaining tissue pieces before starting with the next 3040 mg
of tissue to be processed.
14. Subsequent separation of mitochondria from unbroken cells/
nuclei in a first centrifugation step at 800 g is less efficient if
the homogenate is kept on ice for more than 45 min. A plausible explanation may be a progressive clotting of mitochondria, nuclei, and cell debris over time resulting in marked
mitochondrial loss in the 800 g pellet. Thus, in the case of
multiple isolations subsequent centrifugation steps should not
start later than 3040 min after the initial homogenization.
15. In order to avoid phase intermixing of the Nycodenz density
step gradient we recommend slowly layering the 300 l of 24
or 33 % Nycodenz (for cultured cells or liver tissue, respectively) with a syringe below the 18 % Nycodenz. The phase
boundary is slightly visible for the 18 %/24 % and clearly visible for the 18 %/33 % gradient.
16. If less than 650 g of the crude mitochondrial fraction are
loaded on the Nycodenz gradient, the mitochondrial interphase might be barely visible, overloading (more than 950 g
of the crude mitochondrial fraction) may impair the purification efficiency.
17. To collect the mitochondria, slowly lower a pipette mounted
with a 200 l tip just above the phase boundary, carefully aspirating the interphase. Immediately stop aspiration, the moment
as the mitochondrial band vanishes.
Acknowledgement
We would like to acknowledge E.E. Rojo and A. Simmons for critical
reading of the manuscript. This study was supported in parts by the
Deutsche Forschungsgemeinschaft (DFG) grant RU742/6-1 to H.Z.
97
References
1. Green DR, Kroemer G (2004) The pathophysiology of mitochondrial cell death. Science
305(5684):626629
2. Zischka H, Lichtmannegger J (2014)
Pathological mitochondrial copper overload in
livers of Wilsons disease patients and related
animal models. Ann N Y Acad Sci 1315:615
3. Zischka H et al (2011) Liver mitochondrial
membrane crosslinking and destruction in a rat
model of Wilson disease. J Clin Invest
121(4):15081518
4. Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443(7113):787795
5. Winklhofer KF, Haass C (2010) Mitochondrial
dysfunction in Parkinsons disease. Biochim
Biophys Acta 1802(1):2944
6. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100(1):5770
7. Schmitt S et al (2014) Why to compare absolute numbers of mitochondria. Mitochondrion
19(A):113123
8. Schulz S et al (2013) Progressive stages of
mitochondrial destruction caused by cell toxic
9.
10.
11.
12.
13.
Chapter 9
Dynamic Range Compression with ProteoMiner:
Principles and Examples
Lei Li
Abstract
One of the main challenges in proteomics investigation, protein biomarker research, and protein purity
and contamination analysis is how to efficiently enrich and detect low-abundance proteins in biological
samples. One approach that makes the detection of rare species possible is the treatment of biological
samples with solid-phase combinatorial peptide ligand libraries, ProteoMiner. This method utilizes hexapeptide bead library with huge diversity to bind and enrich low-abundance proteins but remove most of
the high-abundance proteins, therefore compresses the protein abundance range in the samples. This work
describes optimized protocols and highlights on the successful application of ProteoMiner to protein
identification and analysis.
Key words Low-abundance proteins, Proteomics, ProteoMiner, Dynamic range compression,
Plasma, Serum, Saliva, Host cell proteins
Introduction
Concentrations of different proteins in biological samples can span
as much as 12 orders of magnitudes. For example, 99 % of the protein content in plasma and serum samples is only about 20 highabundance proteins, whereas the other thousands of proteins are
less than 1 % of the plasma or serum proteome [1, 2]. After decades
of research and accumulated knowledge on high-abundance proteins due to their relatively easy access, the discovery and analysis of
the rare species in proteomes attracts more and more attentions.
ProteoMiner technology is based on a combinatorial hexapeptide
library bound to chromatographic beads [36]. It can be used to
decrease levels of high-abundance proteins for researchers to enrich and
detect more mid- to low-abundance proteins in complex samples and
unveil the proteomes deeply and comprehensively. When a complex
biological sample is added to the beads, proteins bind to their specific
ligands through combination of various types of interactions including
ionic interaction, hydrophobic interaction, hydrogen bonding and van
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_9, Springer Science+Business Media New York 2015
99
100
Lei Li
Fig. 1 (a) Scheme of the ProteoMiner technology to remove most of high-abundant proteins and enrich lowabundant proteins, thus compressing the protein concentration range in biological samples. (b) Twodimensional gel electrophoresis of human serum samples before and after treatment by ProteoMiner beads,
adapted from [31]. The samples were first separated on a pH 58 IPG strip, then an 816 % Criterion TrisHCl
gel before visualization by the Flamingo Fluorescent Gel Stain
200 l
500 l
1 ml
101
5 ml
250 kD
150 kD
100 kD
75 kD
50 kD
37 kD
25 kD
20 kD
15 kD
Fig. 2 Effect of sample to beads ratio on protein enrichment profile. Different amount of human serum samples
were treated with 20 l ProteoMiner beads in triplicate. The eluates were separated on a 420 % criterion
TrisHCl gel and visualized by Bio-Safe Coomassie Stain. The red arrows indicate increasing amount of proteins with higher sample to beads ratio
Materials
1. Microcentrifuge or vacuum manifold.
2. ProteoMiner Protein Enrichment Large Capacity Kit (BioRad, #163-3007 or #163-3009) and Small Capacity Kit (BioRad, #163-3006 or #163-3008). These kits contain 100 l
(Large Capacity) or 20 l (Small Capacity) of ProteoMiner
beads, PBS wash buffer (150 mM NaCl, 10 mM NaH2PO4,
pH 7.4), lyophilized elution reagent (8 M urea, 2 % CHAPS)
and rehydration reagent (5 % acetic acid), and are intended for
simple one-step elution of the bound proteins.
3. ProteoMiner Sequential Elution Large Capacity Kit (Bio-Rad,
#163-3011) and Small Capacity Kit (Bio-Rad, #163-3010).
These kits contain 100 l (Large Capacity) or 20 l (Small
Capacity) of ProteoMiner beads, PBS wash buffer (150 mM
NaCl, 10 mM NaH2PO4, pH 7.4) and 4 elution reagents
102
Lei Li
Methods
For Subheadings 3.13.5, the ProteoMiner Large Capacity Kits
are used as the example. The bold and italic volumes in parenthesis
are for the ProteoMiner Small Capacity Kits.
3.1 Column
Preparation
and Equilibration
3.3
Sample Wash
103
3.4
Elution
1. After all wash buffers have been removed, replace the bottom
and bottom caps and add 600 l (200 l) deionized water.
Attach top cap and invert end-to-end for 1 min.
2. Remove caps, place column in a collection tube and centrifuge
at 1,000 g for 3060 s to remove water.
If using vacuum up to this point, you will now need to switch
to centrifugation.
3. Attach bottom cap to the column (take caution to ensure the
bottom cap is tightly attached). Add 100 l (20 l) of rehydrated elution reagent to the column and replace top cap.
Lightly vortex for 5 s (see Note 4).
4. Incubate column at room temperature, lightly vortex several
times over a period of 15 min.
5. Remove caps, place in a clean collection tube and centrifuge at
1,000 g for 3060 s. This elution contains your eluted proteins.
6. Repeat steps 57 two more times.
7. Store elution at 20 C or proceed with downstream analysis.
For some downstream applications, we recommend a clean-up
of your sample prior to analysis. See Subheading 3.6 below for
the protocol in details.
3.5 Sequential
Elution (Alternative
to Subheading 3.4
After Subheading 3.3)
1. Carefully add 200 l wash buffer on top cap and all sides of the
column to ensure none of the beads are stuck to the cap and
sides of the column.
2. Centrifuge at 1,000 g for 3060 s. Discard collected material.
If using vacuum up to this point, you will now need to switch
to centrifugation.
3. Attach bottom cap to the column (take caution to ensure the
bottom cap is tightly attached). Add 100 l (20 l) of elution
reagent 1 to spin column. Incubate at room temperature and
lightly vortex several times over a period of 10 min.
4. Remove bottom cap, place column in a collection tube and
centrifuge at 1,000 g for 3060 s to collect the elution. This
elution contains your eluted proteins.
5. Repeat steps 3 and 4 two more times and collect both elutions
in the same tube from the above step.
104
Lei Li
Some downstream applications of the ProteoMiner-treated elutions may require to remove incompatible reagents. Below is an
easy and convenient method using the Micro Bio-Spin P-6 gel column for buffer exchanging the ProteoMiner eluates with compatible reagents.
1. Invert the column several times to resuspend the settled gel
and remove any bubbles.
2. Snap off the tip and place column in a 2 ml microcentrifuge
tube. Remove cap. Allow the excess packing buffer to drain by
gravity to top of gel bed. (If column does not begin to flow,
push cap back into column and remove.) Discard the buffer.
3. Place the column back into the 2 ml tube. Centrifuge for
1 min at 1,000 g to remove the packing buffer.
4. Block the bottom of the Bio-Spin 6 column with the tip and
add 500 l of the downstream application compatible buffer
(for example, the Exchange buffer in Subheading 2). Invert
the column several times to resuspend the settled gel.
5. Remove the bottom tip and place the column in a clean 2 ml
microcentrifuge tube. Remove cap. Allow the excess packing
buffer to drain by gravity to top of gel bed. Discard the buffer.
6. Place the column back into the 2 ml tube. Centrifuge for
1 min at 1,000 g to remove the buffer.
7. Repeat steps 46 two more times.
8. Place the column in a clean 2 ml microcentrifuge tube.
Carefully apply up to 50 l of ProteoMiner eluates to the column and centrifuge for 2 min at 1,000 g. The collected solution is buffer-exchanged to the compatible reagents.
Notes
1. As listed examples in the introduction, variety of sample types
from different species and sources have been used successfully
with the ProteoMiner beads. In principle, the more proteins are
incubated with the ProteoMiner beads, the more degree of
dynamic range compression can be reached, whereas extremely
low abundant proteins can be much enriched compared to the
high abundant proteins. As a guideline, 50 mg of serum or
plasma proteins are recommended for 100 l ProteoMiner
beads. In return, a bit more than 1 mg of proteins can be bound
to and eluted from the beads, so that up to 50-fold dynamic
range compression can be obtained. Depending on the needs
and goals of the specific research, it would be wise to keep in
105
mind of this while designing the experiment and deciding the ratio
between sample and ProteoMiner beads. Much higher sample to
beads ratio has been tested (Fig. 2) with still increasing enrichment
of a few proteins. Regular buffers based on phosphate buffered
saline (PBS) or Tris buffered saline (TBS) with or without mild
detergents in samples are recommended for the ProteoMiner
beads because of their less interference with binding of proteins to
ProteoMiner peptides (see Notes 24 below in more details).
2. Biological samples often contain interfering or incompatible
materials other than proteins which researchers are interested
in. It is crucial to remove them before applying samples onto
ProteoMiner beads. Dialysis, gel filtration, centrifugation and
protein precipitation are a few examples of pretreatment methods, and can be selected according to the sample types and
sizes, protein concentrations, and downstream applications.
The quality of the samples before ProteoMiner treatment can
be simply analyzed by gel electrophoresis or a UV/Vis spectrophotometer. The ProteoMiner process can be viewed as
affinity purification with complex proteinpeptide interactions. Interfering materials and incompatible solutions for
regular protein affinity purification will have some effects on
binding of certain proteins to their specific peptide ligands.
Buffer compatibility for wash and elution steps can often refer
affinity purification guidance and principles.
3. It is recommended to optimize the sample to beads ratio in order
to achieve the desired performance. Higher sample to beads ratio
tends to enrich more mid- to low-abundant proteins (Fig. 2). If
the protein concentration is low in the samples, concentrating
may be needed. Otherwise, the equilibrated ProteoMiner beads
can be taken out of the column and incubated with samples in
another tube. After sample binding is done, the beads can be
spun down and transferred back into the column for wash and
elution. The sample binding incubation time and temperature in
this step are the recommendations for plasma and serum samples.
Incubation at 4 C overnight would be another way researchers
can often use for their specific samples. If using plasma, clumping may occur after 1 h of binding; this is expected and will not
negatively impact the sample preparation. Heparinized plasma is
not compatible with this kit.
4. For 2-D users who plan to use the ProteoMiner treated samples on DIGE, this elution reagent will require clean up and
pH adjustment. As an alternative you may elute with DIGE
buffer. However, this may result in a decreased yield and number of protein spots. For a more complete elution if it would
not interfere with downstream applications, the Laemmli
sample buffer with 1 % SDS and reducing agents like
-Mercaptoethanol or DTT can be added and incubated with
ProteoMiner beads at 95 C for 5 min before elution.
106
Lei Li
References
1. Anderson N, Anderson N (2002) The human
plasma proteome: history, character, and diagnostic prospects. Mol Cell Proteomics 11:
845867
2. Anderson N, Polanski M, Pieper R et al (2004)
The human plasma proteome: a nonredundant
list developed by combination of four separate
sources. Mol Cell Proteomics 3:311326
3. Thulasiraman V, Lin S, Gheorghiu L et al
(2005) Reduction of the concentration difference of proteins in biological liquids using a
library of combinatorial ligands. Electrophoresis
26:35613571
4. Guerrier L, Thulasiraman V, Castagna A et al
(2006) Reducing protein concentration range
of biological samples using solid-phase ligand
libraries. J Chromatogr B 833:3340
5. Boschetti E, Righetti P (2008) Hexapeptide
combinatorial ligand libraries: the march for
the detection of the low-abundance proteome
continues. Biotechniques 44:663665
6. Boschetti E, Righetti P (2013) Low abundance protein discovery: state of the art and
protocols. Elsevier, Amsterdam
7. Castagna A, Cecconi D, Sennels L et al (2005)
Exploring the hidden human urinary proteome via ligand library beads. J Proteome Res
4:19171930
8. Guerrier L, Claverol S, Finzi L et al (2007)
Contribution of solid-phase hexapeptide
ligand libraries to the repertoire of human bile
proteins. J Chromatogr A 1176:192205
9. Guerrier L, Claverol S, Fortis F et al (2007)
Exploring the platelet proteome via combinatorial, hexapeptide ligand libraries. J Proteome
Res 6:42904303
10. Sim C, Bachi A, Cattaneo A et al (2008)
Performance of combinatorial peptide libraries
in capturing the low-abundance proteome of
red blood cells. 1. Behavior of mono- to hexapeptides. Anal Chem 80:35473556
11. Bachi A, Sim C, Restuccia U et al (2008)
Performance of combinatorial peptide libraries
in capturing the low-abundance proteome of
red blood cells. 2. Behavior of resins containing individual amino acids. Anal Chem 80:
35573565
12. D'Ambrosio C, Arena S, Scaloni A et al (2008)
Exploring the chicken egg white proteome
with combinatorial peptide ligand libraries.
J Proteome Res 7:34613474
13. Farinazzo A, Restuccia U, Bachi A et al (2009)
Chicken egg yolk cytoplasmic proteome,
mined via combinatorial peptide ligand libraries. J Chromatogr A 1216:12411252
107
Chapter 10
Qualitative and Quantitative Proteomic Analysis
of Formalin-Fixed Paraffin-Embedded (FFPE) Tissue
Omid Azimzadeh, Michael J. Atkinson, and Soile Tapio
Abstract
Formalin-fixed, paraffin-embedded (FFPE) tissue has recently gained interest as an alternative to fresh/
frozen tissue for retrospective protein biomarker discovery. However, during the formalin fixation proteins
undergo degradation and cross-linking, making conventional protein analysis technologies challenging.
Cross-linking is even more challenging when quantitative proteome analysis of FFPE tissue is planned. The
use of conventional protein labeling technologies on FFPE tissue has turned out to be problematic as the
lysine residue labeling targets are frequently blocked by the formalin treatment. We have established a
qualitative and quantitative proteomics analysis technique for FFPE tissues that combines label-free proteomic analysis with optimized protein extraction and separation conditions.
Key words Label-free proteomics, Biomarker discovery, Mass spectrometry, Formalin-fixed paraffinembedded (FFPE), Peptide modification
Introduction
For decades, formalin-fixed and paraffin-embedded (FFPE) tissue has
been the standard for histopathological analysis due to the ease of
sample preparation and stability in long-term storage. Clinical archives
frequently combine stored FFPE samples with data on diagnosis and
outcome. Such samples hold unique information on biological pathways and cellular processes leading to disease, but suitable molecular
technologies are required to access the information [13].
For a long time, quantitative proteomic studies on archival material have been considered to be an almost impossible task, primarily
due to the harsh and irreversible procedures needed for fixation and
the loss of macromolecular integrity during prolonged storage. The
changes occurring during cross-linking of proteins during the fixation
of FFPE tissue have been investigated in different studies [46]. The
consensus is that the major consequence of formaldehyde fixation is
the generation of a methylol modification at free lysine residues [4, 7].
Several studies have been conducted to evaluate different components
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_10, Springer Science+Business Media New York 2015
109
110
Materials
All solutions are prepared using HPLC grade water. All buffers
contain protease inhibitor cocktail following the manufacturers
instructions (Complete, Roche Diagnostics).
2.1 Buffers
and Solutions
for Sample
Preparation
2.3 Processing
of Gel-Resolved
Proteins and Tryptic
Digestion
111
1. Trypsin (Sigma-Aldrich).
2. 100 % acetonitrile (ACN).
3. 50 mM NH4HCO3 in 30 % ACN (w/v), pH 8.0.
4. 10 mM NH4HCO3, pH 8.0.
5. 80 % (v/v) ACN, 1 % (v/v) trifluoroacetic acid (TFA).
6. 5 % (v/v) ACN, 0.5 % (v/v) TFA.
7. Trypsin digestion equipment: shaker, incubator, and SpeedVac
centrifuge.
8. Peptide separation and identification equipment: Highperformance liquid chromatography (HPLC) and electrospray
ionization tandem mass spectrometry (ESI-MS/MS).
2.4 Experimental
Kits
1. 2D-Clean-Up-Kit (Bio-Rad).
2.5
Software
2. Bradford reagent.
Methods
3.1 Protein
Extraction from FFPE
Tissue
112
3.3 Processing
of Gel-Resolved
Proteins and Tryptic
Digestion
3.4 An Example
of Label-Free
Proteomics on FFPE
Sample
Due to the protein cross-linking during FFPE preparation, conventional labeling approach cannot be used for quantitative proteomic analysis on FFPE tissue. Formaldehyde, used in tissue
fixation, leads to a 30 Da (methylol) modification mainly on lysine
residues and inhibits classical labeling methodology that is targeted
to lysine residues. Alternatively, FFPE proteome profile has been
recently quantified using non-labeling approaches. For label-free
proteomics, the acquired spectra should be identified and quantified using the Progenesis software [16, 17] (see Note 11). To
interpret the observed alterations in proteome, the significantly
differentially expressed proteins in all samples should be analyzed
using bioinformatics tools such as PANTHER or DAVID [19, 20].
Classify the proteins using Gene ontology (GO) categories [21].
113
Fig. 1 Radiation-induced alteration of the cardiac mitochondrial proteome. Significantly differentially regulated
proteins were classified for GO categories Molecular function (a), Biological process (b), and Cellular
compartment (c) using the PANTHER and DAVID bioinformatics tools. The amount of deregulated proteins is
indicated as a number of the total amount of up- or down-regulated proteins at 24 h. The analysis indicated
that ionizing radiation caused impairment of mitochondrial structure and function
114
changes affected the proteins involved in oxidative phosphorylation such as NADH dehydrogenase, cytochrome c, and succinate
dehydrogenase.
Notes
1. To avoid keratin contamination, always wear gloves and cover
the hair during handling samples and gels.
2. To minimize proteolysis of the protein lysates, it is recommended to use protease inhibitors and keep the samples on ice.
3. During precipitation, do not overdry the pellet after removal
of the acetone as this might lead to difficulty in resolubilization of the pellet in the resuspension buffer.
4. If you have problems in determining the protein concentration of the lysate, it may be due to some component in the lysis
buffer. In this case try another protein quantification method.
5. If the protein yield after resolubilization is low, test other solubilization buffers as the result may differ with the types of tissues used. For more information, see the protocols mentioned
in ref. 6.
6. Use ultrapure proteomics grade reagents to prepare buffers for
staining and digestion.
7. For peptide or protein digestion, a ratio of between 1:100 and
1:20 (w/w) of trypsin to substrate is recommended.
8. HPLC setting and proteomics quantification should be handled by trained researchers.
9. To achieve a comprehensive and statically significant interpretation, use at least five biological and technical replicates if
they are available.
10. In order to evaluate the technical variability of mass spectrometry runs of label-free peptide quantifications of FFPE samples, analyze the aliquot of one control sample by repetitive
LC-MS/MS runs at least three replicates.
11. In order to evaluate the degree of the reversion of the crosslinking, the variable 30 Da methylol modification caused by
the cross-linking [7, 22, 23] was set for lysine, histidine, and
arginine residues during searching against the database [4].
Acknowledgements
This work was supported by a grant from the European
Community's Seventh Framework Programme (EURATOM)
contract n232628 (STORE).
115
References
1. Tapio S, Atkinson MJ (2008) Molecular information obtained from radiobiological tissue
archives: achievements of the past and visions
of the future. Radiat Environ Biophys 47(2):
183187
2. Tapio S, Hornhardt S, Gomolka M et al
(2010) Use of proteomics in radiobiological
research: current state of the art. Radiat
Environ Biophys 49(1):14
3. Tapio S, Schofield PN, Adelmann C et al
(2008) Progress in updating the European
radiobiology archives. Int J Radiat Biol
84(11):930936
4. Azimzadeh O, Barjaktarovic Z, Aubele M et al
(2010) Formalin-fixed paraffin-embedded
(FFPE) proteome analysis using gel-free and
gel-based proteomics. J Proteome Res 9(9):
47104720
5. Giusti L, Lucacchini A (2013) Proteomic studies of formalin-fixed paraffin-embedded tissues. Expert Rev Proteomics 10(2):165177
6. Steiner C, Ducret A, Tille JC et al (2014)
Applications of mass spectrometry for quantitative protein analysis in formalin-fixed paraffin-embedded tissues. Proteomics 14(45):
441451
7. Metz B, Kersten GF, Hoogerhout P et al
(2004) Identification of formaldehydeinduced modifications in proteins: reactions
with model peptides. J Biol Chem 279(8):
62356243
8. Magdeldin S, Yamamoto T (2012) Toward
deciphering proteomes of formalin-fixed
paraffin-embedded (FFPE) tissues. Proteomics
12(7):10451058
9. Hatakeyama K, Wakabayashi-Nakao K, Aoki Y
et al (2012) Novel protein extraction approach
using micro-sized chamber for evaluation of
proteins eluted from formalin-fixed paraffinembedded tissue sections. Proteome Sci 10:19
10. Shi SR, Taylor CR, Fowler CB et al (2013)
Complete solubilization of formalin-fixed,
paraffin-embedded tissue may improve proteomic studies. Proteomics Clin Appl 7(34):
264272
11. Xiao Z, Li G, Chen Y et al (2010) Quantitative
proteomic analysis of formalin-fixed and
paraffin-embedded nasopharyngeal carcinoma
using iTRAQ labeling, two-dimensional liquid
chromatography, and tandem mass spectrometry. J Histochem Cytochem 58(6):517527
12. Ono A, Kumai T, Koizumi H et al (2009)
Overexpression of heat shock protein 27 in
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
Chapter 11
Full-Length Protein Extraction Protocols and Gel-Based
Downstream Applications in Formalin-Fixed Tissue
Proteomics
Alessandro Tanca, Sergio Uzzau, and Maria Filippa Addis
Abstract
Archival formalin-fixed, paraffin-embedded (FFPE) tissue repositories and their associated clinical information can represent a valuable resource for tissue proteomics. In order to make these tissues available for
protein biomarker discovery and validation studies, dedicated sample preparation procedures overcoming
the intermolecular cross-links introduced by formalin need to be implemented. This chapter describes a
full-length protein extraction protocol optimized for downstream gel-based proteomics applications.
Using the procedures detailed here, SDS-PAGE, western immunoblotting, GeLC-MS/MS, 2D-PAGE,
and 2D-DIGE can be carried out on FFPE tissues. Technical tips, critical aspects, and drawbacks of the
method are presented and discussed.
Key words FFPE, Paraffin-embedded, Biobanks, Archival tissues, GeLC-MS/MS, Proteomics,
2D-PAGE, DIGE, Electrophoresis, Immunoblotting, Mass spectrometry
Introduction
Tissue biopsies are subjected to formalin fixation and paraffin
embedding (FFPE) in order to enable their microscopic examination in the context of routine diagnostic procedures. Once the
diagnostic process is completed, the residual tissues are stored in
long-term repositories. Clearly, these archives possess a huge
potential for retrospective tissue proteomics studies, thanks to their
matched datasets on the patient clinical records including diagnosis, disease progression, and response to therapy. In addition, when
considering the restrictions due to significant ethical issues, logistic
complexity, or poor availability of rare disease samples, that are
connected with the recovery of sufficient numbers of high-quality,
well-characterized fresh tissues, archived FFPE tissues become an
especially valuable resource [1, 2].
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_11, Springer Science+Business Media New York 2015
117
118
119
Fig. 1 Schematic workflow of the protocol. (a) Protein extraction steps. (b) Downstream gel-based
applications
120
Materials
All protocols require the use of a microcentrifuge and of 1.5 ml
microcentrifuge tubes. All reagents may be purchased from SigmaAldrich, if not otherwise specified.
2.1 Tissue
Deparaffinization
and Rehydration
2.2 Protein
Extraction
and Quantification
2.3
SDS-PAGE
121
(e.g.
SimplyBlue
7. Microwave oven.
2.4
Western Blotting
122
123
1. Clean scalpel.
2. Acetonitrile (ACN).
3. 50 mM ammonium bicarbonate (ABC) (see Note 16).
4. Reducing solution: 10 mM DTT, 50 mM ABC (see Note 5).
5. Alkylating solution: 55 mM IAM, 50 mM ABC.
6. Trypsin, proteomics grade.
7. Air circulation thermostat.
8. 20 % trifluoroacetic acid (TFA).
9. Vacuum centrifuge (e.g. Concentrator plus from Eppendorf).
10. 0.2 % formic acid (FA).
Methods
3.1 Tissue
Deparaffinization
and Rehydration
124
3.2 Protein
Extraction
and Quantification
3.3 Western
Immunoblotting
1. Mix the protein extract(s) (usual load 110 g per well) with
an equal volume of Laemmli sample buffer (see Note 22).
2. Incubate the sample(s) for 5 min at 95 C in a thermoblock,
then spin down briefly.
3. Assemble the apparatus for mini-format polyacrylamide gel
electrophoresis, add an adequate volume of running buffer
and load the sample(s).
4. Connect the gel electrophoresis apparatus to the power supply
and run according to the manufacturers instructions (approx.
30 min at 200 V for Bio-Rad TGX gels).
5. Equilibrate sponges, 3MM paper sheets, polyacrylamide gel
(containing the separated protein sample) and nitrocellulose
membrane by soaking them for a few seconds in transfer
buffer.
6. Assemble a sandwich composed by: sponge, two 3MM
paper sheets, polyacrylamide gel, nitrocellulose membrane,
two 3MM paper sheets, sponge (from the black cathode to the
red anode).
7. Put the sandwich into the transfer apparatus, along with a prefrozen freezer pack and a stir bar.
8. Position the transfer apparatus on a magnetic stirrer and fill it
up with transfer buffer.
9. Connect the transfer apparatus to the power supply and run at
250 mA for (at least) 1 h (see Note 23).
10. Stain the membrane with Ponceau staining for a few seconds
and destain with deionized water until the background is
almost completely white (if necessary, acquire the image with
a scanner) (see Note 11).
11. Block the membrane for 1 h with blocking solution (see Note 24).
12. Remove the blocking solution, pour an adequate volume of
PBS-T and wash the membrane with rapid agitation for 5 min.
125
13. Remove PBS-T, add the primary antibody solution and incubate
with gentle agitation for 1 h (after incubation, collect the primary antibody solution if planning to re-use it) (see Note 24).
14. Wash the membrane with an adequate volume of PBS-T with
rapid agitation for 5 min.
15. Repeat step 14 twice.
16. Remove PBS-T, add the secondary antibody solution and
incubate with gentle agitation for 1 h (after incubation, remove
the secondary antibody solution).
17. Repeat step 14 five times.
18. Wash the membrane with an adequate volume of PBS (alone,
without Tween-20) with rapid agitation for 5 min.
19. Remove excess PBS from the membrane (without allowing it
to dry out completely).
20. Put the membrane on a glass covered by a plastic film, cover it
with an adequate volume of chemiluminescent substrate and
incubate for 5 min.
21. Remove excess substrate from the membrane (without allowing it to dry out completely) and put the membrane between
two tracing paper sheets.
22. Acquire the chemiluminescent signal with a suitable imaging
system (see Note 25).
3.4
GeLC-MS/MS
126
127
2D-DIGE
128
10. Carefully pull off the cover film from the IPG strip gel and
place the strip into the slot, gel-side down, so that the rehydration solution can distribute evenly under the strip.
11. Gently close the lid of the IPGbox and allow the IPG strip gels
to rehydrate at RT for 624 h (see Note 36).
12. Transfer the strips from the IPGbox to the IPGphor manifold,
so that strips are faced up under the cover fluid with the anodic
end (+) pointing at the anode of the IPGphor.
13. Add 150 l deionized water to each paper wick (two per strip),
and position the wicks on each end of the IPG strips so that
one end of the wick overlaps the end of the IPG strip gel.
14. With the electrode cams in the open position, put the electrode assembly on top of all the wicks (the electrode must be
in contact with the wick), then close the lid and start the
appropriate IEF program.
15. Just before the end of the IEF program, prepare the
equilibration-reduction solution by adding 20 mg DTT per
ml of equilibration buffer.
16. Stop the IEF, drain the excess of mineral oil and briefly rinse
out the IPG strips with deionized water (see Note 37).
17. Position each IPG strip in a slot of the equilibration tray, add
an adequate volume of equilibration-reduction solution up to
completely cover the strip and incubate for 15 min with gentle
agitation.
18. Turn the pump valve of the Ettan DALTtwelve to circulate,
then fill the tank to the 7.5 l line with 1 running buffer.
19. On the control unit, adjust the temperature to the desired setting (25 C) and turn the pump on.
20. Prepare the equilibration-alkylation solution by adding 25 mg
IAM per milliliter of equilibration buffer.
21. Remove DTT solution from the IPG strips, add an adequate
volume of equilibration-alkylation solution up to completely
cover the strip and incubate for 15 min with gentle agitation.
22. Carefully unload the cassettes containing the polymerized gels
from the gel caster and rinse the top surface and the glass
plates with deionized water to remove residual n-butanol and
acrylamide.
23. Using forceps, remove the equilibrated IPG strips from the
equilibration solution and rinse them with fresh SDS running
buffer.
24. Slowly pipette agarose solution on the top of each gel, then
position the strip centered on the glass plate, and push against
the plastic backing of the IPG strip (with forceps or a plastic
support) to slide the strip between the two glass plates until it
is into contact with the surface of the slab gel.
129
25. Carefully slide the gels into the tank and fill the upper buffer
chamber with 2.5 l of 2 running buffer (up to the min/max
fluid line) (see Note 38).
26. Close the lid, set the running conditions according to the
desired run length (for instance, 1 W per gel overnight) and
start the run.
27. When the run has finished, unload the gel cassettes from the
electrophoresis chamber, rinse them with deionized water and
proceed with image acquisition using Typhoon laser scanner.
28. Upload the image files into the DeCyder software and carry
out differential analysis according to the software manual.
3.7 Preparation
of a Preparative
Gel, Excision,
and Digestion
of Differential Spots
Notes
1. FFPE tissue blocks should be carefully selected and evaluated
by an expert pathologist in order to assure the presence of a
minimum percentage of cells of the desired tissue type (for
instance, >90 % of neoplastic tissue), and consecutive sections
should be obtained for proteomic analysis. In general, five
5-m-thick tissue sections should provide sufficient material
for a gel-based proteomic analysis. Furthermore, especially for
precious tumor samples, an additional step of manual dissection or laser microdissection can be favorably added to enrich
in cancer cells and keep the amount of adjacent connective and
normal tissue to the minimum.
130
131
132
21. If a clear separation between the residual pellet and the protein
containing supernatant does not occur, collect the greatest
possible volume of supernatant without touching the tissue,
then centrifuge again the remaining wet pellet for at least
15 min at 16,000 g to squeeze it. Finally, collect the new
supernatant and merge it with the one obtained previously.
22. The protein load should be settled depending on the following factors: (a) concentration/abundance of the target antigen
within the protein mixture; (b) specificity of the antibody; (c)
sensitivity of the chemiluminescent substrate and of the signal
detection system. For a mediumlow abundance antigen, a
monoclonal antibody, the Sigma chemiluminescent peroxidase substrate and the VersaDoc imager, 1 g of total protein
should be enough.
23. When interested in high MW proteins, the transfer time may
be extended to 1.52 h.
24. Incubations may be extended up to overnight based on sample or antibody characteristics. For overnight incubations, preincubate the membrane at RT for approx. 30 min, then keep
the membrane at 4 C during the night, and finally equilibrate
the membrane the day after at RT for other 30 min.
25. If using the VersaDoc imaging system, a 5 min exposure is
usually enough to detect a visible signal.
26. The number of slices in which the lane is cut (i.e. the degree
of sample fractionation) is usually proportional to the number
of protein identifications achieved through MS analysis.
Alternatively, the number of protein identifications can be also
increased by increasing the length of the LC separation prior
to MS (a detailed description of the LC-MS/MS analysis
methods is outside the scope of this chapter).
27. Alternatively, samples can be destained using a solution containing 40 % (v/v) ethanol and 75 mM ABC and/or incubating the samples at 56 C and 500 rpm in a thermomixer until
the blue dye is no longer visible.
28. If the gel pieces are not completely shrunk and opaque after
collecting ACN, add fresh ACN up to cover the gel pieces,
incubate for 10 min, collect the new ACN supernatant and
add it to the supernatant mixture belonging to the same gel
slice.
29. In other words, the solution with the lower acrylamide concentration is to be poured in the mixing chamber labeled
light, and the solution with the higher acrylamide concentration in the reservoir chamber labeled heavy. Note that
the level of the solutions in the two chambers will NOT be
equal at this time.
133
30. Do not allow any air bubbles to enter the plates when casting
gradient gels, since bubbles hamper a correct formation of the
gradient.
31. The multi-casting chamber should be stored at RT possibly
overnight to ensure the complete acrylamide polymerization.
In fact, acrylamide polymerization continues in a silent way
even after gels start to visually appear as polymerized.
Afterwards, unused gels can be stored between wet papers at
4 C for up to 2 weeks.
32. Protein precipitation is performed to remove SDS and make
protein solution suitable for IEF separation. Alternatively,
detergent removal spin columns, dialysis membranes, or
MW-cut-off spin filters can be used to eliminate SDS from the
protein extract.
33. At this stage, sample pH should be between 8 and 9 to ensure
maximum labeling efficiency.
34. After this step, cyanine-labeled samples can be stored for at
least 3 months at 80 C in the dark.
35. Ensure that no bubbles are trapped within the strips.
36. Overnight rehydration is recommended to allow high MW
proteins to enter the gel.
37. Focused strips can be stored at 80 C for a few weeks.
38. Lubricate glasses with running buffer to prevent the rubber
tubing from sticking to the cassettes.
39. Alternatively, gels can be stained with fluorescent dyes (e.g.
Sypro Ruby) and their image can be acquired using an imager
suitable for fluorescent signal detection.
References
1. Klopfleisch R, Weiss AT, Gruber AD (2011)
Excavation of a buried treasure DNA,
mRNA, miRNA and protein analysis in formalin fixed, paraffin embedded tissues. Histol
Histopathol 26(6):797810
2. Fowler CB, O'Leary TJ, Mason JT (2013)
Toward improving the proteomic analysis of
formalin-fixed, paraffin-embedded tissue.
Expert Rev Proteomics 10(4):389400
3. Blonder J, Veenstra TD (2009) Clinical proteomic applications of formalin-fixed paraffinembedded tissues. Clin Lab Med 29(1):
101113
4. Tanca A, Pagnozzi D, Addis MF (2012)
Setting proteins free: progresses and achievements in proteomics of formalin-fixed,
paraffin-embedded tissues. Proteomics Clin
Appl 6(12):721
134
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
Chapter 12
Enrichment of Low-Abundant Protein Targets
by Immunoprecipitation Upstream of Mass Spectrometry
Barbara Kaboord, Suzanne Smith, Bhavin Patel, and Scott Meier
Abstract
Immunoprecipitation (IP) is commonly used upstream of mass spectrometry (MS) as an enrichment tool for
low-abundant protein targets. However, several aspects of the classical IP procedure such as nonspecific
protein binding to the isolation matrix, detergents or high salt concentrations in wash and elution buffers,
and antibody chain contamination in elution fractions render it incompatible with downstream mass spectrometry analysis. Here, we discuss two IP workflows that are designed to minimize or eliminate these contaminants: the first employs biotinylated antibodies and streptavidin magnetic beads while the second method
utilizes a traditional antibody that is oriented and cross-linked to Protein AG magnetic beads. Both modified
magnetic supports have low background binding and both antibody immobilization strategies significantly
reduce or eliminate antibody heavy and light chain contamination in the eluent, minimizing potential ion
suppression effects and thereby maximizing detection of target antigens and interacting proteins.
Key words Immunoprecipitation, Co-immunoprecipitation, Mass spectrometry, Proteinprotein
interactions, Biotin, Streptavidin, Magnetic beads, Protein AG, In-solution digestion
Introduction
Immunoprecipitation is widely used to enrich specific protein targets from complex samples for the purpose of evaluating differential
protein expression [14], probing for post-translational modifications [512], or identifying interacting proteins [1316]. An antibody is used to bind an antigen in cell lysates, serum, or tissue
homogenates and the resulting immune complexes are captured on
a solid phase support such as a chromatography resin, magnetic
bead, plastic plate, or membrane. Mass spectrometry is increasingly
becoming the detection methodology of choice for IPs as the technology has the capability to be used for discovery experiments to
identify protein interactors [14, 1619], semi-quantitatively in conjunction with SILAC or TMT-labeling [2025], or quantitatively in
selective reaction monitoring (SRM) assays [2630] (Fig. 1).
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_12, Springer Science+Business Media New York 2015
135
136
Sample
- Cell Culture
- Plasma/Serum
- Tissue
SDS-PAGE
Western blot
Immunoprecipitation
In-Gel/In-Solution Digestion
- Reduction/Alkylation
- Digestion
- Peptide cleanup
AQUA Peptide Spiking
Discovery MS
(LC-MS/MS)
Protein/PTMs
Identification,
Peptide Selection
Targeted MS
(LC-SRM/MS)
Absolute Quantitation
Fig. 1 Experimental workflow for IP to MS applications. Low-abundant proteins in a complex sample matrix typically need to be enriched in order to detect or quantify them. Protein targets are immunoprecipitated, processed
by in-gel or in-solution digestion, and analyzed by LC-MS/MS to identify proteins in the IP/Co-IP eluent, probe
for post-translational modifications, or select candidate peptides. Furthermore, heavy isotope-labeled peptide
standards can be added to the sample for targeted LC-SRM/MS for absolute quantitation. Gel electrophoresis
followed by silver stain or Western blot is still typically used as a validation component to this workflow
IP-MS
137
Fig. 2 Evaluation of resin types and IP strategies using an EGFR immunoprecipitation model system. Direct IP
(antibody pre-immobilized on the solid support followed by incubation with sample) was compared for three
supports (Polyacrylamide (P), Agarose (A) and Magnetic (M)) and four different chemistries of directly coupled
antibody. Additionally, biotinylated antibody and two immobilized streptavidin resins were used in an Indirect
IP method (antibody + sample preincubation before binding to solid support). (a) Capture efficiency was determined by Western blot. (b) EGFR sequence coverage and background proteins were determined by LC-MS/MS
after IP elutions and trypsin digestion. IP using streptavidin magnetic beads resulted in fewer background
proteins identified and higher EGFR sequence coverage
138
AG
+ - + - Direct
- + - + Indirect
Heavy
chain
PP2A
Light
chain
b
Streptavidin
Magnec
Beads
MS results
% Sequence
Coverage
Protein AG
Magnec Beads
IP/Co-IP targets
PP2A, catalytic
subunit (PPP2CA
and PPP2CB)
PP2A, regulatory
subunit (PPP2R1A
and PPP2R1B)
PP4, catalytic
subunit (PPP4C)
Indirect IP
Indirect IP
Direct IP
No Crosslink
Direct IP
With
Crosslink
57%
69%
58%
42%
14%
11%
5%
11%
42%
42%
35%
21%
Fig. 3 Comparison of different IP methods for MS application. (a) Antibody heavy and light chain contamination
is significantly reduced in the Protein AG cross-link and the biotinylated antibody/streptavidin methods compared to the classical Protein AG IP methodology. (b) Indirect and direct IP methods were used to evaluate
enrichment of PP2A, catalytic subunit (PPP2CA/PPP2CB) from 0.5 mg A431 cell lysate using Streptavidin
magnetic beads and Protein AG magnetic beads. IP elutions were processed by in-solution digestion method
and analyzed by LC-MS/MS to assess sequence coverage and identify isoform-specific peptides. MS analysis
of IP methods using streptavidin and Protein AG magnetic bead supports allowed for identification of two PP2A
catalytic subunits (PPP2CA and B), two PP2A regulatory subunits (PPP2R1A and B) and PP4 catalytic subunit
(PPP4C). Protein AG cross-link IP method resulted in slight decrease in PPP2CA recovery as indicated by
Western blot and by the percent sequence coverage obtained in the mass spectrometry analysis
IP-MS
139
Table 1
Detection and quantification of low-abundant targets by MS
Detected by LC-MS/MS
Quantified by LC-SRM/MS
Target
Cell line
Neat
Enriched-IP
Neat
Enriched-IP
EGFR
A431
HEK293
+
+
+
+
AKT1
A431
HEK293
+
+
+
+
AKT2
A431
HEK293
+
+
+
+
AKT3
A431
HEK293
+
+
N/A
N/A
N/A
N/A
PTEN
A431
HEK293
+
+
+
+
PIK3CA
A431
PIK3R2
A431
KRAS
A431
N/A
N/A
STAT1
A431
N/A
N/A
PPP2AC
A431
N/A
N/A
140
Materials
1. Pierce Streptavidin Magnetic Beads (Thermo Fisher Scientific,
Rockford, IL). Supplied as 10 mg/ml; binding capacity
~55 g biotinylated rabbit IgG per mg of beads.
2. N-hydroxysuccinimide-PEG4-biotin
(NHS-PEG4-biotin),
supplied as No-Weigh Format (8 2 mg in microtubes;
MW = 589) (Thermo Fisher Scientific): To make a 20 mM
solution, add 170 l DMF to one 2 mg reagent tube by puncturing the foil seal with the pipette tip. Pipette up and down
to mix. Use immediately (see Note 1).
3. Phosphate-buffered saline (PBS): 0.1 M sodium phosphate,
0.15 M NaCl, pH 7.5.
4. Zeba desalting column (0.5 ml; 7K cut-off) (Thermo Fisher
Scientific).
5. IP Lysis Buffer (Thermo Fisher Scientific): 25 mM TrisHCl,
pH 7.4, 150 mM NaCl, 1 mM EDTA, 1 % NP-40 and 5 %
glycerol.
6. Binding/Wash Buffer: 25 mM ammonium bicarbonate,
pH 8.0.
7. LC-MS grade water.
8. Elution Buffer: 0.5 % formic acid (LC-MS grade), 30 % acetonitrile (LC-MS grade).
9. Pierce Protein AG Magnetic Beads (Thermo Fisher Scientific).
Supplied as 10 mg/ml; binding capacity 5585 g rabbit IgG
per mg magnetic particles.
10. Disuccinimidyl suberate solution (DSS) (Thermo Fisher
Scientific): Dissolve 2 mg DSS in 150 l DMSO or DMF
immediately before use. Once reconstituted, the DSS must be
used immediately. DSS is moisture sensitive and must be
stored desiccated.
11. Modified PBS: PBS containing 0.05 % NP-40 (see Note 2).
12. 0.1 M glycine-HCl, pH 2.0.
13. Denaturation buffer: 6 M Urea, 50 mM TrisHCl, pH 8.0 (see
Note 3). Add 0.36 g urea (sequencing grade) in 0.5 ml
50 mM TrisHCl, pH 8.0, to dissolve then bring volume to
1 ml. Do not warm up to dissolve. Prepare fresh for each use.
14. 0.5 M Bond-Breaker TCEP Solution (Thermo Fisher
Scientific): Prepare a 1:50 working dilution by adding 10 l
0.5 M TCEP to 490 l 50 mM TrisHCl, pH 8.0.
15. 0.5 M Iodoacetimide: Dissolve 37 mg iodoacetamide in 400 l
MS-grade water. Make fresh and cover with foil to protect
from light.
IP-MS
141
Methods
3.1 Biotinylation
of Antibodies
and MagneticStreptavidin
Immunoprecipitation
mmol biotin.
mg antibody
mmol antibody
142
589 mg
170 l
l biotin solution.
mmol biotin 2 mg
IP-MS
143
16. Wash the beads with 3 500 l 25 mM Ammonium bicarbonate, pH 8, by gently vortexing the beads to resuspend during
each wash step (see Note 9).
17. Wash the beads with 2 500 l of LC-MS grade water, by
gently vortexing the beads to resuspend during each wash step
(see Note 10).
18. Elute bound antigen by adding 100 l elution buffer (0.5 %
formic acid, 30 % acetonitrile), and incubating for 5 min
at room temperature with periodic gentle vortexing
(see Note 11).
19. Collect the beads on a magnetic stand and transfer the elution
to a fresh microcentrifuge tube.
20. Dry the elution in a speed vac.
21. Proceed to mass spec sample prep (Subheading 3.3) and/or
resuspend in SDS-PAGE sample buffer for Western blot analysis/validation.
3.2 Cross-linked
IP Method
144
IP-MS
145
The proteins present in the IP elution fractions are typically denatured, reduced, alkylated, and digested into peptides prior to detection by mass spectrometry, because peptides fractionate, ionize,
and fragment more efficiently than intact proteins. In addition, the
resulting peptide mass spectra are easier to interpret during the protein
identification process. Because the protein amount in the IP eluent
is small, an in-solution digestion workflow is preferred as it minimizes sample loss, is fast and is amenable to high-throughput and
automation. Samples are first denatured in a strong chaotrope such
as urea to facilitate complete digestion. Disulfide bonds are reduced
with tris(2-carboxyethyl) phosphine (TCEP) or dithiothreitol
(DTT) and bond reformation is prevented by alkylating free sulfhydryl groups with iodoacetamide. The fully denatured proteins are
then digested with trypsin, although other proteases and protease
combinations can be used (e.g. chymotrypsin, Glu-C, Lys-C). The
resulting peptides are applied to C-18 tips or columns to remove
salts and buffers and to concentrate the sample prior to analysis by
nano or capillary reversed-phase liquid chromatography (LC), electrospray ionization, and tandem mass spectrometry (MS/MS).
The peptides are identified by sequence database searching using
the search algorithm SEQUEST or Mascot. The significance of
each peptide and protein identification is estimated using the software tools, Thermo Scientific Proteome Discoverer 1.4 and
Scaffold 4.0 software to assess percent sequence coverage, spectral
counts, and post-translational modification (PTMs).
1. Suspend dried samples in 10 l denaturation buffer (see Note 17).
2. Add 10 l of 10 mM TCEP to each sample and incubate at
37 C for 30 min. Centrifuge briefly to collect condensate to
bottom of tube.
3. To each 20 l sample of reduced protein add 0.83 l of 0.5 M
iodoacetamide solution (final concentration 20 mM). Incubate
at room temperature for 30 min in the dark (cover sample
tubes with foil OR place samples in a drawer).
4. Add 45 l of 50 mM TrisHCl, pH 8.0 to dilute urea concentration to <1 M.
146
Notes
1. The NHS-ester moiety readily hydrolyzes and becomes nonreactive. Do not prepare solutions for storage. Discard any
unused reconstituted reagent. Suspending the NHS-ester in a
dry solvent such as DMF or DMSO instead of water will slow
the hydrolysis rate, but storage should be limited to <1 month
at 20 C.
2. A very low percentage of detergent facilitates magnetic bead
collection and handling. NP-40 can be substituted with
Tween-20 if desired.
IP-MS
147
148
IP-MS
149
150
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
IP-MS
29. Wu C, Wei W, Li C et al (2012) Delicate analysis of post-translational modifications on
Dishevelled 3. J Proteome Res 11(7):
38293837
30. Rardin MJ, Held JM, Gibson BW (2013)
Targeted quantitation of acetylated lysine peptides by selected reaction monitoring mass
spectrometry. Methods Mol Biol 1077:
121131
31. Herzog F, Kahraman A, Boehringer D et al
(2012) Structural probing of a protein phosphatase 2A network by chemical cross-linking
and mass spectrometry. Science 337:
13481352
32. Kaboord B, Perr M (2007) Isolation of proteins and protein complexes by immunoprecipitation. In: Posch A (ed) Methods in
molecular biology, vol. 424. 2D PAGE: sample
33.
34.
35.
36.
151
Chapter 13
Principles of Protein Labeling Techniques
Christian Obermaier, Anja Griebel, and Reiner Westermeier
Abstract
Protein labeling methods prior to separation and analysis have become indispensable approaches for proteomic
profiling. Basically, three different types of tags are employed: stable isotopes, mass tags, and fluorophores.
While proteins labeled with stable isotopes and mass tags are measured and differentiated by mass
spectrometry, fluorescent labels are detected with fluorescence imagers. The major purposes for protein
labeling are monitoring of biological processes, reliable quantification of compounds and specific detection
of protein modifications and isoforms in multiplexed samples, enhancement of detection sensitivity, and
simplification of detection workflows. Proteins can be labeled during cell growth by incorporation of
amino acids containing different isotopes, or in biological fluids, cells or tissue samples by attaching specific
groups to the -amino group of lysine, the N-terminus, or the cysteine residues. The principles and the
modifications of the different labeling approaches on the protein level are described; benefits and shortcomings of the methods are discussed.
Key words Protein labeling, Quantification, Protein detection, Stable isotopes, Mass tags, Fluorophores, Difference gel electrophoresis, Mass spectrometry, Fluorescence imager
Introduction
Proteomics profiling employs various methods for identification,
monitoring of structural changes, and quantification of expression
levels of proteins in complex mixtures. The most commonly applied
techniques are the separation by gel electrophoresis and chromatography, and further investigation by image analysis, Western
blotting, and mass spectrometry. Lately, a comprehensive overview
on these tools has been published for the example of environmental
microbiology [1].
The different proteomic profiling workflows possess various
methodological shortcomings, which could partly be prevented or
at least reduced by pre-labeling proteins prior to the analysis.
Furthermore, labeling of specific protein moieties offers the possibility of studying some dedicated protein modifications. In the
following we try to give a small overview on methodical issues,
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_13, Springer Science+Business Media New York 2015
153
154
155
Materials
Generally, the use of high-quality electrophoresis/proteomic-grade
chemicals is paramount to achieving successful experiments.
Fluorescent dyes for pre-labeling are available from a number of
specialist suppliers and some fine chemicals companies. Also the
stable isotope labeling kits and the mass tag kits are marketed by
several fine chemical and mass spectrometry companies.
Methods
156
157
158
Fig. 1 (a) Schematic drawing of the DIGE 2-D electrophoresis method principle employing protein labeling with
fluorophores. The sample proteins are pre-labeled with Cy3 and Cy5. An internal standard is created by applying a mixture of all samples labeled with the third dye Cy2. (b) False color representation of the images of
samples and an internal standard, which have been co-separated in the same gel by DIGE. The images have
been acquired with a Typhoon multifluorescent laser scanner (GE Healthcare LifeSciences). (A) Cy2 channel:
pooled standard composed of the two samples; (B) Cy3 channel: mouse liver proteins, control; (C) Cy5 channel:
mouse liver proteins, treated; (D) overlay of all three images; the arrows point to a protein spot which is upregulated in the treated sample; (E) three-dimensional view of this spot in the Cy3 channel (left) and Cy5
channel (right). First dimension: Isoelectric focusing in IPG pH 310, 18 cm; second dimension: SDS polyacrylamide gel electrophoresis in a 12.5 % T gel. From ref. 8
159
160
161
Fig. 2 Schematic drawing of the principle of ICAT. The different samples are reduced with a thiol reagent and
labeled at the free SH groups of their cysteines with the ICAT reagents (light and heavy) via the iodoacetamide
group (IAA). After tryptic digestion of the sample proteins the labeled (biotin-containing) peptides are purified
with a Streptavidin affinity column and submitted to LC/MS for relative quantification and protein identification
162
Fig. 3 Schematic drawing of the principle of SILAC. The cells are grown under different conditions in media
containing different isotope compositions of hydrogen, carbon, and nitrogen. The samples are then mixed
together. The duplex mixture can be fractionated by any protein separation method. After tryptic digestion of
the protein fractions, the peptides are submitted to LC/MS for relative quantification using the defined mass
shifts of the related peptide peaks, and protein identification
163
the samples are denatured, reduced and alkylated prior to the labeling reaction. The tagged samples are mixed in equal amounts and
separated by electrophoretic or chromatographic techniques.
Because all lysines are blocked by the ICPL labels, protein cleavage
is ideally performed by double digest with trypsin and endoproteinase GluC. This measure ensures improved sequence coverage.
The use of Nic-NHS as a label enhances the ionization of the
labeled peptides, and hence the sensitivity of mass spectrometry,
which allows also the detection of low abundant proteins. Figure 4
shows a schematic drawing of the ICPL principle. The method is
compatible with both MALDI and electrospray ionization mass
spectrometry, for quantification with ICPL MS/MS is not required.
Fig. 4 Schematic drawing of the principle of ICPL. The protein samples are labeled with the ICPL reagents
containing different isotope compositions of hydrogen and carbon at the -amino groups of the lysines. The
samples are then mixed together. The multiplex mixture can be fractionated by any protein separation method.
After tryptic digestion of the protein fractions, the peptides are submitted to LC/MS for relative quantification
using the defined mass shifts of the related peptide peaks, and protein identification
164
The evaluation software selects only the peptide peak sets with the
defined mass shifts of 0, 4, 6, and 10 showing differing expression
levels. The ratios of peak intensities of the peptide pairs, triplets or
quadruplets are used to determine the related protein abundances
originating from the different samples. Analyzing quadruplets
instead of triplets or doublets considerably reduces the number of
false-positive multiplets, because randomly occurring sets with the
four defined mass differences are considerably less frequent than
sets with only two or three defined mass shifts (see also Note 4).
ICPL has the advantage, that it yields high sequence coverage, also
when the complexity of the labeled samples has been reduced with
high-resolution separation techniques. This feature is very important for adequate detection of post-translational modifications and
protein isoforms. It offers multiplex analysis of up to four samples,
and it can be applied on any species, cell lysates, or tissue types.
Notes
1. For optimum results, a sample cleanup based on protein precipitation prior to labeling is recommended. It is essential
for labeling, that the sample solution has a pH value between
8 and 9. In order to avoid any interference with the labeling
procedure, the sample solution must not contain any primary
amines, like carrier ampholytes, and reductant, like dithiothreitol.
These reagents are added after labeling has been completed.
2. Saturation labeling is performed for one hour at a pH value of
8.0 and a temperature of 37 C. Like with minimal labeling,
the sample must not contain any carrier ampholytes and reductant. Also here these compounds are added prior to application
on the first dimension.
3. SERVA HPE Lightning Red is compatible with all additives
typically used for sample solubilization and protein extraction,
including carrier ampholytes and reductants like dithiothreitol
(DTT) and dithioerythreitol (DTE). For efficient labeling the
pH value of the sample must be 8 or higher.
4. The ICPL software cannot identify incomplete quadruplets,
which sometimes occur due to the complete absence of a protein in a sample as a result of a complete down-regulation. It is
therefore advised to prepare an internal standard, which is
composed of equal amounts of all samples to be analyzed. This
mixed sample is divided into four aliquots, which receive the
four labels before they are combined again. The internal
standard is treated in the same way like the samples. This will
create equally abundant quadruplets, which are used in the
software data base for the detection and quantification of
incomplete quadruplets.
165
References
1. Whlbrand L, Trautwein K, Rabus R (2013)
Proteomic tools for environmental microbiologya roadmap from sample preparation to
protein identification and quantification.
Proteomics 13:27002730
2. Miller I, Crawford J, Gianazza E (2006)
Protein stains for proteomic applications:
which, when, why? Proteomics 6:53855408
3. Moche M, Albrecht D, Maa S et al (2013)
The new horizon in 2D electrophoresis new
technology to increase resolution and sensitivity. Electrophoresis 34:15101518
4. Zhou S, Bailey MJ, Dunn MJ et al (2005) A
quantitative investigation into the losses of proteins at different stages of a two-dimensional
gel electrophoresis procedure. Proteomics 5:
27392747
5. Go Y-M, Jones DP (2013) The redox proteome. J Biol Chem 288:2651226520
6. Silva JC, Denny R, Dorschel CA et al (2005)
Quantitative proteomic analysis by accurate
mass retention time pairs. Anal Chem 77:
21872200
7. nl M, Morgan ME, Minden JS (1997)
Difference gel electrophoresis: a single gel
method for detecting changes in protein
extracts. Electrophoresis 18:20712077
8. Westermeier R, Scheibe B (2008) Difference
gel electrophoresis based on Lys/Cys tagging.
In: Posch A (ed) Sample preparation and fractionation for 2-D PAGE/proteomics. Methods
in molecular biology, vol 424. Humana,
New York, pp 7385
9. Berendt FJ, Frhlich T, Bolbrinker P et al
(2009) Highly sensitive saturation labeling
reveals changes in abundance of cell cycleassociated proteins and redox enzyme variants
during oocyte maturation in vitro. Proteomics
9:550564
Chapter 14
Isolation of Extracellular Vesicles for Proteomic Profiling
Dong-Sic Choi and Yong Song Gho
Abstract
Extracellular vesicles are nano-sized lipid bilayer vesicles released from most cells, including archaea, bacteria,
and eukaryotic cells. These membrane vesicles play multiple roles in cell-to-cell communication, including
immune modulation, angiogenesis, and transformation of cells by transferring genetic material and functional proteins. They contain specific subsets of proteins, DNA, RNA, and lipids that represent their cellular status. Furthermore, extracellular vesicles are enriched in cell type- or disease-specific vesicular
proteins, especially plasma membrane proteins, which have pathophysiological functions; these vesicular
proteins are considered novel diagnostic biomarkers as well as therapeutic targets. To profile the proteome,
various purification methods of extracellular vesicles have been developed, but density gradient ultracentrifugation is considered the most promising. In this chapter, we describe the isolation of extracellular
vesicles derived from SW480 cells and the preparation of tryptic peptides for mass-spectrometry-based
proteomic analysis.
Key words Exosomes, Ectosomes, Microvesicles, Proteomics, Iodixanol, Ultracentrifugation,
Methanol/chloroform precipitation, Biomarker
Introduction
Nearly all living cells on Earth release vesicles into the extracellular
space. Extracellular vesicles are spherical, lipid-bilayered structures
containing various proteins, nucleic acids, and lipids [13].
Although they have long been considered cellular dust or artifacts
from dead cells, recent research has revealed that extracellular vesicles play various physiological and pathological roles, such as
immune modulation, angiogenesis, and transformation of cells by
transferring genetic material and functional proteins [1, 2]. It is
known that cells release two types of extracellular vesicle: exosomes
and ectosomes [1, 2]. Exosomes, 40100 nm in diameter, are
released via the fusion of multivesicular bodies with the plasma
membrane, followed by the discharge of intraluminal vesicles from
the multivesicular bodies into the extracellular space. Ectosomes,
also known as microvesicles, are 1001,000 nm in diameter and
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_14, Springer Science+Business Media New York 2015
167
168
169
Materials
Prepare all solutions using double-distilled water, to attain a sensitivity of 18 M cm at 25 C, and analytical grade reagents unless
otherwise indicated. Prepare and store all reagents at 4 C. All buffers should be passed through a 0.45 m filter. It is necessary to use
sterile equipment if the final use of the extracellular vesicles will
require sterility (e.g., in vitro cell-based assays or in vivo animal
experiments). In addition, purified extracellular vesicles should be
free of lipopolysaccharide contamination, especially for
immunology-related experiments.
170
2.4 In-Solution
Protein Digestion
Methods
171
Table 1
Rotor information for purification of extracellular vesicles
Rotor
Rotor type
Tubes
Number of rotor
cavities nominal
volume of tube
SW32 Ti
Swinging bucket
Ultra-Clear
6 38.5 mL
28,500 rpm
(100,000 g)
SW41 Ti
Swinging bucket
Ultra-Clear
6 13.2 mL
28,500 rpm
(100,000 g)
40,200 rpm
(200,000 g)
Type 90 Ti
Fixed angle
Polycarbonate
8 10.4 mL
49,200 rpm
(150,000 g)
Conversion of
rpm to g-force
172
3.4 Methanol/
Chloroform
Precipitation
and In-Solution
Digestion
of Extracellular
Vesicles for Proteomic
Profiling (See Note 9)
3.4.1 Methanol/
Chloroform Precipitation
Table 2
Densities of iodixanol solutions
Diluent (L)a
% (w/v) Iodixanol
(g/mL)
1.031
100
1.059
12
88
10
1.078
20
80
14
1.098
28
72
16
1.107
32
68
20
1.127
40
60
24
1.146
48
52
30
1.175
60
40
34
1.194
68
32
40
1.223
80
20
a top
(kDa)
100
75
63
10
top
(kDa)
48
173
bottom
1
10
35
48
CD63
25
CD81
35
20
Density
(g/mL)
Density
(g/mL)
Fig. 1 Purification of SW480-derived extracellular vesicles. (a) Fractions of iodixanol buoyant density gradients
were analyzed by Western blotting. CD63 and CD81, marker proteins of extracellular vesicles, were detected
in the third fraction from the top. (b) Transmission electron microscopy indicated that purified extracellular
vesicles were small, closed vesicles ranging from 50 to 200 nm in diameter. Scale bar, 200 nm
1. Resuspend the precipitated proteins with 100 L protein denaturation buffer and vortex thoroughly to completely dissolve
the proteins.
2. Add 1 L of reduction solution and incubate for 1 h at room
temperature.
3. Add 5 L of alkylation solution and incubate for 30 min at
room temperature in the dark.
4. Add 900 L protein digestion buffer and 4 L of protease
solution (the ratio of trypsin to protein should be 1:20).
5. Incubate for 16 h at 37 C in a shaking incubator.
6. Vacuum dry or lyophilize the sample.
174
Notes
1. To acquire the appropriate amount of isolated extracellular
vesicles (>50 g based on protein amount) for proteomics and
other assays, it is necessary to use as many cells as possible
(i.e., a minimum of 50, 150 mm culture plates, corresponding
to ca. 1 109 cells). In addition, it is better to purify extracellular vesicles from large volumes of conditioned medium,
because the loss of extracellular vesicles is decreased in large
starting volumes.
2. Serum contains lots of extracellular proteins at high concentrations as well as serum-derived extracellular vesicles, which contaminate cultured cells and the culture plate. Without careful
washing, these components are co-purified during isolation of
extracellular vesicles. To entirely remove these contaminants,
use PBS corresponding to 1.25-fold the volume of culture
medium to wash the cells (e.g., 25 mL PBS and 20 mL culture
medium in a 150 mm culture plate).
3. Cell culture with serum-containing media can cause the copurification of serum-abundant extracellular proteins and
175
176
b
Concentration
20
10
0
10
(1 x 106 particles/mL)
Number (%)
a 30
4
3
2
1
0
100
1000
Size (nm)
10000
10
100
1000
10000
Size (nm)
Fig. 2 Size distribution and particle concentration of purified SW480-derived extracellular vesicles. (a) The size
distribution of extracellular vesicles was measured by dynamic light scattering and indicated that the average
diameter is 176.70 7.35 nm. (b) Nanoparticle tracking analysis showed that the average diameter of purified
extracellular vesicles is 172.41 16.42 nm, with a particle concentration of 7.56 0.49 109 particles/g
177
Acknowledgement
We thank Gyeongyun Go for helping in isolation and characterization
of extracellular vesicles and Jaewook Lee for analysis of the extracellular vesicles in transmission electron microscope. This work was
supported by the National Research Foundation of Korea (NRF)
grant funded by the Korea government (MEST) (No. 2014023004
and No. 2012R1A1A2042534).
References
1. Choi DS, Kim DK, Kim YK, Gho YS (2013)
Proteomics, transcriptomics and lipidomics of
exosomes and ectosomes. Proteomics 13:
15541571
2. Choi DS, Kim DK, Kim YK, Gho YS (2014)
Proteomics of extracellular vesicles: exosomes
and ectosomes. Mass Spectrom Rev (in press).
doi:10.1002/mas.21420
3. Kim DK, Kang B, Kim OY, Choi DS, Lee J,
Kim SR, Go G, Yoon YJ, Kim JH, Jang SC,
Park KS, Choi EJ, Kim KP, Desiderio DM, Kim
YK, Lotvall JO, Hwang D, Gho YS (2013)
EVpedia: an integrated database of highthroughput data for systemic analyses of extracellular vesicles. J Extracell Vesicles 2:20384
4. Thery C, Amigorena S, Raposo G, Clayton A
(2006) Isolation and characterization of exosomes from cell culture supernatants and biological fluids. Curr Protoc Cell Biol Chapter 3,
Unit 3:22
5. Muralidharan-Chari V, Clancy J, Plou C,
Romao M, Chavrier P, Raposo G, DSouzaSchorey C (2009) ARF6-regulated shedding of
tumor cell-derived plasma membrane microvesicles. Curr Biol 19:18751885
6. Kato K, Kobayashi M, Hanamura N, Akagi T,
Kosaka N, Ochiya T, Ichik IT (2013)
Electrokinetic evaluation of individual exosomes by on-chip microcapillary electrophoresis with laser dark-field microscopy. Jpn J Appl
Phys 52:06GK10
7. Choi DS, Park JO, Jang SC, Yoon YJ, Jung JW,
Choi DY, Kim JW, Kang JS, Park J, Hwang D,
Lee KH, Park SH, Kim YK, Desiderio DM,
Kim KP, Gho YS (2011) Proteomic analysis of
8.
9.
10.
11.
12.
13.
Chapter 15
A Protocol forExosome Isolation andCharacterization:
Evaluation ofUltracentrifugation, Density-Gradient
Separation, andImmunoaffinity Capture Methods
DavidW.Greening, RongXu, HongJi, BowJ.Tauro,
andRichardJ.Simpson
Abstract
Exosomes are 40150nm extracellular vesicles that are released from a multitude of cell types, and perform
diverse cellular functions including intercellular communication, antigen presentation, and transfer of
tumorigenic proteins, mRNA and miRNA.Exosomes are important regulators of the cellular niche, and
their altered characteristics in many diseases, such as cancer, suggest their importance for diagnostic and
therapeutic applications, and as drug delivery vehicles. Exosomes have been purified from biological fluids
and invitro cell cultures using a variety of strategies and techniques. In this chapter, we reveal the protocol
and key insights into the isolation, purification and characterization of exosomes, distinct from shed
microvesicles and apoptotic blebs. Using the colorectal cancer cell line LIM1863 as a cell model, a comprehensive evaluation of exosome isolation methods including ultracentrifugation (UC-Exos), OptiPrep
density-based separation (DG-Exos), and immunoaffinity capture using anti-EpCAM-coated magnetic
beads (IAC-Exos) were examined. All exosome isolation methodologies contained 40150nm vesicles
based on electron microscopy, and positive for exosome markers (Alix, TSG101, HSP70) based on immunoblotting. This protocol employed a proteomic profiling approach to characterize the protein composition of exosomes, and label-free spectral counting to evaluate the effectiveness of each method in exosome
isolation. Based on the number of MS/MS spectra identified for exosome markers and proteins associated
with their biogenesis, trafficking, and release, IAC-Exos was shown to be the most effective method to
isolate exosomes. However, the use of density-based separation (DG-Exos) provides significant advantages
for exosome isolation when the use of immunoaffinity capture is limited (due to antibody availability and
suitability of exosome markers).
Keywords Exosome, Protocol, Isolation, Purification, EpCAM, Immunoaffinity capture, Density,
Proteomics, Extracellular vesicle, Shed microvesicles
1 Introduction
Almost all cells release different types of membrane microvesicles
and nanovesicles, which have a variety of important physiological
effects. Recently, evidence has emerged that cells naturally release
extracellular vesicles (EVs), which convey critical information in
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI10.1007/978-1-4939-2550-6_15, Springer Science+Business Media New York 2015
179
180
181
(sMV)
Shed microvesicles (sMV)
preparation
preparation
Concentrated culture
medium
ion
medium preparat
preparation
Membrane filtration
(0.1 m pore size)
Ultracentrifugation
(10,000 x g, 30 min)
Supernatant
(soluble factors and crude
exosomes)
PBS wash
Ultracentrifugation
(10,000 x g, 30 min)
sMVs
Ultracentrifugation
(100,000 x g, 1 h)
Ultracentrifugation
(100,000 x g, 1 h)
Crude Exosomes
Fig. 1 Crude exosome and shed microvesicle isolation. The experimental workflow used for crude exosome
and shed microvesicle isolation. Cell media/supernatant or biofluids are processed by differential centrifugation to remove intact cells and cell debris. The resulting supernatant can be processed immediately or stored
(4C for up to 1 week, or 20C). Two different methods for isolation of crude exosomes are presented in this
chapter and include: (1) concentrated culture medium (CCM) preparation, or (2) the isolation of shed microvesicles (sMVs) approach. For the CCM preparation (Subheading3.1), the supernatant is filtered using a membrane 0.1m filter unit and concentrated to ~1.5ml using centrifugal ultrafiltration (100K nominal molecular
weight limit (NMWL) filters). The resulting CCM is further ultracentrifuged at 100,000g to obtain the crude
exosome pellet. For isolation of sMVs (Subheadings3.43.5), the culture medium is centrifuged at 10,000g
for 30min, whereby the supernatant is ultracentrifuged at 100,000g to obtain the crude exosome pellet
(see Note 9)
182
Fig. 2 Strategies for exosome purification. In order to evaluate different strategies for exosome purification and
characterization, we utilized a human cell line model (LIM1863). (a) A confocal microscopy cross-section
through a LIM1863 organoid highlights concentrated syntaxin 3 staining at the apical ring/lumen (red), and
A33 staining at the basolateral cell periphery (green). Scale 20m. (b) Exosomes were isolated from CCM
derived from human colon cancer LIM1863 cells (1.5mg protein) (Subheading3.1) using the following strategies: ultrafiltration combined with ultracentrifugation at 100,000g (UC-Exos, 375g, Subheadings3.1 and
3.4); OptiPrep density gradient centrifugation (DG-Exos, 75g, Subheading3.6); and EpCAM immunoaffinity capture (IAC-Exos, 195g, Subheading3.7)
2 Materials
2.1 Cell Culture
and Concentrated
Culture Medium
(CCM) Preparation
183
Fig. 3 Exosome characterization. UC-Exos (a), DG-Exos (b), and IAC-Exos (c) were characterized by Western
blotting and electron microscopy. For Western blotting (Subheading3.8), each exosome preparation (10g) is
separated by 1D-SDS-PAGE, electrotransferred, and probed with exosome markers Alix, HSP70, and TSG101.
Exosomes from each purification strategy were negatively stained using uranyl acetate and viewed by electron
microscopy (Subheading3.9). The scale bar represents 100nm. Exosome isolation using OptiPrep density
gradient separation demonstrates each of the 12 isolated protein fractions (1.011.30g/ml) stained for protein
quantitation with SYPRO Ruby (d) and immunoblot analysis (10g) (e) revealing the exosomal marker Alix
predominantly detected in fraction 1.09g/ml (exosome-containing fraction)
Exosome
biogenesis
ESCRT
Accessory
ESCRT-II
ESCRT-III
ESCRT-I
Category
TSG101
VPS28
VPS37B
FAM125A
Protein name
7251
51160
79720
93343
1.7
3.2
7.1
5.6
24.3
1.2
1.2
19.4
1.2
4.1
1.2
1.2
2.5
2.3
3.1
1.3
4.4
1.2
1.3
1.2
4.4
13.3
25.5
15.4
1.2
4.8
11.7
7
7.8
4
4.4
4.1
1.3
4.8
1.3
6.7
8.6
2.5
3.9
3.3
2.5
4.8
7.2
5.3
5.3
2.8
1.5
1.6
2.7
28.8
6
1.2
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Exo
Vesicle
RSC (DG/UC)a RSC (IAC/UC)b RSC (IAC/DG)c Cartad pediae
Table 1
Relative quantification of proteins identified in UC-Exos, DG-Exos, and IAC-Exos using label-free-based spectral counting
MFI2
NT5E
XPNPEP2
4241
4907
7512
Syntenin
Trafficking
GTPase
and release
5878
5868
5869
7879
8766
9230
376267
57403
11021
382
27111
6386
RAB5C
RAB5A
RAB5B
RAB7A
RAB11A
RAB11B
RAB15
RAB22A
RAB35
ARF6
SDCBP2
SDCBP
CD9
CD63
CD81
CD82
CD151
TSPAN1
TSPAN14
TSPAN15
TSPAN3
TSPAN5
TSPAN6
TSPAN8
PLSCR1
PLSCR3
CEACAM5
1048
928
967
975
3732
977
10103
81619
23555
10099
10098
7105
7103
Membrane
5359
Architecture 57048
CD59
966
GPI-anchor
Tetraspanin
Category
RAB5C
RAB5A
RAB5B
RAB7A
RAB11A
RAB11B
RAB15
RAB22A
RAB35
ADP-ribosylation factor 6
Syndecan binding protein (syntenin) 2
Syndecan binding protein (syntenin)
Protein name
1.5
1.2
1.2
1
5.1
6.8
3.5
1.2
1.6
4.1
1.8
1.5
1.5
1.2
1.4
10.9
1.2
1.2
1.8
2.2
4.8
1.2
4.1
2.2
1.2
1.2
1.2
4.8
1.6
17.6
1.2
2
3.2
3.2
2
2.7
11.4
6.3
4.4
2.3
5.2
2.7
5.3
2.1
25.8
10.7
3
14.9
11.8
10.8
4.4
33.7
4
51.7
6.3
2.1
2.1
2.8
13
2.2
1.8
6.7
1.3
3.9
3.9
2
13.8
1.7
1.8
5.3
1.4
21.3
4.8
3.5
3.1
31.2
7.6
32.1
18
14.2
6.1
2
7
4.8
12.5
2.8
1.7
1.7
3.4
2.7
1.4
10
8.1
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
(continued)
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Exo
Vesicle
RSC (DG/UC)a RSC (IAC/UC)b RSC (IAC/DG)c Cartad pediae
VAMP3
9341
HLA-A
HLA-C
3105
3107
649853 HLA-A29.1
Syndecan 1
Syndecan 4
Vesicle-associated membrane protein 2
(synaptobrevin 2)
Vesicle-associated membrane protein 3
(cellubrevin)
Protein name
Relative spectral count ratio (Rsc) for proteins identified in DG-Exos, compared with UC-Exos
Relative spectral count ratio (Rsc) for proteins identified in IAC-Exos, compared with UC-Exos
c
Relative spectral count ratio (Rsc) for proteins identified in IAC-Exos, compared with DG-Exos
d
Presence of proteins in the exosome database ExoCarta (http://www.exocarta.org)
e
Presence of proteins in the extracellular vesicle database Vesiclepedia (http://microvesicles.org)
MHC
component
CD44
TFRC
NCSTN
FGA
FGB
FGG
MUC13
PTGFRN
SDC1
SDC4
VAMP2
6382
6385
6844
Syndecan
SNARE
Category
Table 1
(continued)
1.6
3.7
2.2
1.8
2.3
1.2
1.2
1.2
1.2
1.4
3
3.4
6
1.2
2.1
4.2
2.5
3.4
10.6
9.4
2.4
1.3
2.4
9.9
1.8
1.2
4.3
2
1.3
1.6
1.9
4.5
1.5
12.8
11.4
2.9
1.5
1.7
3.3
3.4
4
26
2.5
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Y
Exo
Vesicle
RSC (DG/UC)a RSC (IAC/UC)b RSC (IAC/DG)c Cartad pediae
187
Centrifugation
728
248
122
93
265
171
86
98
Immunoaffinity
627
50.0
Density
571
40.0
30.0
20.0
10.0
TSG101
8.0
CD9
CD63
CD81
20.0
CD82
CD151
TSPAN1
15.0
TSPAN3
TSPAN5
10.0
TSPAN6
TSPAN8
TSPAN14
5.0
0.0
VPS28
VPS37B
FAM125A
6.0
TSPAN15
55.0
FAM49B
50.0
VPS25/EAP20
45.0
VPS4B/SKD1B
2.0
VPS46A/CHMP1A
VPS46B/CHMP1B
VPS60/CHMP5
0.0
UC-Exos
DG-Exos
IAC-Exos
5.0
4.0
3.0
2.0
1.0
0.0
UC-Exos
DG-Exos
IAC-Exos
RAB5A
RAB5B
RAB5C
RAB7A
RAB11A
RAB11B
RAB15
RAB22A
RAB35
IAC-Exos
35.0
Normalised spectral count ratio (Nsc)
VPS24/CHMP3
DG-Exos
40.0
VPS32B/CHMP4B
VPS32C/CHMP4C
UC-Exos
60.0
FAM125B
VPS2B/CHMP2B
4.0
Normalised spectral
count ratio (Nsc)
25.0
Alix
VPS2A/CHMP2A
30.0
30.0
25.0
SDCBP
SDCBP2
20.0
SDC1
SDC4
VAMP2
15.0
VAMP3
CD44
TFRC
10.0
NCSTN
FGA
FGB
5.0
FGG
MUC13
PTGFRN
0.0
UC-Exos
DG-Exos
IAC-Exos
Fig. 4 Semi-quantitative normalized spectral count ratios of selected exosome proteins. For proteomic profiling
of the three different methods for exosome purification, the distribution of identified proteins is shown in a
three-way Venn diagram depicting number of proteins commonly observed in all three datasets (265), and
proteins that are unique to each purification strategy (a). The relative abundance of a protein (containing at
least two significant peptides) within a sample was estimated using semi-quantitative normalized spectral
count ratios (Nsc, Eq.1). For each individual protein, significant peptide MS/MS spectra were summated, and
normalized by the total number of significant peptide MS/MS spectra identified in the sample. The ratio serves
an indicator of protein abundance, i.e., the higher the ratio, the more abundant the protein within the sample.
Protein categories of interest included proteins associated with endosomal sorting complex required for transport (ESCRT) components and cargo selection/sorting in exosomes (b), Rab GTPases involved in docking and/
or fusion of MVBs with the plasma membrane (c), tetraspanins, important for EV formation (d), and other
proteins attributed to exosomal trafficking and recipient cell internalization (e)
188
Table 2
Ultracentrifuge, rotor, and consumable information for exosome and shed microvesicle purification
Speed ( g) Rotor
Exosome preparation
Large scale processing 100,000
OptiPrep
100,000
Small scale (washing) 100,000
10,000
2.3 Protein
Quantitation
2.3.1 Bradford Assay
189
2.3.3 Gel-Based
Densitometry Method
2.4 Shed
Microvesicle
(sMV) Isolation
190
2.6 OptiPrep
Density Gradient
Exosome (DG-Exo)
Isolation
2. Sterile/filtered PBS.
3. TLA-55 fixed angle (small scale/washing)Microcentrifuge
Polypropylene Tube (#357448, Beckman Coulter).
2.7 EpCAM
Immunoaffinity
Capture Exosome
(IAC-Exo) Isolation
191
2.9.1 Transmission EM
2. Sterile/filtered PBS.
4. Formvar coated 200mesh copper grids (ProSciTech).
5. 1% (w/v) aqueous uranyl acetate (ProSciTech).
6. Tecnai F30 electron microscope.
2.9.2 CryoEM
2.10 GeLC-MS/MS
192
3 Methods
In this methodology, two different approaches for isolation of
crude exosomes are presented: (a) concentrated culture medium
(CCM) preparation, and (b) the isolation of shed microvesicles
(sMVs) approach (Fig.1).
3.1 Cell Suspension
Culture andCCM
Preparation
193
1. Confluent cells are washed twice with ice cold PBS and lysed
with 2ml of SDS sample buffer for 10min on ice. Cells are
counted with a hemacytometer.
2. To remove DNA and insoluble cellular debris, lysates are
centrifuged at 435,000g for 30min (TLA-100.2 rotor),
with supernatants collected and stored at 80C (up to 6
months).
3.3 Protein
Quantitation
(See Note 11)
3.3.1 Bradford Assay
(BCA)
1. 5l sample aliquots (CCM/WCL) are solubilized in SDS sample buffer (10l), heated 95C for 5min (closed cap), briefly
centrifuged, and loaded on NuPAGE 412% (w/v) Bis-Tris
Precast gels.
2. 5l BenchMark Protein Ladder (1.7g/l) is loaded into
a separate well on NuPAGE 412% (w/v) Bis-Tris Precast
gels for quantitation. Note that a blank lane should be retained
for background subtraction.
3. Electrophoresis is performed at 150V for 1h in NuPAGE
1 MES running buffer.
4. After power off, the gel is removed from the tank and fixed in
50ml fixing solution for 30min on an orbital shaker and
stained with 30ml SYPRO Ruby for 40min, followed by
destaining in SYPRO destaining solution for 1h.
5. The gel is imaged on a Typhoon 9410 variable mode imager,
using a green (532nm) excitation laser and a 610BP30 emission filter at 100m resolution.
194
1. For isolation of sMVs, the cell culture medium (CM) is centrifuged at 4C (480g for 5min followed by 2,000g for
10min) to remove intact cells and cell debris. Separate tubes
should be used following each centrifugation stage.
2. The CM is further centrifuged at 10,000g for 30min to
remove sMVs (Table2, see Notes 2 and 13).
3. sMVs are resuspended in 1ml of sterile/filtered PBS, before
further centrifugation at 10,000g for 30min to obtain the
washed sMV pellet (see Note 14).
4. sMV pellet can be resuspended in either SDS sample buffer
(for PAGE analysis) or sterile/filtered PBS (EM imaging or
functional studies).
3.5 Ultracentrifuga
tion Exosome (UC-Exo)
Isolation
3.6 OptiPrep
Density Gradient
Exosome (DG-Exo)
Isolation
195
196
3.9.1 Transmission EM
2. Grids are washed twice with water for 5min, and stained with
1% (w/v) uranyl acetate in distilled water for 10min.
3. Imaging is performed at an acceleration voltage of 200kV
using a Gatan UltraScan 1000 (2k2k) CCD (charge-coupled
device) camera coupled to a Tecnai F30 (FEI, Netherlands)
electron microscope.
4. Typically 1020 fields of view are obtained.
3.9.2 CryoEM
3.10 Extraction,
Reduction, Alkylation
1. Exosome samples (20g) lysed in SDS sample buffer, and proteins separated by SDS-PAGE and visualized by Imperial
Protein Stain.
2. Individual gel lanes are placed on a clean glass plate and cut
into equal slices (202mm) using a GridCutter or clean scalpel and individual gel slices subjected to in-gel reduction,
alkylation, and trypsinization (see Note 24).
3. For each band prepare a 1.5ml microcentrifuge tube with
500l of 50mM ammonium bicarbonate/acetonitrile (1/1).
4. Cut excised bands (spots) into cubes (ca. 11mm if you used
1mm thick gel) and transfer gel pieces carefully into low
protein-binding microcentrifuge tubes.
197
1. For tryptic digestion, add enough trypsin to cover the dry gel
pieces (typically, 5060l depending on gel volume). Store all
samples immediately on ice. After 30min, check if all solution
is absorbed and add more trypsin, if necessary. Gel pieces
should be completely covered with trypsin.
2. Leave gel pieces for another 30min to saturate with trypsin
and add 20l of 50mM ammonium bicarbonate to cover the
gel pieces.
3. Place tubes with gel pieces into the thermostat oven and incubate samples overnight at 37C.
4. Withdraw supernatants to low protein binding tubes, use a
pipette with fine gel loader tip (re-use these tips for all following peptide collection steps).
5. Add extraction buffer (100l) to each tube and sonicate for
15min. Collect supernatants into the corresponding tube.
Repeat.
6. Dry down samples (covered with Vacufilm with four pin holes)
in vacuum centrifuge for 15min.
7. For STAGE-Tip desalting, prepare as many desalting columns
as necessary by punching out small disks [2, 3] of C18 Empore
filter using a 22G flat-tipped syringe and ejecting the disks
into P200 pipette tips. Ensure that the disks are securely
wedged in the bottom of the tip. Careful preparation of these
STAGE-Tip devices will ensure effective filtration.
8. Condition columns by forcing methanol through (50l) and
check whether the STAGE-Tips are leaky.
198
3.13 Database
Searching andProtein
Identification
199
1. The relative abundance of a protein within a sample is estimated using semi-quantitative normalized spectral count ratios
(Nsc). For each individual protein, significant peptide MS/MS
spectra (i.e., ion score greater than identity score) are summated, and normalized by the total number of significant MS/
MS spectra identified in the sample (Eq.1)
N sc = (n + f ) / (t - n + f ) ,
(1)
200
Rsc = (nB + f
) (t A - n A + f ) / (n A + f ) (t B - nB + f ) ,
(2)
4 Notes
1. Cells should be grown to reach 7080% confluency for adherent cells, or 6070% of their maximum concentration for cells
grown in suspension. The advantage of utilizing as many cells
as possible ensures a concentrated CM and enrichment of EVs.
When washing adherent cells (typically 10ml), the CM should
be removed and replaced with exosome-production medium
(Cell culture medium B, typically 20ml).
2. Resource for rotor speed and conversion:
https://www.beckmancoulter.com/wsrportal/wsr/researchand-discovery/products-and- services/centrifugation/rotors/
index.htm?t=3
Resource for tubes and adapters:
https://www.beckmancoulter.com/wsrportal/wsr/researchand-discovery/products-and-services/centrifugation/tubesand-adapters/index.htm
3. To isolate CM from adherent cells, the cell culture medium
should be collected under sterile conditions and transferred to
50ml polypropylene centrifuge tubes. CM should be centrifuged at 480g for 5min (the supernatant transferred using a
pipette to a separate 50ml polypropylene centrifuge tubes)
and 2,000g for 10min. The supernatant is collected using a
pipette and stored at 4C.To isolate CM from cell suspensions, the cell culture medium should be collected under sterile
conditions and transferred to 50ml polypropylene centrifuge
tubes. CM should be initially centrifuged at 5min at 200
400g, 4C, before following the protocol for adherent cells
(480g, 5min and 2,000g, 10min).
201
202
203
204
205
206
207
Inaddition, various studies employ differences in their purification and characterization, which inevitably introduces and
potentially confounds interpretation of biochemical data.
26. In this chapter, label-free proteomic quantification was
employed based on spectral counting (SpC). SpC counts the
number of spectra identified for a given peptide in different
biological samples and then integrates the results for all measured peptides of the protein(s) that are quantified [48]. SpC
has become a commonly used approach for measuring protein
abundance in label-free shotgun proteomics.
Acknowledgements
This work was supported, in part, by the National Health &
Medical Research Council of Australia (program grant #487922
(RJS), project grant #1057741 RJS).
Conflict of interest: The authors declare no conflict of interest.
References
1. El Andaloussi S, Mager I, Breakefield XO etal
(2013) Extracellular vesicles: biology and
emerging therapeutic opportunities. Nat Rev
Drug Discov 12:347357
2. Rak J, Guha A (2012) Extracellular vesicles
vehicles that spread cancer genes. Bioessays
34:489497
3. Raposo G, Stoorvogel W (2013) Extracellular
vesicles: exosomes, microvesicles, and friends.
J Cell Biol 200:373383
4. Simpson RJ, Lim JW, Moritz RL etal (2009)
Exosomes: proteomic insights and diagnostic
potential. Expert Rev Proteomics 6:267283
5. Tauro BJ, Greening DW, Mathias RA etal
(2012) Comparison of ultracentrifugation,
density gradient separation, and immunoaffinity capture methods for isolating human colon
cancer cell line LIM1863-derived exosomes.
Methods 56:293304
6. Tauro BJ, Greening DW, Mathias RA etal
(2013) Two distinct populations of exosomes
are released from LIM1863 colon carcinoma
cell-derived organoids. Mol Cell Proteomics
12:587598
7. Ji H, Erfani N, Tauro BJ etal (2008) Difference
gel electrophoresis analysis of Ras-transformed
fibroblast cell-derived exosomes. Electro
phoresis 29:26602671
8. Ji H, Greening DW, Barnes TW etal (2013)
Proteome profiling of exosomes derived from
human primary and metastatic colorectal
208
209
Chapter 16
Chloroplast Isolation and Affinity Chromatography
for Enrichment of Low-Abundant Proteins in Complex
Proteomes
Roman G. Bayer, Simon Stael, and Markus Teige
Abstract
Detailed knowledge of the proteome is crucial to advance the biological sciences. Low-abundant proteins
are of particular interest to many biologists as they include, for example those proteins involved in signal
transduction. Recent technological advances resulted in a tremendous increase in protein identification
sensitivity by mass spectrometry (MS). However, the dynamic range in protein abundance still forms a
fundamental problem that limits the detection of low-abundant proteins in complex proteomes. These
proteins will typically escape detection in shotgun MS experiments due to the presence of other proteins
at an abundance several-fold higher in order of magnitude. Therefore, specific enrichment strategies are
required to overcome this technical limitation of MS-based protein discovery. We have searched for novel
signal transduction proteins, more specifically kinases and calcium-binding proteins, and here we describe
different approaches for enrichment of these low-abundant proteins from isolated chloroplasts from pea
and Arabidopsis for subsequent proteomic analysis by MS. These approaches could be extended to include
other signal transduction proteins and target different organelles.
Key words Chloroplast isolation, Affinity chromatography, Mass spectrometry, Proteomics,
Organellar proteome, ATP-binding protein, Calcium-binding protein
Introduction
Organellar proteomics has become a strong focus in (plant) biology over the last years due to several aspects: Organelles perform
key activities for cellular metabolism and provide the basis for
growth and production of valuable compounds [1]. Moreover, the
use of isolated organelles as starting material does already significantly reduce the complexity of the sample as compared to total
cell extracts. However, in the case of leaves the difference between
isolated chloroplasts and total leaves becomes rather small in terms
of their overall protein composition, thus calling for additional
enrichment strategies for the identification of low-abundant
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_16, Springer Science+Business Media New York 2015
211
212
213
Fig. 1 Scheme of the work-flow and typical profile of a size exclusion chromatography (SEC) of stroma extracts
from isolated chloroplasts on a Superdex S200 gel filtration column. (a) Scheme of the work-flow to stroma
extract. (b) Scheme of a typical elution profile of stromal proteins on a Superdex S-200 size exclusion column.
X-axis shows ml of eluting sample. Y-axis shows OD280 indicating protein content. Fractions eluting after the
large peak of Ribulose 1,5 Bisphosphate Carboxylase/Oxygenase (RuBisCO) are pooled and subjected to: MS
mass spectrometry, IEX ion exchange chromatography, AC affinity chromatography, HIC hydrophobic interaction chromatography; (c) SDS PAGE of 1.44 ml fractions collected from 73 ml to 96 ml of a Supdex S200 16/60
gel filtration column. T total stromal protein extract loaded
Materials
214
2. Leaves from Pea: Pea seedlings are grown for 89 days under
long day conditions (photoperiod of 8 h dark/16 h light at
~70 mol/m2s; 21 C +/5; humidity 7090 %) on vermiculite.
2.2 Chloroplast
Isolation from Pea
(Pisum sativum)
2.3 Chloroplast
Isolation from
Arabidopsis
2.4 Solutions
for Chloroplast Lysis
215
2.8 Hydrophobic
Interaction
Chromatography
(HIC)On
Phenyl-Superose
2.9 ATP/Purvalanol
B Affinity
Chromatography
216
2.11 Eu3+-IDA
Column Affinity
Chromatography
3.1 Chloroplast
Isolation from Pea
(Pisum sativum)
(See Note 6)
217
1. Mix 5 l isolated chloroplast suspension with 5 ml 80 % acetone and centrifugate for 2 min at 3,000 g. Subsequently,
measure the OD645 and OD663 of the supernatant (using glass
or quartz cuvettes) and determine chlorophyll concentration
using the following formula (see Note 11):
218
1. Incubate chloroplasts (amount equivalent to ~20 mg of chlorophyll) in ~1.5 volumes of hypotonic osmotic lysis buffer for
5 min on ice to achieve osmolytic bursting of the envelopes.
2. Centrifuge for 6 min at 12,000 g at 4 C and transfer the
supernatant containing the stromal content to a 50 ml tube.
3. Re-suspend the chloroplast pellet in ~5 ml osmotic lysis buffer
and repeat the extraction step once.
4. Pool the stromal protein extracts and keep on ice until further
treatment.
The thylakoid pellet can be re-suspended in ~5 ml THY buffer
and stored at 80 C for future usage.
1. Pool the fractions of interest from the S200 gel filtration and
concentrate them to a volume of ~500 l using a Centriprep
Centrifugal Filter Unit (NMWL: 10 kDa). After clarification
by centrifugation for 3 min at 16,100 g and 4 C apply the
supernatant to a S75 gel filtration column and perform size
exclusion chromatography on an FPLC system at a flow rate of
0.4 ml/min using solution S200-A as running buffer.
Fractionate the eluate and conduct protein content measurements and/or functional assays as desired. Store protein samples at 4 C until further treatment.
219
3.8 Hydrophobic
Interaction
Chromatography
(HIC)On
Phenyl-Superose
3.9 ATP/Purvalanol
B Affinity
Chromatography
220
3.11 Eu3+-IDA
Column Affinity
Chromatography
Notes
1. Starch granules in the chloroplast will contribute to breaking
of chloroplasts during handling. Therefore prepare chloroplasts in the morning, when starch levels in the chloroplast are
lowest.
2. No titration necessary. Optimal pH is reached by mixing the
two buffer components (MOPS and Tris).
221
222
Acknowledgements
Work in the authors lab is supported by grants from the Austrian
Science Fund (FWF) to MT (P 23435-B12 and P 25359-B21) and
the Marie Curie Initial training network (ITN) CALIPSO (GA
ITN 2013-607607) from the European Union.
References
1. Rolland N, Curien G, Finazzi G, Kuntz M,
Marechal E, Matringe M et al (2012) The biosynthetic capacities of the plastids and integration between cytoplasmic and chloroplast
processes. Ann Rev Genet 46:233264
2. Jarvis P, Lopez-Juez E (2013) Biogenesis and
homeostasis of chloroplasts and other plastids.
Nat Rev Mol Cell Biol 14:787802
3. Shapiguzov A, Vainonen JP, Wrzaczek M,
Kangasjarvi J (2012) ROS-talkhow the apoplast, the chloroplast, and the nucleus get the
message through. Front Plant Sci 3:292
4. Nomura H, Komori T, Uemura S, Kanda Y,
Shimotani K, Nakai K et al (2012) Chloroplastmediated activation of plant immune signalling in Arabidopsis. Nature Commun 3:926
14.
15.
16.
17.
18.
19.
223
Chapter 17
Depletion ofRuBisCO Protein Using theProtamine
Sulfate Precipitation Method
RaviGupta andSunTaeKim
Abstract
Ribulose-1,5-bisphosphate carboxylase/oxygenase (RuBisCO) is a major high-abundant protein (HAP) in
the plant leaves which hinders analysis of low-abundant proteins (LAP). In this chapter, we describe a
highly simple RuBisCO depletion method using protamine sulfate (PS). Addition of 0.1% PS is sufficient
to precipitate the RuBisCO from the leaf extracts of diverse plants including monocots and dicots. Our
results of S
DS-PAGE, Western blotting, and two-dimensional gel electrophoresis showed that both large
and small subunits of RuBisCO were precipitated in the pellet fractions, while LAPs were enriched in the
supernatant fraction after PS precipitation.
Key words RuBisCO, Protamine sulfate, High-abundant proteins, Low-abundant proteins, Two-
dimensional gel electrophoresis
1 Introduction
In the last two decades, the focus of research has been shifted from
genomics to proteomics due to the fact that proteins are the real
executors of the gene expression and thus can provide a better
picture of functional aspects of the cells [1]. As plants perceive
signals from the environment through leaves, one of the major
goals of the scientific community is to dissect the components of
signaling cascades which are initiated in response to stresses or
other environmental cues like photoperiodism, etc, by analyzing
the leaf proteome. However, the major bottleneck for analysis of
leaf proteome is presence of a high-abundant protein (HAP)
RuBisCO, which mask the expression and identification of lowabundant proteins (LAP) and thus hinders their analysis [2]. Mass
spectrometry-based identification of the leaf proteins often results
in the repeated identification of large and small subunits of
RuBisCO, resulting in a noteworthy loss of time and money [3].
RuBisCO is present in the order of 105107 molecules per cell
and therefore, mask the LAPs like phosphatase, kinase, t ranscriptional
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI10.1007/978-1-4939-2550-6_17, Springer Science+Business Media New York 2015
225
226
RaviGupta andSunTaeKim
2 Materials
2.1 Protein
Extraction, RuBisCO
Precipitation,
andProtein
Quantitation
Components
2.2 SDS-PAGE
Components
227
2.3 Immunoblotting
Components
228
RaviGupta andSunTaeKim
3 Methods
3.1 Extraction
ofTotal Proteins
andDifferential
Precipitation
ofRuBisCO
229
Add 1 mL 1% PS solution to 9 mL of
supernatant and incubate on ice for 30 min
Centrifuge at 12,000 g for 10 min at 4 C
230
RaviGupta andSunTaeKim
Rice
M
Arabidopsis
Rice
T
170
130
100
70
P
APx1
APxb
55
40
Cu/Zn SOD
35
DHAR
25
Mn SOD
15
SalT
RubL
10
pH 4
T
pH 7 pH 4
S
pH 7 pH 4
P
pH 7
408
516
139
Rice
Arabidopsis
590
626
283
Fig. 2 Effect of PS on RuBisCO precipitation and corresponding enrichment of LAPs. (a) After 0.1% PS precipitation, equal amount of total (T), supernatant (S) and pellet (P) proteins of rice and Arabidopsis were loaded on
the 12% SDS-PAGE.LAPs were enriched in the supernatant fractions while RuBisCO was enriched in the pellet
fractions. (b) Western blot analysis of rice proteins to show enrichment of LAPs in the supernatant fraction and
RuBisCO depletion in the pellet fraction. 2-DE gel profiles of rice (c) and Arabidopsis proteins (d) after 0.1%
PS precipitation confirming enrichment of LAPs in supernatant and precipitation of RuBisCO in pellet fractions.
Total number of spots identified in 2-DE gels are mentioned at the bottom of the corresponding gels
231
4 Notes
1. Addition of NP-40in the protein extraction buffer solubilizes
the membranes which results in the extraction of membrane
bound proteins also and thus increases the protein yield. In
addition, NP-40 enhances the protein solubility.
2. Initially concentrated HCl can be used to bring the pH of Tris
near 8. After that, diluted HCl should be used to bring the
required pH.While handling HCl, always wear gloves.
3. As phenol is highly volatile and caustic, perform all steps under
Fume Hood only. Always wear gloves and mask while handling
phenol. Avoid contact of phenol to the skin.
232
RaviGupta andSunTaeKim
Acknowledgement
This work was supported by a grant from the Next-Generation
BioGreen 21 Program (SSAC, grant#: PJ011070032015), Rural
Development Administration (RDA), Republic of Korea.
References
1. Agrawal GK, Rakwal R (2011) Rice proteomics:
a move towards expanded proteome coverage
to comparative and functional proteomics
uncovers the mysteries of rice and plant biology.
Proteomics 11:16301649
2. Rose JKC, Bashir S, Giovannoni JJ, Jahn MJ,
Saravnan RS (2004) Tackling the plant proteome: practical approaches, hurdles and
experimental tools. Plant J 39:715733
3. Schulze WX, Usadel B (2010) Quantitation in
mass-spectrometry-based proteomics. Annu
Rev Plant Biol 61:491516
4. Malcevschi A, Marmiroli N (2012) Plant protein analysis. In: Heazlewood J (ed) Proteomic
applications in biology. InTech Manhattan,
NewYork, ISBN: 978-953-307-613-3
5. Krishnan HB, Natarajan SS (2009) A rapid
method for depletion of Rubisco from soybean
(Glycine max) leaf for proteomic analysis of
lower abundance proteins. Phytochemistry
70:19581964
233
12. Laemelli UK (1970) Cleavage of structural
proteins during the assembly of the head of
bacteriophage T4. Nature 227:680685
13. Kim ST, Cho KS, Yu S, Kim SG, Hong JC,
Han CD, Bae DW, Nam MH, Kang KY (2003)
Proteomic analysis of differentially expressed
proteins induced by rice blast fungus and elicitor in suspension-cultured rice cells. Proteomics
3:23682378
Chapter 18
Step-by-Step Preparation of Proteins for Mass
Spectrometric Analysis
Thomas Franz and Xinping Li
Abstract
Nowadays, identification of proteins from biological samples by mass spectrometry is widely used. In principle
there are two scenarios. Proteins are pre-fractionated in some way, e.g. by gel electrophoresis or are
analyzed as complex mixture (shot gun). Shot gun proteomics became recently more popular, because of
technological developments on the mass spectrometer side which allows now the identification of several
thousand proteins from complex biological matrix. However, in many cases it is still useful to separate
proteins first in a gel. But not only mass spectrometer technology made progress. This is also true for the
sample preparation. Recently, protocols and techniques were developed which make the analysis of starting
material in the low microgram range possible and also simplify the whole procedure. Detailed protocols
will be described allowing also inexperienced beginners to get good results.
Key words Shotgun proteomics, In-solution digest, In-gel digest, FASP, STAGE-tip, OASIS, Mass
spectrometry, Sample preparation
Introduction
Mass spectrometry became a valuable tool in biological analysis of
proteins. During the recent years, the sensitivity of the instruments
improved dramatically and this development is still ongoing. We
are close being able to analyze whole proteomes of low microgram
range in a single shot (shotgun analysis). The sample preparation
becomes more and more important. It should be quick, relatively
easy to perform, reproducible and no or minimal loss of sample.
Also recently protein sample preparation for mass spectrometry has
made very much progress toward handling of small samples and
easiness. Multistep procedures are usually not suited for starting
material in the low microgram range. The loss of material during
sample preparation is substantial. Therefore, one-step procedures
are preferred. Inexperienced scientific staff is facing the problem
of old protocols in the literature, incomplete handling procedures
or protocols are scattered throughout several publications making
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_18, Springer Science+Business Media New York 2015
235
236
Materials
Prepare all solutions and buffers using ultrapure water of 18 M cm
at 25 C, use LC-MS grade solvent and reagents of highest purity.
Follow all waste disposal regulations when disposing waste
materials.
237
2.3 Filter-Aided
Sample Preparation
(FASP) [6, 7]
2. Acetonitrile.
3. 30 mM potassium ferricyanide K3[Fe(CN)6] in water.
4. 100 mM sodium thiosulfate in water.
2.4 One-Step
In-Solution Protein
Solubilization,
Reduction, Alkylation
and In-Solution
Digest [8]
2.5 One-Step
Guanidine Method
for Large-Scale
Analysis
2.6 C. elegans
Sample Preparation
238
2.8 Pre-fractionating
with Styrene
Divinyl Benzene
(SDB-RPS) [8]
Methods
1. Use spot cutter to excise the gel plugs from the 2D-gel directly
into the OASIS MTP well. Recommended cutting tip size is
1.5 mm in diameter. Alternatively cut a band by scalpel directly
from the gel, place it at the upper rim of an OASIS MTP well
and cut it in 1 mm3 pieces. Then push them down into the well
by a pipette tip.
2. Remove the liquid from the excision process into the waste
plate (24 10 mL MTP plate) using the positive pressure-96
stand (PPS) (if not otherwise stated 15 PSI of nitrogen pressure was always used).
3. Add 70 L acetonitrile (ACN) and push the gel pieces down
into the solution if necessary.
239
4. Wait for 10 to 15 min until the gel plugs are fully dehydrated
(they will become small and white) (see Note 3).
5. Using the PPS, remove all liquid into waste plate.
3.1.2 Coomassie Gel
Destaining
3.1.4 Reduction
1. Rehydrate gel pieces in 100 L reduction solution and incubate for 1 h at room temperature to reduce the cysteine bridges
in the protein (see Note 5).
2. Remove excess liquid using the PPS.
3. Incubate the gel pieces with 100 L ACN and wait for
1015 min until the gel pieces have dehydrated (see Note 3).
4. Remove all liquid with the PPS.
3.1.5 Alkylation
3.1.6 Application
of Trypsin
1. Remove all liquid with the PPS, then rehydrate the gel pieces
in 33 L digestion solution (contains trypsin) at room
temperature.
2. After 20 min remove remaining buffer with the PPS.
240
1. With the PPS remove the digest solution into the waste plate
(peptides will bind to the HLB column).
2. Wash the OASIS HLB column with 70 L 0.1 % FA.
3. Exchange the waste plate for the collection plate (0.5 mL/
well) (see Note 6).
4. Add 50 L of extraction solution to each well of the Oasis plate
and wait 20 min, then remove the extraction solution into collection plate with the PPS.
5. Add 50 L of acetonitrile to each well and wait 10 min, then
remove the extraction solution into collection plate with the
PPS.
6. Completely dry eluted peptides in the collection plate using a
vacuum centrifuge at 45 C for 60 min.
7. Freeze the dried extracts (peptides) at 20 C or 80 C (for
storage >1 month).
8. Re-dissolve the peptides in 7 L 0.1 % formic acid for ESI-MS
(see Note 7).
3.2 Filter-Aided
Sample Preparation
(FASP) [6, 7]
241
9. Collect the flow through (in case filter was leaking) and add
250 L of buffer B to the residue in the filter.
10. Centrifuge at 12,000 g for 15 min.
11. Add again 250 L buffer B and centrifuge at 12,000 g for
15 min.
12. Alkylation step: add 300 L of buffer C, mix with pipette
carefully and wait 20 min at room temperature in the dark
(see Note 9).
13. Centrifuge at 12,000 g for 15 min.
14. Add 250 L buffer B and centrifuge again (12,000 g for
15 min).
15. Add 200 L buffer D, centrifuge again and repeat this step
once more.
16. Then add 50 L buffer D to the sample followed by 3 g of
Trypsin and mix carefully by pipetting up and down.
17. Fill a plastic sample box partly (two rows) with water and the
digest solution containing closed filter devices are placed in an
empty space. With closed lid place it into an incubator at 37 C
overnight.
18. Following the digest add 200 L buffer D and centrifuge at
10,000 g for 15 min.
19. Place the flow through in a vacuum concentrator for 30 min at
45 C. Around 100 L should be left.
20. Take 2 L of the digested sample for a NanoDrop OD 260/280
measurement [11] with water as reference (see Note 10).
3.3 One-Step
In-Solution Protein
Solubilization,
Reduction, Alkylation
and In-Solution
Digest [8]
This procedure is particular useful for shot gun proteomics of samples like a single fly head (~3 g protein) as described here.
1. Load one single Drosophila melanogaster head to the bottom
of a 0.5 mL centrifugation tube (Eppendorf tube).
2. Add 5 L of Guanidine buffer (see Note 2).
3. Heat sample for 10 min at 95 C.
4. For the homogenization of the single head use a Bandelin
Sonoplus Ultrasonic device with a 1.5 mm Tip. Apply a total of
1.0 kJ energy at 50 % amplitude, which takes about 1 min. At
the same time the head is also mechanically crushed with the
tip of the US stick which acts like a pistil.
5. The homogenate is centrifuged to the bottom of the tube with
a minitable centrifuge at 1,500 g and heated again for 10 min
at 95 C.
6. Again homogenize for 30 s at 50 % amplitude by the Bandelin
Ultrasonic device.
242
For larger samples from 100 g to several mg of protein the onestep procedure in Subheading 3.3 needs to be modified.
1. To ten whole Drosophila melanogaster bodies (about 300 g)
add 100 L of Guanidine buffer.
2. Mechanically homogenize it with an electric mortar in a
1.5 mL centrifugation tube.
3. Boil the mixture for 5 min at 95 C in an Eppendorf Thermo
mixer at 600 rpm.
4. Homogenize further by sonication (five cycles 30 s, 30 s, position high) in a BioruptorTM plus in ice cold water.
5. The homogenate is centrifuged to the bottom of the tube with
a mini table centrifuge at 1,500 g.
6. Boil again for 5 min at 95 C in a Eppendorf Thermo mixer at
600 rpm.
7. Homogenize again with five cycles 30 s, 30 s, position high in
a BioruptorTM plus in ice cold water.
8. Dilute with 900 mL buffer A.
9. Centrifuge for 10 min at 15,000 g.
10. Place 100 L of supernatant on an Omega 30 kDa filter device.
11. Add 5 g Trypsin at a concentration of 1 g/L in 50 mM
acetic acid and incubated overnight at 37 C as described in
Subheading 3.2.
12. Next day centrifuge the tube at 15,000 g for 15 min at room
temperature.
13. Take 2 L of the flow through and measure the protein concentration with NanoDrop OD 260/280 as described and
proceed with STAGE tip procedure Subheading 3.6.
3.5 Preparation
of C. elegans Sample
243
Fig. 1 Centrifuge adapters for 200 L tips and its assembly with a 2 mL tube
244
Fig. 2 Spring-loaded syringe for quick preparation of large batches of STAGE tips
245
Fig. 3 Syringe with orange adapter for the 200 L tip for elution of peptides
3.7 Pre-fractionation
of Protein Digests
with Styrene
Divinyl Benzene
(SDB-RPS) [8]
(see
Notes
1. 0.1 % trifluoroacetic acid in the case of MALDI preparation.
2. It is important to produce the Guanidine/TCEP/CAA/Tris
buffer always fresh, because the components react with each other
over time. First dissolve 5.73 g of GdmCl in 10 mL of Milli-Q
water, then weight the other chemicals into a separate container
and dissolve them with 3,000 L of the GdmCl solution completely and mix it with the rest of the GdmCl solution.
3. It happens that acetonitrile runs through the HLB column
without pressure. In this case just add acetonitrile again after
5 min.
246
4. If gel pieces are not destained, repeat the steps. More than
twice destaining will not improve the result.
5. The reaction can be shortened to 30 min by incubating at 37 C.
6. The PPS stand requires a certain height of the collection plate.
We usually use a stack of three 0.5 mL/96-well collection
plates, where the lower ones are just spacers.
7. For MALDI-MS use 5 L 0.1 % TFA.
8. Some filters may even leak from beginning. To test that, add
200 L buffer B to a couple of filters and centrifuge at 12,000 g
for 5 min. Compare the amount of liquid left in the filter.
Discard the filters with no or significantly less liquid. It is not
unusual that during the procedure the filter starts leaking too.
9. DTT for reduction is in the lysis buffer.
10. This is to calculate how much one needs to take for STAGE tip
peptide desalting and cleaning for MS analysis. Three layers of
membrane have a capacity of 10 g.
11. Dilute at least 1:10, otherwise trypsin digest will not work.
12. 100 L of wet worm pellet corresponds to ~1.8 mg peptides.
13. For SILAC samples with only Lysine label use LysC.
14. Also a hole in the lid of a 2 mL Eppendorf tube with the right
diameter is fine.
15. The amount is calculated from the NanoDrop OD 260/280
measurement.
16. Before you connect the syringe pull the plunger out to the
maximum volume.
17. To squeeze the extraction solution out needs some force.
Hold the tip and press it tightly against the syringe to avoid it
falling off.
References
1. Eckerskorn C, Lottspeich F (1989) Internal
amino acid sequence-analysis of proteins separated by gel-electrophoresis after tryptic digestion in polyacrylamide matrix. Chromatographia
28:9294
2. Rosenfeld J, Capdevielle J, Guillemot JC,
Ferrara P (1992) In-gel digestion of proteins
for internal sequence analysis after one- or twodimensional gel electrophoresis. Anal Biochem
203:173179
3. Franz T, Li X (2012) The OASIS HLB Elution
plate as a one-step platform for manual highthroughput in-gel digestion of proteins and
peptide desalting. Proteomics 12:24872492
247
Chapter 19
Identification of Protein N-Termini Using TMPP
or Dimethyl Labeling and Mass Spectrometry
Jingjing Deng, Guoan Zhang, Fang-Ke Huang, and Thomas A. Neubert
Abstract
Determination of a proteins N-terminal sequence can be important for the characterization of protein
processing. To increase the confidence of protein N-terminal identification, chemical derivatization of the
N-terminal amine group by (N-Succinimidyloxycarbonylmethyl)tris(2,4,6-trimethoxyphenyl)phosphonium bromide (TMPP) or dimethyl labeling followed by mass spectrometric analysis is commonly performed. Using this approach, proteins can be separated by SDS-PAGE, and the protein N-terminus of
interest is labeled by TMPP or dimethyl in-gel before tryptic digestion and LC-MS analysis. The N-terminus
of a protein can thus be easily identified because the N-terminal tryptic peptides are preferentially labeled.
Peptides with N-terminal derivatization produce a better fragmentation pattern during tandem mass spectrometric analysis, which significantly facilitates sequencing of these peptides.
Key words TMPP labeling, Dimethyl labeling, N-terminal sequence analysis, Mass spectrometry
Introduction
Identification of the N-terminal sequence of an intact or cleaved
protein is crucial for its biochemical and structural characterization
[1]. In conventional shotgun proteomic analysis, it is difficult to
identify protein N-termini due to infrequent detection of protein
N-terminal peptides. Several methods have been developed based
on the N-terminal labeling to overcome this limitation, including
TMPP and dimethyl labeling [2, 3].
The TMPP labeling approach is straightforward and has been
successfully applied to different proteins [4]. Two characteristics of
this labeling reagent promote the sensitivity of the method: (1) TMPP
labeling introduces a permanent positive charge resulting in an
enhanced ionization efficiency and thus a better detection of lowabundance peptides; (2) the hydrophobic TMPP group shifts the
retention time of TMPP-derivatized peptides in reversed-phase
chromatography toward a less complex part of the chromatogram,
increasing the sensitivity of detection, especially for short N-terminal
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_19, Springer Science+Business Media New York 2015
249
250
Reduction
and alkylation
In-gel digestion
C
Intensity
TMPP or
dimethyl labeling
m/z
Data analysis
LC-MS/MS
Fig. 1 Flow chart showing the process of protein separation by SDS-PAGE, N-terminal labeling, in-gel digestion, and LC-MS/MS
peptides that otherwise would not be retained on the column [3, 5].
In addition, TMPP is fully compatible with all standard detergents,
chaotropic agents, and reducing conditions used for protein extraction in proteomics [5], which makes TMPP labeling a commonly
used method for protein N-terminal sequencing.
An alternative method based on chemical labeling is dimethylation, which labels peptide N-termini and -amino groups of lysine
with water-soluble formaldehyde via reductive methylation [68].
In MS/MS analysis, this labeling strategy provides a signal enhancement for the a1 and yn 1 ions, which are not detectable from most
of the nonderivatized fragments [8]. Because of its simplicity, as
well as low cost, dimethyl labeling is another promising strategy for
protein N-termini identification.
Due to the relatively simple experimental design (classical 1D
SDS-PAGE, a single chemical derivatization step performed at the
protein level in-gel, followed by protein digestion and LC-MS/
MS, as illustrated in Fig. 1), the two chemical derivation approaches
can be readily used to analyze both purified proteins and highly
complex protein mixtures [5]. Here, N-terminal identification
using TMPP and dimethyl labeling of purified protein hemoglobin
and BSA is described.
Materials
Prepare all solutions using HPLC grade solvents unless indicated
otherwise. Prepare and store all reagents at room temperature
unless indicated otherwise. Diligently follow all waste disposal regulations when disposing waste materials.
2.1
SDS-PAGE
251
2.2 Reduction
and Alkylation
of Excised Protein
Bands
2.3 N-Terminal
Protein Labeling
and Protein Digestion
1. N-terminal labeling solution I: 100 mM succinimidyloxycarbonylmethyl tris(2,4,6-trimethoxyphenyl)phosphonium bromide (TMPP-Ac-OSu). Dissolve 100 mg TMPP in 1.3 ml
ACN (see Note 5).
2. Reducing solution II: 260 mM cyanoborohydride. Dissolve
16.3 mg sodium cyanoborohydride in 1 ml water (see Note 6).
3. Acetate buffer: 100 mM sodium acetate, pH 5. Mix 14.8 ml
0.2 M acetic acid solution (11.5 ml acetic acid in 1 l H2O) and
35.2 ml 0.2 M sodium acetate solution (16.4 g sodium acetate
in 1 l H2O), and fill up to 100 ml with water.
4. N-terminal labeling solution II: 4 % formaldehyde. Dilute a
30 % formaldehyde solution with water.
5. HEPES buffer: 0.1 M HEPES, pH 8.2. Dissolve 23.8 mg
HEPES in 1 ml water. Adjust pH value to 8.2 with sodium
hydroxide.
2.4 Mass
Spectrometric
Analysis
252
Methods
Carry out all procedures at room temperature unless otherwise
specified.
3.1
SDS-PAGE
3.2 Reduction
and Alkylation
of Excised Protein
Bands
253
3.4 Mass
Spectrometric
Analysis
254
2. The Q-Exactive mass spectrometer was operated in datadependent mode using a top ten method. Full MS scans were
acquired in the Orbitrap mass analyzer over a range of 300
1,650 m/z with resolution 70,000 (m/z 200). The target
value was 3.00E + 06. The ten most intense peaks with charge
state 2 were fragmented in the HCD collision cell with normalized collision energy of 27 %, and tandem mass spectra
were acquired in the Orbitrap mass analyzer with resolution
17,500 (m/z 200). The target value was 1.00E + 06. The ion
selection threshold was 1.70E + 04 counts, and the maximum
allowed ion accumulation times were 20 ms for full MS scans
and 60 ms for MS/MS [9].
3. Peak lists were created and searched by Mascot Distiller.
Software setting: semiTrypsin with one missed cleavage; carbamidomethyl (C) as fixed modification; oxidation (M) as variable modification. For TMPP labeling, a mass addition of
572.1811(C29H33O10P) (N-terminus, K and Y) must be
included as a variable modification. For dimethyl labeling, a
mass addition of 28.0313 (C2H4) (N-terminus, K, P and R) as
a variable modification must be included. Peptide tolerance
was set to 10 ppm for peptides and MS/MS tolerance was
0.03 Da (see Note 17).
4. MS/MS spectra of TMPP-labeled peptide VLSPADK
(Hemoglobin A) and TMPP-labeled peptide DTHKSEIAHR
(BSA) are shown in Fig. 2. As illustrated in Fig. 3, dimethyllabeled peptide VLSPADK or DTHKSEIAHR identified the
same N-terminal sequences of Hemoglobin A and BSA respectively. We also found dimethyl labeling to be more sensitive
than TMPP labeling (Detection limit: 0.1 pmol BSA for
dimethyl; 3 pmol BSA for TMPP).
Notes
1. Add 50 l 2-mercaptoethanol to 950 l Laemmli sample buffer. Protein sample is mixed 1:1 (v/v) with the sample buffer.
2. Gel format: 8.6 6.7 cm (W L), 10-well comb for 50 l
samples.
3. SDS is a detergent. Handle it with care and take measures to
prevent inhalation of airborne SDS. It is best to weigh SDS in
a fume hood. Check the pH of the 10 solution. The pH of
the buffer should be between 8.1 and 8.5. Do not adjust pH
with acid or base.
4. Iodoacetamide powder is stored at 2 to 8 C and protected
from light. Iodoacetamide solution is made freshly before use.
255
TMPP-Ac - V L S P A D K
a5
b1b2b3b4 b6
y1
147.11
100
b3
872.37
[M+2H]2+:
651.3027 m/z
y2
262.14
90
a5
1012.47
Relative Abundance
80
b6
1155.49
70
60
50
40
30
y4
430.23
20
b2
785.34
b1
672.26
y3
333.18
10
b4
969.43
0
100
200
300
400
500
600
700
m/z
800
900
1000
1100
1200
1300
y6 y4 y3 y2 y1
TMPP-Ac - D T H K S E I A H R
a2 a3
b1 b2 b3 b4 b6
b1
688.21
b
100
90
[M+4H]4+:
442.1996 m/z
Relative Abundance
80
y3
383.21
70
60
50
40
30
y2
312.18
y1
175.12
a2
761.26
y4
++
496.30 b4
527.71
20
10
b6++
635.75
y6
712.37
b3
926.32
b2
789.261
a3
898.33
b4
1054.41
200
300
400
500
600
m/z
700
800
900
1000
1100
Fig. 2 Detailed characterization of TMPP-labeled N-terminal peptides of Hemoglobin A and BSA. (a) MS/MS
spectrum of the doubly charged N-terminal peptide VLSPADK of Hemoglobin A. Peptide sequence is shown at
the top of the spectrum, with annotation of the matched ions. (b) MS/MS spectrum of the quadruply charged
N-terminal peptide DTHKSEIAHR of BSA. The fragmentation of the derivatized peptide produces a- and b-type
ion series in addition to y-type ions
256
Dimethyl - V L S P A D K
a1
100.11
a1
100
90
[M+2H]2+:
379.2248 m/z
Relative Abundance
80
70
60
50
y4
430.23
40
y6
630.34
30
20
y1
147.11
10
0
100
150
200
y5
517.26
y3
333.18
y2
226.12
250
300
350
400
450
500
550
600
650
m/z
y9
y7 y6 y5 y4 y3 y2 y1
Dimethyl - D T H K S E I A H R
b
100
a1
a1
116.07
90
[M+3H]3+:
407.8804 m/z
80
Relative Abundance
70
60
y3
383.21
50
40
y1
175.12
30
20
y2
312.18
y6
712.37
y4
496.30
y9++
539.79
10
y7
840.46
y5
625.34
0
100
200
300
400
500
600
700
800
900
1000
m/z
Fig. 3 Detailed characterization of dimethyl-labeled N-terminal peptides of Hemoglobin A and BSA. (a) MS/MS
spectrum of the doubly charged N-terminal peptide VLSPADK of Hemoglobin A. (b) MS/MS spectrum of the
triply charged N-terminal peptide DTHKSEIAHR of BSA. Unlike TMPP labeling, the dimethyl labeling strategy
enhances a1 and yn 1 ions
257
7. Pull the green tape gently to remove it from the bottom of the
cassette and remove the comb by pulling upward in one
smooth motion to keep the lane straight.
8. Do not touch the gel with ungloved hands and always use a
new or acid-cleaned dish to avoid contamination.
9. Make sure the staining buffer covers the gel completely.
10. Be sure to use extremely clean surfaces and new scalpels. This
should be done in a flow hood to minimize the possibility of
contamination by dust, hair, flakes of skin, or other forms of dirt.
11. Use polypropylene tubes and low-retention tips to minimize
protein loss by adsorption to tube walls.
12. An increase of temperature (e.g. to 50 C) promotes the
destaining process.
13. Iodoacetamide is unstable and light-sensitive. Prepare iodoacetamide solutions immediately before use and perform alkylation in the dark. Excess reaction time will cause other
functional groups to be labeled.
14. For TMPP labeling, do not use ammonium bicarbonate or Tris
buffer that can decrease the labeling efficiency.
15. For dimethyl labeling, during sample preparation, no buffers and
solutions containing primary amines (such as ammonium bicarbonate and Tris) should be used that can decrease the labeling
efficiency, as formaldehyde can react with these reagents [10].
16. Before loading peptide samples for LC-MS/MS, avoid sample
contamination by keratin, polymers, and detergents. Try to work
as cleanly as possible, especially before tryptic digestion, because
contamination with other proteins can prevent identification of
the protein of interest. The most frequent contaminants are BSA
and human keratin. The keratin comes from dust, small hairs,
and fingerprints. Even a small hair contains overwhelming
amounts of keratin compared to the amount of your sample.
17. Selective N-terminal TMPP derivatization can be achieved by
keeping the solution at pH 8.2, exploiting the weaker basicity
of the N-terminal amine relative to the -amino group of the
lysine side chain [5]. In order to minimize derivatization of
tyrosine residues, 0.1 M hydroxylamine can be added to the
solution to quench the derivatizing reagent 1 h after TMPP
labeling reaction begins [5, 11].
Acknowledgement
This work was supported by NIH Shared Instrumentation Grant
S10RR027990 and NINDS grant P30 NS050276 to T.A.N. We
thank the ABRF Protein Sequencing Research Group (2013) for
helpful protocols, reagents and advice.
258
References
1. Speicher KD, Gorman N, Speicher DW (2009)
N-terminal sequence analysis of proteins and
peptides. Curr Protoc Protein Sci. 57:11.10:
11.10.111.10.31.
2. Gallien S, Perrodou E, Carapito C et al (2009)
Ortho-proteogenomics: multiple proteomes
investigation through orthology and a new
MS-based protocol. Genome Res 19:128135
3. Armengaud J (2009) A perfect genome annotation is within reach with the proteomics and
genomics alliance. Curr Opin Microbiol 12:
292300
4. Huang ZH, Wu J, Roth KD et al (1997) A
picomole-scale method for charge derivatization of peptides for sequence analysis by mass
spectrometry. Anal Chem 69:137144
5. Bertaccini D, Vaca S, Carapito C et al (2013)
An improved stable isotope N-terminal labeling approach with light/heavy TMPP to
automate proteogenomics data validation:
dN-TOP. J Proteome Res 12:30633070
6. Hsu JL, Huang SY, Shiea JT et al (2005)
Beyond quantitative proteomics: signal
enhancement of the a1 ion as a mass tag for
7.
8.
9.
10.
11.
Chapter 20
Optimization ofCell Lysis andProtein Digestion
Protocols forProtein Analysis by LC-MS/MS
DominicWinter, AlirezaDehghani, andHannoSteen
Abstract
Mass spectrometry-based proteomics is the method of choice for analyzing (sub-) proteomes from virtually
any source; with cells grown in culture being the most frequently investigated samples. Using HeLa cells,
we describe a strategy for the optimization of protocols for whole proteome analysis. We cover cell lysis,
protein precipitation and digestion, comparing the results obtained using different parameters and offering
various possibilities for the optimization of a proteomic workflow which can be used for virtually any type
of animal cell grown in culture.
Key words Mass spectrometry, Cell lysis, Protein digestion, Proteomics, Method optimization
1 Introduction
Mass spectrometry-based proteomics has become the major tool
for the identification, characterization, and quantification of proteins. The commonly used strategy, the so-called bottom up
strategy [1], is based on the proteolytic cleavage of the proteins
to be analyzed followed by analysis of the resulting peptides by
mass spectrometry. The extraction of proteins from sample material and the proteolytic digestion are strongly influencing the
number of proteins identified and hence the performance of the
analysis [2, 3]. Dependent on the type of sample analyzed and
the person performing the experiment, buffers, protocols and
techniques vary strongly (e.g. [47]) making it difficult for the
inexperienced operator to choose a particular method. It is often
advisable, if a sufficient amount of material is available, to test
different conditions for sample preparation in order to determine
the optimal parameters for each sample. To achieve optimal
results, several steps of the protocol can be optimized. These
include: harvesting and lysis of cells, extraction and subsequent
proteolytic digestion of proteins and, if in-gel digestions are carried
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI10.1007/978-1-4939-2550-6_20, Springer Science+Business Media New York 2015
259
260
2 Materials
Prepare all solutions (except of PBS and trypsin/EDTA solution)
using HPLC grade water.
2.1 Cell Harvesting
2.3 Protein
Precipitation
261
2.5 Digestion
ofWhole Cell Lysates
262
3 Methods
It is very important to avoid introduction of keratin and other
contaminants such as polyethylene glycol (PEG) during the experimental steps. The keratin proteins may lead to abundant peptide
signals occluding peptides of the sample proteins to be identified,
while other contaminants with surfactant-like properties can significantly affect the ionization of the analytes of interest. Therefore
new plastic ware, pre-stacked pipet tips, LC-MS grade solvents and
high purity chemicals should be used. A laboratory coat and fresh
gloves should be worn and they should be kept clean. Frequent
changes of gloves further reduce the risk of contamination. It
should especially be avoided to bring any surface in contact with
potentially contaminated surfaces, skin, clothing and/or hair that
is, or will be, in direct contact with the sample. In order to minimize sample loss, maximum recovery/low binding 1.5ml tubes
are recommended for all steps. For the optimization of a proteomic
workflow, several parts of the protocol can be altered. An overview
for a possible strategy is depicted in Fig.1.
3.1 Cell Harvesting
263
a
HELA S3 Cells
Lysis Buffers:
RIPA
Protein Precipitaon:
GlyNP40
GCl
Syringe
Sonicator
C/M
TCA
b
Samples prepared with Opmized Lysis and Extracon Procedure
In Soluon
Digeson
Urea
RapiGest
RapiGest+Urea
ProteaseMax
ProteaseMax+Urea
Acetonitrile
In Gel Digeson
No Additives
RapiGest
ProteaseMax
In Gel Digeson
Pepde
Extracon
FA / AA
TFA/HFBA
ACN
Fig. 1 Experimental design for the workflow optimization of the proteomic analysis of HeLa S3 cells. The optimization steps include (a) cell lysis buffers, mechanical cell disruption and protein precipitation as well as (b) protein digestion in
solution and in SDS PAGE gels and peptide extraction from in-gel digests (reproduced from [3] with permission from Wiley)
without disturbing the pellet and wash cells once more with
10ml ice cold PBS followed by centrifugation at 4C for
5min at 500g. If the samples are not to be processed directly,
flash-freeze cells using liquid nitrogen or dry ice and store
at 80C.
3.2 Cell Lysis
264
GlyNP40
GCl
RIPA
TCA C/M
TCA C/M
TCA C/M
TCA C/M
GlyNP40
TCA C/M
265
GCl
TCA C/M
Fig. 2 Coomassie stained SDS-PAGE gels of HeLa S3 protein extracts obtained using combinations of different
lysis buffers (RIPA buffer, GlyNP40 buffer, GCL buffer), lysis methods (syringe and sonicator), and precipitation
methods (TCA and C/M). TCA Trichloroacetic acid precipitation, C/M: chloroform/methanol precipitation (reproduced from [3] with permission from Wiley)
3.3 Protein
Precipitation
266
267
3. Alkylate thiol groups by adding alkylation solution 1 to a final concentration of 1% acrylamide and incubating for 30min at room
temperature. Perform SDS-PAGE electrophoresis (see Notes 16
and 17).
4. Stain the gel using Coomassie blue staining solution. The
staining and destaining steps should each be performed for at
least 2h, preferably overnight (see Notes 18 and 19).
5. For optimization of peptide extraction, choose one region of
the sample lane (e.g. the region between 50 and 80kDa) and
excise the same region from all lanes using a clean scalpel.
Move the big gel piece to a clean surface (e.g. a single use plastic Petri dish), cut the gel bands into small pieces (~1mm3
cubes) and put them in 1.5ml tubes (see Note 20).
6. Add 500l of gel destaining solution and incubate at 25C for
30min at 800rpm in a Thermomixer. If the gel pieces are not
all detached from the bottom of the 1.5ml tube increase the
rpm value or the volume of destaining solution.
7. Discard the liquid and repeat step 6 as long as it takes to
remove the Coomassie from the gel pieces (usually 12 repeats
are sufficient).
8. Discard the liquid, add 500l of 100% ACN and incubate the
samples at 25C for 15min at 800rpm in a Thermomixer.
The gel pieces should shrink and turn white during this step.
9. Discard the liquid and vacuum centrifuge the gel pieces until
they are completely dry. The lids of the 1.5ml tubes have to be
open during vacuum centrifugation.
10. Add 10l of a 1:1 mixture of trypsin solution and 0.1M TEAB
to each sample tube (0.5g trypsin per sample) and incubate
until the gel pieces have taken up all the liquid.
11. Add 50l of 0.1M TEAB and incubate for 30min at 37C.If
the samples are not completely covered by liquid, add as much
0.1M TEAB as needed to cover the samples completely.
12. Incubate the samples overnight at 37C.
13. Transfer the supernatant to a new 1.5ml tube. Do not discard
this solution as the liquid supernatant will contain most of the
peptides to be analyzed.
14. Choose one combination of extraction buffers for each sample
(see Table1). Add 100l of solution A to the 1.5ml tubes containing the gel pieces and incubate at 800rpm, 25C for 15min
in a Thermomixer and transfer the supernatant to the 1.5ml
tubes from step 13. Repeat this step using solutions B and C and
combine all extracts for each individual sample. If 100l are not
sufficient to cover all gel pieces during peptide extraction increase
the volume accordingly until all gel pieces are covered.
15. Vacuum centrifuge the 1.5ml tubes with the pooled supernatants until dryness.
268
Table 1
Combinations of solutions for extracting peptides from gel pieces
Solution A
Solution B
Solution C
Combination 1
OES1
AES 1
100% ACN
Combination 2
OES 2
AES 2
100% ACN
Combination 3
OES 3
AES 3
100% ACN
Combination 4
OES 4
AES 4
100% ACN
Combination 5
OES 5
AES 5
100% ACN
Combination 6
OES 6
AES 6
100% ACN
Combination 7
AES 2
OES 2
OES 7
Combination 8
OES 4
AES 6
100% ACN
Combination 9
OES 1
AES 6
100% ACN
Combination 10
OES 2
AES 6
100% ACN
OES organic extraction solution, AES aqueous extraction solution, ACN acetonitrile
3.5.1 In-Solution
Digestion
269
a
buffer 1
buffer 2
buffer 3
buffer 4
buffer 5
124
128
buffer 8
819
549
607
131
200
solution urea
400
600
800
2749
565
TM
2156
444
solution ProteaseMaxTM
2604
524
2283
470
proteins
2315
360
2716
306
PreteaseMax
2338
370
peptides
794
198
TM
1000
1782
385
solution RapiGestTM
gel
614
111
RapiGestTM
637
160
gel
776
141
no additives
674
139
gel
protiens
569
127
buffer 7
peptides
593
125
buffer 6
buffer 9
602
125
500
1000
1500
2000
2500
3000
Fig. 3 Comparison of different buffers for protein digestion (a) in the gel and (b) in solution as well as for the
extraction of peptides from in-gel digests (reproduced from [3] with permission from Wiley)
1. Prepare protein pellets of 100g of protein, add 1 LDS buffer to the samples and incubate 10min at 95C.
2. Reduce disulfide bonds by adding DTT to a final concentration of 5mM and incubation for 1h at 56C.
3. Alkylate thiol groups by adding alkylation solution 2 to a final
concentration of 15mM IAA and incubation for 45min at
room temperature in the dark.
4. Perform the electrophoresis and let the sample migrate 1cm
into the gel. Optional: Stain the gel using e.g. Coomassie.
270
5. Excise the entire gel area containing the proteins, cut the gel to
pieces and prepare the gel pieces as described in Subheading3.3.
6. Add 1g of trypsin in a sufficient amount of solution (either
TS, RS or PGS) to cover all gel pieces.
7. Incubate the samples overnight at 37C without shaking.
8. Transfer the supernatants to new 1.5ml tubes and perform
peptide extraction following the optimized protocol obtained
from the experiments in Subheading3.3.
9. In order be able to compare the samples to the in-solution
digestion protocols, adjust the sample volume to similar values
as obtained in the in-solution digestion by use of a vacuum
centrifuge. Reduce the ACN content below 5% as this will
otherwise interfere with the binding of the peptides to the
reversed phase material.
10. For the sample containing RapiGest, reduce the sample volume to approximately 100l (take care not to dry down the
sample completely) and add 5l of 100% TFA (final concentration 5%), incubate at 37C at 800rpm for 45min in a
Thermomixer, centrifuge at 20,000g for 30min and transfer
the supernatant to a new tube.
11. Proceed to MS analysis (see Note 21).
In our hands, the highest number of peptide and protein identifications were achieved by in-solution digestion in the presence of
RapiGest (see Fig.3b).
4 Notes
1. One tablet of Protease Inhibitor Cocktail is sufficient for 10 or
50ml of buffer. A 100 stock can be prepared by adding 1
tablet to 100l or 500l HPLC-grade water, respectively, and
kept at -20C for up to 1 year.
2. NP-40 and Triton X-100 are highly viscous detergents. In
order to pipet correct volumes, they should be handled at
room temperature or higher temperatures, pipet tips should be
cut to generate a larger opening and pipetting should be done
slowly. After adding the viscous detergents the solution should
not be vortexed to avoid formation of foam.
3. HEPES, TRIS/HCl, EGTA, KCl, MgCl2, Guanidine-HCl,
NaCl, and SDS can be prepared as stock solutions and kept at
room temperature for several months. TEAB usually ships as
1M stock solution and should be stored at 4C.
4. Highly concentrated acids (TCA, TFA, HFBA, AA, and FA)
cause severe skin burns and eye damage. They should be
handled in the fume hood, safety glasses and gloves should
be worn at all time.
271
272
273
Acknowledgements
This work was supported by the German Academic Exchange
Service (DAAD) and the 7th Framework Program of the European
Union.
References
1. Bogdanov B, Smith RD (2005) Proteomics
by FTICR mass spectrometry: top down and
bottom up. Mass Spectrom Rev 24:
168200
2. Winter D, Hung CW, Jaskolla TW etal (2012)
Enzyme-cleavable tandem peptides for quantitative studies in MS-based proteomics. Proteomics
12:34703474
3. Winter D, Steen H (2011) Optimization of cell
lysis and protein digestion protocols for the
analysis of HeLa S3 cells by LC-MS/MS.
Proteomics 11:47264730
4. Singh S, Winter D, Kirchner M etal (2014)
Co-regulation proteomics reveals substrates
and mechanisms of APC/C-dependent degradation. EMBO J 33:385399
5. Villen J, Gygi SP (2008) The SCX/IMAC
enrichment approach for global phosphorylation analysis by mass spectrometry. Nat Protoc
3:16301638
6. Wisniewski JR, Zougman A, Nagaraj N etal
(2009) Universal sample preparation method
for proteome analysis. Nat Methods 6:
359362
7. Zahedi RP, Lewandrowski U, Wiesner J etal
(2008) Phosphoproteome of resting human
platelets. J Proteome Res 7:526534
Chapter 21
Comprehensive Protocol to Simultaneously
Study Protein Phosphorylation, Acetylation,
and N-Linked Sialylated Glycosylation
Marcella Nunes Melo-Braga*, Mara Ibez-Vea*,
Martin Rssel Larsen, and Katarzyna Kulej
Abstract
Post-translational modifications (PTMs) such as phosphorylation, acetylation, and glycosylation are an
essential regulatory mechanism of protein function and they are associated with a range of biological processes. Since most PTMs alter the molecular mass of a protein, mass spectrometry (MS) is the ideal analytical
tool for studying various PTMs. However, PTMs are generally present in substoichiometric levels and
therefore their unmodified counterpart often suppresses their signal in MS. Consequently, PTM analysis by
MS is a challenging task requiring highly specialized and sensitive enrichment methods. Currently, several
methods have been implemented for PTM enrichment and each of them has its drawbacks and advantages
as they differ in selectivity and specificity toward specific protein modifications. Unfortunately, for most of
the more than 300 known modifications we have none or poor tools for selective enrichment.
Here, we describe a comprehensive workflow to simultaneously study phosphorylation, acetylation,
and N-linked sialylated glycosylation from the same biological sample. The protocol involves an initial
titanium dioxide (TiO2) step to enrich for phosphopeptides and sialylated N-linked glycopeptides followed
by glycan release and post-fractionation using sequential elution from immobilized metal affinity chromatography (SIMAC) to separate mono-phosphorylated and deglycosylated peptides from multiphosphorylated ones. The IMAC flow-through and acidic elution is subsequently subjected to a next
round of TiO2 enrichment for further separation of mono-phosphopeptides from deglycosylated peptides.
In addition, the acetylated peptides present in the first TiO2 flow-through are enriched by immunoprecipitation (IP). Finally, the samples are fractionated by hydrophilic interaction liquid chromatography (HILIC)
to reduce sample complexity and increase the coverage during LC-MS/MS analysis. This allows the analysis of multiple types of modifications from the same highly complex biological sample without decreasing
the quality of each individual PTM study.
Key words Protein post-translational modification (PTM) enrichment, Phosphorylation, Acetylation,
Sialic acid (SA)-glycosylation, Immunoprecipitation (IP), TiSH Comprising of titanium dioxide
(TiO2), Sequential elution from immobilized metal affinity chromatography (SIMAC) and hydrophilic interaction liquid chromatography (HILIC), Liquid chromatography coupled with tandem
mass spectrometry (LC-MS/MS)
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_21, Springer Science+Business Media New York 2015
275
276
Introduction
Proteins are frequently modified by post-translational modifications
(PTMs) such as phosphorylation, acetylation, and glycosylation.
PTMs regulate protein structure, function and lifetime, modulating
their activity in dynamic cells. Aberrations of protein regulation by
modifications lead to the development of various disorders and diseases. Despite the pivotal role of PTMs in a range of biological
processes, PTM analysis by mass spectrometry (MS) is a challenging task since modified peptides are present in substoichiometric
amount in comparison to their unmodified counterpart in proteolytic digests from cells or tissues. However, recent advances in
PTM enrichment methods combined with the new generation of
MS instrumentation have tremendously contributed to large-scale
studies of PTMs.
Nowadays, several techniques exist for phosphopeptide
enrichment prior to MS analysis. The most extensively used methods involve metal ions for the binding of negatively charged phosphopeptides, i.e. immobilized metal affinity chromatography
(IMAC) [1, 2], and metal oxide affinity chromatography such as
titanium dioxide (TiO2) [35]. However, studies comparing various phosphopeptide enrichment methods showed that each
method allows the isolation of distinct subset of phosphopeptides
whereas none of the methods are able to provide a whole phosphoproteome, even though they partially overlap [6]. On the
other hand, this is highly dependent on the individuals performing the analysis and on the numerous protocols that exist for the
phosphopeptide enrichment. The possibility to combine the
strength of different enrichment methods by serial performance
has substantially improved the phosphoproteomic field. For
instance, the combination of IMAC and TiO2 chromatography,
known as Sequential elution from IMAC (SIMAC), has been
employed to separate mono-phosphorylated peptides from multiply phosphorylated peptides in large-scale studies [7]. IMAC has
a stronger selectivity for multi-phosphopeptides than for monophosphopeptides, leading to a better characterization of the first
ones. Following these enrichment methods, a new multi-dimensional large-scale phosphopeptide enrichment strategy, known as
the TiSH protocol was proposed to study phosphorylation from
low amount of starting material (300 g). The TiSH strategy
includes an initial TiO2 enrichment followed by SIMAC and
hydrophilic interaction liquid chromatography (HILIC) fractionation to decrease the complexity of mono-phosphopeptide fraction [8]. The TiSH approach lead to several advances: (1) the
TiO2-based pre-enrichment step improves the enrichment specificity compared to a setup utilizing only SIMAC; (2) TiO2 is more
robust than IMAC being compatible with several chemical reagents
[9], and (3) HILIC fractionation of the mono-phosphorylated
277
Materials
All the solutions and buffers should be prepared with Milli-Q water
(UHQ), analytical grade reagents, and highest purity chemicals.
Organic solutions should be prepared fresh or stored for no more
than 2 weeks before performing the protocol to avoid changes in
buffers composition.
278
1. Lysis and reduction: 6 M urea, 2 M thiourea, 10 mM dithiotreitol (DTT), phosphatase inhibitors, 0.1 mM sodium pervanadate, and 0.04 AU of Lys-C (lysyl endopeptidase, WAKO
Pure Chemical Industries, Ltd., Osaka, Japan) (see Note 1).
2. Alkylation: 20 mM iodoacetamide in 20 mM triethylammonium bicarbonate (TEAB).
3. Digestion: 2 % trypsin (Sigma porcine trypsin, St. Louis, MO, US)
(see Note 2).
4. Stop digestion: 100 % formic acid.
2.3 Desalting/
Concentration
of the Peptide Mixture
on Reversed-Phase
(RP) Columns
279
2.6 Enrichment
of Multiphosphorylated
Peptides by Sequential
Elution from IMAC
Beads (SIMAC)
2.7 Separation
of MonoPhosphopeptides
from Deglycosylated
Peptides by Second
TiO2 Enrichment
2.8 Acetyl-Lysine
Immunoprecipitation
(IP)
2.10 nanoLC-MS/MS
Analysis
280
Data Analysis
Methods
The complete workflow for the multiple PTM enrichment strategy
described in this chapter is illustrated in Fig. 1. The workflow consists of an initial simultaneous enrichment of phosphopeptides and
N-linked sialylated glycopeptides using the first TiO2 step. The
flow-through (FT) from this step is used to enrich acetylated peptides by IP. The K-Ac antibodies capture acetylated peptides,
whereas the non-modified peptides are present in the IP-FT. After
enzymatically release of N-linked glycan structures from the
sialylated N-linked glycopeptides in the TiO2 eluate, the multiphosphopeptides are separated from the mono-phosphopeptides
and deglycosylated peptides by SIMAC. Three fractions are
obtained from IMAC: (I) IMAC-FT, (II) an acidic elution fraction
(pH 2.3), and (III) a basic elution fraction (pH 11.3). The FT
and acidic elution that contains deglycosylated peptides and monophosphopeptides are combined and subjected to a second milder
TiO2 chromatographic step to separate both types of modified
peptides. Finally, in order to decrease the complexity of monophosphopeptides, deglycosylated peptides and non-modified peptides, HILIC fractionation is performed prior LC-MS/MS analysis.
Here, a total of 500 g of whole HeLa cell lysate was used for a
qualitative analysis to illustrate the average outcome of the protocol; although, the protocol can be optimized to different amounts
(lower or higher) and applied also for quantitative studies.
281
Fig. 1 Enrichment workflow. An initial titanium dioxide (TiO2) step is performed to enrich phosphopeptides and
sialylated N-linked glycopeptides followed by glycan release. Then, SIMAC is performed to separate monophosphorylated and deglycosylated peptides from multi-phosphorylated peptides. The IMAC-FT together with
the IMAC acidic elution is subjected to a second round of TiO2 enrichment to separate mono-phosphopeptides
from deglycosylated peptides. In addition, the first TiO2-FT is used to enrich acetylated peptides by IP. Finally,
the samples are fractionated by HILIC prior to LC-MS/MS analysis
282
3.3 Desalting/
Concentration
of the Peptide Mixture
on HLB/RP Cartridge
(See Notes 7 and 8)
283
284
3.6 Enrichment
of Multiphosphorylated
Peptides by SIMAC
3.7 Separation
of MonoPhosphopeptides
from Deglycosylated
Peptides by Second
TiO2 Enrichment
285
3.9
Acetyl-Lysine IP
286
1. Prepare a micro-column using p200 tip by plugging the constricted end with C8 material filter. Pack 2 cm column with R3
material slurry in 100 % ACN by applying air pressure using a
syringe.
2. Equilibrate the column with 50100 L of 0.1 % TFA.
3. Load the acidified sample (pH < 2.0) into the column and
make a gently air pressure. It is important to perform this step
slowly. In case the sample is dried, add 80 L of 0.1 % TFA to
reconstitute the sample prior to load. Collect the FT.
4. Pass again the FT through the micro-column to increase the
binding of peptides.
5. Wash the column with 100 L of 0.1 % TFA.
6. Elute the peptides into a new low-binding Eppendorf tube
with 100 L of 60 % ACN/0.1 % TFA.
7. Lyophilize the sample in a vacuum centrifuge.
3.11 HILIC
Fractionation of MonoPhosphopeptides,
Deglycosylated
Peptides and Nonmodified Peptides
1. Reconstitute the sample in 45 L of HILIC solvent B by adding first 0.45 L of 10 % TFA and then 4.05 L of milli-Q
water. Vortex and finally add 40.5 L of 100 % ACN slowly to
avoid precipitation. This step should be performed immediately prior to fractionation since the acetonitrile evaporates fast
altering the efficiency of the method.
2. Inject 40 L onto the microHPLC HILIC column.
3. Fractionate the peptide mixture using the gradient described
in Table 1.
4. Collect the fractions according to Table 2.
287
Table 1
HILIC gradient
Time (min)
Concentration of Sol. B
(90 % ACN, 0.1 %TFA)
Flow (L/min)
00:00
100
12
08:60
100
12
09:00
95
35:00
60
39:00
42:00
46:00
100
48:00
100
The gradient used for fractionation of all obtained modified and unmodified peptide
samples by HILIC microHPLC, except for the multi-phosphorylated peptides
Table 2
Timeline of HILIC fraction collector
Time (min)
Trigger mode
00:00
Time-based
10
10:00
Time-based
14:00*
Time-based
36:00**
Time-based
48:00
off
The timeline used to collect all peptide samples beside the multi-phosphorylated peptides. In addition, alteration to *20:00 and **40:00 was done for the sample containing
mono-phosphorylated peptides
1. Reconstitute the dried samples (multi-phosphopeptides, acetylated peptides as well as the HILIC fractions from monophosphopeptides, deglycosylated peptides and non-modified
peptides) in 0.1 % formic acid.
2. Inject 5 L of sample onto an in-house packed Reprosil-Pur
C18-AQ (3 m; Dr. Maisch GmbH, Germany) pre-column
288
Fig. 2 HILIC chromatogram. The figure illustrates the HILIC chromatogram of non-modified peptides in order to
show how fractions were pooled in the present work. Asterisk denote individual fractions
Data Analysis
289
serine, threonine, tyrosine phosphorylation; asparagine and glutamine deamidation. Protein grouping was performed, in order to
avoid presence of different proteins identified by non-unique peptides. Only peptides with up to q-value of 0.01 (Percolator),
Mascot and Sequest HT rank 1, Mascot and Sequest HT search
engine 1, a Sequest HT Cn of 0.1, a cut-off value of Mascot score
22 and a cut-off value of XCorr score greater than 1.5, 2, 2.5 and
3 for charge states of +1, +2, +3, and +4, respectively, were considered for further analysis.
3.14
Results
Fig. 3 Results obtained from the analysis of phosphopeptides, sialylated N-linked glycopeptides and acetylated
peptides using the optimized workflow. The number of phosphorylated peptides (mono- and multiply phosphorylated peptides), sialylated N-linked glycopeptides and acetylated peptides identified from 500 g of HeLa
cell Lys-C/tryptic digestion using the combination of TiO2, SIMAC, and K-Ac IP enrichment methods
290
Notes
1. Protease inhibitor cocktail can be added if needed.
2. The trypsin is purified by benzamidine sepharose affinity to
guarantee the same proteolytic activity for the entire batch,
as well as to significantly reduce the level of autodigestion
products.
3. The choice between POROS Oligo R3 or Oasis HLB cartridge
depends on the quantity of material. For peptide samples with
quantity higher or equal to 500 g, the Oasis HLB cartridge is
often a choice.
4. To avoid peptide precipitation add to the lyophilized sample
0.45 L of 10 % TFA and then 4.05 L of milli-Q water. Vortex
and finally add 40.5 L of 100 % ACN. The final concentration
of dissolution buffer should be 90 % ACN/0.1 % TFA.
5. Any protein extraction, denaturation, and digestion method is
compatible with the present simultaneous PTMs enrichment
strategy.
6. Mix by vortexing and pipette cell pellet up and down after adding lysis buffer. Cell lysate will form a very viscous solution due
to the presence of nuclear DNA.
7. POROS Oligo R3 micro-columns or Oasis HLB cartridges
desalting step is required if the buffer solutions are not compatible with labeling methods such as iTRAQ, TMT, dimethyl
labeling; or prior to determination of protein concentration by
AAA analysis.
8. TiO2 enrichment might be performed directly after the protein
digestion step since TiO2 chromatography is compatible with
most of the commonly used buffer solutions [9].
9. A labeling strategy such as iTRAQ, TMT or dimethyl labeling
can be introduced in this step for quantitative studies.
Remember to check the compatibility of buffer solutions with
the specific labeling reagents.
10. To reduce nonspecific binding to the TiO2 resin, it is important
to adjust the amount of TiO2 beads to the amount of sample.
The optimal quantity is 0.6 mg TiO2 beads per 100 g peptide
solution [8].
11. In case the sample was lyophilized before, it is crucial to reconstitute the sample in a small volume of 0.1 % TFA. Moreover,
the TiO2 loading buffer should be added slowly to avoid peptide precipitation.
12. Centrifugation time is not critic.
13. This step can be done up to three times, depending on the
sample complexity.
291
6.
7.
8.
9.
292
Chapter 22
Protein Profiling and Phosphoprotein Analysis
by Isoelectric Focusing
Giuseppina Maccarrone and Michaela D. Filiou
Abstract
Protein profiling enables the qualitative characterization of a proteome of interest. Phosphorylation is
a post-translational modification with regulatory functions in a plethora of cell processes. We present
an experimental workflow for simultaneous analysis of the proteome and phosphoproteome with no
additional enrichment for phosphoproteins/phosphopeptides. Our approach is based on isoelectric
focusing (IEF) which allows the separation of peptide mixtures on an immobilized pH gradient (IPG)
according to their isoelectric point. Due to the negative charge of the phosphogroup, most of the
phosphopeptides migrate toward acidic pH values. Peptides and phosphopeptides are then identified
by mass spectrometry (MS) and phosphopeptide spectra are manually checked for the assignment of
phosphorylation sites. Here, we apply this methodology to investigate synaptosome extracts from
whole mouse brain. IEF-based peptide separation is an efficient method for peptide and phosphopeptide identification.
Key words Isoelectric focusing, Immobilized pH gradient, Mass spectrometry, Peptides,
Phosphopeptides, Proteome profiling, Phosphoproteome, Synaptosomes, Mouse brain
Abbreviations
CID
IEF
IPG
LC-ESI-MS/MS
MS
pI
S
T
Y
Collision-induced dissociation
Isoelectric focusing
Immobilized pH gradient
Liquid chromatography-electrospray ionization-tandem mass spectrometry
Mass spectrometry
Isoelectric point
Serine
Threonine
Tyrosine
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_22, Springer Science+Business Media New York 2015
293
294
Introduction
Mass spectrometry (MS)-based proteome profiling provides a
protein map for the biological material under investigation, facilitating functional and quantitative analyses. Phosphorylation is a
post-translational modification that occurs in approximately onethird of all proteins and is involved in many biological processes in
health and disease [1].
For proteome and phosphoproteome analysis, effective fractionation methods prior to MS are crucial to ensure high proteome
coverage through peptide and phosphopeptide identification.
Here, we describe how isoelectric focusing (IEF), which has been
traditionally used to separate proteins in the first dimension of
2D-gel electrophoresis, can be used as an effective fractionation
method both for peptides and phosphopeptides. Peptide mixtures
are loaded onto an immobilized pH gradient (IPG) strip and upon
application of electric current, peptides are separated according to
their isoelectric point (pI). The unique characteristic of IEF compared to other fractionation methods is the ability to simultaneously
analyze peptides and phosphopeptides. This is due to the addition
of a negatively charged phosphogroup to a peptide sequence which
results in a decrease of its pI. As a consequence, phosphopeptides
migrate toward the acidic part of the IPG strip [2].
MS analysis of the whole and the acidic part of the IPG strip
allows the investigation of the proteome and the phosphoproteome, respectively, with no additional step for phosphopeptide
enrichment. The pI focusing position of each peptide on the IPG
strip after IEF can be used to confirm subsequent MS-based peptide identifications and/or validate the presence of post-translational
modifications [35]. When applied to brain tissue, IEF has been
shown to result in increased proteome coverage compared to
1D-SDS gel electrophoresis [6]. In this chapter, we provide a
detailed guide to proteome and phosphoproteome analysis by
IEF. As study material we use synaptosomes, which are artificially
isolated synapses [7], extracted from whole mouse brain. We chose
brain synaptosomes for two reasons; (i) their proteome is highly
complex (ii) phosphorylation plays a critical role in the regulation
of synaptic function and neurotransmission [8]. In this protocol,
we describe the following steps: (1) synaptosome enrichment from
brain tissue and synaptosome enrichment quality control by
Western blot, (2) sample preparation for IEF and IEF, (3) MS, (4)
MS raw data analysis for peptide and phosphopeptide identification
and assignment of phosphorylation sites. The experimental workflow is shown in Fig. 1. Mouse synaptosome analysis by IEF
resulted in the identification of up to 3,000 proteins and 118 phosphoproteins [6].
295
Brain tissue
Synaptosome enrichment
quality control
Synaptosome extraction
Reduction &
carboxymethylation
In-gel digestion
IEF
3.5
4.5
Peptide extraction
Extracted/cleaned peptides
LC ESI-MS/MS
Database search
Peptide/Phosphopeptide identification
Proteome profiling
Phosphoproteome profiling
Manual MS spectra validation
Phosphosite assignment
Fig. 1 Experimental set-up of IEF-based analysis of proteome and phosphoproteome from mouse brain
synaptosomes
296
Materials
2.1 Synaptosome
Enrichment and
Western Blot for
Enrichment Quality
Control
2.1.1 Synaptosome
Enrichment
2.2
IEF
2.3
ESI-LC-MS/MS
297
1. Nanoflow HPLC-2D system (Eksigent) including a trap column Zorbax SB300, 5 mm 0.3 mm, packing material 5 m
RP-C18 (Agilent Technologies).
2. LTQ Orbitrap XL mass spectrometer (Thermo Fisher).
3. Nano electrospray ionization source (Thermo Fisher).
4. Picofrit self-packed column 75 m 15 cm i.d., 10 m tip
(New Objective), packing material 3 m RP-C18.
5. Acetonitrile.
6. Formic acid.
2.4 MS Data
Analysis Software
Methods
3.1 Synaptosome
Enrichment
and Western Blot
for Enrichment Quality
Control
3.1.1 Synaptosome
Enrichment
298
3.2
IEF
299
10. After IEF completion, the IPG strip is removed from the tray
and mineral oil is drained by vertically holding the IPG strip.
11. The IPG strip is cut in 4 mm pieces. Each gel piece is transferred in a sample tube prefilled with 100 l 5 % formic acid.
12. Peptides from each gel piece are extracted with 50 l 5 % formic acid three times (15 min vortexing and 15 min sonication)
and the three supernatants are combined. To remove the residual mineral oil from the combined supernatant, the peptide
extracts are overlaid with hexane, mixed briefly and the upper
part of the organic solvent is discarded. The aqueous phase is
kept and the oil extraction with hexane (50 l) is repeated
three times. After oil extraction, the aqueous phase is kept and
lyophilized (see Note 10).
13. The resulting pellet is oil extracted with 10 l hexane and left
to dry overnight. The following day, pellets are dissolved in
25 l 5 % formic acid, desalted with OMIX tips according to
the manufacturers instructions and lyophilized (see Note 11).
3.3
ESI-LC-MS/MS
3.4
Data Analysis
1. MS raw files are searched against a decoy mouse database utilizing BioWorks and SEQUEST. Search parameters are as follows: peptide mass tolerance, 20 ppm; fragment ion mass
tolerance, 1 Da; enzyme, trypsin; missed cleavage sites, up to
two; only tryptic peptides allowed; static modification, cysteine
carboxyamidomethylation; variable modification, methionine
oxidation.
300
Notes
1. There are different options for IPG lengths and pH ranges
according to the application of interest. For high amounts of
starting material to be loaded, longer IPG strips are more
appropriate. For phosphorylation studies, IPG strips with a pH
range toward more acidic values are recommended [2, 10].
2. A minimum amount of approx. 30 mg brain tissue is required
to obtain a synaptosomal fraction. When studying brain regions
with lower tissue weight, pooling is necessary to study the synaptosomal proteome.
3. The crude synaptosome fraction at the interface of the
1.2 M/0.8 M sucrose gradient appears as a white cloud.
301
EGDGSATTDAAPATSPK
680.39
100
95
90
85
80
Relative Abundance
75
70
65
60
55
50
45
40
30
25
20
313.12
15
10
5
y6-98
y3-98
y*3
y*
b15-98
y*10
y*7
35
y9*
751.33
y*11
1038.55
937.44
y5*
583.24
* +2
b*11
15
976.37
706.76
411.12
1245.71
y11-98
1041.50
1314.58
b14
1139.57
y12
*
y13
y*14
b*15
1354.58
1412.68
y*15
1210.71 1297.56
1470.11
244.17
300
400
500
600
700
800
900
1000
1100
1200
1300
1400
1500
1600
m/z
Fig. 2 ESI-MSMS spectrum of Neuromodulin phosphorylated peptide EDGSATTDAAPATSPK. The single- and
double-charged b- and y-fragment ions identified by SEQUEST algorithm are shown. The phosphorylation site
at S15 is unambiguously determined by the loss of the phosphoric acid (H3PO4) from the y3 ion (y3-98) and by
the absence of the phosphoric acid loss peak from the b14 ion fragment. Phosphorylation at S15 is ascertained
by multiple fragment ions, e.g. y3-98, y6-98, y11-98, b15-98. The phosphorylation site is underlined in the peptide sequence. b- and y-fragment ions are highlighted in blue and red, respectively. Peaks marked by asterisk
(*) denote phosphorylated fragments
302
303
Acknowledgments
This work is funded by the Max Planck Society. M.D.F. is supported by a grant from the Deutsche Forschungsgemeinschaft (FI
1895/1-1). We thank Chris Turck for useful comments.
References
1. Cohen P (2001) The role of protein phosphorylation in human health and disease. The Sir
Hans Krebs Medal Lecture. Eur J Biochem
268:50015010
2. Maccarrone G, Kolb N, Teplytska L, Birg I,
Zollinger R, Holsboer F, Turck CW (2006)
Phosphopeptide enrichment by IEF. Electrophoresis 27:45854595
3. Cargile BJ, Bundy JL, Freeman TW, Stephenson
JL Jr (2004) Gel based isoelectric focusing of
peptides and the utility of isoelectric point in protein identification. J Proteome Res 3:112119
4. Uwaje NC, Mueller NS, Maccarrone G, Turck
CW (2007) Interrogation of MS/MS search
data with an pI Filter algorithm to increase
protein identification success. Electrophoresis
28:18671874
5. Xie H, Bandhakavi S, Roe MR, Griffin TJ
(2007) Preparative peptide isoelectric focusing
as a tool for improving the identification of
lysine-acetylated peptides from complex mixtures. J Proteome Res 6:20192026
6. Filiou MD, Bisle B, Reckow S, Teplytska L,
Maccarrone G, Turck CW (2010) Profiling
of mouse synaptosome proteome and phosphoproteome by IEF. Electrophoresis 31:
12941301
7. Schrimpf SP, Meskenaite V, Brunner E,
Rutishauser D, Walther P, Eng J, Aebersold R,
8.
9.
10.
11.
12.
Chapter 23
Principles and Examples of Gel-Based Approaches
for Phosphoprotein Analysis
Birgit Steinberger and Corina Mayrhofer
Abstract
Methods for analyzing the phosphorylation status of proteins are essential to investigate in detail key
cellular processes, including signal transduction and cell metabolism. The transience of this post-translational
modification and the generally low abundance of phosphoproteins require specific enrichment and/or
detection steps prior to analysis. Here, we describe three gel-based approaches for the analysis of differentially expressed phosphoproteins. These approaches comprise (1) the sequential fluorescence staining of
two-dimensional (2-D) gels using Pro-Q Diamond and SYPRO Ruby dyes to visualize and quantify
phosphoproteins in total cellular lysates as well as (2) affinity enrichment of phosphoproteins in conjunction with sequential fluorescence staining of the 2-D gels and (3) affinity enrichment of proteins prior to
pre-electrophoretic fluorescence labeling and 2-D gel electrophoresis.
Key words Phosphoproteins, 2-D gel electrophoresis, IMAC, Pro-Q Diamond, SYPRO Ruby, 2-D
fluorescence difference gel electrophoresis
Introduction
The reversible modification of proteins, in particular of serine, threonine,
and tyrosine residues, with one or more phosphate groups is a key
regulator of a multitude of cellular processes including growth and
differentiation [1]. Moreover, protein phosphorylation/dephosphorylation driven by specific kinases/phosphatases is involved in the
information transmission within a cell and is part of the communication system of cells with their environment [2]. Thus, the unambiguous characterization of the phosphorylation status of proteins in
response to a certain stimuli or environmental changes is crucial to
gain a better knowledge of cell signaling processes and in addition of
vital biological events. Various methods are available to characterize
and identify these post-translational modified proteins. Beside gelfree, mass spectrometry-based approaches that enable the study
of the modified amino acids on the peptide as well as on the protein level [3], different approaches based on two-dimensional gel
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_23, Springer Science+Business Media New York 2015
305
306
307
Fig. 1 Overview of different gel-based approaches to analyse phosphoproteins. In the first approach (light
grey ), total cell lysates are directly separated by 2-D GE and the gels are sequentially post-stained using a
phosphospecific stain, visualizing the phosphoproteins content followed by a total fluorescence stain. The
second approach (grey ) consists of an enrichment step to purify the phosphoproteins using affinity chromatography and subsequent separation by 2-D GE and sequential post-staining. In the third described approach
(dark grey ), affinity-enriched proteins are labeled with fluorescence dyes for differential analysis prior to gel
electrophoresis. The sample preparation steps include sample clean up and the use of a protein concentration
assay. Copyright Wiley-VCH Verlag GmbH & Co. KGaA. Modified with permission from [16]
309
identification (see Fig. 3) [16]. Moreover, tagging of the IMACenriched proteins with fluorescent dyes can further be used for
quantitation by 2-D DIGE. To sum up, one should choose an
adjusted approach to analyze the phosphoproteome depending on
the available biological samples.
310
Materials
Prepare all buffers and solutions using ultrapure water (e.g.
obtained from a Milli-Q system, Millipore, Bedford, MA; MHQ
water, 18 M cm conductivity at 25 C). Store all solutions at
4 C, unless otherwise stated.
2.2 Affinity
Enrichment Using
IMAC Columns
2.3 Sample
Preparation (Protein
Precipitation
and Protein Labeling)
311
2.4 Two-Dimensional
Gel Electrophoresis
(2-D GE)
312
313
2.6 Image
Acquisition
and Analysis
Methods
314
3.2 Affinity
Enrichment Using
IMAC Columns
3.3 Sample
Preparation (Protein
Precipitation
and Protein Labeling)
315
3.4 Two-Dimensional
Gel Electrophoresis
(2-D GE)
316
9. At this point the strips can either be stored in, for example, a
15 mL tube or a disposable rehydration tray at 80 C or
immediately continue with step 10.
10. Put the strips with the gel side up into a disposable rehydration
tray and cover each single strip with 2 mL reduction equilibration buffer. Incubate at room temperature for 19 min with
gentle agitation (see Note 20).
11. Immediately remove the strips and put it into a new disposable
rehydration tray. Cover each strip with 2 mL alkylation equilibration buffer. Incubate at room temperature for 19 min with
gentle agitation.
12. During step 11, melt the 1 % agarose solution (sealing solution) and store at 70 C till use.
13. Remove the strips from the rehydration tray by holding one
side of the strip with forceps and carefully align each strip onto
the top of a second-dimension SDS polyacrylamide gel (for
example: CriterionTM, Bio Rad) without any air bubbles
between the strip and the gel.
14. Load the molecular weight standard into the designated well
(see Note 21).
15. Quickly overlay the strip with 12 mL sealing solution. Avoid
introducing air bubbles. Let the sealing solution solidify for
approximately 2 min.
16. Separate the proteins in 1 SDS running buffer.
17. Open the gel plates with the use of a spatula. Carefully transfer
the gels into a cleaned container, add some water to prevent
the gels from drying out and immediately continue either with
steps 18 or 19.
18. In case of pre-electrophoretic labeling, visualize the fluorescence images of the gels, as stated in Subheading 3.6, step 1.
19. In case of sequential post staining, handle the gels as described
in Subheading 3.5 (see Note 22).
3.5 Sequential Gel
Staining
3.5.1 Pro-Q Diamond
Staining
317
1. After image acquisition, proceed with the silver staining protocol to visualize the proteins (according to [17]).
2. Put the gels in a new tray and wash them in water for 30 min.
3. Incubate the gels for 1 min in sensitizing solution. Remove the
sensitizing solution.
4. Wash three times for 20 s with water. Remove the water after
each washing step (see Note 24).
5. Add the silver nitrate solution and incubate the gel for 20 min
at 4 C in the dark.
6. Repeat step 3.
7. Add development solution and remove it when the solution
turns slightly turbid. Add fresh development solution and
incubate the gel until it is sufficiently stained. Discard the
solution.
8. Wash with water for 20 s. Discard the water.
9. Incubate the gels in stop solution for 5 min. Remove the stop
solution.
10. Wash the gels for 10 min with water. Repeat this step twice.
Discard the water after each washing step.
11. The gels can now be stored in storage solution at 4 C.
318
3.6 Image
Acquisition
and Analysis
Notes
1. The use of phosphatase/protease inhibitor cocktails is essential
to prevent the degradation of proteins and the removal of
phosphate groups from proteins.
2. The volume of the lysis buffer is dependent on the wet weight
cell pellet. Prepare the lysis buffer volume according to the
weight of each cell pellet as well as to the number of samples.
Add protease and phosphatase inhibitors to the lysis buffer
freshly before use.
3. Other protein concentration assays can be used as well.
However, no single available protein assay is compatible with
all buffer components. Therefore, select an appropriate assay
method that is most compatible with your samples and the
buffer components (e.g. BCA Protein AssayReducing Agent
Compatible, Thermo Fisher).
4. For each column load you need approximately 25 mL Lysis/
Binding/Wash buffer containing 0.25 % CHAPS as well as
5 mL elution buffer containing 0.25 % CHAPS. Prepare the
volume according to the number of samples.
5. Other systems can be used as well, such as ReadyPrep 2-D
Cleanup Kit (Bio Rad).
6. Other commercial available fluorescent dyes can be used as
well, e.g. Refraction-2D G-dyes (NH DyeAGNOSTICS,
Halle, Saxony-Anhalt, Germany).
7. Prepare the equilibration buffer at least 3 h prior to use, considering that it takes the buffer components a while to dissolve
completely. To avoid carbamylation, do not heat urea containing buffers above 37 C. Furthermore, add only small amounts
of water at once, to prevent that the total volume of 200 mL
will not be exceeded.
8. It is more convenient to prepare a 10 running buffer: 0.25 M
Tris, 1.92 M glycine and 1 % SDS (w/v). Weigh 30.3 g Tris,
319
320
Acknowledgements
This work was funded by Life Science Calls, Niedersterreichische
Forschungs-und BildungsgmbH (NFB).
References
1. Machida M, Kosako H, Shirakabe K et al
(2007) Purification of phosphoproteins by
immobilized metal affinity chromatography
and its application to phosphoproteome analysis. FEBS J 274:15761587
2. Raggiaschi R, Lorenzetto C, Diodato E et al
(2006) Detection of phosphorylation patterns
in rat cortical neurons by combining phosphatase treatment and DIGE technology.
Proteomics 6:748756
321
12. Talvas J, Obled A, Sayd T et al (2008) Phosphoproteomic approach to identify new targets of
leucine deprivation in muscle cells. Anal
Biochem 381:148150
13. Chen A, McEwen ML, Sun S et al (2010)
Proteomic and phosphoproteomic analyses of
the soluble fraction following acute spinal cord
contusion in rats. J Neurotrauma 27:263274
14. Stasyk T, Morandell S, Bakry R et al (2005)
Quantitative detection of phosphoproteins by
combination of two-dimensional difference gel
electrophoresis and phosphospecific fluorescent staining. Electrophoresis 26:28502854
15. Tang W, Deng Z, Oses-Prieto JA et al (2008)
Proteomics studies of brassinosteroid signal
transduction using prefractionation and twodimensional DIGE. Mol Cell Proteomics 7:
728738
16. Steinberger B, Besenfelder U, Brem G et al
(2013) Comparison of gel-based phosphoproteomic approaches to analyse scarce oviductal
epithelial cell samples. Proteomics 13:1216
17. Blum H, Beier H, Gross HJ (1987) Improved
silver staining of plant proteins, RNA and
DNA in polyacrylamide gels. Electrophoresis
8:9399
Chapter 24
Neutral Phosphate-Affinity SDS-PAGE System
for Profiling of Protein Phosphorylation
Emiko Kinoshita-Kikuta, Eiji Kinoshita, and Tohru Koike
Abstract
In this chapter, we describe a standard protocol for phosphate-affinity SDS-PAGE that uses a dizinc(II)
complex of the phosphate-binding molecule Phos-tag in conjunction with a neutral-pH gel system (Zn2+
Phos-tag SDS-PAGE) to detect shifts in the mobilities of phosphoproteins. A previous protocol for affinity
electrophoresis that uses polyacrylamide-bound Mn2+-Phos-tag and Laemmlis buffer system under conditions of alkaline pH has limitations in separating certain phosphoproteins. The current protocol provides
major improvements in separation and detection of various phosphorylated protein species. We here
introduce two neutral-pH gel systems buffered with BisTrisHCl and TrisAcOH, respectively, for Zn2+
Phos-tag SDS-PAGE, and we also discuss their characteristics on the basis of comparative studies on phosphorylation profiling of proteins with a wide range of molecular masses. Each analytical procedure, from the
beginning of gel preparation to the end of electrophoresis, requires 2.55 h with either buffer system.
Key words Phos-tag, Affinity electrophoresis, Protein phosphorylation, Phosphoproteomics, Neutral
SDS-PAGE
Introduction
1.1 General
Background
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_24, Springer Science+Business Media New York 2015
323
324
325
Fig. 1 Phosphate-affinity SDS-PAGE for the detection of shifts in mobilities of phosphoprotein species.
(a) Structure of acrylamide-bound Phos-tag ligand and scheme for reversible capture of a phosphomonoester
dianion (ROPO32) by polyacrylamide-bound Phos-tag. (b) Schematic representation of the principle of
phosphate-affinity SDS-PAGE. (c) Flow schemes summarizing strategies for the analysis of phosphorylated
proteins and peptides by phosphate-affinity 2-DE analysis and by MS analysis, respectively
326
327
328
1.5 Alternative
Protocol for Zn2+
Phos-tag SDS-PAGE
for the Analysis
of Large
Phosphoproteins
with Molecular
Masses in Excess
of 200 kDa
329
330
331
2.1 Sample
Preparation
2.1.1 Dephosphorylation
of Standard
Phosphoproteins
2.1.2 Preparation
of Cell Lysate
332
2.2.2 Zn2+Phos-tag
SDS-PAGE Using a Bis
TrisHCl Buffer System
333
2.3
Gel Staining
2.4
Electroblotting
2.5 Equipment
for Electrophoresis
and Blotting
334
Methods
3.1 Sample
Preparation
3.1.1 Preparation
of Standard
Phosphoproteins
Dephosphorylated by AP
3.1.2 Preparation
of a Lysate from HeLa Cells
1. Grow HeLa cells in culture medium under a humidified atmosphere of 5 % CO2 and 95 % air at 37 C.
2. Treat the cells (~107 cells) with 2.0 M actinomycin D for 2 h.
3. Wash the cells twice with TBS.
4. Lyse the cultures in 0.50 mL of lysis buffer (1).
5. Sonicate briefly the lysate samples, boil them for 5 min, and
store at 20 C.
6. Subject an aliquot (10 L) of the resulting sample solution to
Phos-tag SDS-PAGE.
3.1.3 Preparation
of a Lysate from A431
Cells
1. Grow A431 cells in culture medium under a humidified atmosphere of 5 % CO2 and 95 % air at 37 C.
2. Treat the cells (~107 cells) with 250 ng/mL of EGF for 0 (no
treatment), 2, 5, 10, 30, 60, 90, or 120 min.
3. Wash each culture twice with TBS.
4. Lyse the cultures in 0.50 mL of lysis buffer (1).
5. Sonicate briefly the lysate samples, boil them for 5 min, and
store at 20 C.
6. Subject an aliquot (10 L) of the resulting solution to Phostag SDS-PAGE.
3.2
Electrophoresis
3.2.1 Preparation
of the Zn2+Phos-tag
SDS-PAGE Gel
(See Note 20)
1. Clean the glass plates for casting the gels. It is vital that the
glass plates (1 mm thick, 9 cm wide, and 9 cm long for minislab gels) are washed thoroughly with a rinsable detergent, and
that they are subsequently thoroughly rinsed with distilled
water before the gels are cast.
2. Select an appropriate gel type for Zn2+Phos-tag SDSPAGE. For the analysis of 10200 kDa phosphoproteins, a gel
buffered with either BisTrisHCl or TrisAcOH gel can be
used. Cast this gel according to the procedures described in
steps 310 (below). For the analysis of large phosphoproteins
with molecular masses of more than 200 kDa, a TrisAcOH
gel should be used. Cast this gel according to the procedures
described in steps 1116 below.
3. Prepare the separating gel solution for the analysis of 10200 kDa
phosphoproteins. As a typical example of the Zn2+Phos-tag gel
335
Problem
Step
Steps 5 and 9 in
Subheading 3.2.1
Step 13 in
Subheading 3.2.1
Step 16 in
Subheading 3.2.1
Step 7 in
Subheading 3.4
Table 1
Troubleshooting advice
The agarosepolyacrylamide
composite gel is partially melted
during electroblotting because of
the generation of heat
Agarose gelling
Possible reasons
Solution
336
Emiko Kinoshita-Kikuta et al.
1. Contamination by small-molecule
substances, such as inorganic salts
or surfactants
2. Contamination by viscous
genomic DNA
3. Contamination by cell debris or
insoluble biomaterials
4. Impurities, such as lipids or small
molecules derived from cells,
interfere with the interaction
between the Zn2+Phos-tag and
phosphoprotein
Step 8 in
Subheading 3.4
Solution
Possible reasons
Problem
Step
338
Fig. 2 Schematic representation of the relationship between the degree of migration (Rf value) and the molecular weight for myosin heavy chain (205 kDa) and
ATM (350 kDa) in 2.54.0 % (w/v) polyacrylamide slab gels. The Rf value of myosin heavy chain was determined by means of CBB gel staining. The Rf value of
ATM was determined by Western blotting analysis of HeLa cell lysate using the
anti-ATM antibody. The Rf value of 1.0 is defined as the position of the BPB dye
339
3.3
Gel Staining
3.4
Electroblotting
1. When the run is complete, remove the gel from the apparatus
and soak it in the blotting buffer containing 1 mM EDTA for
30 min (see Note 31).
2. Soak the gel in the blotting buffer without EDTA for a further
1030 min.
3. Prepare the poly(vinylidene difluoride) (PVDF) membrane
(e.g., Fluorotrans W, Nippon Pall, Tokyo, Japan) by cutting it
to the same size as the gel and soak it for 30 s in 100 %
MeOH. Then, incubate the membrane in the blotting buffer
for more than 30 min.
4. Prepare four pieces of blotting paper (e.g., 3MM; Whatman,
Maidstone, UK) by cutting them to the same size as the gel.
5. To form a blotting sandwich on the electroblotting screen
attached to the electroblotting equipment, assemble the gel,
PVDF membrane, and 3MM paper as follows. Soak the blotting sponge attached to the electroblotting equipment in the
blotting buffer and place it on the electroblotting screen. Then
soak two pieces of 3MM paper in the blotting buffer and place
them on the sponge; these are followed sequentially by the gel
and PVDF membrane. Avoid incorporating air between the
various layers. Then place two more sheets of 3MM paper and
340
Typical Results
The current Zn2+Phos-tag SDS-PAGE produced better separations of phosphorylated species of various proteins than did the
previous Mn2+Phos-tag SDS-PAGE. Comparative studies were
performed by using several molecular-weight markers (1497 kDa)
and four standard phosphoproteins: -casein (24 kDa), -casein
(24 kDa), ovalbumin (45 kDa), and pepsin (35 kDa) (Fig. 3).
Differences in the degrees of migration and banding patterns
between the phosphorylated forms of the proteins and their
dephosphorylated counterparts, obtained by treatment with AP,
Fig. 3 (continued) Zn2+Phos-tag (right-hand panel). The figures in parts (a) and (b) are reproduced from ref. [45]
with the permission of the publisher, Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany. (c) 10 % (w/v)
Polyacrylamide gel containing 0 M (left-hand panel) or 100 M polyacrylamide-bound Mn2+Phos-tag (righthand panel). The figures in part (c) are reproduced from ref. [42] with permission of the publisher, Wiley-VCH
Verlag GmbH & Co. KGaA, Weinheim, Germany. Lanes 111 correspond to (1) -casein (1.0 g), (2) -casein
treated with AP for 5 min (1.5 g), (3) -casein treated with AP for 12 h (1.0 g), (4) -casein (1.0 g), (5)
-casein treated with AP for 5 min (1.5 g), (6) -casein treated with AP for 12 h (1.0 g), (7) molecular-weight
markers (APRO marker low range; APRO Science, catalogue number SP-0110) [from the top: phosphorylase b
(97 kDa), BSA (66 kDa), ovalbumin (45 kDa), carbonic anhydrase (29 kDa), trypsin inhibitor (21 kDa), and lysozyme (14 kDa)], (8) ovalbumin (1.0 g), (9) ovalbumin treated with AP for 12 h (1.0 g), (10) pepsin (1.0 g),
and (11) pepsin treated with AP for 12 h (1.0 g). The Rf value of 1.0 is defined as the position of the BPB dye.
The gels were stained with CBB dye. Apparent molecular weights of marker bands are indicated on the right-hand
side of the left panel. By using the Zn2+Phos-tag gel buffered with BisTrisHCl, three distinct phosphorylated
species of ovalbumin could be separated, as indicated by arrowheads (see lane 8 in part (b); from the top:
species phosphorylated at both Ser-68 and Ser-344, species monophosphorylated at Ser-68, and species
monophosphorylated at Ser-344, respectively) [47]
Fig. 3 Comparison of mobilities of standard phosphoproteins in Zn2+Phos-tag gels buffered with TrisAcOH
and BisTrisHCl and in Mn2+Phos-tag gel. (a) TrisAcOH system on 12 % (w/v) polyacrylamide gel containing 0 M (left-hand panel) or 100 M polyacrylamide-bound Zn2+Phos-tag (right-hand panel). (b) BisTris
HCl system on 8.0 % (w/v) polyacrylamide gel containing 0 M (left-hand panel) or 100 M polyacrylamide-bound
342
343
344
345
Fig. 4 (continued) The separated sample lane was cut, and then the part corresponding to 80250 kDa was subjected to Zn2+Phos-tag SDS-PAGE [4 % (w/v)
polyacrylamide, 50 M Zn2+Phos-tag] buffered with BisTrisHCl as the second dimension. The Phos-tag gel was strengthened with 0.5 % (w/v) SeaKem
Gold agarose. All the spots appeared as smeared images after gel staining,
because the BisTris-buffered gel did not have adequate sieving properties at
concentration of less than 4 % (w/v) polyacrylamide. The Rf value of 1.0 is
defined as the position of the BPB dye. Figures are reproduced from ref. [45] with
permission of the publisher
Fig. 5 Comparison of mobility of phosphorylated ERK between the BisTrisHCl buffer system and TrisAcOH
buffer system. (a) A431 whole lysate (, 10 g proteins) and lysate from EGF-stimulated cells (+, 10 g proteins) were subjected to Zn2+Phos-tag SDS-PAGE [8.0 % (w/v) polyacrylamide and 25 M Zn2+Phos-tag]
using the BisTrisHCl buffer system or TrisAcOH buffer system. The gels were analyzed by Western blotting with
347
Notes
1. All reagents and solvents used are purchased at the highest
commercial quality available and used without further purification. All aqueous solutions are prepared by using deionized
and distilled water.
2. Hydrochloric acid (HCl) is dangerously irritating to the skin,
eyes, and mucous membranes. When handing this chemical,
work in a chemical fume hood and wear gloves, eye protection,
and a mask.
Fig. 5 (continued) a phosphorylation-independent anti-ERK antibody (Millipore, Bedford, MA). The lysate from
the EGF-stimulated cells (10 g proteins) was analyzed by 2-DE, consisting of normal SDS-PAGE [8.0 % (w/v)
polyacrylamide] as the first dimension and Zn2+Phos-tag SDS-PAGE [8.0 % (w/v) polyacrylamide, 25 M
Zn2+Phos-tag] as the second dimension. The Rf value of 1.0 is defined as the position of the BPB dye. (b) The
time-course of phosphorylation of ERK after stimulation with EGF (250 ng/mL) was analyzed by Zn2+Phos-tag
SDS-PAGE [8.0 % (w/v) polyacrylamide and 25 M Zn2+Phos-tag] using the BisTrisHCl buffer system or the
TrisAcOH buffer system. Each lane contains 10 g of proteins. The gels were analyzed by Western blotting
with the dual-phosphorylation-specific anti-pT202/Y204 antibody (Cell Signaling Technology, Danvers, MA, USA),
and then the same blot was reprobed with the phosphorylation-independent anti-ERK antibody. (c) A431 whole
lysate (control, 10 g proteins) and lysate from EGF-stimulated cells (10 g proteins, 250 ng/mL of EGF for
5 min) were subjected to Zn2+Phos-tag SDS-PAGE [8.0 % (w/v) polyacrylamide and 25 M Zn2+Phos-tag]
using the BisTrisHCl buffer system or TrisAcOH buffer system, followed by immunoblotting with the
phospho-site-specific anti-pT202 antibody or anti-pY204 antibody (left and center panels, respectively, in both
systems) (Signalway Antibody, Pearland, TX, USA). Lysates from EGF-stimulated cells (10 g proteins, 250 ng/
mL of EGF for 5 min) pretreated with calyculin A (100 nM, 30 min) (Cell Signaling Technology), pervanadate [a
mixture of 1.0 mM sodium orthovanadate (Sigma-Aldrich) and 3.0 mM H2O2 (Nacalai Tesque), 30 min], or
PD98059 (100 M, 60 min) (Calbiochem, La Jolla, CA, USA) were similarly analyzed with the phosphorylationindependent anti-ERK antibody (right panels in the both systems). Figures are reproduced from ref. [45] with
permission of the publisher
348
349
350
351
Acknowledgments
This work was supported in part by KAKENHI Grants (24590050,
25293005, 25560417, 25117718, and 26460036) and a research
grant from the Takeda Science Foundation.
352
References
1. Hunter T (2000) Signaling: 2000 and beyond.
Cell 100:113127
2. Olsen JV, Blagoev B, Gnad F et al (2006)
Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 127:
635648
3. Ubersax JA, Ferrell JE Jr (2007) Mechanisms
of specificity in protein phosphorylation. Nat
Rev Mol Cell Biol 8:530541
4. Manning G, Whyte DB, Martinez R et al
(2002) The protein kinase complement of the
human genome. Science 298:19121934
5. Kerk D, Templeton G, Moorhead GB (2008)
Evolutionary radiation pattern of novel protein
phosphatases revealed by analysis of protein
data from the completely sequenced genomes
of humans, green algae, and higher plants.
Plant Physiol 146:351367
6. Brognard J, Hunter T (2011) Protein kinase
signaling networks in cancer. Curr Opin Genet
Dev 21:411
7. Martin L, Latypova X, Terro F (2011) Posttranslational modifications of tau protein:
implications
for
Alzheimers
disease.
Neurochem Int 58:458471
8. Kosako H, Nagano K (2011) Quantitative
phosphoproteomics strategies for understanding protein kinase-mediated signal transduction
pathways. Expert Rev Proteomics 8:8194
9. Schmelzle K, White FM (2006) Phosphoproteomic approaches to elucidate cellular signaling networks. Curr Opin Biotechnol 17:
406414
10. Stasyk T, Dubrovska A, Lomnytska M et al
(2005) Phosphoproteome profiling of transforming growth factor (TGF)- signaling:
abrogation of TGF1-dependent phosphorylation of transcription factor-II-I (TFII-I)
enhances cooperation of TFII-I and Smad3 in
transcription. Mol Biol Cell 16:47654780
11. Agrawal GK, Thelen JJ (2006) Large scale
identification and quantitative profiling of
phosphoproteins expressed during seed filling
in oilseed rape. Mol Cell Proteomics 5:
20442059
12. Kinoshita E, Takahashi M, Takeda H et al
(2004) Recognition of phosphate monoester
dianion by an alkoxide-bridged dinuclear
zinc(II) complex. Dalton Trans 11891193
13. Kinoshita-Kikuta E, Kinoshita E, Yamada A
et al (2006) Enrichment of phosphorylated proteins from cell lysate using a novel phosphateaffinity chromatography at physiological pH.
Proteomics 6:50885095
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
353
354
Chapter 25
Enrichment and Identification of Bacterial Glycopeptides
by Mass Spectrometry
Nichollas E. Scott and Stuart J. Cordwell
Abstract
Large-scale analysis of protein N- and O-linked glycosylation by mass spectrometry has traditionally been
performed in eukaryotes by parallel approaches aimed at elucidating glycan structures (glycomics) and
their formerly glycosylated peptides (glycoproteomics) without reference to their intact state. Such analyses depend heavily on commercial glycosidases (e.g. protein N-glycosidase F) that can remove glycans
from the peptide backbone for separate analyses. Bacterial glycosylation has only recently been identified
as a widespread phenomenon. In many cases however, unique bacterial sugars preclude enzymatic removal,
therefore ultimately requiring a site-specific approach for intact glycopeptide analysis. Here, we describe
protocols for the enrichment of bacterial glycopeptides using zwitterionichydrophilic interaction liquid
chromatography (ZIC-HILIC) and their analysis using liquid chromatography coupled to electrospray
ionization tandem mass spectrometry (LC-MS/MS) with collision-induced dissociation (CID) and higher
energy collisional dissociation (HCD) fragmentation for glycan structure elucidation and glycopeptide
identification.
Key words Glycoprotein, Hydrophilic interaction liquid chromatography (HILIC), Mass spectrometry,
Membrane proteins, Proteomics
Introduction
1.1 Protein
Glycosylation
in Bacteria
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_25, Springer Science+Business Media New York 2015
355
356
Over the last decade there have been a number of newly characterized general glycosylation systems identified in a wide range of
bacterial species [1, 2, 5, 6, 9, 10], which have facilitated the
elucidation of glycan structures and their protein targets.
Glycosylation should not be regarded as a single discrete modification but rather as a class of several possible types, broadly clustered into N- and O-linkages (although rarer linkages such as
C-mannosylation also occur), that can occur in both gram-negative and -positive organisms and are characterized by the enzymatic formation of a chemical linkage between a carbohydrate
chain or monomer and protein substrates by the actions of oligosaccharyltransferases (OSTases) or GTases. Addition of glycans to
protein can also be via en bloc or sequential transfer. For en bloc
transfer, the glycan is typically assembled on a membrane-bound
lipid carrier (e.g. the C. jejuni N-linked and Neisseria meningitidis O-linked systems) and added upon completion to synthesized
proteins within the periplasmic space; while sequential transfer
implies the addition of a single sugar to protein potentially followed by additional monosaccharides (e.g. Haemophilus influenzae high molecular weight adhesin 1 (HMW1) glycosylation and
C. jejuni flagellin glycosylation [11, 12]).
The OSTases and GTases, which appear to be present across
bacterial genera, typically target protein substrates at similar consensus motifs (e.g. the D/E-X-N-X-S/T [X Pro] of N-linked
glycosylation in C. jejuni), or in disordered protein regions (e.g.
the low complexity or repeat regions targeted by ligase-like
O-linked systems). Glycosylation events can involve the attachment of multiple carbohydrates, ranging from single hexose or
N-acetylhexosamine sugars or bacterial-specific carbohydrates such
as pseudaminic acid or other nonulosonic acid derivatives [2, 11,
13], to complex glycan structures [2, 5, 6, 10]. Further complexity
is generated by the addition of non-carbohydrate moieties such as
methylated aspartic acid, phosphate, and phosphoethanolamine
357
358
Materials
2.1 Generation
of Bacterial Cells
2.2 Preparation
of Protein Samples
2.4 Desalting
of Peptides
359
2.6 ZIC-HILIC
Enrichment
of Glycopeptides
2.7
LC-MS/MS
360
Data Analysis
1. MASCOT (Matrix Science) search engine for peptide identification (proprietary software).
2. Expert Annotation website (http://www.biochem.mpg.de/
mann/tools/).
3. Xcalibur MS data viewer (Thermo Scientific).
4. GPMAW 8.2 (Lighthouse Data; proprietary software).
5. Proteome Discoverer (Thermo Scientific; proprietary software).
6. Microsoft Excel (Microsoft; proprietary software).
3
3.1
Methods
Bacterial Growth
1. Grow bacterial strain of interest to the required phase or density and harvest cells by centrifugation at 2,000 g for 10 min
at 4 C.
2. Wash cell pellets two times with ice-cold PBS and collect cells
by centrifugation at 2,000 g for 10 min at 4 C.
3. Freeze dry cells and store at 80 C for long-term storage. To
ensure adequate material, >100 mg of freeze-dried cells should
be generated.
3.2 Preparation
of Protein Samples
3.2.1 Whole Cell Lysates
361
3.4 Desalting
of Peptides (See Note 10)
362
Fig. 1 Construction of ZIC-HILIC STAGE tips. (a) Excision of C8 disk, using a Kel-F Hub NDL blunt needle generating a 2 mm C8 disk; (b) Insertion of C8 disk into P10 tip using the Plunger assembly enables the C8 disk to be
lodged at the end of the P10 tip; (c) Packing of ZIC-HILIC material onto the C8 disk [I) shows an unpacked tip;
II) a tip with only the C8 disk; and III) the final ZIC-HILIC tip to be used for glycopeptide enrichment containing
0.5 cm of packed ZIC-HILIC material]
3.6 ZIC-HILIC
Enrichment
of Glycopeptides
1. Resuspend peptides in ZIC-HILIC buffer B, 200 g of peptide to 100 L of ZIC-HILIC buffer B and vortex. Store at
4 C.
2. Prepare ZIC-HILIC column for glycopeptide binding, wash
with four washes of 50 L ZIC-HILIC buffer A, four washes
of 50 L ZIC-HILIC buffer C and four washes of 50 L ZICHILIC buffer B (see Note 11).
3. Load peptides onto ZIC-HILIC STAGE tip column and wash
ten times with 50 L ZIC-HILIC buffer B (see Note 12).
4. Elute glycopeptides with three washes of ZIC-HILIC buffer A.
5. Dry eluted glycopeptides by vacuum centrifugation and store
at 20 C prior to analysis.
3.7
LC-MS/MS
363
Data Analysis
Fig. 2 Data analysis of ZIC-HILIC enriched glycopeptides. (a) Extracted ion chromatogram of the diagnostic oxonium ion 204.07204.09 m/z, the glycopeptide
979.46704 m/z, charge state +3, is highlighted in red; (b) CID fragmentation of the
glycopeptide 979.46704 m/z containing the glycan HexNAc-Hex2-HexNAc-258;
365
a precursor mass tolerance of 10 ppm and an MS/MS fragment mass tolerance of 0.05 Da for HCD scans and 0.6 Da
for CID scans.
4. Export MS/MS scan events that do not result in peptide identification to Microsoft Excel.
5. Use the mgf graph module in GPMAW 8.2 to identify possible glycopeptides in this list that contain the diagnostic oxonium 204.086 m/z ion (see Note 17).
6. Manually inspect the CID and HCD scan events of each potential glycopeptide using Xcalibur to confirm that these scans of
the same peptide are dissimilar (Fig. 2b, c).
7. For HCD scan events, the deglycosylated peptide and the peptide plus one carbohydrate are typically the most abundant
ions and can be used to determine a putative deglycosylated
mass of the glycopeptide (Fig. 2d).
8. To facilitate a glycopeptide assignment from an HCD scan,
ions below the mass of the predicted deglycosylated peptide
are extracted within Xcalibur using the Spectrum List function.
Ions with a deconvoluted mass above that of the deglycosylated peptide and those corresponding to known carbohydrate
oxonium ions are removed and the resulting peak list used to
generate a .mgf file that can be searched with MASCOT, using
the same parameters defined for the initial search (step 3).
9. To further validate glycopeptide matches, HCD scan events
are annotated using the Expert Annotation tool (Fig. 2e).
Notes
1. Avoid heating urea/thiourea solution to decrease the risk of
protein carbamylation [21].
2. Decant TFA or use glass pipette, avoid the introduction of
plastic tips into the stock solution.
3. An alterative method for the construction of ZIC-HILIC columns using constricted GELoader tips in place of C8 support
has also been reported, for instructions see [22].
4. Alterative ion pairing agents can be used and have been shown
to augment the efficiency of HILIC enrichment [23]. With
certain samples these agents can be more effective than FA and
should be considered during optimization.
366
5. For complete assignment of glycopeptides (absolute site localization independent of any sequence motif constraints), CID,
HCD and electron transfer dissociation (ETD) approaches can
be used provided the instrumentation supports these fragmentation techniques. Alternatively, if only the peptide sequence
and confirmation of glycosylation are required, beam-type
fragmentation on Q-Tof or Q-Exactive instrumentation may
be used.
6. An alterative to Benzonase nuclease is Cryonase (Clontech
Laboratories Inc., Takara Bio Group), which provides enhanced
nuclease digestion at 4 C.
7. Ensure the sample remains chilled during sonication and is at
4 C before beginning another round of sonication.
8. With each round of sonication the amount of extracted protein
should decrease, for most gram-negative bacteria we have
examined 5 rounds results in effective solubilization of membrane material.
9. Alternative enzymes may also be useful for generation of glycopeptides of suitable length conducive to their analysis
(e.g. GluC, AspN). Importantly, due to the steric hindrance
provided by larger glycans, non-specific proteinases such as
thermolysin, pepsin and proteinase-K can also be used to
generate peptides of adequate length to enable glycopeptide
identification [24]. If these enzymes are used, the corresponding digestion buffer may also require alteration to
ensure maximum efficiency.
10. The presence of salts and urea/thiourea severely compromises
the enrichment of glycopeptides using ZIC-HILIC. For optimal results ensure the effective clean-up of peptide samples.
11. Do not allow the ZIC-HILIC resin to become dry during the
glycopeptide enrichment process, as this appears to severely
compromise the final glycopeptide yield.
12. For complex peptide lysates, 10 washes with ZIC-HILIC buffer B is effective for the removal of the majority of nonglycosylated peptides, but for less complex samples such as
single proteins, 3 washes are typically sufficient. The number of
wash steps should be varied to assess the effect on the enrichment of glycopeptides for any new sample type.
13. A microflow LC system can also be used but it should be noted
that the use of high flow rates would lead to a loss of sensitivity.
14. As the addition of a glycan increases the mass of a peptide but
does not increase the observed charge state, to increase the
chances of selecting a glycopeptide the lower threshold of the
MS1 mass window can be increased to 500 m/z, thus
5001,800 m/z.
367
Acknowledgments
This work was supported by an Australian Research Council
Discovery Project Grant to S.J.C. (ARC DP 110103573). N.E.S.
is supported by a National Health and Medical Research Council
of Australia (NHMRC) Overseas (Biomedical) Early Career
Fellowship (APP1037373) and a Michael Smith Foundation for
Health Research Trainee Postdoctoral Fellowship (award # 5363).
References
1. Iwashkiw JA, Vozza NF, Kinsella RL, Feldman
MF (2013) Pour some sugar on it: the expanding world of bacterial protein O-linked glycosylation. Mol Microbiol 89:1428
2. Nothaft H, Szymanski CM (2010) Protein
glycosylation in bacteria: sweeter than ever.
Nat Rev Microbiol 8:765778
3. Szymanski CM, Burr DH, Guerry P (2002)
Campylobacter protein glycosylation affects
host cell interactions. Infect Immun 70:
22422244
4. Howard SL, Jagannathan A, Soo EC, Hui JP,
Aubry AJ, Ahmed I, Karlyshev A, Kelly JF,
Jones MA, Stevens MP, Logan SM, Wren BW
(2009) Campylobacter jejuni glycosylation
island important in cell charge, legionaminic
acid biosynthesis, and colonization of chickens. Infect Immun 77:25442556
5. Iwashkiw JA, Seper A, Weber BS, Scott NE,
Vinogradov E, Stratilo C, Reiz B, Cordwell SJ,
Whittal R, Schild S, Feldman MF (2012)
Identification of a general O-linked protein glycosylation system in Acinetobacter baumannii
6.
7.
8.
9.
368
19.
20.
21.
22.
23.
24.
25.
26.
Chapter 26
In-Gel Peptide IEF Sample Preparation for LC/MS Analysis
Tom Berkelman, Sricharan Bandhakavi, and Aran Paulus
Abstract
The technique of proteolytically digesting a sample and identifying its protein components by liquid
chromatography followed by mass spectrometry (LC-MS) is a widely used analytical tool. Prior fractionation by isoelectric focusing (IEF) may be performed to increase the depth of proteome coverage. Here,
we describe a method for in-gel IEF separation of a proteolytic digest that utilizes commercially available
immobilized pH gradient (IPG) strips and a widely used IEF instrument.
Key words LC-MS, Isoelectric focusing, IEF, Proteomics, Off-gel, In-gel, Immobilized pH
gradient
Introduction
The separation of proteolytically digested samples by reversedphase liquid chromatography coupled to mass spectrometry with
electrospray ionization (LC-MS) is a powerful and widely used
strategy for proteomic analysis. This technique can detect thousands of individual peptides, and by referencing a database, identify
their proteins of origin. Termed bottom-up or shotgun proteomics, this is a powerful profiling method for proteins present in
a biological sample [1]. Additional depth of coverage can be
obtained by preceding LC-MS with an orthogonal upstream fractionation [2] generating fractions of lower complexity that are analyzed separately. Orthogonal separation techniques that are used for
this purpose include strong cation exchange chromatography (SCX)
[3], hydrophilic interaction chromatography (HILIC) [4], and isoelectric focusing (IEF) [58]. The depth of proteome coverage
obtainable with IEF can be higher than that obtained with chromatographic methods [9, 10] and IEF has become widely used for
this application. IEF has the additional benefit of providing a search
constraint for peptide identification, since the isoelectric point (pI)
of a peptide can be predicted directly from its sequence [1113].
IEF fractionation of sample digests is predominantly performed
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_26, Springer Science+Business Media New York 2015
369
370
using immobilized pH gradient (IPG) strips due to the reproducibility and versatility afforded by commercial pre-cast IPG gels covering various pH ranges. The separation may either be performed
in an off-gel mode or in an in-gel mode. In the off-gel
mode, the digest is electrophoretically fractionated through a series
of sample cups placed in contact with the IPG gel [8, 14]. The
separated peptides are recovered in solution directly from the sample cups. In the in-gel mode, the digest is loaded into an IPG gel
and separated within the gel matrix [10, 12, 13, 1517]. The separated peptides are eluted from excised sections of IPG gel. Whereas
the off-gel method offers a certain degree of convenience, in-gel
separations have several advantages. Peptide yields have been found
to be higher and the separation is more rapid [18]. Additionally,
in-gel instrumentation allows greater flexibility in the selection of
IPG strip length and pH range. There is also no restriction with
in-gel separations on the proportion of the length of IPG strip
represented by each fraction. An off-gel fraction, by necessity, represents a section of the gel corresponding to the footprint of a
single sample cup. The pH range of an in-gel separation is determined by the spacing of the cuts made. The number of fractions
generated can therefore be tailored more closely to the needs of
the experimenter. Since the distribution of peptide pI is variable
across the pH scale, it can be advantageous to cut the IPG strip
into sections of variable length such that each eluted fraction contains a similar number of peptides, thereby taking fullest advantage
of the downstream analysis methodology [19]. This strategy is not
possible with off-gel IEF. The following method describes in-gel
preparative IEF using the PROTEAN i12 IEF System, a widely
used instrument which controls voltage and current independently
to each IPG strip, allowing multiple pH ranges, or samples of differing conductivity to be run simultaneously.
Materials
Reagents and prepared buffer stocks are from Bio-Rad or SigmaAldrich except when indicated. Reagent quality should be
reagent grade or better. Water should be of the highest purity
possible. Measures should be taken to avoid keratin contamination (see Note 1).
2.1 Lysate
Preparation
(See Note 2)
2.2 Reduction
and Alkylation
371
1. 1.2 M dithiothreitol (DTT): Weigh 185.1 mg of DTT and dissolve in sufficient water for 1 ml of solution. The solution may
be stored frozen in aliquots.
2. 600 mM iodoacetamide: Weigh 111.0 mg of iodoacetamide
and dissolve in sufficient water for 1 ml of solution.
2.3
Digestion
2.4 Desalting
and Concentration
of Peptide Digest
2.6
Peptide Elution
1. Hexane.
2. Razor blades.
3. 0.1 % TFA, 35 % acetonitrile: Mix 6.5 ml of 0.1 % TFA in water
with 3.5 ml of 0.1 % TFA in acetonitrile.
4. 0.1 % TFA, 80 % acetonitrile: Mix 2 ml of 0.1 % TFA in water
with 8 ml of 0.1 % TFA in acetonitrile.
Methods
3.1 Lysate
Preparation
372
3.3
Digestion
3.4 Desalting
and Concentration
of Peptide Digest
1. Equilibrate four 3 ml Empore C18 solid phase extraction cartridges for de-salting. Place each cartridge in a 15 ml conical
centrifuge tube. Apply 1 ml of methanol to each column and
centrifuge at 1,500 g for 1 min in a fixed-angle rotor. Discard
the flow-through and repeat with 0.5 ml 0.1 % TFA, 70 % acetonitrile followed by 0.5 ml 0.1 % TFA in water.
2. Apply one-quarter of the digested sample to each column
(roughly 1 ml each) and centrifuge at 135 g for 10 min.
Discard the flow-through.
3. Wash each column three times with 0.5 ml 0.1 % TFA, centrifuging as in step 2 and discarding the flow-through after each
centrifugation.
4. Elute the peptides from each column with 0.5 ml 0.1 % TFA,
70 % acetonitrile, centrifuging as in step 2 and reserving the
eluates. Repeat the elution and pool all of the eluates.
5. Determine the absorbance at 280 nm of the pooled eluates,
using 0.1 % TFA, 70 % acetonitrile to blank the spectrophotometer. Estimate the peptide concentration and yield by
assuming that a 1 mg/ml peptide solution has an absorbance
at 280 nm of 1.1 (see Note 6).
6. Dry the peptides down by vacuum centrifugation.
373
3.6
Peptide Elution
1. Following electrophoresis, briefly rinse each IPG strip in a beaker of hexane (see Note 13).
2. Print out a cutting template consisting of 12 parallel 1 cm lines
placed 9 mm apart from one another (see Fig. 1).
3. Place a clean sheet of glass over the template. Place each IPG strip
gel-side up on the glass over the template as shown in Fig. 1. Cut
through the gel and backing over each line with a razor blade,
using a fresh blade for each cut, generating twelve 9 mm gel sections. Keep track of which section corresponds to which fraction.
4. Place each section in a 1.5 ml tube and discard the 5 mm pieces
from each end of the IPG strip (the sites of electrode contact).
374
Site of electrode
wick contact
1
pH 3
Site of electrode
wick contact
2
10
11
pH 10
Fig. 1 Use of template for cutting IPG strip to generate IEF fractions. Each IPG strip is placed on a clean sheet
of glass over a template comprising twelve parallel 1 cm lines placed 9 mm apart so that the ends of the IPG
gel extended 5 mm beyond the outermost lines. The IPG strip is cut over each line. Fraction numbers are
indicated
5. Add 150 l 0.1 % TFA in water to each tube. Incubate the tube
with the gel section and 0.1 % TFA for 1.5 h with gentle shaking. Remove the liquid and reserve (see Note 14).
6. Repeat step 5 using 0.1 % TFA, 35 % acetonitrile. Pool the
removed liquid with the eluted material from step 5.
7. Repeat step 6 using 0.1 % TFA, 70 % acetonitrile. Pool the
removed liquid with the eluted material from steps 5 and 6.
8. Dry the pooled material by vacuum centrifugation.
3.7 Solid Phase
Extraction Clean-up
of Peptide Fractions
375
Notes
1. Clean technique is practiced throughout in order to minimize
keratin contamination. Solutions containing urea, iodoacetamide, ammonium bicarbonate or enzyme are prepared immediately before use.
2. The technique may be applied to any mammalian cell line cultured by any method.
3. The basic procedure may be scaled up or down, depending on
the number of IPG strips to be run.
4. Reduction and alkylation of cysteine thiols prior to digestion
and LC-MS enables more effective digestion and simplifies MS
analysis as all cysteine residues will be modified with an acetamido group. The sample is reduced with DTT and alkylated
with an excess of iodoacetamide.
5. A two-step digestion is employed utilizing Lys-C at an enzyme
to protein ratio of 1:50 followed by trypsin at an enzyme to
protein ratio of 1:100. Lys-C cleaves at lysine residues and is
compatible with 8 M urea and thus can be applied in undiluted lysis solution. Trypsin cleaves at both lysine and arginine residues and digests optimally only when the urea
concentration is 2 M or lower. The reaction is therefore
diluted prior to addition of trypsin. This procedure ensures
more complete digestion than trypsin alone. If desired, the
digest may be directly diluted and treated with trypsin, omitting the Lys-C digestion step, however digestion may not be
as efficient or complete.
6. The portion of the sample used to measure the absorbance at
280 nm should be added back to the pooled eluates following
the measurement. The cuvette used for the measurement
should therefore be thoroughly cleaned before use, or a disposable, UV transparent cuvette should be used.
7. Handle IPG strips from the ends using forceps.
8. Other pH ranges may be used.
9. Do not write on the IPG strip backing. The ink will be extracted
in subsequent steps and contaminate the sample.
10. The sample is applied to the entire length of the immobilized
pH gradient by rehydration into a dry IPG strip. As voltage is
applied, each peptide in the sample focuses to a narrow zone in
the pH gradient corresponding to its pI.
11. Mineral oil prevents evaporation of the sample solution.
12. Additional details of the sample application and IEF procedure
may be found in the user manual for the PROTEAN i12 IEF
System.
376
3500
Peptide identifications
3000
2500
2000
1500
1000
500
0
1
10
11
Fraction number
Fig. 2 Peptide distribution across the separation. Peptide fractions from a HeLa cell lysate were generated as
described. Each fraction was dissolved in 10 l 0.1 % formic acid, 2 % acetonitrile. Half of each fraction was
analyzed by LC-MS as described in [21]. Peptides found uniquely in a single fraction are indicated in a darker
shade. Peptides not restricted to a single fraction are indicated in a lighter shade (see Note 17)
377
10.00
9.00
Average pI
8.00
7.00
6.00
5.00
4.00
3.00
1
10
11
Fraction
Fig. 3 Average pI of peptides found in each fraction. The isoelectric point of each identified peptide was calculated from the peptide sequence using the compute pI/MW tool on the ExPasy server (www.expasy.org), which
is based on the method described in [22]. The average pI for each reaction is shown. Error bars show the
standard deviation within each fraction
Fig. 4 Distribution of LC-MS peptide identifications from the unfractionated digest and following two different
IEF fractionation methods. Identical quantities of digested HeLa cell lysate were fractionated as described here
(in-gel), and by off-gel IEF as described in [8] using an IPG strip of the same length and pH gradient. LC-MS
analysis was performed as described in [21]. An identical quantity of unfractionated digest was analyzed the
same way (Unfractionated). The Venn diagram shows the total number of peptide identifications generated by
each method, the number of identifications unique to each method, and the identifications in common among
the different methods (see Note 18)
378
11.
12.
13.
14.
15.
16.
17.
18.
379
Chapter 27
Western Blotting Using In-Gel Protein Labeling
as a Normalization Control: Stain-Free Technology
Jennifer E. Gilda and Aldrin V. Gomes
Abstract
Western blotting is a commonly used laboratory technique for semi-quantifying protein amounts. It is
important when quantifying protein expression to account for differences in the amount of total protein
loaded onto the gel using a loading control. Common loading controls include housekeeping proteins,
such as -actin or GAPDH, quantified by Western blot, or total protein, quantified using a stain such as
Coomassie Brilliant Blue or Ponceau S. A more recently developed method for total protein quantification
utilizes stain-free technology, which has a linear dynamic detection range and allows for protein detection
on both gels and membranes. Here, we describe the theory and use of stain-free gels for total protein
quantification and normalization of Western blots.
Key words Stain-free technology, Western blotting, Loading control, Data normalization,
Immunoblotting
Introduction
Western blotting is a highly valuable laboratory technique used to
quantify protein levels [1]. When carried out properly, Western
blots can be used to accurately semi-quantify amounts of specific
proteins in a complex sample; however, several considerations must
be taken into account to ensure the accuracy and quality of the blot
[2, 3]. One important consideration is the amount of protein in
each lane, which may vary due to errors in protein concentration
determination, loading volume, or differences in transfer efficiency
[4]. To account for these potential differences, the amount of the
target protein should be normalized to the amount of protein
present in each lane using a loading control.
A commonly used loading control is the expression of a housekeeping protein, such as -actin, glyceraldehyde 3-phosphate
dehydrogenase (GAPDH), or -tubulin, quantified by Western
blot. The rationale for using housekeeping proteins is that they are
constitutively expressed and necessary for maintaining cell viability;
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_27, Springer Science+Business Media New York 2015
381
382
Materials
All solutions should be prepared with ultrapure water (18 M)
and analytical grade reagents. All reagents are prepared and stored
at room temperature unless otherwise indicated. Sodium azide is
not added to any of the solutions and all waste disposal protocols
should be followed when discarding Western blotting waste.
383
2.2
Protein Transfer
Corporation,
Texas,
USA)
384
Methods
All procedures are carried out at room temperature unless otherwise noted.
3.1 Sample
Preparation
and SDS-PAGE
385
Table 1
Gel loading pattern
Lane
1 2
7 8
10 11 12 13 14 15 16 17 18
10 20 30 40 50
10 20 30 40 50
10 20 30 40 50
Volume (L)
10 10 15 20 25
10 10 15 20 25
10 10 15 20 25
Concentration (g/L)
386
3.2
Protein Transfer
3.3
Immunostaining
Fig. 2 Detection of total protein in rat heart homogenates on nitrocellulose membrane using the stain-free
method. (a) Detection of 1050 g total heart protein on nitrocellulose membrane using stain-free imaging. (b)
Quantification of total protein detected on the membrane versus amount of protein loaded onto the gel (n = 3
for each data point). Data are represented as mean standard error
387
3. Incubate with shaking in peroxidase-conjugated rabbit antimouse IgG secondary antibody for 1 h (see Note 19). Pour off
the secondary antibody and rinse briefly with TBST, then rinse
three times for 5 min in TBST with shaking.
4. To image, lift the membrane with forceps, taking care to touch
only the side of the membrane. Allow the TBST to drain off
the membrane so that it does not dilute the ECL substrate;
however, take care not to dry the membrane. The edge of the
membrane may be dabbed with a Kimwipe to remove excess
liquid. Place the membrane on the imaging surface, laying
from one edge to the other to exclude bubbles.
5. Combine equal parts of Clarity western peroxide reagent and
Clarity western luminol/enhancer reagent. Mix the solution
and pipette it onto the membrane (protein side up) so that
every part of the membrane is evenly covered with ECL substrate (see Note 20). Either proceed directly to the imaging
step (incubation in between addition of the ECL substrate and
imaging is not necessary) or incubate the ECL covered membrane for 2 min. If the ECL reagent is left on the membrane
for 2 min, then remove the excess ECL reagent and cover the
membrane with a thin sheet protector to prevent drying.
6. Image the blot with the Chemi Hi Sensitivity setting (Image
Lab software) and then adjust the exposure time and sensitivity/resolution settings as needed. If the target protein (Fig. 3)
388
Notes
1. Unused sample buffer may be stored at 20 C for up to 3
months or at 80 C for more than 6 months.
2. Any suitable pre-stained protein standard may be used; however
it should be noted that some protein standards give very strong
signals when imaged using the stain-free method, likely due to a
high tryptophan content of certain standards or the dye used to
stain them. This may interfere with total protein quantification.
3. Do not adjust the pH with acid or base. Once diluted to a 1
buffer the pH of the buffer should be 8.38.6.
4. Polyvinylidene difluoride (PVDF) membranes may also be
used with stain-free gels. We noticed that when the total protein on the membrane is imaged there is a slightly higher background with PVDF membranes than nitrocellulose membranes.
However, if fluorescently labeled secondary antibodies are
used, low fluorescence PVDF membranes are recommended.
5. 10 TBS can be stored at room temperature for at least 6 months
(10 TBS will crystallize if stored at 4 C).
6. Unused BSA should be stored at 4 C and may be used for up
to approximately 1 week after preparation. If the primary
antibody is to be used again, it is recommended that freshly
prepared BSA is used, as the BSA will limit the amount of
time in which the antibody can be used.
7. We have found that blocking and diluting certain antibodies
with solutions of BSA in TBST gives lower background than
when milk in TBST is used, though certain antibodies work
better with milk. If the antibody being used gives a high level
of background, the protocol may be optimized by testing the
antibody with solutions of BSA and milk to see which gives
lower background and stronger signal.
8. We recommend storing a small volume of ECL substrate at
room temperature for regular use, and the remainder at 4 C
for long-term storage. Using cold ECL reagent decreases the
sensitivity of the blot.
389
390
18. Many primary antibodies may be used more than once. If you
wish to reuse the antibody, you may store it at 4 C for up to
approximately 1 week. The antibody should not be used if the
BSA or milk has gone bad.
19. The quality of the secondary antibody is important for obtaining a high-quality Western blot. If consistently poor results
are obtained, we recommend trying a different secondary
antibody.
20. The volume of ECL substrate required to evenly cover the
membrane will depend on the area of the membrane. For an
uncut midi membrane, 56 mL ECL substrate is sufficient. If
a clear plastic support is used to cover the membrane during
imaging, a smaller volume of ECL substrate can be used, as the
plastic support will spread the ECL substrate over the membrane. If a long exposure time (90 s or longer) is required, it is
recommended to use a plastic support to prevent the membrane from drying during imaging. The ECL reagent should
not be left on the membrane for longer than 5 min as this will
result in a shorter time frame for which the maximum signal is
obtained.
Acknowledgement
This work was supported by the National Institutes of Health
Grants HL080101 and HL096819.
References
1. Kurien BT, Scofield RH (2006) Western blotting. Methods 38:283293
2. Towbin H, Staehelin T, Gordon J (1979)
Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci
U S A 76:43504354
3. Murphy RM, Lamb GD (2013) Important
considerations for protein analyses using antibody based techniques: down-sizing Western
blotting up-sizes outcomes. J Physiol 591:
58235831
4. Posch A, Kohn J, Oh K, Hammond M, Liu N
(2013) V3 stain-free workflow for a practical,
convenient, and reliable total protein loading
control in western blotting. J Vis Exp 82:
50948
391
Chapter 28
2-D Western Blotting for Evaluation of Antibodies
Developed for Detection of Host Cell Protein
Tom Berkelman, Adriana Harbers, and Sricharan Bandhakavi
Abstract
Recombinant proteins generated for therapeutic use must be substantially free of residual host cell protein
(HCP). The presence of host cell protein (HCP) is usually assayed by ELISA using a polyclonal antibody
mixture raised against a population of proteins derived from the host cell background. This antibody
should recognize as high a proportion as possible of the potential HCPs in a given sample. A recommended method for evaluating the assay involves two-dimensional electrophoretic separation followed by
Western blotting.
We present here a method using commercial anti-HCP antibody and samples derived from Chinese
Hamster Ovary (CHO) cells. The 2-D electrophoresis procedure gives highly reproducible spot patterns
and entire procedure can be completed in less than 2 days. Software analysis enables the straightforward
generation of percent coverage values for the antibody when used to probe HCP-containing samples.
Key words HCP, Host cell protein, 2-D Electrophoresis, Western blotting, Anti-HCP
Introduction
Most therapeutic proteins are produced by cultured cells which
have been engineered through recombinant DNA technology to
produce the relevant protein. The protein is either produced in the
host cell or secreted into the medium. In the case of intracellular
protein production, the host cells are harvested and lysed, and the
protein of interest is purified from the lysate. In the case of secreted
protein, the protein of interest is purified from the culture medium
in which the cells have been grown. In either case, the starting
material for purification of the protein of interest contains other
proteins from the host cell line, and these so-called host cell proteins (HCP) are potential impurities in the final drug substance.
HCP impurities can induce an immune response in the patient, so
their presence needs to be minimized. Validation and quality
control testing of any therapeutic protein needs to include a test to
show that HCP contamination is at a minimal level.
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_28, Springer Science+Business Media New York 2015
393
394
Materials
Reagents and prepared buffer stocks are from Bio-Rad Laboratories
or Sigma-Aldrich except when indicated. Reagent quality should
be reagent grade or better. Water should be of the highest purity
possible.
395
Fig. 1 Workflow for evaluation of antibodies developed for detection of host cell
proteins
396
2.1 Sample
Preparation
2.2 2-D
Electrophoresis,
First-Dimension IEF
2.3 2-D
Electrophoresis,
Equilibration
and SecondDimension SDS-PAGE
397
2.4 Staining
and Blotting
398
Methods
3.1 Sample
Preparation
3.2 2-D
Electrophoresis,
First-Dimension IEF
399
3.3 2-D
Electrophoresis,
Equilibration,
and SecondDimension SDS-PAGE
400
1. There should be at least two gels run for each sample. One is
to be stained and the other is to be blotted.
2. Carefully open the cassette for the gel to be stained and use a
spatula to separate the agarose overlay, including the IPG strip,
from the polyacrylamide gel.
3. Carefully peel the gel from the cassette and place it in a tray
containing 100 ml Oriole Fluorescent Gel Stain. Place the lid
on the staining tray incubate with gentle staining or rocking
for 90 min (see Note 26).
401
4. For each gel to be blotted open a transfer pack and place the
anode stack in the middle of the base of the Trans-Blot Turbo
cassette. The blotting membrane should be at the top of the
stack.
5. Carefully open the cassette for the gel to be blotted and use a
spatula to separate the agarose overlay, including the IPG strip,
from the polyacrylamide gel. Peel the gel from the cassette and
carefully place it on the anode stack, covering the blotting
membrane. Use the roller to remove any air bubbles that may
be trapped between the gel and the membrane, or within the
transfer stack (see Note 27). Place the cathode stack on top of
the gel and roll once again to remove trapped air.
6. Place the lid on the cassette and lock it into place by turning
the green knob clockwise. Ensure that the locking pins fully
engage their locking slots.
7. Turn on the Trans-Blot Turbo instrument and slide the cassette into a cassette bay.
8. Select TURBO, then select 2 MINI OR 1 MIDI GEL, then
select RUN for the occupied cassette bay(s). The transfer will
take 7 min. At the end of the run, RUN COMPLETE will
appear on the screen.
9. Disassemble the cassette. Remove and discard the cathode
stack. Carefully lift the gel and place it in a tray containing
100 ml Oriole Fluorescent Gel Stain. Place the lid on the staining tray incubate with gentle staining or rocking for 90 min
(see Note 28).
10. Using forceps, remove the membrane and rinse it briefly with
distilled or de-ionized water (see Note 29).
11. Float each membrane, protein side down, in a plastic tray containing 7 % acetic acid, 10 % methanol.
12. Wash each membrane in distilled or deionized water four times
for 5 min each.
13. Stain each membrane for 15 min with 20 ml SYPRO Ruby
Protein Blot Stain (see Note 30).
14. Wash each membrane in distilled or deionized water two to
three times for 1 min.
15. Capture images of the stained blots with the ChemiDoc MP
imager. Use the imager settings for SYPRO Ruby and use the
longest possible exposure time that does not result in image
saturation.
16. When each gel (both the unblotted gel and the replicate blotted gel) has been in Oriole stain for 90 min, wash briefly in
distilled or deionized water, and capture a fluorescent image
with the ChemiDoc MP imager. Use the imager settings for
Oriole stain. For the unblotted gel, use the longest possible
402
exposure time that does not result in image saturation. Use the
same exposure time for the replicate blotted gel. Ensure that
the zoom setting for all images is the same as used for the
stained membrane.
3.5 Immunodetection
3.6 Software
Analysis
and Generation
of a Match Rate
for Assessment
of Antibody Coverage
403
1. For each sample, use Image Lab software (used to capture the
images on the ChemiDoc MP) to open the images for the
SYPRO Ruby-stained blot and the immunodetected blot.
Crop both images to the edge of the membrane. If necessary,
first rotate each image so that the edges of the membrane are
parallel with the frame of the image. This will ensure that the
stained blot image and the Western blot image are in registry
for analysis with PDQuest software. Export the images for
analysis. (File > Export > Export for Analysis). This creates
16 bit tiff images.
2. Launch PDQuest. Open both of the files exported from Image
Lab.
3. Crop the images to include only the area containing well
resolved protein spots. Use the Advanced Crop function to
ensure that the cropped images are in registry with each other.
Place the crosshair on a recognizable feature common to both
images (Edit > Image > Advanced Crop > Define Crop Area,
see Fig. 2).
4. Create a new experiment (File > New Experiment). Select
both of the cropped images. Move through the next set of
screens by clicking Next. This will select the default options.
5. Follow the instructions on the Spot Detection Parameter
Wizard. Increase the sensitivity to maximize the number of
spots identified (see Fig. 3).
6. Select Proceed. In the next dialog box, select the cropped
image of the stained gel as the Master gel. Select Add
unmatched spots from all gel images to the Master gel. This
will create an artificial image containing all of the detected spots
from both the stained blot and the immunodetected Western
blot (see Fig. 4).
7. Choose default values by clicking Next through the successive
dialog boxes. The software will calculate matches and display
the Master image, the blot images and an Experiment Summary
with match rates (see Fig. 5). Match Rate 2 for the Western
blot image is the ratio of the number of protein spots recognized by the antibody to the total number of proteins detectable in the sample (see Fig. 6).
8. Choose View > Spot > Show Crosshairs or press F5 to show
the detected spots (see Fig. 7).
9. Show matched spots by selecting elect Analyze > Analysis Set
Manager, then Create and Matching (see Fig. 8). The
resulting images will have identified spots marked with a colored + and matched spots marked with an x in a different
color. This will aid in the determination of whether or not a
spot identification or match is valid (see Fig. 9).
404
405
406
408
409
Notes
1. Solutions containing urea are prepared immediately before use.
2. It is convenient to use a concentrated stock solution of
Bromophenol Blue, but this dye will not dissolve in unbuffered water. It is therefore prepared in Tris base solution.
3. Other pH ranges may be used. pH 310 NL (non-linear) was
chosen for this experiment because it covers most of the range
of protein pIs. This non-linear gradient is flatter in the region
of the gradient where most proteins are found, thereby spreading them out and allowing resolution of more individual
proteins.
4. Equilibration buffer is highly viscous and it may be difficult to
stir efficiently. Allow several hours for the urea to dissolve into
the liquid components.
5. The amount of Equilibration solution with DTT or iodoacetamide may be scaled up or down depending on the number of
IPG strips run. These solutions may be prepared from aliquoted
Equilibration buffer (see Subheading 2.3 step 3). Frozen tubes
of Equilibration buffer should be allowed to thaw slowly in a
beaker of water (do not heat).
410
411
alone generally results in a cleaner, higher resolution 2-D pattern [9]. In this experiment, the secreted proteins, which are
generally very soluble, are separated in a solution containing
urea alone. Cell lysate protein, which contains more hydrophobic proteins, is separated in a solution containing a urea/
thiourea mixture.
16. Each of the sample solution components has a specific role in
promoting high resolution IEF separation. Urea and thiourea
are protein denaturants that promote the complete unfolding
of polypeptide chains so that all ionizable groups are exposed
to the solution. CHAPS is a detergent that prevents protein
aggregation and promotes solubility. DTT is a thiol reductant
that breaks disulfide bonds within and between polypeptide
chains, and maintains proteins in a fully reduced state. Carrier
ampholytes enhance protein solubility by minimizing protein
aggregation due to chargecharge interactions. Bromophenol
blue is a tracking dye. Its inclusion is not necessary, but clearance of the dye from the IPG strip during electrophoresis provides a visual confirmation that electrical current is being
delivered to the IPG strip appropriately.
17. Each sample is run in replicate and separated on two seconddimension gels, one of which is stained and the other blotted.
18. Recombinant therapeutics can be expressed either as secreted
proteins recovered from the culture medium or as cellular proteins that are recovered from cell lysates. The HCPs found in
either case are expected to differ from each other. Cell culture
medium, however, is expected to contain some proteins that
are released from cells that lyse during growth and transfer.
This experiment analyzes reactivity of an antibody directed
against secreted HCP in both a secreted protein- and cell
lysate-derived samples (see Figs. 13 and 14).
19. Handle IPG strips from the ends using forceps.
20. The sample is applied to the entire length of the immobilized
pH gradient by rehydration into a dry IPG strip. As voltage is
applied, each peptide in the sample focuses to a narrow zone in
the pH gradient corresponding to its pI.
21. Mineral oil prevents evaporation of the sample solution.
22. Additional details of the sample application and IEF procedure
may be found in the user manual for the PROTEAN i12 IEF
System.
23. It will take at least 10 min for the agarose sealing solution to
melt. It is best to start this prior to equilibrating the IPG strips
for the second dimension.
24. Although size standards are not strictly required for this application, they may be applied to the gel at this stage if desired.
412
Fig. 13 CHO cell lysate probed with anti-CHO HCP: Stained gels, stained blot, and Western blot. CHO cell lysate
was separated in duplicate by 2-D electrophoresis. One gel was stained with Oriole Fluorescent gel stain (a).
The second gel was transferred to PVDF membrane and stained with Oriole following transfer (b). The PVDF
membrane was stained with SYPRO Ruby Blot Stain to visualize total protein (c). Following staining, the blot
was immunodetected with anti-CHO HCP and visualized by chemiluminescence (d)
413
Fig. 14 CHO cell secreted protein probed with anti-CHO HCP: Stained gels, stained blot and Western blot. CHO
cell secreted protein was separated in duplicate by 2-D electrophoresis. One gel was stained with Oriole
Fluorescent gel stain (a). The second gel was transferred to PVDF membrane and stained with Oriole following
transfer (b). The PVDF membrane was stained with SYPRO Ruby Blot Stain to visualize total protein (c).
Following staining, the blot was immunodetected with anti-CHO HCP and visualized by chemiluminescence (d)
414
Chapter 29
Free Flow Electrophoresis for Separation of Native
Membrane Protein Complexes
Lutz Andreas Eichacker, Gerhard Weber, Ute Sukop-Kppel,
and Robert Wildgruber
Abstract
This chapter describes the technology of free flow electrophoresis (FFE) and protocols to separate membrane
protein complexes for proteome analysis. FFE is a highly versatile technology applied in the field of protein
analysis. It is superior to native PAGE due to its fast continuous processing of sample at high resolution.
Additionally, the dynamic separation range from ions, peptides, to proteins, protein complexes, up to
organelles, and whole cells makes it the method of choice in the analysis of proteins. FFE is carried out in an
aqueous medium without inducing any solid matrix, such as acrylamide, so that it simplifies the analysis of
protein complexes for the downstream analysis. Here, we describe the novel zone electrophoresis interval
method (IZE-FFE) for separation of protein complexes from the thylakoid membrane of Arabidopsis thaliana by charge only. Protein complexes isolated by IZE FFE were characterized according to molecular weight
by Blue Native PAGE and were proteins stained with coomassie.
Key words Free flow electrophoresis, Thylakoid membrane, Protein complexes, Blue Native PAGE
Abbreviations
BN
FFE
HPMC
iZE
PAGE
ZE
Introduction
The molecular mass ranges of thylakoid membrane protein
complexes that regulate biogenetic processes cover several orders
of magnitude. Isolation and characterization of protein complexes
and analysis by single particle electrotomography or structure
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_29, Springer Science+Business Media New York 2015
415
416
417
Fig. 1 FFE-separated protein complexes visualized after native BN-PAGE. Thylakoid membranes corresponding
to 300 g Chl were solubilized in 300 L detergent mixture 1 and 2. Supernatant 2 was applied to Free Flow
Electrophoresis. The FFE fractions containing the solubilized protein complexes were then concentrated and
subsequently loaded onto native BN-PAGE. Chl-protein complexes and standard proteins (kDa) were stained
with colloidal Coomassie G250 and detected by white light scanning
Materials
2.1 Preparation
of Thylakoid
Membranes (Rosette
Leaves
from Arabidopsis
thaliana)
2.2 Solubilization
Buffers
for the Membranes
418
2.3
FFE Media
2.4
BlueNative-PAGE
2.5 Analysis
of Thylakoid
Membrane Complexes
419
Methods
The FFE system performs electrophoretic separations in patented
and well-defined separation solutions without the use of a solid
(e.g. polyacrylamide) matrix. The gel-free basis enables the highresolution separation of charged or chargeable entities like cell
organelles, protein complexes and proteins, or peptides on a fast,
preparative, and continuous basis. Fluid-phase separation is operated in the three modes zone electrophoresis (ZE), including the
novel iZE method, isoelectric focusing (IEF), and isotachophoresis.
The system enriches low-abundant proteins with high reproducibility. The sample is applied using a peristaltic pump and is induced
into a separation chamber consisting of two parallel plates. Under
laminar flow, the sample is transported within a thin (0.20.4 mm)
film of aqueous medium formed between the two plates. The plates
are bordered by two electrodes that generate a high-voltage electric
field perpendicular to the laminar flow. Charged particles like ions,
peptides, proteins, organelles, membrane fragments, or whole cells
that are deflected in this electric field are separated and seamlessly
fractionated into microtiter plates.
The iZE method described in this chapter delivers excellent
resolution for the separation of protein complexes and membrane
proteins. Compared to IEF separations, media require no addition
of polymers like Hydroxy-ethyl-propyl-cellulose (HPMC) to suppress electroendosmotic flow. This enables an easy concentration
of the separated samples by ultra-filtration and ensures highest
compatibility for any downstream technology like, e.g. native and
SDS-PAGE, HPLC, MS, and ELISA.
The protocol describes how protein assemblies from the thylakoid membrane of Arabidopsis thaliana can be prepared for iZEFFE separation. Separation by iZE-FFE is based on the charge of
the protein assemblies. The resolution of the iZE-FFE protocol is
visualized using blue native PAGE as a native PAGE approach. In
general, blue-native PAGE is intended to separate protein complexes in a state that reflects the physiologically relevant functional
state of the protein assembly [4]. It is therefore advisable to minimize protein complex degradation through sample preparation on
ice and fast electrophoresis at 4 C. Solubilization of thylakoid
membranes resulted in the formation of membrane assemblies of
chlorophyll-binding thylakoid membrane protein complexes that
were charge separated by iZE-FFE within 6 min. The composition
of the FFE fractions was analyzed in a BN-PAGE approach. Here,
one to several protein bands have been determined per FFE fraction. Unique protein compositions were identified per every
second FFE fraction that was loaded for BN-PAGE separation.
Especially, five distinctive maxima were determined after the FFE
fractionation (Fig. 1, fractions 35, 37, 49, 53 and 59). BN-PAGE
420
421
by
centrifugation
at
by
centrifugation
at
FFE Method
422
1. Place the sample to the ice cooled sample holder on the FFE
system (see Notes 37).
2. Clean interior surface of chamber: move separation chamber to
an upright position and open the pairs of chamber clamps
simultaneously. Use the following sequence for cleaning
(H2Odist 18 n/20 S):water isopropanolpetroletherisopropanolwater (see Note 8).
3. Apply the spacer (0.2 mm) without covering the inlets, apply
the electrode membrane (soaked in Glycerol/Isopropanol,
mix 1:1) and the filter paper (0.3 mm), soaked in distilled
water onto the membrane.
4. Close separation chamber: lift the Plexiglas side of the chamber
to meet the mirrored side, first fasten central and adjacent
clamps loosely (release chamber clamps by two turns each).
Then fasten central and adjacent clamps tight, close top and
bottom clamps, place fractionation plate on top of fractionation housing, finally make sure that all inlets of media and
counterflow are free and are not covered by the spacer.
5. Fill separation chamber with degassed dist. water: open (lift)
valves at the top of the chamber to degas the chamber, fasten
wedge clamps (I1I9) of the media pump and place tubes in
reservoir filled with H2Odist, switch on medium pump on
control board. When the chamber is totally flushed and free of
air add counter flow by closing wedge clamps on pump.
6. Set up the Electrode Buffer, Stabilization Buffers and counter
flow buffer and separation buffers.
7. Connect anode (+) and cathode () electrolytes, connect plastic cover and start electrode pump.
8. Adjust the media flow rate to 120 mL/h and start the media
flow, wait for 15 min to flush the buffers in.
9. Adjust the voltage to 1,600 V, set the current to 150 mA.
10. Set the sample flow rate to 4,000 L/h.
11. Set the media pump flow rate to 16.4 digits.
12. Turn on media pump.
13. Turn on sample pump and flush the sample in for 70 s, then
switch the sample pump to backwards direction for 2 s and
turn off the sample pump.
14. Wait 20 additional seconds, and then turn on the high
voltage.
15. Wait 30 s, and then adjust the media pump to 3.6 units.
16. Wait 4 min, then turn the high voltage off and elute the
separated samples with a media pump speed of 28.8 units.
423
17. After 100 s the sample can be collected with a MTP or deep
well plate, therefore place the MTP in the fraction collection
device and collect for 120 s.
18. If more separated sample is needed, repeat the steps 817.
3.4 Concentration
of FFE Fractions
3.5 Electrophoresis
of Protein Complexes
from FFE Fractions
on BN PAGE
Notes
1. Digitonin is dissolved by heating the solution to 100 C. After
heating, Digitonin can immediately be mixed with other detergents. The rest of the detergent mixture can be stored at
20 C. Always reheat solutions before usage.
424
425
References
1. Schgger H, Von Jagow G (1991) Blue native
electrophoresis for isolation of membrane protein complexes in enzymatically active form.
Anal Biochem 199:223231
2. Reisinger V, Eichacker LA (2012) Native DIGE
of fluorescent plant protein complexes. In:
Cramer R, Westermeier R (eds) Difference gel
electrophoresis (DIGE). Humana Press,
New York, pp 343353
3. Krause F (2006) Detection and analysis of
protein-protein interactions in organellar and
prokaryotic proteomes by native gel electrophoresis: (membrane) protein complexes and supercomplexes. Electrophoresis 27:27592781
4. Wittig I, Karas M, Schgger H (2007) High resolution clear native electrophoresis for in-gel functional assays and fluorescence studies of membrane
protein complexes. Mol Cell Proteomics 6:
12151225
Chapter 30
Three-Dimensional Electrophoresis for Quantitative
Profiling of Complex Proteomes
Sergio Mauro, Bertrand Colignon, Marc Dieu, Edouard Delaive,
and Martine Raes
Abstract
Quantitative 2D-gel-dependent proteomics became feasible with 2D fluorescence difference gel electrophoresis
(2D-DIGE), and this technique has gained wide acceptance because it has eliminated the gel to gel variations
and greatly facilitated the quantitative comparisons across gels for many different experimental conditions.
However, the co-migration of several proteins in the same spot is still a major limitation which detracts from
the accuracy of comparative quantification and prevents unambiguous post-translational modifications
(PTMs) detection.
A protocol based on traditional polyacrylamide gel IEF sample fractionation, and followed by two
consecutive SDS-PAGE electrophoreses alleviates co-migration limitations. The use of two different buffer
systems for SDS-PAGE is central to the proposed approach.
Key words 2D-DIGE, 3D-DIGE, Western blot, Co-migration, Complex proteomes
Introduction
The early work of Tiselius demonstrated the value of electrophoretic
fractionation as an analytical technique for separating proteins [1].
Since this pioneering work, the technique has seen many refinements resulting in increased resolving power.
The introduction of inert support materials such as paper,
agarose, and acrylamide greatly improved the resolving power by
eliminating the continuous mixing of the sample components associated with the current-driven convection.
Among the various separation media, polyacrylamide gel has
gained the most widespread use. In addition to preventing thermal
convection, the polyacrylamide matrix acts as a molecular size
selective sieve increasing the resolving power. Therefore, during
polyacrylamide gel electrophoresis (PAGE) protein separation is
based on a combination of charge, shape, and size.
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_30, Springer Science+Business Media New York 2015
427
428
3D Differential Electrophoresis
429
Materials
Unless indicated otherwise all solutions are prepared with Milli-Q
water (resistivity, 18.2 M cm at 25 C).
2.1 Sample
Preparation
and Isoelectric
Focusing
430
1. Typhoon 9400.
2. DeCyder 2-D Differential Analysis Software v6.5.
3D Differential Electrophoresis
431
Methods
3.1 Sample
Preparation
and Isoelectric
Focusing
3.1.1 Reconstitution
of CyDye Fluor
for Saturation DIGE-Based
Analysis (See Note 1)
3. For analytical 2D-DIGE gels, add 50 L DMF so as to reconstitute a 2 mM working dye solution (Cy3 and Cy5 are available).
2. For preparative 2D-DIGE gels, add 20 L DMF so as to reconstitute a 20 mM working dye solution (only Cy3 is available).
4. Vortex for 30 s.
5. Centrifuge for 30 s at 12,000 g.
6. If not used directly the solutions may be stored at 20 C up
to 3 months.
432
Protein Labeling
for Preparative Gels
IPG-Strip Rehydration
3D Differential Electrophoresis
433
3.2.1 Equilibration
of IPG-Strips
3.2.2 2D Difference
Gel Electrophoresis
434
6. Wet the upper buffer chamber seals with some cathode buffer
and slide the chamber into place.
7. Fill the upper chamber with cathode buffer (approximately
1.2 L).
8. Fill the lower chamber with anode buffer up to the fill line.
9. Run the gels initially at constant 80 V for 1 h and then at
12 mA/gel overnight (1.5 W/gel) until the bromophenol
blue front reaches the end of the gel.
3.2.3 Two- and ThreeDimensional Gel
Electrophoresis Image
Capture and Analysis
3.2.4 Quantitative
3D-DIGE for Comparative
Proteomics Using
Analytical 2D-DIGE Gels
Fig. 1 The work flow for 3D-DIGE analysis for quantitative profiling of complex proteomes. The 3D-DIGE protocol
begins with the 2D-DIGE analytical gel analysis which generates the list of informative spots. These spots are
excised from a preparative 2D-DIGE gel and further electrophoresed in a different gel system. The resolved individual
components are analyzed in downstream applications such as mass spectrometry and Western blot analyses
436
Fig. 2 Analytical 3D-DIGE of total protein extracts of control and heat-treated leaves of Solanum tuberosum.
(a). Resolution of Solanum tuberosum leaves protein extracts by 2D-DIGE. Extracts from internal standard and
heat-treated leaves (42 C, 1 h) samples were differentially labeled with the Cy5 and Cy3 and separated by
two-dimensional electrophoresis on 24 cm (pH 47) IPG strips and 12.5 % polyacrylamide gels. Cy3-labeled
heat-treated sample and Cy5-labeled internal standard are shown. The arrow indicates the position of the spot
displayed in b. (b). Resolution of a spot selected from the picking list. The spots of interest (see arrow in a) from
control (C) and heat-treated leaves (T) were excised from two different analytical DIGE gels and reelectrophoresed in a NuPage Tris acetate 7 % gel. Each spot resolved into two, S1 and S2. A 3D view of the
fluorescence signal is shown. (c). Manual quantification of the changes in S1 and S2 abundance. The NuPage
Tris acetate 7 % gel was scanned using the Typhoon 9400 and the fluorescence intensity volumes were estimated with the DeCyder Image Analysis Software. After normalization, it appeared that S2 accounted for the
observed signal reduction. To confirm fluorescent signal preservation, we summed the volumes of all 3D spots
for each condition and compared them to the corresponding spot volumes on 2D gel. These results show the
relative conservation of total fluorescence and global variation (average volume ratio: 1.87 for 2D gel and
1.67 for 3D gel). The minor difference can be explained by a loss of material due to the fact that: (a) only the
cylinder surrounding the top of the peak of 2D spot was picked; (b) after the 3D run, quantification does not
include small smears into the spot volume estimation. An Anova test with p-value < 0.05 was realized with SAS
8.0 (SAS Institute Inc., Cary, North Carolina, USA). Error bars represent SD values from three replicates
3D Differential Electrophoresis
437
3.4 Western
Blot Analysis
438
Fig. 3 3D-DIGE-based detection and identification of low-abundance proteins. 2D Western blot analysis of
control leaves protein extracts was performed with a polyclonal antibody against SUMO1 protein (Primary
antibody, dilution 1/800; secondary antibody, dilution 1/2,000). Over 70 SUMO-protein conjugate spots were
detected. One of these spots (a) was excised and re-electrophoresed in a Bis-Tris Glycine 412 % gel.
It resolved into three 3D-components a, b and c (b). A Western blot analysis using the anti-SUMO1 polyclonal
antibody identified spot c as a SUMO-protein conjugate (c)
8. Use the 2D-DIGE gel and PVDF images to create a picking list.
9. Run a new preparative 2D-DIGE gel and excise the spots according to the picking list.
10. Fractionate the protein content of these gel fragments as in
Subheading 3.2.4.
11. Scan the gel to visualize the individual proteic components.
12. Perform a new Western blot (steps 36) to detect SUMO
conjugate.
13. For the MS identification of the SUMO targets duplicate the
gel (go to step 9).
Notes
1. Our protocol requires in-gel protein detection without fixation. Although several methods that fulfill that requirement
are available [18, 19], we described a DIGE-based procedure
which combines high-sensitivity protein detection and differential protein expression analysis.
Two procedures for protein labeling are available for DIGE
application. In the minimal labeling procedure two protein
samples and the internal standard are separately labeled with
the spectrally resolvable cyanine dyes Cy2, Cy3 or Cy5 and
mixed before electrophoretic fractionation. The minimal labeling fluorophors contain a N-hydroxysuccinimidyl ester reactive
3D Differential Electrophoresis
439
440
17.
18.
19.
20.
Chapter 31
A Bead-Based Multiplex Sandwich Immunoassay
to Assess the Abundance and Posttranslational
Modification State of -Catenin
Nicola Groll, Cornelia Sommersdorf, Thomas O. Joos, and Oliver Poetz
Abstract
A system-wide analysis of cell signaling involves detecting and quantifying a range of proteins and their
posttranslational modification states in the same cellular sample. We propose a protocol for a miniaturized,
bead-based array and describe its efficiency in characterizing the different forms and functions of -catenin.
The protocol provides detailed instructions for cell culture and bead array assays that enable insights into
complex networks at the systems level.
Key words Bead-based array, Sandwich immunoassay, co-immunoprecipitations, Proteinprotein
interaction assay, -catenin, Wnt-pathway
Introduction
Protein arrays have become powerful tools to investigate the status
of signaling pathways in cells and tissues. Bead-based multiplexed
assays can be performed with hundreds of samples, thereby enabling
time-resolved pathway activities of stimulated or perturbed cells.
The data can be used to infer the structure of underlying signaling
networks. Protein array technology is well-suited for such investigations because it can simultaneously measure many different proteins
as well as entails minimal amounts of material [14].
Over the past decade, bead-based arrays have become
well-established in cell signaling research (Fig. 1). This technology
platform has been used to analyze signaling networks in a timeresolved manner. The polystyrene beads have an internal fluorescent color code that facilitates the measurement of up to 500
features. The surface coating includes paramagnetic material and
carboxyl groups that allow easy conjugation of proteins, peptides,
oligonucleotides, and carbohydrates. For a panel of sandwich
immunoassays, the beads are usually coated with different capture
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_31, Springer Science+Business Media New York 2015
441
442
Assay type
Protein-protein interaction
assay
Sandwich immunoassay
Ba
it
Analyte
Detector antibody
Detection system
anti-non pSer33Ser37Thr41
anti-pSer33Ser37Thr41
anti-pSer45
anti-pSer552
anti-pSer675
anti-pan -catenin
phosphorylated and total
-catenin
PE
Ecad
Bead
7-9
Bead
1-6
Capture antibody/
bait protein
PE
PE
Assay set-up
Coip
GST-TCF4
GST-ICAT
GST-ECT
transcriptionally active
free -catenin
Bead
10
ms E-cadherin
E-cadherin-bound
-catenin
anti-mouse-PE conjugate
Fig. 1 The -catenin bead array panel employs antibodies and known interaction partners of -catenin to
study the phosphorylation status and its complexation status within the same multiplex assay system.
Differentially phosphorylated -catenin is measured with bead array-based sandwich immunoassays. By utilizing different phosphorylation state and sequence-specific antibodies and a single pan-specific antibody
serving as the capture molecule, it is possible to reliably discriminate and detect various phosphorylated
-catenin forms. The recombinantly expressed binding partners of -catenin, ECT (E-cadherin cytosolic tail),
ICAT (inhibitor of -catenin), and TCF4 (T-cell factor 4) are used as bait proteins in a proteinprotein interaction
assay for binding and detecting transcriptionally active -catenin. The miniaturized co-immunoprecipitation
(Coip) allows -catenin in complex with E-cadherin to be detected by fishing E-cadherin. In all assay set-ups,
the same antibody specific for the -catenin C-terminus is used as a detector
443
changes in concentration, phosphorylation and complexation associated with other factors such as cadherins. After activation of the
Wnt receptor, a free pool of -catenin accumulates in the cytoplasm, translocates into the nucleus and then functions as a transcriptional co-activator [6, 7].
The Wnt/-catenin signaling pathway plays an important role
in the regeneration of adult tissues and in embryonic development
[8]. Being a proto-oncogene, -catenin is a key factor in carcinogenesis. Mutations in the -catenin gene or in other genes involved
in -catenin regulation, such as APC or Axin, lead to hyperactivation of the Wnt signaling pathway and are implicated in the genesis
of common cancers, such as colorectal cancer and hepatocellular
carcinoma [911]. Wnt/-catenin signaling also impacts the epithelial mesenchymal transition (EMT), which is reportedly involved
in the invasion of tissue and formation of metastasis [12].
Here, we propose a protocol using three different assay setupssandwich immunoassays, proteinprotein interaction and
co-immunoprecipitationsin one panel to assess and quantify
-catenin, its phosphorylation and functional mode in a single
sample. The sandwich immunoassay allows the measurement of
total -catenin as well as the degree of phosphorylation at six different sites. For this purpose, one pan-specific -catenin antibody,
one de-phosphorylation and four phosphorylation-specific antibodies served as capture molecules. Thus discrimination and detection of non-phosphorylated, differentially phosphorylated and
total -catenin is carried out.
In miniaturized GST-pull down (GST pull down) assays,
cellular interaction partners of -catenin are used as baits to detect
-catenin in its free non-complexed form. The -catenin which
forms a complex with these baits is transcriptionally active, as previously described [4].
A capture antibody toward E-cadherin allows co-immunoprecipitation of the E-cadherin/-catenin complex [13]. This membrane complex reflects the integrity of the cell. Finally, all the analytes
are detected by an antibody which binds to the C-terminal region of
-catenin and a phycoerythrin-conjugated secondary antibody. This
enables a readout of the various assays and allows the different pools
and states of -catenin to be monitored at the same time.
Materials
444
4. Dimethylsulfoxide (DMSO).
5. Compound of interest in DMSO, e.g. proteasome inhibitor
MG132 (Calbiochem).
6. Phosphate-buffered saline (PBS).
7. Lysis buffer: 150 mM NaCl, 1 % w/v Triton X-100,
Phosphatase Inhibitor Cocktail II (Sigma Aldrich), Phosphatase
Inhibitor Cocktail III (Sigma Aldrich), Complete Protease
Inhibitor Cocktail (Roche Applied Science), 50 mM TrisHCl,
pH 7.4, 2,5 U/ml Benzonase Nuclease (Novagen).
8. BCA (bicinchoninic acid) protein assay reagent.
2.2 Generation
of Bead Array
2.2.1 Covalent Coupling
of Capture Antibodies
to Microspheres
2.2.2 Non-covalent
Coupling of Capture
Antibodies and Bait
Proteins to Microspheres
445
1. KingFisher
Flex
(ThermoScientific).
Magnetic
Particle
Processors
Methods
446
447
448
50 l
Time-out
80 s
Total beads
10,000
449
Before carrying out the assay procedure, the protein extracts need
to be normalized to protein concentration. Due to the cell type
and the quantity of the target protein (in this case: -catenin)
being proportional to total protein, it is advisable to determine
the optimal amounts in separate, preliminary experiments. We
suggest that a dilution series of cell culture lysates is made to identify a suitable range of lysate amounts for detecting the target
proteins. We recommend using 1025 g of total protein, as this
is usually appropriate for analyses (see Note 8).
Here we describe how the assay can be performed by using a
magnetic bead transfer system ([14] King Fisher, Thermo Fisher
Scientific) which enables semi-automated washing and incubation
steps. Of course, it is also possible to use a magnetic plate separator
for manual washing and incubation in a 96-well plate.
1. Prepare a bead-master mix by diluting each population of
coated beads in assay buffer in order to ensure that there are
2,000 beads for each population in 20 l of fully mixed bead
solution. Vortex the diluted beads to mix them.
2. Choose an appropriate protein amount of each sample, and
adjust each sample to the same protein concentration with
assay buffer at a volume of 40 l. Keep the samples on ice.
3. Combine 20 l bead mix with 40 l of diluted cell sample in
one well of the 96-well microplate (half-area, flat-bottomed
microplate, with a non-binding surface, is subsequently
referred to as the assay plate).
4. Use 40 l assay buffer as a reagent control sample.
5. Seal the plate with a microplate sealing film.
6. Incubate the samples containing the beads overnight in a
96-well plate shaker at 4 C and 750 rpm.
7. Prepare the secondary antibody solution at least 2 h before
assay processing to minimize the background signal. Goat and
donkey sera can be used to minimize the cross-reactivity of the
polyclonal antibodies. Dilute the PE-conjugated donkey antimouse IgG in assay buffer to a final concentration of 2.5 g/ml.
450
7,50015,000
Sample volume
100 l
100
Table 1
Program of the semiautomatic, magnetic bead transfer system (King Fisher, Thermo Fisher Scientific)
Step
Plate
Assay step
Duration
Sample plate
Bead collection
10 s
Washing plate 1
Washing
1 min
Washing plate 2
Washing
1 min
60 min
Washing plate 3
Washing
1 min
Washing plate 4
Washing
1 min
45 min
Washing plate 2
Washing
1 min
Washing plate 4
Washing
1 min
10
Elution plate
Bead elution
20 s
1200
anti-total -catenin
1200
451
anti-nonpSer33/37/Thr41 -catenin
4500
MFI [AU]
MFI [AU]
600
600
0
0.1
10
3000
1500
300
300
PE
MFI [AU]
900
900
0.1
Time [h]
1
Time [h]
10
anti-pSer552 b-catenin
Bead
1-6
2000
0.1
1
Time [h]
anti-pSer675 -catenin
10
anti-pSer45
2000
450
1000
8000
300
150
500
MFI [AU]
1500
MFI [AU]
MFI [AU]
1500
0
0.1
1
Time [h]
10
500
0
0.1
1
Time [h]
0.1
10
60 GST-TCF4
GST-ICAT
1000
40
1
Time [h]
10
1
Time [h]
10
GST-ECT
N
PE
4000
40
MFI [AU]
Bead
7-9
MFI [AU]
30
6000
MFI [AU]
Ba
it
20
20
10
2000
0.1
1
time [h]
10
anti-ms E-cadherin
MFI [AU]
1
time [h]
10
0.1
20
PE
Bead
10
0
0.1
DMSO
MG132
Ecad
10
0.1
1
Time [h]
10
Fig. 2 Snapshots of different forms of -catenin monitored by a bead array assay. Hep 70.4 cells were treated
with the proteasome inhibitor MG132 and DMSO as a control, in intervals of 5, 30, 60, 180, 360, 720, and
1,440 min. The experiment was performed in triplicate. From each sample, a protein extract of 20 g was
analyzed by the -catenin bead array assay panel
Notes
1. Growth rate depends considerably on cell line and incubation
parameters.
2. The purpose of the assay described here is to examine a
molecular snapshot of a single protein. The conditions were
optimized, in particular for extracting the native -catenin and
its diverse forms as well as the intact E-cadherin complexes.
Generally, the extraction conditions, for instance, those including additives of the lysis buffer, can influence the native protein. It is thus necessary to optimize the appropriate detergent,
salt concentration and inhibitors if other proteins or pathways
shall be analyzed.
452
3.
4.
5.
6.
7.
8.
453
Chapter 32
Identification of SUMO E3 Ligase-Specific Substrates
Using the HuProt Human Proteome Microarray
Eric Cox, Ijeoma Uzoma, Catherine Guzzo, Jun Seop Jeong,
Michael Matunis, Seth Blackshaw, and Heng Zhu
Abstract
The functional protein microarray is a powerful and versatile systems biology and proteomics tool that
allows the rapid activity profiling of thousands of proteins in parallel. We have recently developed a human
proteome array, the HuProt array, which includes ~80 % of all the full-length proteins of the human proteome. In one recent application of the HuProt array, we identified numerous SUMO E3 ligase-dependent
SUMOylation substrates. For many SUMO E3 ligases, only a small number of substrates have been identified and the target specificities of these ligases therefore remain poorly defined. In this protocol, we outline
a method we developed using the HuProt array to screen the human proteome to identify novel SUMO
E3 ligase substrates recognized by specific E3 ligases.
Key words Microarray, SUMO, Proteomics, E3 ligases, Posttranslational modifications
Introduction
The functional protein microarray is a powerful and versatile
systems biology and proteomics tool that allows the rapid activity
profiling of thousands of proteins in parallel. Applications of functional protein microarrays range from the identification of proteinbinding properties, to surveying targets of posttranslational
modifications, to uncovering novel enzymatic activities. Since the
development of the yeast proteome microarray over 10 years ago
[1], more recent work has seen the development of complete and
near-complete proteome arrays representing viruses, bacteria, and
plants [24]. However, most existing human protein microarrays
are comprised of only a minority of the human proteome [59].
We have recently developed a human proteome microarray, the
HuProt array, which includes nearly 20,000 full-length human
proteins [10]. The proteins used to generate this microarray
were expressed in yeast and purified under native conditions.
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_32, Springer Science+Business Media New York 2015
455
456
2
2.1
457
Materials
Equipment
2.2 Purification
of the SUMO
E1 Enzyme
2.3 Purification
of the SUMO
E2 Enzyme
2.4 Common
Reagents
for Expression
and Purification
of SUMO Protein
and SUMOylation
Enzymes
458
2.6 On-Chip
SUMOylation Assay
Methods
3.1 Purification
of SUMO Protein
3.2 Purification
of SUMO E1 Enzyme
459
3.3 Purification
of SUMO E2 Enzyme
460
8. During centrifugation prepare glutathione sepharose by washing 2 ml 50 % glutathione sepharose with 1 PBS in 50 ml
tube. Repeat twice.
9. Bind protein by mixing supernatant with glutathione sepharose, rotate at 4 C for 1 h or longer.
10. Wash sepharose by spinning down sepharose, discard supernatant, resuspend agarose in 1 ml of E2 wash buffer. Transfer
sepharose to column; wash with 12 ml of E2 wash buffer.
11. Cleave UBC9 from GST tag by transferring beads bound with
GST-precission protease and GST-UBC9 to the same column.
Parafilm column to prevent leakage. Incubate with shaking at
4 C overnight.
12. Allow sepharose to settle. Remove supernatant containing
purified untagged UBC9. Apply 4 ml of E2 wash buffer and
repeat.
13. Dialyze overnight against 2 l enzyme dialysis buffer.
14. Concentrate protein using a micron centrifugal filter (10 kDa
MWCO) by spinning 20 min at 500 g. Aliquot and freeze
with liquid nitrogen and store at 80 C.
3.4 SUMO Antibody
Labeling
3.5 On-Chip
SUMOylation Assay
461
Notes
1. The humidity chamber is made by placing a folded paper towel
in the bottom of an empty pipette tip box (we usually use a
USA scientific stacked rack made for 200 l tips, e.g., #11111206), adding 1 in. of ddH2O, and replacing the tip holder on
462
the box. The arrays will sit on the tip holder and be covered
with the lid to maintain a humid environment.
2. SUMOylation of many proteins occurs in the absence of E3
ligases at relatively high E1 and E2 concentrations; therefore
E1 and E2 concentrations are usually used in the low end of
this range when using an E3 ligase. However, it is advisable to
establish the best SUMOylation conditions using a conventional tube-based assay, particularly E1 and E2 concentrations,
with the SUMO E3 ligase of interest and a known substrate.
The best result will allow clear visualization of enhanced
SUMOylation in the presence of the E3 ligase, before proceeding with the assay on-chip.
3. To minimize evaporation and potential photobleaching of any
fluorescently labeled protein beacons included on the protein
microarray, it is advised to wrap four-well plate with a layer of saran
wrap, then a layer of aluminum foil, before overnight incubation.
4. It is critical to strike the right balance of removing excess liquid
from the array while not over-drying at this step. The goal is to
remove enough liquid so as not to dilute the reaction mix,
while also not allowing the top (protein surface) to not become
completely dry. The best results can be obtained by being
ready to add reaction mix immediately, i.e., reaction mix tube
open, pipet set to correct volume and already loaded with a
pipet tip, after placing the chip on the surface of the tip holder.
5. Ensure that bubbles are pushed out of the reaction mix while
adding coverslip to the top surface of the array. One reliable
strategy is to first rest one end of the coverslip on the edge of
the array, then gradually lower the other end of the coverslip to
push out the bubbles.
6. 1 % SDS is used in order to stringently wash the array and remove
any non-covalently bound SUMO from the arrayed proteins.
7. Removing the coverslip is best achieved by gently sliding the
coverslip straight along the array with only the raised edges of
the coverslip contacting the outside edges of the chip, where
no proteins are printed. Allowing the raised edges of the coverslip to move across the array (contacting the region where
proteins are printed) can scratch the surface of the chip, causing high background signal in the scratched region, resulting
in lost information about proteins in the path of the scratch.
8. A wash with high purity water is essential to remove residual
salts that can result in high background when scanning.
9. To get the most meaningful information from your protein
microarray experiment, it is important to scan the chip with
the appropriate gain and power settings. A useful starting point
using GenePix software would be PMT Gain = 600,
Power = 100 %. Usually the gain value can then be raised or
463
Chapter 33
Amyloid-Binding Proteins: Affinity-Based Separation,
Proteomic Identification, and Optical Biosensor Validation
Alexei Medvedev, Olga Buneeva, Arthur Kopylov, Oksana Gnedenko,
Alexis Ivanov, Victor Zgoda, and Alexander A. Makarov
Abstract
The amyloid-beta peptide is considered as a key player in the development and progression of Alzheimers
disease (AD). Although good evidence exists that amyloid-beta accumulates inside cells, intracellular brain
amyloid-beta-binding proteins remain poorly characterized. Here we describe a protocol for affinity-based
profiling of amyloid-beta-binding proteins of rat brain, their proteomic identification and validation by a
surface plasmon resonance (SPR)-based analysis. It includes: (a) SPR-based selection of immobilization
conditions for beta-amyloid coupling and choice of appropriate resin for preparation of an affinity sorbent;
(b) immobilization of beta-amyloid on the selected resin; (c) preparation of biological samples (rat brain
homogenate) for affinity-based separation; (d) isolation of rat brain proteins by affinity chromatography
using amyloid-beta as the affinity ligand; (e) sample preparation for proteomic analysis; (f) proteomic
analysis and protein identification; (g) SPR-based validation of the interaction of identified proteins with
immobilized amyloid-beta.
Key words Amyloid-beta, Amyloid-beta-binding proteins, Affinity-based proteomic profiling, Rat brain
Introduction
The amyloid-beta peptide 142 formed during proteolytic processing of the amyloid precursor protein (APP) is considered as a key
player in the development or progression of Alzheimers disease
(AD) and other pathologies associated with formation of protein
aggregates in the central nervous system [16]. Although much
attention is paid to formation of extracellular amyloid plaques and
protein aggregates as well as to corresponding mechanisms of their
toxicity, good evidence exists that intracellular amyloid-beta can
accumulate intraneuronally and contribute to disease progression
[4, 713]. This suggests importance of amyloid-beta interaction
with particular intracellular targets. Indeed, interaction of amyloid-beta with intracellular catalase caused inactivation of this
enzyme and accumulation of hydrogen peroxide inside cells [14].
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_33, Springer Science+Business Media New York 2015
465
466
467
Materials
2.1 SPR-Based
Biosensor
carbodiimide
468
2.2 Components
for Affinity-Based
Separation
2.3 Proteomic
Analysis
2.4
Instrumentation
469
Methods
3.1 SPR-Based
Selection
of Immobilization
Conditions for BetaAmyloid Coupling
Fig. 2 SPR-based selection of immobilization conditions for amyloid-beta coupling and validation of amyloidbeta interaction with glyceraldehyde-3-phosphate dehydrogenase. (a) Optimization of pH for buffer used for
amyloid-beta immobilization: 1amyloid-beta peptide (100 g/mL) in immobilization buffer B, 2HBS-EP
471
3.2 Immobilization
of Amyloid-Beta
Peptide
3.2.1 Binding
Measurements
3.3 Immobilization
of Amyloid Beta
Protein Fragment 142
on Affi-Gel 10
Fig. 2 (continued) buffer, pH 7.4, 3amyloid-beta peptide (100 g/mL) in immobilization buffer A. At pH 4.0
(immobilization buffer A) amyloid-beta demonstrates much better interaction with the surface of the optical
biosensor chip. (b) Immobilization of amyloid-beta peptide (100 g/mL) in 10 mM sodium acetate, pH 4.0
onto the surface of the optical biosensor chip. Arrows indicate injections: 1EDC/NHS; 2HBS-EP buffer;
3100 g/mL amyloid-beta in immobilization buffer A; 4blocking solution. (c) Interaction of GAPDH
(glyceraldehyde-3-phosphate dehydrogenase) with immobilized amyloid-beta peptide. Results are corrected
versus control channel (without amyloid-beta peptide)
472
3.6 Sample
Preparation
for Proteomic Analysis
473
3.7 Liquid
Chromatography:
Mass Spectrometry
474
3.9 SPR-Based
Validation
of the Interaction
of Identified Proteins
with Amyloid- Beta
475
Further validation of the interaction between immobilized amyloidbeta and proteins identified by mass spectrometry can be obtained
using various methods. We recommend the SPR-based biosensor
analysis with purified proteins of interest (see Figs. 1 and 2).
Advantages of the SPR-based validation consist in the possibility of
repeated use of the optical biosensor cuvette with immobilized
amyloid-beta.
Notes
1. Recent results of isoelectric focusing studies indicate that low
molecular forms of monomeric amyloid-beta (142 and 140)
are characterized by a pI value of ca. 5, while at pI of 66.5
aggregates of high molecular weight are detected [17]. This
suggests that it is better to use more acidic solutions of amyloidbeta to prevent its aggregation.
2. Several studies have recognized glyceraldehyde-3-phosphate
dehydrogenase as the amyloid-binding protein (e.g., [18, 21]),
and therefore it can be used as a probe for interaction with
immobilized amyloid-beta.
3. Various intracellular highly abundant proteins (e.g., cytokeratins) may nonspecifically bind to a resin even without an immobilized affinity sorbent (see for example, ref. 22) and therefore
a control batch of the ligand-free sorbent has to be subjected
to all the same treatments but without amyloid-beta. The list
of identified amyloid-binding proteins will have to be corrected for possible presence of proteins nonspecifically bound
to the control sorbent.
4. Use sequencing grade trypsin only for mass spectrometry
experiments due to its high specificity and low contamination
of alpha-chymotrypsin. Also it is strongly recommended to use
modified trypsin (with acetylated lysins) to prolong activity of
enzyme and slow down its autolysis.
5. In the case of the use of non-sequencing grade trypsin its catalytic activity may be rather low; if so, it is recommended to add
up to 0.003 M of calcium chloride to increase ionic strength of
reaction mixture rather than sodium salts to minimize further
cationization during LC-MS analysis. However, it should be
noted that native trypsin may undergo autolysis, generating
pseudotrypsin, which exhibits a broadened substrate specificity
including chymotrypsin-like activity (see comments [23]). In
addition, trypsin may be contaminated with chymotrypsin,
and this fact has to be taken into consideration especially when
high enzyme/substrate ratios are used.
476
Acknowledgments
This work was supported by the Russian Science Foundation (grant
no. 14-24-00100) within the project The role of structural polymorphism of amyloid-beta in initiation of Alzheimers disease.
References
1. Schmechel DE, Saunders AM, Strittmatter WJ,
Crain BJ, Hulette CM, Joo SH, Pericak-Vance
MA, Goldgaber D, Roses AD (1993) Increased
amyloid beta-peptide deposition in cerebral
cortex as a consequence of apolipoprotein E
genotype in late-onset Alzheimer disease. Proc
Natl Acad Sci U S A 90:96499653
2. Saido TC, Iwatsubo T, Mann DM, Shimada H,
Ihara Y, Kawashima S (1995) Dominant and
differential deposition of distinct beta-amyloid
peptide species, A beta N3(pE), in senile
plaques. Neuron 14:457466
3. Hardy J, Selkoe DJ (2002) The amyloid
hypothesis of Alzheimers disease: progress and
problems on the road to therapeutics. Science
297:353356
4. LaFerla FM, Green KN, Oddo S (2007)
Intracellular amyloid- in Alzheimers disease.
Nat Rev Neurosci 8:499509
5. Masters CL, Selkoe DJ (2012) Biochemistry of
amyloid -protein and amyloid deposits in
Alzheimer disease. Cold Spring Harb Perspect
Med 2(6):a006262
6. Upadhaya A, Kosterin I, Kumar S, von Arnim
CA, Yamaguchi H, Fndrich M, Walter J, Thal
DR (2014) Biochemical stages of amyloid-
peptide aggregation and accumulation in the
human brain and their association with symptomatic
and
pathologically
preclinical
Alzheimers disease. Brain 137(pt 3):887903
7. Li QX, Maynard C, Cappai R, McLean CA,
Cherny RA, Lynch T, Culvenor JG, Trevaskis
J, Tanner JE, Bailey KA, Czech C, Bush AI,
Beyreuther K, Masters CL (1999) Intracellular
accumulation of detergent-soluble amyloidogenic A beta fragment of Alzheimers disease
precursor protein in the hippocampus of aged
transgenic mice. J Neurochem 72:24792487
8. Wirths O, Multhaup G, Czech C, Blanchard V,
Moussaoui S, Tremp G, Pradier L, Beyreuther K,
Bayer TA (2001) Intraneuronal A beta accumulation precedes plaque formation in betaamyloid precursor protein and presenilin-1
double-transgenic mice. Neurosci Lett 306:
116120
9. Butterfield DA, Sultana R, Poon HF (2006)
Redox proteomics: a new approach to investigate oxidative stress in Alzheimers disease. In:
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
477
Chapter 34
Proteomic Profiling by Nanomaterials-Based
Matrix-Assisted Laser Desorption/Ionization Mass
Spectrometry for High-Resolution Data and Novel Protein
Information Directly from Biological Samples
Suresh Kumar Kailasa and Hui-Fen Wu
Abstract
Qualitative and quantitative analyses of global proteome samples derived from biocomplex mixtures are
very important to understand the cellular functions in cell biology. Matrix-assisted laser desorption/
ionization mass spectrometry (MALDI-MS)-based proteomics has recently become one of the most
informative and attractive core technologies in proteomics. Particularly, nanomaterials-based MALDI
mass spectrometric methods are quickly becoming a critical miniaturized bioanalytical tool for detecting
and discerning proteins from biocomplex samples. These MALDI-developed strategies allow highthroughput identification of proteins from highly complex mixtures including accurate mass measurement of peptides derived from total proteome digests and peptides/proteins separations from various
samples. The nanomaterials-integrated MALDI-MS technologies in protein arrays hold much promise
for interrogating the diverse and immense proteome in cell biology. As a result, nanomaterials-assisted
MALDI-MS-based proteomic workflow, including sample preparation, information on the local molecular composition, relative abundance and spatial distribution of peptides and proteins and their analysis,
should make the technology more easily available to a broad community and turn it into a powerful
methodology for bioanalysts.
Key words Nanomaterials, MALDI-MS, Proteomics, Sample preparation, Protein digestion
Introduction
A cornerstone of proteomics is the ability to monitor and to
characterize proteins in a cell or any biological sample. Mass spectrometry has become the bioanalytical technique for proteome
analyses, which has the ability to probe the covalent structure of
proteins. Among the mass spectrometric-based proteomics,
MALDI-MS-based proteomics is mature and an attractive technology for characterization and quantification of proteins in a biological sample or system [1]. Although MALDI-MS has been known
as a powerful bioanalytical technique for protein analysis, it still
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6_34, Springer Science+Business Media New York 2015
479
480
The unique optical and physico-chemical properties of nanomaterials have been explored to be used as matrices for analysis of peptides and proteins with high mass accuracy and reduced background
signals because nanomaterials act as functional probes for generation of biomolecular mass spectra which produce better mass spectra than the conventional organic matrices [2]. The small nature
and unique physico-chemical properties of nanomaterials allow
them to enter/cross cellular membranes and their high surface area
to volume ratio provides a possibility to trap trace analytes with
high degree from complex samples. Furthermore, nanostructures
acted as good candidates to absorb incoming laser radiation and to
transfer the absorbed energy to the analytes, promoting their
desorption with high mass accuracy and with reduced background
noises, and eliminating the need for the organic matrix. Due to the
high dispersion ability of nanomaterials, nanomaterials-conjugated
analytes are efficiently and readily transferred between instruments,
which allow to detect ultratrace biomolecules with minimal sample
loss. A wide variety of nanomaterials including carbon-based nanostructures (carbon nanotubes, graphite, graphene), metallic (Au,
Ag, and Pt NPs), metal oxides (TiO2, ZnO, SiO2, Fe3O4, and
ZrO2), and semiconductor (ZnS, CdS, CdTe, and HgTe, etc.)
nanoparticles have been used as matrices for the analysis of biomolecules by MALDI-MS with good accuracy and high mass resolution [2, 57]. Due to their strong UV absorption cross-section of
481
1.3 Nanomaterials
as Extracting
and Preconcentrating
Probes
Recent trends in MALDI-MS-based proteomics have been intensively focused on the improvement in quality of mass spectra of
target proteins, introduction of new technological developments
with MALDI-MS and especially in miniaturization, simplification,
and automation of the whole analytical procedure, to extract and
concentrate trace proteins in microvolume samples [12, 13]. Prior
to the identification of target proteins/peptides by MALDI-MS,
sample preparation step is essentially required to isolate them from
the sample matrix, and to enrich them to a concentration level
detectable by MALDI-MS. To achieve the objective, substantial
482
483
484
2.2 Preparation
of Hydrophobic
Nanomaterials
1. AgNO3, Se powders and octadecylamine (ODA), octadecanethiol (ODT) and 11-mercaptoundecanoic acid (MUA) were
used for the synthesis of functionalized silver selinide nanoparticles (Ag2Se NPs) [18]. The formed NPs were washed with
ethanol and then dispersed in toluene using an ultrasonicator
for further use.
2. Surface-modified BaTiO3 NPs were synthesized according to
the literature procedure [19]. Briefly, BaTiO3 NPs (0.5 g) surfaces were hydroxylated by refluxing with 200 mL of H2O2 for
4 h at 106 C. The hydroxylated BaTiO3 NPs were filtered and
then washed with deionized water for two times. The hydroxylated BaTiO3 NPs were directly added into 250 mL round
bottom flask that contained 50 mL of 50 % (v/v) aqueous
alcohol along with 0.5 (w/v) of 12-hydroxy octadecanoic acid
(HOA). The reaction mixture was heated at 90 C under vigorous stirring for 3 h. The reaction mixture was cooled to
room temperature and HOA-modified BaTiO3 NPs were
treated with 1 N HCl until the pH became 56 in order to
agglomerate the nanopowder and to facilitate NPs filtration.
The surface-modified BaTiO3 NPs were washed with deionized water and acetone in sequence, several times. The surfacemodified BaTiO3 NPs were dried in vacuum oven for 24 h and
then dispersed in toluene by ultrasonication for 10 min.
2.3
Bacteria Growth
Methods
3.1 Functionalized
Nanomaterials
as Matrices
for Peptides
and Proteins
485
486
3.4 Nanomaterials
as Extracting
and Concentrating
Probes for Enrichment
of Peptides
and Proteins
487
Among different possible bioanalytical approaches, nanomaterialsbased MALDI mass spectrometry-based proteomics is increasingly
used to acquire the data important for understanding these processes. For example, Mn2+-doped ZnS-cysteine NPs-based
MALDI-MS was successfully used for the analysis of peptide mixture of Leu-enk (3.0 M), Met-enk (3.0 M), and HW6 (2.1 M)
(Fig. 1). It can be observed that Mn2+-doped ZnS-cysteine NPsbased MALDI-MS provided remarkable mass spectra for peptide
mixture, corresponding to the mass peaks of peptides at m/z 578.2,
595.3, 615.6, 906.6, and 929.5 for [Leu-enk + Na]+, [Metenk + Na]+, [Leu-enk + K]+, [HW6 + H]+, and [HW6 + Na]+, respectively. All ion signals were generated with sodium and potassium
adducts while only HW6 peptide was generated as protonated
ions. This reason is due to the alkali metals have greater affinity
toward the small molecules in MALDI-MS and also due to the
nanomaterials as the matrix lacking of the proton sources.
Moreover, large proteins were successfully detected in E. coli by
using HgTe nanostructures as matrices in MALDI-MS (Fig. 2).
The peaks at m/z 10,770, 21,539, 10,728, and 21,454 that correspond to the adducts [zSSAT1 + 2H]2+, [zSSAT1 + H]2+, [zSSAT1
R101A + 2H]2+, and [zSSAT1R101A + H]+, respectively, resulting
from the recombinant proteins of zSSAT1 or zSSAT1 R101A
under individual plasmids transformed into E. coli strain BL21.
Although zSSAT1 and zSSAT1 R101A share 99.5 % amino acid
sequence homology and only one amino acid variation, our
approach allowed differentiation of them, showing its potential for
the rapid detection of proteins. Interestingly, HgTe nanostructuresbased MALDI-MS was effectively detected protein G in both the
488
Fig. 1 MALDI-mass spectra of peptide mixture (Leu-enk, Met-enk, and HW6) using (a) CHCA and (b) Mn2+doped ZnS-cysteine nanoparticles as the matrices. Peak identities: m/z 578.2, 595.3, 615.6, 906.6 and 929.5
which are attributed to the [Leu-enk + Na]+, [Met-enk + Na]+, [Met-enk + K]+, [HW6 + H]+ and [HW6 + Na]+.
Reproduced from ref. 20 with permission from Royal Chemical Society
absence and presence of Brij 76, but the protein G IgG complex
was detectable only in the presence of 0.1 % Brij 76 [22]. The signals at m/z 26,023 and 13,019 represent the [protein G + H]+ and
[protein G + 2H]2+ adducts. The mass peaks at m/z 74,885, 49,984,
26,023, 13,019, 86,585, and 58,297 correspond to the
[IgG + 2H]2+, [IgG + 3H]3+, [protein G + H]+, [protein G + 2H]2+,
[IgG+ protein G + 2H]2+, and [IgG + protein G + 3H]3+ adducts
(Fig. 3). Using this approach, singly and multiply charged (1+, 2+,
3+, and 6+) adducts of IgG were observed and this approach successfully allowed to detect protein G (m/z 26,023) and IgG (m/z
149,931) at concentrations as low as 2 M (1 pmol) and 5 M
(2.5 pmol).
489
Fig. 2 Mass spectra of solutions containing HgTe nanostructures and the recombination proteins (0.01 g/L)
(a) zSSAT1 and (b) zSSAT1 R101A. SALDI-MS was performed in the linear mode. The peaks at m/z 10,770,
21,539, 10,728, and 21,454 represent the adducts [zSSAT1 + 2H]2+, [zSSAT1 + H]+, [zSSAT1R101A + 2H]2+, and
[zSSAT1 R101A + H]+, respectively. Reproduced from ref. 7 with permission from The American Chemical Society
Fig. 3 Mass spectra of IgG, protein G, and their complexes, recorded through SALDI-MS using HgTe nanostructures (a) in the absence and (b and c) in the presence of 0.1 % Brij 76. The samples were prepared in 20 mM
ammonium citrate (pH 5.0) containing 1 M Zn(II). The signals at m/z 149,931, 74,885, 49,984, 24,997,
26,023, 13,019, 86,585, and 58,297 represent the adducts [IgG + H]+, [IgG + 2H]2+, [IgG + 3H]3+, [IgG + 6H]6+,
[protein G + H]+, [protein G + 2H]2+, [IgG + protein G + 2H]2+, and [IgG + protein G + 3H]3+, respectively.
Reproduced from ref. 22 with permission from The American Chemical Society
Fig. 4 MALDI-TOF mass spectra of microwave tryptic-digested peptides originated from BSA (6.0 M) using
(a) 2,5-DHB as the conventional matrix and (b) QDs-DDTC as the affinity probes. Reproduced from ref. 14 with
permission from Elsevier
491
a
5000
7 2000
4000
1000
13
3000
Absolute intensity
1000
x10
1600
17
15
2000 1
0
4
14
1800
2000
2200
2400
22
10
15
4000
12 13
2000
11
1600
16
18
17
14
1800
19
2000
2200
22
10
1250
1500
2400
23
24
0
1000
21
20
1
1 2
21
20
1750
2000
m/z
2250
2500
2750
3000
3250
Fig. 5 (a) Direct MALDI mass spectrum of microwave tryptic digest of -casein (4.0 106 M). (b) MALDI mass
spectrum of microwave tryptic digest of -casein using BaTiO3 NPs (12 mg/mL) as enrichment probes.
Reproduced from ref. 21 with permission from Springer
492
Table 1
List of identified phosphopeptides by MALDI-MS from the microwave tryptic digest of non-fat milk
(50-fold dilution) using BaTiO3 NPs as affinity probes
Sr. No
Peak number
Observed m/z
Phosphopeptide sequences
M1
924.4
DIGpSESTEDQAMEDIK (-S1/5873)
M2
976.2
YKVPQLEIVPNpSAEER (-S1/119134)
M3
1,003.3
NANEEEYSIGpSpSpSEEpSAEVATEEVK (-S2/6185)
M4
1,027.8
DIGpSEpSTEDQAMEDIKQ (-S1/4359)
M5
1,103.5
GNAEGpSpSDEEGKLVIDEPAK (-S1/180188)
M6
1,251.6
TKVIPYVRYL (-S2/213222)
M7
1,266.8
YLGYLEQLLR (-S1/91100)
M8
1,293.7
QMEAEpSIpSpSpSEEIVPNpSVEQ (-S1/7493)
M9
1,337.5
VNELpSKDIGpSEpSTEDQAMEDIK (-S1/5273)
10
M10
1,383.7
FFVAPFPEVFGK (-S1/3849)
11
M11
1,593.6
TVDMEpSTEVFTKK (-S2/153165)
12
M12
1,637.2
YLGYLEQLLRLKK (-S1/106118)
13
M13
1,641.3
FFVAPFPEVFGKEK (-S1/3851)
14
M14
1,759.8
HQGLPQEVLNENLLR (-S1/2337)
15
M15
1,769.3
LYQGPIVLNPWDQVK (-S2/114128)
16
M16
1,847.4
DIGpSETEDQAMEDIK (-S1/5873)
17
M17
1,927.6
DIGpSEpSTEDQAMEDIK (-S1/119134)
18
M18
2,105.0
TDAPSFSDIPNPIGSENSEK (-S1/189208)
19
M19
2,235.2
HPIKHQGLPQEVLNENLLR (-S1-(1937))
20
M20
2,618.7
NTMEHVpSpSpSEESIIpSQETYK (-S1/1736)
21
M21
2,678.0
VNELpSKDIGpSEpSTEDQAMEDIK (-S1/5273)
Reproduced from ref. 21 with permission from Springer. pS refers to phosphorylated serine unit
493
Fig. 6 MALDI mass spectra of identified hydrophobic proteins in E. coli by using (a) HOA-modified BaTiO3 NPsassisted LLME along with SA (0.5 M) as the matrix and (b) SA (0.5 M) as the matrix without HOA-modified
BaTiO3 NPs. Reproduced from ref. 19 with permission from Elsevier
494
Notes
1. Nanomaterials exhibited high surface area, good photostability, which results to improve the solubility in aqueous media
and their conjugation with biomolecules.
2. The mass limit when SALDI-MS was used with HgTe nanostructures as matrixes reaches up to 150,000 Da (IgG), much
higher than those obtained using other kinds of nanomaterials.
The HgTe nanostructures-based MALDI-MS allowed to
detect weak drugprotein complexes (BSA/Y and hCAI-ACZ
complexes). Importantly, HgTe nanostructures allow the analyses of proteins and their complexes under mild conditions and
with greater tolerance toward salts.
3. BaTiO3 NPs are good candidatures for effective enrichment of
phosphopeptides from microwave tryptic digest of -casein.
Moreover, after centrifuging of BaTiO3 NPsphosphopeptides
conjugates from microwave tryptic digests, the trapped phosphopeptides are directly identified by MALDI-MS without
further washing procedure step. The entire procedure was
completed within 60 min.
4. The high degree of protein digestion was observed by using
following microwave conditions (microwave power: 210 W
and microwave irradiation time: 50 s) and trypsin-to-protein
ratio (1:30, w/w) was used for the digestion of cytochrome c,
lysozyme, and BSA proteins in the present study.
5. The BaTiO3 NPs acted as concentrating probes for phosphopeptides from microwave tryptic digest of -casein, since trace
levels of phosphopeptides (12, 16, 18, 19, and 20) are effectively enriched and identified without non-phosphorylated
peaks. It confirms that surfaces of BaTiO2 NPs have high capability to adsorb phosphopeptides from microwave tryptic
digest of -casein. It can be proved that BaTiO3 NPs have welldispersed in solution which can improve signal intensities, S/N
ratio and allowed unambiguous trapping of low abundance
phosphopeptides (Fig. 2).
6. The binary matrix approach applying Au NP-SDME microdroplets which can be homogeneously mixed with the organic
matrix (CHCA or SA) to form homogeneous crystals and thus
favorable for sensitive detection of peptides and proteins at low
concentration in MALDI-MS. In addition, the Au NP-SDME
can also serve as multifunctional nanoprobes for ionization,
enrichment, preconcentration, and desalting purposes for a
variety of peptides and proteins. Moreover, the sample can be
directly deposited onto the MALDI target plates and directly
sent for MALDI-MS analysis without the need for any further
washing steps or elution processes.
495
Acknowledgement
The authors greatly acknowledge Elsevier, Royal Chemical Society,
Springer, and American Chemical Society for giving copyright permission to reuse figures and some of the text for this chapter.
References
1. Godovac-Zimmermann J, Brown LR (2001)
Perspectives for mass spectrometry and functional proteomics. Mass Spectrom Rev 20:157
2. Chiang C-K, Chen W-T, Chang H-T (2011)
Nanoparticle-based mass spectrometry for the
analysis of biomolecules. Chem Soc Rev 40:
12691281
3. Zhu Z-J, Rotello VM, Vachet RW (2009)
Engineered nanoparticle surfaces for improved
mass spectrometric analyses. Analyst 134:
21832188
4. Kailasa SK, Cheng K-H, Wu H-F (2013)
Semiconductor nanomaterials-based fluorescence spectroscopic and matrix-assisted laser
desorption/ionization (MALDI) mass spectrometric approaches to proteome analysis.
Materials 6:57635795
5. Kailasa SK, Kiran K, Wu H-F (2008)
Comparison of ZnS semiconductor nanoparticles capped with various functional groups as
the matrix and affinity probes for rapid analysis
of cyclodextrins and proteins in surface-assisted
laser desorption/ionization time-of-flight mass
spectrometry. Anal Chem 80:96819688
6. Kailasa SK, Wu H-F (2011) Semiconductor
cadmium sulphide nanoparticles as matrices for
peptides and as co-matrices for the analysis of
large proteins in matrix-assisted laser desorption/ionization reflectron and linear time-offlight mass spectrometry. Rapid Commun Mass
Spectrom 25:271280
7. Chiang C-K, Yang Z, Lin Y-W, Chen W-T, Lin
H-J, Chang H-T (2010) Detection of proteins
and protein-ligand complexes using HgTe
8.
9.
10.
11.
12.
13.
14.
496
15.
16.
17.
18.
INDEX
A
Acetone precipitation .......................................................410
Affinity chromatography ................................. 160, 211222,
307309, 319, 467, 472
Affinity electrophoresis.....................................................327
Alkylation ..........128, 159, 170, 173, 196197, 206, 236239,
241242, 251253, 257, 261, 262, 267269, 272,
273, 278, 282, 312, 316, 371, 372, 375, 468, 473
Amyloid-binding proteins ........................................465475
Antibody........................................... 24, 26, 30, 33, 121, 131,
132, 135139, 141143, 147149, 196, 279, 285,
286, 291, 298, 301, 325, 327, 331, 338, 343345, 347,
383, 384, 386390, 394, 398, 402411, 430, 437,
438, 442445, 448450, 452, 457, 460, 462
APV Gaulin .......................................................................2
B
Bead-based array ......................................................441, 442
Bead beater .......................................................................2, 4
Bead impact ......................................................................27
Bead mill ..........................................................................57
Benzonase.......................... 337, 358, 360, 366, 444, 446, 457
Bicinchoninic acid (BCA) protein assay ........... 310, 313, 444
BioNeb cell disruption........................................................16
Bis-Tris SDS PAGE gels .......................... 328, 329, 344, 345
Blue native electrophoresis ...............................................419
Bradford protein assay .............................. 296, 396, 398, 472
Branson sonifier................................................................296
C
Carbamidomethylation ..................................... 288, 363, 474
Carbamylation ............................................ 30, 271, 318, 365
Carrier ampholytes ............................122, 159, 164, 396, 411
Cell culture
CHO-K1 cells ............................................................398
Hep 70.4 cells .............................................................451
LIM1863 cells ............................................ 180, 182, 192
primary skeletal muscle cells .........................................57
SW 480 cells ....................................... 168170, 173, 176
Cell disruption
bead impact methods ..................................................27
electromotive field ........................................................18
high pressure batch ................................... 2, 3, 1013, 19
high pressure flow ..................................... 2, 3, 1316, 19
low pressure ...........................................2, 3, 7, 16, 17, 19
D
Data normalization
stain-free technology ..........................................381390
western blotting ..................................................381390
Detergents .................................... 28, 30, 105, 110, 119, 120,
124, 130, 133, 136, 146148, 168, 203, 250,
254, 257, 265, 270, 311, 334, 410, 411, 417, 421,
423, 424, 428, 451
Dimethylformamide (DMF) ........................... 122, 127, 131,
140, 144, 146, 311, 315, 429, 431
Anton Posch (ed.), Proteomic Profiling: Methods and Protocols, Methods in Molecular Biology, vol. 1295,
DOI 10.1007/978-1-4939-2550-6, Springer Science+Business Media New York 2015
497
PROTEOMIC PROFILING
498 Index
Disulfide bridges ..............................................................157
Dithioerythritol (DTE) ....................................................164
Dithiothreitol (DTT) .................................67, 105, 110, 120,
122, 123, 128, 130, 145, 157, 159, 164, 191, 195, 197,
214, 216, 226, 236, 237, 246, 251, 252, 261, 262, 266,
268, 269, 271273, 278, 296, 298, 311, 312, 358, 361,
371, 372, 375, 396, 397, 400, 409411, 424, 429, 430,
432, 433, 457, 468, 485, 486
DMEM. See Dulbeccos modified Eagle medium
DMF. See Dimethylformamide
Dounce homogenizer .......................................................384
Droplet low pressure nebulizer ...........................................17
Droplets impingement .......................................................16
DTT. See Dithiothreitol
Dulbeccos modified Eagle medium (DMEM) ........... 45, 48,
201, 260, 278, 280, 331, 443
Dynamic range compression.......................................99105
Dyno-Mill ............................................................................6
E
Ectosome ..................................................................167, 168
Electron microscopy ............ 94, 173, 175, 183, 191, 196, 206
Electrophoresis
affinity electrophoresis ................................................327
blue native PAGE .......................................................419
characterization of extracellular vesicles..............167168
2-D electrophoresis ................................... 158, 394, 396,
397, 399400, 410, 412, 413
differential in gel electrophoresis (DIGE) ............. 61, 63,
105, 156158, 430, 431, 433, 434, 436, 438, 439
free flow electrophoresis .....................................415424
isoelectric focusing ............................... 51, 119, 122, 154,
156, 158, 176, 231, 293302, 311, 319, 369, 419, 428,
429, 431, 432, 475
SDS-PAGE ................................................ 267, 312, 429
zone electrophoresis .................................... 416, 419, 421
Electrospray ionization (ESI) .......................... 111, 145, 154,
163, 362, 369
Escherichia coli (E. coli) ............. 7, 73, 452, 484, 487, 492, 493
ESI. See Electrospray ionization
Exosome
density-gradient separation ................................179206
electron microscopy ............................ 183, 191, 196, 206
immunoaffinity capture methods........................179206
ultracentrifugation ..............................................179206
F
Filter aided sample preparation (FASP) .............. 6670, 237,
240241, 473
mass spectrometry ..............................................237, 240
Formalin fixed paraffin embedded (FFPE) ...... 109114, 120
Free flow electrophoresis (FFE) ...............................415424
French press ..............................................................2, 1012
G
Glycoprotein.............................................................205, 357
enrichment .................................................................357
Glycosidases .....................................................................356
Glycosylation ....................................275291, 324, 355357,
366, 367, 456
Gradient centrifugation
exosomes .............................................................180, 181
lymphocytes ............................................................3341
mitochondria .................................................... 83, 93, 94
H
Heat stabilization of proteins .......................................2131
HeLa cells ..........147, 280, 289, 334, 338, 342, 371, 376, 377
HILIC. See Hydrophilic interaction liquid chromatography
Homogenizer................................................. 79, 13, 15, 78,
80, 82, 89, 9196, 131, 321, 384, 459, 472
Horseradish peroxidase (HRP)................................. 398, 402
western blotting ..................................................398, 402
Host cell protein
antibody coverage ....................................... 398, 403408
CHO cells ...................................394, 396, 410, 412, 413
2-D electrophoresis ..... 394, 396, 399400, 410, 412, 413
PDQuest ............................................................398, 403
ProteoMiner ...............................................................100
western blotting ..................................................393414
Human plasma ...................................................................56
Human serum................................................... 100, 101, 482
Hydrophilic interaction liquid chromatography
(HILIC) ................................276, 279281, 286, 288
I
ICPL. See Isotope-coded protein label
IEF. See Isoelectric focusing
Immobilized metal affinity chromatography
(IMAC) ........................................276, 279281, 284,
289, 291, 306, 308310, 314
Immobilized pH gradient (IPG) ..................... 100, 122, 127,
128, 154, 158, 231, 294, 296, 298, 300302, 311, 315,
319, 370, 371, 373, 375, 377, 396, 399401, 411, 412,
429, 430, 432, 436
Immunoaffinity ...........................33, 137, 139, 168, 179207
Immunoassay ...................................57, 60, 62, 100, 441452
multiplexing ........................................................441452
Immunoblotting .......................................118, 124, 204, 206,
227228, 327, 331, 340, 343
Immunoprecipitation
biotin ...........................................137, 141, 143, 147, 148
crosslinking .................................................................138
magnetic beads ....................135, 137, 138, 140, 142149
mass spectrometry ..............................................135149
protein AG ................................. 137140, 143, 147149
L
Label-free proteomics....................................... 112114, 207
Liquid chromatography couples to mass spectrometry
(LC-MS) .................... 28, 71, 72, 140, 141, 143, 144,
48, 149, 160163, 236, 262, 369378, 469,
474, 475
Loading buffer........................................... 73, 148, 216, 220,
227, 278, 279, 282286, 290, 291
SDS-PAGE ........................................................227, 229
Lowry protein assay ..........................................................264
Lymphocytes
density gradient centrifugation ...............................3341
magnetic cell sorting ...............................................3341
sample preparation........................................................34
Lysis buffer .............................................45, 47, 67, 110, 111,
114, 140, 142, 144, 147, 148, 214, 216, 218, 220, 231,
237, 240, 246, 263, 265, 271, 272, 282, 290, 310, 313,
318, 319, 331, 332, 334, 370, 372, 417, 420, 444, 446,
447, 451, 457459, 468, 473
Lysis solution ....................................................................375
Lysosome ............................................................................76
M
Maldi. See Matrix-assisted laser desorption ionization
Mass spectrometry (MS)
electrospray ionization (ESI) ............................. 111, 112,
145, 154, 163, 240, 299, 301, 362, 369
Maldi .................................................. 163, 246, 479495
tandem .........................................111, 145, 155, 254, 474
Matrix-assisted laser desorption ionization
(Maldi) .................... 22, 163, 236, 245, 272, 479495
Matrix-assisted laser desorption ionization
time-of-flight mass spectrometry
(MALDI-TOF-MS) .............................. 82, 485, 490
Membrane proteins ...................159, 401, 402, 419, 428, 493
complexes ...........................................................415424
isolation with FFE ..................................... 416, 417
Microarray ........................................................ 455463, 481
human protein (HuProt) ......................................55463
Microfluidizer........................................................... 2, 15, 16
N
Nanomaterials, MALDI MS....................................479495
N-terminal sequence analysis ...................................249, 254
O
Organelle isolation ........................75, 76, 211, 212, 221, 419
chloroplast, mitochondria ................75, 76, 211, 212, 221
P
Paraffin embedding ..........................................................117
Parr cell disruption bomb .......................................10, 1213
PBS. See Phosphate-buffered saline
Peptides
desalting.......................................................... 70, 71, 246
fractionation by isoelectric focusing ....................369, 373
glycopeptides .......................277283, 289, 291, 355367
phosphopeptides ........................................ 276283, 288,
289, 291, 294, 300, 302, 324, 326, 486, 491, 492, 494
reversed-phase (RP)-chromatography ............... 154, 160,
168, 176, 249, 369, 474
shotgun proteomics .......................70, 118, 207, 324, 369
Peroxisome .........................................................................76
Phenylmethylsulfonyl fluoride (PMSF) ................... 468, 472
protease inhibitor ................................................468, 472
Phosphatase inhibitor ........................................ 26, 147, 278,
296, 297, 310, 318, 319, 345, 444, 446
Phosphate-buffered saline (PBS) .................... 36, 3840, 45,
46, 49, 50, 57, 61, 63, 68, 101, 105, 121, 124, 125,
140, 142144, 147, 169, 170, 174, 188195, 202,
204, 260, 262, 263, 271, 358, 360, 370, 371, 396, 398,
444446, 448, 449, 457, 458, 460
Phosphopeptides
enrichment .......... 276, 278280, 282283, 294, 324, 494
identification............................................... 294, 300, 486
Phosphoproteins
affinity electrophoresis ................................................327
enrichment .................................................................326
fluorescent gel staining ...............................................318
Phosphorylation ........................................ 2227, 31, 56, 58,
114, 147, 275291, 294, 300302, 305, 306,
323351, 442, 443, 447, 448, 456
Phos-tag technology .................................................325326
Plasma proteins .................................................. 99101, 104
PROTEOMIC PROFILING
500 Index
Polytron ........................................................................2, 8, 9
Posttranslational modifications (PTM)
acetylation...........................................................276, 277
glycosylation ....................................... 276, 277, 324, 456
phosphorylation ........... 276, 277, 289, 294, 323, 324, 456
Preadipocytes ..........................................................4450, 52
Protease inhibitors ................ 50, 84, 110, 114, 214, 216, 260,
270, 278, 290, 310, 318, 358, 360, 444, 457, 468, 472
Protein assays
bicinchoninic acid (BCA) assay .................. 310, 313, 444
Bradford assay............................................ 111, 171, 172,
188189, 193, 203, 264, 296, 298, 396, 398, 472
detergent compatible (DC) assay....................... 120, 124,
130, 370, 372
Lowry assay ................................................................264
NanoDrop .......................................... 237, 238, 241246
Protein complexes..............168, 415417, 419421, 423, 424
Protein depletion ......................................................225232
Protein digestion
alkylation ..................... 170, 261, 262, 268, 269, 272, 273
filter aided sample preparation (FASP) ......... 6669, 237,
240241, 473
in-gel digest ........................................118, 119, 129, 236,
238240, 250, 259261, 263, 266270, 325, 331
in-solution digest ....................................... 136, 138, 145,
147, 172174, 237, 241242, 260, 266, 268270
Lys-C .................................................145, 278, 289, 296,
298, 302, 358, 361, 371, 372, 375
OASIS ................. 236, 238240, 278, 290, 358, 361, 362
reduction.....................................170, 173, 206, 236, 237,
239, 241242, 261, 262, 266, 268, 272, 278, 282, 375
STAGE-tip................................................. 197, 361, 362
trypsin.................................................111, 112, 114, 123,
137, 141, 145, 146, 163, 168, 170, 173, 176, 197, 236,
237, 239, 240, 246, 253, 261, 262, 267, 268, 278, 282,
290, 358, 361, 371, 372, 375
Protein extraction .................................... 110112, 117133,
164, 216, 220, 226, 231, 250, 266, 290, 431
Protein fractionation..........................139, 176, 245, 294, 427
Protein interaction ............................................ 442, 443, 452
Protein labeling
dimethyl labeling ........................................ 249257, 290
fluorescent dyes.................................... 79, 133, 154159,
309, 315, 318, 324, 428
stable isotopes ............................................. 155, 160164
(2,4,6-trimethoxyphenyl) phosphonium bromide
(TMPP) labeling ..........................................249257
Protein microarray ............................................ 455, 461, 462
Protein N-termini ....................................................249257
Protein precipitation
acetone ................................................................260, 265
Clean-up kit ................................111, 127, 311, 315, 410
methanol/chloroform......................... 168, 170, 172174,
176, 260, 265266, 473
trichloroacetic acid (TCA)...........218, 226, 260, 265, 266
R
Reducing agents ...............................105, 119, 120, 124, 130,
318, 348, 483
Reversed-phase high performance liquid chromatography
(RP-HPLC) ......................................... 160, 198, 278
Ribulose 1,5 Bisphosphate Carboxylase/Oxygenase
(RuBisCO)
high abundant protein ................................................225
plant leaves .................................................................226
protamine sulfate ................................................225232
protein depletion ................................................225232
Rotor/stator homogenizers ...............................................79
S
Sample cleanup......................................... 118, 157, 164, 307
Sample preparation .....................................22, 24, 44, 57, 61,
6574, 105, 109, 110, 160, 202, 206, 235237,
257, 259, 278, 280281, 294, 307, 311, 315,
331332, 334, 369378, 384385, 396, 398,
416, 419421, 424, 429433, 443447, 467, 473,
481, 482
Secretome analysis ........................................................46, 59
Shotgun proteomics...............6574, 118, 207, 249, 324, 369
SILAC. See Stable isotope labeling with amino acids in cell
culture
Silver staining ............................................ 61, 154, 156, 219,
220, 232, 266, 272, 313, 317, 318
Solid phase extraction (SPE) ........................... 238, 243, 244,
358, 361, 371, 372, 374, 376
Sonicator
bath sonicator .......................................................68, 398
tip sonicator .................................................. 31, 264, 271
SPE. See Solid phase extraction
SPR. See Surface plasmon resonance
Stable isotope labeling with amino acids in cell culture
(SILAC) ................................135, 160162, 246, 357
Stain-free technology ...............................................381390
Surface plasmon resonance (SPR) ................... 326, 466, 467,
469471, 475
Swiss-Prot ................................................................280, 288
Sypro Ruby ........................ 133, 308, 320, 401, 403, 412, 413
T
TCEP. See Tris(2-carboxyethyl) phosphine
Tissue grinder ...................................................................2, 9
Tissue homogenization ................................ 118, 84, 85, 88
Tris(2-carboxyethyl) phosphine (TCEP)................. 140, 145,
157, 170, 237, 245, 273, 429, 431, 432, 439
U
Ultracentrifugation ........................... 167172, 175, 179207
Ultrafiltration ...........................................................181, 182
Urine proteomics ................................................................66
V
Vacuum centrifuge ........................................... 123, 126, 192,
197, 240, 245, 252, 253, 267, 270, 283, 285, 286,
291, 359
W
Western blotting
data normalization ......................................................387
house keeping proteins .......................................381, 382
immunostaining .......................................... 384, 386388
loading control ....................................................381, 382
stain-free technology ..........................................381390
Wheaton Potter-Elvehjem tissue grinder .............................9
Z
Zone electrophoresis (ZE)........................................ 416, 419