The Surfaceome: Methods and Protocols
The Surfaceome: Methods and Protocols
Kenneth R. Boheler
Rebekah L. Gundry Editors
The
Surfaceome
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Kenneth R. Boheler
School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong,
SAR, China; Stem Cell & Regenerative Medicine Consortium, School of Biomedical Sciences,
LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
Rebekah L. Gundry
Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
Editors
Kenneth R. Boheler Rebekah L. Gundry
School of Biomedical Sciences Department of Biochemistry
LKS Faculty of Medicine Medical College of Wisconsin
The University of Hong Kong Milwaukee, WI, USA
Hong Kong, SAR, China
Stem Cell & Regenerative Medicine
Consortium, School of Biomedical Sciences
LKS Faculty of Medicine
The University of Hong Kong
Hong Kong, SAR, China
The plasma membrane is the gateway through which cells sense and respond to their
microenvironment. Critical to this process are cell surface proteins that span (transmem-
brane) or are anchored/embedded in the plasma membrane. Cell surface proteins perform
diverse functions, including nutrient and ion transport, intra- and intercellular communica-
tion, receptor signaling, and enzymatic reactions. Altogether, the collection of proteins that
reside at the cell surface (i.e., surfaceome) facilitates interactions with pathogens, binding of
chemical messengers, and transmission of signaling cascades, and it is required for cell
migration, adhesion, and survival. Surfaceome content, including protein identity and
modifications, differs among cell types and is dynamic during development and disease
states. For these reasons, and the fact that cell surface proteins are accessible, the surfaceome
is a rich source of drug and immunotherapy targets and contains unique markers that can be
used to identify cell types, disease states, and cellular phenotypes. Despite their critical
functions in health and disease, cell surface proteins have historically been understudied in
most cell and tissue types. This is due, in part, to the challenges posed by their relatively low
abundance when compared to intracellular proteins, their hydrophobic nature, and the
difficulty in biophysically purifying plasma membrane proteins without contamination
from intracellular membrane components. Moreover, high-quality antibodies are currently
available for a limited subset of cell surface proteins.
Considering these challenges, the development and dissemination of modern methods
and technologies that enable the study of cell surface proteins will undoubtedly advance a
broad range of research efforts, including our understanding of cellular differentiation and
development, host-pathogen interactions, and metastatic processes, and will lead to the
development of new treatments for disease. In this volume of Methods in Molecular
Biology: The Surfacome, we have assembled 19 chapters that cover a variety of methods
ranging from molecular and cellular biology to proteomics to bioinformatics. The overall
aim of this edition is to provide state-of-the-art techniques and tools to assess the surfa-
ceome content, modifications, and function. The volume does not include standard
approaches extensively reviewed elsewhere, nor does it include methods to analyze lipids
and glycans, which are key components of the plasma membrane and worthy of separate
volumes dedicated to their study. While most of the methods described in this volume are
generally applicable to any cell type, some chapters focus on specific cell types and/or
specific molecule classes of interest. These latter chapters are designed to illustrate the
application of these procedures and protocols in defined systems, but the approaches should
be applicable across a broad range of cells. Altogether, we hope this collection of methods
will facilitate the study of cell surface protein biology and function and lead to the discovery
of new drug and immunotherapy targets for treating disease and new immunophenotyping
markers for studying cellular function, differentiation, and disease. The chapters are
arranged in four parts, beginning with discovery-based and then targeted strategies for
cataloging surfaceome content, moving to functional assays for specific protein and cell
types, and ending with computational approaches.
Part I focuses on discovery-based approaches for cataloging surfaceome content and
includes methods to analyze the surfaceome of bacteria, avian embryos, and mammalian
systems. Chapters in this part focus on modern proteomic methods that offer the ability to
v
vi Preface
specifically target cell surface proteins with limited interference from intracellular membrane
proteins. These include surface membrane protein enrichment techniques, using proteases
to “shave” proteins from the surface of bacteria to identify surface-exposed proteins, and
exploiting the avian system to study developmental changes in cell surface proteins, includ-
ing bioinformatics-based techniques to translate to human orthologs. Subsequent chapters
describe the Cell Surface Capture Technology, a targeted analytical approach to specifically
identify cell surface N-glycoproteins, the use of iron oxide nanoparticles to enrich plasma
membrane proteins, and methods to profile secreted proteins and exosomes in cell culture, a
topic that has recently gained attention across a variety of research disciplines.
Part II focuses on targeted approaches to analyze the surfaceome. The chapters in this
part include methods to overexpress specific targets in Sf9 cells and an approach to generate
bispecific antibodies that are valuable for targeting cancer and somatic cells. Also included is
a tutorial chapter on flow cytometry and its application to immunophenotyping to assist
novices in their pursuit of surface proteins. The last chapter in this part provides an example
of how ELISA and flow cytometry are applied to detecting the G protein-coupled receptor
CXCR4, a strategy particularly valuable for investigators interested in G proteins and in drug
repurposing.
Part III focuses on cell-based functional analyses. This part begins with a review on
voltage-dependent sodium channels and methods for high content electrophysiological
analyses. Methods are then described for the evaluation of vascular endothelial cell functions
and approaches to study signal transduction of surface receptor tyrosine kinase in neurons. A
comprehensive analysis of cell polarity, using retinal pigmented epithelium as a model
system, is then described, including techniques for immunostaining for apical and basolat-
eral membrane markers, polarized cytokine secretion, fluid transport, phagocytosis, and
identification of plasma membrane proteins through cell surface capturing technologies as
described in the first part. This part finishes with a description of methods that take
advantage of extracellular matrix components to capture mesenchymal stromal cells under
flow, model disease states, and ultimately analyze cell-matrix interactions through the use of
3D microtissues.
Part IV focuses on computational approaches in surfaceome studies and describes a new
web-based platform, Targets-search, that incorporates information from a variety of sources
including the Cell Surface Protein Atlas and online drug databases, to facilitate identification
of surface proteins that are informative for a particular cell type or disease and known drugs
that interact with these proteins.
In closing, we would like to thank Springer for its support, dedication to this project,
and patience in developing this book. We also wish to especially thank all of the authors for
their time, energy, and valuable contributions. With their efforts, we have assembled what
we hope will be a valuable resource for those research laboratories working to advance the
study of surface protein biology.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345
Contributors
ix
x Contributors
Abstract
The bacterial surfaceome, comprising outer membrane-sorted and/or associated (i.e., cell transporters),
cell surface-exposed (i.e., adhesins) and extracellularly secreted proteins (i.e., toxins), has been character-
ized in bacterial pathogens, such as Bordetella pertussis (Bp) to provide information for use in development
of diagnostic and prevention strategies. This protein subset has clinical significance, as these bacterial
proteins are often associated with attachment to host cells, microbial pathogenesis and antibody-mediated
immunity. Here we describe classical surface membrane protein enrichment techniques, followed by
proteomic methodologies, such as gel-free protein separation and antibody-affinity capture technologies
in combination with nano-liquid chromatography mass spectrometry, for the identification and characteri-
zation of Bp surfaceome proteins.
Key words Carbonate extraction, Membrane proteins, Sample solubilization, Antibody affinity,
Immunoprecipitation, Gel-free, Mass spectrometry, nLC-MS/MS, Bioinformatics
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_1, © Springer Science+Business Media, LLC 2018
3
4 Yulanda M. Williamson et al.
2 Materials
2.1 Enriched Outer 1. Bordet-Gengou agar plates (Becton, Dickinson and Company,
Membrane Isolation Sparks, MD, USA).
Method 2. Stainer-Scholte media (SS- or SSM) (see Note 1): To prepare
1 L of SSM, add the following to 900 mL of water: 10 g
casamino acids, 10.7 g L-glutamic acid (Na salt), 0.24 g L-
proline, 2.5 g NaCl, 0.5 g KH2PO4, 0.2 g KCl, 0.1 g
MgCl2·6H2O, 0.02 g CaCl2, 6.1 g Tris·Cl and 2,6-o-
dimethyl-beta-cyclodextrin (heptakis). Adjust the pH to 7.6
using concentrated HCl, and next add water up to 1 L (broth
should be light yellow in color). Sterilize by autoclaving and
dispense 100 mL aliquots into 1000 mL screw-capped flasks.
The media can be stored up to a month at room temperature.
3. Carbonate buffer solution: ice-cold 100 mM sodium carbonate
(pH 11) (see Note 2).
4. Wash solution: 50 mM Tris–HCl buffer (pH 8) (see Note 3).
6 Yulanda M. Williamson et al.
3 Methods
3.1 Enriched Outer 1. Grow Bp Tohama I (T) strain on Bordet-Gengou agar and then
Membrane Isolation subculture into 1000 mL screw-capped flasks containing
Method: Cell 100 mL of SS-liquid media (see Note 1).
Harvesting 2. Cultures were maintained at 36 C with shaking (200 rpm)
until the OD650nm reaches 0.5–1.0.
3. Collect Bp bacterial cells from 100 mL of liquid culture by
centrifugation at 8000 g for 30 min at 4 C.
8 Yulanda M. Williamson et al.
3.3 Antibody Affinity 1. All experiments are performed in accordance to project specific
Capture Technology: animal protocol # 1642, “Production of Antibodies to Borde-
Preparation tella pertussis in New Zealand White Rabbits” approved by the
and Purification Institutional Animal Care and Use Committee (IACUC), Cen-
of Rabbit Anti-Bp ters for Disease Control and Prevention, Atlanta,
Immune Sera Georgia, USA.
2. For the antibody affinity capture technology, carry out all
procedures at room temperature unless otherwise specified.
3. Immunize New Zealand white rabbits, intraperitoneally, with
1 107 cfu Bp T strain; administer a comparable dosage every
2 weeks for 6 weeks (see Note 16).
Identification of Novel Bordetella pertussis Antigens 9
4. Bleeds are performed via a rabbit ear artery. The sampling site is
disinfected (marginal ear artery area is wiped with an alcohol
swab), and a 25 gauge butterfly needle-capped syringe (5 cc)
inserted into the artery as distally (toward the tip of the ear) as
possible.
5. Approximately 25 mL of blood is drawn from each rabbit
biweekly and dispensed in a K2EDTA-containing vacutainer.
6. Blood was collected before the first immunization (pre-bleed)
representing the normal rabbit sera negative control (NRS) and
after the third immunization for the rabbit anti-Bp immune
sera experiments (see Note 16).
7. After the third immunization, collect all remaining blood (via
exsanguination) from rabbits then euthanize and sacrifice (see
Note 17).
8. Prepare Bp immune sera by centrifuging vacutainers containing
blood into the Beckman Coulter, Inc. centrifuge for 20 min at
1200 g (see Note 18).
9. Purify rabbit anti-Bp immune sera using the Melon Gel® IgG
purification kit.
10. Determine the IgG concentrations using Easy-Titer® IgG
assay kit (see Note 19).
3.4 Rabbit Immuno- 1. Determine the optimal concentrations of antigen (Ag) and
precipitation (IP) antibody (Ab) for rabbit IP (see Note 20).
2. In an Eppendorf tube, combine purified IgG (2 μg) rabbit
antisera (Ab) with Bp T EMF (80 μg) (Ag) then adjust to a
final volume of 200 μL using x% NP40/PBS solution.
(x ¼ 0.22%, 0.45%, or 0.90% v/v). The detergent concentra-
tion should be optimized (see Note 21), and the concentration
that yields the largest quantity of target proteins should be used
for final data analysis.
3. Prepare two Eppendorf™ tubes to be used as negative controls
in a similar manner.
4. Combine purified NRS (2 μg) and Bp T EMF (80 μg) in one
tube and place Bp T EMF (80 μg) alone in the second tube (see
Note 22).
5. Place all samples (Bp T immune complex, NRS, Bp T EMF
only) on a Hula mixer® and allow them to rotate at 10 rpm at
room temperature for 1.5 h (see Note 23).
6. During the 1.5 h incubation period, wash three aliquots con-
taining 200 μL Protein G Dynabeads® (per sample, ~4 108
beads) with PCB. Resuspend each aliquot in 400 μL of PCB,
place each sample on the magnetic rack, and remove the super-
natant. Repeat 2. After the final wash, leave the bound Dyna-
beads® on the magnetic rack.
10 Yulanda M. Williamson et al.
3.5 Outer Membrane 1. EMF (10 μg) or EMF-antibody (IP) complexes (50 μL) are
Protein Identification: treated with 0.1% RG (10 μL) and heated at 100 C for 5 min
Protein Digestion and cooled at 4 C for 5 min.
2. After incubation, the protein containing tubes are briefly spun
at 10,000 g. The denatured proteins are digested with
sequence-grade trypsin (10 μg) and incubated at 37 C over-
night (18 h) (see Note 9).
3. To inactivate the RG, 1 M HCl (final concentration 175 mM)
is added to each tube and incubated for 30 min at 37 C,
followed by centrifugation at 10,000 g (15 min) (see Note
25).
4. The supernatant containing antibody-captured EMF tryptic
peptides is transferred to a fresh Eppendorf tube and dried
(see Note 26) to concentrate the peptide pool.
5. Equal volumes of tryptic peptides and a 0.1% formic acid, 50%
acetonitrile solution are transferred to vials, capped and stored
at 70 C until needed (see Note 27).
Identification of Novel Bordetella pertussis Antigens 11
= Antigen (Ag)
= Protein G DynaBeads®
BIOINFORMATICS
Fig. 1 Rabbit immunoprecipitation (IP) method coupled with nLC-ESI MS/MS workflow. Schematic of how IP
sample was isolated then analyzed by mass spectrometry, scaffold analysis, KEGG, and PSORTb
Fig. 2 Direct and indirect IP methods. Invitrogen™ Protein G Dynabeads® includes a Direct and an Indirect IP
method: In the direct method, the antibody and Protein G Dynabeads® are incubated, cross-linked, then the
antigen is added. In the indirect method, the antigen and antibody are added, incubated, and the Protein G
Dynabeads® are added later
12 Yulanda M. Williamson et al.
3.6 Liquid 1. Protein tryptic digests are loaded on a C18 packed silica capil-
Chromatography, ESI lary column and separated using a nanoflow reverse phase
Tandem Mass gradient at 400 nL/min (see Notes 28 and 29) [7, 8].
Spectrometry, 2. The peptides are separated by the reverse phase column and
and Bioinformatics ionized before being transferred to the mass spectrometer, for
MS and MS/MS acquisition (see Note 30) [7, 8].
3. For protein identification, mass spectrum raw files are extracted
and searched using a database search algorithm using specified
parameters (see Note 31).
4. MS/MS peptide and protein identifications are validated using
Scaffold (see Note 32). See Fig. 3 for a visual example of an
MS/MS spectrum profile [13, 14].
5. Bioinformatic tools can be further utilized to categorize iden-
tified proteins based on gene ontology/function or predicted
subcellular localization (see Note 33) [6, 15].
4 Notes
Fig. 3 Sequence of a known Bordetella membrane surface protein, virulence factor and immunogen,
filamentous hemagglutinin (FHA); and the scaffold mass spectrum b and y ion profiles for the tryptic peptide
14 Yulanda M. Williamson et al.
Fig. 3 (continued) GALALDGGAGVALQSAK digested from this protein. (a) Yellow highlights represent addi-
tional FHA MS/MS detected tryptic peptides identified within the FHA amino acid sequence (partial sequence
snapshot—amino acid 1–1150). Green highlights in the sequence represent deamidation of the amino acid—
glutamine (Q). MS/MS FHA detected peptide/protein identification corresponding to (b) Gel-free Bp T EMF and
(c) Indirect Bp T EMF-IP capture
Identification of Novel Bordetella pertussis Antigens 15
30. Ion source voltages for capillary columns are at flow rates of
300–600 nL/min and are generally between 1.7 and 2.3 kV.
The mass spectrometer was programmed to perform data-
dependent acquisition by scanning the mass range from m/z
400 to 1600 at a nominal resolution setting of 60,000 for
parent ion acquisition. For MS/MS analysis the mass spec-
trometer chose the top 16 most intense ions with two or
more charges. Singly charged ions were rejected for MS/MS
as these ions are likely due to detergents or other sample
additives. The tandem mass spectrometer implemented ulti-
mately is researcher-dependent, but one that incorporates a
hybrid mass spectrometer such as the LTQ-Orbitrap instru-
mental systems is ideal for surfaceome protein identification
and biomarker discovery.
31. Mascot Distiller (data extraction) and Daemon (mass spectrum
matching) (Matrix Science, London, UK; version 2.2.1.0) is
preferred. Suggested parameters for the mascot search include
digestion agent (trypsin), missed cleavages (two); fragment ion
tolerance mass (0.50 Da), precursor ion tolerance (200 ppm),
variable modification (oxidation). The database utilized is
researcher-dependent, but in this instance an NCBI nonredun-
dant database specific for the organism/typed-strain of choice
(Bp) was implemented.
32. Scaffold_4.0.6.1 (or current version) is preferred. Proteome
Software, Portland, OR, USA.
33. PSORTb can be used to detect subcellular localization of Bp
EMF proteins http://www.psort.org/psortb/. Utilize KEGG
identifiers in conjunction with NCBI gi accession numbers to
discover functions of each of the identified proteins.
Acknowledgments
References
1. Bordet J, Gengou O (1906) Le microbe de la (2012) A gel-free proteomic-based method
coqueluche. Ann Inst Pasteur 20:48–68 for the characterization of Bordetella pertussis
2. Hulbert RR, Cotter PA (2009) Laboratory clinical isolates. J Microbiol Methods
maintenance of Bordetella pertussis. Curr Pro- 90(2):119–133
toc Microbiol 15(4B):1.1–1.9. https://doi. 9. Laemmli UK (1970) Cleavage of structural
org/10.1002/9780471729259.mc04b01s15 proteins during the assembly of the head of
3. Parkhill J, Sebaihia M, Preston A, Murphy LD, bacteriophage t4. Nature 227:685–689
Thomson N, Harris DE et al (2003) Compar- 10. Molloy MP (2008) Isolation of bacterial cell
ative analysis of the genome sequences of Bor- proteins using carbonate extraction. Methods
detella pertussis, Bordetella parapertussis and Mol Biol 424:397–401
Bordetella bronchioseptica. Nat Genet 11. Hulbert RR, Cotter PA (2009) Laboratory
35:32–40 maintenance of Bordetella pertussis. Curr Pro-
4. Matoo S, Cherry JD (2005) Molecular patho- toc Microbiol 15:4B.1.1–4B.1.9
genesis, epidemiology, and clinical manifesta- 12. Yang L, Zhang H, Bruce JE (2009) Optimiz-
tions of respiratory infections due to Bordetella ing the detergent concentration conditions for
pertussis and other Bordetella subspecies. Clin immunoprecipitation (IP) coupled with
Microbiol Rev 18(2):326–382 LC-MS/MS identification of interacting pro-
5. Shrivastava R, Miller JF (2009) Virulence fac- teins. Analyst 134:755
tor secretion and translocation by Bordetella 13. Keller A, Nesvizhskii AI, Kolker E, Aebersold R
species. Curr Opin Microbiol 12(1):88–93 (2002) Empirical statistical model to estimate
6. Tefon B, Maass S, Ozcengiz E, Becher D, the accuracy of peptide identifications made by
Hecker M, Ozcengiz G (2011) A comprehen- MS/MS and database search. Anal Chem
sive analysis of Bordetella pertussis surface pro- 74:5383–5392
teome and identification of new immunogenic 14. Nesvizhskii AI, Keller A, Kolker E, Aebersold R
proteins. Vaccine 29(19):3583–3595 (2003) A statistical model for identifying pro-
7. West R, Whitmon J, Williamson YM, teins by tandem mass spectrometry. Anal Chem
Moura H, Nelson M, Melnick N et al (2012) 75:4646–4658
A rapid method for capture and identification 15. Yu NY, Wagner JR, Laird MR, Mellj G, Rey S,
of immunogenic proteins in Bordetella pertussis Lo R et al (2010) PSORTb 3.0: improved
enriched membranes fractions: a fast-track protein subcellular localization prediction
strategy applicable to other microorganisms. J with refined localization subcategories and pre-
Proteome 75(6):1966–1972 dictive capabilities for all prokaryotes. Bioinfor-
8. Williamson YM, Moura H, Simmons K, matics 26(13):1608–1615
Whitmon J, Melnick N, Woolfitt A et al
Chapter 2
Abstract
Surface proteins are essential molecules for the interplay between cells and the environment. They partici-
pate in many biological processes including transport, adhesion, cell–cell recognition, signaling, and other
cell interactions. In pathogenic microorganisms, these molecules may act as virulence or cytotoxicity
factors. Analyzing the set of surface proteins is critical to understand these processes and to identify possible
targets that can be the starting point for other studies or discoveries (e.g., vaccines or diagnostics). Here I
describe a proteomic procedure to identify in a fast and reliable way a set of surface-exposed proteins in
bacteria, the methodology of which can be adapted to other biological systems (unicellular fungi, parasites).
The protocol presented here involves “shaving” the cells cultured in broth with proteases followed by liquid
chromatography–tandem mass spectrometry (LC/MS/MS) and analysis of the generated peptides. This
method overcomes some important limitations of the first-generation, gel based proteomics techniques,
and the “shaving” approach allows one to identify which domains from identified proteins are more
accessible to proteases. These identified proteins have the highest potential to be recognized by antibodies,
and thus permits the identification of potential epitopes or antigens.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_2, © Springer Science+Business Media, LLC 2018
21
22 Manuel J. Rodrı́guez-Ortega
2 Materials
3 Methods
3.1 “Shaving” 1. Grow the bacterial culture to the desired OD600, normally
Protocol for Gram- corresponding to mid-exponential phase (see Notes 5 and 6).
Positive Bacteria 2. Pellet the bacteria normally by centrifugation at 3500 g,
10 min, 4 C.
3. Resuspend the pelleted bacteria in PBS (see Note 7). Repellet
the bacteria as in step 2.
4. Repeat step 3 two more times.
5. Resuspend the bacterial pellet in PBS/30% sucrose, pH 7.4 in a
1.5 mL low-binding tube at a ratio of 800 μL buffer per each
100 mL of initial bacterial culture.
Protease Shaving of Bacterial Cells 25
3.2 Cleaning Cleaning can be done with a vacuum manifold system or by push-
the “Surfome” ing the liquids through cartridge resin with a 5 mL tip pipette,
with Oasis® HLB using a 5 mL pipette for generating pressure (see Note 12). Accord-
Extraction Cartridges ing to the manufacturer’s instructions, the use of the cartridges
involves the following steps: equilibration, sample loading, wash-
ing, and elution.
1. Equilibrate Oasis® HLB extraction cartridges with 0.6 mL of
80% acetonitrile.
2. Add 0.6 mL of 0.1% formic acid.
3. Load the sample (in our hands, loading 150 μL of the “sur-
fome” of any streptococcal species mixed with 450 μL PBS
works well) (see Note 13).
4. Wash the sample twice with 0.6 mL of 2% acetonitrile/0.1%
formic acid.
5. Elution in three steps with 0.6 mL of each of the following
solutions (see Note 14):
(a) 10% acetonitrile/0.1% formic acid
(b) 20% acetonitrile/0.1% formic acid
(c) 50% acetonitrile/0.1% formic acid
6. Dry in a speed-vacuum system (see Note 15).
7. Resuspend the pellet in 100 μL 2% acetonitrile/0.1% formic
acid (you can divide it among three tubes, or resuspend the first
tube, then transfer the volume to the second one and resus-
pend, and then transfer it to the third one and resuspend).
Keep the sample in a low-binding tube.
8. At this stage, the sample is ready for MS/MS analysis (see
Note 16). Otherwise, it can be stored at 20 C for some
months (see Notes 17 and 18).
26 Manuel J. Rodrı́guez-Ortega
4 Notes
References
1. Navarre WW, Schneewind O (1999) Surface protein antigens in children. J Proteome
proteins of gram-positive bacteria and mechan- 126:228–233
isms of their targeting to the cell wall envelope. 8. Olaya-Abril A, Jimenez-Munguia I, Gomez-
Microbiol Mol Biol Rev 63(1):174–229 Gascon L, Obando I, Rodriguez-Ortega MJ
2. Grandi G (2006) Genomics and proteomics in (2015) A pneumococcal protein array as a plat-
reverse vaccines. Methods Biochem Anal form to discover serodiagnostic antigens
49:379–393 against infection. Mol Cell Proteomics 14
3. Rabilloud T, Chevallet M, Luche S, Lelong C (10):2591–2608
(2010) Two-dimensional gel electrophoresis in 9. Olaya-Abril A, Jimenez-Munguia I, Gomez-
proteomics: past, present and future. J Prote- Gascon L, Rodriguez-Ortega MJ (2014) Sur-
ome 73(11):2064–2077 fomics: shaving live organisms for a fast pro-
4. Wu CC, MacCoss MJ, Howell KE, Yates JR teomic identification of surface proteins. J
3rd (2003) A method for the comprehensive Proteome 97:164–176
proteomic analysis of membrane proteins. Nat 10. Doro F, Liberatori S, Rodriguez-Ortega JJ,
Biotechnol 21(5):532–538 Rinaudo CD, Rosini R, Mora M et al (2009)
5. Rodriguez-Ortega MJ, Norais N, Bensi G, Surfome analysis as a fast track to vaccine discov-
Liberatori G, Capo S, Mora M et al (2006) ery: identification of a novel protective antigen
Characterization and identification of vaccine for Group B Streptococcus hypervirulent strain
candidate proteins through analysis of the COH1. Mol Cell Proteomics 8(7):1728–1737
group A Streptococcus surface proteome. Nat 11. Gomez-Gascon L, Luque I, Olaya-Abril A,
Biotechnol 24(2):191–197 Jimenez-Munguia I, Orbegozo-Medina RA,
6. Olaya-Abril A, Gomez-Gascon L, Jimenez- Peralbo E, Tarradas C, Rodriguez-Ortega MJ
Munguia I, Obando I, Rodriguez-Ortega (2012) Exploring the pan-surfome of Strepto-
MM (2012) Another turn of the screw in coccus suis: looking for common protein anti-
shaving Gram-positive bacteria: optimization gens. J Proteome 75(18):5654–5666
of proteomics surface protein identification in 12. Bohle LA, Riaz T, Egge-Jacobsen W, Skaugen M,
Streptococcus pneumoniae. J Proteome 75 Busk OL, Eijsink VG, Mathiesen G (2011) Iden-
(12):3733–3746 tification of surface proteins in Enterococcus fae-
7. Jimenez-Munguia I, van Wamei WJ, Olaya- calis V583. BMC Genomics 12:135
Abril A, Garcia-Cabrera E, Rodriguez-Ortega 13. Tjalsma H, Lambooy L, Hermans PW, Swin-
MM, Obando I (2015) Proteomics-driven kels DW (2008) Shedding & shaving: disclo-
design of a multiplex bead-based platform to sure of proteomic expressions on a bacterial
assess natural IgG antibodies to pneumococcal face. Proteomics 8(7):1415–1428
Chapter 3
Abstract
We present a protocol for the specific labeling and isolation of proteins from the membrane surface of
endothelial cells and the surrounding extracellular matrix of organs, experimental wounds and tumors using
chicken embryos. Proteins are deglycosylated on streptavidin resin and purified after gentle elution and
trypsin digestion. Peptides are analyzed by spectroscopy and reverse proteomic fingerprinting. The major
advantages of this protocol include reductions in both the background and overrepresentation of single
proteins that would otherwise mask less well-represented proteins in the mass spectroscopy analysis. We also
present methods to identify putative vascular and endothelial cell targets from isolated chicken membranes
and extracellular proteins. The use of human genome and transcriptome data facilitates this analysis.
Human orthologs of isolated chicken proteins are identified using best hit BLAST searches against the
Human Reference Sequence Database. The expression of Human orthologs is then assessed for endothelial
and non-endothelial cell enrichment using second generation RNA-seq sequenced libraries. Scanning of
the published literature then provides a ranking score of those genes most likely involved in cancer or having
a link to angiogenesis.
Key words Proteomics chick embryo, Membrane and matrix proteome, Chorio-allantoic membrane,
Mass spectrometry, Protein mapping, In vivo biotinylation
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_3, © Springer Science+Business Media, LLC 2018
31
32 Witold W. Kilarski et al.
2 Materials
2.3 Organ Lysis 1. Lysis buffer: Prepare a solution composed of 1% sodium dode-
and Purification cyl sulfate (SDS), 0.1% Triton X-100 (Sigma), 5 mM ethylene-
of Biotinylated diaminetetraacetic acid (EDTA) (Sigma), 10 mM dithiothreitol
Peptides (DTT), 20 mM Tris, 150 mM NaCl and adjust pH to 7.5 with
HCl, and add 2 concentrated inhibitor cocktail (see Note 1).
2. Sonicator-homogenizer.
3. Washing buffer: Prepare a solution composed of 1% SDS, 0.1%
Triton X-100 (Sigma), 5 mM EDTA (Sigma), 10 mM iodoa-
cetamide, 20 mM Tris, 150 mM NaCl and adjust the pH to 7.5
with HCl.
4. 1 M iodoacetamide in water (Bio-Rad Laboratories, Hercules,
CA, USA).
5. Streptavidin (ST)-sepharose beads (High Performance)
(GE Healthcare Life Sciences, Marlborough, MA, USA) (see
Note 2).
6. Streptavidin-sepharose washing column: 10 mL syringe,
clogged with cotton gauze secured with perforated socket
and placed in 50 mL tube.
7. Sepharose washing buffer: Prepare a solution composed of
0.1% SDS, 20 mM Tris, 150 mM NaCl and adjust the pH to
7.5 with HCl.
Mapping the Avian Extracellular and Membrane Proteome 35
3 Methods
3.1 Embryo Culture 1. Fertilized brown leghorn eggs should be obtained from a local
farmer.
2. On the day of delivery place eggs horizontally inside the incu-
bator and culture without tilting for 3 days at 38 C and 75%
humidity (see Note 6).
3. On day 4, gently move eggs to a biological safety cabinet (BSC).
4. Crack the shell by hitting the bottom side of the egg on the edge
of an open petri culture dish using a single decisive motion.
5. Gently open the bottom side of the shell and transfer the
contents into the dish, taking special care not to break the
egg yolk (see Notes 7 and 8). Discard the shell and close the
lid. Dishes where the yolk is broken after embryo transfer from
the egg should be immediately discarded as biological waste.
6. Gently and without unnecessary shaking, return the closed
petri dish containing the unbroken egg yolk to the incubator.
7. Continue embryo culture for an additional 7 days.
8. During the incubation period, check the embryos every 3 days
to eliminate dead embryos. This can be determined with the
naked eye while observing the embryos in the incubator with-
out opening the dish, but avoid unnecessary plate movements
during the embryo examinations. Dead embryos loose
completely the red appearance of blood vessels in the yolk
and later CAM. Gently remove and discard dead embryos.
9. To prevent embryo deaths after day 10, 0.5 mL of a 100 mM
calcium carbonate slurry (in water) can be added onto the
embryo CAM. The addition of this slurry to the embryo serves
Mapping the Avian Extracellular and Membrane Proteome 37
3.1.2 Optional Tumor 1. Wash confluent monolayer of tumor cells (e.g., glioblastoma,
Cells Implantation U87 cell line) twice with PBS and trypsinize them for 3 min
at RT.
2. Cells should be then washed twice in cultivation media (4.5 g/
L glucose DMEM with 20 mM HEPES) containing 10%
serum, followed by a third wash with PBS.
3. Dissociate the cells using a trypsin-based solution according to
the protocols provided by ATCC.
4. After centrifugation (3 min at 4 oC, 800 g), the media/
supernatant should be removed by aspiration and discarded.
5. The pellet slurry should be applied directly onto CAM wounds
covered with nylon grid as described above.
6. Return dishes with embryos to the incubator for additional
6 days.
3.1.3 Perfusion and In 1. All procedures can be performed on the bench, and all solu-
Vivo Biotinylation tions should be prewarmed to 37 C.
of Endothelial Surfectome 2. On day 16 of incubation, embryos should be anesthetized by
of Chicken Embryo Organs application of 500 μL of the Anesthesia solution (assuming an
egg weight of approximately 25 g) directly on the CAM surface
(see Note 10).
38 Witold W. Kilarski et al.
3.2 Organ Lysis 1. Pool selected organs from 5 to 15 biotinylated embryos. Use
and Purification 15 or 50 mL conical tubes, depending on organ size.
of Biotinylated 2. Weigh the pooled organs and add 40 μL of lysis buffer per
Proteins milligram of wet tissue. Because tissue with lysis buffer heats
during sonication, tissue together with lysis buffer should not
exceed 10% of maximum tube volume.
3. Sonicate the mixture at room temperature in a sonicator-
homogenizer at medium amplitude settings until it becomes
frothy
4. Boil samples for 15 min at 95 C and after cooling to room
temperature, add 1 M iodoacetamide (to acetylate and perma-
nently protect free thiols) to a final concentration of 20 mM
and leave at room temperature for an additional 30 min.
5. Cool sample at room temperature, but do not place sample on
ice as SDS precipitates below 15 C.
6. Spin sample at 14,000 g for 5 min at room temperature.
7. Collect the soluble fraction and filter through a 0.45 μm
syringe filter (see Note 12).
8. Mix 1 mL of lysate with 100 μL of streptavidin (ST)-linked
beads prewashed twice with lysis buffer supplemented with
iodoacetamide at a final concentration of 20 mM.
9. Incubate with constant rotation (on a Tube Rotator) at room
temperature for 1 h.
10. Quick spin the slurry at 1000 g for 3 min and transfer the
beads with washing buffer to a new 15 mL conical tube.
11. Perform two additional washes with 15 mL washing buffer by
spinning the slurry for 3 min at 1000 g.
12. From this point all equipment used for the washing and elution
procedures of biotinylated proteins must be silanized or the
plasticware should be made of materials characterized by
low-binding affinity.
13. Transfer the bead slurry to a WC using SDS washing buffer.
40 Witold W. Kilarski et al.
3.5 Bioinformatics 1. We recommend that the user work with individuals who are
Procedures to Identify adept in the use of databases to identify endothelium enriched
Endothelium Enriched genes and vascular targets.
Genes and Potential 2. We also provide protocols in the Supplement that are intended
Vascular Targets as a guide for someone with limited bioinformatics experience
who is willing to learn and use a command line based Linux
operating system.
3. The bioinformatics protocols involve four main themes, all of
which are provided in the Supplement to this chapter.
(a) Human ortholog identification of chicken proteins.
(b) Endothelial gene expression assessment of human
orthologs.
(c) Literature abstract scanning and the generation of a rank-
ing score.
(d) Combining results and investigation of the best candidate
genes.
4. These methods were performed using the Ubuntu 12.04.5
OS. It is assumed that a user has access to such a machine
(or similar) and is able to navigate directories and run simple
commands.
5. For further information, tutorials are available online for
Linux, including one at http://www.ee.surrey.ac.uk/Teach
ing/Unix/.
4 Notes
Acknowledgment
Supplementary Materials
Bioinformatics Mass spectrum peak lists are assigned to Uniprot [19] or Refseq
Procedures to Identify [20], chicken proteins using the Mascot Daemon software package
Endothelial Enriched (Matrix Science). FASTA sequences of Uniprot and Refseq proteins
Genes are the starting data. To facilitate identification of endothelial and
putative vascular target genes, the superior annotation and data of
44 Witold W. Kilarski et al.
the human transcriptome and genome are utilized. The first step in
the procedure is to assign putative human orthologs to the chicken
proteins. This is done using the BLAST database search alignment
algorithm [21] as performed previously [22]. This methods finds
the best Human protein hit to a chicken protein, using a stringent
expectation value cutoff of 1e6. This can be accomplished as
follows:
– Identification of chicken proteins human ortholog
will print the full path to the location on the file system (pwd
means present working directory) where the programs are,
and this location should be entered into the accompanying
Perl script.
5. Download and unzip the supplementary files that accom-
pany this article. Edit these paths into the Perl script
provided, called “find_human_chick_ortholog.pl”. The
Ubuntu path variable can also be adjusted to include the
BLAST bin location, by typing
export PATH=$PATH:‘pwd‘
Decompress with
zcat human*protein.faa.gz > human.protein.faa
zcat human*protein.gpff.gz > human.protein.gpff
The size of the data now is ~6.4Gb, so delete any files not
needed with the command;
rm human*protein.faa.gz human*protein.gpff.gz
46 Witold W. Kilarski et al.
(http://hgdownload.soe.ucsc.edu/goldenPath/hg19/
bigZips). Specifically, the FASTA sequences of chromo-
somes are downloaded with
wget --content-disposition http://hgdownload.soe.
ucsc.edu/goldenPath/hg19/bigZips/chromFa.tar.gz
library(edgeR)
# Read in the counts table that was generated by
FeatureCounts into a data frame
dframe<-read.table("Endo_counts.txt", sep¼"\t", head-
er¼T, row.names¼1 skip=1)
# Make a data matrix from the data frame that only
contains the raw read to transcript counts (removes data
not needed)
datamat<-data.matrix(dframe[,-c(1:5)])
# Remove very low expressed genes, with anything >¼ raw
count of 20 across the six samples
counts_small <- datamat[rowSums(datamat)>20,]
# Create a DGElist object that holds the data.
dob <- DGEList(counts ¼ counts_small, group¼c(rep
("nonEndo",3),rep("Endo",3)))
# Calculate the normalization factors to scale the raw
counts of varying library sizes
dob <- calcNormFactors(dob)
# Estimates the common dispersion parameter (see the
edgeR documentation)
dob <- estimateCommonDisp(dob,verbose¼T)
# This function uses empirical Bayes to assess the
tagwise negative binomial dispersions (see the edgeR
manual)
dob <- estimateTagwiseDisp(dob)
# Perform endothelial versus non-endothelial library
contrast, so up regulated genes are enriched in en-
dothelial cells.
diff_ex <- exactTest(dob, pair¼c("nonEndo", "Endo"))
# Make a table of all the differentially expressed
genes results
out_table<-topTags(diff_ex,n¼Inf)[[1]]
write.table(out_table, sep¼"\t", quote¼F, fi-
le¼"Endo_output.exact.test.txt", col.names¼NA)
finish the R session by typing;
q()
type n if prompted to save the session
References
sequencing experiments. Nat Methods for assigning sequence reads to genomic fea-
10:71–73 tures. Bioinformatics 30:923–930
33. Dobin A, Davis CA, Schlesinger F, Drenkow J, 35. Robinson MD, McCarthy DJ, Smyth GK
Zaleski C, Jha S, Batut P, Chaisson M, Gingeras (2010) edgeR: a Bioconductor package for dif-
TR (2013) STAR: ultrafast universal RNA-seq ferential expression analysis of digital gene
aligner. Bioinformatics 29:15–21 expression data. Bioinformatics 26:139–140
34. Liao Y, Smyth GK, Shi W (2014) feature-
Counts: an efficient general purpose program
Chapter 4
Abstract
Human stem cells and their progeny are valuable for a variety of research applications and have the potential
to revolutionize approaches to regenerative medicine. However, we currently have limited tools to permit
live isolation of homogeneous populations of cells apt for mechanistic studies or cellular therapies. While
these challenges can be overcome through the use of immunophenotyping based on accessible cell surface
markers, the success of this process depends on the availability of reliable antibodies and well-characterized
markers, which are lacking for most stem cell lineages. This chapter outlines an iterative process for the
development of new cell surface marker barcodes for identifying and selecting stem cell derived progeny of
specific cell types, subtypes, and maturation stages, where antibody-independent identification of cell
surface proteins is achieved using a modern chemoproteomic approach to specifically identify
N-glycoproteins localized to the cell surface. By taking advantage of a large repository of available cell
surfaceome data, proteins that are unlikely to confer cell type specificity can be rapidly eliminated from
consideration. Subsequently, targeted quantitation by mass spectrometry can be used to refine candidates of
interest, and a bioinformatic visualization tool is key to mapping experimental data to candidate protein
sequences for the purpose of epitope selection during the antibody development phase. Overall, the process
of developing cell surface barcodes for immunophenotyping is iterative and can include multiple rounds of
discovery, refinement, and validation depending on the phenotypic resolution required.
Key words N-glycoproteins, Proteomics, Cell surface, Cell surface protein atlas, Immunophenotyp-
ing, Barcode, Mass spectrometry, Epitope selection, Extracellular domain
1 Introduction
Human stem cells and their progeny are valuable for a variety of
research applications and have the potential to revolutionize thera-
peutic options for treating intractable diseases. Specifically, stem
cells and their progeny can be used to study molecular dynamics
during very early stages of human development that are impossible
to study in vivo [1] and to study potentially toxic effects of
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_4, © Springer Science+Business Media, LLC 2018
57
58 Chelsea M. Fujinaka et al.
Fig. 1 Iterative process for identifying extracellular epitopes and developing cell surface marker barcodes for
live immunophenotyping. In the discovery phase, combining the CSC-Technology with the Cell Surface Protein
Atlas provides a rapid method for identifying proteins that may be informative for identifying or selecting a
specific cell type. Once putative candidates are selected, the refinement phase includes antibody-independent
quantitation by targeted mass spectrometry to further refine those markers that are potentially most
informative. Subsequently, Protter is used to visualize extracellular domains and guide epitope selection for
antibody development. In the validation phase, monoclonal antibodies are validated and functional and
molecular measurements can be performed on the cells identified by the antibodies in order to correlate
the functional phenotype with the cell surface phenotype
2 Materials
2.1 Cell Collection 1. Phosphate buffered saline (PBS, 1): 1 PBS pH 7.4. Add
900 mL water to 100 mL 10 PBS, pH 7.4 (Quality
Biological, Gaithersburg, MD, USA). Store at 4 C.
2. Liberase TH dissociation solution: Liberase TH (Roche Diag-
nostics, Indianapolis, IN, USA) (see Note 1).
3. PBS with 0.1% fetal bovine serum (FBS): 1 PBS pH 7.4, 0.1%
(v/v) FBS. Add 1 mL FBS (Sigma-Aldrich Chemical Company,
Cell Surface Capture Technology 63
St. Louis, MO, USA) to 100 mL 10 PBS and fill to 1 L with
water. Store at 4 C.
4. 15 and 50 mL conical tubes.
5. Low speed centrifuge capable of holding 15 and 50 mL conical
tubes.
2.2 Oxidation 1. Labeling buffer: 1 PBS, 0.1% (v/v) FBS, pH 6.5. Add 500 μL
and Biotinylation FBS to 50 mL 10 PBS, fill to 500 mL with water, and adjust
of Cell Surface the pH to 6.5 using 85% (w/v) H3PO4. Store at 4 C (see
Oligosaccharides Notes 2 and 3).
2. NaIO4 stock solution: 195 mM NaIO4 in labeling buffer.
Weigh 0.01 g NaIO4 (Sigma-Aldrich) in a microcentrifuge
tube, add 240 μL labeling buffer and vortex until dissolved
(see Note 2).
3. Biocytin hydrazide, 25 mg aliquots (Biotium, Hayward, CA,
USA) (see Note 4).
4. Laboratory rocker.
3 Methods
3.1 Collect Cells 1. Begin with a culture of approximately 108 cells (see Note 6). If
and Oxidize working with adherent cells, wash cells with room temperature
Extracellular PBS to remove as many dead cells as possible. If working with
Oligosaccharides cells in suspension, proceed to step 3.
2. Detach cells from the culture plate using a trypsin-free solu-
tion. The choice of this solution may vary by cell type, and can
include collagenases, enzyme free, and/or EDTA solutions.
Scraping may be possible, but this must be first tested with a
small aliquot of cells, and cells should be stained with trypan
blue to evaluate plasma membrane integrity (see Notes 1
and 7).
3. Transfer cells and solution into a 50 mL conical tube. Centri-
fuge at 500 g at 4 C for 3 min to collect cells.
4. Aspirate liquid and gently resuspend cells by first tapping
against benchtop to loosen the pellet. Add 10 mL of cold
PBS with 0.1% FBS and pipette up/down three times (pipette
tip close to wall of tube to break up clumps). Remove a 50 μL
66 Chelsea M. Fujinaka et al.
3.2 Biotinylate 1. Aspirate liquid. Gently resuspend cells by first tapping against
Extracellular the benchtop to loosen the pellet. Add 4 mL cold labeling
Oligosaccharides buffer.
2. Add 1 mL cold labeling buffer to one vial of biocytin hydrazide
(25 mg). Vortex briefly until resuspended. Add all of this to the
cell solution. Use 1 mL of cold labeling buffer to rinse the
biocytin hydrazide vial and subsequently add this to the cell
suspension. The final concentration will be approximately
10 mM biocytin hydrazide (5–10 mM is optimal).
3. Place tube in ice on a rocker to agitate slowly for 60 min.
4. Add PBS with 0.1% FBS up to 50 mL and invert several times
to mix. Centrifuge 500 g at 4 C for 3 min to collect cells.
5. Aspirate liquid. Gently resuspend cells by first tapping against
benchtop to loosen the pellet. Add PBS with 0.1% FBS up to
50 mL. Centrifuge at 500 g at 4 C for 3 min to collect cells.
Repeat for a total of two washes.
6. At this step, either flash-freeze and store at 80 C for short
term storage or proceed with next step directly.
3.3 Lyse Cells, 1. Aspirate PBS, and ensure that all the PBS is removed. Resus-
Remove Nuclei, pend cells in 4 mL hypotonic lysis buffer. Set on ice for 10 min
and Enrich Membranes to swell the cells making it easier to lyse them.
2. Using a serological pipette, transfer cell solution to Orange
Tube (M tube) for GentleMacs. Homogenize cells twice,
using method D01 both times (see Note 8).
3. Centrifuge GentleMacs tube at 800 g at 4 C for 10 min to
remove nuclei/debris. Transfer supernatant to a new 15 mL
conical tube, and save on ice until step 5.
Cell Surface Capture Technology 67
3.5 Glycopeptide 1. Place an empty Mobicol column on the Vacman, using the
Capture and Elution stopcock as shown in Fig. 2a. Rinse Mobicol with
5 500 μL acetonitrile to remove polymer contamination,
then rinse with 5 500 μL 100 mM ammonium bicarbonate.
2. Add 350 μL streptavidin bead slurry to Mobicol column. Wash
beads with 3 500 μL 100 mM ammonium bicarbonate.
68 Chelsea M. Fujinaka et al.
Fig. 2 Vacuum manifold setup. Shown are the configurations for using the Mobicol (A, B) for glycopeptide
capture and washing and the MicroSpin column (C) used for peptide desalting and concentration
3.6 Desalting 1. Cut off approximately 25 mm of a P200 pipette tip and use as
and Concentrating an adaptor to place the MicroSpin column on the vacuum
manifold (Fig. 2c).
2. Rinse column with 5 200 μL 100% HPLC grade acetonitrile
then 10 200 μL HPLC grade 0.1% TFA.
3. Place column into a clean 1.5 mL low-binding
microcentrifuge tube.
4. Add 200 μL of peptide sample to the top of the column. Do
not add any pellet if there is one. Using a P200 pipette, slowly
“push” sample through the column, from top to bottom.
Repeat until all of the sample is through the column twice.
5. Place column on vacuum manifold. Rinse column with
10 200 μL HPLC grade 0.1% TFA, then 10 200 μL
HPLC grade water.
6. Place column into a clean 0.5 mL low-binding microcentrifuge
tube. Add 100 μL of elution solution. Use a P200 pipette,
slowly “push” sample through the column, from top to bot-
tom. Centrifuge tube/column at 1000 g for 30 s to collect
remaining eluent.
7. Place the sample in a SpeedVac or similar instrument until all of
the liquid has evaporated.
8. Resuspend peptides in 12 μL LC-MS grade 0.1% formic acid in
water.
Fig. 3 Exploiting the Cell Surface Protein Atlas to identify proteins of interest. (a) Annotated MS/MS spectrum
of a representative peptide from Integrin Beta 1 that was identified in human pluripotent stem cell derived
72 Chelsea M. Fujinaka et al.
3.8 Prioritization 1. To begin the selection of candidate makers that may be infor-
of Epitopes for Marker mative for a particular cell type, we utilize ProteinCenter to
and Antibody archive all data generated by the CSC-Technology. This work-
Development flow enables comparisons of cell surface N-glycoproteins
observed among various experimental conditions (e.g., cell
types, organ types, and developmental stages). We maintain
an in-house repository of data from three sources: data gener-
ated in-house, data from the Cell Surface Protein Atlas (Atlas
[24]; downloaded from http://wlab.ethz.ch/cspa/), and data
from other publications that may not be contained in the Atlas.
While proteins of interest for distinguishing cellular pheno-
types can be identified by directly including the desired cell
types for comparison within a quantitative CSC-Technology
experiment, the Atlas may still add value as a freely available
repository populated with data from a variety of cell types.
2. To establish a custom database of cell surface N-glycoproteins,
data from the Atlas and other publications are imported into
ProteinCenter via .csv file format. Data generated in-house are
imported as prot.xml files.
3. Once all data are imported into ProteinCenter, select the
desired datasets and create a comparison. At this stage, con-
taminating proteins identified in the experiment can be elimi-
nated by data filtering to keep only those proteins identified by
peptides containing a deamidated asparagine within the con-
served sequence motif (NxS/T/C) for N-glycosylation. To
eliminate redundancy and enable comparisons among datasets
that may have been generated using different protein data-
bases, cluster data within the comparison by “indistinguishable
proteins,” which will group proteins that cannot be
Fig. 3 (continued) cardiomyocytes via the CSC-Technology. One identified site of N-glycosylation (N115) within
the sequence motif is indicated. Spectrum generated using Proteome Discoverer 2.1 (ThermoFisher Scien-
tific). Inset table shows m/z values for fragment ions predicted to result from the proposed peptide sequence
NPCTSEQNCTSPFSYK. Values in red or blue were observed in the MS/MS spectrum. (b) Screenshot of
ProteinCenter repository of CSC-Technology data for 59 human cell types. Color denotes that a protein (one
protein/row) was identified in a particular cell type (one cell type/column). Data are shown for 20 cell surface
N-glycoproteins, including those ubiquitously observed across most cell types, and those that are observed
less commonly among those cell types represented in the Atlas. (c) Hierarchical clustering of 15 human cell
types using 1323 cell surface N-glycoproteins identified via CSC-Technology can successfully cluster similar
cell types
Cell Surface Capture Technology 73
Fig. 4 CSC-Technology data can inform subsequent quantitative studies and antibody development. (a)
Quantitative data for two cell surface proteins, Integrin Beta 1 and Glucose Transporter 3 across three time
points of human pluripotent stem cell derived cardiomyocyte differentiation. Quantitation was achieved using
parallel reaction monitoring to target both proteins in the same experiment and data were analyzed using
Skyline [37]. Top row shows fragment ion chromatograms used for quantitation, and integrated data are
summarized in the chart below. (b) Illustrations of the transmembrane topology of Glypican-1 and Integrin
Beta 1 highlighting the extracellular peptides identified by CSC-Technology, including sites of N-glycosylation
as well as additional annotated sequence features that may be considered during epitope selection for
antibody development. Image generated using Protter
4 Notes
Acknowledgments
References
1. Mummery CL, Zhang J, Ng ES, Elliott DA, non-human primate hearts. Nature 510
Elefanty AG, Kamp TJ (2012) Differentiation (7504):273–277
of human embryonic stem cells and induced 5. Schwartz SD, Hubschman JP, Heilwell G,
pluripotent stem cells to cardiomyocytes: a Franco-Cardenas V, Pan CK, Ostrick RM et al
methods overview. Circ Res 111(3):344–358 (2012) Embryonic stem cell trials for macular
2. Braam SR, Tertoolen L, van de Stolpe A, degeneration: a preliminary report. Lancet 379
Meyer T, Passier R, Mummery CL (2010) Pre- (9817):713–720
diction of drug-induced cardiotoxicity using 6. Bryder D, Rossi DJ, Weissman IL (2006)
human embryonic stem cell-derived cardio- Hematopoietic stem cells: the paradigmatic
myocytes. Stem Cell Res 4(2):107–116 tissue-specific stem cell. Am J Pathol 169
3. Liang P, Lan F, Lee AS, Gong T, Sanchez- (2):338–346
Freire V, Wang Y et al (2013) Drug screening 7. Shizuru JA, Negrin RS, Weissman IL (2005)
using a library of human induced pluripotent Hematopoietic stem and progenitor cells: clin-
stem cell-derived cardiomyocytes reveals ical and preclinical regeneration of the hema-
disease-specific patterns of cardiotoxicity. Cir- tolymphoid system. Annu Rev Med
culation 127(16):1677–1691 56:509–538
4. Chong JJ, Yang X, Don CW, Minami E, Liu 8. Clark G, Stockinger H, Balderas R, van Zelm
YW, Weyers JJ et al (2014) Human embryonic- MC, Zola H, Hart D, Engel P (2016) Nomen-
stem-cell-derived cardiomyocytes regenerate clature of CD molecules from the tenth human
Cell Surface Capture Technology 77
leucocyte differentiation antigen workshop. 20. Ducret A, Kux van Geijtenbeek S, Roder D,
Clin Transl Immunol 5(1):e57 Simon S, Chin D, Berrera M et al (2015) Iden-
9. Turac G, Hindley CJ, Thomas R, Davis JA, tification of six cell surface proteins for specific
Deleidi M, Gasser T, Karaoz E, Pruszak J liver targeting. Proteomics Clin Appl 9
(2013) Combined flow cytometric analysis of (7–8):651–661
surface and intracellular antigens reveals sur- 21. Wollscheid B, Bausch-Fluck D, Henderson C,
face molecule markers of human neuropoiesis. O’Brien R, Bibel M, Schiess R, Aebersold R,
PLoS One 8(6):e68519 Watts JD (2009) Mass-spectrometric identifi-
10. Dubois NC, Craft AM, Sharma P, Elliott DA, cation and relative quantification of N-linked
Stanley EG, Elefanty AG, Gramolini A, Keller cell surface glycoproteins. Nat Biotechnol 27
G (2011) SIRPA is a specific cell-surface (4):378–386
marker for isolating cardiomyocytes derived 22. Apweiler R, Hermjakob H, Sharon N (1999)
from human pluripotent stem cells. Nat Bio- On the frequency of protein glycosylation, as
technol 29(11):1011–1018 deduced from analysis of the SWISS-PROT
11. Kelly OG, Chan MY, Martinson LA, Kadoya K, database. Biochim Biophys Acta 1473(1):4–8
Ostertag TM, Ross KG et al (2011) Cell- 23. Gahmberg CG, Tolvanen M (1996) Why
surface markers for the isolation of pancreatic mammalian cell surface proteins are glycopro-
cell types derived from human embryonic stem teins. Trends Biochem Sci 21(8):308–311
cells. Nat Biotechnol 29(8):750–756 24. Bausch-Fluck D, Hofmann A, Bock T, Frei AP,
12. Bye CR, Jonsson ME, Bjorklund A, Parish CL, Cerciello F, Jacobs A et al (2015) A mass
Thompson LH (2015) Transcriptome analysis spectrometric-derived cell surface protein
reveals transmembrane targets on transplanta- atlas. PLoS One 10(3):e0121314
ble midbrain dopamine progenitors. Proc Natl 25. Boheler KR, Bhattacharya S, Kropp EM,
Acad Sci U S A 112(15):E1946–E1955 Chuppa S, Riordon DR, Bausch-Fluck D et al
13. Cordwell SJ, Thingholm TE (2010) Technol- (2014) A human pluripotent stem cell surface
ogies for plasma membrane proteomics. Prote- N-glycoproteome resource reveals markers,
omics 10(4):611–627 extracellular epitopes, and drug targets. Stem
14. Elschenbroich S, Kim Y, Medin JA, Kislinger T Cell Rep 3(1):185–203
(2010) Isolation of cell surface proteins for 26. Domon B, Gallien S (2015) Recent advances in
mass spectrometry-based proteomics. Expert targeted proteomics for clinical applications.
Rev Proteomics 7(1):141–154 Proteomics Clin Appl 9(3–4):423–431
15. Kitchen P, Day RE, Salman MM, Conner MT, 27. Omasits U, Ahrens CH, Muller S, Wollscheid B
Bill RM, Conner AC (2015) Beyond water (2014) Protter: interactive protein feature visu-
homeostasis: diverse functional roles of mam- alization and integration with experimental
malian aquaporins. Biochim Biophys Acta proteomic data. Bioinformatics 30
1850(12):2410–2421 (6):884–886
16. Danzer C, Eckhardt K, Schmidt A, 28. Mallanna SK, Cayo MA, Twaroski K, Gundry
Fankhauser N, Ribrioux S, Wollscheid B et al RL, Duncan SA (2016) Mapping the cell-
(2012) Comprehensive description of the surface N-glycoproteome of human hepato-
N-glycoproteome of mouse pancreatic beta- cytes reveals markers for selecting a homoge-
cells and human islets. J Proteome Res 11 neous population of iPSC-derived hepatocytes.
(3):1598–1608 Stem Cell Rep 7(3):543–556
17. Tyleckova J, Valekova I, Zizkova M, 29. Kropp EM, Bhattacharya S, Waas M, Chuppa
Rakocyova M, Marsala S, Marsala M, Gadher SL, Hadjantonakis AK, Boheler KR, Gundry
SJ, Kovarova H (2016) Surface RL (2014) N-glycoprotein surfaceomes of
N-glycoproteome patterns reveal key proteins four developmentally distinct mouse cell
of neuronal differentiation. J Proteome types. Proteomics Clin Appl 8(7–8):603–609
132:13–20 30. Gundry RL, Riordon DR, Tarasova Y,
18. Sun B, Ma L, Yan X, Lee D, Alexander V, Chuppa S, Bhattacharya S, Juhasz O et al
Hohmann LJ et al (2013) N-glycoproteome (2012) A cell surfaceome map for immunophe-
of E14.Tg2a mouse embryonic stem cells. notyping and sorting pluripotent stem cells.
PLoS One 8(2):e55722 Mol Cell Proteomics 11(8):303–316
19. Hofmann A, Thiesler T, Gerrits B, Behnke S, 31. Bausch-Fluck D, Hofmann A, Wollscheid B
Sobotzki N, Omasits U et al (2015) Surfa- (2012) Cell surface capturing technologies for
ceome of classical Hodgkin and non-Hodgkin the surfaceome discovery of hepatocytes.
lymphoma. Proteomics Clin Appl 9 Methods Mol Biol 909:1–16
(7–8):661–670
78 Chelsea M. Fujinaka et al.
32. Hofmann A, Gerrits B, Schmidt A, Bock T, 37. MacLean B, Tomazela DM, Shulman N,
Bausch-Fluck D, Aebersold R, Wollscheid B Chambers M, Finney GL, Frewen B et al
(2010) Proteomic cell surface phenotyping of (2010) Skyline: an open source document edi-
differentiating acute myeloid leukemia cells. tor for creating and analyzing targeted proteo-
Blood 116(13):e26–e34 mics experiments. Bioinformatics 26
33. Schiess R, Mueller LN, Schmidt A, Mueller M, (7):966–968
Wollscheid B, Aebersold R (2009) Analysis of 38. Eng JK, Jahan TA, Hoopmann MR (2013)
cell surface proteome changes via label-free, Comet: an open-source MS/MS sequence
quantitative mass spectrometry. Mol Cell Pro- database search tool. Proteomics 13(1):22–24
teomics 8(4):624–638 39. Craig R, Beavis RC (2004) TANDEM: match-
34. Kall L, Canterbury JD, Weston J, Noble WS, ing proteins with tandem mass spectra. Bioin-
MacCoss MJ (2007) Semi-supervised learning formatics 20(9):1466–1467
for peptide identification from shotgun prote- 40. Keller A, Nesvizhskii AI, Kolker E, Aebersold R
omics datasets. Nat Methods 4(11):923–925 (2002) Empirical statistical model to estimate
35. Taus T, Kocher T, Pichler P, Paschke C, the accuracy of peptide identifications made by
Schmidt A, Henrich C, Mechtler K (2011) MS/MS and database search. Anal Chem 74
Universal and confident phosphorylation site (20):5383–5392
localization using phosphoRS. J Proteome 41. Nesvizhskii AI, Keller A, Kolker E, Aebersold R
Res 10(12):5354–5362 (2003) A statistical model for identifying pro-
36. Dorfer V, Pichler P, Stranzl T, Stadlmann J, teins by tandem mass spectrometry. Anal Chem
Taus T, Winkler S, Mechtler K (2014) MS 75(17):4646–4658
Amanda, a universal identification algorithm
optimized for high accuracy tandem mass spec-
tra. J Proteome Res 13(8):3679–3684
Chapter 5
Abstract
Enrichment of the plasma membrane represents one valuable method to characterize the surfaceome, along
with other plasma membrane and structural proteins. Currently, the overlapping densities of many subcel-
lular organelles hinder enrichment of the plasma membrane by centrifugation. However, external access to
the plasma membrane of intact cells allows the attachment of a nanoparticle pellicle to enhance its density
and facilitate enrichment. We describe the synthesis of iron oxide nanoparticles, attachment of the pellicle to
suspended cells, and recovery of plasma membrane proteins for proteomic analysis.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_5, © Springer Science+Business Media, LLC 2018
79
80 Rebecca L. Rose et al.
Table 1
Densities of subcellular organelles (adapted from ref. 8)
2 Materials
2.1 Iron Oxide 1. For the preparation of iron oxide nanoparticles prepare
Nanoparticle 0.5 mM FeCl3·6H2O and 1.5 mM sodium acetate
Construction (C2H3NaO2) in 25 mL of propylene glycol.
2. For coating the iron oxide particles with alumina, prepare
0.01 M Al(NO3)3/0.1 M K(NO3) as a 10 mL aqueous
solution.
3. Glassware required: 100 mL round bottom flask with a 24/40
ground glass joint (catalog number UX-34706-10; Cole-
Parmer, Vernon Hills, IL, USA) connected to the condenser
via a female 24/40 ground glass joint.
4. Centrifugal Filters (Cat No. UFC500396, Millipore, Billerica,
MA, USA).
2.3 Tissue Culture 1. Human multiple myeloma cells (ATCC RPMI 8226).
Supplies 2. Tissue culture incubator with CO2 regulator.
3. Refrigerated tissue culture compatible centrifuge for 15 and
50 mL tubes.
4. 75 cm2 cell culture flask.
5. RPMI 1640 medium.
6. Fetal Bovine Solution (FBS).
7. Hemocytometer.
8. Light microscope (200 magnification).
9. 50 mL conical tube.
10. 3 mL syringe with 21G needle.
3 Methods
3.1 Iron Oxide 1. To begin the nanoparticle preparation dilute FeCl3·6H2O and
Nanoparticle sodium acetate in 25 mL of propylene glycol to a final concen-
Construction [6] tration of 0.5 mM and 1.5 mM, respectively.
(See Fig. 1) 2. Add 500 μL of deionized water to this mixture and reflux for
12 h in a 100 mL round bottom flask at room temperature; be
sure to fit a condenser to the round bottom flask via a female
24/40 ground glass joint. Particles will form (see Note 2).
3. Collect and wash the particles four times with water via centri-
fugation (8500 g for 30 s) in Millipore centrifugal filter.
4. Resuspend the particles in 10 mL of 0.01 M Al(NO3)3/0.1 M
K(NO3) for 12 h at room temperature on a rocking platform.
5. Collect the particles via centrifugation (8500 g for 10 min),
discard the supernatant, and resuspend the pellet in PMCBA
for use in cell coating and pellicle construction.
Nanoparticles to Enrich Plasma Membrane Proteins 83
Fig. 3 Scanning electron microscope images. (a) Human multiple myeloma cell, (b) cell coated with alumina-
coated iron oxide nanoparticles, (c) PAA cross-linked coated cell, (d) fragment of pellicle-coated plasma
membrane (modified from ref. 5)
3.4 Preparation 1. Precipitate 100 μg of protein from the extraction sample using
of Samples for High a standard chloroform–methanol procedure (see Note 18).
Performance Liquid 2. Resuspend the protein pellet in 100 μL 8 M urea and 50 mM
Chromatography NH4HCO3.
(HPLC)-Mass 3. Reduce protein sulfhydryl groups by adding 2 μL of 1 M DTT
Spectrometry stock solution for a final concentration of 20 mM DTT. Incu-
(See Note 17) bate the sample at 56 C for 20 min in a water bath.
4. Alkylate protein thiol groups by adding 4 μL of the 1 M IAA
stock solution to the sample for a final concentration of 40 mM
IAA. Incubate at room temperature for 20 min.
5. Add protease Lys-C to a final concentration of 1:50 enzyme–-
substrate and incubate at 37 C for 3 h.
6. Following Lys-C digestion dilute the approximately 8 M urea
solution to 1.6 M urea with 50 mM NH4HCO3.
7. Add trypsin to a final concentration of 1:25 enzyme–substrate
and incubate at 37 C for 16 h.
8. Desalt and concentrate samples using C18 spin columns.
Recover peptides in 85% acetonitrile (see Notes 19 and 20).
9. Freeze samples at 80 C to prepare for lyophilization.
10. Lyophilize samples until the final volume is negligible and
resuspend in 100 μL of 0.1% formic acid for analysis via
HPLC interfaced to a tandem mass spectrometer.
4 Notes
Acknowledgment
References
1. Chaney LK, Jacobson BS (1983) Coating cells 5. Choksawangkarn W, Kim SK, Cannon JR,
with colloidal silica for high yield isolation of Edwards NJ, Lee SB, Fenselau C (2013) Enrich-
plasma membrane sheets and identification of ment of plasma membrane proteins using nano-
transmembrane proteins. J Biol Chem particle pellicles: comparison between silica and
258:10062–10072 higher density nanoparticles. JProteome Res
2. Stolz DB, Jacobson BS (1992) Examination of 12:1134–1141
transcellular protein polarity of bovine aortic 6. Bai X, Son SJ, Zhang S, Liu W, Frank JA,
endothelial cells in vitro using the cationic col- Venkatesan T, Lee SB (2008) Synthesis of super-
loidal silica microbead membrane-isolation pro- paramagnetic nanotubes as MRI contrast agents
cedure. J Cell Sci 103:39–51 and for cell labeling. Nanomedicine 3:163–166
3. Rabar AM, Fenselau C (2004) Integration of 7. Choksawangkarn W, Graham LM, Burke M, Lee
Jacobson’s pellicle method into proteomic stra- SB, Ostrand-Rosenberg S, Fenselau C, Edwards
tegies for plasma membrane proteins. J Prote- N (2016) Peptide-based systems analysis of
ome Res 3:1267–1277 inflammation induced myeloid-derived suppres-
4. Durr E, Yu J, Krasinska K, Carver LA, Yates JR, sor cells reveals diverse signaling pathways. Pro-
Testa JE, Oh P, Schnitzer J (2004) Direct pro- teomics 16:1881–1888
teomic mapping of the lung microvascular endo- 8. Lodish H, Berk A, Zipursky SL, Matsudaira P,
thelial cell surface in vivo and in cell culture. Nat Baltimore D, Darnell J (2000) Molecular cell
Biotechnol 22:985–992 biology, 4th edn. W.H. Freeman, New York
Chapter 6
Abstract
Secreted proteins are of tremendous biological interest since they can act as ligands for receptors to activate
downstream signalling cascades or be used as biomarkers if altered abundance is correlated with a specific
pathological state. Proteins can be secreted either as soluble molecules or as part of extracellular vesicles
(i.e., exosomes or microvesicles). The complete proteomic profiling of secretomes requires analysis of
secreted proteins and extracellular vesicles. Hence, the method described here enriches for microvesicles,
exosomes, and secreted proteins from conditioned media using differential centrifugation. The three
fractions are then analyzed by mass spectrometry-based proteomics for in-depth characterization and
comparison of the protein secretome of cell lines.
Key words Extracellular vesicles, Exosomes, Microvesicles, Secreted proteins, Secretome, Proteo-
mics, Mass spectrometry
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_6, © Springer Science+Business Media, LLC 2018
91
92 Ankit Sinha et al.
Fig. 1 Workflow for the proteomic characterization of MVs, exosomes, and secreted proteins from CM. (a)
Conditioned media is obtained through 48 h incubation in serum-free media. (b) MVs, exosomes, and secreted
proteins are obtained through differential centrifugation of the CM. (c) Protein content of all fractions is
analyzed using LC-MS/MS based proteomics
2 Materials
2.4 TFE Based 1. Denaturation buffer: 50% (v/v) TFE in PBS (pH 7.4).
Digestion of Proteins 2. Bicinchoninic acid (BCA) assay for protein determination.
96 Ankit Sinha et al.
2.5 Preparation 1. Desalting tips: OMIX C18 pipette tips (Agilent Technologies,
of Samples for Mass Santa Clara, CA, USA).
Spectrometry 2. Vacuum centrifuge.
3. Priming solution: 80% acetonitrile and 0.1% formic acid in
water.
4. Washing solution: 0.1% formic acid in water.
5. Elution solution: 60% acetonitrile and 0.1% formic acid in
water.
3 Methods
3.1 Obtaining The suggested volumes are for monolayer adherent cells growing in
Conditioned Media 15 cm dishes. All cells are incubated at 37 C in 5% CO2 condition.
1. Culture cells in ten 15-cm dishes using appropriate cell culture
medium, beginning with a confluence of 30–40% (see Note 3).
2. Cells should be cultured to a confluence level where they can be
projected to reach 70–80% confluence within the next 48 h (see
Note 4).
3. Once the adhered cells have reached the predetermined con-
fluence level; aspirate and discard culture media and gently
wash the cells three times with 10 mL of room
temperature PBS.
4. Add 20 mL of secretome media to each plate and incubate at
37 C for 48 h.
5. After 48 h, carefully aspirate and pool together the conditioned
media (using a 25 mL serological pipette) from the ten plates in
four 50 mL conical tubes. Store conditioned media in an ice
bath at 4 C.
Profiling of Secreted Proteins, Exosomes and Microvesicles 97
3.3 Digestion 1. Perform BCA assay to determine the total amount of protein
of Proteins present in the MVs, exosome and conditioned media fractions.
2. Aliquot the conditioned media based on protein amount
equating to total protein in the exosome and MV fraction (see
Note 7).
3. Add an equal volume of TFE to the conditioned media (see
Note 8).
4. Heat denature all the samples at 60 C for 120 min.
5. Reduce proteins by adding DTT to a final concentration of
5 mM. Incubate at 60 C for 30 min.
98 Ankit Sinha et al.
3.5 Analysis Below we describe the proteomics approach that has provided good
of Peptides Using results in our hands. Briefly, in bottom-up MS driven shotgun
Tandem Mass proteomics, digested peptides are analyzed using liquid chroma-
Spectrometry tography coupled to tandem mass spectrometry (LC-MS/MS).
This involves separation of peptides based on hydrophobicity (use
of reverse phase is highly recommended) using a nano-flow ultra-
performance liquid chromatography system (such as Easy-nLC
1000) coupled to a heated 50-cm column packed with 2-μm Pep-
Map C18 particles. The liquid chromatography system is
connected to a high-resolution Orbitrap tandem mass spectrometer
(such as QExactive). Considering that LC-MS/MS systems from
different vendors will have their own specific settings, the method
described below should be used as guidelines.
1. Add 10–13 μL of Buffer A to reach a sample final volume of
~15 μL.
2. Measure total peptide amount using a NanoDrop.
Profiling of Secreted Proteins, Exosomes and Microvesicles 99
4 Notes
In our hands, this protocol has produced data with high precision
and robustness. We recently used this exosome isolation method
for quantitative comparisons across various ovarian cancer cell lines
[21]. We strongly advise the use of MS grade reagents for Subhead-
ing 3.3 and onward. The following notes are provided as a sugges-
tion for users of this protocol.
1. Phenol red is used as a pH indicator in cell culture media. Cells
are cultured in media without phenol red for only 48 h, and
hence pH should not drastically change over such a short time
period.
100 Ankit Sinha et al.
Acknowledgment
References
1. Hanahan D, Weinberg RA (2011) Hallmarks 10. Yoon YJ, Kim OY, Gho YS (2014) Extracellular
of cancer: the next generation. Cell vesicles as emerging intercellular communica-
144:646–674 somes. BMB Rep 47:531–539
2. Tlsty TD, Coussens LM (2006) Tumor stroma 11. Beach A, Zhang HG, Ratajczak MZ, Kakar SS
and regulation of cancer development. Annu (2014) Exosomes: an overview of biogenesis,
Rev Pathol 1:119–150 composition and role in ovarian cancer. J Ovar-
3. Schiarea S, Solinas G, Allavena P, Scigliuolo ian Res 7(1):14
GM, Bagnati R, Fanelli R, Chiabrando C 12. Raposo G, Stoorvogel W (2013) Extracellular
(2010) Secretome analysis of multiple pancre- vesicles: exosomes, microvesicles, and friends. J
atic cancer cell lines reveals perturbations of key Cell Biol 200:373–383
functional networks. J Proteome Res 13. Saleem SN, Abdel-Mageed AB (2014) Tumor-
9:4376–4392 derived exosomes in oncogenic reprogram-
4. Xu BJ, Yan W, Jovanovic B, An AQ, Cheng N, ming and cancer progression. Cell Mol Life
Aakre ME et al (2010) Quantitative analysis of Sci 72:1–10
the secretome of TGF-beta signaling-deficient 14. Peinado H, Aleckovic M, Lavotshkin S,
mammary fibroblasts. Proteomics Matei I, Costa-Silva B, Moreno-Bueno G et al
10:2458–2470 (2012) Melanoma exosomes educate bone
5. Mathias RA, Wang B, Ji H, Kapp EA, Moritz marrow progenitor cells toward a
RL, Zhu HJ, Simpson RJ (2009) Secretome- pro-metastatic phenotype through MET. Nat
based proteomic profiling of Ras-transformed Med 18:883–891
MDCK cells reveals extracellular modulators of 15. Lu M, Huang B, Hanash SM, Onuchic JN,
epithelial-mesenchymal transition. J Proteome Ben-Jacob E (2014) Modeling putative thera-
Res 8:2827–2837 peutic implications of exosome exchange
6. Luo X, Liu Y, Wang R, Hu H, Zeng R, Chen H between tumor and immune cells. Proc Natl
(2011) A high-quality secretome of A549 cells Acad Sci U S A 7:E4165–E4174
aided the discovery of C4b-binding protein as a 16. Lundholm M, Schroder M, Nagaeva O,
novel serum biomarker for non-small cell lung Baranov V, Widmark A, Mincheva-Nilsson L,
cancer. J Proteome 74:528–538 Wikstrom P (2014) Prostate tumor-derived
7. Oksvold MP, Neurauter A, Pedersen KW exosomes down-regulate NKG2D expression
(2015) Magnetic bead-based isolation of exo- on natural killer cells and CD8þ T cells: mech-
somes. Methods Mol Biol 1218:465–481 anism of immune evasion. PLoS One 9:
8. Thery C, Zitvogel L, Amigorena S (2002) Exo- e108925
somes: composition, biogenesis and function. 17. Luga V, Zhang L, Viloria-Petit AM, Ogunjimi
Nat Rev Immunol 2:569–579 AA, Inanlou MR, Chiu E et al (2012) Exo-
9. Huang X, Yuan T, Tschannen M, Sun Z, somes mediate stromal mobilization of auto-
Jacob H, Du M et al (2013) Characterization crine Wnt-PCP signaling in breast cancer cell
of human plasma-derived exosomal RNAs by migration. Cell 151:1542–1556
deep sequencing. BMC Genomics 14:319 18. Shimoda M, Principe S, Jackson HW, Luga V,
Fang H, Molyneux SD et al (2014) Loss of the
102 Ankit Sinha et al.
Timp gene family is sufficient for the acquisi- 23. Baietti MF, Zhang Z, Mortier E, Melchior A,
tion of the CAF-like cell state. Nat Cell Biol Degeest G, Geeraerts A et al (2012) Syndecan-
16:889–901 syntenin-ALIX regulates the biogenesis of exo-
19. Valenti R, Huber V, Filipazzi P, Pilla L, somes. Nat Cell Biol 14:677–685
Sovena G, Villa A et al (2006) Human tumor- 24. Ebert MP, Korc M, Malfertheiner P, Rocken C
released microvesicles promote the differentia- (2006) Advances, challenges, and limitations in
tion of myeloid cells with transforming growth serum-proteome-based cancer diagnosis. J
factor-beta-mediated suppressive activity on T Proteome Res 5:19–25
lymphocytes. Cancer Res 66:9290–9298 25. Eichelbaum K, Winter M, Berriel Diaz M,
20. Aliotta JM, Sanchez-Guijo FM, Dooner GJ, Herzig S, Krijgsveld J (2012) Selective enrich-
Johnson KW, Dooner MS, Greer KA et al ment of newly synthesized proteins for quanti-
(2007) Alteration of marrow cell gene expres- tative secretome analysis. Nat Biotechnol
sion, protein production, and engraftment into 30:984–990
lung by lung-derived microvesicles: a novel 26. Cox J, Mann M (2008) MaxQuant enables
mechanism for phenotype modulation. Stem high peptide identification rates, individualized
Cells 25:2245–2256 p.p.b.-range mass accuracies and proteome-
21. Sinha A, Ignatchenko V, Ignatchenko A, wide protein quantification. Nat Biotechnol
Mejia-Guerrero S, Kislinger T (2014) 26:1367–1372
In-depth proteomic analyses of ovarian cancer 27. Deutsch EW, Mendoza L, Shteynberg D,
cell line exosomes reveals differential enrich- Farrah T, Lam H, Tasman N et al (2010) A
ment of functional categories compared to the guided tour of the Trans-Proteomic Pipeline.
NCI 60 proteome. Biochem Biophys Res Proteomics 10:1150–1159
Commun 445:694–701 28. Zhang J, Xin L, Shan B, Chen W, Xie M, Yuen
22. Lee TH, D’Asti E, Magnus N, Al-Nedawi K, D et al (2012) PEAKS DB: de novo sequencing
Meehan B, Rak J (2011) Microvesicles as med- assisted database search for sensitive and accu-
iators of intercellular communication in can- rate peptide identification. Mol Cell Proteo-
cer--the emerging science of cellular ‘debris’. mics 11:M111.010587
Semin Immunopathol 33:455–467
Part II
Abstract
The cation-dependent mannose 6-phosphate receptor (CD-MPR) is a single-pass type I membrane protein.
This protein functions to transport lysosomal enzymes displaying phosphomannosyl residues from the
Golgi complex and the cell surface to the lysosome. This glycosylated protein contains three disulfide
bridges in its 159-residue extracytoplasmic domain. One of the problems with studying eukaryotic mem-
brane proteins is isolating sufficient quantities. Structural studies typically require several hundred milli-
grams of highly purified protein. Here we present a method to isolate milligram quantities of CD-MPR/
Asn81 suitable for structural studies.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_7, © Springer Science+Business Media, LLC 2018
105
106 Linda J. Olson and Nancy M. Dahms
2 Materials
2.1 Instruments 1. MaxQ™ 4000 orbital benchtop shaker (Thermo Fisher Scien-
and Other General tific, Waltham, MA, USA) or similar.
Materials 2. Standard tissue culture hood.
3. Inverted microscope.
4. EVOS® FL Auto Imaging System.
5. ÄKTA Start chromatography system (GE Healthcare Life
Sciences, Pittsburgh, PA, USA).
6. NanoDrop 2000c (Thermo Fisher Scientific, Waltham, MA,
USA).
7. Branson 450 or similar sonicator .
8. Low speed centrifuge capable of holding a JLA8.1000 rotor
and a JA-20 rotor or similar (Beckman Coulter, Indianapolis,
IN, USA).
9. Ultracentrifuge capable of holding a Ti-75 rotor or similar
(Beckman Coulter).
10. 20 mL syringe and large bore (20G) needle.
Transmembrane Proteins from Sf9 Cells 107
2.2 Insect Cells Sf9 Expression Systems (Expression Systems, Davis, CA, USA)
(www.ExpressionSystems.com) (see Note 1).
2.3 Media 1. ESF 921 Medium: This is a protein-free insect cell medium
and Transfection appropriate for cell propagation as well as protein production
Reagents (www.ExpressionSystems.com).
2. BestBac 2.0 v-cath/chiA Deleted Baculovirus Cotransfection
Kit: This kit contains the viral DNA backbone, transfection
medium and transfection reagent (www.ExpressionSystems.
com).
2.4 Vectors 1. pVL1392: This transfer vector may be obtained from several
and Plasmid sources including Expression Systems (www.
Purification expressionsystems.com).
2. PureYield™ Plasmid Miniprep System (Promega, Madison,
WI, USA): Transfection efficiency is dependent on quality of
the plasmid DNA (see Note 2).
3 Methods
3.1 Cloning Gene Although companies have developed different vectors for expres-
of Interest sing foreign proteins in Sf9 cells, we have chosen to remain with
one of the early transfer vectors, pVL1392. This transfer vector
3.1.1 Modification contains features for growth and maintenance in standard E. coli
of pVL1392 strains such as DH5α and JM109. This 9.8 kb plasmid contains the
pMB1 origin of replication from pUC19 for high copy number
growth. Sandwiched between the 50 and 30 -viral sequences required
for homologous recombination is the very strong polyhedron
promoter, which drives recombinant protein expression. This vec-
tor is lacking one key feature needed for recombinant protein
expression: a cleavable signal sequence which directs the recombi-
nant protein to the endoplasmic reticulum for entry into the secre-
tory pathway, an essential targeting signal for secreted and
membrane-bound proteins. To overcome this limitation, we have
chosen to use the honeybee melittin signal peptide: 5-
0
-MKFLVNVALVFMVVYISYIYA-30 (Fig. 1) [6].
3.1.2 Cloning CD-MPR The DNA encoding the signal peptide was synthesized and
into Modified pVL1392 digested as indicated in Fig. 1. Following successful incorporation
of the DNA encoding the signal sequence, the plasmid was digested
with EcoRI and XbaI and treated with FAST-alkaline phosphatase
(AP). The purified PCR product encoding CD-MPR/Asn81 was
also digested with the above two restriction enzymes. The digests
were subjected to standard electrophoresis on a 1% low melting
point agarose gel and the desired bands excised. Standard cloning
procedures were followed and final clones were subjected to DNA
sequencing to verify the sequence.
Fig. 1 Schematic diagram depicting modifications made to the standard pVL1392 transfer vector. The DNA
sequence encoding the melittin signal peptide (translated DNA sequence shown in red) along with a octa-His
tag and GS from an added restriction site (blue) and CD-MPR (black) has been modified to remove
glycosylation at all but one (N81Q) position that was previously determined to be critical for proper folding [5]
Transmembrane Proteins from Sf9 Cells 109
3.2 Culturing Sf9 Sf9 cells obtained in suspension culture were propagated and fro-
Insect Cells zen as directed by the company. Cells were propagated by thawing a
1 mL vial of 50 106 cells at 37 C and diluting the cells into a
250 mL sterile storage bottle containing 30 mL ESF 921 medium.
1. Cells are grown at 27 C in suspension cultures while shaking at
140 rpm in a MaxQ™ 4000 orbital benchtop shaker.
2. Cells were seeded in ESF 921 medium at a density of
0.75–1 106cells/mL in either 250 mL or 1 L sterile Corn-
ing® storage bottles. The 250 mL bottles contained
30–100 mL of liquid culture while the 1 L bottles contained
400 mL suspension culture (see Note 3).
3. Cells were passaged when they reached a density of
4–6 106cells/mL (typically every 2–3 days).
3.3 Transfection The transfection protocol is based on the published protocol from
of Sf9 Cells Expression Systems with modifications to utilize a 24-well Falcon
plate. All steps are carried out in a standard tissue culture hood
using sterile technique.
1. Seed cells at a density of 1.8 105 cells/well of a 24-well
Falcon plate. Allow the cells to adhere to the plate ~30 min
while transfection solutions are made (see Note 4).
2. Combine gently in a polypropylene tube (solution A) 0.4 μg of
CD-MPR/pVL1392, 1 μL BestBac 2.0, v-cath/chiA Deleted
linearized viral DNA, 20 μL Transfection Medium.
3. Combine together in a polypropylene tube (solution B) 1–2 μL
supplied transfection reagent and 20 μL Transfection Medium.
4. Let the solutions sit for 5 min in the hood.
5. Combine solution B into A and incubate in the tissue culture
hood for ~30 min at room temperature.
6. Add 160 μL Transfection Medium to the tube containing the
combined solutions A and B.
7. Gently remove the medium from the plated adherent cells and
replace with the solution from step 6.
8. Place tray in a tight-sealing, small plastic container and incubate
at 27 C 4–5 h (see Note 5).
9. Remove the transfection mix and feed cells with 600 μL ESF
921.
10. Place tray back in plastic container being sure it is sealed and
incubate at 27 C without shaking.
11. Collect medium containing P-0 virus after 5 days, centrifuge
5 min at 500 g to remove any cells (see Note 6).
110 Linda J. Olson and Nancy M. Dahms
3.5 Titering of Virus Determination of the optimal infection conditions for protein
Stocks expression is critical. The amount of virus stock required to infect
a given number of cells will vary from batch to batch of virus as well
as with the age of the viral stock. The amount of virus stock needed
to infect a given number of Sf9 cells is determined by infecting a
series of 30 mL cultures each containing 1 106 cells/mL.
1. Seed four 250 mL Corning® bottles with 30 mL of culture
containing 1 106 cells/mL.
2. Leave one bottle uninfected as a control and infect the other
three bottles with increasing volumes of virus stock: 25, 50 and
250 μL is typical.
3. Count the cells 3 and 4 days post infection (see Note 3) and
determine which flask(s) did not grow after infection. This
determination will indicate how much virus needs to be
added to arrest cell growth (see Note 8).
3.6 Expression The key to protein production in large quantities is having a large
and Purification of Full number of cells to infect at the same time (see Note 9). This
Length CD-MPR procedure involves two independent steps involving infection of
Sf9 cells and protein purification.
1. For the infection of Sf9 cells, count cells and infect with the
amount of virus determined above to allow one doubling (see
Note 10).
2. Cultures are grown as before with shaking at 140 rpm at 27 C.
It is important to monitor cell density, determined through
counting, until the culture has stopped growing.
3. Cells are ready for harvest 48 h after they have stopped divid-
ing, which typically occurs on the third day post-infection.
Transmembrane Proteins from Sf9 Cells 111
Fig. 2 Immunodetection of CD-MPR in Sf9 cells following infection with recombinant baculovirus. Infected and
uninfected control Sf9 cells grown in suspension culture are allowed to adhere 30 min to a 24-well plate. After
attachment, cells are washed with PBS and fixed by incubation on ice for 20 min in the presence of 10%
formalin in PBS. Cells were rinsed with PBS plus 0.1% BSA before being incubated overnight at 4 C in the
dark in a solution of PBS plus 0.1% BSA and a 1:100 dilution of anti-CD-MPR (NMD) [4]. Cells were then rinsed
with PBS plus 0.1% BSA two times prior to incubation with a 1:500 chicken anti-rabbit Alexa 864 antibody.
Control, uninfected cells showed no detectable levels of fluorescence and are not shown. Images were taken
using a 40 objective, the bar representing 100 μm
Infected cells appear larger, but cell death should be less than
10% (Fig. 2).
4. Cells are harvested by centrifugation at 1000 g for 15 min in
a JLA8.1000 rotor. Cell pellets can be either frozen at 80 C
for future use or processed immediately.
5. For protein purification, cell pellets from 1.2 L of Sf9 culture
are resuspended in 150 mL of cold lysis buffer: 20 mM Tris,
pH 7.8 at 4 C, 300 mM NaCl, and 0.1 mM PMSF.
6. Cells are lysed by three 10 s bursts of sonication on ice (see
Note 11).
7. Cell debris is removed by centrifugation at 500 g for 10 min
in a JA-20 rotor.
8. The supernatants are collected and centrifuged at 80,000 g
for 60 min in a Ti-75 rotor (Beckman Coulter).
9. The membrane pellets are resuspended in 100 mL of wash
buffer: 20 mM Tris, pH 7.5 at 22 C, 150 mM NaCl.
112 Linda J. Olson and Nancy M. Dahms
3.7 Preliminary We have used several standard techniques to evaluate if the length
Characterization of solubilization and subsequent longer exposure to higher deter-
of Purified Protein gent concentrations had any effect on the quality or quantity of the
transmembrane protein, CD-MPR/Asn81, being isolated. As
described here, four steps are involved: (1) SDS-PAGE analysis,
(2) Protein quantification, (3) Treatment with PNGaseF, and
(4) SPR analysis of binding affinity.
1. Following elution from the HisTrap excel Ni column, the
results of the purification are analyzed by SDS-PAGE. As this
is a standard laboratory protocol, details are not provided.
2. Proteins were visualized after staining with Coomassie Blue
(Fig. 3) (see Note 12).
3. For Protein Quantification, the Bio-Rad protein assay solution
is used to measure total protein concentration.
4. For this, water (800 μL) is aliquoted into borosilicate glass tubes.
5. An aliquot (10 μL) of the100 mM eluate (Subheading 3.6, step
17) is added to the water.
Transmembrane Proteins from Sf9 Cells 113
Fig. 5 SPR studies characterizing the interaction between CD-MPR/Asn81 and GAA. Increasing concentrations
(10, 20, 40, 80, 120, 200, and 400 nM) of the CD-MPR/Asn81 protein are flowed over the GAA monoester
coupled surface for 120 s followed by buffer and regeneration with 10 mM M6P. The response at equilibrium
(Req) of the sensorgrams are plotted versus the concentration of protein and fit to a 1:1 binding isotherm
(inset). Data from the 1 h detergent solubilization protocol are shown. The time of solubilization with 1% Triton
X-100/0.1% sodium deoxycholate did not significantly affect binding affinity, and resulted in similar kD values
of 100 and 81 nM, respectively, for 1 h and overnight incubation times
Transmembrane Proteins from Sf9 Cells 115
4 Notes
1. Sf9 cells isolated from the ovarian tissue of the fall army worm,
Spodoptera frugiperda, were selected for their faster growth rate
and ability to reach higher densities than some of the other
established lepidopteran insect cell lines. This cell line is able to
be maintained in monolayer cultures for transfections as well as
in shake culture for protein production. They are suitable for
use in all phases of protein generation from transfection, to
virus propagation, to protein production [8].
2. This kit from Promega has provided very reliable results.
3. Cells are counted using a standard hemocytometer. Cells are
split on Mondays and Wednesdays at 1 106 cells/mL. Cells
are split at a lower density of 0.75 106 cells/mL on Fridays to
avoid overgrowth by Monday.
4. In order to ensure cells are at ~75% confluence, plate wells at
the calculated number of cells and also 1.5 and 2 times the
calculated number of cells.
5. If nonshaking incubator is not available to incubate the plate, a
shaking floor incubator can be utilized provided some type of
stable, nonmoving platform can be installed. In the MaxQ™
4000 orbital benchtop shaker, enough stationary floor space is
provided to rest the legs of a homemade metal shelf. This shelf
is tall enough to allow bottles to rest on the shaking platform
underneath.
6. Failure to remove cells may result in higher cell densities and
lower infection rates.
7. In our hands, viral stocks grown in this manner and having
these final cell counts will yield a virus stock of 1 109 virus
units/mL.
8. Also note which flask doubled in cell number since it is often
advantageous to allow the cells to double once after infection in
order to increase cell numbers and accordingly protein yields.
9. If six bottles, each with 400 mL of cells at 4 106 cells/mL,
are needed on Friday and today is Monday, counting back,
halving the volume each day until the current day will deter-
mine how many cells are needed (Friday: 6 400, Thursday:
3 400, Wednesday: 1.5 400, Tuesday: 300 mL, Monday:
150 mL at 4 106 cells/mL today).
10. If your 25 μL virus inoculum resulted in no cell growth then
(30 mL (1 106 cells/mL))/0.025 mL ¼ 1.25 109 infect-
ing units/mL.
11. The extent of cell disruption was evaluated by pipetting 20 μL
onto a transparent surface and viewing by a light microscope.
116 Linda J. Olson and Nancy M. Dahms
Acknowledgment
References
1. Dahms NM, Hancock MK (2002) P-type lec- cation-dependent mannose 6-phosphate recep-
tins. Biochim Biophys Acta 1572:317–340 tor: effects on ligand binding, intracellular tar-
2. Ghosh P, Dahms NM, Kornfeld S (2003) Man- getting and association with binding
nose 6-phosphate receptors: new twists in the immunoglobulin protein. Biochem J
tale. Nat Rev Mol Cell Biol 4:202–213 295:841–848
3. Roberts DL, Weix DJ, Dahms NM, Kim J-JP 6. Tessier DC, Thomas DY, Khouri HE, Laliberte
(1998) Molecular basis of lysosomal enzyme VY (1991) Enhanced secretion from insect cells
recognition: three-dimensional structure of the of a forgein protein fused to the honeybee melit-
cation-dependent mannose 6-phosphate recep- tin signal peptide. Gene 98:177–183
tor. Cell 93:639–648 7. Chavez CA, Bohnsack RN, Kudo M, Gotschall
4. Dahms NM, Brzycki-Wessell MA (1995) RR, Canfield WM, Dahms NM (2007) Domain
Expression and characterization of functional 5 of the cation-independent mannose
bovine cation-dependent mannose 6-phosphate 6-phosphate receptor preferentially binds phos-
receptors in baculovirus-infected insect cells. phodiesters (mannose 6-phosphate N-acetylglu-
Arch Biochem Biophys 317:497–503 cosamine ester). Biochemistry 46:12604–12617
5. Zhang Y, Dahms NM (1993) Site-directed 8. Jarvis D (2009) Baculovirus-insect cell expres-
removal of N-glycosylation sites in the bovine sion systems. Methods Ezymol 463:191–222
Chapter 8
Abstract
The common strategy for making bispecific antibodies (BsAbs) involves combining the variable domains of
the desired monoclonal antibodies (mAbs) into a single bispecific structure. Bispecific immunotherapeutics
has generated many different formats of BsAbs including chemical heteroconjugation of two complete
molecules or fragments of monoclonal antibodies, quadroma, F(ab)2, diabodies, tandem diabodies, and
single-chain antibodies (scFv). This chapter describes the process of generating activated T cells and arming
T cells with heteroconjugated BsAbs to target cancer cells.
Key words Bispecific antibodies, Monoclonal antibodies, Immunotherapy, Armed T cells, Cancer
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_8, © Springer Science+Business Media, LLC 2018
117
118 Archana Thakur et al.
2 Materials
2.1 Tumor Cell Lines 1. Obtain SK-BR-3 (see Note 1), a Her2/neu-positive breast
adenocarcinoma cancer lines and Raji (see Note 2), and a
Her2/neu-negative Burkitt’s lymphoma (as a negative control)
from a commercial source (e.g., ATCC, Rockville, MD, USA).
2. Prepare Iscove’s modified Dulbecco’s medium (IMDM) (Life
Technologies, Grand Island, NY, USA) to culture and maintain
the SK-BR-3 cell line by adding 2 mM L-glutamine, 10% fetal
bovine serum (FBS), and 1% penicillin/streptomycin
antibiotics.
3. Prepare RPMI-1640 medium (Life Technologies, Grand
Island, NY, USA), culture and maintain the Raji cell line by
adding 2 mM L-glutamine, 10% fetal bovine serum (FBS)
(BioWhittaker, Walkersville, MD, USA), and 1% penicillin/
streptomycin antibiotics.
4. Trypsin–EDTA solution (0.05%) (Life Technologies).
Bispecific Antibodies 119
2.2 T-Cell Activation, 1. Obtain Ficoll-Hypaque for isolation of peripheral blood mono-
Expansion nuclear cells by density gradient centrifugation.
and Cryopreservation 2. Dilute whole blood 1:1 with phosphate buffered saline (PBS),
pH 7.4.
3. Prepare Complete RPMI-1640 medium (Life Technologies)
by supplementing with 10% FBS, 2 mM L-glutamine, 1% peni-
cillin/streptomycin and store it at 4 C.
4. Prepare 100 μg/mL Muromonab-CD3 (OKT3) stock in ster-
ile water and store it at 4 C.
5. Prepare 1 106 IU/mL interleukin-2 (IL-2) stock (Chiron,
Emeryville, CA, USA) and (see Note 3) in sterile water and
store it at 4 C.
6. Obtain trypan blue to determine viability by trypan blue
exclusion.
7. Obtain cell culture grade dimethyl sulfoxide (DMSO) for
freezing.
3 Methods
3.1 Generation 1. Slowly overlay (see Note 4) 1:1 PBS diluted blood from a
and Cryopreservation normal healthy donor on Ficoll (one part Ficoll and two parts
of Activated T Cells diluted blood) to isolate PBMC by Ficoll-Hypaque density
gradient centrifugation.
2. Centrifuge for 20 min (400 g) with the brake OFF.
3. Harvest PBMC (See Note 5) at the interface of the PBS and
Ficoll layer into a fresh tube.
4. Add PBS to fill the tube to wash the cells.
5. Centrifuge cell suspension 4–5 min (300–400 g) at 4 C,
discard supernatant.
6. Resuspend the cell pellet in complete RPMI-1640 and perform
a cell count (See Note 6) and viability analysis.
7. Culture PBMC in RPMI-1640 into culture flasks adjusting
concentration 1 106 cells/mL.
8. Activate T cells in the PBMC with 20 ng/mL OKT3 from
stock OKT3 solution (100 μg/mL).
9. Bring IL-2 concentration to 100 IU/mL in the final volume.
10. Expand for 14 days (see Note 7) in the presence of 100 IU/mL
of IL-2 in RPMI-1640 supplemented with 10% FCS
[7, 8]. Adjust cell concentration to 1 106 cells/mL along
with adding 100 IU/mL of IL-2 as T cells proliferate.
11. On day 14, collect ATC in 50 mL conical tubes and centrifuge
at 400 g for 10 min at room temperature to pellet the cells,
decant culture supernatant.
12. Cryopreserve ATC by control rate freezing in medium contain-
ing 10% DMSO and 90% FBS.
3.3 Flow Cytometry 1. Take 1 106 activated T cells in 100–150 μL of 1% BSA in PBS
to Detect BsAb Binding buffer in two separate 5 mL polystyrene round bottom flow
to Effector Cells tube and add 10 μL of each FITC or PE-conjugated anti-CD3
and anti-CD4 antibody in one tube and 10 μL of each FITC or
Bispecific Antibodies 121
Fig. 1 Targeted killing of cells by bispecific antibody armed T cells. This figure illustrates heteroconjugation of
anti-CD3 antibody with anti-Her2 antibody to produce bispecific antibody (BsAb) capable of binding to
activated T-cells (ATC) and targeted killing of tumor
3.4 Detection of Dual 1. Detach SK-BR-3 (an adherent cell line) cells from the plate
Binding Specificity either by scraping or by enzymatic dissociation using trypsin.
of OKT3 anti-Her2 (a) By scraping: Remove culture supernatant, wash with 1%
Bispecific Antibody BSA in PBS then use scraper to dislodge the adherent
cells.
(b) By trypsinization: Add 1–5 mL 0.05% trypsin–EDTA
solution depending on the size of the culture flask, for
25 cm flask add 1 mL 0.05% trypsin–EDTA solution;
incubate at 37 C for 3 min followed by adding 10%
FBS in PBS to stop enzymatic reaction.
2. Collect cells in a conical tube, centrifuge at 450 g for 5 min.
3. Re-suspend pellet in 1 mL of flow buffer; count and adjust cell
concentration to 1 106 cells in 100 μL flow buffer.
122 Archana Thakur et al.
Fig. 2 Flow cytometry analysis of BsAB binding. (a) Binding of ATC armed with OKT3 anti-Her2 to ATC. A
total of 1 106 ATC were armed with 50 ng of OKT3 anti-Her2BsAb. Antibody bound to the surface of ATC
was detected with directly conjugated goat anti-mouse specific antibodies and analyzed by flow cytometry as
percent positive cells, 96% ATC show binding to BsAb. (b) A total of 1 106 SK-BR-3 were armed with 50 ng
of OKT3 anti-Her2BsAb. Antibody bound to the surface of SK-BR-3 was detected with directly conjugated
goat anti-mouse specific antibody and analyzed by flow cytometry show 92% binding of BsAb
3.5 In Vitro 1. Resuspend target SK-BR-3 cells at 0.4 106 cells/mL (total of
Cytotoxicity Assay 10 mL to make one 96-well plate.
to Assess the Function 2. Plate target cells in triplicate at 4 104 target cells/well in
of BsAb Armed ATC 100 μL volume in a flat-bottomed microtiter plate and incubate
Against Adherent Cells overnight at 37 C.
3. Prepare 100 μCi of 51Cr (see Note 10) containing culture
medium (20 μL of stock solution at 5 mCi/mL) and replace
the culture media in the SK-BR-3 plate after overnight incuba-
tion with 51Cr containing media and incubate the plate for
additional 4 h at 37 C.
4. Decant media in appropriate disposal container; wash cells
2 and add 100 μL complete IMDM media in each well.
Bispecific Antibodies 123
5. Add ATC armed (see Note 11) with OKT3 anti-Her2 BsAb
as effector cells in 100 μL volume at effector to target ratios
(E/T) of 25:1, 12.5:1, 6.25:1, and 3.12:1. Incubate overnight
at 37 C. Set-up spontaneous and total release controls in each
plate.
6. Spontaneous Release: Incubate target cells alone (replace effec-
tor cells by adding 100 μL of medium in triplicate).
7. Total Release: Incubate target cells with 2% SDS (replace effec-
tor cells by 100 μL of 2% SDS).
8. After completion of incubation, centrifuge plate and collect
100 μL aliquot of the supernatant to detect radioactivity on a
COBRA-II gamma counter, output of radioactivity is displayed
in counts per minute (cpm).
9. Calculate the percentage of specific lysis using the standard
formula [(experimental spontaneous release)/(maximum
load spontaneous release) 100] and expressed as the
mean of triplicate samples.
3.6 In Vitro 1. For nonadherent Raji cells, label 4 106 cells with 100 μCi of
Cytotoxicity Assay
51
Cr (20 μL of stock solution at 5 mCi/mL) for 1 h at 37 C.
to Assess the Function 2. Wash cells 2 and re-suspend in RPMI-1640 medium.
of BsAb Armed ATC
3. Add 104 target cells/well of a round-bottom 96-well plate in
Against Nonadherent 100 μL RPMI-1640.
Cells
4. Add ATC armed (see Note 11) with OKT3 anti-CD20 BsAb
as an effector cells in 100 μL volume at effector to target ratios
(E/T) of 25:1, 12.5:1, 6.25:1, and 3.12:1.
5. Incubate plate for 4 h at 37 C. Set-up spontaneous and total
release controls in each plate. And follow steps 6–9 as above for
cytotoxicity against adherent targets.
4 Notes
References
1. Baselga J, Tripathy D, Mendelsohn J, Multinational study of the efficacy and safety
Baughman S, Benz CC, Dantis L et al (1996) of humanized anti-HER2 monoclonal anti-
Phase II study of weekly intravenous recombi- body in women who have HER2-
nant humanized anti-p185HER2 monoclonal overexpressing metastatic breast cancer that
antibody in patients with HER2/neu- has progressed after chemotherapy for meta-
overexpressing metastatic breast cancer. J Clin static disease. J Clin Oncol 17:2639–2648
Oncol 14:737–744 3. Lundin J, Kimby E, Bjorkholm M, Broliden
2. Cobleigh MA, Vogel CL, Tripathy D, Robert PA, Celsing F, Hjalmar V et al (2002) Phase
NJ, Scholl S, Fehrenbacher L et al (1999) II trial of subcutaneous anti-CD52
Bispecific Antibodies 125
treatment with autologous T lymphocytes T cells and low dose IL-2, after stem cell trans-
retargeted by a bispecific monoclonal antibody. plant for high risk non-Hodgkin’s lymphoma: a
J Natl Cancer Inst 87:1463–1469 pilot study. Bone Marrow Transplant 49
25. Bolhuis RLH, Lamers CHJ, Goey SH, Egger- (73–9):2014
mont AMM, Trimbos JBMZ, Stoter G et al 28. Lum LG, Thakur A, Liu Q, Deol A,
(1992) Adoptive immunotherapy of ovarian Al-Kadhimi Z, Ayash L et al (2013) CD20-
carcinoma with BSMAb-targeted lymphocytes: targeted T cells after stem cell transplantation
a multicenter study. Int J Cancer 7:78–81 for non-Hodgkin lymphoma. Biol Blood Mar-
26. Renner C, Bauer S, Sahin U, Jung W, van row Transplant 19:925–933
Lier R, Jacobs G, Held G, Pfreundschuh M 29. Baeuerle PA, Kufer P, Lutterbuse R (2003)
(1996) Cure of disseminated xenografted Bispecific antibodies for polyclonal T-cell
human Hodgkin’s tumors by bispecific mono- engagement. Curr Opin Mol Ther 5:413–419
clonal antibodies and human T cells: the role of 30. Loffler A, Kufer P, Lutterbuse R, Zettl F, Dan-
human T-cell subsets in a preclinical model. iel PT, Schwenkenbecher JM et al (2000) A
Blood 87:2930–2937 recombinant bispecific single-chain antibody,
27. Lum LG, Thakur A, Pray C, Kouttab N, CD19 x CD3, induces rapid and high
Abedi M, Deol A, Colaiace WM, Rathore R lymphoma-directed cytotoxicity by unstimu-
(2014) Multiple infusions of CD20-targeted lated T lymphocytes. Blood 95:2098–2103
Chapter 9
Abstract
Human pluripotent stem cells (hPSCs) have great potential for use in regenerative medicine and cell
replacement therapies; however, prior to clinical application, cultured cell populations need to be screened
to ensure the quality of the culture, as well as the capacity of these pluripotent cells to differentiate into
desired cell types. Flow cytometry, utilizing antibodies recognizing targets restricted to the hPSC surfa-
ceome, offers an invaluable tool for high-throughput validation of hPSC lines. Here we describe the
immunophenotyping of live human embryonic stem cell (hESC, H9) and human induced pluripotent
stem cell (hiPSC, KB3) lines by flow cytometry using a panel of antibodies identified as either stem cell
reference markers (CD90, EpCam) or reported as being prevalent or restricted (c-Kit, HPI-1, Integrin α6,
Semaphorin-6A) to these cells. The protocols described here with hPSCs are also applicable to differen-
tiated hPSC progeny and should be instrumental in the immunophenotyping and isolation of well-defined
homogeneous cell populations useful in regenerative medicine.
Key words Embryonic stem cells, Induced pluripotent stem cells, Surfaceome, Immunophenotyping,
Flow cytometry, Antibody
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_9, © Springer Science+Business Media, LLC 2018
127
128 Daniel R. Riordon and Kenneth R. Boheler
2 Materials
Table 1
List of antibodies (primary and secondary) used in the current protocols
Amount of Amount of
primary secondary
antibody per antibody per
Antigen Isotype 1 106 cells Secondary 1 106 cells
(clone) Fluorochrome Company control in 100 μL anti body in 100 μL
CD49f PE eBioScience Rat 0.13 μg N/A N/A
(GoH3) IgG2a
CD90 PE BD Mouse 4.0 μg N/A N/A
Pharmingen IgG1
CD117 PE Bio Legend Mouse 0.25 μg N/A N/A
(104D2) IgG1
CD326 Alexa Fluor Bio Legend Mouse 0.25 μg N/A N/A
(9C4) 647 IgG2b
HAI-1 PE eBioScience Mouse 0.25 μg N/A N/A
(9B10) IgG2a
SEMA6A R&D Systems Goat 0.5 μg Donkey 0.125 μg
IgG anti-goat
IgG-PE
2.5 Antibody 1. Antibodies. See Table 1 for antibodies used in the current
Labeling of Cell protocol.
Surface Antigens 2. Isotype Controls. See Table 1 for Isotype controls used in the
current protocol.
3. Borosilicate glass disposable 900 pipette.
4. Phosphate Buffered Saline (PBS), pH 7.4, without calcium,
without magnesium.
5. Cell Staining Solution for Flow Cytometry (see item 5 of
Subheading 2.4).
6. Round-Bottom polystyrene tube: 5 mL.
7. Serological Pipettes: 5 and 10 mL.
8. Human Trustain FcX™ Fc Blocking Solution (Catalog number
422301; BioLegend, San Diego, CA, USA).
9. Platform Rocker.
10. 10% normal goat serum in 1 PBS Secondary Antibody Block-
ing Solution (Catalog number 50062Z; ThermoFisher Scien-
tific, Waltham, MA, USA).
Flow Cytometry and Immunophenotyping 133
2.7 Flow Cytometry 1. The current protocol used a FACSCanto II flow cytometer
Analysis (BD Biosciences, Franklin Lakes, NJ, USA).
2. Single peak Rainbow beads (catalog number RFP-30-5A;
Spherotech Inc., Green Oaks, IL, USA).
3. Compensation beads (catalog number 557640; Beckton Dick-
inson Immunocytometry Systems).
4. 0.5% paraformaldehyde (PFA).
3 Methods
The protocols described here are applicable for both flow cytome-
try analysis and sorting of hPSC (see Note 8). All procedures should
be performed using aseptic techniques in a Biological Safety Cabi-
net/Tissue Culture Hood unless otherwise noted.
3.1 Coating Cell 1. Slowly thaw 1 aliquot of hESC qualified matrix (Matrigel®) at
Culture Plates 4 C for 30 min.
with hESC Qualified 2. Transfer 25 mL of cold DMEM/F12 medium to a 50 mL
Matrix centrifuge tube on ice.
3. Add an aliquot of hESC qualified matrix to 25 mL of chilled
DMEM/F12 medium and mix well immediately before coat-
ing plates (see Note 9).
4. Add 2 mL hESC-qualified Matrix in culture medium per
9.5 cm2 culture well (one well of a 6-well plate) growth areas.
5. Swirl plate to ensure even coating, and store at 4 C for Matri-
gel or at 37 C for Geltrex. Before using, allow hESC-qualified
Matrix coated plates to set, covered, for 30 min at room
temperature (see Notes 10 and 11).
6. Immediately prior to plating cells, aspirate the excess hESC
qualified matrix (see Note 12).
3.4 Titration All steps should be performed on ice and samples protected from
of Antibodies light.
for Percent Positive For Fluorochrome-conjugated Primary Antibodies:
Measurements 1. Determine the concentration and volume of the antibody stock
solutions and recommended antibody concentration for use in
flow cytometry analysis from the manufacturer’s product data
sheet (see Note 25).
2. For each antibody and isotype control, number 6–8 microcen-
trifuge tubes and place on ice.
136 Daniel R. Riordon and Kenneth R. Boheler
Table 2
Antibody serial dilution scheme
Volume and
Cell wash source of Working Working stock Ab staining Concentration in
Tube solution primary Ab stock concentration reaction volume staining reaction
no. volume (μL) (μL) dilution (μg/mL) (μL/106 cells) (μg/106 cells)
1 0 30 of stock 0 200 20 4.0
2 20 20 of tube 2 100 20 2.0
1 dilution
3 20 20 of tube 4 50 20 1.0
2 dilution
4 20 20 of tube 8 25 20 0.5
3 dilution
5 20 20 of tube 16 12.5 20 0.25
4 dilution
6 20 20 of tube 32 6.25 20 0.125
5 dilution
7 20 20 of tube 64 3.125 20 0.0625
6 dilution
8 20 20 of tube 128 1.5625 20 0.03125
7 dilution
Example of a serial dilution scheme for a PE conjugated CD90 antibody with a master stock concentration of 0.2 mg/mL
for staining 1 105 cells
3.5 Antibody All steps should be performed on ice and samples protected from
Labeling of Cell light.
Surface Antigens
1. Resuspend cells prepared in Subheading 3.3 in cold Wash
on Live Cells Solution so that final concentration of cells is 1 106 total
cells in 95 μL. Using a P200 pipette, gently triturate to disag-
gregate cells.
2. Block cells by adding 5 μL Human Trustain FcX™ Fc Blocking
Solution for every 1 106 cells and mix by gently flicking tube
with finger, then incubate for 10 min on ice, gently rocking.
3. Gently triturate cell solution using P200 pipette to ensure
homogeneous mixture of cells then aliquot 1 106 cells
(100 μL) per tube into 5 mL round bottom polystyrene tubes
on ice (see Note 28).
4. Add primary antibody or isotype control at its optimal concen-
tration (as determined in Subheadings 3.4 and 3.8) then incu-
bate for 60 min on ice with gentle rocking (see Note 29).
Include an unstained control.
5. Add 3 mL cold Wash Buffer, then collect cells by centrifugation
at 200 g for 5 min at 4 C.
6. Aspirate solution being careful not to disturb cell pellet.
7. Repeat washing steps 5 and 6 for a total of two washes follow-
ing antibody labeling.
8. If a primary antibody directly conjugated to a fluorochrome is
used, proceed directly to Subheading 3.6. Continue as follows
for labeling with a secondary antibody conjugated to a
fluorochrome.
9. Resuspend cells in 100 μL secondary antibody blocking solu-
tion using a P200 pipette with gentle trituration.
10. Add secondary antibody, gently tap tube to mix, and then
incubate for 30 min on ice, gently rocking.
11. Add 3 mL cold Wash Buffer, then collect cells by centrifugation
at 200 g for 5 min at 4 C.
12. Aspirate solution being careful not to disturb cell pellet
13. Repeat washing steps 11 and 12 for a total of two washes after
secondary antibody labeling.
3.7 Flow Cytometry 1. Prior to running samples, alignment beads and calibration
Analysis standards should be run to ensure that the data are reproduc-
ible from day to day. This may be routinely run by an experi-
enced user or in a core facility (if so, go to step 16 below), but if
not, then beads should be run to ensure good instrument
performance.
2. Place diluted single peak Rainbow beads onto the sample inser-
tion tube and initiate data acquisition.
3. While observing FSC versus SSC and all other fluorescence
parameter combinations, adjust the instrument according to
the manufacturer’s instructions to ensure the narrowest CV
with the highest signal intensity. Tolerance ranges are estab-
lished for the coefficient of variance and fluorescent intensity as
well as all instrument parameters.
4. Adjust the voltages for each photomultiplier tube (PMT) to
achieve the predetermined intensity levels for the bead
population.
5. Collect and save all single parameter histograms for subsequent
analyses.
6. The signal-to-background ratio (S/B) is obtained by using the
eight peak beads (i.e., the median channel of each peak) divided
by the median channel. Each PMT will have a characteristic
S/B, and the correct tolerance plotted against time should be
in the range of 10%.
7. If multiple fluorophores are run, then compensation beads
(mouse anti-K beads) will also need to be run as described in
https://www.bdbiosciences.com/documents/BD_
FACSDiva_setup_system.pdf.
8. Latex beads coated with anti-mouse K antibody are used with
each antibody conjugate to determine the compensation
matrix for polychromatic flow cytometry. Into a conical tube
add 40 μL of compensation beads and the volume of previously
Flow Cytometry and Immunophenotyping 139
Fig. 1 Gating strategy for selective live cells. Dot blots showing light scatter profiles of hiPSC and hESC gated for
the selection of live cells and exclusion of debris. In this instance, cells have not been stained for viability, and all
cells are included, excluding those with a very low SSC value. These likely correspond to dead or damaged cells.
50,000 events were collected and the gated population used for determination of cells positive for marker proteins
140 Daniel R. Riordon and Kenneth R. Boheler
and eliminate debris and cell aggregates (see Fig. 1) (see Note
33). If there are multiple isotype controls we begin with
unstained controls and then optimize settings to maximize
data acquisition of all isotype controls tested.
18. Ideally, the scatter plots should show an equal distribution of
cells distributed along a 45 angle relative to the ordinate and
abscissa.
19. Visualize the cell distribution of isotype controls and
fluorochrome-labeled cells to determine threshold values to
limit the number of events acquired by the flow cytometer. If
using total counts, then the threshold value is low and most
events will be acquired; however, population gating may be
useful to eliminate events such as cell debris and dead cells (see
Note 34).
20. Adjust the voltages to ensure that the signals with the isotype
control can be readily visualized. Ideally, the peak signal will be
well defined when viewed as a histogram, and it will have an
even distribution, and a signal on the Fluorochrome log scale
that is minimal.
21. Using fluorochrome-labeled cells, check to see if there is a
signal above that seen with the isotype control. Normally, we
test each antibody singly on a specific cell type before attempt-
ing any multilabeling experiments. Establish optimum baseline
PMT gains (see Note 35) to maximize resolution. Use the
minimum intensity required to achieve a histogram of fluores-
cence, which clearly displays both left and right edges of the
peak(s).
22. Record settings, as this will be valuable for future repeat experi-
ments. We recommend multiple repeats with an n > 4 for each
cell surface protein examined.
23. Once these thresholds have been determined, maintain the
laser voltage settings of each fluorochrome when analyzing
each corresponding antibody labeled sample. It is inappropri-
ate to alter these voltages during data acquisition among sam-
ples, either when determining optimal antibody dilutions or
when performing analyses.
24. Collect a minimum of 10,000 events; however, a higher acqui-
sition may be needed for multicolor analyses.
25. Acquire data using flow cytometry for each antibody tested;
however, it may be necessary to adjust laser voltage settings for
each fluorochrome using the appropriate isotype controls.
26. If multicolor parameters are assessed, then the voltage settings
should be set to maximize data acquisition.
27. If displaying multiple parameters with multiple fluorochromes,
then 2D, 3D, and other plots may be necessary for analyzing
Flow Cytometry and Immunophenotyping 141
Fig. 2 Titration of CD90-PE antibody using H9 hESC. The data shown here reflect titration results from pilot
tests with three independent antibodies. Data from the antibody showing the best results are shown, but at
three concentrations. Histograms of CD90 in H9 cells illustrating that the optimal signal to noise ratio (S/N) is
achieved using 4.0 μg antibody. MFI median fluorescence intensity, S/N MFI positive/MFI negative
3.9 Analysis of Data 1. Data will need to be exported and analyzed using software.
There are a number of software packages that can be used, and
we recommend that you discuss with your Flow Cytometry
Core, which package may be apt. Examples include FCS
Express, FloJo, and free software from Flowing Software.
142 Daniel R. Riordon and Kenneth R. Boheler
4 Notes
Count
Count
CD90
85.7% 86.0%
Count
Count
EpCam (CD326)
C-Kit (CD117)
49.5% 89.7%
Count
Count
Integrin α6 (CD49f)
74.9% 90.2%
Count
Count
SEMA6A
84.1% 90.8%
Count
Count
HAI-1
Isotype Control
Anbody
hiPSC(KB3) hESC (H9)
Fig. 3 Characterization of hiPSC and hESC by flow cytometry. Histograms of live KB3 hiPSC and H9 hESC
showing populations positive for (a) Stem cell reference markers and (b) markers of pluripotency
144 Daniel R. Riordon and Kenneth R. Boheler
30. The small volume may take several minutes to wet the entire
area of the strainer. This step can be performed during final
wash/centrifugation steps.
31. Straining of cells is necessary to eliminate large aggregates that
could clog the flow cytometer. Tubes can be tapped gently on
the benchtop to aid the collection of cell solution through
strainer and into the tube. Return cells to ice as quickly as
possible.
32. Forward-scattered light (FSC) is proportional to cell surface
area or cell size. Side-scattered light (SSC) is proportional to
cell granularity or internal complexity.
33. Dead cells can nonspecifically bind many antibody conjugates.
These signals can erroneously be counted as positively labeled
cells; therefore, gating strategies should be used to gate out
these cells. Intercalated dyes like propidium iodide (PI) and live
cell assays like Calcein AM can be useful for identifying and
gating dead cells.
34. Preliminary tests should be run to determine whether a cell
type can be effectively labeled using a specific antibody. For this
type of analysis, we usually use the cells from a single well of a
6-well plate and test the antibody using the manufacturer’s
recommendations. Antibody dilutions must then be per-
formed with more cells to optimize conditions (see Subhead-
ings 3.4 and 3.8).
35. Optimal baseline PMT gains need to be established empirically
at the outset, and should be determined based on the advice of
a FC Core facility or with the help of someone who is well
acquainted with flow cytometry.
36. It is possible for multiple parameters/signals to be measured;
however, if multiple fluorochromes are being assessed simulta-
neously, then quenching must be considered. Contact a FC
Core or experienced user for assistance.
37. If using a fluorochrome-conjugated secondary antibody, it is
best to titrate both the neat, primary antibody, as well as the
fluorochrome-conjugated secondary antibody. Begin by label-
ing the cells with the primary antibody at the manufacturer’s
recommended concentration (typically 1.25–20 ng/mL final
dilution) and then perform a series of secondary antibody
dilutions similar to the method outlined for the titration of
primary antibodies. Once the optimal concentration has been
determined for the secondary antibody, perform a titration of
the primary antibody as described. The listed concentration of
a fluorochrome-conjugated antibody includes both the anti-
body as well as the fluorochrome; therefore, the concentration
of each antibody required will likely be less.
148 Daniel R. Riordon and Kenneth R. Boheler
Acknowledgments
References
1. Thomson JA, Itskovitz-Eldor J, Shapiro SS, 8. Chin MH, Mason MJ, Xie W, Volinia S,
Waknitz MA, Swiergiel JJ, Marshall VS, Jones Singer M, Peterson C, Ambartsumyan G,
JM (1998) Embryonic stem cell lines derived Aimiuwu O, Richter L, Zhang J et al (2009)
from human blastocysts. Science Induced pluripotent stem cells and embryonic
282:1145–1147 stem cells are distinguished by gene expression
2. Tachibana M, Amato P, Sparman M, Gutierrez signatures. Cell Stem Cell 5:111–123
NM, Tippner-Hedges R, Ma H, Kang E, 9. Narsinh KH, Sun N, Sanchez-Freire V, Lee AS,
Fulati A, Lee HS, Sritanaudomchai H et al Almeida P, Hu S, Jan T, Wilson KD, Leong D,
(2013) Human embryonic stem cells derived Rosenberg J et al (2011) Single cell transcrip-
by somatic cell nuclear transfer. Cell tional profiling reveals heterogeneity of human
153:1228–1238 induced pluripotent stem cells. J Clin Invest
3. Takahashi K, Tanabe K, Ohnuki M, Narita M, 121:1217–1221
Ichisaka T, Tomoda K, Yamanaka S (2007) 10. Tang C, Lee AS, Volkmer JP, Sahoo D, Nag D,
Induction of pluripotent stem cells from adult Mosley AR, Inlay MA, Ardehali R, Chavez SL,
human fibroblasts by defined factors. Cell Pera RR et al (2011) An antibody against
131:861–872 SSEA-5 glycan on human pluripotent stem
4. Park IH, Zhao R, West JA, Yabuuchi A, cells enables removal of teratoma-forming
Huo H, Ince TA, Lerou PH, Lensch MW, cells. Nat Biotechnol 29:829–834
Daley GQ (2008) Reprogramming of human 11. Boheler KR, Bhattacharya S, Kropp EM,
somatic cells to pluripotency with defined fac- Chuppa S, Riordon DR, Bausch-Fluck D, Bur-
tors. Nature 451:141–146 ridge PW, Wu JC, Wersto RP, Chan GC et al
5. Yu J, Vodyanik MA, Smuga-Otto K, (2014) A human pluripotent stem cell surface
Antosiewicz-Bourget J, Frane JL, Tian S, Nie J, N-glycoproteome resource reveals markers,
Jonsdottir GA, Ruotti V, Stewart R et al (2007) extracellular epitopes, and drug targets. Stem
Induced pluripotent stem cell lines derived from Cell Rep 3:185–203
human somatic cells. Science 318:1917–1920 12. Koivuniemi R, Makela J, Hokkanen ME,
6. Narsinh KH, Plews J, Wu JC (2011) Compari- Bruelle C, Ho TH, Ola R, Korhonen L,
son of human induced pluripotent and embry- Schroder J, Kataoka H, Lindholm D (2013)
onic stem cells: fraternal or identical twins? Mol Hepatocyte growth factor activator inhibitor-
Ther 19:635–638 1 is induced by bone morphogenetic proteins
7. Ghosh Z, Wilson KD, Wu Y, Hu S, and regulates proliferation and cell fate of neu-
Quertermous T, Wu JC (2010) Persistent ral progenitor cells. PLoS One 8:e56117
donor cell gene expression among human 13. van Galen P, Kreso A, Mbong N, Kent DG,
induced pluripotent stem cells contributes to Fitzmaurice T, Chambers JE, Xie S, Laurenti E,
differences with human embryonic stem cells. Hermans K, Eppert K et al (2014) The
PLoS One 5:e8975 unfolded protein response governs integrity of
Flow Cytometry and Immunophenotyping 149
the haematopoietic stem-cell pool during Howden SE, Diol NR, Propson NE et al
stress. Nature 510:268–272 (2011) Chemically defined conditions for
14. Yu KR, Yang SR, Jung JW, Kim H, Ko K, Han human iPSC derivation and culture. Nat Meth-
DW, Park SB, Choi SW, Kang SK, Scholer H ods 8:424–429
et al (2012) CD49f enhances multipotency and 16. Beers J, Gulbranson DR, George N, Siniscalchi
maintains stemness through the direct regula- LI, Jones J, Thomson JA, Chen G (2012) Pas-
tion of OCT4 and SOX2. Stem Cells saging and colony expansion of human plurip-
30:876–887 otent stem cells by enzyme-free dissociation in
15. Chen G, Gulbranson DR, Hou Z, Bolin JM, chemically defined culture conditions. Nat Pro-
Ruotti V, Probasco MD, Smuga-Otto K, toc 7:2029–2040
Chapter 10
Abstract
G protein-coupled receptors (GPCRs) are cell surface receptors that relay extracellular signals to the inside
of the cells. C-X-C chemokine receptor 4 (CXCR4) is a GPCR that undergoes receptor internalization and
recycling upon stimulation with its cognate ligand, C-X-C chemokine 12 (CXCL12). Using this receptor/
ligand pair we describe the use of two techniques, enzyme-linked immunosorbent assay (ELISA) and flow
cytometry, widely used to quantify GPCR internalization from the plasma membrane and its return to the
cell surface by recycling.
Key words G Protein-coupled receptor (GPCR), C-X-C chemokine receptor 4 (CXCR4), C-X-C
chemokine ligand 12 (CXCL12), Receptor internalization, ELISA, Flow cytometry
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_10, © Springer Science+Business Media, LLC 2018
151
152 Amanda M. Nevins and Adriano Marchese
or internalized from the cell surface via endocytosis [7]. For most
GPCRs, a large fraction of the internalized receptor is rapidly
recycled to the plasma membrane leading to the resensitization of
signaling [8–11].
The GPCR that we use here, as an example of internalization
and recycling, is the C-X-C chemokine receptor 4 (CXCR4). Che-
mokine receptors constitute the largest branch of the γ subfamily of
rhodopsin-like GPCRs and, along with their ligands, chemotactic
cytokines, are involved in the direction of leukocyte trafficking
[3, 12–14]. Stimulation of CXCR4 by its cognate ligand,
CXC-chemokine ligand 12 (CXCL12), plays a role in cancer metas-
tasis and progression, HIV infection, and WHIM syndrome [3, 12,
15–18]. Monitoring the internalization and recycling of CXCR4
following CXCL12 stimulation using techniques targeted at the
quantification of cell surface receptor expression is one way to assess
the role of receptor trafficking and recycling in cellular fate. An
enzyme-linked immunosorbent assay (ELISA) is a plate-based tech-
nique that employs an enzyme, like alkaline phosphatase, to detect
an immobilized antigen after incubation with a substrate, yielding a
measurable product [19]. Flow cytometry detects fluorescence
emitted from cell-bound fluorophores upon excitation as they
pass in front of a light source [20]. In this chapter, these techni-
ques, ELISA (1.1 and 1.3) and flow cytometry (1.2), are described
for the quantification of CXCL12-stimulated surface-CXCR4
internalization and recycling.
2 Materials
2.1 Cell Lines 1. Human embryonic kidney cells (HEK293) (Microbix, Tor-
onto, CA) stably expressing tagged CXCR4 [9].
2. Human cervical cancer cells with high levels of endogenous
CXCR4 expression (HeLa, ATCC, Manassas, VA, USA).
2.3 ELISA Materials 1. Assay medium: DMEM, 0.1% BSA, 20 mM HEPES, 1 mM Ca+2.
and Reagents 2. 100 mM Ca+2 stock.
3. Ca+2/Mg+2-free PBS.
4. PBS supplemented with 1 mM Ca+2.
5. Monoclonal mouse M1 anti-FLAG® antibody (Sigma-Aldrich,
St. Louis, MO, USA).
6. Ethylenediaminetetraacetic acid (EDTA).
7. 10 μM stock of CXCL12 (CXCR4 ligand/agonist) (Protein
Foundry, Milwaukee, WI, USA) (see Note 1).
8. AMD3100—CAS 155148–31-5 (CXCR4 antagonist) (Sigma-
Aldrich).
9. Fixing solution: 3.7% paraformaldehyde (PFA) in PBS.
10. Alkaline phosphatase-conjugated goat anti-mouse antibody
(Sigma-Aldrich).
11. P-Nitrophenyl phosphate (developing solution) (Bio-Rad
Laboratories, Hercules, CA, USA).
12. Diethanolamine buffer (Bio-Rad Laboratories).
13. 0.4 M NaOH.
14. Plate reader to measure absorbance. We use the FlexStation
3 multi-mode plate reader from Molecular Devices (Sunnyvale,
CA, USA).
2.4 Flow Cytometry- 1. CellStripper® (Fisher Scientific Co., Pittsburgh, PA, USA).
Specific Material 2. Trypan Blue.
and Reagents
3. Flow buffer: PBS supplemented with 0.1% BSA.
4. PE-conjugated anti-CXCR4 (Catalog No: #306506 (clone
12G5); BioLegend, San Diego, CA, USA).
5. IgG2a κ-isotype control (Catalog No: #400212; BioLegend,
San Diego, CA, USA).
6. Fixing solution: 3.7% paraformaldehyde (PFA) in PBS.
7. 10 μM stock of CXCL12 (CXCR4 ligand/agonist) (see Note 1).
8. Round bottom test tubes suitable for flow cytometry
(BD Bioscience, Franklin Lakes, NJ, USA).
9. Our flow cytometry facility is equipped with a FACSCalibur
flow cytometer and FlowJo software (BD Biosciences, San Jose,
CA, USA).
154 Amanda M. Nevins and Adriano Marchese
3 Methods
after this wash, and leave plates on ice to process in parallel with
the internalization (I45) wells.
10. Following the 45 min incubation with either vehicle or
CXCL12, aspirate medium from the internalization (I45)
wells. Fix and wash cells in the I45 wells as described for control
wells in steps 8 and 9. Cover with aluminum foil to prevent
exposure to light and leave on ice to process in parallel with the
I45 wells.
11. After washes incubate each plate with 300 μL of alkaline
phosphatase-conjugated goat anti-mouse antibody (see Note
10) for 1 h at RT.
12. Post-incubation, wash cells three times with 500 μL PBS
(þ1 mM Ca+2). Incubate plates with 250 μL developing solu-
tion that has been diluted in diethanolamine buffer for
5–15 min (see Note 11).
13. Quench the reactions of each well with 100 μL of 0.4 M
NaOH. Take 100 μL aliquots of each condition and measure
the absorbance at 405 nm using a plate reader.
14. Calculate the proportion of receptor internalized by dividing
the amount of surface receptor by the total number of receptor
present at the cell surface prior to treatment with CXCL12
according to the following formula:
3.2 CXCR4 Surface 1. Culture HeLa cells in 10-cm dishes as described in Subheading
Expression 3.1 until 90–95% confluent (see Note 2).
and Internalization: 2. Wash cells once with 10 mL incomplete DMEM. Subsequently,
Flow Cytometry serum-starve the cells in 10 mL of fresh incomplete DMEM for
at least 3 h (see Note 5).
3. Wash cells twice with 10 mL PBS and detach cells from plates
using a nonenzymatic dissociation buffer such as CellStripper®
(Corning) (see Note 12). Aspirate medium, add 2 mL of
CellStripper® and incubate cells at 37 C for 10 min or until
detached (see Note 13).
4. Add 8 mL cold (4 C) (see Note 6) flow buffer prepared as
specified in Subheading 2.4 and transfer cells to a conical
156 Amanda M. Nevins and Adriano Marchese
4 Notes
Acknowledgment
References
1. Fredriksson R, Lagerstrom MC, Lundin LG, 8. Bhandari D, Trejo J, Benovic JL, Marchese A
Schioth HB (2003) The G-protein-coupled (2007) Arrestin-2 interacts with the ubiquitin-
receptors in the human genome form five protein isopeptide ligase atrophin-interacting
main families. Phylogenetic analysis, paralogon protein 4 and mediates endosomal sorting of
groups, and fingerprints. Mol Pharmacol the chemokine receptor CXCR4. J Biol Chem
63:1256–1272 282:36971–36979
2. Tesmer JJ (2016) Hitchhiking on the heptahe- 9. Malik R, Marchese A (2010) Arrestin-2-
lical highway: structure and function of 7TM interacts with the endosomal sorting complex
receptor complexes. Nat Rev Mol Cell Biol required for transport machinery to modulate
17:439–450 endosomal sorting of CXCR4. Mol Biol Cell
3. Bachelerie F, Ben-Baruch A, Burkhardt AM, 21:2529–2541
Combadiere C, Farber JM, Graham GJ, 10. Malik R, Soh UJ, Trejo J, Marchese A (2012)
Horuk R, Sparre-Ulrich AH et al (2014) Inter- Novel roles for the E3 ubiquitin ligase
national Union of Basic and Clinical Pharma- atrophin-interacting protein 4 and signal trans-
cology. [corrected]. LXXXIX. Update on the duction adaptor molecule 1 in G protein-
extended family of chemokine receptors and coupled receptor signaling. J Biol Chem
introducing a new nomenclature for atypical 287:9013–9027
chemokine receptors. Pharmacol Rev 66:1–79 11. Marchese A, Benovic JL (2001) Agonist-
4. Van Rhee AM, Jacobson KA (1996) Molecular promoted ubiquitination of the G protein-
architechture of G protein-coupled receptors. coupled receptor CXCR4 mediates lysosomal
Drug Dev Res 37(1):1–38 sorting. J Biol Chem 276:45509–45512
5. Rajagopal S, Ahn S, Rominger DH, Gowen- 12. Chevigne A, Fievez V, Szpakowska M,
MacDonald W, Lam CM, Dewire SM, Violin Fischer A, Counson M, Plesseria JM, Schmit
JD, Lefkowitz RJ (2011) Quantifying ligand JC, Deroo S (2014) Neutralising properties of
bias at seven-transmembrane receptors. Mol peptides derived from CXCR4 extracellular
Pharmacol 80:367–377 loops towards CXCL12 binding and HIV-1
6. Whalen EJ, Rajagopal S, Lefkowitz RJ (2011) infection. Biochim Biophys Acta
Therapeutic potential of beta-arrestin- and G 1843:1031–1041
protein-biased agonists. Trends Mol Med 13. Moser B, Willimann K (2004) Chemokines:
17:126–139 role in inflammation and immune surveillance.
7. Neel NF, Schutyser E, Sai J, Fan GH, Rich- Ann Rheum Dis 63(Suppl 2):ii84–ii89
mond A (2005) Chemokine receptor internali- 14. Moser B, Wolf M, Walz A, Loetscher P (2004)
zation and intracellular trafficking. Cytokine Chemokines: multiple levels of leukocyte
Growth Factor Rev 16:637–658 migration control. Trends Immunol 25:75–84
164 Amanda M. Nevins and Adriano Marchese
15. Balabanian K, Lagane B, Pablos JL, Laurent L, 25. Pinilla-Macua I, Sorkin A (2015) Methods to
Planchenault T, Verola O, Lebbe C, Kerob D study endocytic trafficking of the EGF recep-
et al (2005) WHIM syndromes with different tor. Methods Cell Biol 130:347–367
genetic anomalies are accounted for by 26. Kershaw T, Wavre-Shapton ST, Signoret N,
impaired CXCR4 desensitization to CXCL12. Marsh M (2009) Analysis of chemokine recep-
Blood 105:2449–2457 tor endocytosis and intracellular trafficking.
16. Brelot A, Heveker N, Adema K, Hosie MJ, Methods Enzymol 460:357–377
Willett B, Alizon M (1999) Effect of mutations 27. Bonecchi R, Savino B, Borroni EM,
in the second extracellular loop of CXCR4 on Mantovani A, Locati M (2010) Chemokine
its utilization by human and feline immunode- decoy receptors: structure-function and
ficiency viruses. J Virol 73:2576–2586 biological properties. Curr Top Microbiol
17. Forster R, Kremmer E, Schubel A, Breitfeld D, Immunol 341:15–36
Kleinschmidt A, Nerl C, Bernhardt G, Lipp M 28. Borroni EM, Mantovani A, Locati M, Bone-
(1998) Intracellular and surface expression of cchi R (2010) Chemokine receptors intracellu-
the HIV-1 coreceptor CXCR4/fusin on vari- lar trafficking. Pharmacol Ther 127:1–8
ous leukocyte subsets: rapid internalization and 29. Wilson DF, Chance B (1967) Azide inhibition
recycling upon activation. J Immunol of mitochondrial electron transport. I. The aer-
160:1522–1531 obic steady state of succinate oxidation. Bio-
18. Ramsey DM, McAlpine SR (2013) Halting chim Biophys Acta 131:421–430
metastasis through CXCR4 inhibition. Bioorg 30. Carnec X, Quan L, Olson WC, Hazan U, Dra-
Med Chem Lett 23:20–25 gic T (2005) Anti-CXCR4 monoclonal antibo-
19. Engvall E (1980) Enzyme immunoassay dies recognizing overlapping epitopes differ
ELISA and EMIT. Methods Enzymol significantly in their ability to inhibit entry of
70:419–439 human immunodeficiency virus type 1. J Virol
20. Givan AL (2011) Flow cytometry: an introduc- 79:1930–1933
tion. Methods Mol Biol 699:1–29 31. Veldkamp CT, Seibert C, Peterson FC, De la
21. Anselmo A, Mazzon C, Borroni EM, Cruz NB, Haugner JC III, Basnet H, Sakmar
Bonecchi R, Graham GJ, Locati M (2014) TP, Volkman BF (2008) Structural basis of
Flow cytometry applications for the analysis of CXCR4 sulfotyrosine recognition by the che-
chemokine receptor expression and function. mokine SDF-1/CXCL12. Sci Signal 1:ra4
Cytometry A 85:292–301 32. Veldkamp CT, Ziarek JJ, Peterson FC, Chen Y,
22. Sloane AJ, Raso V, Dimitrov DS, Xiao X, Volkman BF (2010) Targeting SDF-1/
Deo S, Muljadi N, Restuccia D, Turville S CXCL12 with a ligand that prevents activation
et al (2005) Marked structural and functional of CXCR4 through structure-based drug
heterogeneity in CXCR4: separation of HIV-1 design. J Am Chem Soc 132:7242–7243
and SDF-1alpha responses. Immunol Cell Biol 33. Ziarek JJ, Getschman AE, Butler SJ, Taleski D,
83:129–143 Stephens B, Kufareva I, Handel TM, Payne RJ,
23. Alekhina O, Marchese A (2016) Beta-arrestin1 Volkman BF (2013) Sulfopeptide probes of the
and signal-transducing adaptor molecule CXCR4/CXCL12 interface reveal oligomer-
1 (STAM1) cooperate to promote focal adhe- specific contacts and chemokine allostery. ACS
sion kinase autophosphorylation and chemo- Chem Biol 8:1955–1963
taxis via the chemokine receptor CXCR4. J 34. Kalatskaya I, Berchiche YA, Gravel S, Limberg
Biol Chem 2016:M116.757138 BJ, Rosenbaum JS, Heveker N (2009)
24. Veldkamp CT, Koplinski CA, Jensen DR, AMD3100 is a CXCR7 ligand with allosteric
Peterson FC, Smits KM, Smith BL, Johnson agonist properties. Mol Pharmacol
SK, Lettieri C et al (2016) Production of 75:1240–1247
recombinant chemokines and validation of
refolding. Methods Enzymol 570:539–565
Part III
Abstract
Integral to the cell surface is channels, pumps, and exchanger proteins that facilitate the movement of ions
across the membrane. Ion channels facilitate the passive movement of ions down an electrochemical
gradient. Ion pumps actively use energy to actively translocate ions, often against concentration or voltage
gradients, while ion exchangers utilize energy to couple the transport of different ion species such that one
ion moves down its gradient and the released free energy is used to drive the movement of a different ion
against its electrochemical gradient. Some ion pumps and exchangers may be electrogenic, i.e., the ion
transport they support is not electrically neutral and generates a current. Functions of these pore-forming
membrane proteins include the establishment of membrane potentials, gating of ions flows across the cell
membrane to elicit action potentials and other electrical signals, as well as the regulation of cell volumes.
The major forms of ion channels include voltage-, ligand-, and signal-gated channels. In this review, we
describe mammalian voltage dependent Na (NaV) channels.
Key words Ion channel, Electrophysiology, Stem cells, Optical recording, Seizure, Arrhythmia
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_11, © Springer Science+Business Media, LLC 2018
167
168 Gordon F. Tomaselli and Federica Farinelli
N Na+
I II III IV S
S
N
β1
+
+
N C
N EFL C
Ankyrin G C
L/S N4L IQ
syn
C
Fig. 1 Topology of the Na channels (left). The domains I–IV wrap around a central ion permeable pore (right)
3 Methods
3.1 Structural Ion current recording, cellular and tissue electrophysiology gener-
and Functional ate a rich tapestry of data that in combination with molecular and
Readouts protein chemical analyses facilitates a detailed functional assessment
of ion channels. These studies are essential to our understanding
the roles of ion channels in excitation, contraction, secretion, and
other essential biological functions. Moreover, such methods allow
for the study of derangements in channel function and the contri-
bution of such derangements to disease pathogenesis in rare inher-
ited traits and more common acquired diseases. Finally ion channels
are the targets of a number of drugs and toxins that are used for
everything from treatment of disease to biological warfare. Chan-
nels are attractive targets for treatment of disease but carry a serious
liability of potentially life-threatening side effects.
172 Gordon F. Tomaselli and Federica Farinelli
LATERAL MEMBRANE
N
SIV
Ank G
PDZ
Syntrophin
Dystrophin
INTERCALATED DISK
N
SIV
Ank G
SAP97
PDZ
PDZ
PDZ
Plakophilin-2
Fig. 2 Schematic of the proteins involved in localization of Na channels at the lateral membrane (top) or the
intercalated disk (bottom). A different constellation of proteins with PDZ binding domains and ankyrin G
binding proteins. Modified from Petitprez et al., 2011
Fig. 3 Total internal reflectance fluorescence (TIRF, left) and superresolution fluorescence microscopy (SRFM,
center) localization of NaV1.5 channels in cardiac myocytes. High magnification SFRM image (right) from
inside the white box shows co-localization of NaV1.5 and N-cadherin. The scale bar in the two images on the
right and that on the left are 5 mm and 1 mm, respectively. Image from Agullo-Pascual et al., 2014
3.3 Electrical The main function of ion channels is to signal by way of modulating
and Optical Recording transmembrane voltage. The ability to directly record currents from
cells and tissues provides an unmatched tool for characterizing
channel function and correlating functional features with molecular
structure. Moreover, voltage recording using electrodes or optical
dyes affords a means to study the function of a channel or channels
in the context of other electrogenic molecules.
3.4 Voltage Early voltage recording in the squid giant axon attested to the
and Patch Clamp importance of the transmembrane movement of Na+ in the gener-
Recording ation and conduction of the action potential [73]. Arguably the
single most importance advance for the quantitative study of ionic
currents was the voltage clamp developed over 65 years ago
[74–76] and a cornerstone method still used today. Using voltage
clamp recording with the proper ionic conditions currents carried
by different ions (Na+, K+, etc.) can be studied in detail. In this
method the experimenter controls the voltage across the membrane
and a feedback amplifier injects current to keep the membrane
voltage constant (Fig. 4a). Clamping the membrane voltage at
NaV Channels - Review 175
A B
50
a1 a2 V (mV)
0
-160 -110 -60 -10 40
-10 mV
-50
1 nA
-90 3 ms
5 mS -100
-150
I (nA)
i = -1.9 pA
-200
1.2
I/Imax
1
a3 a4 0.8
0.6
0.4
1nA
0.2
2ms
V (mV)
0
-150 -100 -50 0
Fig. 4 Electrophysiological recording of Na currents. (a). Configuration for whole cell current recording. (a1)
Whole cell recordings of Na currents expressed in mammalian tissue culture cells held at a holding voltage of
120 mV and stepped to a range of depolarizing test voltages. (a2) Current-voltage (IV) relationship, peak
currents plotted over a range of test voltages. (a3) Currents recorded at a test voltage of 20 mV over a range of
holding voltages to generate (a3), the steady-state inactivation curve. (b). Configuration for cell-attached
current recording (top) and exemplar single Na channels expressed in a membrane patch of an HEK-293 cell
A B
5 ms
C 20 mV 10 pA/pF
200 ms
0
control E-4031
20 mV
200 ms
Fig. 5 Na channels in adult ventricular myocytes. (a) Adult ventricular cardiomyocyte, the scale bar is 50 mm.
(b) Action potential (AP) recorded from an adult ventricular myocyte, the rapid upstroke is mediated by Na
currents. (c) The effect of a potassium channel blocking drug, E4031 on ventricular APs
A Non-transduced WT NaV1.5
150
6 200 6
150 100
0 0
100
50
50
-6 -6
CV = 20.2 cm/s CV = 28.1 cm/s
0 0
-5 0 5 -5 0 5
B
200 ms
Fig. 6 Neonatal rat ventricular myocytes (NRVM) in monolayer culture. (a) Isochronal maps of 2D monolayer
cultures of nontransduced (left) and NRVMs infected with a lentivirus expressing human NaV1.5 channels. (b)
The corresponding optical action potentials (APs). Infecting cells with Na channels hastens conduction in
monolayer cultures
Fig. 7 Optical action potentials (APs) and isochronal maps recorded in monolayers of control hiPSC-VCMs after
29 days in culture paced from a point source. The optical APs (left) shown are recorded from the black boxes in
the voltage maps (right). Slowing of conduction occurs at faster pacing rates. Loss of 1:1 capture occurred at
pacing CL < 500 ms
3.5 High-Throughput The introduction of a planar glass electrode rather than a pipette
Patch Clamp over a decade ago [82] ushered in the era of high-throughput
Recording automated patch clamp recording [83]. The planar patch
approaches allow for more automated ion current studies than
180 Gordon F. Tomaselli and Federica Farinelli
4 Conclusions
References
1. Makielski JC, Sheets MF, Hanck DA, January channel expressed by sensory neurons. Nature
CT, Fozzard HA (1987) Sodium current in 379:257–262
voltage clamped internally perfused canine car- 5. George AL Jr, Komisarof J, Kallen RG, Barchi
diac Purkinje cells. Biophys J 52:1–11 RL (1992) Primary structure of the adult
2. Noda M, Shimizu S, Tanabe T, Takai T, human skeletal muscle voltage-dependent
Kayano T, Ikeda T, Takahashi H, Nakayama sodium channel. Ann Neurol 31:131–137
H et al (1984) Primary structure of Electroph- 6. Gellens ME, George AL Jr, Chen LQ,
orus electricus sodium channel deduced from Chahine M, Horn R, Barchi RL, Kallen RG
cDNA sequence. Nature 312:121–127 (1992) Primary structure and functional
3. Dib-Hajj SD, Tyrrell L, Black JA, Waxman SG expression of the human cardiac tetrodotoxin-
(1998) NaN, a novel voltage-gated Na channel, insensitive voltage-dependent sodium channel.
is expressed preferentially in peripheral sensory Proc Natl Acad Sci U S A 89:554–558
neurons and down-regulated after axotomy. 7. Ahmed CM, Ware DH, Lee SC, Patten CD,
Proc Natl Acad Sci U S A 95:8963–8968 Ferrer-Montiel AV, Schinder AF, McPherson
4. Akopian AN, Sivilotti L, Wood JN (1996) A JD, Wagner-McPherson CB et al (1992) Pri-
tetrodotoxin-resistant voltage-gated sodium mary structure, chromosomal localization, and
NaV Channels - Review 181
33. Zhou J, Shin HG, Yi J, Shen W, Williams CP, Ca/calmodulin kinase IIdelta is required for
Murray KT (2002) Phosphorylation and puta- late I(Na) augmentation leading to cellular Na
tive ER retention signals are required for pro- and Ca overload. Circ Res 108:555–565
tein kinase A-mediated potentiation of cardiac 45. Wagner S, Dybkova N, Rasenack EC,
sodium current. Circ Res 91:540–546 Jacobshagen C, Fabritz L, Kirchhof P, Maier
34. Smith RD, Goldin AL (1997) Phosphorylation SK, Zhang T et al (2006) Ca2þ/calmodulin-
at a single site in the rat brain sodium channel is dependent protein kinase II regulates cardiac
necessary and sufficient for current reduction Naþ channels. J Clin Invest 116:3127–3138
by protein kinase A. J Neurosci 17:6086–6093 46. Ma J, Luo A, Wu L, Wan W, Zhang P, Ren Z,
35. Li M, West JW, Numann R, Murphy BJ, Zhang S, Qian C, Shryock JC, Belardinelli L
Scheuer T, Catterall WA (1993) Convergent (2012) Calmodulin kinase II and protein
regulation of sodium channels by protein kinase C mediate the effect of increased intra-
kinase C and cAMP-dependent protein kinase. cellular calcium to augment late sodium cur-
Science 261:1439–1442 rent in rabbit ventricular myocytes. Am J
36. Hallaq H, Yang Z, Viswanathan PC, Fukuda K, Physiol Cell Physiol 302:C1141–C1151
Shen W, Wang DW, Wells KS, Zhou J, Yi J, 47. Koval OM, Snyder JS, Wolf RM, Pavlovicz RE,
Murray KT (2006) Quantitation of protein Glynn P, Curran J, Leymaster ND, Dun W et al
kinase A-mediated trafficking of cardiac sodium (2012) Ca2þ/calmodulin-dependent protein
channels in living cells. Cardiovasc Res kinase II-based regulation of voltage-gated
72:250–261 Naþ channel in cardiac disease. Circulation
37. Frohnwieser B, Chen LQ, Schreibmayer W, 126:2084–2094
Kallen RG (1997) Modulation of the human 48. Herren AW, Weber DM, Rigor RR, Margulies
cardiac sodium channel alpha-subunit by KB, Phinney BS, Bers DM (2015) CaMKII
cAMP-dependent protein kinase and the phosphorylation of NaV1.5: novel in vitro
responsible sequence domain. J Physiol 498 sites identified by mass spectrometry and
(Pt 2):309–318 reduced S516 phosphorylation in human
38. Hallaq H, Wang DW, Kunic JD, George AL Jr, heart failure. J Proteome Res 14:2298–2311
Wells KS, Murray KT (2012) Activation of pro- 49. Aiba T, Hesketh GG, Liu T, Carlisle R, Villa-
tein kinase C alters the intracellular distribution Abrille MC, O’Rourke B, Akar FG, Tomaselli
and mobility of cardiac Naþ channels. Am J GF (2010) Naþ channel regulation by Ca2þ/
Physiol Heart Circ Physiol 302:H782–H789 calmodulin and Ca2þ/calmodulin-dependent
39. Frohnwieser B, Weigl L, Schreibmayer W protein kinase II in guinea-pig ventricular myo-
(1995) Modulation of cardiac sodium channel cytes. Cardiovasc Res 85:454–463
isoform by cyclic AMP dependent protein 50. Aiba T, Barth AS, Hesketh GG, Hashambhoy
kinase does not depend on phosphorylation of YL, Chakir K, Tunin RS, Greenstein JL, Win-
serine 1504 in the cytosolic loop interconnect- slow RL et al (2013) Cardiac resynchronization
ing transmembrane domains III and therapy improves altered Na channel gating in
IV. Pflugers Arch 430:751–753 canine model of dyssynchronous heart failure.
40. Cantrell AR, Ma JY, Scheuer T, Catterall WA Circ Arrhythm Electrophysiol 6:546–554
(1996) Muscarinic modulation of sodium cur- 51. Ptacek LJ, George AL Jr, Griggs RC, Tawil R,
rent by activation of protein kinase C in rat Kallen RG, Barchi RL, Robertson M, Leppert
hippocampal neurons. Neuron 16:1019–1026 MF (1991) Identification of a mutation in the
41. Hammarstrom AK, Gage PW (1998) Inhibi- gene causing hyperkalemic periodic paralysis.
tion of oxidative metabolism increases persis- Cell 67:1021–1027
tent sodium current in rat CA1 hippocampal 52. Kumar D, Ambasta RK, Kumar P (2014)
neurons. J Physiol 510(Pt 3):735–741 Mutational consequences of aberrant ion chan-
42. Ju YK, Saint DA, Gage PW (1996) Hypoxia nels in neurological disorders. J Membr Biol
increases persistent sodium current in rat ven- 247:1083–1127
tricular myocytes. J Physiol 497 53. Waxman SG, Merkies IS, Gerrits MM,
(Pt 2):337–347 Dib-Hajj SD, Lauria G, Cox JJ, Wood JN,
43. Saint DA (2006) The role of the persistent Na Woods CG et al (2014) Sodium channel
(þ) current during cardiac ischemia and hyp- genes in pain-related disorders: phenotype-
oxia. J Cardiovasc Electrophysiol 17(Suppl 1): genotype associations and recommendations
S96–S103 for clinical use. Lancet Neurol 13:1152–1160
44. Wagner S, Ruff HM, Weber SL, Bellmann S, 54. Suetterlin K, Mannikko R, Hanna MG (2014)
Sowa T, Schulte T, Anderson ME, Grandi E Muscle channelopathies: recent advances in
et al (2011) Reactive oxygen species-activated
NaV Channels - Review 183
genetics, pathophysiology and therapy. Curr 65. Angelides KJ, Elmer LW, Loftus D, Elson E
Opin Neurol 27:583–590 (1988) Distribution and lateral mobility of
55. Veerman CC, Wilde AA, Lodder EM (2015) voltage-dependent sodium channels in neu-
The cardiac sodium channel gene SCN5A and rons. J Cell Biol 106:1911–1925
its gene product NaV1.5: role in physiology 66. Garrido JJ, Giraud P, Carlier E, Fernandes F,
and pathophysiology. Gene 573:177–187 Moussif A, Fache MP, Debanne D, Dargent B
56. Tan BH, Iturralde-Torres P, Medeiros- (2003) A targeting motif involved in sodium
Domingo A, Nava S, Tester DJ, Valdivia CR, channel clustering at the axonal initial segment.
Tusie-Luna T, Ackerman MJ, Makielski JC Science 300:2091–2094
(2007) A novel C-terminal truncation SCN5A 67. Bennett V, Baines AJ (2001) Spectrin and
mutation from a patient with sick sinus syn- ankyrin-based pathways: metazoan inventions
drome, conduction disorder and ventricular for integrating cells into tissues. Physiol Rev
tachycardia. Cardiovasc Res 76:409–417 81:1353–1392
57. Medeiros-Domingo A, Kaku T, Tester DJ, 68. Aiba T, Farinelli F, Kostecki G, Hesketh GG,
Iturralde-Torres P, Itty A, Ye B, Valdivia C, Edwards D, Biswas S, Tung L, Tomaselli GF
Ueda K et al (2007) SCN4B-encoded sodium (2014) A mutation causing brugada syndrome
channel beta4 subunit in congenital long-QT identifies a mechanism for altered autonomic
syndrome. Circulation 116:134–142 and oxidant regulation of cardiac sodium cur-
58. Grant AO, Carboni MP, Neplioueva V, Starmer rents. Circ Cardiovasc Genet 7:249–256
CF, Memmi M, Napolitano C, Priori S (2002) 69. Makara MA, Curran J, Little SC, Musa H,
Long QT syndrome, Brugada syndrome, and Polina I, Smith SA, Wright PJ, Unudurthi SD
conduction system disease are linked to a single et al (2014) Ankyrin-G coordinates interca-
sodium channel mutation. J Clin Invest lated disc signaling platform to regulate cardiac
110:1201–1209 excitability in vivo. Circ Res 115:929–938
59. Lupoglazoff JM, Cheav T, Baroudi G, 70. Shy D, Gillet L, Ogrodnik J, Albesa M, Verkerk
Berthet M, Denjoy I, Cauchemez B, AO, Wolswinkel R, Rougier JS, Barc J et al
Extramiana F, Chahine M, Guicheney P (2014) PDZ domain-binding motif regulates
(2001) Homozygous SCN5A mutation in cardiomyocyte compartment-specific NaV1.5
long-QT syndrome with functional two-to- channel expression and function. Circulation
one atrioventricular block. Circ Res 89: 130:147–160
E16–E21 71. Agullo-Pascual E, Reid DA, Keegan S,
60. Kyndt F, Probst V, Potet F, Demolombe S, Sidhu M, Fenyo D, Rothenberg E, Delmar M
Chevallier JC, Baro I, Moisan JP, Boisseau P (2013) Super-resolution fluorescence micros-
et al (2001) Novel SCN5A mutation leading copy of the cardiac connexome reveals
either to isolated cardiac conduction defect or plakophilin-2 inside the connexin43 plaque.
Brugada syndrome in a large French family. Cardiovasc Res 100:231–240
Circulation 104:3081–3086 72. Agullo-Pascual E, Lin X, Leo-Macias A,
61. Bezzina C, Veldkamp MW, van Den Berg MP, Zhang M, Liang FX, Li Z, Pfenniger A, Lub-
Postma AV, Rook MB, Viersma JW, van Lan- kemeier I et al (2014) Super-resolution imag-
gen IM, Tan-Sindhunata G et al (1999) A sin- ing reveals that loss of the C-terminus of
gle Na(þ) channel mutation causing both connexin43 limits microtubule plus-end cap-
long-QT and Brugada syndromes. Circ Res ture and NaV1.5 localization at the intercalated
85:1206–1213 disc. Cardiovasc Res 104:371–381
62. Waxman SG (2006) Axonal conduction and 73. Hodgkin AL, Katz B (1949) The effect of
injury in multiple sclerosis: the role of sodium sodium ions on the electrical activity of giant
channels. Nat Rev Neurosci 7:932–941 axon of the squid. J Physiol 108:37–77
63. Long C, McAnally JR, Shelton JM, Mireault 74. Marmont G (1949) Studies on the axon mem-
AA, Bassel-Duby R, Olson EN (2014) Preven- brane; a new method. J Cell Physiol
tion of muscular dystrophy in mice by 34:351–382
CRISPR/Cas9-mediated editing of germline 75. Hodgkin AL, Huxley AF, Katz B (1949) Ionic
DNA. Science 345:1184–1188 currents underlying activity in the giant axon of
64. Long C, Amoasii L, Mireault AA, McAnally JR, the squid. Arch Sci Physiol 3:129–150
Li H, Sanchez-Ortiz E, Bhattacharyya S, Shel- 76. Cole KS (1949) Dynamic electrical character-
ton JM et al (2016) Postnatal genome editing istics of the squid axon membrane. Arch Sci
partially restores dystrophin expression in a Physiol 3:253–258
mouse model of muscular dystrophy. Science 77. Hamill OP, Marty A, Neher E, Sakmann B,
351:400–403 Sigworth FJ (1981) Improved patch-clamp
184 Gordon F. Tomaselli and Federica Farinelli
techniques for high-resolution current record- and adult fibroblast cultures by defined factors.
ing from cells and cell-free membrane patches. Cell 126:663–676
Pflugers Arch 391:85–100 81. Takahashi K, Tanabe K, Ohnuki M, Narita M,
78. Dykxhoorn DM, Lieberman J (2005) The Ichisaka T, Tomoda K, Yamanaka S (2007)
silent revolution: RNA interference as basic Induction of pluripotent stem cells from adult
biology, research tool, and therapeutic. Annu human fibroblasts by defined factors. Cell
Rev Med 56:401–423 131:861–872
79. Deschenes I, Armoundas AA, Jones SP, Toma- 82. Fertig N, Blick RH, Behrends JC (2002)
selli GF (2008) Post-transcriptional gene Whole cell patch clamp recording performed
silencing of KChIP2 and Navbeta1 in neonatal on a planar glass chip. Biophys J
rat cardiac myocytes reveals a functional associ- 82:3056–3062
ation between Na and Ito currents. J Mol Cell 83. Farre C, Fertig N (2012) HTS techniques for
Cardiol 45:336–346 patch clamp-based ion channel screening -
80. Takahashi K, Yamanaka S (2006) Induction of advances and economy. Expert Opin Drug Dis-
pluripotent stem cells from mouse embryonic cov 7:515–524
Chapter 12
Abstract
Considerable interest has been raised to develop human pluripotent stem cell-derived cardiomyocytes
(hPSC-CMs) as a model for drug discovery and cardiotoxicity screening. High-content electrophysiological
analysis of currents generated by transmembrane cell surface ion channels has been pursued to complement
such emerging applications. Here we describe practical procedures and considerations for accomplishing
successful assays of hPSC-CMs using an automated planar patch-clamp system.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_12, © Springer Science+Business Media, LLC 2018
185
186 Chi-Wing Kong et al.
2 Materials
2.1 Human Human embryonic stem cell (hESC) line, HES2 (ESI, passages
Embryonic Stem Cell 35–55), was maintained in its pluripotent state and subsequently
Derived differentiated into cardiomyocytes with our published protocol for
Cardiomyocytes (CM) ventricular specification [10]. Although the differentiation proto-
col is not described here, the differentiated cardiospheres typically
contain >90% Troponin T type 2 (cardiac) (TNNT2+) cells that can
be isolated following enzymatic digestion.
2.4 Patch Clamp All solutions were prepared in double distilled water and filtered to
Recording Solutions remove impurities right after preparation. For convenience, all
extra- and intra-cellular solutions can be stored as aliquots at
20 C for 3 months (see Note 1).
1. Sealing solution containing (in mM) 150 NaCl, 4 KCl, 1.2
CaCl2, 1 MgCl2, and 10 HEPES (4-(2-hydroxyethyl)-1-piper-
azineethanesulfonic acid). The pH was adjusted to 7.4 with
NaOH. Freshly prepared sealing solution can be stored at room
temperature, showing consistent performance when used
within 3 days (see Note 2).
2. Sodium current (INa): The extracellular solution contained
(in mM) 50 NaCl, 110 K-aspartate, 1.8 CaCl2, 1 MgCl2,
10 D-Glucose, 10 HEPES, and 0.001 Nifedipine. The pH
was adjusted to 7.4 with CsOH. The intracellular solution
contained (in mM): 135 CsCl, 10 NaCl, 2 CaCl2, 5 EGTA,
10 HEPES, and 5 MgATP, and the pH was adjusted to 7.2
with CsOH.
3. L-type Ca2+ current (ICaL): The extracellular solution
contained (in mM): 160 tetraethylammonium chloride
(TEA-Cl), 1 MgCl2, 5 CaCl2, 10 D-Glucose, and 10 HEPES.
The pH was adjusted to 7.4 with CsOH. The intracellular
solution contained (in mM): 145 CsCl, 5 NaCl, 2 CaCl2,
5 MgATP, 10 HEPES, and 5 EGTA, the pH was adjusted to
7.2 with CsOH. Tetrodotoxin (TTX, 0.01 mM) and
4-aminopyridine (4-AP, 2 mM) were included in the external
bath during recording.
4. Rapid delayed rectifier potassium current (IKr): The extracellu-
lar solution contained (in mM): 140 KCl, 15 NaCl, 1 MgCl2,
1.2 CaCl2, 0.002 Nifedipine, and 10 HEPES, and the pH was
adjusted to 7.4 with NaOH. The intracellular solution
contained (in mM): 110 K-aspartate, 20 KCl, 5 MgATP,
1 EGTA, 1 MgCl2, and 10 HEPES, 5 Na2-phosphocreatine,
and 0.1 NaGTP. The pH was adjusted to 7.4 with KOH.
188 Chi-Wing Kong et al.
3 Methods
3.3 Automated Patch The detailed operation procedures of the PatchXpress 7000A or
Clamping equivalent, and data analysis can be found in the user manual
provided by the manufacturer of the high-content electrophysiology
system employed by users. The PatchXpress 7000A automated patch
clamp system employed here is equipped with PatchXpress Com-
mander 2.0. All the patch settings are cell type-specific and require
empirical optimizations (see Note 12) as needed. Table 1 sum-
marizes performance indices such as the cell-detection rate, the
whole-cell rate, the average giga-seal resistance and membrane resis-
tance in this assay of HES2-CMs.
On average, our assays of cardiac ion currents in hESC-CMs
started ~20 min after cell recovery in the sealing solution.
1. Whole-cell currents were sampled at 10 kHz and low-pass
filtered at 2 kHz.
2. Data were transferred automatically into a database and can be
analyzed using the DataXpress software.
3. Figure 2 summarizes the characteristics of three ionic currents
(INa, ICaL, IKr) recorded in our assay of HES2-CMs.
4. The voltage protocols applied to probe for the three cardiac
ionic currents illustrated as examples are detailed as below:
High-content Electrophysiological Analyses 191
Table 1
Summary of performance indices and cell parameters in our HES2-derived cardiomyocytes assay
a Voltage (mV)
1.0 1.0
Current density (pA/pF)
G/Gmax
-40
I/Imax
0.6
50 ms 0.5
50 mV -60
0.4
-80
-100 0.2
200 pA
–120 mV -120 0.0 0.0
1 ms
-140 -100 -80 -60 -40 -20 0
Voltage (mV)
b Voltage (mV)
1.0 1.0
Current density (pA/pF)
G/Gmax
0.6
I/Imax
0.6
-5
100 ms 0.4 0.4
60 mV
-10 0.2 0.2
200 pA 0.0 0.0
–50 mV 10 ms -15 -60 -40 -20 0 20
Voltage (mV)
Voltage (mV)
c
[K+]o 140mM/[Na+]o 15mM
Tail current density (pA/pF)
IKr -10
500 ms
60 mV
-20
-60 mV -110 mV
-30
-160 mV
Fig. 2 Characteristics of three cardiac ionic currents (INa, ICaL, IKr) recorded in our automated patch clamp
assay of HES2-CMs. The fidelity of the currents recorded with the automated patch clamp assay described is
illustrated. Left: Representative (a) INa and (b) ICaL traces elicited by voltage protocol as shown in the inset.
Middle: peak I–V plots of the two currents. Right: Steady-state inactivation and activation relations. (n ¼ 7 for
both currents). (c) Left: representative IKr traces after subtraction of E4031-insensitive current, elicited by
voltage protocol as shown in the inset. Right: activation relation of IKr (n ¼ 11). Adapted from Weng et al.
(Reprinted with permission from STEM CELLS & DEVELOPMENT Volume 23, Issue 14, 2014, pp. 1704–1716,
published by Mary Ann Liebert, Inc., New Rochelle, NY)
192 Chi-Wing Kong et al.
4 Notes
Acknowledgements
References
1. Thomson JA, Itskovitz-Eldor J, Shapiro SS, in human pluripotent stem cells. BioEssays 35
Waknitz MA, Swiergiel JJ, Marshall VS, Jones (3):281–298
JM (1998) Embryonic stem cell lines derived 7. Li RA (2012) The use of induced pluripotent
from human blastocysts. Science 282 stem cells for disease modeling: what are the
(5391):1145–1147 promises and hurdles? Drug Discov Today Dis
2. Takahashi K, Tanabe K, Ohnuki M, Narita M, Model 9(4):e143–e145
Ichisaka T, Tomoda K, Yamanaka S (2007) 8. Burridge PW, Thompson S, Millrod MA,
Induction of pluripotent stem cells from adult Weinberg S, Yuan X, Peters A et al (2011) A
human fibroblasts by defined factors. Cell 131 universal system for highly efficient cardiac dif-
(5):861–872 ferentiation of human induced pluripotent
3. Fox IJ, Daley GQ, Goldman SA, Huard J, stem cells that eliminates interline variability.
Kamp TJ, Trucco M (2014) Stem cell therapy. PLoS One 6(4):e18293
Use of differentiated pluripotent stem cells as 9. Lian X, Zhang J, Azarin SM, Zhu K, Hazeltine
replacement therapy for treating disease. Sci- LB, Bao X et al (2012) Directed cardiomyocyte
ence 345(6199):1247391 differentiation from human pluripotent stem
4. Yu J, Vodyanik MA, Smuga-Otto K, cells by modulating Wnt/beta-catenin signal-
Antosiewicz-Bourget J, Frane JL, Tian S et al ing under fully defined conditions. Nat Protoc
(2007) Induced pluripotent stem cell lines 8(1):162–175
derived from human somatic cells. Science 10. Weng Z, Kong CW, Ren L, Karakikes I,
318(5858):1917–1920 Geng L, He J et al (2014) A simple, cost-
5. Belmonte JC, Ellis J, Hochedlinger K, Yama- effective but highly efficient system for deriving
naka S (2009) Induced pluripotent stem cells ventricular cardiomyocytes from human plurip-
and reprogramming: seeing the science otent stem cells. Stem Cells Dev 23
through the hype. Nat Rev Genet 10 (14):1704–1716
(12):878–883 11. Hamill OP, Marty A, Neher E, Sakmann B,
6. Rajamohan D, Matsa E, Kalra S, Crutchley J, Sigworth FJ (1981) Improved patch-clamp
Patel A, George V, Denning C (2013) Current techniques for high-resolution current record-
status of drug screening and disease modelling ing from cells and cell-free membrane patches.
Pflugers Arch 391(2):85–100
Chapter 13
Abstract
Vascular endothelial transient potential (TRP) channels, located mostly on the plasma membrane of cells,
are critical in regulatory and pathophysiological circumstances. The objective of this chapter is to describe
several well-established approaches, ranging from function to molecular assays, to investigate the mecha-
nistic role of TRP channels in vascular endothelial cells. We show experimental procedures and representa-
tive figures on the following methods: (1) Isolation and culture of vascular endothelial cells,
(2) examination of electrophysiological activity of TRP channel by patch-clamping with whole-cell config-
uration and its function in vascular tone and blood flow by isometric tension and isobaric diameter
measurements, and Laser Doppler flowmetry, (3) detection of TRP channel-mediated intracellular Ca2+
imaging by using fluorescent microscopy, and (4) determination of TRP channel interaction by coimmu-
noprecipitation, double immunofluorescence staining and Förster resonance energy transfer (FRET)
detection.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_13, © Springer Science+Business Media, LLC 2018
195
196 Yung Wui Tjong and Xiaoqiang Yao
2 Materials
2.2 Cell Culture 1. Endothelial basic medium with 0.02% collagenase IA: Dilute
Components 2 mg of collagenase IA (Sigma-Aldrich) in 10 mL of endothe-
lial basic medium (Lonza, Walkersville, MD, USA).
2. Endothelial growth medium with 1% bovine brain extract:
Dilute 0.05 g bovine brain extract in 5 mL of endothelial
growth medium and add 50 μL of 100 Penicillin–Streptomy-
cin (100 U/mL penicillin and 100 μg/mL streptomycin)
(Invitrogen, Carlsbad, CA, USA).
3. PBS: 80 mM of Na2HPO4, 20 mM of NaH2PO4, 100 mM of
NaCl. Weigh reagents and transfer to the beaker and followed
by adding water up to 900 mL. Mix and adjust pH with HCl to
pH 7.4. Transfer the solution to the 1-L graduated cylinder
and make up to 1 L with H2O and store at 4 C.
4. Reagents and equipment: 25-cm2 tissue culture flasks, CO2
tissue culture incubator.
2.4 Fluorescent 1. Fluroscent dyes (or probes): Prepare 0.02% pluronic F127 by
Measurement adding 0.2 mg of pluronic 127 to 1 mL of H2O.
of Intracellular 2. Normal physiological salt solution (NPSS): 140 mM NaCl
Calcium Concentration (Sigma-Aldrich), 1 mL KCl (Sigma-Aldrich), 1 mM of CaCl2
([Ca2+]i) (Sigma-Aldrich), 1 mM of MgCl2 (Sigma-Aldrich), 10 mM
glucose (Sigma-Aldrich), and 5 mL HEPES (Sigma-Aldrich).
Add 500 mL of water to a 1-L glass beaker. Weigh reagents and
transfer to the beaker followed by the addition of water up to
900 mL. Mix and adjust with HCl to pH 7.4. Transfer the
solution to the 1-L graduated cylinder and make up to 1 L with
H2O and store at 4 C.
3 Methods
3.2 Patch Clamp 1. Prepare pipette solution and bath solution (see Subheading 2.3,
with Whole-Cell items 1 and 2).
Configuration 2. Seed the cells on the coverslips.
3. Place a coverslip with adherent cells into the special flow cham-
ber that is exposed to steady laminar flow with the flow rate at
2 mL/min. The distance between the flow inlet and the cells is
maintained constant. Shear stress is estimated at the range of
0.5–1 dyn/cm2.
4. Prepare glass micropipette by pulling it with P-97 micropipette
puller (see Subheading 2.1, item 1). Fill the recording micropi-
pette with bath solution.
5. Bring the recording micropipette onto the cell until the pipette
tip is in contact with the cell membrane.
6. Apply the suction through the pipette pressure tubing to make
a gigaseal on the cell membrane (see Note 1).
7. Attain the whole-cell configuration by rupturing the mem-
brane patch within the micropipette. Compensate cell capaci-
tance and series resistance using the controls on the amplifier.
8. Apply successive voltage pluses from 80 mV to þ80 mV for
100 ms duration and record the whole cell current density
(pA/pF) by using EPC-10 patch clamp amplifier (Fig. 1).
9. The whole-cell current data is analysed with PulseFit software
(see Subheading 2.1, item 1) (Fig. 1).
3.3 Fluorescence 1. Prepare 0.02% of pluronic F127 and normal physiological salt
Measurement solution (NPSS) (see Subheading 2.4, items 1 and 2).
of Intracellular 2. Seed the cells on the coverslips and load with 10 mM of the
Calcium Concentration fluorescence probe (see Subheading 2.4, item 2) for 30 min
([Ca2+]i) with NPSS solution in the dark at 37 C.
3. Place the coverslip with cells into the recording chamber filled
with NPSS solution and place the recording chamber on the
stage of an inverted microscope (see Subheading 2.1, item 2).
202 Yung Wui Tjong and Xiaoqiang Yao
[Ca2+]i (nM)
200
100
0
0 400 800 1200 1600 2000
Time (seconds)
3.4 Laser Doppler 1. SD rats (c.a. 280–320 g) are anaesthetized with ketamine
Flowmetry (35 mg/kg) plus xylazine (7 mg/kg).
2. After performing a midline laparotomy, approximately
two-thirds of the rat mesenteric arterial bed is gently placed
into a petri-dish chamber and bathed in Krebs solution.
3. The local blood perfusion of the rat mesenteric arterial bed is
assessed with a Laser Doppler perfusion system (see Subheading
2.1, item 3) (Fig. 4).
4. The acquisition was made in high resolution mode with 5 s
interval. The pixel resolution of image is 760 568. The
digital color-coded images are analyzed to quantify blood
flow in the region from mesenteric vascular beds.
5. Blood pressure of rats is simultaneously monitored through a
pressure transducer inserted in common carotid artery. When
needed, the bathing solution is changed to ones that contain
4 μM 4α-PDD (Fig. 4).
3.5 Isometric 1. Sacrifice the male C57BL mice (~5 weeks old) by cervical
Tension Measurement dislocation.
2. Isolate the thoracic aorta and place it into an ice-chilled Kreb-
s–Henseleit solution (see Subheading 2.6, item 1) bubbled
with 95% O2 and 5% CO2 gas mixture.
3. Remove the fat and peripheral tissues under a dissection
microscope.
4. Cut the aorta into 2 mm segments.
5. Mount the aortic rings onto two thin stainless steel holders
(supplied by isometric tension myograph; see Subheading 2.1,
204 Yung Wui Tjong and Xiaoqiang Yao
Fig. 4 Role of TRPV4-KCa2.3 pathway in the control of local blood flow in mesenteric bed ex vivo. Representa-
tive images of Laser Doppler studies in rat mesenteric arteries from the fourth-order to the end in response to
4α-PDD
SNAP
2.5 mN
5 min
3.7 Western Blotting 1. Calibrate protein samples to equal amounts based on the stan-
dard curve obtained by Lowry assay (see Subheading 2.1, item 6).
206 Yung Wui Tjong and Xiaoqiang Yao
a Phe b Phe
Flow
Vessel diameter (µm) 400
Fig. 6 Representative traces showing the effect of T1E3, a TRPC1 blocking antibody, on flow-induced vascular
isobaric diameter changes in isolated mice mesenteric arteries [13]. Arteries were preincubated with
preimmune IgG (1:50) or T1E3 (1:50) overnight. The solid bar on the top of the trace indicates the period
when intraluminal flow (Krebs solution with 1% BSA) was applied. The arteries were preconstricted with
phenylephrine (Phe)
a b
IP: pre-immune Anti-C1 IP: pre-immune Anti-V4
IB: Anti-V4 Anti-V4 IB: Anti-C1 Anti-C1
KDa KDa
148- 148-
98-
98-
3. Place the coverslips with the cells into a chamber 12–24 h after
transfection.
4. Mount the chamber on an inverted microscope equipped with
a CCD camera and three-cube FRET filter set.
5. Subtract the average background signal.
6. Capture the fluorescence images of the transfected cells at
CFP-, YFP-, and FRET-channels respectively.
7. Calculate the FRET ratio (FR) (Fig. 8) by the equation shown
as follows:
FR ¼ FAD/FA ¼ [SFRET(DA) RD1 SCFP(DA)]/
RA1 [SYFP(DA) RD2 SCFP(DA)]
where FAD represents the total YFP emission with 430/425-
nm excitation, and FA represents the direct YFP emission with
500/520-nm excitation. In SCUBE(SPECIMEN), CUBE indi-
cates the filter cube (CFP, YFP, or FRET), and SPECIMEN
indicates whether the cell is expressing donor (D, CFP), accep-
tor (A, YFP), or both (DA). RD1 ¼ SFRET(D)/SCFP(D),
RD2 ¼ SYFP(D)/SCFP(D), and RA1 ¼ SFRET(A)/SYFP(A) are
predetermined constants that require measurement of the
bleed-through of the emission of only CFP- or YFP-tagged
molecules into the FRET channel and the emission of only
CFP-tagged molecules into the YFP channel.
4 Notes
Fig. 8 FRET detection for the interaction between TRPV4 and TRPC1. Horizontal
axes indicate FRET ratio of living cells expressing the indicated constructs
[13]. Each point represents the FRET ratio of a single cell. The red lines and
error bars indicate the average FRET ratio values and SE. When the FRET ratio is
1, there is no FRET; when the FRET ratio is greater than 1, there is FRET. Data are
given as mean SE (n ¼ 50–81). GIRK G-protein-activated inwardly rectifying
K+ channels
Acknowledgments
This work was supported by grants from the Hong Kong Research
Grant Committee TBRS T13-706/11, AoE/M-05/12,
CUHK478413 and by the China National Science Foundation
31470912.
210 Yung Wui Tjong and Xiaoqiang Yao
References
1. Vay L, Gu C, McNaughton PA (2012) The 12. Minamitani H, Okada E (1993) Microscopic
thermo-TRP ion channel family: properties laser Doppler velocimeter measuring blood
and therapeutic implications. Br J Pharmacol velocity in single microvessel. Keio J Med
165:787–801 42:186–190
2. Hecquet CM, Zhang M, Mittal M, Vogel SM, 13. Ma X, Qiu S, Luo J, Ma Y, Ngai CY et al (2010)
Di A et al (2014) Cooperative interaction of trp Functional role of vanilloid transient receptor
melastatin channel transient receptor potential potential 4-canonical transient receptor poten-
(TRPM2) with its splice variant TRPM2 short tial 1 complex in flow-induced Ca2þ influx.
variant is essential for endothelial cell apopto- Arterioscler Thromb Vasc Biol 30:851–858
sis. Circ Res 114:469–479 14. Kobori T, Smith GD, Sandford R, Edwardson
3. Brayden JE, Earley S, Nelson MT, Reading S JM (2009) The transient receptor potential
(2008) Transient receptor potential (TRP) channels TRPP2 and TRPC1 form a heterote-
channels, vascular tone and autoregulation of tramer with a 2:2 stoichiometry and an alter-
cerebral blood flow. Clin Exp Pharmacol nating subunit arrangement. J Biol Chem
Physiol 35:1116–1120 284:35507–35513
4. Holzer P, Izzo AA (2014) The pharmacology 15. Shen B, Cheng KT, Leung YK, Kwok YC,
of TRP channels. Br J Pharmacol Kwan HY et al (2008) Epinephrine-induced
171:2469–2473 Ca2þ influx in vascular endothelial cells is
5. Hellmich UA, Gaudet R (2014) Structural mediated by CNGA2 channels. J Mol Cell Car-
biology of TRP channels. Handb Exp Pharma- diol 45:437–445
col 223:963–990 16. Masters SC (2004) Co-immunoprecipitation
6. Owsianik G, Talavera K, Voets T, Nilius B from transfected cells. Methods Mol Biol
(2006) Permeation and selectivity of TRP 261:337–350
channels. Annu Rev Physiol 68:685–717 17. Herman B, Krishnan RV, Centonze VE (2004)
7. Kwan HY, Huang Y, Yao X (2007) TRP chan- Microscopic analysis of fluorescence resonance
nels in endothelial function and dysfunction. energy transfer (FRET). Methods Mol Biol
Biochim Biophys Acta 1772:907–914 261:351–370
8. Mathar I, Vennekens R, Meissner M, Kees F, 18. Latif R, Graves P (2000) Fluorescent probes:
Van der Mieren G et al (2010) Increased cate- looking backward and looking forward. Thy-
cholamine secretion contributes to hyperten- roid 10:407–412
sion in TRPM4-deficient mice. J Clin Invest 19. Du J, Ma X, Shen B, Huang Y, Birnbaumer L
120:3267–3279 et al (2014) TRPV4, TRPC1, and TRPP2
9. Lo CY, Tjong YW, Ho JC, Siu CW, Cheung SY assemble to form a flow-sensitive heteromeric
et al (2014) An upregulation in the expression channel. FASEB J 28:4677–4685
of vanilloid transient potential channels 20. Ma X, Cao J, Luo J, Nilius B, Huang Y et al
2 enhances hypotonicity-induced cytosolic Ca (2010) Depletion of intracellular Ca2þ stores
(2)(þ) rise in human induced pluripotent stem stimulates the translocation of vanilloid tran-
cell model of Hutchinson-Gillford Progeria. sient receptor potential 4-c1 heteromeric chan-
PLoS One 9:e87273 nels to the plasma membrane. Arterioscler
10. Entin-Meer M, Levy R, Goryainov P, Landa N, Thromb Vasc Biol 30:2249–2255
Barshack I et al (2014) The transient receptor 21. Leung PC, Cheng KT, Liu C, Cheung WT,
potential vanilloid 2 cation channel is abundant Kwan HY et al (2006) Mechanism of
in macrophages accumulating at the peri- non-capacitative Ca2þ influx in response to
infarct zone and may enhance their migration bradykinin in vascular endothelial cells. J Vasc
capacity towards injured cardiomyocytes fol- Res 43:367–376
lowing myocardial infarction. PLoS One 9: 22. Wong CO, Sukumar P, Beech DJ, Yao X
e105055 (2010) Nitric oxide lacks direct effect on
11. Hamill OP, Marty A, Neher E, Sakmann B, TRPC5 channels but suppresses endogenous
Sigworth FJ (1981) Improved patch-clamp TRPC5-containing channels in endothelial
techniques for high-resolution current record- cells. Pflugers Arch 460:121–130
ing from cells and cell-free membrane patches.
Pflugers Arch 391:85–100
Chapter 14
Abstract
Receptor tyrosine kinases (RTK) belong to a major class of cell surface receptors that transduce extracellular
signals to elicit diverse intracellular responses. Upon binding to specific ligand, the RTKs become dimerized
and autophosphorylated at tyrosine residues. This creates binding sites to recruit specific signaling inter-
mediates and hence trigger distinct signaling events. The cellular response to a given RTK may be modified
through the regulation of membrane insertion and receptor internalization. Here we use Trk receptor and
its ligand, the neurotrophin brain-derived neurotrophic factor (BDNF), as an example to illustrate the
approaches (coimmunoprecipitation and biotinylation) to study the surface expression and signal transduc-
tion mediated by this class of RTK in the nervous system.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_14, © Springer Science+Business Media, LLC 2018
211
212 Kwok-On Lai and Nancy Y. Ip
1.3 Regulation of Trk The number of RTK proteins expressed on cell surface is a crucial
Receptor Surface determinant of the cellular responsiveness to its cognate ligand, and
Expression the surface expression of RTK can be regulated at the level of
membrane insertion and receptor internalization. RTK internaliza-
tion is an important event following ligand binding, and can affect
ligand function by down-regulating the receptor signaling and/or
interaction with specific signaling proteins in distinct cellular com-
partments. For example, it was found that enhancing the rate of
NGF-TrkA internalization or disrupting TrkA endocytosis can dif-
ferentially affect the trophic and differentiation action of NGF on
PC12 cells [16, 17]. Interestingly, both the membrane insertion
and BDNF-induced internalization of TrkB is facilitated by neuro-
nal activity [18, 19]. This suggests that the response to BDNF is
greater in active neurons or synapses, and is consistent with the
Surface Receptor Tyrosine Kinase TrkB in Neurons 215
Fig. 1 Schematic diagram illustrating the coimmunoprecipitation of TrkB and the signaling protein TIAM1, a
guanine nucleotide exchange factor (GEF) for the small GTPase Rac1. Upon treatment of cortical neurons with
BDNF, TIAM1 is recruited to TrkB and this interaction depends on phosphorylation of TrkB at Ser-478 by the
serine/threonine kinase Cdk5 [14]. The interaction between TrkB and TIAM1 can be demonstrated by
immunoprecipitation by anti-TIAM1 antibody. After cell lysis in a mild detergent, the associated TrkB can
be coimmunoprecipitated with TIAM1 by the antibody. After captured by the Protein A sepharose beads, the
association between the two proteins is disrupted by elution with sample buffer and boiling. TrkB and TIAM1
will be separated by SDS-PAGE based on difference in molecular weight, and Western blotting with antibodies
against TrkB and TIAM1 will detect two different bands. In cortical neurons carrying a S478A mutation in
which the serine residue is substituted to alanine and hence deficient in phosphorylation, TIAM1 fails to
interact with TrkB, and the receptor will therefore not be captured by the TIAM1 antibody and Protein A
Sepharose beads. As a result, the band corresponding to TrkB will be absent in the Western blot
216 Kwok-On Lai and Nancy Y. Ip
2 Materials
2.1 Reagents 1. Medium for neurons: Primary cortical neurons were cultured
in Neurobasal medium (ThermoFisher Scientific, Carlsbad,
CA, USA) supplemented with 2% B27 (ThermoFisher Scien-
tific), 1 mM L-glutamine (ThermoFisher Scientific) and 10 mM
D-glucose (Sigma-Aldrich Chemical Company, St. Louis, MO,
USA). HEK-293T cells were cultured in Minimal Essential
Medium (MEM) (ThermoFisher Scientific) þ 10% fetal bovine
serum (FBS, ThermoFisher Scientific).
2. Dulbecco’s phosphate-buffered saline (DPBS) (ThermoFisher
Scientific).
3. Bovine serum albumin (BSA; Sigma-Aldrich).
4. Brain-derived neurotrophic factor (BDNF; PeproTech, Rocky
Hill, NH, USA).
5. Lipofectamine plus (ThermoFisher Scientific).
6. Bradford reagent (Bio-Rad, Hercules, CA, USA).
7. Buffer A for coimmunoprecipitation: 20 mM Tris, 50 mM
NaCl, 1 mM EDTA, 1 mM NaF, 0.5% Nonidet P-40
(NP-40) (v/v). A 2 solution is prepared.
8. Radioimmunoprecipitation assay (RIPA) buffer: 12 mM
sodium deoxycholate, 0.1% SDS; 1% NP-40 in Dulbecco’s
phosphate-buffered saline (DPBS). A 2 solution was
prepared, which was stored at 4 C. Dilute to 1 by DPBS
before use.
9. Protease and phosphatase inhibitors (purchased from Sigma-
Aldrich except sodium fluoride):
(a) Soybean trypsin inhibitor.
(b) Leupeptin.
(c) Aprotinin.
(d) Antipain.
(e) Okadaic acid.
(f) Benzamidine.
(g) Sodium orthovanadate.
(h) Phenylmethylsulfonyl fluoride (PMSF).
(i) Sodium fluoride. (A-Tech Global Science Limited, Hong
Kong, SAR China).
(j) Beta-glycerol phosphate.
10. To make RIPA plus protease and phosphatase inhibitors, pre-
pare the RIPA buffer with the following concentrations of
inhibitors:10 μg/mL soybean trypsin inhibitor, 10 μg/mL
leupeptin, 10 μg/mL aprotinin, 2 μg/mL antipain, 30 nM
Surface Receptor Tyrosine Kinase TrkB in Neurons 217
2.2 Tissue Culture 1. 293T (ATCC), primary rat cortical neurons (see Note 1).
Supplies 2. 60 and 100 mm tissue culture dishes.
3. Bel-Art Cell scrapers (VWR, Randor, PA, USA).
4. Eppendorf tubes (1.5 mL).
2.3 Antibodies Commercially available antibodies used were: TrkB (Western blot-
ting, BD Biosciences); TIAM1, phospho-S6K-Ser411 (Santa
Cruz); Phospho-TrkA (Y490), phospho-S6K-Thr-389, phospho-
S6-S235/236, S6K, S6 ribosomal protein were from Cell Signaling
Technology; secondary antibodies for Western blotting were horse-
radish peroxidase-conjugated goat antibodies to rabbit or mouse
and were purchased from Cell Signaling Technology.
218 Kwok-On Lai and Nancy Y. Ip
2.4 Equipment 1. CO2 Tissue culture incubator (NuAire, Plymouth, MN, USA).
2. Refrigerated microcentrifuge with speed up to 16,000 g.
3. Equipment to run SDS-PAGE gels (Gel caster: GE Healthcare
Life Sciences, Marlborough, MA, USA; SDS gel tank: Hoefer
Inc., Hollison, MA, USA; Protein transfer: Bio-Rad).
4. Cold room holding a Laboratory rocker (use cold room instead
of refrigerator).
5. Laboratory rocker.
6. Wheaton overhead stirrer with a 15 mL tissue grinder.
7. Spectrophotometer or microplate reader measuring absor-
bance at 595 nm.
8. pH meter.
9. Power Supply (similar to a MINI-300 or equivalent).
10. Weighing scale.
3 Methods
3.1 Phosphorylation 1. Primary rat cortical neurons (1.5 106 cells) were cultured
of Signaling Proteins onto 60 mm culture dishes. BDNF treatment was performed at
in Cortical Neurons 13–14 DIV (see Note 1).
2. Aspirate the medium, add 4 mL serum-free neurobasal
medium to each dish and incubate at 37 C for 2 h. This
starvation can lower the baseline phosphorylation of the recep-
tor and signaling proteins and make it easier to see the induc-
tion by BDNF.
3. Dilute BDNF in 0.1% BSA (in DPBS) to 100 ng/μL. Add 4 μL
BDNF or 0.1% BSA (control) for 5–30 min.
4. To extract total proteins, aspirate the medium. Immediately
put the dish on ice. Add 4 mL ice-cold DPBS.
5. Aspirate the DPBS. Add 0.25 mL ice-cold 1 RIPA plus
protease and phosphatase inhibitors. Collect the lysate using
cell scrapper, and transfer the lysate onto eppendorf tube.
6. Rock the lysate in cold room for 45 min.
7. Centrifuge the sample at 16,000 g at 4 C for 10 min.
8. Collect the supernatant. Measure the protein concentration by
Bradford reagent. The concentration of proteins should be
about 2 μg/μL.
9. Add 5 SDS sample buffer to the lysate. Boil the samples at
>95 C for 6 min right before SDS-PAGE (see Note 2).
Surface Receptor Tyrosine Kinase TrkB in Neurons 219
3.2 Coimmuno- Interaction between RTK and signaling proteins can be examined
precipitation Between either in transfected 293T cells, in primary rat cortical neurons, or
RTK and Signaling in whole brain homogenate. Both 293T cells and primary rat
Proteins (Fig. 1) cortical neurons (5 106 cortical neurons) were cultured onto
100 mm dishes. For transfected 293T cells, transfection was per-
formed using Lipofectamine plus. Cell lysate was collected 24 h
after transfection.
1. To extract total proteins from 293T cells or cortical neurons,
aspirate the medium. Immediately put the dish on ice. Add
4 mL ice-cold DPBS.
2. Aspirate the DPBS. Add 0.6 mL ice-cold 1 Buffer A that
contains the various protease and phosphatase inhibitors listed
in Subheading 3.1, step 5. Then follow steps 6–8 of Subhead-
ing 3.1 above.
3. Coimmunoprecipitation of HEK293T cell or whole brain
lysate was performed in Buffer A. Lysate (1 mg for
HEK293T cells, 2 mg for cultured cortical neurons) was
diluted to 600 μL with buffer A, and incubated with the
corresponding antibody (1–2 μg) in cold room rocking
overnight.
4. Gently vortex the protein A–Sepharose (if the antibody for
immunoprecipitation is polyclonal) or protein G–Sepharose
(if the antibody for immunoprecipitation is monoclonal) for
1 min. Aliquot 40 μL of the sepharose to each eppendorf tube.
Wash the sepharose 3 times with 0.6 mL DPBS. Centrifuge at
600 g at 4 C for 1 min between each wash.
5. Add the cell lysate in step 3 to the sepharose, and incubate the
samples in a cold room with constant rocking for 1 h.
6. Wash the sepharose with 0.6 mL buffer A (plus the protease
and phosphatase inhibitors) three times. Centrifuge at 600 g
at 4 C for 1 min between each wash.
7. After the last wash (see Note 3), resuspend the sepharose beads
with 35 μL 2 SDS sample buffer. Boil the samples at >95 C
for 6 min. Centrifuge at 16,000 g at room temperature for
1 min, and transfer the samples to a new eppendorf tube. Run
the samples in SDS-PAGE.
The coimmunoprecipitated proteins can be detected by West-
ern blot (see Notes 4 and 5).
3.3 Coimmuno- 1. If brain homogenate is used as the starting material, weigh the
precipitation Between mouse brains and place the tissues into the chilled 15 mL glass
RTK and Signaling tube for tissue grinder (see Note 6). Add 3 volumes (e.g., 3 mL
Proteins in Whole to 1 g brain tissues) of ice-cold DPBS that contains the protease
Brain Lysate and phosphatase inhibitors as indicated in Subheading 3.1,
step 5.
220 Kwok-On Lai and Nancy Y. Ip
2. Homogenize the tissues for 1 min, then chill on ice for 1 min;
repeat the homogenization step two more times.
3. Centrifuge at 1000 g at 4 C for 5 min.
4. Collect supernatant. Add equal volume of 2 buffer A with
protease and phosphatase inhibitors.
5. Rocking for 45 min at 4 C.
6. Spin down at 16,000 g for 10 min at 4 C.
7. Collect supernatant (homogenate). The homogenate (2 mg
proteins) is subjected to coimmunoprecipitation as in steps
3–7 of Subheading 3.2.
4 Notes
Acknowledgment
References
1. Ip NY, Yancopoulos GD (1996) The neurotro- signal transduction pathways. Annu Rev Cell
phins and CNTF: two families of collaborative Biol 10:251–337
neurotrophic factors. Ann Rev Neurosci 13. Cheung ZH, Chin WH, Chen Y, Ng YP, Ip NY
19:491–515 (2007) Cdk5 is involved in BDNF-stimulated
2. Lai KO, Fu WY, Ip FC, Ip NY (1998) Cloning dendritic growth in hippocampal neurons.
and expression of a novel neurotrophin, NT-7, PLoS Biol 5:e63
from carp. Mol Cell Neurosci 11:64–76 14. Lai KO, Wong AS, Cheung MC, Xu P, Liang Z,
3. Lu Y, Christian K, Lu B (2008) BDNF: a key Lok KC et al (2012) TrkB phosphorylation by
regulator for protein synthesis-dependent LTP Cdk5 is required for activity-dependent struc-
and long-term memory? Neurobiol Learn tural plasticity and spatial memory. Nat Neu-
Mem 89:312–323 rosci 15:1506–1515
4. Park H, Poo MM (2013) Neurotrophin regu- 15. Miyamoto Y, Yamauchi J, Tanoue A, Wu C,
lation of neural circuit development and func- Mobley WC (2006) TrkB binds and tyrosine-
tion. Nat Rev Neurosci 14:7–23 phosphorylates Tiam1, leading to activation of
5. Longo FM, Massa SM (2013) Small-molecule Rac1 and induction of changes in cellular mor-
modulation of neurotrophin receptors: a strat- phology. Proc Natl Acad Sci U S A
egy for the treatment of neurological disease. 103:10444–10449
Nat Rev Drug Discov 12:507–525 16. Saragovi HU, Zheng W, Maliartchouk S,
6. Reichardt LF (2006) Neurotrophin-regulated DiGugliemo GM, Mawal YR, Kamen A et al
signalling pathways. Philos Trans R Soc Lond B (1998) A TrkA-selective, fast internalizing
Biol Sci 361:1545–1564 nerve growth factor-antibody complex induces
7. Benedetti M, Levi A, Chao MV (1993) Differ- trophic but not neuritogenic signals. J Biol
ential expression of nerve growth factor recep- Chem 273:34933–34940
tors leads to altered binding affinity and 17. Zhang Y, Moheban DB, Conway BR,
neurotrophin responsiveness. Proc Natl Acad Bhattacharyya A, Segal RA (2000) Cell surface
Sci U S A 90:7859–7863 Trk receptors mediate NGF-induced survival
8. Kaplan DR, Hempstead BL, Martin-Zanca D, while internalized receptors regulate
Chao MV, Parada LF (1991) The trk proto- NGF-induced differentiation. J Neurosci
oncogene product: a signal transducing recep- 20:5671–5678
tor for nerve growth factor. Science 18. Du J, Feng L, Yang F, Lu B (2000) Activity-
252:554–558 and Ca(2þ)-dependent modulation of surface
9. Minichiello L (2009) TrkB signalling pathways expression of brain-derived neurotrophic factor
in LTP and learning. Nat Rev Neurosci receptors in hippocampal neurons. J Cell Biol
10:850–860 150:1423–1434
10. Minichiello L, Calella AM, Medina DL, 19. Du J, Feng L, Zaitsev E, Je HS, Liu XW, Lu B
Bonhoeffer T, Klein R, Korte M (2002) Mech- (2003) Regulation of TrkB receptor tyrosine
anism of TrkB-mediated hippocampal long- kinase and its internalization by neuronal activ-
term potentiation. Neuron 36:121–137 ity and Ca2þ influx. J Cell Biol 163:385–395
11. Gruart A, Sciarretta C, Valenzuela- 20. Zhao L, Sheng AL, Huang SH, Yin YX,
Harrington M, Delgado-Garcia JM, Mini- Chen B, Li XZ et al (2009) Mechanism under-
chiello L (2007) Mutation at the TrkB PLC lying activity-dependent insertion of TrkB into
{gamma}-docking site affects hippocampal the neuronal surface. J Cell Sci
LTP and associative learning in conscious 122:3123–3136
mice. Learn Mem 14:54–62 21. Zeitelhofer M, Vessey JP, Xie Y, Tubing F,
12. van der Geer P, Hunter T, Lindberg RA (1994) Thomas S, Kiebler M et al (2007) High-
Receptor protein-tyrosine kinases and their efficiency transfection of mammalian neurons
via nucleofection. Nat Protoc 2:1692–1704
Chapter 15
Abstract
Surface proteins localized on the apical and basal plasma membranes are required for a cell to sense its
environment and relay changes in ionic, cytokine, chemokine, and hormone levels to the inside of the cell.
In a polarized cell, surface proteins are differentially localized on the apical or the basolateral sides of the
cell. The retinal pigment epithelium (RPE) is an example of a polarized cell that performs a variety of
functions that are dependent on its polarized state including trafficking of ions, fluid, and metabolites across
the RPE monolayer. These functions are absolutely crucial for maintaining the health and integrity of
adjacent photoreceptors, the photosensitive cells of the retina. Here we present a series of approaches to
identify and validate the polarization state of cultured primary human RPE cells using immunostaining for
RPE apical/basolateral markers, polarized cytokine secretion, electrophysiology, fluid transport, phagocy-
tosis, and identification of plasma membrane proteins through cell surface capturing technology. These
approaches are currently being used to validate the polarized state and the epithelial phenotype of human
induced pluripotent stem (iPS) cell derived RPE cells. This work provides the basis for developing an
autologous cell therapy for age-related macular degeneration using patient specific iPS cell derived RPE.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_15, © Springer Science+Business Media, LLC 2018
223
224 Vladimir Khristov et al.
2 Materials
Table 1
Human fetal RPE medium components for preparation of 500 mL medium
2.4 Electro- 1. Ringer’s solution: The control Ringer’s solution used for apical
physiology and basal bath perfusion has the following composition
(in mM): NaCl 116.5, NaHCO3 23, KCl 5, MgCl2 0.5, Glu-
cose 5, Taurine 2, CaCl2 1.8, and sucrose 10. Prior to use, all
solutions are bubbled continuously with 8% CO2
(pH 7.4 0.05), and maintained at 37 1.0 C.
Polarized State of Human RPE 227
2.5 Polarized 1. Luminex xMAP magnetic bead assay kit (Millipore, Billerica,
Cytokine Secretion MA, USA).
Measurement 2. Luminex 200 analyzer (Luminex, Austin, TX, USA).
3. Bioplex manager TM 6.0 (Bio-Rad, Hercules, CA, USA).
2.6 Vectorial Fluid 1. 11 mm Trephine tissue hole punch (Lines Industries, Concord,
Transport CA, USA).
Measurement 2. 205 μm thickness Nylon mesh (SEFAR, Buffalo, NY, USA).
3. Capacitive probe (MT Instruments, Latham, NY, USA).
4. Agar bridges: 4.5% MEMα-agar bridges were used (catalog
number A7002) (Sigma, St. Louis, MO, USA) (see Note 2).
€
5. Modified Ussing chamber (custom-made).
€
6. Ussing chamber water jacket (custom-made).
7. Voltage-Current Clamp (VCC600; Physiologic Instruments).
6. 1 PBS.
7. 0.25% Trypsin–EDTA (ThermoFisher Scientific).
8. Fluorescence-activated cell sorting (FACS) buffer composed of
0.1% BSA (MP Biomedicals) in 1 PBS.
9. 40 μm cell strainer (BD Biosciences).
10. 40 ,6-diamidine-20 -phenylindole dihydrochloride (DAPI) solu-
tion (ThermoFisher Scientific).
11. Zeiss 780 Confocal Microscope or equivalent fluorescence
microscope with 20 magnification objective.
2.10 Western Blot 1. Trans-Blot Turbo RTA Transfer Kit, LF PVDF (catalog num-
ber 1704272) (Bio-Rad).
2. Trans-Blot Turbo Transfer System (catalog number 1704150)
(Bio-Rad).
3. Criterion Cell (catalog number 1656001XTU) (Bio-Rad).
230 Vladimir Khristov et al.
3 Methods
3.1 Maintaining 1. Start with RPE cells maintained in primaria flasks in an incuba-
and Splitting RPE cells tor (37 C, 5% CO2).
(See Note 5) [7–9] 2. To split cells from single 25 cm2 primaria flask, trypsinize cells
in 5 mL trypsin–EDTA for 20 min at 37 C, centrifuge at
400 g for 5 min, and resuspend the cells in 10 mL 15%
FBS containing RPE medium.
3. Put cell suspension into two 25 cm2 primaria flasks and keep
the flask in an incubator (37 C, 5% CO2) overnight.
4. Replace the medium after 1 day with 5% FBS containing RPE
medium. Change medium every 2 days.
5. After 4–5 weeks, trypsinize cells in 5 mL trypsin–EDTA for
20 min at 37 C, centrifuge at 400 g for 5 min, and resus-
pend the cells in 15% FBS containing RPE medium.
6. In some cases, repeat the trypsinization procedure a second
time to collect the cells that do not detach after the first
trypsinization.
7. Before seeding, coat 12 or 75 mm transwells with human
extracellular matrix (100 μg in 2 mL HBSS per well) and
expose them to UV light for 2 h in the hood.
8. Seed the cells onto clear cell culture inserts at a density of
1.5 105 cells per 12 mm diameter transwell for functional
analysis or at 1.0–1.5 107 per 75 mm diameter transwell for
CSC analysis.
9. The same protocol (excluding coating with ECM) is used to
culture cells on the flasks to generate the P1 population of cells
(see Note 6).
10. RPE cells are used for experiments when they reach a total
tissue resistance of >300 Ω cm2 (see Subheading 3.3.1) and
are uniformly pigmented.
3.2 Ensuring RPE A polarized confluent RPE monolayer has structural features (such
Monolayer Polarization as apical processes/basal infoldings, apically localized pigment
granules and basally located nuclei) and special characteristic prop-
erties (such as polarized distribution of certain ion channels, trans-
porters, and receptors on RPE apical or basolateral membranes to
mediate specific electrical responses, ability of the monolayer to
Polarized State of Human RPE 231
3.2.2 RPE Protein Marker Differentially localized proteins provide specialized functions in
Immunostaining different compartments of a cell (see Note 7). For instance,
EZRIN an apically located protein is a structural component of
RPE apical processes [10] and COLLAGEN IV a basally located
protein is a part of RPE basement membrane [11]. Figure 2 shows
expected localization of EZRIN and COLLAGEN IV in a mono-
layer of primary RPE cells.
1. Rinse RPE cells grown as a polarized monolayer on 12 mm
transwell three times in 1 PBS for 1 min each.
2. Incubate in fixation solution for 15 min exactly.
3. Rinse three times in 1 PBS for 1 min each.
4. Embed RPE monolayer in OCT cryosectioning medium based
on manufacturer specifications.
5. Make 10 μm cryosections on cryostat following manufacturer
instructions, and mount on slides.
6. Incubate in blocking and permeabilization solution 1 h at RT.
7. Add primary antibody (diluted 1:100–1:1000) diluted in
blocking and permeabilization solution, and incubate over-
night at 4 C.
8. Wash three times in washing solution for 15 min each.
9. Add fluorophore-conjugated secondary antibody diluted in
ICC buffer, and incubate for 1 h at room temperature in the
dark, with gentle shaking.
Fig. 2 Polarized distribution of known RPE markers in the monolayer. Immunofluorescence localization of RPE
basolateral marker Collagen IV (a) and apical marker Ezrin (b) in primary cultures hfRPE grown on transwell
inserts. DAPI (blue) labels the nuclei located close to the basolateral membrane
Polarized State of Human RPE 233
10. Wash three times in washing solution for 15 min each, with
gentle shaking.
11. Mount samples on glass slides using Fluoromount-G and ana-
lyze via microscopy.
3.3.1 Functional 1. Take out a primary cultured hfRPE transwell and cut out a
Validation of Cell Surface piece of RPE monolayer with a 7 mm skin biopsy punch.
Receptors and Channels 2. Mount the RPE monolayer on a modified Ussing € chamber (see
Using Electrophysiology Note 8) [12, 14, 15]. Perfuse Ringer’s solution (pH 7.4,
37 C) to the apical and basal baths of the chamber separately.
3. Connect the calomel electrodes in series with agar bridges to
Ringer’s solution of the chamber to make electrical contact
with each bathing solution.
4. Measure the transepithelial potential (TEP) of the tissue under
CURRENT mode of the Voltage-Current Clamp (see Note 9).
5. Pass 2–4 μA current pulse (bipolar, with a period of 3 s applied
at 40 s intervals) across the tissue mounted in the Ussing€
chamber, measure the resultant changes in TEP to obtain the
total tissue resistance (RT).
6. Dissolve ATP or ClC2 channel activator (lubiprostone) in
Ringer’s solution to reach the final working concentration,
and perfuse it into either the apical or basal bath to detect the
activity of P2Y2 receptors or ClC2 channels on the RPE mem-
brane (see Figs. 3 and 4).
3.3.2 Polarized Cytokine RPE is located between the neural retina and the choroid. These
Secretion Measurement three different cell layers form the homeostatic unit at the back of
the eye to maintain visual function. The polarized secretion of
VEGF on the basal side and PEDF on the apical side by RPE
monolayer is required for the maintenance of the health and integ-
rity of choroid and retina respectively [13, 16] (see Note 10). The
234 Vladimir Khristov et al.
Fig. 3 Differential electrical responses evoked by apical or basal application of ATP. The continuous trace
represents transepithelial potential (TEP) and open squares represent total tissue resistance (RT). The apical or
basal bath perfusion of 100 μM ATP is indicated by black bars above the graph. A time scale bar is located at
the bottom of the graph. In the native tissue, ATP acts on purinergic P2Y2 receptors on RPE apical membrane,
increasing intracellular IP3 and thus stimulating Ca2+ release from the endoplasmic reticulum, to cause
activation of basolateral membrane Ca2+-activated Cl channels followed by a decrease in apical membrane
K+ conductance, thus leading to electrical responses across the RPE monolayer [14]. Consistent with this,
upon the apical application in the primary cultures RPE, ATP produced a dramatic biphasic response in TEP
and a markedly decrease in RT; but induced much smaller TEP responses with little RT changes after basal
perfusion, suggesting the majority of P2Y2 receptors are located on RPE apical membrane
Fig. 4 Differential electrical responses evoked by apical or basal application of the ClC2 channel activator. The
ClC2 activator (10 μM) produced a similar increase in TEP and a decrease in RT upon either the apical or basal
bath application, suggesting the basal localization of ClC2 channels in RPE. The activation of ClC2 on the RPE
basal membrane leads to the efflux of intracellular Cl and depolarization of RPE basolateral membrane, thus
causing TEP increase and RT drop. The larger effect induced by apical application may be due to the much
larger RPE apical surface area
Polarized State of Human RPE 235
6000
APICAL BASAL
4000
3000
2000
1000
0
Line A Line B Line C
Fig. 5 Polarized cytokine secretion to determine the polarity of iPSC-derived RPE monolayer. RPE from three
different RPE samples, namely Line A, B, and C, were tested for VEGF secretion on apical and basal side. RPE
monolayers were fed with fresh media 24 h before the media collection. The collected media was spun down
and used for the analysis. The results demonstrate the RPE monolayers grown in vitro secrete the VEGF
cytokine in a manner replicating in vivo function
3.3.3 Vectorial Fluid An essential function of the RPE monolayer is unidirectional trans-
Transport Measurement port of water from the subretinal space to the choriocapillaris. This
€
fluid flow (JV) can be measured (Fig. 6) using a modified Ussing
chamber as described previously [17, 18].
1. Set incubator temperature to 39 C, CO2 to 5% and relative
humidity to 50%.
€
2. Set Ussing chamber water jacket to 36 C.
3. Place all solutions to be used in an incubator at least 1 h ahead
of time to equilibrate the CO2 and temperature conditions.
236 Vladimir Khristov et al.
Fig. 6 Fluid transport response induced by apical application of ATP. Top panel: Transepithelial fluid transport
(JV). Bottom panel: Black trace represents transepithelial potential (TEP) and red trace represents transe-
pithelial resistance (RT). Application of ATP is indicated by the black bar above the top graph. The apical
(AP) application of 100 μM ATP activated the P2Y2 receptors on RPE apical membrane, which resulted in a
reversible increase in apical-to-basal fluid adsorption. Time scale bar indicated 50 min
3.3.4 Phagocytosis RPE cells are among the most active phagocytic cells in the human
Assay body. Primary RPE should possess the same phagocytic machinery
as native RPE. Apically localized phagocytic receptors allow healthy
RPE phagocytose photoreceptor outer segments (POS), remove
metabolic waste and recycle visual pigment. The ability of RPE cells
to phagocytose POS in vitro is indicative of a healthy, polarized
monolayer [16, 19] (Fig. 7).
Polarized State of Human RPE 237
Fig. 7 Phagocytic capability of polarized primary hfRPE monolayer. Fluorescently labeled photoreceptor outer
segments (POS) were observed in hfRPE cells indicating its normal phagocytic capability
3.4 Cell Surface Extracellular, N-linked glycoproteins were labeled and captured
Capturing (CSC) using CSC technology [20–22] (see Note 11). The
Technology CSC-technology uses meta-sodium periodate to oxidize carbohy-
drates on intact cells and followed by covalent chemical labeling of
oxidized carbohydrate-containing proteins with biotin for affinity
enrichment. To selectively identify apical or basolateral surface
238 Vladimir Khristov et al.
Fig. 10 Immunostaining of a novel RPE cell maker. Immunofluorescence localization of a novel RPE marker,
SLC5A12 (red) and F-actin (green) in cultured hfRPE (a) and fibroblasts (b)
Fig. 11 Western blot of a novel RPE marker GRIK1 is enriched in the membrane and cytosolic fractions of
hfRPE cells. (A) Nuclear wash buffer, (B) nuclear fraction, (C) mitochondrial fraction, (D) cytosolic fraction,
(E) membrane fraction
240 Vladimir Khristov et al.
Fig. 12 Western blot of known RPE marker GPR143 is enriched in the membrane
fraction of hfRPE cells. (A) Nuclear wash buffer, (B) nuclear fraction,
(C) mitochondrial fraction, (D) cytosolic fraction, (E) membrane fraction
3.4.1 Cell Surface 1. Labeling of human fetal RPE cells is performed on either the
Capturing (See Note 12) apical or the basal plasma membranes.
2. Aspirate the cultivation medium and wash both the apical and
basolateral membranes of RPE cells in transwells with labeling
buffer. Repeat twice to remove debris and as many dead cells as
possible.
3. Oxidize either the apical or the basal membrane by treating the
designated membrane with 0.2 mg/mL sodium metaperiodate
for 15 min at 4 C in the dark with gentle rotation. Incubate
the other membrane with labeling buffer (see Note 13).
4. Wash cells twice to remove periodate solution. Add 4 mg/mL
biocytin hydrazide or labeling buffer to the respective mem-
branes of the RPE cells for 1 h at 4 C.
5. Gently wash the cells twice with labeling buffer. Collect all RPE
cells by gently scraping the transwell with a cell scraper. Add
additional labeling buffer and continue scraping until all cells
have been isolated.
6. Scraped cells should be transferred to a 50 mL Falcon tube
using a sterile Pasteur pipette.
7. Centrifuge the cells at 800 g. Discard supernatant and wash
with labeling buffer. Recentrifuge.
8. Aspirate the supernatant and resuspend the pellet in 4 mL of
hypotonic lysis buffer on ice. Incubate 15 min.
9. Transfer cells to a Dounce homogenizer and gently homoge-
nize on ice through 35 strokes of the teflon pestle.
10. Transfer the homogenized cells to a 15 mL polypropylene tube
on ice and centrifuge at 800 g for 10 min. Remove tube and
place on ice (see Note 14).
11. Transfer the supernatant, without disturbing the pellet, to a
new tube. Resuspend the pellet in an additional 2 mL hypo-
tonic lysis buffer and rehomogenize the resuspended pellet on
ice with 35 strokes of the teflon pestle (see Note 15).
12. Repeat steps 10 and 11. After the third homogenization and
centrifugation, it usually becomes possible to distinguish
between the cell pellet and the supernatant.
Polarized State of Human RPE 241
3.4.2 Mass 1. For each biological sample, analyze three technical replicates by
Spectrometry, Database liquid chromatography tandem mass spectrometry (LC-MS/
Search, and Data MS) on an LTQ-Orbitrap Velos or equivalent as previously
Processing reported [35, 36].
2. Use three independent search engines, Sequest, X!Tandem
[37], and Morpheus [38], followed by manual validation of
spectra as previously described [35]. Compare PSC (Protein
surface classification) to non-PSC surface proteomes and incor-
poration of transcriptome data using ProteinCenter™.
3. Confirm the unambiguous identification of an occupied glyco-
site based on the identified peptide meeting the following three
criteria: (1) peptide was captured by streptavidin beads, indi-
cating it was originally attached to a biotin-labeled oligosaccha-
ride structure, (2) the capture peptide contains a deamidation
(0.98 Da shift; conversion of N!D), and (3) the deamidation
occurred at asparagine within the N-glycosylation consensus
amino acid sequence motif (NxS/T). These criteria allow for
the efficient filtering of protein “contaminants” derived from
intracellular membranes and determination of the orientation
of transmembrane proteins within the membrane. Proteins that
do not meet these criteria are not analyzed further.
3.4.3 Bioinformatics 1. The above approach when applied to cultured human fetal RPE
to Identify Potential RPE cells has yielded over 1800 cell surface proteins. Of these,
Surface Markers 488 proteins have been detected only on the apical membrane,
Polarized State of Human RPE 243
3.4.4 Validation of Cell 1. Scrape and resuspend cells from one T25 flask of primary
Surface Markers Through human cells using 1 mL Lysis Buffer.
Western Blot 2. Ensure complete cell lysis by passing the lysate through a 25 G
needle 100 times using a 1 mL syringe.
3. Leave lysate containing tube on ice for 20 min.
4. Centrifuge the nuclear pellet (P1) at 720 g for 5 min. The
nuclear pellet should be washed once by adding 500 μL frac-
tionation buffer again.
5. Disperse the pellet with a pipette and pass through a 25 G
needle 10 times.
6. Centrifuge again at 720 g for 10 min. Remove the fraction-
ation buffer and resuspend the nuclear pellet in the nuclear
buffer.
7. Sonicate the nuclear pellet briefly (3 s) on ice at a power setting
of 2-continuous to fractionate chromatin.
8. Remove the supernatant and place in a fresh 1.5 mL
microcentrifuge tube.
9. Centrifuge the supernatant again at 10,000 g. Move the
supernatant containing the cytosolic and membrane fractions
to a fresh 1.5 mL microcentrifuge tube.
244 Vladimir Khristov et al.
4 Notes
Acknowledgment
References
1. Strauss O (2005) The retinal pigment epithe- 11. Braisted JE, Essman TF, Raymond PA (1994)
lium in visual function. Physiol Rev 85 Selective regeneration of photoreceptors in
(3):845–881 goldfish retina. Development 120
2. Sparrow JR, Hicks D, Hamel CP (2010) The (9):2409–2419
retinal pigment epithelium in health and dis- 12. Baron R, Joseph RD, Owen T, Tennyson J,
ease. Curr Mol Med 10(9):802–823 Miller S, Ballester GE (1991) Imaging Jupiter’s
3. Chiba C (2014) The retinal pigment epithe- aurorae from H3þ emissions in the 3-4 micro-
lium: an important player of retinal disorders meters band. Nature 353:539–542
and regeneration. Exp Eye Res 123:107–114 13. Sonoda S, Sreekumar PG, Kase S, Spee C, Ryan
4. Reichhart N, Strauss O (2014) Ion channels SJ, Kannan R, Hinton DR (2010) Attainment
and transporters of the retinal pigment epithe- of polarity promotes growth factor secretion by
lium. Exp Eye Res 126:27–37 retinal pigment epithelial cells: relevance to
5. Lehmann GL, Benedicto I, Philp NJ, age-related macular degeneration. Aging
Rodriguez-Boulan E (2014) Plasma mem- (Albany NY) 2(1):28–42
brane protein polarity and trafficking in RPE 14. Maminishkis A, Jalickee S, Blaug SA, Rymer J,
cells: past, present and future. Exp Eye Res Yerxa BR, Peterson WM, Miller SS (2002) The
126:5–15 P2Y(2) receptor agonist INS37217 stimulates
6. Kay P, Yang YC, Paraoan L (2013) Directional RPE fluid transport in vitro and retinal reat-
protein secretion by the retinal pigment epithe- tachment in rat. Invest Ophthalmol Vis Sci 43
lium: roles in retinal health and the develop- (11):3555–3566
ment of age-related macular degeneration. J 15. Peterson WM, Meggyesy C, Yu K, Miller SS
Cell Mol Med 17(7):833–843 (1997) Extracellular ATP activates calcium sig-
7. Maminishkis A, Chen S, Jalickee S, Banzon T, naling, ion, and fluid transport in retinal pig-
Shi G, Wang FE et al (2006) Confluent mono- ment epithelium. J Neurosci 17(7):2324–2337
layers of cultured human fetal retinal pigment 16. Kevany BM, Palczewski K (2010) Phagocytosis
epithelium exhibit morphology and physiology of retinal rod and cone photoreceptors. Physi-
of native tissue. Invest Ophthalmol Vis Sci 47 ology (Bethesda) 25(1):8–15
(8):3612–3624 17. Li R, Maminishkis A, Banzon T, Wan Q,
8. Maminishkis A, Miller SS (2010) Experimental Jalickee S, Chen S, Miller SS (2009) IFN
models for study of retinal pigment epithelial {gamma} regulates retinal pigment epithelial
physiology and pathophysiology. J Vis Exp fluid transport. Am J Physiol Cell Physiol 297
45:2032 (6):C1452–C1465
9. Miyagishima KJ, Wan Q, Corneo B, Sharma R, 18. Li R, Wen R, Banzon T, Maminishkis A, Miller
Lotfi MR, Boles NC et al (2016) In pursuit of SS (2011) CNTF mediates neurotrophic factor
authenticity: induced pluripotent stem cell- secretion and fluid absorption in human retinal
derived retinal pigment epithelium for clinical pigment epithelium. PLoS One 6(9):e23148
applications. Stem Cells Transl Med 5 19. Mazzoni F, Safa H, Finnemann SC (2014)
(11):1562–1574 Understanding photoreceptor outer segment
10. Kivela T, Jaaskelainen J, Vaheri A, Carpen O phagocytosis: use and utility of RPE cells in
(2000) Ezrin, a membrane-organizing protein, culture. Exp Eye Res 126:51–60
as a polarization marker of the retinal pigment 20. Boheler KR, Bhattacharya S, Kropp EM,
epithelium in vertebrates. Cell Tissue Res 301 Chuppa S, Riordon DR, Bausch-Fluck D et al
(2):217–223 (2014) A human pluripotent stem cell surface
N-glycoproteome resource reveals markers,
Polarized State of Human RPE 247
extracellular epitopes, and drug targets. Stem 30. Chen YY, Lu QJ, Lu QX, Wang NL (2011)
Cell Rep 3(1):185–203 Gene expression profile changes caused by the
21. Doerr A (2009) Snapshots of the cell surface. dysfunction of Mer during retinal pigment epi-
Nat Methods 6(6):401–401 thelium phagocytosis. Chin Med J 124
22. Wollscheid B, Bausch-Fluck D, Henderson C, (8):1145–1155
O’Brien R, Bibel M, Schiess R, Aebersold R, 31. Mandal MN, Vasireddy V, Jablonski MM,
Watts JD (2009) Mass-spectrometric identifi- Wang X, Heckenlively JR, Hughes BA, Reddy
cation and relative quantification of N-linked GB, Ayyagari R (2006) Spatial and temporal
cell surface glycoproteins. Nat Biotechnol 27 expression of MFRP and its interaction with
(4):378–386 CTRP5. Invest Ophthalmol Vis Sci 47
23. Strunnikova NV, Maminishkis A, Barb JJ, (12):5514–5521
Wang F, Zhi C, Sergeev Y et al (2010) Tran- 32. Lin H, Kenyon E, Miller SS (1992)
scriptome analysis and molecular signature of Na-dependent pHi regulatory mechanisms in
human retinal pigment epithelium. Hum Mol native human retinal pigment epithelium.
Genet 19(12):2468–2486 Invest Ophthalmol Vis Sci 33(13):3528–3538
24. Surace EM, Angeletti B, Ballabio A, Marigo V 33. Hollborn M, Petto C, Steffen A, Trettner S,
(2000) Expression pattern of the ocular albi- Bendig A, Wiedemann P, Bringmann A, Kohen
nism type 1 (Oa1) gene in the murine retinal L (2009) Effects of thrombin on RPE cells are
pigment epithelium. Invest Ophthalmol Vis Sci mediated by transactivation of growth factor
41(13):4333–4337 receptors. Invest Ophthalmol Vis Sci 50
25. Bachner D, Schroder D, Gross G (2002) (9):4452–4459
mRNA expression of the murine glycoprotein 34. Sugasawa K, Deguchi J, Okami T,
(transmembrane) nmb (Gpnmb) gene is linked Yamamoto A, Omori K, Uyama M, Tashiro Y
to the developing retinal pigment epithelium (1994) Immunocytochemical analyses of distri-
and iris. Brain Res Gene Expr Patterns 1 butions of Na, K-ATPase and GLUT1, insulin
(3–4):159–165 and transferrin receptors in the developing ret-
26. Adijanto J, Banzon T, Jalickee S, Wang NS, inal pigment epithelial cells. Cell Struct Funct
Miller SS (2009) CO2-induced ion and fluid 19(1):21–28
transport in human retinal pigment epithelium. 35. Gundry RL, Riordon DR, Tarasova Y,
J Gen Physiol 133(6):603–622 Chuppa S, Bhattacharya S, Juhasz O et al
27. Young A, Powelson EB, Whitney IE, Raven (2012) A cell surfaceome map for immunophe-
MA, Nusinowitz S, Jiang M et al (2008) notyping and sorting pluripotent stem cells.
Involvement of OA1, an intracellular GPCR, Mol Cell Proteomics 11(8):303–316
and G alpha i3, its binding protein, in melano- 36. Gundry RL, White MY, Murray CI, Kane LA,
somal biogenesis and optic pathway formation. Fu Q, Stanley BA, Van Eyk JE (2009) Prepara-
Invest Ophthalmol Vis Sci 49(7):3245–3252 tion of proteins and peptides for mass spec-
28. Stamer WD, Bok D, Hu J, Jaffe GJ, McKay BS trometry analysis in a bottom-up proteomics
(2003) Aquaporin-1 channels in human retinal workflow. Curr Protoc Mol Biol Chapter 10:
pigment epithelium: role in transepithelial Unit10.25
water movement. Invest Ophthalmol Vis Sci 37. Craig R, Beavis RC (2004) TANDEM: match-
44(6):2803–2808 ing proteins with tandem mass spectra. Bioin-
29. Lambooij AC, van Wely KH, Lindenbergh- formatics 20(9):1466–1467
Kortleve DJ, Kuijpers RW, Kliffen M, Mooy 38. Wenger CD, Coon JJ (2013) A proteomics
CM (2003) Insulin-like growth factor-I and search algorithm specifically designed for
its receptor in neovascular age-related macular high-resolution tandem mass spectra. J Prote-
degeneration. Invest Ophthalmol Vis Sci 44 ome Res 12(3):1377–1386
(5):2192–2198
Chapter 16
Abstract
We present a method to capture mesenchymal stromal cells (MSCs) by adhesion to extracellular matrix
(ECM) molecules under flow conditions. The technique simulates a physiological system and exploits the
natural biological interactions of cells, through integrin receptors, with their ECM. The system offers an
insight into how MSCs could be targeted/localized to the site of interest (graft) following intravenous
injection.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_16, © Springer Science+Business Media, LLC 2018
249
250 Teresa Massam-Wu et al.
2 Materials
2.1 Ibidi Equipment 1. ibidi Pump System (catalog number 10902) (ibidi GmbH,
Munich, Germany).
2. Fluidic unit (catalog number 10903) (ibidi).
3. ibidi Pump (catalog number 10905) (ibidi).
4. PumpControl software (catalog number 10908) (ibidi).
2.2 Ibidi Labware 1. ibidi μ-slides I 0.6 Luer, tissue culture treated and sterilised
(catalog number 80186) (ibidi).
2. Perfusion set WHITE (catalog number 10963) (ibidi) (see
Note 1).
3. Dehydrated silica beads (see Note 2).
2.3 Microscopes 1. Live cell imaging microscope. Images were acquired on a Zeiss
and Camera Axiovert 200M live cell-imaging microscope (Carl Zeiss AG,
Oberkochen, Germany) under brightfield with an LED light
source (Zeiss Colibri) using a 20/0.30 LD A-Plan objective
(Zeiss). Images were taken at 500 ms intervals on a CoolSnap
HQ2 CCD camera (Photometrics).
2. High content imaging system Pathway Bioimager 855 (Beck-
ton Dickenson and Company, Sparks, MD, USA). Images were
acquired on a Pathway Bioimager 855 (Beckton Dickenson)
with laser autofocus using a 4/0.13 UPlan FLN objective
and the following filter setup: DAPI Ex. 360/10, Di. 400D
CLP, Em. 435 LP. Images were collected from the entire chan-
nel on an Orca ER camera (Hammamatsu Photonics, Hama-
matsu, Japan) with an offset from the channel center and a
montage of 3 26 was created without gaps. An exposure time
of 0.6 ms and threshold masks were applied to each image
using the automatic feature of the software. The images were
then processed and analyzed through the BD Data Explorer
software.
2.4 Other Equipment 1. CASY cell counter (Roche Diagnostics, Indianapolis, IN, USA)
(see Note 3).
2. Vacuum desiccator.
3. NanoDrop 2000 spectrophotometer (ThermoFisher Scientific,
Carlsbad, CA, USA).
4. HiSeq2500 System (Illumina, San Diego, CA, USA).
5. Beckman Coulter Cyan ADP (Beckman Coulter, Inc., Brea,
CA, USA). Samples were analyzed on a Beckman Coulter Cyan
ADP running Summit software version 4.3. Both Alexa Fluor
488 and Phycoerythrin were excited using the 488 nm laser.
ECM-Based Capture of MSCs 251
2.5 Image Analysis 1. Download Fiji image analysis software from http://fiji.sc/Fiji.
Software [2] 2. Download a media player such as VLC Media player from
http://www.videolan.org/vlc/index.en_GB.html.
2.6 Mesenchymal 1. Human MSCs (Catalog number PT-2501) (Lonza, Inc., Allen-
Stem Cells (MSCs) dale, NH, USA).
2.9 Antibodies MSCs for flow cytometry were labeled with either unconjugated
primary antibodies:
1. Mouse IgG2b κ Isotype Control (catalog number 555743).
2. CD14 [MP9] (catalog number 347490).
3. CD45 [2D1] (catalog number 347460).
4. CD73 [AD2] (catalog number 550256).
5. CD166 (catalog number 559260) (BD Pharmingen).
6. CD34 (catalog number CBL555) (Chemicon).
7. CD90 [5E10] (catalog number 328102) (Biolegend) and sec-
ondary Alexa Fluor 488 Donkey anti-Mouse IgG antibody
(catalog number A-21202) (Life Technologies)
(or)
8. Primary Stro-1 antibodies (catalog number MAB1038) (R&D
Systems) and secondary Alexa Fluor 488 Goat anti-Mouse IgM
antibody (catalog number A-21042) (Life Technologies)
or
9. Directly conjugated Phycoerythrin antibodies: PE Mouse
IgG2b κ Isotype Control (catalog number), CD29 (catalog
number 555443), CD44 [G44-26] (catalog number 555479)
(BD Pharmingen), CD105 [166707] (catalog number
FAB10971P) (R&D Systems).
3 Methods
3.2 Live Cell Imaging All experiments conducted at 37 C unless otherwise stated.
of MSCs Captured by
1. Dehydrate the silica beads in the drying bottle (orange) (see
Laminin-521 and FN III Note 2).
7–14 Under Flow
Conditions
2. Coat μ-slides I (600 μm channel height) Luer with 150 μL of
100 nM ECM molecules in PBS with or without CaCl2/
MgCl2 (see Note 4) overnight at 4 C (see Note 8).
3. Block unbound surfaces by gently exchanging the protein
solution with 10 mg/mL BSA for 1 h at room temperature
(see Note 9).
254 Teresa Massam-Wu et al.
A B
C
BODIPY DAPI Merged
Control
Adipogenesis
ä
Fig. 1 Classification of MSCs. Human mesenchymal stem cells (MSCs) expressing (a) negative (CD14, CD34,
and CD45) and (b) positive markers (CD29, CD44, CD73, CD90, CD105, CD166, and Stro-1) by flow cytometry
that conform to the criteria defined by the Mesenchymal and Tissue Stem Cell Committee of the International
Society for Cellular Therapy [5]. IgG1 and IgM control peaks are highlighted in red. (c) Adipogenic and (d)
Osteogenic differentiation of human mesenchymal stromal cells (MSCs) in vitro for 28 days using the human
MSC functional identification kit (catalog number SC006) (R&D Systems). Adipogenic MSCs were stained with
BODIPY 493/503 (green), Osteogenic MSCs with osteocalcin (green) and nuclei stained with DAPI (blue).
Un-treated Human MSCs were cultured in basal medium for 28 days. Scale bar ¼ 50 mm. Representative data
and images from two independent experiments
256 Teresa Massam-Wu et al.
positive pressure
Direction of flow
Extracellular matrix
Mesenchymal stem
molecule coated
cells in suspension
surface
Fig. 2 Schematic illustration of the ibidi fluidic pump system under positive pressure. Adapted from ibidi
17. Reattach the clamp to the tubing and disconnect the slide from
the perfusion system.
18. Remove unattached cells in the channel by gently washing once
with 500 μL PBS without CaCl2/MgCl2 (see Note 9).
3.4 Quantitation 1. Image the number of cells attached imaged using a BD Path-
of Immobilized way microscope with a 4 objective (see Note 17).
Cells (see Fig. 3b) 2. Quantify the number of cells attached using Fiji image analysis
software.
3. Open the TIFF image in Fiji.
4. Go to IMAGE then ADJUST the THRESHOLD.
5. Slide bar to the LEFT or RIGHT to ensure nuclei are distinct
then go to APPLY.
ECM-Based Capture of MSCs 257
3.5 RNA Extraction 1. Extract the RNA as per manufacturer’s instructions (see
Note 19).
2. Quantify and assess the purity of the extracted RNA sample
using a NanoDrop spectrophotometer (see Note 20).
3.6 Generating AVI Videos are generated from images from the first 7 min of a 10 min
Videos of Images experiment and are 2.5 the speed of real time.
Captured in a 10 min
1. Open Fiji imaging software.
Experiment
2. DRAG and DROP file of images onto symbol.
258 Teresa Massam-Wu et al.
4 Notes
Acknowledgment
References
1. Ball SG, Worthington JJ, Canfield AE, Merry (2005) Homotypic fibrillin-1 interactions in
CLR, Kielty CM (2014) Mesenchymal stromal microfibril assembly. J Biol Chem 280
cells: inhibiting PDGF receptors or depleting (6):5013–5021
fibronectin induces mesodermal progenitors 5. Dominici M, Le Blanc K, Mueller I, Slaper-
with endothelial potential. Stem Cells 32 Cortenbach I, Marini FC, Krause DS et al
(3):694–705 (2006) Minimal criteria for defining multipotent
2. Schindelin J, Arganda-Carreras I, Frise E, mesenchymal stromal cells. The International
Kaynig V, Longair M, Pietzsch S et al (2012) Society for Cellular Therapy position statement.
Fiji: an open-source platform for biological- Cytotherapy 8(4):315–317
image analysis. Nat Methods 9(7):676–682 6. Kroutvar M, Guttenberg Z (2013) Ibidi Pump
3. Cain SA, Baldock C, Gallagher J, Morgan A, Bax System Instructions version 1.5.1, revision
DV, Weiss AS, Shuttleworth CA, Kielty CM 3. http://ibidi.com/fileadmin/products/
(2005) Fibrillin-1 interactions with heparin: instruments/I_1090X_PumpSystem/IN_
implications for microfibril and elastic fiber 1090X_pump_system.pdf. Accessed 09 Jan
assembly. J Biol Chem 280(34):30526–30537 2015
4. Marson A, Rock MJ, Cain SA, Freeman LJ,
Morgan A, Mellody K, Shuttleworth CA et al
Chapter 17
Abstract
Use of experimentally derived induced pluripotent stem cells (iPSCs) has led to the development of cell
models for differentiation, drug testing and understanding disease pathogenesis. For these models to be
informative, reprogrammed cell lines need to be adequately characterized and shown to preserve all of the
critical characteristics of pluripotency and differentiation. Here, we report a detailed protocol for the
generation of iPSCs from human fibroblasts containing mutations in COL3A1 using a Sendai virus
mediated integration-free reprogramming approach. We describe how to characterize the putative iPSCs
in vivo and in vitro to ensure potency and differentiation potential. As an example of how these mutations
may affect cell surface and extracellular matrix (ECM) interactions, we provide protocols for the differenti-
ation of these cells into smooth muscle cells to illustrate how different cell types may display cell autono-
mous differences in collagen receptors that may affect their phenotype. These cells, when applied to
mechanical model systems (see Chapter 18 by Bose et al.) facilitate an assessment of stiffness and stress-
strain relationships useful for understanding how extracellular matrix dysfunction and its interactions with
surface proteins contribute to disease processes.
Key words Induced pluripotent stem cells, Reprogramming, Differentiation, Smooth muscle cells,
Collagen, Integrin
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_17, © Springer Science+Business Media, LLC 2018
261
262 Jiaozi He et al.
medium [21], and the resulting cells are useful for the study of
collagen and cell surface collagen receptors. Depending on the
cultivation medium, these SMCs can be either proliferative or
contractile. The latter cells, when applied to the three dimensional
model system described in the next chapter of this book (Fabrica-
tion and mechanical properties measurements of 3D microtissues for
the study of cell-matrix interactions), allow for an assessment of
stiffness and stress-strain relationships useful for understanding
how extracellular matrix dysfunction and its interactions with sur-
face proteins contribute to disease processes.
2 Materials
2.3 Tissue Culture 1. Tissue culture plates and dishes (96-, 24-, 6-well plates), Corn-
Reagents and Media ing brand (Sigma-Aldrich).
2. Ultra-Low attachment 6 well plate for suspension cultures
(catalog number 27145; StemCell Technologies, Cambridge,
MA, USA).
3. Dermal fibroblasts (see Note 1).
4. Fetal Bovine Serum (FBS) .
5. Ham’s F12 Nutrient Mix (catalog number 11765-047; Ther-
moFisher Scientific).
6. Dulbecco’s Modified Eagle Medium (DMEM) with high glu-
cose, GlutaMAX™ supplement, and HEPES (catalog number
10564-011; ThermoFisher Scientific).
Human iPS Cells to Model 267
3 Methods
Table 1
Primer sequences for characterization of putative hiPSCs and Progeny
(continued)
272 Jiaozi He et al.
Table 1
(continued)
16. To passage cells, aspirate the medium and wash one time with
either D-PBS or DMEM/F12 medium. Aspirate wash solution
and discard.
17. Add 1 mL of TrypLE and incubate at 37 C for 3–7 min.
Observe until cells are visibly coming off of the plate as large
aggregates when shaken back and forth.
18. Add 1 mL of FG medium, and triturate the mixture to dislodge
any remaining cells. No scraping is necessary.
19. Transfer the cells to a 15 mL conical tube containing 7 mL of
FG medium.
20. Rinse the well with 1 mL of FG medium and transfer this
medium with any remaining cells to the conical tube.
21. Centrifuge cells at 200 g for 5 min.
22. Aspirate and discard the supernatant.
23. Resuspend the cells in FG medium and passage 1:3 onto
untreated six well plates. No RI is needed.
24. Once the cells have been passaged and expanded at least one
time after thawing, the cells can be passaged with TrypLE as in
steps 16–20. Before centrifuging, take an aliquot and count
cells with a hemocytometer.
3.2 Reprogramming 1. On Day 0 (D0), plate 150,000 fibroblasts onto three wells each
of Fibroblasts of an untreated 6-well plate containing 2 mL FG medium (see
to Putative iPSCs Note 6).
(See Fig. 1) 2. On Day 1, remove one set of CytoTune™ 2.0 Sendai virus
tubes from the 80 C freezer.
3. Thaw each tube by first immersing the bottom of the tube in a
37 C water bath for 5–10 s, and then remove the tube from
the water bath and allow it to thaw at room temperature.
4. Once thawed, quick spin the tube and place it on ice. Check
certificate of analysis for # of particles/tube and follow the
companies instructions for infection, i.e., approximately
3 106 CIU/virus.
5. Aspirate medium from fibroblasts and wash one time with
2 mL prewarmed DGGS to remove serum.
6. Take pictures of fibroblasts on D0.
7. Aspirate DGGS medium and add 1 mL prewarmed DGGS to
each well to be reprogrammed, including to a negative control
well where no SV will be added to the fibroblasts.
8. Add CytoTune SV according to the manufacturer’s instruc-
tions directly into fibroblast media (see Note 7).
9. After addition of the virus, move the plate back and forth to
ensure that the virus is well distributed throughout the 1 mL of
274 Jiaozi He et al.
Fig. 1 Generation of putative hiPSC lines from fibroblasts using Sendai virus (Sv). In this figure, fibroblasts are
plated and then infected with Sv or left uninfected at the time points indicated. Within 2–3 days, the
morphology of Sv infected cells is dramatically different than that of uninfected cells. By day 6, considerable
cell death can be observed, and the density sometimes decreases by days 8–9. Around days 12–14, early
signs of colonies can be observed, which are clearly discernible usually around days 17–18. We normally
allow the colonies to grow until days 20–22 before picking the cells and transferring the colonies into 96- or
48-well plates. Once these have expanded, the colonies can be passaged with Accutase or EDTA and
expanded. A typical colony at the second passage (P2) in shown
medium. Rinse all tips and pipettes that come into contact with
virus with a 10% bleach solution and discard.
10. Incubate fibroblasts at 37 C for 24 h.
11. Aspirate medium and feed cells with 2 mL of fresh DGGS/
well. CytoTune contaminated medium should be treated with
10% bleach before disposal, and all pipettes and tips should be
similarly treated or disposed after autoclaving or incineration.
12. Take pictures of fibroblasts on Day 2.
13. On Day 3, add 2 mL of fresh DGGS/well and incubate at
37 C.
14. On Day 4, observe the cells and note any morphological dif-
ferences, particularly relative to the uninfected fibroblasts.
15. On Day 5, add 2 mL of fresh DGGS medium to each well and
incubate at 37 C. From this day forth, medium may need to be
changed every day (see Note 8).
Human iPS Cells to Model 275
3.3 Selection 1. After 5–8 passages, individual clones (5–10) are picked and
of Reprogrammed expanded clonally from each original isolate, as in Subheading
Cells Lacking Sendai 3.2, steps 34–39, except that medium should be changed
Virus and Mycoplasm daily. The isolation of selected colonies should be based on
morphology, with good colonies showing relatively smooth
edges and a high nucleus to cytoplasmic ratio.
2. Once each colony has been isolated and expanded sufficiently
to passage 1:2 or 1:3, make a duplicate set of plates for each
colony on separate 24-well plates. The first plate will be for
continued cultivation, expansion, freezing and immunostain-
ing if required. The second plate will be RNA isolation or
immunostaining and subsequent analysis for the presence of
Sendai virus.
3. Continue to passage the cultivation plate as needed to maintain
clonal lines.
4. For RNA, wash each nearly confluent well of the duplicate plate
with D-PBS, and lyse according to the manufacturer’s instruc-
tions (see Note 15).
5. Prepare RNA as a solution in ultrapure water (at neutral pH),
quantify using a NanoDrop spectrophotometer, and adjust the
278 Jiaozi He et al.
3.4 Freezing 1. Label 2 or more cryotubes for each clonal line to be preserved
and Thawing with date, cell line name and passage number.
of Putative iPSC
Clones
Human iPS Cells to Model 279
Fig. 2 Quality control of selected putative iPSCs: (a) Immunostaining of an early (left) and late (right) passage
putative hiPSC clone stained for Sv. In the early passage colonies, many individual colonies can be observed
that contain Sv. (b) Using a standard mycoplasma PCR reaction with controls, we verified that our clones from
both control (057) and vEDS (0197) hiPSC clones do not contain mycoplasma
10. Once the cells have been stored in liquid nitrogen for at least
24 h, we generally thaw one tube to ensure cell recovery.
11. To thaw cells, place cryovial up to the top level of cells in a
37 C water bath without submerging the cap. Mix gently in
the water bath until nearly thawed.
12. Once the last vestige of ice has thawed, dry the exterior of the
tube and spray the outside of the vial with 70% ethanol and
open in the laminar flow hood.
13. Once opened, gently transfer the 1 mL of thawed putative
iPSCs into a sterile 15-mL conical tube using a 5-mL sterile
pipette.
14. Slowly add 8 mL of E8 medium with RI drop-wise to cells in
the 15-mL conical tube. While adding the medium, gently
move the tube back and forth to mix the cells. This technique
reduces osmotic shock to the cells.
15. Rinse the vial with 1 mL of E8 medium with RI and add to the
15-mL tube with cells.
16. Centrifuge the cells at 200 g for 5 min. Aspirate and discard
the supernatant.
17. Resuspend the cell pellet in 2 mL of E8 medium plus RI by very
gently pipetting the cells up and down in the tube a few times.
Transfer to a 6 well plate (1–2 106 cells/well) coated with
Geltrex as described earlier.
18. Place in the 37 C incubator overnight and the following
morning, check to ensure cells have survived. If the cells sur-
vived, then these either can be discarded or the cells expanded
for subsequent analysis. If expansion is required, then replace
the cultivation medium with E8 lacking RI, and passage as
needed (see Note 17).
Characterization of Putative human iPS Cell Lines
3.5 PCR Analysis 1. Using cDNA prepared in Subheading 3.3, steps 6–8, prepare
of Pluripotency the master mix to evaluate the RNA levels of pluripotency
Associated transcription factors (OCT4, SOX2, NANOG, and ZFP42)
Transcription Factors and of lineage restricted cell markers (Ectoderm: PAX6,
SOX1, OTX2, GBX2; Mesoderm: Brachyury (T), Goosecoid;
Endoderm: FOXA2, AFP, SOX17).
2. Perform qPCR as described under Subheading 3.3, steps
10–16, except using the primers specific for these target
genes as described in Table 1.
3. As a control, use RNA prepared from an established hiPSC line
or from a line of human embryonic stem cells.
Human iPS Cells to Model 281
Fig. 3 Characterization of putative hiPSC clones. (a) Immunostaining for pluripotency markers, showing
nuclear staining of NANOG, and also staining of TRA-1-60 and SSEA4, which should be surface proteins
present on good quality hiPSCs. (b) Example of the neuroepithelial differentiation protocol used to test
differentiation potential of putative hiPSC clones. In this example, the neuroepithelial cells were positive for
PAX6 and Tuj1. Similar analyses were also performed to test for mesodermal and endodermal lineages. (c)
H&E staining of sections generated from a teratoma generated in NOD-SCID mice, showing the presence of
various lineages. Upon full analysis by a pathologist, we were able to show that all three germ layer lineages
were equally represented in these tumors
284 Jiaozi He et al.
27. RNA should also be analysed for the presence of lineage mar-
kers and the absence of pluripotency markers, as described
under Subheading 3.5.
Directed Differentiation to Ectodermal Cells
28. For differentiation of putative human iPSCs to ectodermal
lineages, we perform a modified neural induction step origi-
nally provided to us by Dr. Tilo Kunuth (Edinburgh) to pro-
duce PAX6 positive cells, although any differentiation protocol
that produce ectodermal lineages are suitable (see Note 26).
29. For ectodermal cell differentation, take cells from step 6 and
passage using accutase. Add 1 mL of accutase to the 6-well
plate and incubate for 5–7 min.
30. Add 1 mL of medium to dilute the accutase, followed by
trituration using a 5 mL pipette. Transfer the cells to a 15 mL
conical tube, remove an aliquot for cell counting and then
centrifuge the cells at 300 g for 3 min. Aspirate the medium.
31. Resuspend the cells in a volume of E8 medium containing
~4 105 cells/mL of medium.
32. Add 1 mL of cell suspension to a newly prewarmed plate
containing 1 mL of E8 medium with 2 RI.
33. Change the medium every day for 3 days or until the con-
fluency is ~90–95%.
34. At this time (Day 0), change the medium to 3N medium (a 1:1
mixture of N2 medium and B27-Neurobasal Medium) supple-
mented with 100 nM LDN 193189 and 10 μM SB431542.
Feed daily with 2 mL N3 medium until Day 5.
35. On Day 6, feed cells with N3 medium supplement only with
100 nM LDN 193189 until Day 11. Change medium daily. If
cells begin to peel off from plates, then the cultures may need
additional glutamine. This can be accomplished by doubling
the amount of GlutaMAX in the medium.
36. Cells can be passaged at this time onto glass coverslips and
allowed to plate for 2–3 days or used to make RNA suitable
for qPCR (see Table 1).
37. Immunocytochemistry can be performed using antibodies to
PAX6 and Tuj1 as described in Subheading 3.6, using the
antibodies and dilutions according to the manufacturer.
38. Following staining and microscopy, one expects that >90% of
the cells should be PAX6+ and many of the cells should be
Tuj1+ (Fig. 3b). Alternatively, flow cytometric analyses of
PAX6 or nestin can be performed.
39. RNA should also be analysed for the presence of lineage mar-
kers and the absence of pluripotency markers, as described
under Subheading 3.5.
286 Jiaozi He et al.
3.9 PCR Analysis 1. For PCR analyses of cell surface proteins or extracellular matrix
of Cell Surface components, we use quantitative PCR techniques, as described
Receptors and ECM in Subheading 3.3, steps 12–17.
Components 2. Many investigators may wish to design primers de novo, and
there are numerous sites and software available for this proce-
dure. To minimize testing of primers, we prefer to use pre-
tested/predesigned primers, which have been deposited to
PrimerDepot for the research community.
3. For selection of predesigned primers, go to PrimerDepot
http://primerdepot.nci.nih.gov. Input the RefSeq ID (such as
NM_181501, for integrin alpha1) or HUGO gene name (such
as ITGA1). This website is for human genes ONLY. For quan-
titative PCR, choose a pair of primers with amplicon sizes
ranging from ~80 to 120 bp and estimated genomic amplicon
size over 1000 bp.
4. Search the location of right and left primers in Ensembl
Genome Browser http://www.ensembl.org/index.html (see
Note 36). The reverse primer must be reverse complement
and visualized in software such as SnapGene Viewer.
(a) Choose primers that span two exons, which make geno-
mic amplification impossible for the desired length.
(b) If (a) is not possible, then choose right and left primers in
different exons.
(c) Alternatively, both (a) and (b) can be ignored, if genomic
DNA is eliminated during the RNA extraction procedure.
292 Jiaozi He et al.
Fig. 4 Analysis of paraxial mesoderm derived smooth muscle cells generated from verified hiPSCs from
control and vEDS patients. (a) Immunostaining of three smooth muscle markers, as indicated in the figure. (b)
Normalized qPCR analysis of transcripts analyzed from a control (2003-071-057, clone 1) and vEDS (0197)
human iPSC lines. In these analyses, we noted a difference in MYH11 transcript abundance between the
control and diseased smooth muscle cells cultivated in the presence of high FBS. (c) Example of analyses
performed on collagen isoforms (COL1A1 and COL3A1) and collagen receptors (not shown) after differentiation
to paraxial mesoderm derived smooth muscle cells. Unlike fibroblasts, COL3A1 was reduced relative to control
lines, suggesting some abnormal regulation of this transcript that might contribute to the phenotype observed
in patients with some vEDS point mutations
Human iPS Cells to Model 293
Table 2
Primer sequences designed to analyze collagen and collagen receptors
Gene NCBI Forward PRIMER Reverse PRIMER Start End Product length
ITGA1 NM_181501 CTCACTGTTGTTCTACGCTGC CGGAGAACCAATAAGCACCCA 107 253 147
ITGB1 NM_002211 TTTGAGCAAACACACAGCAA GAGTCGCGGAACAGCAG 66 189 124
ITGA11 NM_012211 ACTCAACCTGGGAAGGGTCA GCTCCCACACTCATGAGACC 342 480 139
DDR2 NM_006182 AGATAGGCAGCAGCAGGAAC AGAGGCCAGCTGTTTTTGAG 93 228 136
COL1A1 NM_000088 CCCCGAGGCTCTGAAGGTC GGAGCACCATTGGCACCTTT 1204 1334 131
COL3A1 NM_000090 GCAGGGTCTCCTGGTTCAAA CGGGACCCATTTCGCCTTTA 1183 1318 136
Human iPS Cells to Model 295
4 Notes
Table 3
Actual example of CytoTune Sv concentrations used for reprogramming
Recommended
Titer (CIU/mL) transduction CTL-0182 Well CTL-0182 Well CTL-0182 Well
transduction volume in μL A 1.5 105 B 1.5 105 C 1.5 105
Component volume for 106 cells cells plated cells plated cells plated
CytoTune™ 1.1 108 45 25 18 0
2.0 human
KOS
CytoTune™ 1.1 108 45 25 18 0
2.0 human
c-Myc
CytoTune™ 1.1 108 27 15 10.8 0
2.0
Human Klf4
Example of the volumes of CytoTune™2.0 added to each well of fibroblasts for reprogramming. In this example,
fibroblast line CTL-0182 was plated at a density of 150,000 cells. The CytoTune SV is added at two concentrations for
reprogramming and we run one well as a negative control. When observing whether the infections have been successful,
visual comparisons with the uninfected cells are useful. KOS—Klf4, Oct4, Sox2. Volumes are in microliter
13. Passage with EDTA is a fast and easy way to passage putative
human iPS cells. Cells must, however, be 70–85% confluent, as
densities above 90% are difficult to passage. Through the che-
lator activity of EDTA, the removal of calcium and magnesium
from cells causes adhesions between cells to be lost, allowing
for rapid dissociation, even at room temperature. Dissociations
are not always complete, but for routine passaging, the plating
of small aggregates can be beneficial. The addition of RI is not
required for passaging of cells by EDTA; however, there are
reports that chromosomal aberrations may occur with long-
term cultivation and that these aberrations can be minimized
by the addition of RI. Once lines are well established and stocks
frozen in liquid nitrogen, the addition of RI to iPSC lines may
no longer be required when passaging with EDTA. To passage
cells using EDTA, aspirate medium, followed by addition of
EDTA (2 mL of a 6 well plate) to rinse wells two times at room
temperature. Add EDTA a third time to the well (1 mL) and
incubate the plates at room temperature for 3–5 min for small
aggregates or 7–9 min for single cells. Once cells begin to show
distinct separations within the colonies, remove the EDTA by
aspiration. Add E8 medium and immediately triturate to break
any clumps of cells into small aggregates. The addition of
calcium back to the cells caused rapid reaggregation, so it is
imperative to dissociate the cells quickly. Once cells are disso-
ciated, ideally into small aggregates, transfer to a Geltrex-
coated well and plate with E8 þ RI.
14. Depending on the initial isolation, colony expansion can be
readily achieved within 4–5 passages, but for novices it may
take a few additional passages for successful expansion. This is
not a problem, as the colonies still contain residual SV. With
the CytoTune 2 SV, it usually takes 8–14 passages to eliminate
the SV, and in some cases colony isolation may be required. It is
also important to realize that not every picked putative colony
will be suitable for expansion. In cases where numerous clones
are being expanded, pick those colonies that more resemble
embryonic stem cells, characterized by smooth edges, very
tightly packed cells, and cells with a high nuclear to cytoplasmic
ratio.
15. Isolation of RNA is beyond the scope of this chapter, but many
kits are available that permit high quality RNA isolates (e.g.,
RNeasy Mini Kits from Qiagen).
16. Although a single frozen stock may be acceptable, we recom-
mend making a minimum of three tubes of frozen cells. The
first is to ensure proper freezing, the second is maintained for
storage and thawing, while the third serves as a backup in case
there is contamination of the cells upon thawing. Ideally
1 to 3 million cells/cryotube should be stored; however, we
298 Jiaozi He et al.
Acknowledgment
This work was supported by a grant from the Hong Kong Research
Grant Committee General Research Fund (Project number
17100214) and by generous support from the Huey Foundation.
Human iPS Cells to Model 301
References
1. Takahashi K, Yamanaka S (2006) Induction of terminally differentiated circulating T cells.
pluripotent stem cells from mouse embryonic Cell Stem Cell 7:11–14
and adult fibroblast cultures by defined factors. 12. Anokye-Danso F, Anokye-Danso F, Trivedi
Cell 126:663–676 CM, Trivedi CM, Juhr D, Juhr D et al (2011)
2. Takahashi K, Tanabe K, Ohnuki M, Narita M, Highly efficient miRNA-mediated reprogram-
Ichisaka T, Tomoda K et al (2007) Induction ming of mouse and human somatic cells to
of pluripotent stem cells from adult human pluripotency. Cell Stem Cell 8:376–388
fibroblasts by defined factors. Cell 13. Warren L, Manos PD, Ahfeldt T, Loh Y-H,
131:861–872 Li H, Lau F et al (2010) Highly efficient repro-
3. Cahan P, Daley GQ (2013) Origins and impli- gramming to pluripotency and directed differ-
cations of pluripotent stem cell variability and entiation of human cells with synthetic
heterogeneity. Nat Rev Mol Cell Biol modified mRNA. Cell Stem Cell 7:618–630
14:357–368 14. Gazit Y, Jacob G, Grahame R (2016) Ehlers-
4. Sugiura M, Kasama Y, Araki R, Hoki Y, Danlos syndrome-hypermobility type: a much
Sunayama M, Uda M et al (2014) Induced neglected multisystemic disorder. Rambam
pluripotent stem cell generation-associated Maimonides Med J 7:e0034
point mutations arise during the initial stages 15. Kim ST, Brinjikji W, Lanzino G, Kallmes DF
of the conversion of these cells. Stem Cell Rep (2016) Neurovascular manifestations of
2:52–63 connective-tissue diseases: a review. Interv
5. Mekhoubad S, Bock C, de Boer AS, Kiskinis E, Neuroradiol 22:624–637
Meissner A, Eggan K (2012) Erosion of dosage 16. Mak KM, Png CYM, Lee DJ (2016) Type V
compensation impacts human iPSC disease collagen in health, disease, and fibrosis. Anat
modeling. Cell Stem Cell 10:595–609 Rec (Hoboken) 299:613–629
6. Nakagawa M, Koyanagi M, Tanabe K, 17. Hershenfeld SA, Wasim S, McNiven V,
Takahashi K, Ichisaka T, Aoi T et al (2008) Parikh M, Majewski P, Faghfoury H et al
Generation of induced pluripotent stem cells (2016) Psychiatric disorders in Ehlers-Danlos
without Myc from mouse and human fibro- syndrome are frequent, diverse and strongly
blasts. Nat Biotechnol 26:101–106 associated with pain. Rheumatol Int
7. Soldner F, Hockemeyer D, Beard C, Gao Q, 36:341–348
Bell GW, Cook EG et al (2009) Parkinson’s 18. Beridze N, Frishman WH (2012) Vascular
disease patient-derived induced pluripotent Ehlers-Danlos syndrome: pathophysiology,
stem cells free of viral reprogramming factors. diagnosis, and prevention and treatment of its
Cell 136:964–977 complications. Cardiol Rev 20:4–7
8. Sommer CA, Sommer AG, Longmire TA, 19. Frank M, Albuisson J, Ranque B, Golmard L,
Christodoulou C, Thomas DD, Gostissa M Mazzella J-M, Bal-Theoleyre L et al (2015)
et al (2010) Excision of reprogramming trans- The type of variants at the COL3A1 gene
genes improves the differentiation potential of associates with the phenotype and severity of
iPS cells generated with a single excisable vec- vascular Ehlers–Danlos syndrome. Eur J Hum
tor. Stem Cells 28:64–74 Genet 23:1657–1664
9. Kaji K, Norrby K, Paca A, Mileikovsky M, 20. Daheron L, D’Souza S (2012) Blood – SeV
Mohseni P, Woltjen K (2009) Virus-free induc- derived fibroblast generated iPSCs. In: Stem-
tion of pluripotency and subsequent excision of Book [Internet]. Cambridge, MA, Harvard
reprogramming factors. Nature 458:771–775 Stem Cell Institute, pp 2008–2012
10. Woltjen K, Michael IP, Mohseni P, Desai R, 21. Cheung C, Bernardo AS, Trotter MWB, Ped-
Mileikovsky M, H€am€al€ainen R et al (2009) ersen RA, Sinha S (2012) Generation of human
piggyBac transposition reprograms fibroblasts vascular smooth muscle subtypes provides
to induced pluripotent stem cells. Nature insight into embryological origin-dependent
458:766–770 disease susceptibility. Nat Biotechnol
11. Seki T, Yuasa S, Oda M, Egashira T, Yae K, 30:165–173
Kusumoto D et al (2010) Generation of 22. Bhattacharya S, Burridge PW, Kropp EM,
induced pluripotent stem cells from human Chuppa SL, Kwok W-M, Wu JC et al (2014)
302 Jiaozi He et al.
High efficiency differentiation of human plu- instability in the human embryonic stem cell
ripotent stem cells to cardiomyocytes and char- epigenome. Hum Mol Genet 16:1253–1268
acterization by flow cytometry. J Vis Exp 26. Cheung C, Bernardo AS, Pedersen RA, Sinha S
23:52010 (2014) Directed differentiation of embryonic
23. Fazeli A, Liew C-G, Matin MM, Elliott S, Jean- origin-specific vascular smooth muscle sub-
meure LFC, Wright PC et al (2011) Altered types from human pluripotent stem cells. Nat
patterns of differentiation in karyotypically Protoc 9:929–938
abnormal human embryonic stem cells. Int J 27. Stadtfeld M, Apostolou E, Akutsu H,
Dev Biol 55:175–180 Fukuda A, Follett P, Natesan S et al (2010)
24. Draper JS, Smith K, Gokhale P, Moore HD, Aberrant silencing of imprinted genes on chro-
Maltby E, Johnson J et al (2004) Recurrent mosome 12qF1 in mouse induced pluripotent
gain of chromosomes 17q and 12 in cultured stem cells. Nature 465:175–181
human embryonic stem cells. Nat Biotechnol 28. Gutierrez-Aranda I, Ramos-Mejia V, Bueno C,
22:53–54 Munoz-Lopez M, Real PJ, Mácia A et al
25. Allegrucci C, Wu Y-Z, Thurston A, Denning (2010) Human induced pluripotent stem cells
CN, Priddle H, Mummery CL et al (2007) develop teratoma more efficiently and faster
Restriction landmark genome scanning identi- than human embryonic stem cells regardless
fies culture-induced DNA methylation the site of injection. Stem Cells 28:1568–1570
Chapter 18
Abstract
Cell interactions with the extracellular matrix (ECM) are critical to cell and tissue functions involving
adhesion, communication, and differentiation. Three-dimensional (3D) in vitro culture systems are an
important approach to mimic in vivo cell–matrix interactions for mechanobiology studies and tissue
engineering applications. This chapter describes the use of engineered microtissues as 3D constructs in
combination with a magnetic tissue gauge (μTUG) system to analyze tissue mechanical properties. The
μTUG system is composed of poly(dimethylsiloxane) (PDMS) microwells with vertical pillars in the wells.
Self-assembled microtissues containing cells and ECM gel can form between the pillars, and generate
mechanical forces that deform the pillars, which provides a readout of those forces. Herein, detailed
procedures for microfabrication of the PDMS μTUG system, seeding and growth of cells with ECM gels
in the microwells, and measurements of the mechanical properties of the resulting microtissues via magnetic
actuation of magnetic sphere-tagged μTUGs are described.
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_18, © Springer Science+Business Media, LLC 2018
303
304 Prasenjit Bose et al.
Fig. 1 Schematic of microtissue stretching using a magnetic tweezer. (a) Microtissue suspended between two
flexible PDMS micropillars that are deflected by the microtissue’s contractile force. (b) When a magnetic field
is generated by the magnetic tweezer, a magnetic force Fmag is applied to the magnetic sphere bonded on the
top of one of the pillars, and strains are exerted on the microtissue
2 Materials
2.1 Equipment 1. Contact mask aligner (Model 200; Optical Associates, San
and Instruments Jose, CA, USA) or equivalent.
2. Programmable spin-coating system like the WS-400B-NPP-
Lite system (Laurell Technologies Corporation, North Wales,
PA, USA).
3. Vacuum desiccator capable of achieving P ¼ 25 Torr pressure
(Bel-Art 230 mm or equivalent).
4. Vacuum pump with ~140 L/min (5.0 Cfm) capacity (model
6912; FJC, Mooresville, NC, USA) or similar.
5. Oven: Set to 65 C for PDMS curing.
6. Hot plates: One in cleanroom for photoresist processing and
one for PDMS processing.
7. Stereo-microscope.
8. Cell culture hood.
306 Prasenjit Bose et al.
2.2 Photolithography 1. Mylar masks: patterns printed at 40,000 dots per inch (DPI).
2. Blank glass mask plate: 400 400 to fit in mask aligner (see Note 2).
3. Silicon wafers: h100i Si wafers with 300 diameter and thickness
350–400 μm (see Note 2).
4. Precoated glass plate: 400 400 glass plate precoated with chro-
mium and 0.5 μm of positive photoresist, AZ 1518 (Nanofilm
Inc., Westlake Village, CA, USA).
Microtissues to Study Cell-Matrix Interactions 307
2.3 Replica Molding 1. Poly(dimethyl siloxane) (PDMS) and curing agent Sylgard
and μTUG Device 184 elastomer kit (catalog number 2065662; Dow Corning
Fabrication Corporation, Auburn, MI, USA).
2. Smooth-Cast® 305, Parts A and B (Smooth-On Inc., Macun-
gie, PA, USA).
3. Tridecafluoroctyltrichlorosilane (catalog number 78560-45-9;
UCT Specialties, Bristol, PA, USA).
4. Nickel spheres: Ni powder 74–116 μm (mesh size: 150 þ 200;
catalog number 7440-02-0; Alfa Aesar, Ward Hill, MA, USA).
5. Fluorescent beads: Fluoresbrite YG carboxylate microspheres
2 μm diameter (catalog number 09847-5; Polysciences Inc.,
Warrington, PA, USA) diluted 1:3000 in 100% Ethyl Alcohol
200 proof, Absolute, Undenatured, A.C.S./USP Grade).
6. Plastic culture dishes and plates: P35 (35 mm) culture dishes
(catalog number 353001 or similar), 12-well plates (catalog
number 353043 or similar), 24-well plates (catalog number
353226 or similar; BD Biosciences, Bedford, MA, USA).
7. 44 mm aluminum weighing dishes (catalog number 25433-
016; VWR International, Radnor, PA, USA).
8. Precision nonmagnetic tweezers with 50 μm tips, razor blades,
50 mL centrifuge tubes.
2.4 Microtissue 1. 0.2% Pluronic® F-127: Prepare from mixing Pluronic® F-127
Seeding and Culture powder (catalog number P2443; Sigma-Aldrich Chemical
Company) or 10% Pluronic® F-127 solution (catalog number
P6866; Thermo Fisher Inc.) with deionized water or
Phosphate-Buffered Saline (PBS, pH 7.4) and followed by
308 Prasenjit Bose et al.
Table 1
Example of creating 2 mL of extracellular matrix solution with 2.5 mg/mL
of collagen and 2 mg/mL of fibrinogen
3 Methods
Fig. 2 Schematics of the fabrication of SU-8 master mold. (a) Deposit photoresist base layer, first layer and
blocking layer on Si wafer. (b) Expose under first layer mask. (c) Deposit second layer of SU-8. (d) Expose
under second layer mask after alignment. (e) Develop with SU-8 developer. (f) Image of an SU-8 master mold
array on a silicon wafer and a magnified image of one microwell
Fig. 3 Mask set to produce an array of μTUG devices in a P35 culture dish. (a)
Bottom layer mask used to define the pillars’ stems. (b) Top layer mask that
defines the pillars’ heads. A magnified view of one microwell on each mask is
shown. The complimentarily shaded, circumscribed star patterns in the bottom
corners are used for mask and substrate alignment as described in Note 16. The
micro-wells are 800 μm 400 μm in size, and the horizontal pillar-to-pillar
spacing is 500 μm. In the well shown magnified, the pillar stems’ cross-sectional
dimensions are 150 μm 50 μm, and those of the heads’ are 200 μm 90 μm.
This yields pillars with effective spring constants k ¼ 2 μN/μm. The other three
groups of μTUGs (listed top to bottom) had pillar stem dimensions of
150 μm 26 μm, 150 33 μm, and 150 μm 42 μm, pillar head
dimensions of 200 μm 65 μm, 200 μm 75 μm, and 200 μm 80 μm,
and corresponding spring constants k ¼ 0.25 μN/μm, 0.5 μN/μm, and 1 μN/μm,
respectively. All quoted k values assume a 10:1 PDMS mixing ratio (Young’s
modulus ¼ 1.6 MPa)
17. Soft bake at 95 C for 3.5 h, and then allow the wafer to cool to
room temperature.
18. Preparation of blocking layer: Deposit a 20 μm thick layer
from a v/v mixture of 30% S1813 in SU-8 2010 on the wafer
via the following spin coater settings: 10 s at 500 rpm þ 30 s at
1000 rpm.
19. Soft bake at 95 C for 30 min. Remove and allow to cool to
room temperature.
20. Once the sample cools to room temperature, expose it under
the mask aligner using the first layer chrome mask. Keep the
total dose at 700 mJ/cm2 and insert a low pass filter in the path
of the light to block out UV rays of wavelength below 350 nm
(see Note 14). The high dose penetrates the blocking layer and
exposes the first layer.
21. Do a post-exposure bake at 95 C for 12 min (see Note 15).
22. Preparation of final layer: Deposit the final layer of 50 μm of
SU-8 2050 using the following spin coater settings: 60 s at
500 rpm þ 60 s at 1500 rpm.
23. Soft bake at 95 C for 3 h.
24. Once the sample cools to room temperature, expose the rest of
the wafer using the second layer mask. Use the mask aligner to
align patterns on the mask to the patterns on the wafer (see
Note 16).
25. The UV light dose at this step should be between
100–110 mJ/cm2 in the presence of the low pass filter (see
Note 17).
26. Follow this exposure with a post exposure bake at 95 C for
12 min. After a few minutes of cooling down, the wafer is ready
for development.
27. Develop the wafer in a glass petri dish filled with PGMEA for
45 min on a stirrer or shaker (see Note 18).
28. Once the development is done, wash the wafer in IPA for 90 s,
and then dry using nitrogen gas (see Note 19).
29. The master fabrication is now complete.
3.2 Replica Molding 1. Generation of negative molds. The ‘negative’ molds required
and PDMS Device to make the final devices are made from PDMS. The process is
Fabrication sketched in Fig. 4a–c.
2. Combine PDMS and curing agent in a 20:1 ratio (see Note
20), and mix thoroughly for 20 min to obtain a uniform
solution.
3. Centrifuge the solution in a 50 mL tube at 720 g (see Note
21) for 1 min to remove any bubbles.
Microtissues to Study Cell-Matrix Interactions 313
Fig. 4 Schematics of replica molding and PDMS device fabrication. (a) Silanization of SU-8 master molds. (b)
Cast PDMS and cure at 65 C. (c) Peel off PDMS negative mold. (d) Silanization of PDMS negative mold. (e)
Cast PDMS and cure at 65 C. (f) Peel off the PDMS devices
4. Place the μTUG master flat in a large plastic weighing dish, and
add the PDMS þ curing agent solution on top to a depth of
3–5 mm.
5. Insert the entire dish into a vacuum desiccator, and hold at
P ¼ 25 Torr for 1 h to remove all trapped air.
6. Bake in a 65 C convection oven for 24 h to cure the PDMS.
7. Separate the PDMS mold from the master (see Note 22) and
trim away excess PDMS around the features to form squares
~1.75 1.75 cm2.
8. Generation of Smooth-Cast® master copies. To make these
durable masters, attach negative molds (pattern side facing
up) in 44 mm aluminum weighing dishes using double-sided
tape (see Note 23).
9. Prepare a 1:1 volume mixture of Smooth-Cast 305 A and
305 B, mix for 30 s, add on top of the molds to cover them,
and degas for 5 min using a vacuum desiccator at P ¼ 25 Torr
(see Note 24).
10. The Smooth-Cast mixture hardens within 3–4 h at room tem-
perature and the molds can be peeled off (use ethanol for
lubrication) (see Note 25).
11. μTUG device production from molds. The final μTUG devices
are made from PDMS, as outlined in Fig. 4d–f and described in
subsequent steps. The PDMS to curing agent ratio may be
varied to control the pillars’ stiffness.
314 Prasenjit Bose et al.
12. Place a mold made from the masters or the Smooth-Cast copies
in a glass petri dish (pattern side up), clean with ethanol on a
shaker for 5 min, and dry with nitrogen gas (see Note 26).
13. To remove any organic impurities and to make the surface
hydrophilic, clean the molds using an oxygen plasma prior to
each use. We recommend a power of 25 W and pressure of
450 mTorr for 60 s.
14. To “silanize” the molds, place the plasma-cleaned molds in a
vacuum desiccator, together with a few drops of tridecafluor-
octyltrichlorosilane on a separate coverslip.
15. Seal the desiccator under vacuum (P ¼ 25 Torr) and expose the
molds to the silane fumes for at least 12 h. This creates a thin
layer of silane over the PDMS mold to facilitate separation of
the device from the mold.
16. Prepare a mixture of PDMS and curing agent. Together with
the pillar geometry, the PDMS to curing agent ratio deter-
mines the pillar stiffness of the μTUGs [10]. The Young’s
modulus for 20:1, 10:1, and 4:1 mixtures is 0.54, 1.6, and
4 MPa, respectively.
17. To produce μTUG devices in a P35 culture dish, add 2 mL of
PDMS solution to the dish, followed by quick degassing
(10 min at P ¼ 25 Torr).
18. Heat the dish on a hot plate at 65 C for 20 min. This step is
done to harden this bottom layer of PDMS somewhat (see
Note 27).
19. While the bottom layer of PDMS in the petri dish is curing,
pour 1 mL of PDMS on top of the silane-treated mold and
degas (~15 min at P ¼ 25 Torr).
20. Invert the mold and gently push down through the liquid
PDMS and onto the slightly hardened PDMS in the petri dish.
21. Top off the sides of the mold with excess PDMS. It may be
necessary to degas the dish for 2–3 h to remove air bubbles.
22. After all the bubbles are removed, cure the PDMS in a 65 C
oven for 24 h.
23. Once the dishes are cured, the molds are peeled off slowly from
the device. This is done by making incisions along the edge of
the mold device interface from the top using a thin tweezer or a
razor blade, and then prying off the mold from one or multiple
sides using the flat edge of the tweezer or blade. Squirting
ethanol in the fissures is highly recommended as it facilitates
the separation and prevents the μTUG pillars from breaking.
24. To create μTUG devices in 12- or 24-well plates, cut the molds
into smaller cubes with surface area near 1 cm2, and employ the
Microtissues to Study Cell-Matrix Interactions 315
3.4 Cell Seeding 1. Detach and dissociate the cells by Trypsin, Accutase or TrypLE,
neutralize with fresh media and count the cells (see Note 32.
For fibroblasts, adjust the cell concentration to
~300,000 cells/mL and take 1 mL of cell solution (see Note
33). This volume varies for 12- and 24-well plates, depending
on the device size. Centrifuge at 180 g for 5 min, resuspend
the cells in fresh media and keep the cells on ice).
2. Place all the ECM buffer components and the soluble ECM
(1 M NaOH, 10 M199, 5% NaHCO3, 250 mM HEPES,
S-DI water, collagen and (if needed) fibrin or fibrinogen) on ice.
3. Place μTUG device(s) on a cold ice pack.
4. Determine the desired concentration of matrix components
(collagen, fibrin and/or fibrinogen.) Add calculated volumes
of each component to a 15 mL conical tube (see items 8 and
9 of Subheadings 2.4 and Table 1) and mix thoroughly by
repeatedly and gently pipetting (see Note 34).
5. Add 1 mL of ECM mixture to μTUGs in P35 dish or 250 μL
per well in 12-well or 24-well plates.
6. Place μTUGs along with ice pack in a vacuum desiccator to
degas (2–3 min at P ¼ 25 Torr) (see Note 35), or pipet up and
down swiftly to destroy any air bubbles that may have formed
in the collagen solution.
7. Spin down the cell solution from step 1 at 180 g for 3 min,
and replace the media solution with 500 μL of the ECM
mixture for P35 dishes to reach the desired cell number. This
volume varies for 12- and 24-well plates, depending on the
device size (see Note 36).
8. Transfer the above cell-ECM solution to the μTUGs and mix
by pipetting (see Note 37).
9. Spin the μTUGs at 237 g for 90 s at 9 C (see Note 38) to
pull the cells down into the wells.
10. For P35 dishes, rotate the dishes by 90 and spin at 237 g for
90 s at 9 C to even out effects on the distribution of the
solution due to the tangential acceleration. This additional
spinning step is optional for 12- and 24-well plates.
11. Transfer the μTUGs on ice pack to the cell culture hood.
12. Hold the ice pack slightly tilted towards you.
13. Aspirate the collagen mixture. Start with the aspirator at one of
the top corners of the device, and draw the aspirator horizon-
tally to the other top corner to detach the liquid from the
upper sidewall of the dish. Then gradually slide the aspirator
Microtissues to Study Cell-Matrix Interactions 317
down along the side, staying away from the microwells (see
Note 39).
14. Invert the dish or plates so that they are now upside down, and
spin at 40–60 g for 15 s at 9 C (see Note 40).
15. Keep the μTUGs inverted on an ice block when transferring
from centrifuge to hood.
16. Add 500 μL of H2O or PBS in the lid of the P35 dish or 3 mL
to 12-well or 24-well plates.
17. Incubate the μTUGs with the lid down (inverted) for
10–15 min at 37 C until the collagen is polymerized (see
Note 41).
18. Aspirate water and gently add medium at the corner (1 mL for
P35 dish and 250 μL per well for 12-well or 24-well plates).
19. Place the dishes or plates in the incubator for culturing to allow
the microtissues to form.
3.6 Contractility The contractile forces exerted by the microtissues are obtained by
Measurements tracking the deflection of the pillars to which the tissues are
adhered. This is achieved by measuring the positions of the fluores-
cent microbeads on the top of the pillars or by tracking the edges of
the pillars through imaging.
1. To record the initial compaction and formation of the micro-
tissues, which typically takes 12–24 h, mount the P35 dish or
multi-well plate containing the microtissues in a live-cell cham-
ber mounted on top of the microscope stage at 37 C with the
CO2 concentration set between 5–10% depending on the cell
type used (see Note 44).
2. To maximize the ability to resolve the pillars’ motion, choose a
microscope objective so that one microtissue fills the field of
view (typically 4 or 10). However, make sure that static
318 Prasenjit Bose et al.
Fig. 5 Magnetic tweezer system. (a) shows a schematic and (b) shows a photograph of the unit mounted on a
microscope. The long, tapered rod is an iron core that acts as a magnetic pole tip projecting into a P35 μTUG
sample dish. The tweezer solenoid surrounds the core between the arms of the aluminum bracket on a 3-axis
micromanipulator and is encased in an aluminum heat-sinking block. A second manipulator and coil assembly
(unused in this application) is shown at left in (b) without its core. The image in (b) is reproduced from A. S. Liu,
Ph.D. Thesis, the Johns Hopkins University (2015). Used by permission
3.8 Data Reduction For both contractility measurements (Subheading 3.6) and stress–-
and Analysis strain measurements (Subheading 3.7), the tissue force is measured
from the deflection of the nonmagnetic pillar(s), using the relation
F ¼ kδ, where k is the pillar’s bending constant, and δ is the
measured pillar deflection. The value of k is calculated for small
deflections from Euler-Bernoulli beam theory. For pillars with
heads, the bending constant is given by, k ¼ a2 ð6EI 3La Þ
, where E is
the Young’s modulus of PDMS, I is the area moment of inertia of
the pillar stem’s cross section, a is the height measured to the center
of the pillar head, and L is the total height of the pillar (see Note 55)
[10]. Strain is measured either from the length change of the
microtissue, or from measurements of the local strain in the central
region of the microtissues via a texture correlation algorithm (see
Note 56) [18].
1. Pillar tracking: fluorescent labeling. When relatively small
numbers of images (<~150) are acquired, and the pillars are
fluorescently labeled, import images into ImageJ and convert
images into a stack.
2. Use the Spot Tracker plugin [19] to analyze the changes of
particle locations at different time point.
3. For larger fluorescently labeled data sets, use automated parti-
cle tracking approaches [20].
4. Pillar tracking: white light. We use custom image analysis
written in the software package Igor Pro (Wavemetrics) to
track the motion of identifiable features, such as the edge of a
moving pillar (see Note 57).
Microtissues to Study Cell-Matrix Interactions 321
4 Notes
25. The resulting hardened plastic shapes have the same pattern as
the original masters and can be utilized to make new molds
following the procedure given in step 1 above.
26. Typically, between four to eight molds are treated simulta-
neously to make multiple μTUG devices in P35 dishes.
27. If the PDMS at the bottom of the dish is not cured well, then
the mold will sink all the way to the bottom of the dish when
inverted onto the dish. This makes peeling off the mold from
the completed devices difficult. On the other hand, if the
PDMS is over-cured, then inverting the mold onto the surface
can often lead to trapping of air bubbles.
28. Some optimization of this process is needed, as tracking is
easiest with a small number of isolated beads in each pillar. As
an alternate approach, fluorescent beads can also be glued to
the pillar tops by hand with PDMS after the μTUGs are
fabricated.
29. Dissolution of nickel into cell culture media was measured and
the levels does not show any negative effects on biocompatibil-
ity of the tissues [12].
30. The duration of Pluronic® treatment varies between 1–30 min
depending on the contractility of the cells and the concentra-
tion of ECM. Shorter times can be used when working with
more contractile cells, such as fibroblasts, which start compact-
ing the matrix within a few minutes after polymerization. In
contrast, longer duration Pluronic® treatments are needed for
low contractility cell types, such as smooth muscle cells, that
compact tissues over the course of up to 48 h. For example, it
takes 1–2 min of treatment for 3T3 fibroblasts in 1 mg/mL of
collagen, while 7–8 min treatment is needed for the same cells
in 2.5 mg/mL of collagen. Note that the microtissues may slip
off the pillar caps if the treatment is excessive.
31. Although passivation of the devices and seeding of the cells are
presented as separate methods, we suggest combining both
procedures in order to shorten the time. Typically, we start
with sterilizing the devices for 10–15 min. During this time, we
trypsinize and count the cells, and aliquot the number of cells
needed for the experiment. Then, we treat the devices with
Pluronic®, and during the incubation time we make the colla-
gen solution.
32. Cells may require different dissociation reagents. We use Accu-
tase for SMCs and cardiac fibroblasts, TrypLE for cardiomyo-
cytes, and Trypsin for fibroblasts.
33. In this step, one needs to define the total number of cells
needed for the experiments. For fibroblasts, 1 mL of
300,000 cells/mL is enough for one μTUG array in a P35
Microtissues to Study Cell-Matrix Interactions 325
Acknowledgements
References
1. Chen CS, Tan J, Tien J (2004) Mechanotrans- 2. Hynes RO (2002) Integrins: bidirectional,
duction at cell-matrix and cell-cell contacts. allosteric signaling machines. Cell
Annu Rev Biomed Eng 6:275–302 110:673–687
328 Prasenjit Bose et al.
3. Schwartz MA, Ginsberg MH (2002) Networks study collective 3D cell mechanics in long-
and crosstalk: integrin signalling spreads. Nat term cultures (invited). J Appl Phys
Cell Biol 4:E65–E68 115:172616
4. Ciobanasu C, Faivre B, Le Clainche C (2013) 14. Zhao R, Chen CS, Reich DH (2014) Force-
Integrating actin dynamics, mechanotransduc- driven evolution of mesoscale structure in engi-
tion and integrin activation: the multiple func- neered 3D microtissues and the modulation of
tions of actin binding proteins in focal tissue stiffening. Biomaterials 35:5056–5064
adhesions. Eur J Cell Biol 92:339–348 15. Liu AS, Wang H, Copeland CR, Chen CS,
5. Schiller HB, Fassler R (2013) Mechanosensi- Shenoy VB, Reich DH (2016) Matrix visco-
tivity and compositional dynamics of cell- plasticity and its shielding by active mechanics
matrix adhesions. EMBO Rep 14:509–519 in a microtissue model: in situ experiments and
6. Kuo JC (2013) Mechanotransduction at focal mathematical modeling. Sci Rep 6:33919
adhesions: integrating cytoskeletal mechanics 16. Sakar MS, Eyckmans J, Pieters R, Eberli D,
in migrating cells. J Cell Mol Med 17:704–712 Nelson BJ, Chen CS (2016) Cellular forces
7. Wozniak MA, Chen CS (2009) Mechanotrans- and matrix assembly coordinate fibrous tissue
duction in development: a growing role for repair. Nat Commun 7:11036
contractility. Nat Rev Mol Cell Biol 10:34–43 17. Xu F, Zhao R, Liu AS, Metz T, Shi Y, Bose P,
8. Schmeichel KL, Bissell MJ (2003) Modeling Reich DH (2015) A microfabricated magnetic
tissue-specific signaling and organ function in actuation device for mechanical conditioning
three dimensions. J Cell Sci 116:2377–2388 of arrays of 3D microtissues. Lab Chip
9. Shamir ER, Ewald AJ (2014) Three- 15:2496–2503
dimensional organotypic culture: experimental 18. Zhao R, Simmons CA (2012) An improved
models of mammalian biology and disease. Nat texture correlation algorithm to measure sub-
Rev Mol Cell Biol 15:647–664 strate–cytoskeletal network strain transfer
10. Legant WR, Pathak A, Yang MT, Deshpande under large compressive strain. J Biomech
VS, McMeeking RM, Chen CS (2009) Micro- 45:76–82
fabricated tissue gauges to measure and manip- 19. Sage D, Neumann FR, Hediger F, Gasser SM,
ulate forces from 3D microtissues. Proc Natl Unser M (2005) Automatic tracking of indi-
Acad Sci U S A 106:10097–10102 vidual fluorescence particles: application to the
11. Legant WR, Chen CS, Vogel V (2012) Force- study of chromosome dynamics. IEEE Trans
induced fibronectin assembly and matrix remo- Image Process 14:1372–1383
deling in a 3D microtissue model of tissue 20. Crocker JC, Grier DG (1996) Methods of dig-
morphogenesis. Integr Biol 4:1164–1174 ital video microscopy for colloidal studies. J
12. Zhao R, Boudou T, Wang WG, Chen CS, Colloid Interface Sci 179:298–310
Reich DH (2013) Decoupling cell and matrix 21. Saltzman WM, Parkhurst MR, Parsons-
mechanics in engineered microtissues using Wingerter P, Zhu WH (1992) Three-
magnetically actuated microcantilevers. Adv dimensional cell cultures mimic tissues. Ann
Mater 25:1699–1705 N Y Acad Sci 665:259–273
13. Zhao R, Boudou T, Wang WG, Chen CS,
Reich DH (2014) Magnetic approaches to
Part IV
Abstract
Integration and analysis of high content omics data have been critical to the investigation of molecule
interactions (e.g., DNA–protein, protein–protein, chemical–protein) in biological systems. Human pro-
teomic strategies that provide enriched information on cell surface proteins can be utilized for repurposing
of drug targets and discovery of disease biomarkers. Although several published resources have proved
useful to the analysis of these interactions, our newly developed web-based platform Targets-search has the
capability of integrating multiple types of omics data to unravel their association with diverse molecule
interactions and disease. Here, we describe how to use Targets-search, for the integrated and systemic
exploitation of surface proteins to identify potential drug targets, which can further be used to analyze gene
regulation, protein networks, and possible biomarkers for diseases and cancers. To illustrate this process, we
have taken data from Ewing’s sarcoma to identify surface proteins differentially expressed in Ewing’s
sarcoma cells. These surface proteins were then analyzed to determine which ones were known drug
targets. The information suggested putative targets for drug repurposing and subsequent analyses illu-
strated their regulation by the transcription factor EWSR1.
Key words Surface protein, Web-based platform, Targets-search, Drug target, Molecular marker
1 Introduction
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2_19, © Springer Science+Business Media, LLC 2018
331
332 Bin Yan et al.
2 Materials
input Genes or
proteins
Search
Database
Surface proteins: Drug-target-disease: Disease and Cancer: Gene regulation: Protein networks
Drugbank OMIM TFBS: transcription
Human surfaceome PPI: protein
TTD Biomarkers factor target genes
Cell surface protein protein
ChemBL Driver genes containing binding
atlas (CSPA) interactions
PharmGKB sites
Interface
Interaction tables Interaction tables with Interaction tables Interaction Interaction
with cell surface drug/chemicals and with cancer tables with tables with
proteins in different targeted pathways detection, diagnosis transcriptional protein
cell types and diseases and prognosis gene regulation networks
Output
Integrations
Fig. 1 Workflow of the web-based Targets-search platform. The platform can integrate information from cell
surface proteins, drug-target, transcriptional regulation, protein networks, and human disease/cancer for data
analysis and interpretation
334 Bin Yan et al.
3 Methods
Fig. 3 An example study of Targets-search using surfaceome data of Ewing sarcoma (EWS) vs. Mesenchymal
stem cells (MSCs) from [13]. The results demonstrate that differentially expressed surface proteins not only
have been targeted by approved or testing drugs, acted as cancer biomarkers, but also are regulated by
EWS-related TFs
3.2 Example Study: 1. The first step is to prepare input data (see Fig. 3).
Human Cancer 2. Using published RNA-seq data [13], we identified two input
datasets: 185 genes upregulated in EWS and 316 genes upre-
gulated in MSCs (or downregulated in EWS) with >2.0-fold
changes in abundance and an adjusted p-value below 0.05.
3. Before submitting the input data, we analyzed biological pro-
cesses of the two input gene sets using DAVID. The results
show that 37 of the 185 upregulated and 80 of the 316 down-
regulated genes involve cell surface receptor linked signal trans-
duction events (see Note 14).
4. The second step is to identify how many authenticated surface
proteins are among the 185 upregulated genes in EWS. Copy
and paste these genes to the Input Window, check the box
listed “CSPA” on the Input Window, and search by pressing
“Search.”
5. The Output Window indicates that 59 proteins are detected as
human CSC-surface proteins.
6. Similar to the step above, there were 137 surface proteins
identified from 316 downregulated genes in EWS. These out-
put lists can be exported as .txt files.
Targets-Search for Surface Proteins 339
14. From the output above, we can select the target genes that only
are regulated by the three TFs. The result showed that EWSR1
can bind to 25 promoters of upregulated genes in EWS. Of
these, 12 are surface proteins, and NRP1, CA11, and DDR1
are targets of approved drugs. Furthermore, seven EWSR1
target genes are cotargeted by NF-κB and STAT3 both of
which have been implicated in EWSR1 regulation
[14, 15]. Of note, the surface protein leucine-rich repeat and
Ig domain protein 1 (LINGO1), expressed in over 90% of
EWS, was found to carry antibody conjugated with drugs to
kill Ewing sarcoma cells [13].
15. To link the input genes to protein network, the seventh step is
to check the database box “PPI.” After submission of the two
input lists, the outputs provide a list of all proteins that can be
connected to the input genes. As a result, the derived gene–-
protein interactions can establish protein networks linking the
upregulated genes in EWS and protein modifications.
16. From the outputs generated using the above procedures, Fig. 3
displays a multi-dimensional picture showing how the surface
proteins/genes differentially expressed in EWS link with drug
target, transcriptional gene regulation, protein networks, and
diseases/cancer. The main upregulated (FGFR4, PTPRS, and
ALK) and downregulated (EGFR, SERPINE1, etc.) genes/
proteins in EWS cells are targeted by approved drugs and were
reported as cancer biomarkers. EWSR1 was predicted to bind
to the upregulated NRP1, CA11, and DDR1 that are also
targeted by drugs. This result demonstrates the surface pro-
teins/genes identified by high throughput sequencing in EWS
could be potential targets for drug discovery and reveals their
regulatory components.
4 Notes
14. Using the bioinformatics tool DAVID, we can classify the input
genes to different function categories, gene ontology
(GO) and KEGG pathway. Among the 185 and 316 genes, of
80 and 128 among these genes were found to locate at plasma
membrane or cell surface respectively. By GO biological pro-
cess annotation, there were corresponding 37 and 80 genes
that involve cell surface receptor linked signal transduction.
Acknowledgment
References
1. Bausch-Fluck D, Hofmann A, Bock T, Frei AP, 8. Papadatos G, Overington JP (2014) The
Cerciello F, Jacobs A, Moest H, Omasits U ChEMBL database: a taster for medicinal che-
et al (2015) A mass spectrometric-derived cell mists. Future Med Chem 6:361–364
surface protein atlas. PLoS One 10:e0121314 9. Thorn CF, Klein TE, Altman RB (2010) Phar-
2. da Cunha JP, Galante PA, de Souza JE, de macogenomics and bioinformatics:
Souza RF, Carvalho PM, Ohara DT, Moura PharmGKB. Pharmacogenomics 11:501–505
RP, Oba-Shinja SM et al (2009) Bioinformatics 10. Cheng WC, Chung IF, Chen CY, Sun HJ, Fen
construction of the human cell surfaceome. JJ, Tang WC, Chang TY, Wong TT, Wang HW
Proc Natl Acad Sci U S A 106:16752–16757 (2014) DriverDB: an exome sequencing data-
3. Yan B, Guan D, Poon NYE, Li MJ, Wang P, base for cancer driver gene identification.
Tse WL, Wu SCM, Sham PC, Xia Z, Gundry Nucleic Acids Res 42:D1048–D1054
RL, Wang J, Boheler KR (in preparation). Tar- 11. Guan D, Shao J, Zhao Z, Wang P, Qin J,
getCSP, a comprehensive resource can priori- Deng Y, Boheler KR, Wang J, Yan B (2014)
tize targeting cell surface proteins for exploring PTHGRN: unraveling post-translational hier-
multi-interactions among molecules, drugs, archical gene regulatory networks using PPI,
and diseases ChIP-seq and gene expression data. Nucleic
4. Wishart DS, Knox C, Guo AC, Shrivastava S, Acids Res 42:W130–W136
Hassanali M, Stothard P, Chang Z, Woolsey J 12. Burchill SA (2008) Molecular abnormalities in
(2006) DrugBank: a comprehensive resource Ewing’s sarcoma. Expert Rev Anticancer Ther
for in silico drug discovery and exploration. 8:1675–1687
Nucleic Acids Res 34:D668–D672 13. Town J, Pais H, Harrison S, Stead LF,
5. LawV, KnoxC, Djoumbou Y, JewisonT, Guo Bataille C, Bunjobpol W, Zhang J, Rabbitts
AC, LiuY, Maciejewski A, Arndt D et al (2014) TH (2016) Exploring the surfaceome of
DrugBank 4.0: shedding new light on drug Ewing sarcoma identifies a new and unique
metabolism. Nucleic Acids Res 42: therapeutic target. Proc Natl Acad Sci U S A
D1091–D1097 113:3603–3608
6. Chen X, Ji ZL, Chen YZ (2002) TTD: Thera- 14. Javelaud D, Besancon F (2001) NF-kappa B
peutic Target Database. Nucleic Acids Res activation results in rapid inactivation of JNK
30:412–415 in TNF alpha-treated Ewing sarcoma cells: a
7. Zhu F, Shi Z, Qin C, Tao L, Liu X, Xu F, mechanism for the anti-apoptotic effect of
Zhang L, Song Y et al (2012) Therapeutic NF-kappa B. Oncogene 20:4365–4372
target database update 2012: a resource for 15. Lai R, Navid F, Rodriguez-Galindo C, Liu T,
facilitating target-oriented drug discovery. Fuller CE, Ganti R, Dien J, Dalton J, Billups C,
Nucleic Acids Res 40:D1128–D1136 Khoury JD (2006) STAT3 is activated in a
344 Bin Yan et al.
subset of the Ewing sarcoma family of tumours. 17. Li MJ, Sham PC, Wang J (2010) FastPval: a fast
J Pathol 208:624–632 and memory efficient program to calculate very
16. Levy S, Hannenhalli S (2002) Identification of low P-values from empirical distribution. Bio-
transcription factor binding sites in the human informatics 26:2897–2899
genome sequence. Mamm Genome
13:510–514
INDEX
Kenneth R. Boheler and Rebekah L. Gundry (eds.), The Surfaceome: Methods and Protocols, Methods in Molecular Biology,
vol. 1722, https://doi.org/10.1007/978-1-4939-7553-2, © Springer Science+Business Media, LLC 2018
345
THE SURFACEOME: METHODS AND PROTOCOLS
346 Index
Extracellular matrix (ECM)........................ 249, 251, 303 K
Extracellular matrix molecule-based capture...... 249–259
Extracellular secreted proteins.......................................... 3 Kegg identifiers ...........................................................4, 19
Extracellular vesicles (EVs).............................. 92–94, 100 Kinase................................. 131, 170, 173, 211–221, 238
F L
Fibroblast......................................... 62, 73, 92, 129, 212, Laminin..................... 142, 249, 251, 253, 255–257, 293
238, 239, 243, 262, 265, 270, 271, 273–277,
M
281, 292, 293, 304, 316, 317, 324, 325
Fibronectin .................................................................... 251 Magnetic actuation ....................................................... 315
Flow cytometry ......................... 22, 26, 61, 73, 118–120, Mass spectrometry (MS).......................... 4, 5, 11, 12, 22,
122, 124, 128, 129, 131–134, 136, 138–141, 32, 57–70, 73, 74, 76, 81, 86, 93, 95, 96, 98, 99,
143, 145, 147, 152, 153, 155, 156, 161, 249, 224, 241, 332, 341
252, 253, 255, 291, 299, 300 ESI-MS/MS ................................................................ 4
Fluid transport ......................vi, 224, 227, 235, 236, 245 HPLC-MS/MS ......................................................... 89
Fluorescence-activated cell sorting (FACS)................130, LC-MS/MS...................................68, 89, 93, 98, 241
131, 133, 138, 162, 228 nLC-MS/MS ................................................. 9, 11, 65
Förster resonance energy transfer (FRET) .................196, Mechanobiology ........................................................... 303
206, 208 Membrane enrichment .............................................62, 63
Membrane protein ...................... 4, 7, 10, 15, 22, 58–60,
G 73, 79, 80, 89, 180, 298
Gel-free systems ..........................................................4, 14 Membrane and matrix proteome ................................... 31
Glycopeptide capture ............................ 62, 64, 67–69, 76 Mesenchymal stromal cells(MSCs) .................... 255, 257,
Glycopeptide elution....................................62, 64, 67–69 BNF–259
Glycoprotein.................................. 62, 79, 113, 167, 237, Microfabrication............................................................ 303
241, 243, 341 Microtissues................................ 263, 303–314, 316–327
G protein-coupled receptor (GPCR).................. 151–163 Microvesicles (MVs)................................................91–100
Molecular marker ................................................. 331–343
H Monoclonal antibodies (mAbs)......................58, 60, 117,
144, 156
High-content automated electrophysiology ............... 185 Monolayer ....................................... 33, 37, 96, 115, 130,
131, 133–135, 178, 179, 223, 224, 230–238, 244
I
Immobilized cells ........................................ 249, 256, 257 N
Immunoaffinity ............................................................... 94 Nanoparticles........................................................vi, 79–89
Immunocytochemistry............................... 128, 224, 226, Neuron vi, 168, 170, 172, 173, 179, 186, 211–221, 284
262, 281, 282, 284, 285, 287, 291, 299 N-glycoproteins.....................................59, 60, 62, 70, 72
Immunohistochemistry (IHC).................................35, 39 N-linked antennas ........................................................... 40
Immunophenotyping................................. 57–70, 73, 74, Non-adherent cells.........................................97, 118, 201
76, 127–134, 136, 138–140, 142, 144–147
Immunoprecipitation (IP) ......................... 9, 11, 16, 208, O
215, 217, 219
Induced pluripotent stem cells (iPSCs) ................. 62, 73, Oligosaccharides.......................................... 59, 63, 65, 66
128, 178, 261–291, 293, 294, 296–300 O-linked antennas ........................................................... 40
Integrin ..................... 71, 73, 74, 94, 129, 238, 249, 290 Optical recording ................................................. 173, 178
Intercellular signaling ..................................................... 95
P
Internalization ...........152, 154–156, 158, 160–162, 214
Intracellular calcium concentration............ 198, 201, 202 Pellicles ......................................................................79–89
Ion channel.........................................167, 168, 171–173, Peptide separation ............................................................. 4
175–177, 180, 185, 224, 230 Peptide-N-glycosidase F (PNGaseF) ..................... 59, 69,
In vitro cytotoxicity assay ........................... 118, 122, 123 70, 111, 113, 114, 229, 242
In vivo biotinylation..................................................37–39 Phagocytosis ........................................224, 227, 236, 237
THE SURFACEOME: METHODS AND PROTOCOLS
Index 347
Phosphorylation ..................................170, 213–215, 218 Streptococcus pyogenes.................................................22, 24
Plasma membrane (PM) ............................ 43, 59, 65, 73, Stress–strain relations........................................... 263, 305
74, 79–89, 92, 152, 163, 172, 223, 231, 240, Subcellular localization ................................ 4, 12, 19, 58,
245, 333, 339, 343 169, 172, 174
Polarization ..................................................224, 230–233 Surface expression ...................................... 180, 212, 214,
Polymerase chain reaction (PCR) ........................ 14, 108, 215, 341
128, 172, 258, 262–264, 266, 278, 279, 290, Surface protein .................................3, 13, 21–28, 58, 59,
293, 341 61, 62, 74, 140, 185, 220, 224, 237, 238, 241,
Proteases .................................................... 14, 21–28, 241 262, 263, 282, 283, 298, 304, 331–334, 336,
Protein enzymatic cleavage............................................... 4 338–340, 342
Protein interaction ............................ 196, 241, 331, 332, Surfaceome .......................................3–19, 22, 57–70, 73,
334, 337, 340 74, 76, 79, 129, 261–291, 293, 294, 296–300,
Protein mapping.............................................................. 31 303, 332, 334, 341
Protein-protein interactions ...............331, 332, 334, 337 Surfome ........................................................22–25, 27, 28
Proteolytic digestion .................................................33, 35
Proteomics....................................... 5, 22, 23, 58, 73, 75, T
93, 94, 97, 98, 229, 241, 244, 332 Targets-search ..................................................62, 332–337
Purification ....................................... v, 6, 8, 9, 16, 33–35,
Transcriptome .................................................44, 47, 241,
39, 41, 59, 105–115 333, 336
Transfection...............................106, 107, 109, 110, 115,
R
158, 208, 219
Receptor internalization ..................................... 155, 158, Transient receptor potential (TRP) channel...... 195–206,
160–162, 214 208, 209
Receptor tyrosine kinase ...................................... 211–221 Transmembrane protein ...............................79, 105–115,
Reprogramming ......................................... 262, 263, 265, 195, 239, 241, 303, 333, 334, 336, 341, 342
270–273, 275–277, 293 Transmission electron microscopy ............. 224, 226, 231
Retinal pigment epithelium (RPE) ....................BNF–246 Trans-proteomic pipeline..........................................76, 99
RNA ................................47, 49, 92, 172, 177, 178, 249, Tryptic peptides....................................... 4, 10, 14, 17, 18
252, 257–259, 270, 274, 279, 284–287, 290, Tumor cell implantation ................................................. 37
291, 297, 333, 338, 339
RNA-seq data .............................................. 333, 338, 339 U
Ultracentrifugation ..................................... 8, 15, 94, 100
S
Sample solubilization ........................................................ 3 V
Secreted proteins .............................................. 79, 91–100 Vascular endothelial cell......................195–206, 208, 209
Secretome ............................................91, 92, 94–96, 100
Vascular endothelium............................................. 31, 196
Seizure ........................................................................... 171 Voltage dependent Na channels ..................168–171, 180
Sf9 cells ................................................................. 105–115
Shaving ......................................................................21–28 W
Sialidase......................................................................35, 40
Signal transduction .................................... 195, 211–221, Web-based platform ............................................. 332, 333
331, 338, 343 Western ................................. 16, 41, 128, 196, 197, 199,
Smooth muscle cells (SMCs).............................. 261–294, 200, 205–207, 215, 217, 219, 220, 224, 229,
296–300, 324, 325 238, 239, 243, 244
Sodium channels .................................................. 168–171 Wound healing .................................................32, 37, 325
Stem cells .............................................. 57–70, 73, 74, 76,
127–134, 136, 138–140, 142, 144–147, 180,
185, 191, 251, 255, 279, 297, 338, 339