Imunosenecence
Imunosenecence
Immunose-
necence
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
Albert C. Shaw
Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine,
New Haven, CT, USA
Editor
Albert C. Shaw
Section of Infectious Diseases
Department of Internal Medicine
Yale School of Medicine
New Haven, CT, USA
The United Nations estimates that, by 2050, the number of adults over age 60 will rise to
over two billion worldwide and will exceed the number of individuals under age 15 for the
first time in human history. This aging of the worldwide population has profound social
implications and will undoubtedly influence the distribution and delivery of healthcare;
notably, older adults are at increased risk for organ-specific dysfunction such as cardiovas-
cular and renal disease, as well as increased rates of neurodegeneration and epithelial
malignancies, to name a few examples. Older adults are also at risk for increased morbidity
and mortality from infectious diseases and poor responses to vaccinations. In some cases,
this increased risk is for specific infectious syndromes, such as sepsis, or reactivation of vari-
cella zoster virus infection or tuberculosis. At the same time, aging of adults with chronic
viral infections such as HIV disease will result in immunologic changes that reflect the
convergence of immune dysregulation of chronic infection and of aging. This burden of
acute and chronic disease in older adults in part results from age-associated changes in the
immune system, or immunosenescence. This volume contains protocols employed by
experts in the field to study the protean effects of immunosenescence on innate and adap-
tive immune responses and includes cell biology and biochemical methods for analyses of
telomere dysfunction, autophagy, and protein oxidation. Genomic approaches for the
analysis of antigen receptor repertoire, microRNAs, and DNA methylation are also dis-
cussed. While in no way comprehensive, this volume is intended to provide a mixture of
basic and advanced protocols that will be useful for immunologists in general and investi-
gators in aging biology in particular.
This edition of Methods in Molecular Biology would not have been possible without the
efforts of the chapter authors, and I am grateful to them for taking time from their busy
schedules to contribute. I would also like to thank Professor John Walker, the Editor-in-
Chief of this series, for inviting me to edit this volume and for his constant support, and
would also like to thank Patrick Marton, David Casey, and the team at Humana Press. We
all hope that this edition will facilitate cross-fertilization and future advances aimed at
improving the health of older adults.
v
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 265
Contributors
ix
x Contributors
Abstract
Neutrophils are present within minutes to the site of aggression in the body making them one of the first
cells of the immune system to be in contact with incoming threats. The cell functions of neutrophils are
elicited through the engagement of surface receptors, some of which are located in a specific region of the
membrane called lipid rafts, a functionally segregated region of the membrane enriched with cholesterol
and distinct species of sphingomyelin and glycerophospholipids. Lipid rafts are relatively resistant to
detergent extraction and this can be taken advantage of to isolate them from the rest of the cell membrane.
This chapter will describe a reliable method to obtain lipid rafts from detergent-resistant membrane
fractions of human neutrophils. Cells are lysed in an HEPES solution containing 0.5 % Triton X-100,
supernatants are mixed with a 42 % sucrose solution, which is then overlaid with a 35 % and 5 % sucrose
solution. The gradient is centrifuged for 16 h and the resulting fractions can be further analyzed by immu-
noblotting or subjected to immunoprecipitation.
Key words Human, Neutrophils, Sucrose, Lipid raft, Flotillin, Detergent-resistant membrane
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_1, © Springer Science+Business Media New York 2015
1
2 Carl Fortin and Tamas Fülöp
2 Materials
2.2 Solutions 1. Solution A: PBS 1× pH 7.4 containing all the inhibitors men-
Required tioned in Subheading 2.1 (see Note 3).
for Neutrophil Lysis 2. Solution B: 25 mM HEPES, 100 mM NaCl, 2 mM EDTA,
pH 6.9. Add about 100 mL water to a glass beaker. Weight
1.49 g HEPES, 1.46 g NaCl, 0.146 g EDTA, and add to the
beaker. Mix until the solution is colorless and adjust the pH to
6.9. Put the solution into a 250-mL graduated cylinder and
complete to 250 mL with water. Filter the solution through a
250 mL 0.45 μm filter unit. Discard after 3 months.
2.3 Sucrose Gradient 1. 85 % sucrose solution (see Note 4): Weigh 42.5 g sucrose and
add to a glass beaker containing 10 mL of solution B. Mix
overnight with a small stir bar and, when the sucrose is dis-
solved, complete to 50 mL with solution B. Discard after 3
months.
2. 35 % sucrose solution (see Note 5): Weigh 17.5 g sucrose and
add to a glass beaker containing 25 mL of solution B. Mix with
a small stir bar until the sucrose is dissolved and complete to
50 mL with solution B. Discard after 3 months.
3. 5 % sucrose solution (see Note 5): Weigh 2.5 g sucrose and add
to a glass beaker containing 25 mL of solution B. Mix with a
small stir bar until the sucrose is dissolved and complete to
50 mL with solution B. Store the solution at 4 °C and discard
after 3 months.
2.4 Centrifugation Tubes: 1 mL polycarbonate thick wall centrifuge tubes (11 × 34 mm,
Beckman) were used with the TLA-120.2 rotor in a Beckman
Optima MAX centrifuge.
3 Methods
3.1 Neutrophil When working with primary cells, such as neutrophils, it is essential
Stimulation to follow some guidelines to prevent accidental cell activation and
reduce variability between donors. As neutrophils as extremely
sensitive to endotoxins, the entire cell isolation procedure must be
carried out under endotoxin-free conditions. Therefore, this means
using sterile, apyrogenic plasticware, sterilized and baked glassware
(LPS survives autoclaving), and low-endotoxin serum/FBS.
Equally important is to avoid heat shock, which can activate the cells.
4 Carl Fortin and Tamas Fülöp
3.3 Cell Lysis 1. Pellet the cells by doing a quickspin (4–5 s at max speed).
2. Remove supernatant by aspiration (see Note 6).
3. Resuspend the cells in 150 μL solution B containing freshly
made inhibitors (Subheading 2.1) and 0.5 % Triton X-100.
4. Incubate 10 min on ice.
5. Centrifuge 5 min at max speed to pellet cell debris.
6. Proceed immediately to gradient preparation (Subheading 3.4).
4 Notes
Acknowledgments
References
1. Mantovani A, Cassatella MA, Costantini C, 8. Zech T, Ejsing CS, Gaus K, de Wet B,
Jaillon S (2011) Neutrophils in the activation Shevchenko A, Simons K, Harder T (2009)
and regulation of innate and adaptive immu- Accumulation of raft lipids in T-cell plasma
nity. Nat Rev Immunol 11:519–531 membrane domains engaged in TCR signal-
2. Borregaard N (2010) Neutrophils, from ling. EMBO J 28:466–476
Marrow to Microbes. Immunity 33:657–670 9. Simons K, Gerl MJ (2010) Revitalizing mem-
3. Scapini P, Lapinet-Vera JA, Gasperini S, brane rafts: new tools and insights. Nat Rev
Calzetti F, Bazzoni F, Cassatella MA (2000) Mol Cell Biol 11:688–699
The neutrophil as a cellular source of chemo- 10. Gallagher SR (2012) One-dimensional SDS
kines. Immunol Rev 177:195–203 gel electrophoresis of proteins. Curr Protoc
4. Fulop T, Larbi A, Douziech N, Fortin C, Protein Sci 10(1):1–44, Chapter 10, Unit 10 1
Guerard KP, Lesur O, Khalil A, Dupuis G 11. Gordon JA (1991) Use of vanadate as protein-
(2004) Signal transduction and functional phosphotyrsine phosphatase inhibitor.
changes in neutrophils with aging. Aging Cell Methods Enzymol 201:477–482
3:217–226 12. Sitrin RG, Emery SL, Sassanella TM, Blackwood
5. Fortin CF, Lesur O, Fulop T Jr (2007) Effects RA, Petty HR (2006) Selective localization of
of TREM-1 activation in human neutrophils: recognition complexes for leukotriene B4 and
activation of signaling pathways, recruitment Formyl-Met-Leu-Phe within lipid raft microdo-
into lipid rafts and association with TLR4. mains of human polymorphonuclear neutro-
Int Immunol 19:41–50 phils. J Immunol 177:8177–8184
6. David A, Fridlich R, Aviram I (2005) The 13. Fortin CF, Sohail A, Sun Q, McDonald PP,
presence of membrane proteinase 3 in neutro- Fridman R, Fulop T (2010) MT6-MMP is
phil lipid rafts and its colocalization with present in lipid rafts and faces inward in living
FcyRIIIb and cytochrome b558. Exp Cell Res human PMNs but translocates to the cell sur-
308:156–165 face during neutrophil apoptosis. Int Immunol
7. Bournazos S, Hart SP, Chamberlain LH, 22:637–649
Glennie MJ, Dransfield I (2009) Association of 14. Hill WG, An B, Johnson JP (2002)
FcyRIIa (CD32a) with lipid rafts regulates Endogenously expressed epithelial sodium
ligand binding activity. J Immunol 182: channel is present in lipid rafts in A6 cells.
8026–8036 J Biol Chem 277:33541–33544
Chapter 2
Abstract
Natural killer (NK) cells represent a subpopulation of lymphocytes involved in innate immunity, defined
recently as group 1 of innate lymphoid cells (ILCs). NK cells are cytotoxic lymphocytes with a relevant role
in the destruction of transformed cells as virus-infected or tumor cells, as well as the regulation of the
immune response through cytokine and chemokine production that activates other cellular components of
innate and adaptive immunity. In humans, NK cell subsets have been defined according to the level of
expression of CD56. Aging differentially affects NK cell subsets and NK cell function. Here, we describe
protocols for the delineation of NK cell subsets and the analysis of their functional capacity using multipa-
rametric flow cytometry.
Key words NK cell subsets, Density gradient separation, Flow cytometry, CD107a/b degranulation
assay, Cytotoxicity
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_2, © Springer Science+Business Media New York 2015
9
10 Raquel Tarazona et al.
2 Materials
2.1 Cells, Media, 1. Whole blood collected in sodium heparin vacutainer tubes
and Solutions (2–3 tubes by donor) (see Note 1).
2. Sterile Phosphate Buffered Saline (PBS) 1×.
3. NK cell-susceptible target cells (e.g. K562 or 721.221) are
required for functional experiments.
4. Density gradient cell separation medium for mononuclear cells
as Histopaque-1077 containing Ficoll and sodium diatrizoate
(Sigma-Aldrich).
5. Complete culture medium (RPMIc): RPMI-1640 supple-
mented with 10 % heat-inactivated fetal bovine serum (FBS)
and 2 mM L-glutamine or 1 % of Glutamax (GIBCO) and pen-
icillin (50 U/ml)-streptomycin (50 U/ml).
6. Materials for freezing cells: Cold Freezing Media (Heat inacti-
vated and filtered FBS with 20 % of Dimethylsulfoxide
(DMSO)). Cryovials, freezing container (e.g. Mr. Frosty,
Nalgene) and isopropyl alcohol.
7. Optional, for NK cell isolation: NK cell-negative isolation kit
(Miltenyi) or Rosette Sep human NK cell enrichment cocktail
(StemCell Technologies, Grenoble, France).
8. Materials for proliferation assays: CellTrace CFSE proliferation
kit, from Invitrogen. Prepare CFSE aliquots at a concentration
of 5 mM (10 μl). Store at −20 °C.
9. Human recombinant IL-2 [15] in vials containing 50,000 IU/ml.
Store at −20 °C.
10. Flow cytometry buffer.
11. Trypan Blue solution (0.4 %), Neubauer chamber and
coverslips.
12. Plasticware: 30 ml sterile universal tubes, sterile pipettes, 15 ml
sterile polypropylene conical tubes and 96-well plates, U
bottom.
3 Methods
3.1 Peripheral Blood 1. Dilute whole blood from each vacutainer at least 2× with PBS.
Mononuclear Cell Carefully layer the diluted blood suspension over the
(PBMC) Isolation by Histopaque separation medium (7 ml, ~ 3:1 proportion) in a
Gradient Density 30 ml universal tube.
2. Centrifuge at 400 × g for 20 min at room temperature (RT) in
a swinging bucket rotor without brake.
3. Aspirate the mononuclear cell layer at the interphase and
deposit it in a new 30 ml tube. Fill with 1× PBS to the brim
and centrifuge at 300 × g for 5 min.
4. Discard supernatant and resuspend cell pellet in 10 ml of PBS.
Centrifuge at 300 × g for 5 min and then discard supernatant
and resuspend cell pellet again in 10 ml of PBS.
5. Count cells in a Neubauer chamber using Trypan blue for
assessment of viability.
Flow Cytometry of NK Cells in the Elderly 13
3.3 Flow Cytometry The protocol and antibody panels presented here are standardized
Analysis of NK Cell for the analysis on a 7-color MACSQuant flow cytometer (Miltenyi).
Phenotype Antibody incubation was performed for 20 min at 4 °C unless
otherwise specified. We recommend performing titration experi-
ments to obtain the optimal concentration for each antibody batch.
1. For the analysis of human NK cell subsets: CD56brightCD16negative,
CD56brightCD16+, CD56dimCD16+ and CD56negative CD16+
populations, PBMCs (0.3 × 106 to 106) obtained as indicated
in Subheading 3.1 were labeled with anti-CD3PerCP, anti-
CD56 PE-Cy7, and anti-CD16 APC-Cy7.
2. Further characterization of NK cell subsets is done by using
mAb against different surface markers such as CD57 (clone
TB03, Miltenyi Biotec), a marker of activated/senescent cells
that we include in all tubes, and antibodies against activating
and inhibitory receptors such as NKG2D, NKp46, NKp30,
DNAM-1 and CD94 together with NKG2A or NKG2C. Table 1
is a representative multicolor antibody panel used for seven-
color flow cytometry analysis of NK cells.
14 Raquel Tarazona et al.
Table 1
Example of an antibody panel for seven-color flow cytometry analysis of NK cells
APC-Vio770
VioBlue FITC PE PerCP PE-Cy7 APC or APC-Cy7
Isotope controls
Phenotype CD57 Biotin/ DNAM-1 NKp30 CD3 CD56 NKp46 CD16
antibiotin VB
CD57 Biotin/ CD94 NKG2C CD3 CD56 NKG2D CD16
antibiotin VB
CD57 Biotin/ CD85j Pan-KIR2D CD3 CD56 NKG2A CD16
antibiotin VB
Analysis of CD57 Biotin/ Granzyme B TNF-α CD3 CD56 INF-γ CD16
effector antibiotin VB
molecules and CD57 Biotin/ INF-γ TNF-α CD3 CD56 CD107a CD16
degranulation antibiotin VB
CD57 Biotin/ Perforin Granzyme A CD3 CD56 CD107a CD16
antibiotin VB
3.4 NK Cell As CD107a expression has been directly correlated with CD8+ T
Cytotoxicity cell and NK cell cytotoxicity, this marker is used for the degranula-
and Cytokine tion assays.
Production 1. Use either freshly isolated PBMCs or cryopreserved PBMCs
in Response to K562 (see Subheading 3.2).
and 721.221 Cell Lines
2. PBMCs are resuspended at 1 × 106 cells/ml in RPMIc and
rested overnight at 37 °C in a standard incubator (humidified
CO2 atmosphere).
3. The following day, PBMCs are placed in a 96-well plate at
1.5 × 106 cells/ml (200 μl final volume). First, anti-CD16
APC-Vio770 (Miltenyi Biotec) is added to all wells and the
plate is incubated for 15 min at RT. Next, Anti-CD107a-APC
(BD Biosciences) antibody is added. The recommended anti-
body dilution for CD107a conjugate is 1:10 for up to 107
cells/100 μl. For each individual a positive control containing
Flow Cytometry of NK Cells in the Elderly 15
104
4000 4000
CD16 APC-Cy7
SSC-A
SSC-A
2000 2000
102
1000 1000
101
100 100
80 R4 80 R5
60 60
40 40
20 20
0 0
10
1
102 103 104 105 101 102 103 104 105
CD57 VioBlue
Fig. 1 Representative analysis of NK cell subsets by flow cytometry. Lymphocyte gating is performed according
to FSC and SSC parameters, then CD3-negative lymphocytes are selected and the analysis of CD56 and/or
CD16 expression is represented in a dot plot in order to select the different NK cell subsets. Regions R1 to R5
represent, total NK cells, CD56negative CD16+, CD56dim CD16+, CD56bright CD16+ and CD56bright CD16negative NK cells
respectively. Histograms represent CD57 expression for each subset
3.5 Analysis of Cell 1. Separate and isolate PBMCs from blood samples as described
Proliferation by CFSE in Subheading 3.1 (see Note 7).
Staining 2. Count the cells with a Neubauer chamber and calculate viability
with Trypan blue solution.
3. Take a few cells (~200,000 cells) to perform a pre-purification
analysis (using surface markers CD3 and CD56 and their
corresponding isotype controls).
4. Purify cells using a purification kit and count purified cells
obtained (see Subheading 2.1.7).
5. Take a few cells to make a post-purification cytometry
(~100,000 cells) using anti-CD3 and anti-CD56 mAb and
quantify its efficiency (see Note 8).
6. Centrifuge the remaining volume at 300 × g, 5 min (to remove
any residual separation solution after purification). Decant and
resuspend the pellet.
7. Add to the pellet 1 ml of PBS + CFSE (1 μM final concentra-
tion) and incubate for 3 min at RT in the dark and shake the
sample (manually mixing).
8. Stop reaction by adding 10 ml of cold medium and centrifuge
at 200 × g for 10 min at 20 °C. Decant and resuspend in 10 ml
of RPMIc, wash twice.
9. Resuspend the pellet in RPMIc to a final concentration of
250,000 cell/200 μl per well (just over 106/ml). Usually we
use four wells per donor: two for isotype control (stimulated
and unstimulated) and two for sample (stimulated and
unstimulated).
10. Add 2 μl of rhIL-2 (so that the final concentration of rhIL-2 is
500 IU/ml) to the corresponding wells and incubate at 37 °C
and 5 % CO2 during 24 h.
11. The next day, take 50 μl from one of the unstimulated wells
(best use cells from the isotype control well), acquire the cells
in the cytometer and set the setting (value) for FL1 (CFSE
fluoresces on this channel). Thus, we establish the beginning
of proliferation. To do this, we set the voltage for FL1 between
103 and 104. If we have sufficient cells also set the voltages for
the rest of the channels (since unlabeled cells may also emit
auto-fluorescence).
12. Incubate for 5 days.
13. At the end of 5 days, centrifuge the plate at 300 × g for 5 min
at 20 °C. Decant the supernatant and wash twice with 200 μl
of cold sterile 1× PBS. Label the cells with mAb against CD56,
Flow Cytometry of NK Cells in the Elderly 17
CD3 and CD16 (in any color except FITC, which corresponds
to CFSE) and other surface markers of interest, like CD57.
14. Wash with 200 μl of cold sterile 1× PBS. Centrifuge at 300 × g
for 5 min at 10 °C, decant and resuspend with 200 μl of 1×
PBS or cytometry buffer. Acquire the cells the same day.
15. Analyze the data using the FlowJo software (Tree Star Inc
V.7.2.1, Ashland, OR. USA). With this program you can cal-
culate the “proliferation index” which represents the number
of cell divisions, not counting the cells that have not entered
into division.
4 Notes
Acknowledgement
References
1. Cooper MA, Fehniger TA, Caligiuri MA (2001) uncoupled from NK-cell education. Blood
The biology of human natural killer-cell subsets. 116:3853–3864
Trends Immunol 22:633–640 9. Lopez-Verges S, Milush JM, Pandey S, York
2. Poli A, Michel T, Theresine M, Andres E, VA, Arakawa-Hoyt J, Pircher H, Norris PJ,
Hentges F, Zimmer J (2009) CD56bright Nixon DF, Lanier LL (2010) CD57 defines a
natural killer (NK) cells: an important NK cell functionally distinct population of mature NK
subset. Immunology 126:458–465 cells in the human CD56dimCD16+ NK-cell
3. Romagnani C, Juelke K, Falco M, Morandi B, subset. Blood 116:3865–3874
D’Agostino A, Costa R, Ratto G, Forte G, 10. Fauriat C, Just-Landi S, Mallet F, Arnoulet C,
Carrega P, Lui G, Conte R, Strowig T, Moretta Sainty D, Olive D, Costello RT (2007)
A, Munz C, Thiel A, Moretta L, Ferlazzo G Deficient expression of NCR in NK cells from
(2007) CD56bright. J Immunol 178: acute myeloid leukemia: evolution during
4947–4955 leukemia treatment and impact of leukemia
4. Chan A, Hong DL, Atzberger A, Kollnberger cells in NCRdull phenotype induction. Blood
S, Filer AD, Buckley CD, McMichael A, Enver 109:323–330
T, Bowness P (2007) CD56bright human NK 11. Sanchez-Correa B, Gayoso I, Bergua JM,
cells differentiate into CD56dim cells: role of Casado JG, Morgado S, Solana R, Tarazona R
contact with peripheral fibroblasts. J Immunol (2012) Decreased expression of DNAM-1 on
179:89–94 NK cells from acute myeloid leukemia patients.
5. Gayoso I, Sanchez-Correa B, Campos C, Immunol Cell Biol 90:109–115
Alonso C, Pera A, Casado JG, Morgado S, 12. Sanchez-Correa B, Morgado S, Gayoso I,
Tarazona R, Solana R (2011) Immunosenescence Bergua JM, Casado JG, Arcos MJ, Bengochea
of human natural killer cells. J Innate Immun ML, Duran E, Solana R, Tarazona R (2011)
3:337–343 Human NK cells in acute myeloid leukaemia
6. Solana R, Tarazona R, Gayoso I, Lesur O, patients: analysis of NK cell-activating recep-
Dupuis G, Fulop T (2012) Innate immunose- tors and their ligands. Cancer Immunol
nescence: effect of aging on cells and receptors Immunother 60:1195–1205
of the innate immune system in humans. Semin 13. de Andrade LF, Smyth MJ, Martinet L (2014)
Immunol 24:331–341 DNAM-1 control of natural killer cells func-
7. Solana R, Pawelec G, Tarazona R (2006) Aging tions through nectin and nectin-like proteins.
and innate immunity. Immunity 24:491–494 Immunol Cell Biol 92:237–244
8. Bjorkstrom NK, Riese P, Heuts F, Andersson 14. Alter G, Malenfant JM, Altfeld M (2004)
S, Fauriat C, Ivarsson MA, Bjorklund AT, CD107a as a functional marker for the identifi-
Flodstrom-Tullberg M, Michaelsson J, cation of natural killer cell activity. J Immunol
Rottenberg ME, Guzman CA, Ljunggren Methods 294:15–22
HG, Malmberg KJ (2010) Expression pat- 15. Lahm HW, Stein S (1985) Characterization of
terns of NKG2A, KIR, and CD57 define a recombinant human interleukin-2 with micro-
process of CD56dim NK-cell differentiation methods. J Chromatogr 326:357–361
Chapter 3
Abstract
Because the incidence of organ fibrosis increases with age, various fibrosing disorders are projected to
account for significant increases in morbidity, mortality, and healthcare costs in the years to come.
Treatments for these diseases are scarce and better understanding of the immunopathogenesis of fibrosis
and its relationship to aging are sorely needed. One area of interest in this field is the role that fibrocytes
might play in the development of tissue remodeling and fibrosis. Fibrocytes are mesenchymal progenitor
cells presumed to be of monocyte origin that possess the tissue remodeling properties of tissue resident
fibroblasts such as extracellular matrix production and α-SMA-related contractile properties, as well as the
immunologic functions typically attributed to macrophages including production of cytokines and chemo-
kines, antigen presentation, regulation of leukocyte trafficking, and modulation of angiogenesis. Fibrocytes
could participate in the development of age-related fibrosing disorders through any or all of these functions.
This chapter presents methods that have been developed for the study of circulating human fibrocytes.
Protocols for the quantification of fibrocytes in the human circulation will be presented along with discus-
sion of the technical challenges that are frequently encountered in this field. It is hoped that this informa-
tion will facilitate further investigation of the relationship between fibrocytes, aging, and fibrosis, and
perhaps uncover new areas of study in these difficult-to-treat and deadly diseases.
Key words Aging, Collagen, Extracellular matrix, Fibrocyte, Fibrosis, Flow cytometry
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_3, © Springer Science+Business Media New York 2015
19
20 Xinyuan Hu et al.
Fig. 1 Representative imaging of fibrocytes from an aged subject. Panel (a) shows an image of a fibrocyte
obtained from the peripheral blood monocytes of a healthy but aged individual. This image was generated
using Amnis image stream technology in which flow cytometry is combined with real-time confocal micros-
copy. Top left: brightfield; top right: FITC-detected Pro-Collagen Iα1 (Pro-Col Iα1, pseudocolored yellow here).
Bottom left: PE-detected CD45, bottom right: merged image of CD45 and Pro-Col Iα1. (b) Brightfield image of
cultured fibrocytes. The spindle-shaped cells demonstrate the typical morphology of fibrocytes
1.3 Fibrocytes: Because human and murine fibrocyte precursors copurify with
Differentiation peripheral blood monocytes that express CD14 [40], fibrocytes are
and Homing considered to be of monocytic origin, though lineage tracing stud-
ies will be required to definitively address this question. Enrichment
for CD11b(+) CD115(+) Gr1(+) expression enhances fibrocyte
outgrowth from cultured murine monocytes; these effects require
direct contact with activated CD4+ lymphocytes and occur via an
mTOR-PI3 kinase-dependent pathway [24]. Human and rodent
fibrocyte precursors also express the Fcγ receptor [41], which
responds to aggregated IgG by inducing fibrocyte outgrowth.
22 Xinyuan Hu et al.
Table 1
Markers used to identify fibrocytes
Marker Expression
Adhesion and motility
CD9, CD11a, CD11b, CD11c, CD43 Intermediate to high
CD164, Mac2, LSP-1, CD29, CD44, CD81 Intermediate to high
ICAM-1, CD49 complex, CD34 Intermediate to high
Cell surface enzymes
CD10, CD172a, FAP Intermediate to high
CD13, Prolyl-4-hydroxylase Intermediate to high
Scavenging receptors and host defense
CD14, CD68, CD163, CD206, CD209, Conflicting reports
CD35, CD36
Fcγ receptors
CD16, CD32a, CD32b, CD32c Intermediate to high
Chemokine receptors
CCR2, CCR5, CCR4, CCR7, CCR9 Intermediate to high
CXCR1, CXCR4, CXC3R1 Intermediate to high
Antigen presentation
CD45, CD80, CD86, MHCI, MCHII Low to intermediate
Extracellular matrix
Collagen-I/III/IV, vimentin, tenascin Low to intermediate
Fibronectin, α-SMA Conflicting reports
Collagen V High
Glycosaminoglycans
Perlecan, Versican, Hyaluronan Intermediate to high
Decorin Low to intermediate
Miscellaneous
Semaphorin 7a, CD115, Thy 1.1, CD105 Low to intermediate
2 Materials
Fig. 2 Potential contributions of fibrocytes to the pathogenesis of age-related fibrosis. Fibrocyte precursors
copurify with CD14+ monocytes and possess immunomodulatory functions including production of chemo-
kines and cytokines, antigen presentation, extracellular matrix production, wound contraction, paracrine regu-
lation of angiogenesis, and regulation of leukocyte trafficking. Any or all of these functions might contribute to
the age-related immune abnormalities
3 Methods
3.1.2 Staining of Cells 1. Place approximately 1 × 106 cells/tube. Prepare the following
for CD45 and Pro- tubes for CD45 and Pro-Collagen Iα1 staining (see Note 4):
Collagen-Iα1 Expression (a) No Stain.
(b) CD45 PE (2 μL) + Intracellular isotype control + FITC
labeled secondary antibody.
(c) CD45 PE (2 μL), Rat Anti-human Procol-I and FITC
labeled secondary antibody (1 μL).
FACS-Based Detection of Circulating Human Fibrocytes 27
3.1.3 Flow Cytometry 1. Open Cell Quest, connect to the cytometer and make sure the
of Stained Samples setup box is checked.
2. Voltages for the FACS Calibur are set using the non-stain
control.
(a) Adjust the Forward Scatter (FSC) (controls lateral move-
ment of events on graph if FSC is x-axis) and Side Scatter
(SSC) (controls vertical tilt if SSC is y-axis) to record as
many of the cells of interest as possible.
28 Xinyuan Hu et al.
3.2 Analysis of Flow 1. Open the flow cytometry analysis software (in this case,
Cytometry Data FlowJo).
3.2.1 FACS Analysis 2. First a gate is set for all of the live cells based on the FSC and
for Fibrocytes SSC and applied to all samples (Fig 3a, see Note 10).
a 1k
FSC-Height. SSC-Height subset
b 104
73.6%
800 03
CD45-PE
600
SSC
02
400
01
200 FL1-FITC.FL2-CD45-PE subset
98.7%
0 100
0 200 400 600 800 1k 100 101 102 103 104
FSC-H::FSC-Height
FSC Intracellular Isotype-FITC
c 104 Q1 Q2
d 10 4
Q1 Q2
e 4 Q1 Q2
10 86.4%
99.4% 0.637% 99.5% 0.506% 13.6%
103 03 103
CD45-PE
CD45-PE
CD45-PE
102 02 102
101 01 101
Q4 Q3 Q4 Q3 Q4 Q3
0.00% 0.00% 0.00% 0.00% 0.00% 0.00%
100 100 100
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
Intracellular Isotype-FSC Pro-Coll α1-FITC Pro-Coll α1-FITC
Fig. 3 FACS-based identification of fibrocytes in human PBMCs. (a) The live cell gate is set based on forward
(FSC) vs. side scatter (SSC). (b, c) FITC-detected intracellular isotype control (X Axis) vs. Anti-CD45-PE (Y axis).
The negative gate for PE is set (b). PE-stained CD45+ve cells are selected and the negative gate for intracel-
lular FITC staining is chosen (c). (d) This gate is then applied to the sample stained with FITC-detected Pro-
Collagen- Iα1 (X Axis) vs. Anti-CD45-PE. The dual positive cells in the right upper quadrant represent fibrocytes.
The proportion of dual positive cells in the right upper quadrant in the negative control (c) is subtracted from
the proportion of dual positive cells in the sample stained with Pro-Collagen-Iα1 in a young (d) or aged (e)
subject to determine the overall percentage of double positive cells
FACS-Based Detection of Circulating Human Fibrocytes 29
4 Notes
Acknowledgements
References
1. Murray PJ, Wynn TA (2011) Protective and 15. Schmidt M, Sun G, Stacey MA et al (2003)
pathogenic functions of macrophage subsets. Identification of circulating fibrocytes as pre-
Nat Rev Immunol 11:723–737 cursors of bronchial myofibroblasts in asthma.
2. Murray LA, Rubinowitz A, Herzog EL (2012) J Immunol 171:380–389
Interstitial lung disease: is interstitial lung dis- 16. Wang CH, Huang CD, Lin HC et al (2008)
ease the same as scleroderma lung disease? Increased circulating fibrocytes in asthma with
Curr Opin Rheumatol 24:656–662 chronic airflow obstruction. Am J Respir Crit
3. Homer RJ, Elias JA, Lee CG et al (2011) Care Med 178:583–591
Modern concepts on the role of inflammation 17. Nihlberg K, Larsen K, Hultgardh-Nilsson A
in pulmonary fibrosis. Arch Pathol Lab Med et al (2006) Tissue fibrocytes in patients with
135:780–788 mild asthma: A possible link to thickness of
4. Gross TJ, Hunninghake GW (2001) Idiopathic reticular basement membrane? Respir Res
pulmonary fibrosis. N Engl J Med 345: 7:50
517–525 18. Vakil V, Sung JJ, Piecychna M et al (2009)
5. Bucala R, Spiegel LA, Chesney J et al (1994) Gadolinium-containing magnetic resonance
Circulating fibrocytes define a new leukocyte image contrast agent promotes fibrocyte differ-
subpopulation that mediates tissue repair. Mol entiation. J Magn Reson Imaging 30:
Med 1:71–81 1284–1288
6. Reilkoff RA, Bucala R, Herzog EL (2011) 19. Falk E (2006) Pathogenesis of atherosclerosis.
Fibrocytes: emerging effector cells in chronic J Am Coll Cardiol 47:C7–C12
inflammation. Nat Rev Immunol 11:427–435 20. Nikam VS, Wecker G, Schermuly R et al
7. Galligan CL, Siminovitch KA, Keystone EC (2011) Treprostinil inhibits adhesion and dif-
et al (2009) Fibrocyte activation in rheumatoid ferentiation of fibrocytes via camp and rap
arthritis. Rheumatology (Oxford) 49: dependent erk inactivation. Am J Respir Cell
640–651 Mol Biol 45:692–703
8. Douglas RS, Afifiyan NF, Hwang CJ et al 21. Murray LA, Chen Q, Kramer MS et al (2010)
(2009) Increased generation of fibrocytes in Tgf-beta driven lung fibrosis is macrophage
thyroid-associated ophthalmopathy. J Clin dependent and blocked by serum amyloid p.
Endocrinol Metab 95:430–438 Int J Biochem Cell Biol 43:154–162
9. Peng X, Mathai SK, Murray LA et al (2011) 22. Phillips RJ, Burdick MD, Hong K et al (2004)
Local apoptosis promotes collagen production Circulating fibrocytes traffic to the lungs in
by monocyte-driven cells in transforming response to cxcl12 and mediate fibrosis. J Clin
growth factor β1-induced lung fibrosis. Invest 114:438–446
Fibrogenesis Tissue Repair 4:12–25 23. Kisseleva T, Uchinami H, Feirt N et al (2006)
10. Mathai SK, Gulati M, Peng X et al (2010) Bone marrow-derived fibrocytes participate in
Circulating monocytes from systemic sclerosis pathogenesis of liver fibrosis. J Hepatol 45:
patients with interstitial lung disease show an 429–438
enhanced profibrotic phenotype. Lab Invest 24. Niedermeier M, Reich B, Gomez RM et al
90:812–823 (2009) Cd4+ t cells control the differentiation
11. Gan Y, Reilkoff RA, Peng X et al (2011) Role of gr1+ monocytes into fibrocytes. Proc Natl
of semaphorin 7a signaling in transforming Acad Sci U S A 106:17892–17897
growth factor β1-induced lung fibrosis and 25. Katebi M, Fernandez P, Chan ES et al (2008)
scleroderma-related interstitial lung disease. Adenosine a2a receptor blockade or deletion
Arthritis Rheum 63:2484–2494 diminishes fibrocyte accumulation in the skin
12. Moeller A, Gilpin SE, Ask K et al (2009) in a murine model of scleroderma, bleomycin-
Circulating fibrocytes are an indicator for poor induced fibrosis. Inflammation 31:299–303
prognosis in idiopathic pulmonary fibrosis. Am 26. Haudek SB, Cheng J, Du J et al (2010)
J Respir Crit Care Med 179:588–594 Monocytic fibroblast precursors mediate fibro-
13. Mehrad B, Burdick MD, Zisman DA et al sis in angiotensin-ii-induced cardiac hypertro-
(2006) Circulating peripheral blood fibrocytes phy. J Mol Cell Cardiol 49:499–507
in human fibrotic interstitial lung disease. 27. Haudek SB, Xia Y, Huebener P et al (2006)
Biochem Biophys Res Commun 353:104–108 Bone marrow-derived fibroblast precursors
14. Mehrad B, Burdick MD, Strieter RM (2009) mediate ischemic cardiomyopathy in mice.
Fibrocyte cxcr4 regulation as a therapeutic tar- Proc Natl Acad Sci U S A 103:18284–18289
get in pulmonary fibrosis. Int J Biochem Cell 28. Haudek SB, Trial J, Xia Y et al (2008) Fc recep-
Biol 41:1708–1718 tor engagement mediates differentiation of
32 Xinyuan Hu et al.
cardiac fibroblast precursor cells. Proc Natl 39. Bianchetti L, Barczyk M, Cardoso J et al
Acad Sci U S A 105:10179–10184 (2012) Extracellular matrix remodeling prop-
29. Buday A, Orsy P, Godo M et al (2010) erties of human fibrocytes. J Cell Mol Med
Elevated systemic tgf-beta impairs aortic vaso- 3:483–495
motor function through activation of nadph 40. Abe R, Donnelly SC, Peng T et al (2001)
oxidase-driven superoxide production and Peripheral blood fibrocytes: differentiation path-
leads to hypertension, myocardial remodel- way and migration to wound sites. J Immunol
ing, and increased plaque formation in 166:7556–7562
apoe(−/−) mice. Am J Physiol Heart Circ 41. Pilling D, Buckley CD, Salmon M et al (2003)
Physiol 299:H386–H395 Inhibition of fibrocyte differentiation by serum
30. Xu J, Gonzalez ET, Iyer SS et al (2009) Use of amyloid p. J Immunol 171:5537–5546
senescence-accelerated mouse model in 42. Castano AP, Lin SL, Surowy T, Nowlin BT
bleomycin-induced lung injury suggests that et al (2009) Serum amyloid p inhibits fibrosis
bone marrow-derived cells can alter the out- through fc gamma r-dependent monocyte-
come of lung injury in aged mice. J Gerontol A macrophage regulation in vivo. Sci Transl Med
Biol Sci Med Sci 64:731–739 1:5ra13
31. Scholten D, Reichart D, Paik YH et al (2011) 43. Bellini A, Marini MA, Bianchetti L et al (2011)
Migration of fibrocytes in fibrogenic liver Interleukin (il)-4, il-13, and il-17a differen-
injury. Am J Pathol 179:189–198 tially affect the profibrotic and proinflamma-
32. Zhang H, Maric I, DiPrima MJ, Khan J, tory functions of fibrocytes from asthmatic
Orentas RJ, Kaplan RN, Mackall CL (2013) patients. Mucosal Immunol 5:140–149
Fibrocytes represent a novel MDSC subset cir- 44. Shao DD, Suresh R, Vakil V et al (2008)
culating inn patients with metastatic cancer. Pivotal advance: Th-1 cytokines inhibit, and
Blood 122:1105–1113 th-2 cytokines promote fibrocyte differentia-
33. Van Deventer HW, Wu PQ, Bergsstralh DT, tion. J Leukoc Biol 83:1323–1333
Davis BD, O’Connor BP, Ting J, Serody JS 45. Moore BB, Murray L, Das A et al (2006)
(2008) C-C chemokine receptor 5 on pulmo- The role of ccl12 in the recruitment of fibro-
nary fibrocytes facilitates migration and pro- cytes and lung fibrosis. Am J Respir Cell Mol
motes metastasis via matrix metalloproteinase Biol 35:175–181
9. Am J Pathol 173:253–264 46. Sakai N, Wada T, Yokoyama H et al (2006)
34. Van Deventer HW, Palmieri DA, Ping Q, Secondary lymphoid tissue chemokine (slc/
McCook EC, Serody JS (2013) Circulating ccl21)/ccr7 signaling regulates fibrocytes in
fibrocytes prepare the lung for cancer metasta- renal fibrosis. Proc Natl Acad Sci U S A
sis by recruiting Ly-6C+ monocytes via CCL2. 103:14098–14103
J Immunol 190:4861–4867 47. Quan TE, Cowper S, Wu SP et al (2004)
35. Yang L, Scott PG, Giuffre J et al (2002) Circulating fibrocytes: Collagen-secreting cells
Peripheral blood fibrocytes from burn patients: of the peripheral blood. Int J Biochem Cell
identification and quantification of fibrocytes Biol 36:598–606
in adherent cells cultured from peripheral 48. Lazova R, Gould Rothberg BE, Rimm D et al
blood mononuclear cells. Lab Invest 82: (2009) The semaphorin 7a receptor plexin c1
1183–1192 is lost during melanoma metastasis. Am J
36. Pilling D, Fan T, Huang D et al (2009) Dermatopathol 31:177–181
Identification of markers that distinguish 49. Chesney J, Metz C, Stavitsky AB et al (1998)
monocyte-derived fibrocytes from mono- Regulated production of type i collagen and
cytes, macrophages, and fibroblasts. PLoS inflammatory cytokines by peripheral blood
One 4:e7475 fibrocytes. J Immunol 160:419–425
37. Chesney J, Bacher M, Bender A et al (1997) 50. Balmelli C, Alves MP, Steiner E et al (2007)
The peripheral blood fibrocyte is a potent Responsiveness of fibrocytes to toll-like
antigen-presenting cell capable of priming receptor danger signals. Immunobiology 212:
naive t cells in situ. Proc Natl Acad Sci U S A 693–699
94:6307–6312 51. Hartlapp I, Abe R, Saeed RW et al (2001)
38. Bellini A, Mattoli S (2007) The role of the Fibrocytes induce an angiogenic phenotype
fibrocyte, a bone marrow-derived mesenchy- in cultured endothelial cells and promote
mal progenitor, in reactive and reparative fibro- angiogenesis in vivo. FASEB J 15:
ses. Lab Invest 87:858–870 2215–2224
FACS-Based Detection of Circulating Human Fibrocytes 33
Abstract
Cutaneous wound healing is a complex physiological process. This process can be altered by multiple
physiological and pathological factors. Multiple pathophysiological disturbances act to impair resolution of
cutaneous wound injury, including obesity, diabetes, peripheral vascular disease, and advanced age. As our
longevity increases without a concomitant increase in healthy living years, it is plausible to assume that
problematic wound closure will continue to consume a large portion of our health care resources.
Furthermore, advanced age is associated with numerous alterations in the innate and adaptive immune
responses that complicate outcomes following cutaneous injury, trauma, or infection. Thus, models that
examine the impact of advanced age on cutaneous wound repair will be of great benefit to the develop-
ment of potential therapeutics that target age-related aberrancies in tissue repair. Herein, we detail two
animal models of tissue injury, excisional wound injury and burn injury, that can be used to evaluate
wound healing in the context of advanced age. We also describe modifications of these methods to exam-
ine wound infection following either excisional or burn injury. Lastly, we discuss methods of subsequent
tissue analysis following injury. Models described below can be further adapted to genetically engineered
murine strains to study the effects of aging and other co-morbidities on wound healing.
Key words Wounding healing, Animal model, Aging, Excisional biopsy, Burn injury, Infection
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_4, © Springer Science+Business Media New York 2015
35
36 Aleah L. Brubaker et al.
Fig. 1 Excisional wound by punch biopsy. Lift the skin in the mid-dorsal line and fold over a hockey puck (a). Push
the punch biopsy tool through the folded skin, such that the puck is visible on the other side. This will create a
wound on each side of the dorsal midline (b). Repeat this procedure for the desired number of wounds (c)
1.1 Excisional Excisional wound injury models allow for evaluation of the phases
Wound Healing of wound healing outlined above. In these models, a full thickness,
cutaneous injury results in removal of the murine epidermis,
dermis, hypodermis and the smooth muscle layer known as the
panniculus carnosus. Depending on the interest of the researchers,
the number and size of wounds can be varied using a standard
dermal punch biopsy. Briefly, young and aged mice of the desired
strains and age ranges are anesthetized and the fur is removed
either by shaving with an electric animal clipper, hair removal
cream or plucking. The skin is then lightly cleansed with ethanol,
and the wounds are induced by lifting the murine skin in the dorsal
midline and folding it over a hockey puck. Then 2–6 wounds, from
3 to 8 mm in diameter, can be created using dermal punch biop-
sies, allowing for symmetrical and identical sized wounds on either
side of the dorsal midline (Fig. 1). This method also allows for full
excision of the epidermis, dermis, hypodermis, and panniculus car-
nosus as mentioned above. Mice can then be sacrificed at early
(3 h–3 days), mid (5–10 days), or late (14 days onward) time
points to evaluate the different phases of wound repair described
above [1, 2]. At sacrifice, wounds are removed with a larger punch
biopsy than used to injure the animal to allow for consistent
removal of intact skin around the wound margin (Fig. 2). During
dissection of the pelt of the mouse, it is important to carefully
remove the tissue around the wound site to ensure removal of the
entire wound matrix. This requires special attention during the
early phases of wound repair when the granulation tissue is easily
disrupted. After removal of the tissue, the wounds can be analyzed
as detailed later in the chapter.
Fig. 2 Wound harvesting. Carefully dissect the dorsal pelt from the mouse using scissors and forceps (a). Using
scissors, cut around the wound site to ensure inclusion of all granulation tissue. Using forceps, provide counter
traction while cutting directly on top of the muscular layer to include epidermis, dermis, hypodermis, and
panniculus carnosa (b). Place the excised specimen on a hockey puck (c). Using a punch biopsy 2 mm larger
than the size used to inflict the wounds, remove the wound tissue and surrounding wound margins (d–e)
Fig. 3 Scald burn apparatus and burn template. The apparatus consists of a metal
container for water, heating plate, templates, and paper towels for drying mice
after burn injury. The template is selected based on mouse weight in order to
achieve a 12–15 % total body surface area burn. The inferior surface of the
template has a portion removed that corresponds to the needed size. Hardware
cloth is used to reinforce the bottom of the template
Fig. 4 Scald burn administration. Immerse the dorsum, template lined area of
the mouse in the water bath for 8 s. Push gently on the abdomen to ensure the
dorsum of the animal is flush with the burn template. The tail should remain out
of the template and above the water
2 Materials
2.1 Prototypical 1. BALB/c: Young 10–16 weeks, Aged 18–22 months [20, 21].
Animal Strains 2. C57BL/6: Young 8–14 weeks, Aged 25–32 months [22].
in Aging Studies
3. CBA: Young 8–14 weeks, Aged 25–27 months [23].
2.3 Burn Wound 1. Anesthesia solution: Ketamine (100 mg/mL solution), xylazine
Injury and Isolation (100 mg/mL) and sterile saline. Prior to injury, combine 1 mL
of Wound Tissue of stock ketamine, 0.2 mL of stock xylazine and 3.8 mL of
sterile, 0.9 % normal saline in a 15 mL polypropylene conical
tube. Mix by inverting the tube.
2. Sterile 0.9 % normal saline, warmed to 37 °C (Hospira Inc.,
Lake Forest, IL).
3. Electric animal hair clippers (see Subheading 2.2).
4. Hot plate.
5. Thermometer.
6. Two Metal pans with lid (1 quart, ~21.5 × 11.5 cm pans, Fisher
Scientific, Pittsburgh, PA).
7. Deionized water.
Experimental Approaches to Tissue Injury and Repair in Advanced Age 41
2.9 OCT Embedding 1. Tissue-Tek O.C.T compound (Sakura Finetek, Torrance, CA).
of Wound Tissue 2. Straight edge razor (Personna America Safety Razor Co.,
Verona, VA).
3. Disposable base molds, 15 mm × 15 mm × 5 mm (Fischer
HealthCare, Houston, TX).
3 Methods
3.2 Burn Wound 1. Prepare anesthesia mixture prior to use on day of injury as
Injury and Isolation described in Subheading 3.1 and load into single use syringe.
of Wound Tissue Weigh the mice. For a 20–25 g mouse, 100 μL of solution will
result in 100 mg/kg ketamine/10 mg/kg xylazine as desired.
2. Grasp the mouse at the nape of the neck and tail. Tilt the head
of the mouse downwards. Inject desired volume of anesthetic
i.p. in the left lower quadrant based on the weight of the animals
to achieve 100 mg ketamine/10 mg xylazine as desired.
3. Prepare the water bath by filling two metal containers with
deionized water. Heat one container to 90–92 °C for scald
burn injury. Keep the thermometer in the water bath at all times
to ensure the temperature remains tightly controlled from
mouse to mouse. The second container will be maintained at
ambient temperature for sham injury (Fig. 3).
4. Once the mouse no longer responds to firm pressure applied
to the hind limb, shave the dorsum with an electric clipper.
Be sure to shave over the entire area of anticipated injury, from
the nape of the neck to the tail. Shave beyond where the injury
itself will be placed, as skin will contract after burn. Alternate
methods of hair removal mentioned above can be used but will
not be discussed here.
5. Based on the animal’s weight, select the appropriate burn tem-
plate to give a 12–15 % total body surface area (TBSA) burn
(Fig. 3). When placing the mouse in the burn template, ensure
that the dorsum of the animal is flush with the burn template
by gently pushing on the abdomen. Ensure the tail remains out
of the template and above the water.
6. Immerse the dorsum, template lined area of the mouse in the
water bath for 8 s (Fig. 4). Watch the timer carefully to
ensure the time in the water is a constant 8 s for each mouse.
We recommend this temperature and timing to ensure a full
thickness burn.
7. Immediately remove the mouse and blot the dorsal scald area
with a paper towel to prevent continued scald injury.
8. Resuscitate the mouse by injecting 1 mL of warmed, sterile
saline i.p. in the left lower quadrant.
Experimental Approaches to Tissue Injury and Repair in Advanced Age 45
9. Return the mouse to a clean cage and place the cages on heat-
ing pads for 3–4 h, or until the mice are aroused from anesthe-
sia. Given that these studies involve evaluation the inflammatory
stages of wound healing, no analgesics are given as they can
interfere with the inflammatory immune process.
10. Sacrifice mice by CO2 inhalation at desired time point to evalu-
ate the various phases of wound healing as delineated above.
11. To harvest the wounds, carefully dissect the dorsal pelt from
the mouse. Remove an extra 4 mm of tissue from the margin
of the burn wound to include all granulation tissue.
12. Place the pelt on a hockey puck. Use a 5–8 mm punch biopsy
to remove the burn tissue at the wound margin, such that half
of the punch biopsy contained burn wound and the remaining
half contains intact skin from the wound margin.
13. Process the tissue as described below.
3.3 Models 1. Follow the steps above under Subheading 3.1 through step 6.
of Combined Wound 2. Immediately after injury, pipette desired CFU/mL in 10 μL
Injury and Infection: directly into each open wound bed. Be sure to change pipette
Excisional Wound tips between each wound on a given mouse.
Injury (a) S. aureus growth and inoculation: S. aureus of the desired
and Staphylococcus strain can be grown overnight in tryptic soy broth (TSB) at
aureus Infection 37 °C under constant agitation. The next day, 1 mL of S.
aureus in TSB is resuspended in 2 mL fresh TSB and incu-
bated at 37 °C for 2 h under constant agitation to ensure
mid-logarithmic growth at the time of application to cuta-
neous wounds. Bacterial concentration (CFU/mL) is then
determined by absorbance at 600 nm and the final inocu-
lum confirmed by back-plating on mannitol salt agar (MSA;
BD Diagnostics, Sparks, MD).
3. Resume the protocol under Subheading 3.1 at step 7.
Evaluation of bacterial colonization at the time of sacrifice is
described below.
3.4 Burn Wound 1. Follow the steps above under Subheading 3.2 through step 8.
Injury and Topical (a) P. aeruginosa growth and inoculation: P. aeruginosa of the
Pseudomonas desired strain can be grown overnight on centrimide agar
aeruginosa Infection plates (BD Diagnostics, Sparks, MD) at 37 °C. The next day,
inoculate sterile saline with one loop of P. aeruginosa from
centrimide agar plate. Bacterial concentration (CFU/mL)
is then determined by absorbance at 600 nm and the final
inoculum confirmed by back-plating on centrimide agar
plates (BD Diagnostics, Sparks, MD).
2. After towel drying the mouse to prevent further scald injury,
slowly pipette desired bacteria concentration in CFU/mL in
46 Aleah L. Brubaker et al.
3.6 Measurement 1. Attach the digital camera onto the camera stand at a fixed
of Wound Closure by distance above the height of the hockey puck. We recommend
Digital Photography a fixed distance of 20 cm.
and Image Analysis 2. Remove the dorsal pelt as described above in Subheading 3.2.
3. Place the pelt flat on the hockey puck. Align a metric ruler with
the wounds in the frame of the photograph.
Experimental Approaches to Tissue Injury and Repair in Advanced Age 47
3.7 Wound Cell A detailed procedure for isolation and staining of wound cells can be
Isolation for Flow found in our previously published manuscript [24]. The procedure
Cytometry below only details the initial processing for tissue for flow cytom-
etry following wound excision. Details of subsequent processing
and staining can be carried out as described [24].
1. Prepare “Dispase Solution.”
2. Remove dorsal pelt and excise wounds from mice at desired
time-points following excisional cutaneous injury as described
above. Be sure to not use any ethanol in washing of the pelt as
ethanol may disrupt the cell membrane and promote lysis. If
you wish to wash the pelt, do so in sterile PBS.
3. Collect 2–3 wounds per animal and determine weight of sam-
ples in grams.
4. Mince wounds into small pieces (<2 mm × 2 mm) with a
straight razor.
5. Place wound pieces into a 24-well culture plate (1 well/
animal) filled with 15.4 mL “Dispase Solution”/gram of tissue
(see Note 2).
6. Incubate plate overnight at 4 °C on a rotating shaker at a gentle
setting.
7. Follow remaining step as outlined [24].
48 Aleah L. Brubaker et al.
3.8 RNA Isolation 1. Immediately following removal of the wound tissue, add 1–2
from Wound Tissue wounds to a 1.7 mL Eppendorf tube and snap freeze in liquid
nitrogen.
2. Store at −80 °C until ready to process tissue for RNA.
3. Using the RNA Easy Kit (Qiagen), isolate RNA as per the
manufacturer’s instructions (see Note 3).
3.9 Protein Isolation 1. Prepare Lysis Buffer: 9.9 mL BioRad Lysis Buffer, 40 μL
from Wound Tissue BioRad Factor I, 20 μL BioRad Factor 2, and 40 μL PMSF.
2. Place frozen tissue samples on dry ice.
3. Add 1 mL of Lysis Buffer to each polypropylene tube and keep
on wet ice.
4. Using forceps, remove tissue and place in polypropylene tube
and homogenize for 30 s until no tissue bits are left. Return to
ice bucket.
5. Spin the polypropylene tubes at 300 × g at 4 °C for 5 min.
6. Transfer the supernatant to 1.7 mL Eppendorf tubes.
7. Sonicate samples at a 30 % setting for 10 s.
8. Centrifuge at 4 °C for 10 min at 2655 × g (5000 rpm using
an Eppendorf centrifuge 5417 R, Eppendorf, Hamburg,
Germany).
9. Remove the supernatant, avoiding the pellet, with a 1 mL
single-use syringe.
10. Filter the supernatant through a 25 μM single use syringe filter
into a clean 1.7 mL Eppendorf tube. Maintain tube on ice.
11. Aliquot sample into fresh 1.7 mL Eppendorf tubes.
12. Snap freeze in liquid nitrogen and store at −80 °C until use.
3.10 OCT Embedding 1. Place a small amount of OCT compound into the Tissue Tray.
of Wound Tissue Allow to lightly cover the bottom of the cassette.
2. Using the straight razor, cut the outer 1/3 of wound.
3. Place the wound so the straight edge is flush with edge of tis-
sue cassette.
4. Cover the tissue with OCT, avoiding bubbles.
5. Freeze immediately by placing cassette on dry ice.
6. Store at −80 °C until use.
4 Notes
Acknowledgements
The authors thank Jessica Palmer for her critical review and discussion
of this manuscript. This work was supported by NIH R21
AI073987 (E.J.K.), R01 AG018859 (E.J.K.), T32 AG031780
(P.L.W.), T32 GM008750 (R.L.G.) and by the Dr. Ralph and
Marian C. Falk Medical Research Trust (E.J.K.).
Experimental Approaches to Tissue Injury and Repair in Advanced Age 51
References
1. Singer AJ, Clark RA (1999) Cutaneous wound 14. Slater H, Gaisford JC (1981) Burns in older
healing. N Engl J Med 341:738–746 patients. J Am Geriatr Soc 29:74–76
2. Reinke JM, Sorg H (2012) Wound repair and 15. Linn BS (1980) Age differences in the severity
regeneration. Eur Surg Res 49:35–43 and outcome of burns. J Am Geriatr Soc 28:
3. Gillitzer R, Goebeler M (2001) Chemokines in 118–123
cutaneous wound healing. J Leukoc Biol 16. Clayton MC, Solem LD, Ahrenholz DH
69:513–521 (1995) Pulmonary failure in geriatric patients
4. Kim MH, Liu W, Borjesson DL et al (2008) with burns: the need for a diagnosis-related
Dynamics of neutrophil infiltration during group modifier. J Burn Care Rehabil 16:
cutaneous wound healing and infection using 451–454
fluorescence imaging. J Invest Dermatol 17. Ely EW, Wheeler AP, Thompson BT et al
128:1812–1820 (2002) Recovery rate and prognosis in older
5. Daley JM, Brancato SK, Thomay AA et al persons who develop acute lung injury and the
(2010) The phenotype of murine wound mac- acute respiratory distress syndrome. Ann Intern
rophages. J Leukoc Biol 87:59–67 Med 136:25–36
6. Nissen NN, Polverini PJ, Koch AE et al (1998) 18. Dancey DR, Hayes J, Gomez M et al (1999)
Vascular endothelial growth factor mediates ARDS in patients with thermal injury. Intensive
angiogenic activity during the proliferative phase Care Med 25:1231–1236
of wound healing. Am J Pathol 152:1445–1552 19. Le HQ, Zamboni W, Eriksson E et al (1986)
7. Swift ME, Kleinman HK, DiPietro LA (1999) Burns in patients under 2 and over 70 years of
Impaired wound repair and delayed angiogen- age. Ann Plast Surg 17:39–44
esis in aged mice. Lab Invest 79:1479–1487 20. Nomellini V, Brubaker AL, Mahbub S et al
8. Sen CK, Gordillo GM, Roy S et al (2009) (2012) Dysregulation of neutrophil CXCR2
Human skin wounds: a major and snowballing and pulmonary endothelial ICAM-1 promotes
threat to public health and the economy. age-related pulmonary inflammation. Aging
Wound Repair Regen 17:763–771 Dis 3:234–247
9. Kaye KS, Anderson DJ, Sloane R et al (2009) 21. Birjandi SZ, Ippolito JA, Ramadorai AK et al
The effect of surgical site infection on older (2011) Alterations in marginal zone macro-
operative patients. J Am Geriatr Soc 57:46–54 phages and marginal zone B cells in old mice.
10. Vowden KR, Vowden P (2009) The preva- J Immunol 186:3441–3451
lence, management and outcome for patients 22. Zook EC, Krishack PA, Zhang S et al (2011)
with lower limb ulceration identified in a Overexpression of Foxn1 attenuates age-
wound care survey within one English health associated thymic involution and prevents the
care district. J Tissue Viability 18:13–19 expansion of peripheral CD4 memory T cells.
11. Christensen K, Doblhammer G, Rau R et al Blood 118:5723–5731
(2009) Ageing populations: the challenges 23. Bhattacharyya TK, Thomas JR (2004)
ahead. Lancet 374:1196–1208 Histomorphologic changes in aging skin:
12. Brubaker AL, Palmer JL, Kovacs EJ (2011) observations in the CBA mouse model. Arch
Age-related dysregulation of inflammation and Facial Plast Surg 6:21–25
innate immunity: lessons learned from rodent 24. Brubaker AL, Schneider DF, Palmer JL, Faunce
models. Aging Dis 2:346–360 DS, Kovacs EJ (2011) Wound cell isolation for
13. Panda A, Arjona A, Sapey E et al (2009) flow cytometric and functional analysis neutro-
Human innate immunosenescence: causes and phils and natural killer T cells as negative regula-
consequences for immunity in old age. Trends tors of wound healing. J Immunol Methods
Immunol 30:325–333 373:161–166
Chapter 5
Abstract
By facilitating the simultaneous analysis of parameters from diverse cell lineages and biological pathways,
multicolor flow cytometry is integral to many studies in human immunology—particularly those in older
individuals—where sample amounts may be limiting. Studies in human cohorts require particular attention
to fluorochrome panel design and procedures to standardize instrument performance; reproducible instru-
ment conditions (over time and between centers) are crucial to accurate comparisons and conclusions in
the analysis of heterogeneous groups of human subjects. Here, we describe procedures for multicolor digi-
tal flow cytometry, our experience in flow cytometry panel design and our approach in standardizing
instrument performance using BD Biosciences hardware and software (BD Biosciences, San Jose, CA).
These techniques allow for the generation of accurate and precise data in a variety of settings.
Key words Multicolor flow cytometry, Translational immunology, Application settings, Flow cytometry
standardization, Flow cytometry panel design
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_5, © Springer Science+Business Media New York 2015
53
54 Samit R. Joshi et al.
2 Methods
2.1 Selection 1. The investigator should identify the specific cytometer, available
of Color Matrix lasers, and installed filters at their institution. This information
can be entered into an online spectral viewer allowing for an
approximation of the spectral overlap between wavelengths
(e.g. http://www.bdbiosciences.com/research/multicolor/
spectrum_viewer/index.jsp). In our experience, employing
more than 8–9 colors can lead to an increase in spectral overlap
and subsequent difficulties in compensation. However, others
have developed 15-color panels with success [6]. Consider that
even though an instrument may support n colors, given the
need to maintain resolution of populations, optimal panel design
may only employ a portion of the available colors (e.g. n − 2).
Sufficient time should be dedicated to panel design to optimize
results (see Note 1).
2.4 Create 1. Since many institutions, including our own, use BD Flow
Application Settings Cytometers (BD Biosciences, San Jose, CA), we will take this
opportunity to briefly describe key features in BD FACSDiva
v6.0 software. Like many others, our center utilizes Cytometer
Setup and Tracking (CST) beads for reproducible instrument
setup and tracking of performance on a daily basis. We find this
leads to decreased variability and improved data quality. Using
the information gathered from the CST baseline cytometer
support, Application settings can be established for a particular
assay. We find the use of Application settings allows for consis-
tent settings, less spillover, higher resolution, and more repro-
ducible data by automatically adjusting Photomultiplier Tube
(PMT) voltages based upon daily instrument performance. In
our experience, the use of both Application settings and CST
beads will keep a consistent Mean Fluorescence Intensity
(MFI) for a particular parameter over time despite any potential
minor variations in instrument performance. This is particularly
56 Samit R. Joshi et al.
Fig. 1 Sample cytometer baseline report. Red arrows indicate baseline maximum linearity and electronic noise
rSD for each respective parameter, respectively. The operator should calculate one-half the maximum linearity
and 2.5–3× the rSD (this can be done separately on paper or using computer software)
Fig. 2 Lymphocyte gate (P1) in global worksheet. Note adjustments using FSC,
SSC, and actual gate can be adjusted when acquiring PBMCs
Fig. 3 Table of statistics (rSD). Creation of a table of statistics only with rSD values (red arrow). Notice how our
sample experiment only uses six flurochromes
58 Samit R. Joshi et al.
Fig. 4 Adjust parameter voltage. While recording events, the voltage of each parameter can be adjusted
individually (red arrow)
Fig. 5 Using unstained PBMCs to optimize voltage based on 2.5–3× the baseline rSD. A new tube (red arrow)
is created after each adjustment of the voltage for the respective parameter’s rSD (in this case V450, green
arrow) to be within 2.5–3× the baseline value
Fig. 6 Table of statistics (median) creation of a new table of statistics only with median values (red arrow)
60 Samit R. Joshi et al.
Fig. 7 Creation of individual histograms. Creation of individual histograms for each respective parameter (each
with their own gate on their own bright population) showing only the P1 population (in our case PBMCs stained
for CD8 using a v450-conjugated antibody, red arrow). Note: prior to running the single flurochrome stained
PBMCs, this is a reasonable estimate of where the bright CD8+ population will be. This can be modified during
acquisition of the PBMCs (see Fig. 8)
Fig. 8 Individual histogram when recording PBMCs stained with a v450-conjugated CD8+ antibody. The table
of statistics shows the Median (P2 gate) of PBMCs stained for CD8 using a v450-conjugated antibody, red
arrow. Notice how the initial position of the P2 gate has changed from Fig. 7 to the actual position of the bright
population
2.5 Run 1. When conducting a new experiment, first apply the Application
Compensation settings to the experiment by right clicking on cytometer
settings and then clicking on Application settings. The optimized
voltages (created above) will be imported.
2. Use unstained PBMCs and single color stained CompBeads (BD
Biosciences, San Jose, CA) to create a compensation matrix.
With the designated Application settings, we find we do not
need to further adjust our PMT voltages and subsequent
spectral overlap should be optimized. With low spillover the
operator should achieve the best resolution possible. Finally,
the operator can record their experiment.
62 Samit R. Joshi et al.
Fig. 9 Individual histogram when recording PBMCs stained with APC-conjugated CD8+ antibody. The table of
statistics shows the Median (P3 gate) of PBMCs stained for CD8 using a APC-conjugated antibody, red arrow.
Again, notice how the P3 gate is on the bright population
3 Notes
Acknowledgements
References
1. Sugar IP, Gonzalez-Lergier J, Sealfon SC (2011) RR, Shaw AC (2010) Age-associated decrease
Improved compensation in flow cytometry by in TLR function in primary human dendritic
multivariable optimization. Cytometry A cells predicts influenza vaccine response. J
79(5):356–360. doi:10.1002/cyto.a.21062 Immunol 184(5):2518–2527. doi:10.4049/
2. Kalina T, Flores-Montero J, van der Velden VH, jimmunol.0901022
Martin-Ayuso M, Bottcher S, Ritgen M, Almeida 5. van Duin D, Allore HG, Mohanty S, Ginter S,
J, Lhermitte L, Asnafi V, Mendonca A, de Tute Newman FK, Belshe RB, Medzhitov R, Shaw
R, Cullen M, Sedek L, Vidriales MB, Perez JJ, te AC (2007) Prevaccine determination of the
Marvelde JG, Mejstrikova E, Hrusak O, expression of costimulatory B7 molecules in acti-
Szczepanski T, van Dongen JJ, Orfao A (2012) vated monocytes predicts influenza vaccine
EuroFlow standardization of flow cytometer responses in young and older adults. J Infect Dis
instrument settings and immunophenotyping 195(11):1590–1597. doi:10.1086/516788
protocols. Leukemia 26(9):1986–2010. 6. Mahnke YD, Roederer M (2007) Optimizing a
doi:10.1038/leu.2012.122 multicolor immunophenotyping assay. Clin Lab
3. Maino VC, Picker LJ (1998) Identification of Med 27(3):469–485. doi:10.1016/j.
functional subsets by flow cytometry: intracellu- cll.2007.05.002, v
lar detection of cytokine expression. Cytometry 7. Edinger M (2009) Multicolor flow cytometry—
34(5):207–215 principles of panel design. http://www.bd.
4. Panda A, Qian F, Mohanty S, van Duin D, com/videos/bdb/webinars/multicolor_flow_
Newman FK, Zhang L, Chen S, Towle V, principals_of_panel_design/. Accessed 21 Feb
Belshe RB, Fikrig E, Allore HG, Montgomery 2013
Chapter 6
Abstract
Flow cytometry is an invaluable technique that can be used to phenotypically and functionally characterize
immune cell populations ex vivo. This technology has greatly advanced our ability to gain critical insight
into age-related changes in immune function, commonly known as immune senescence. Rodents have been
traditionally used to investigate the molecular mechanisms of immune senescence because they offer the
distinct advantages of an extensive set of reagents, the presence of genetically modified strains, and a short
lifespan that allows for longevity studies of short duration. More recently, nonhuman primates (NHPs), and
specifically rhesus macaques, have emerged as a leading translational model to study various aspects of
human aging. In contrast to rodents, they share significant genetic homology as well as physiological and
behavioral characteristics with humans. Furthermore, rhesus macaques are a long-lived outbred species,
which makes them an ideal translational model. Therefore, NHPs offer a unique opportunity to carry out
mechanistic studies under controlled laboratory conditions (e.g., photoperiod, temperature, diet, and med-
ications) in a species that closely mimics human biology. Moreover similar techniques (e.g., activity record-
ing and MRI) can be used to measure physiological parameters in NHPs, making direct comparisons
between NHP and human data sets possible. In addition, the outbred genetics of NHPs enables rigorous
validation of research findings that goes beyond proof of principle. Finally, self-selection bias that is often
unavoidable in human clinical trials can be completely eliminated with NHP studies. Here we describe flow
cytometry-based methods to phenotypically and functionally characterize innate immune cells as well as T
and B lymphocyte subsets from isolated peripheral blood mononuclear cells (PBMC) in rhesus macaques.
Key words Flow cytometry, Lymphocyte, B cell, T cell, Regulatory T cell, Rhesus macaque, Age,
Proliferation, Cytokine, Dendritic cell, Monocyte, Natural killer cell, Pattern recognition receptor
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_6, © Springer Science+Business Media New York 2015
65
66 Christine Meyer et al.
2 Materials
Table 1
Innate cell antibody information for use in rhesus macaques
Table 2
Intranuclear and intracellular antibody information
for use in rhesus macaques
Table 3
Rhesus macaque B and T cell antibody panel
3 Methods
3.1 Delineating The innate immune system is the first line of defense against patho-
Innate Immune Cells gens and its action is mediated by several immune cell subsets that
by Flow Cytometry include granulocytes (e.g., neutrophils), natural killer (NK) cells,
dendritic cells (DC), and monocytes/macrophages. Because little
information is available for rhesus macaque granulocytes, they are
not discussed here.
1. Add 2 × 106 peripheral blood mononuclear cells (PBMC) per
well in a 96-well round bottomed plate (see Notes 1 and 2).
Rhesus PBMC are isolated from whole blood by centrifuga-
tion over Ficoll (density 1.077) exactly as described for human
blood. PBMC yields per milliliter blood are also comparable
between rhesus and humans.
2. Add 150 μl of PBS to each well (washing step) and spin down
plate at 900 × g for 3 min. Discard the supernatant and briefly
vortex the plate (see Notes 3 and 4).
3. Repeat step 2 with 200 μl of PBS.
4. Add surface antibody cocktail described in Table 1 diluted to
50 μl of PBS (see Note 5) resuspend, and incubate for 20 min
at 4 °C in the dark.
Flow Cytometry of Immunosenescence in Non-Human Primates 69
5. Add 150 μl of PBS to each well and spin down plate at 900 × g
for 3 min at 4 °C. Discard supernatant, and briefly vortex
plate.
6. Repeat step 5 with 200 μl of PBS.
7. Resuspend cells in 100 μl fixation buffer. Incubate for 30 min
at 4 °C (see Note 6).
8. Add 100 μl of PBS to each well, spin down at 900 × g for 3 min
at 4 °C, discard supernatant, and briefly vortex pellet. Repeat
this wash step with 200 μl of PBS.
9. Resuspend cells in 100 μl of PBS and transfer to microtiter
tubes for acquisition on flow cytometer. Samples may be
stored at 4 °C for up to 48 h until they can be acquired.
10. Acquire samples on LSR II or similar instrument and analyze
data using FlowJo or comparable software. Since monocytes
represent 3–5 % and DC 1–2 % of the PBMC, it is important
to acquire at least 1 × 106 events.
11. Following acquisition by flow cytometer, samples can be
analyzed as depicted in Fig. 1: First, exclude AQUA LIVE/
DEAD+ cells (dead), CD3 (T cells), and CD20 (B cells) to
define the live “non-lymph” population. From here, classical
monocytes are defined as CD14hiCD16- cells, whereas
nonclassical monocytes are defined as CD14loCD16+ cells [1];
DCs are defined as HLA-DR+CD14- cells and can be further
subdivided using CD123 and CD11c markers into CD123+
plasmacytoid DCs (pDCs) and CD11c+ myeloid DCs (mDCs)
[2]; NK cells are defined as CD14-HLA-DR-CD8α+ and can
be subdivided into a major CD16+ cytolytic population and a
minor CD16- cytokine-producing population. Two major
differences between human and rhesus macaque NK cells
should be noted here: (1) rhesus macaque NK cells, but not
human NK cells, express high-levels of CD8α [3]; and (2)
human but not rhesus macaque NK cells express CD56. CD56
is expressed by monocytes in rhesus macaques [4].
a b c
CD20
non-lymph
FSC
d e CD16+
monocytes
CD16
DC CD16-
monocytes
HLA-DR CD14
g f
pDC
IFNγ- Cytotoxic
CD8a
NK cells
CD123
NK cells
mDC
CD16 CD11c
Fig. 1 Delineation of innate immune cells using flow cytometry. Live PBMCs are discriminated by forward scat-
ter (FSC) and side scatter (SSC) measuring size and granularity, respectively (a), and Live/Dead gating (b). CD3
to CD20− cells are identified as non-lymph cells (c). Monocytes and DCs are HLA-DR+ (d). CD14 and CD16
co-staining can be used to delineate major monocyte/macrophage subpopulations (e). HLA-DR+ CD14− cells
can be gated to delineate DCs (e), which are further subdivided into CD123+ plasmacytoid DC (pDC) and
CD11c+ myeloid DCs (mDC) (f). NK cells are identified as HLA-DR− cells that are CD8α+ and are subdivided
into a major CD16+ cytotoxic subset and a minor CD16− IFNγ-producing subset (g)
Non-Lymph gate
a b c
Monocyte pDC
CD123
CD14
CD3
mDC
DC
IL-6
13.4
96.2 3.2 17.8 60.1
TNFa IFNa
Fig. 2 Cytokine production by innate immune cells. PBMC were incubated for 6 h with either RP-10 alone
(“unstimulated”), or in the presence of LPS or Imiquimod. Non-lymph cells are identified as CD3 to CD20− cells
(a). Cells were then stained for surface markers to delineate monocytes (HLA-DR+ CD14+ cells) or DCs (HLA-DR+
CD14−) (b), with CD123 and CD11c used to delineate pDC and mDC respectively (c). Cells were stained intracel-
lularly for TNFα, IL-6, and IFNα. Representative TNFα/IL-6 costaining by monocytes unstimulated (d) or incu-
bated with LPS (e) and representative IFNα staining by pDCs unstimulated or stimulated with Imiquimod (f)
1. Add 1 × 106 PBMCs per well into a 96-well plate (see Note 6,
Table 3).
2. Spin plates at 900 × g for 3 min. Discard supernatant and vor-
tex plate (see Note 3).
3. Add surface antibodies described in Table 3 in 50 μl of PBS
per well and incubate in the dark at 4 °C for 30 min.
4. Wash plate with 150 μl of PBS, spin for 3 min at 900 × g at
4 °C. Discard supernatant and vortex plate.
5. Repeat step 4 with 200 μl of PBS.
6. If staining panel requires a secondary antibody (see Note 10)
dilute in 50 μl of PBS per well and incubate plate in dark for
20 min at 4 °C.
7. Add 150 μl of PBS to the wells and spin for 3 min at 900 × g at
4 °C. Discard supernatant and vortex plate.
8. Repeat step 7 using 200 μl of PBS.
Flow Cytometry of Immunosenescence in Non-Human Primates 73
a b c
exhausted
SSC
CD3
IgD
Fig. 3 Flow-cytometric gating strategy to delineate B cell populations. Live PBMC are delineated as previously
described (a). B cells are identified as CD3− CD20+ (b). Naïve B cells are IgD+ CD27−, marginal zone-like
(MZ-like) B cells are IgD+ CD27+, and memory B cells are IgD− CD27+ (c)
74 Christine Meyer et al.
a b CD4 c
transitional CM
naïve CM CM
EM EM
CD28
CD28
CD4
d CD4 e
CD25+
CD127+
T reg
CD25
CD127 Foxp3
Fig. 4 Flow-cytometric gating strategy to delineate T cell populations. Lymphocytes are separated into CD4+
and CD8+ T cells (a). CD4 T and CD8 T cells are subdivided into naïve CD95− CD28+, central memory (CM,
CD95+ CD28+) and effector memory (EM, CD95+ CD28−) T cells (b). The addition of CCR7 allows further
subdivision of the memory CD95+ T cells population and the identification of transitional CM T cells CD95+
CD28+ CCR7− (c). T regulatory (T reg) cells are defined as CD4+ CD25+ CD127− (d). Confirmation is provided
by the expression of Foxp3+ in this population (e)
3.4 Monitoring One of the hallmarks of the adaptive immune response is a robust
Immune Cell proliferative burst. Ki67 is a nuclear protein that is upregulated
Proliferation Through during the G2–S phase of the cell cycle [11], and is an excellent
the Detection of Ki67 marker for determining kinetics and magnitude of a proliferative
Expression burst. Rhesus macaques are an outbred population with a complex
Flow Cytometry of Immunosenescence in Non-Human Primates 75
a b
EM
CD28
CD4
d
CD8 CD95 50
30
20
30.8
4.34 10
0
-14 3 7 10 14 17 21 28
Ki67 days post infection
Fig. 5 Measuring T cell proliferation via analysis of Ki67 expression. Lymphocytes are separated into CD4+ and
CD8+ T cells (a). CD4 EM T cell subsets are delineated based on CD28 and CD95 expression (b). Ki67+ expres-
sion in CD4 EM T cells increases dramatically 14 days post-infection with simian varicella virus (SVV) com-
pared to day 0 post-infection (c). Representative example of CD4 EM T cell proliferative burst following SVV
infection (average values of six animals, d)
a 24.1
b 0.265 0.0294 3 22.2
0dpi 14dpi
IFNg
62.3
CD4
CD8 TNFa
30
IL-2
20
10
Fig. 6 Measuring frequency of antigen-specific T cells using intracellular cytokine production by flow cytom-
etry. Lymphocytes are separated into CD4+ and CD8+ T cells (a). Representative examples of the frequency of
IFNγ/TNFα− producing (b) and IL-2/MIP-1β− producing (c) CD4 T cells at 0 and 14 days post-infection with
simian varicella virus (SVV) following ex vivo stimulation with SVV lysate are shown. Frequency of antigen-
specific T cells can be reliably monitored over the course of SVV infection using ICS as demonstrated in panel
d with the monitoring of the frequency of IFNγ+ CD4 T cells (average of six animals)
4 Notes
References
1. Passlick B, Flieger D, Ziegler-Heitbrock HW of memory B cells expressing costimulatory
(1989) Identification and characterization of a molecules in the resting state. J Immunol
novel monocyte subpopulation in human 167:5669–5677
peripheral blood. Blood 74:2527–2534 8. Weller S, Braun MC, Tan BK et al (2004)
2. Coates PT, Barratt-Boyes SM, Zhang L et al Human blood IgM “memory” B cells are cir-
(2003) Dendritic cell subsets in blood and culating splenic marginal zone B cells
lymphoid tissue of rhesus monkeys and their harboring a prediversified immunoglobulin
mobilization with Flt3 ligand. Blood 102: repertoire. Blood 104:3647–3654
2513–2521 9. Pitcher CJ, Hagen SI, Walker JM et al (2002)
3. Ramothea L, Webster RPJ (2005) Delineation Development and homeostasis of T cell mem-
of multiple subpopulations of natural killer cells ory in rhesus macaque. J Immunol 168:
in rhesus macaques. Immunology 115:206–214 29–43
4. Carter LL, Murphy KM (1999) Lineage- 10. Picker LJ, Reed-Inderbitzin EF, Hagen SI
specific requirement for signal transducer and et al (2006) IL-15 induces CD4 effector mem-
activator of transcription (Stat)4 in interferon ory T cell production and tissue emigration in
gamma production from CD4(+) versus nonhuman primates. J Clin Invest 116:
CD8(+) T cells. J Exp Med 189:1355–1360 1514–1524
5. Asquith M, Haberthur K, Brown M et al 11. Gerdes J, Lemke H, Baisch H et al (1984) Cell
(2012) Age-dependent changes in innate cycle analysis of a cell proliferation-associated
immune phenotype and function in rhesus human nuclear antigen defined by the mono-
macaques (Macaca mulatta). Pathobiol Aging clonal antibody Ki-67. J Immunol 133:
Age Relat Dis. doi:10.3402/pba.v2i0.18052 1710–1715
6. Kawai T, Akira S (2011) Toll-like receptors and 12. Fujiwara T, Oda K, Yokota S et al (1988)
their crosstalk with other innate receptors in Brefeldin A causes disassembly of the Golgi
infection and immunity. Immunity 34:637–650 complex and accumulation of secretory pro-
7. Bar-Or A, Oliveira EM, Anderson DE et al teins in the endoplasmic reticulum. J Biol
(2001) Immunological memory: contribution Chem 263:18545–18552
Chapter 7
Abstract
The standard for single-cell analysis of phenotype and function in recent decades has been fluorescence
flow cytometry. Mass cytometry is a newer technology that uses heavy metal ions, rather than fluoro-
chromes, as labels for probes such as antibodies. The binding of these ion-labeled probes to cells is quan-
titated by mass spectrometry. This greatly increases the number of phenotypic and functional markers that
can be probed simultaneously. Here, we review topics that must be considered when adapting existing flow
cytometry panels to mass cytometry analysis. We present a protocol and representative panels for surface
phenotyping and intracellular cytokine staining (ICS) assays.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_7, © Springer Science+Business Media New York 2015
81
82 Michael D. Leipold et al.
103 103
DNA1:Ir191
DNA1:Ir191
102 102
Intact singlets
71.6
101 101
0 0
104 Monocytes
30.7 104
103
CD33:Er166
103
DNA1:Ir191
102
102
Live cells
96.1
101
101
0 Lymphocytes
69.1 0
The metals that are sold as part of antibody labeling kits are
of very high purity (98% and higher in most cases). As a practi-
cal matter, this means that “compensation” analogous to fluo-
rescent antibodies is not needed, as most of the signal will be
of the specified mass, with little to no signal at “M+1” or
another contaminating mass. However, metal salts from other
commercial sources may be of lesser purity. For example, the
chemistries of the lanthanides (Ln) are sufficiently similar that
undesired lanthanides (often La139) can be contaminants in
purchased salts since they are difficult to purify using only
chemical methods. There are currently no labeling kits con-
taining Gd157 due to purity concerns about the available salts.
If using these less-pure isotopes, some caveats to consider
include the following:
(a) Consider using them for “dump” channels or exclusion
markers. If only events that are negative for the label in
question are subjected to further analysis, the impurities
present should not cause any issues.
(b) Put a lower-abundance marker at a less-pure “M” so that
the absolute spillover (usually up to 0.5–1% of “M” sig-
nal) is reduced. If “M” is a less pure isotope and is labeling
a high-abundance marker, do not put a low-abundance
marker at the M+1 position. Aim for at least medium-
abundance so that positive and negative populations can
still be clearly resolved if there is isotopic “spillover.”
(c) For channels that have significant spillover, use combina-
tions of markers that label mutually exclusive populations.
For instance, put a T-cell-specific marker at M+1 when
using a B-cell-specific marker labeled with a less-pure iso-
tope M.
2. Impurities from the sample or environment. There are several
sources of impurities to guard against. When in doubt, a highly
diluted aliquot of the suspected stock can be injected into the
mass cytometer in tuning/liquid mode and observed for
contamination.
(a) Many laboratory dish soaps have high levels of barium
(AM 130-138). Barium tends to persist even after multi-
ple rinses. This is a problem even if these masses are not
being used in the experiment, as it leads to detector aging,
and can result in oxidation signals in M+16 channels (see
point #3 below). It is therefore generally advised to store
mass cytometry buffers in brand-new plastic or glass ves-
sels that have never been through laboratory wash.
(b) Low levels of mercury, lead, or tin can sometimes be
found in lab buffers, especially those made with "house"
distilled water rather than reverse osmosis (e.g., MilliQ)
Multiparameter Phenotyping of Human PBMCs Using Mass Cytometry 87
Fig. 2 Representative comparisons of fluorescence (top row) and mass cytometry (bottom row) for the same
antibody combinations on cryopreserved PBMC from the same donor. Parent populations are shown at the top
of each column. Left to right: naïve and memory B cell sub-populations, CD4+ and CD8+ T cells, and NK cells.
In each case, the staining patterns differ somewhat, especially with regard to background staining of negative
populations; but overall frequencies of each gated population are very similar. All data was collected using
Data Dual (Dd) calibration
4 4
10 104 104 CD8+ cells 10 104
CD20+ cells 25
Monocytes
3 17.3 3 3
103 6.95 10 10 103 10
2 2 2
102 10 10 102 10
NK cells
36.1
1 1
10 101 101 10 101
NonB NonT B cells
CD3+ cells
CD8:Nd144
6.5 93.4
CD33:Er166
CD20:Dy164
CD56:Yb174
CD19:Nd142
75.4
0 Lymphocytes 0 0 CD4+ cells
0 0
Michael D. Leipold et al.
91.7 50.3
1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4
0 101 102 103 104 0 10 10 10 10 0 10 10 10 10 0 10 10 10 10 0 10 10 10 10
Live cells CD14:Sm154 Lymphocytes CD3:Nd150 CD3+ cells CD4:Nd143 CD20+ cells CD19:Nd142 NonB NonT CD16:Sm149
2
10 102 102 102 102
1 1
10 101 101 101 10
CD28:Er167
CD38:Eu151
Treg
CD85j:Sm147
CD127:Ho165
0 0 0 0 0
CD45RA:Dy162
4.91
6.02 13.7 0.114 0.435 87.4 4.44 96.1 0.795
CD4+ cells CD25:Yb176 CD4+ cells CCR7:Gd160 CD4+ cells CD27:Sm152 CD4+ cells CD94:Gd156 CD4+ cells HLADR:Lu175
Fig. 3 Representative staining of various cell-surface (a) and intracellular (b) markers in a 38-antibody panel that includes eight intracellular cytokines. Plots show stain-
ing from a positive control, stimulated with PMA+ionomycin, but the panel has been successfully used with antigen-specific stimulations as well. Resolution of surface
and intracellular markers is generally quite good. The parent population is shown at the bottom of each plot. All data was collected using Data Dual (Dd) calibration
4 IL-2+ 4 4 4
10 10 10 10
0.104
3 3 3 3 MIP1b+
10 10 IFNg+ 10 IL-17+ 10
0 5.62e-3 0.0281
CD4+ T cells
2 2 2 2
10 10 10 10
unstimulated
1 1 1 1
10 10 10 10
IL-2:Gd157
IFNg:Gd158
IL-17:Dy164
MIP1b:Nd148
CD107a+
0 0 0 GMCSF+ 0
0.177 TNFa+ 0.141 CD69+
0.127 2.56
1 2 3 4 1 2 3 4 1 2 3 4 1 2 3 4
0 10 10 10 10 0 10 10 10 10 0 10 10 10 10 0 10 10 10 10
4 IL-2+ 4 4 4
10 10 10 10
30.1
2 2 2 2
CD4+ T cells
10 10 10 10
stimulated with
1 1 1 1
10 10 10 10 PMA + ionomycin
IL-2:Gd157
IFNg:Gd158
IL-17:Dy164
MIP1b:Nd148
Fig. 3 (continued)
Multiparameter Phenotyping of Human PBMCs Using Mass Cytometry
91
92 Michael D. Leipold et al.
2 Materials
3.1 Sample 1. Two million viable cells per well (as measured by dye exclusion
Collection method such as Vicell; rested, if desired) (see Note 3).
3.4 Data Acquisition 1. Start the machine. Warm up and tune as in Ref. [14].
and Analysis 2. Acquire data as in Ref. [14]. The length of the run will be
dependent upon the volume of your sample: dilution with
MilliQ water to ~1 million cells/mL is recommended for
minimizing doublets as well as maximizing sample throughput
(see Note 10).
3. Analyze data using third-party flow analysis software such as
FlowJo (Treestar) or Cytobank (see Note 11 and Fig. 1). Note
that some display settings may need to be altered for proper
viewing.
4 Notes
Acknowledgments
The authors thank Sean Bendall for helpful discussions, and Sheena
Gupta and Meena Malipatlolla for contributing example data.
Development of this protocol was funded in part by grant 2 U19
AI057229 S4 from the National Institutes of Health.
References
1. Kalisky T, Quake SR (2011) Single-cell 7. Behbehani GK, Bendall SC, Clutter MR, Fantl
genomics. Nat Methods 8:311–314 WJ, Nolan GP (2012) Single-cell mass cytom-
2. Ornatsky O, Bandura D, Baranov V, Nitz M, etry adapted to measurements of the cell cycle.
Winnik MA, Tanner S (2010) Highly multipa- Cytometry A 81:552–566
rametric analysis by mass cytometry. J Immunol 8. Fienberg HG, Simonds EF, Fantl WJ, Nolan
Methods 36:1–20 GP, Bodenmiller B (2012) A platinum-based
3. Bandura DR, Baranov VI, Ornatsky OI, covalent viability reagent for single-cell mass
Antonov A, Kinach R, Lou X, Pavlov S, cytometry. Cytometry A 81:467–475
Vorobiev S, Dick JE, Tanner SD (2009) Mass 9. Maecker HT (2009) Multiparameter flow
cytometry: technique for real time single cell cytometry monitoring of T cell responses.
multitarget immunoassay based on inductively Methods Mol Biol 485:375–391
coupled plasma time-of-flight mass spectrom- 10. Maecker HT, Frey T, Nomura LE, Trotter J
etry. Anal Chem 81:6813–6822 (2004) Selecting fluorochrome conjugates for
4. Newell EW, Sigal N, Bendall SC, Nolan GP, maximum sensitivity. Cytometry A 62:169–173
Davis MM (2012) Cytometry by time-of- 11. Rundberg Nilsson A, Bryder D, Pronk CJ
flight shows combinatorial cytokine expression (2013) Frequency determination of rare popu-
and virus-specific cell niches within a contin- lations by flow cytometry: a hematopoietic stem
uum of CD8(+) T cell phenotypes. Immunity cell perspective. Cytometry A 83:721–727
36:142–152 12. Ornatsky OI, Kinach R, Bandura DR, Lou X,
5. Bendall SC, Simonds EF, Qiu P, Amir el AD, Tanner SD, Baranov VI, Nitz M, Winnik MA
Krutzik PO, Finck R, Bruggner RV, Melamed R, (2008) Development of analytical methods for
Trejo A, Ornatsky OI, Balderas RS, Plevritis SK, multiplex bio-assay with inductively coupled
Sachs K, Pe'er D, Tanner SD, Nolan GP (2011) plasma mass spectrometry. J Anal At Spectrom
Single-cell mass cytometry of differential immune 23:463–469
and drug responses across a human hematopoi- 13. Lovelace P, Maecker HT (2010) Multi-
etic continuum. Science 332:687–696 parameter intracellular cytokine staining.
6. Bodenmiller B, Zunder ER, Finck R, Chen TJ, Methods Mol Biol 699:165–178
Savig ES, Bruggner RV, Simonds EF, Bendall 14. Leipold MD, Maecker HT (2012) Mass
SC, Sachs K, Krutzik PO, Nolan GP (2012) cytometry: protocol for daily tuning and run-
Multiplexed mass cytometry profiling of ning cell samples on a CyTOF mass cytometer.
cellular states perturbed by small-molecule J Vis Exp. doi:10.3791/4398
regulators. Nat Biotechnol 30:858–867
Chapter 8
Imaging Immunosenescence
Feng Qian and Ruth R. Montgomery
Abstract
To demonstrate effects of aging visually requires a robust technique that can reproducibly detect small
differences in efficiency or kinetics between groups. Investigators of aging will greatly appreciate the ben-
efits of Amnis ImageStream technology (www.amnis.com/), which combines quantitative flow cytometry
with simultaneous high-resolution digital imaging. Imagestream is quantitative, reproducible, feasible with
limited samples, and it facilitates in-depth examination of cellular mechanisms between cohorts of samples.
Key words Flow cytometry, Microscopy, Image analysis, Immune response, Signal transduction,
Aging, Monocyte, Dendritic cell, Neutrophil
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_8, © Springer Science+Business Media New York 2015
97
98 Feng Qian and Ruth R. Montgomery
2 Materials
Table 1
Antibody panel for cell lineage staining of signaling pathways
3 Methods
Fig. 1 Gating strategy for human monocytes from PBMCs. Monocytes are distinguished in gates with normal
nuclear dye DAPI intensity and high DAPI aspect ratio (R1) which are labeled with lineage marker CD14 (R2).
A high correlation of NF-κB with DAPI nuclear dye localization is reflected in a high similarity score and indi-
cates the degree of activation in CD14+ monocytes (R3)
12. To label cell surface lineage markers, we first remove the stim-
ulation medium by centrifuging samples at 300 × g for 10 min
at 4 °C, then remove the supernatant taking care to avoid dis-
turbing the pellet.
13. Wash cells once by resuspending samples in 200 μl cold
Staining Buffer, centrifuge at 300 × g for 10 min at 4 °C, and
discard the supernatant.
14. Label cells by resuspending samples in 50 μl Staining Buffer
containing specific antibodies or isotype controls and incubate
for 20 min at 4 °C protected from light. A sample staining
panel is shown in Table 1 (see Note 3).
15. At the end of the staining period, wash cells by adding 200 μl
cold Staining Buffer, and centrifuge at 300 × g for 10 min at
4 °C. Remove the supernatant.
16. Fix cells by adding 0.1 ml 4 % paraformaldehyde in PBS for
10 min at RT protected from light.
102 Feng Qian and Ruth R. Montgomery
Fig. 2 Translocation of NF-κB in human monocytes after stimulation. The translocation of NF-κB (p65) into the
nucleus after stimulation (R3) is depicted in populations of untreated and stimulated cells. The median value
of similarity scores is 1.16 in untreated sample and 2.34 in stimulated sample (a). Digital images collected
simultaneously of the untreated or flagellin-stimulated cell populations show representative cells and the
intensity of NF-κB translocated into the cell nucleus (b)
29. Design a gating strategy template for the study and apply the
gates equivalently across all study samples (see Note 4).
30. For data processing, identify monocytes using Amnis IDEAS
software and follow gates as shown in Fig. 1. To distinguish
in-focus single cells from debris, gate on events with a normal
nuclear dye DAPI intensity and high aspect ratio (defined as
the ratio of the width of a shape to its height) for DAPI (R1).
31. To identify monocytes, choose cells gated with high intensity
labeling of the CD14 marker (R2).
32. To identify activated cells after stimulation, the similarity score
of NF-κB and the nuclear dye DAPI will be calculated in
CD14+ cells. Similarity measures the degree to which two
images (NF-κB/DAPI) are correlated on a pixel-by-pixel
basis. A high correlation of NF-κB with DAPI, indicating co-
localization of the two markers, is reflected in a high
“Similarity” score (R3). After stimulation, the treated cells
were determined to have a significantly higher similarity score
of NF-κB p65 with DAPI, indicating that NF-κB p65 translo-
cated into the nucleus after stimulation. Representative images
of cells are shown in Fig. 2.
33. The labeling and gating strategy described here works well for
both monocytes and dendritic cells and should be effective for
other cell types that show nuclear translocation of NF-κB after
stimulation.
34. Differences between subject groups will be readily apparent
through dot plots, histograms, and cell images. Data represen-
tative of the study cohort may be presented as populations in
histogram format, and/or representative images (where they
are particularly compelling). Data from within a defined
gate—such as the “Similarity” score—can be tabulated for
each study subject and/or group and is suitable for statistical
determinations.
4 Notes
Acknowledgement
References
1. Higashi T, Watanabe W, Matsunaga S (2012) 4. George TC, Fanning SL, Fitzgerald-Bocarsly P
Application of visualization techniques for cell et al (2006) Quantitative measurement of
and tissue engineering. J Biosci Bioeng 115: nuclear translocation events using similarity
122–126 analysis of multispectral cellular images obtained
2. Herbert S, Soares H, Zimmer C et al (2012) in flow. J Immunol Methods 311:117–129
Single-molecule localization super-resolution 5. Ploppa A, George TC, Unertl KE et al (2011)
microscopy: deeper and faster. Microsc ImageStream cytometry extends the analysis of
Microanal 18:1419–1429 phagocytosis and oxidative burst. Scand J Clin
3. Masedunskas A, Milberg O, Porat-Shliom N Lab Invest 71:362–369
et al (2012) Intravital microscopy: a practical 6. Qian F, Montgomery RR (2012) Quantitative
guide on imaging intracellular structures in live imaging of lineage specific Toll-like receptor
animals. Bioarchitecture 2:143–157 mediated signaling in monocytes and dendritic
106 Feng Qian and Ruth R. Montgomery
cells from small samples of human blood. J Vis monocytes of systemic lupus erythematosus
Exp 62:e3741 patients is triggered by circulating autoantigens
7. Qian F, Wang X, Zhang L et al (2012) Age- independent of type I interferons. Arthritis
associated elevation in TLR5 leads to increased Rheum 64:788–798
inflammatory responses in the elderly. Aging 9. Schluchter MD, Elashoff JD (1990) Small-
Cell 11:104–110 sample adjustments to tests with unbalanced
8. Stone RC, Feng D, Deng J et al (2012) repeated measures assuming several covariance
Interferon regulatory factor 5 activation in structures. J Statist Comput Simul 37:69–87
Chapter 9
Abstract
Aging impairs humoral immune responses, leading to increased frequency and severity of infectious
diseases and reduced protective effects of vaccination. We have identified B-cell biomarkers that are reduced
by aging and that can be used as predictive markers of the response of an individual to vaccination. The
identification of these biomarkers will have an impact on the development of effective vaccines to protect
the elderly from infections and other debilitating diseases.
Key words Activation-induced cytidine deaminase, Class switch recombination, Antibody vaccine
response
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_9, © Springer Science+Business Media New York 2015
107
108 Daniela Frasca et al.
2 Materials
2.1 B-Cell Isolation We perform this procedure using the MACS Cell separation proto-
col from Miltenyi Biotec (Miltenyi), that is why we buy most of the
materials and reagents from this company.
1. Suspension of human PBMC in complete medium (RPMI
1640, supplemented with 10 % FBS, 10 μg/ml Pen-Strep,
1 mM Sodium Pyruvate, and 2 × 10–5 M 2-Mercaptoethanol
and 2 mM L-glutamine).
2. CD19 Microbeads (Miltenyi Biotec).
3. MACS buffer. A solution of 0.5 % fetal bovine serum (FBS) or
0.5 % Bovine Serum Albumin in 1× Phosphate-Buffered Saline
(PBS).
AID and Memory B Cells in Vaccine Response 109
2.3 mRNA Extraction 1. μMACS mRNA isolation kit (Miltenyi), containing Oligo
(dT) Microbeads, Lysis/Binding, Wash and Elution buffers.
2. Lysate clear columns and μMACS columns (Miltenyi).
3. μMACS separator (Miltenyi).
2.6 In Vivo Antibody This can be measured by the hemagglutination inhibition assay
Response (HAI), which is the most established correlate with vaccine protec-
tion [7, 10, 17, 18]. The technique is not described here but it will
be used below for correlations with our B-cell-specific biomarkers
(see Figs. 1 and 2).
110 Daniela Frasca et al.
0.4
0.3
0.2
0.1
p=0.0030
0.0
0 100 200 300 400
Vaccine-specific HAI
(reciprocal of the titers
after vaccination)
8
(% of CD19+)
p=0.0081
0
0 100 200 300 400
Vaccine-specific HAI
(reciprocal of the titers
after vaccination)
3 Methods
3.1 Isolation This procedure should be performed under a laminar flow hood to
of Human B Cells preserve sterility and in a BSL-2 laboratory, implementing all the
by Positive Selection adequate safety measures for working with potentially infectious
with CD19 Microbeads human samples. All personnel should be immunized with the
Hepatitis B vaccine.
Positive selection of B cells is the best protocol to obtain highly
purified B cells, without other contaminating cell types (T cells,
NK cells, monocytes, macrophages). In long-term cultures (2–7
days), B cells isolated with this procedure, compared to B cells
isolated with negative selection, give comparable results. Negative
selection should be preferred only in studies on signal transduction
performed in minutes from selection (5–30 min), in order to avoid
effects of antibody cross-linking of cell surface proteins.
1. Resuspend PBMC in MACS buffer at a concentration of
107 cells/80 μl of buffer (see Note 2).
2. Add CD19 Microbeads (20 μl per 107 cells), mix well, and incu-
bate for 15–20 min at 4 °C. Usually the B-cell yield is about
2–10 % of the total PBMC. A minimum of 105 B cells can be
used for activation in culture and reliable detection of AID.
3. Wash cells with 5 ml of buffer, by centrifuging at 300 × g for
10 min. Discard supernatant and resuspend in 500 μl of MACS
buffer.
4. Place the MS columns (one per sample) in the MACS separa-
tor, rinse the columns with 500 μl of MACS buffer and apply
cell suspension onto the column.
5. Wash the column twice with 500 μl of MACS buffer.
6. Remove column from the separator and place it in a centrifuge
tube for collection of the adherent fraction.
7. Add 1 ml of complete RPMI onto the column, and collect
CD19+ cells by pushing the plunger into the column. Count
the cells by Trypan blue exclusion dye with a hemocytometer.
3.3 mRNA Extraction 1. Extract mRNA from cultured cells using the μMACS mRNA
from Activated Human isolation kit. Before starting, the Lysis/Binding buffer and the
B Cells Wash buffer should be brought to room temperature, whereas
the Elution buffer must be heated at 65 °C in a water bath.
112 Daniela Frasca et al.
4 Notes
References
1. Okazaki IM, Kinoshita K, Muramatsu M et al 11. Khurana S, Frasca D, Blomberg B et al (2012)
(2002) The AID enzyme induces class switch AID activity in B cells strongly correlates with
recombination in fibroblasts. Nature 416: polyclonal antibody affinity maturation in-vivo
340–345 following pandemic 2009-H1N1 vaccination
2. Yoshikawa K, Okazaki IM, Eto T et al (2002) in humans. PLoS Pathog 8:e1002920
AID enzyme-induced hypermutation in an 12. Sayegh CE, Quong MW, Agata Y et al (2003)
actively transcribed gene in fibroblasts. Science E-proteins directly regulate expression of
296:2033–2036 activation-induced deaminase in mature B
3. Pone EJ, Zan H, Zhang J et al (2010) Toll-like cells. Nat Immunol 4:586–593
receptors and B-cell receptors synergize to 13. Gardner EM, Bernstein ED, Dran S et al
induce immunoglobulin class-switch DNA (2001) Characterization of antibody responses
recombination: relevance to microbial anti- to annual influenza vaccination over four years
body responses. Crit Rev Immunol 30:1–29 in a healthy elderly population. Vaccine
4. Stavnezer J, Guikema JE, Schrader CE (2008) 19:4610–4617
Mechanism and regulation of class switch recom- 14. LeMaoult J, Szabo P, Weksler ME (1997)
bination. Annu Rev Immunol 26:261–292 Effect of age on humoral immunity, selection
5. Muramatsu M, Nagaoka H, Shinkura R et al of the B-cell repertoire and B-cell develop-
(2007) Discovery of activation-induced cyti- ment. Immunol Rev 160:115–126
dine deaminase, the engraver of antibody 15. McElhaney JE, Effros RB (2009)
memory. Adv Immunol 94:1–36 Immunosenescence: what does it mean to
6. Rada C, Williams GT, Nilsen H et al (2002) health outcomes in older adults? Curr Opin
Immunoglobulin isotype switching is inhib- Immunol 21:418–424
ited and somatic hypermutation perturbed 16. Amanna IJ, Carlson NE, Slifka MK (2007)
in UNG-deficient mice. Curr Biol 12: Duration of humoral immunity to common
1748–1755 viral and vaccine antigens. N Engl J Med
7. Frasca D, Diaz A, Romero M et al (2010) 357:1903–1915
Intrinsic defects in B cell response to seasonal 17. Murasko DM, Bernstein ED, GardnerE M
influenza vaccination in elderly humans. et al (2002) Role of humoral and cell-medi-
Vaccine 28:8077–8084 ated immunity in protection from influenza
8. Frasca D, Landin AM, Lechner SC et al (2008) disease after immunization of healthy elderly.
Aging down-regulates the transcription factor Exp Gerontol 37:427–439
E2A, activation-induced cytidine deaminase, 18. Skowronski DM, Tweed SA, De Serres G
and Ig class switch in human B cells. J Immunol (2008) Rapid decline of influenza vaccine-
180:5283–5290 induced antibody in the elderly: is it real, or is
9. Gibson KL, Wu YC, Barnett Y et al (2009) it relevant? J Infect Dis 197:490–502
B-cell diversity decreases in old age and is cor- 19. Schmittgen TD, Livak KJ (2008) Analyzing
related with poor health status. Aging Cell real-time PCR data by the comparative C(T)
8:18–25 method. Nat Protoc 3:1101–1108
10. Frasca D, Diaz A, Romero M et al (2012) 20. Frasca D, Diaz A, Romero M et al (2011) Age
Unique biomarkers for B-cell function predict effects on B cells and humoral immunity in
the serum response to pandemic H1N1 influ- humans. Ageing Res Rev 10:330–335
enza vaccine. Int Immunol 24:175–182
Chapter 10
Abstract
One of the most noticeable changes in T-cell immunity with aging is the expansion of memory CD8+ T
cells, with a decline in naïve phenotype T cells that reflects both diminished thymopoiesis and the effects
of chronic antigenic stimulation with age. Flow cytometry is a useful tool in evaluating immune cells
including the phenotype characteristics of different T-cell subsets. Here, we show flow cytometric methods
measuring the different subsets of human CD8+ T cells that change with aging.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_10, © Springer Science+Business Media New York 2015
115
116 Min Sun Shin and Insoo Kang
2 Materials
CCR7
2000
CD6
2
SSC
10
50K 0
0
0 0
2 3 4 5 2 3 4 5 2 3 4 5
0 50K 100K 150K 200K 250K 0 10 10 10 10 0 10 10 10 10 0 10 10 10 10
e CD8+ T cells
60 60 60 60
Isotype
40 40 40 40
20 20 20 20
0 0 0 0
3 4 5 3 4 5 3 4 5 3 4 5
0 10 10 10 0 10 10 10 0 10 10 10 0 10 10 10
Fig. 1 Identification of different CD8+ T cell subsets with differential expression of IL-7Rα in human peripheral
blood using flow cytometry. Peripheral blood was drawn into a heparinized tube from a healthy control after
obtaining informed consent. Peripheral blood mononuclear cells (PBMCs) were purified by Ficoll-Paque centrifu-
gation. PBMCs were stained with antibodies to CD3, CD4, CD8, CD45RA, CCR7 and IL-7Rα or isotype antibodies.
Stained cells were analyzed on an LSRII® flow cytometer. (a) Lymphocytes were identified by forward and side
light scatters. (b–c) CD4+ and CD8+ T cells were identified based on CD3, CD4 and CD8 expression. (d) Naïve
(CD45RA+ CCR7+), central memory (CM, CD45RA− CCR7+), CD45RA− effector memory (EM, CD45RA− CCR7-)
and CD45RA+ effector memory (CD45RA+ CCR7−) CD8+ T cells are identified. (e) IL-7Rα expression by differ-
ent CD8+ T cell subsets is detected based on staining with antibodies to IL-7Rα (open) and isotype control
Table 1
Antibodies used in analyzing T-cell phenotype
3 Methods
4 Notes
References
1. Reiner SL (2007) Development in motion: memory and effector CD8+ T cells. Mech
helper T cells at work. Cell 129:33–36 Ageing Dev 125:615–618
2. Lee N, Shin MS, Kang I (2012) T-cell biology 8. Lee WW, Shin MS, Kang Y et al (2012) The
in aging, with a focus on lung disease. J relationship of cytomegalovirus (CMV) infec-
Gerontol A Biol Sci Med Sci 67:254–263 tion with circulatory IFN-alpha levels and IL-7
3. Starr TK, Jameson SC, Hogquist KA (2003) receptor alpha expression on CD8(+) T cells in
Positive and negative selection of T cells. Annu human aging. Cytokine 58:332–335
Rev Immunol 21:139–176 9. Sallusto F, Lenig D, Forster R et al (1999)
4. Murphy KM, Stockinger B (2010) Effector T Two subsets of memory T lymphocytes with
cell plasticity: flexibility in the face of changing distinct homing potentials and effector func-
circumstances. Nat Immunol 11:674–680 tions. Nature 401:708–712
5. Cui W, Kaech SM (2010) Generation of effec- 10. Kim HR, Hong MS, Dan JM et al (2006)
tor CD8+ T cells and their conversion to Altered IL-7R{alpha} expression with aging
memory T cells. Immunol Rev 236:151–166 and the potential implications of IL-7 therapy
6. Shin MS, Lee N, Kang I (2011) Effector T-cell on CD8+ T-cell immune responses. Blood
subsets in systemic lupus erythematosus: 107:2855–2862
update focusing on Th17 cells. Curr Opin 11. Soloski MJ, Chrest FJ (2013) Multi-parameter
Rheumatol 23:444–448 flow cytometry for discovery of disease mecha-
7. Hong MS, Dan JM, Choi JY et al (2004) Age- nisms in rheumatic diseases. Arthritis Rheum
associated changes in the frequency of naive, 65:1148–1156
Chapter 11
Abstract
The antibody response to vaccination has been the industry and regulatory standard for evaluating influenza
vaccine efficacy. Although antibodies are an important defense mechanism providing sterilizing immunity,
in older adults, the cellular immune response is also needed for clinical protection against the serious com-
plications of influenza. Thus, the demonstration of enhanced antibody responses as a strategy for advanc-
ing new influenza vaccines through the standard clinical development pipeline may fail to translate to
enhanced protection in the older population. In peripheral blood mononuclear cells (PBMC) challenged
with live influenza virus, an increase in the interferon-γ:interleukin-10 (IFN-γ:IL-10) ratio and the level of
the cytolytic mediator, granzyme B (GrzB), correlates with protection against influenza in vaccinated older
adults. This chapter provides detailed methods for measuring these cell-mediated immune responses,
which have been validated according to the International Conference on Harmonisation (ICH) guide-
lines. These immune correlates could be combined with antibody responses to improve the prediction of
enhanced protection in vaccine trials in the older population.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_11, © Springer Science+Business Media New York 2015
121
122 Janet E. McElhaney and Beth Gentleman
2 Materials
2.5 Reagent 1. All the Influenza virus strains detailed are routinely used to
Preparation for Virus stimulate PBMC samples but other virus strains or stimulants
Stimulation such as Concanavalin A can be used.
2. For each virus strain (stock tube and dilution tube), resuspend
and mix well with micropipette before creating aliquots or
stimulating cultures. Virus particles tend to settle to the
bottom of the tube and may not reach full infectivity if insuf-
ficiently distributed.
3. Thaw each stock tube quickly by rolling in between hands and
once thawed, create aliquots (to limit freeze/thaw cycles) of
the appropriate size based on the number of stimulations to
be performed per week, and then immediately refreeze and
store at −80 °C. Do not create aliquots smaller than 100 μL.
Also, if all of the aliquot is not used, do not refreeze as more
Cytokine and Granzyme B Assessment of Influenza Response 125
Tris-HCI 3.63 g
Triton X-100 2 mL
Deionized H2O 100 mL
NaCl 1.753 g
Sucrose 100 g
CHAPS 1g
HEPES 23.83 g
Deionized H2O 400 mL
3 Methods
3.1 Stimulation 1. After isolation and counting of PBMC, the final cell concentration
of PBMC with Live is to be adjusted to 3 × 106 viable PBMC/mL suspended in
Influenza Virus AIM V medium (see Note 5).
2. Distribute 0.6 mL of 3 × 106 cells/mL cell suspension to the
appropriate number of wells in the tissue culture plate pre-
pared for virus stimulation. Each culture well is stimulated
with only one virus strain; therefore, when testing multiple
strains of virus, set up the necessary number of cultures to test
all the required strains/subject (final number of cells per well:
3 × 106 cells/mL × 0.6 mL/well = 1.8 × 106 cells/well).
3. Stimulate each well with virus (MOI = 2) by placing pipette tip
directly into culture and injecting the contents. Place the TC
plate in a 37 °C CO2 incubator for 20 h and then harvest the
culture supernatants and cell lysates (see Note 6).
3.2 Harvesting Cell 1. Remove cell culture plates from 37 °C CO2 incubator, and
Culture Supernatants inspect cultures under microscope to observe changes to
and Cell Lysates PBMC. It is not necessary to centrifuge the plate as the cells
will have all settled to the bottom of the culture wells within
the 20 h incubation period.
2. Working in the biological safety cabinet, remove the superna-
tants and transfer to 2 mL cryogenic tubes. To avoid drawing
up cells at the bottom of the well and remove as much super-
natant as possible, tilt the plate towards you, at a gentle/slight
angle so as to not draw the cells down, and get as close to the
bottom of the well as possible without touching it. Then, draw
up the supernatant from the front corner of the well very
slowly. It’s important that only approximately ≤20 μL of
supernatant remains in the well to minimize dilution of the
lysis buffer to be added.
3. During the collection process, tubes with cell supernatants
should be placed on ice and then frozen at −80 °C for later use
in assay for cytokines.
4. After removing the supernatant, add 200 μL Cell Lysis Buffer
to the well containing 1.8 × 106 cells/culture. If the cell quan-
tity/culture changes, adjust the amount of Cell Lysis Buffer
added accordingly. Process at room temperature. To ensure no
drying of the cells after you have removed the supernatant,
immediately add the Cell Lysis Buffer and process each well
individually before moving on to the next (see Note 7). To
effectively lyse all the cells, scrape the bottom of the well thor-
oughly (approximately minimum of 1 min scrapping/well) with
the pipette tip (use a motion as if coloring), being sure to scrape
along the edges as well and then resuspend a couple times, from
the front corner of the well, to fully collect all the lysate and
transfer this suspension to 1.5 mL microcentrifuge tubes.
During this step it is also important to avoid the formation of
Cytokine and Granzyme B Assessment of Influenza Response 129
bubbles, which will hinder the removal of all the lysate from the
well. To avoid bubbles, use a fluid motion and avoid picking
up the pipette tip, keeping it in constant contact with the well
bottom. The well should appear clean after successful removal
of cells (see Note 8).
5. Once the lysate has been transferred to a tube, let each lysate
sit for approximately 5–10 min at room temperature to allow
the Triton X-100 to fully permeate the cells. If processing
larger batches of cultures, after 5–10 min at room temperature,
transfer the lysate tubes to ice and then freeze within 30 min of
collection to preserve GrzB activity. Freeze the lysates in open
racks at −80 °C to ensure quick freezing and then once frozen
transfer to freezer storage boxes. Do not store at −20 °C under
any conditions.
6. When ready, continue to GrzB/Protein and Multiplex
Cytokine assays.
3.3 Lysate 1. Prepare the cell lysates by completing three freeze/thaw cycles,
Preparation vortexing after each thaw cycle:
(a) Place tubes in −80 °C freezer until the lysates are frozen
(~5 min in open racks to allow faster freeze cycles).
(b) Immediately place the tubes into a 37 °C water bath for
~2 min or until the frozen lysates have thawed. Do not
leave unattended in water bath.
(c) Vortex the samples vigorously (high speed for 30 s).
(d) Immediately repeat the freeze/thaw cycle two more times.
2. Using a refrigerated microcentrifuge, centrifuge samples at top
speed (~16,000 × g) for 5 min to 4 °C to keep the lysates cold
and to pellet cell debris, nuclei, and DNA.
3. Remove most of the lysate while safely avoiding contact with
the pellet (~150 μL of lysate is removed if the original volume
of Cell Lysis Buffer used to lyse the cells was 200 μL as leaving
behind 50 μL is best to fully avoid the pellet—contact with
the pellet introduces contaminants which introduces back-
ground activity to the GrzB assay) and transfer to two fresh-
labeled tubes (100 μL dispensed into one tube and 50 μL in
other tube). When splitting lysate between tubes, resuspend
well in between to dispense the same concentration of protein
into the tubes. One aliquot can be used for initial GrzB/
Protein assays (need minimum of ~70 μL for both assays) and
the other for later repeats if necessary. For this entire step all
original sample tubes and transfer tubes (precool) need to be
on ice (see Note 9).
4. Continue to keep samples on ice, to preserve GrzB activity, and
proceed directly to GrzB/Protein assays or freeze at −80 °C
until later use (limit to one freeze/thaw cycle) (see Note 10).
130 Janet E. McElhaney and Beth Gentleman
3.4 Determination Assay Principle: Lysates are transferred to microtiter plates following
of Granzyme B Activity three freeze/thaw cycles and incubated with the substrate IEPDpNA
(Part A: Granzyme (Isoleucine-Glutamate-Proline-Aspartate-paraNitroanalide). GrzB (a
Assay) serine protease) levels are measured by cleavage of the substrate,
IEPDpNA at the aspartate (D) residue. The released paranitroanalide
(pNA) forms a yellow colored end product within 20 h at 37 °C in a
humidified container.
1. Biomol Standard Preparation:
(a) Standards are prepared from Biomol GrzB units; initial
stock is supplied as 100 U/μL in 50 μL total starting vol-
ume. Begin with a 1:67 dilution for the top (A) standard
by adding 328.3 μL cell lysis buffer containing 2 μg/μL
BSA (BSA preserves the GrzB enzyme activity) to a 5 μL
stock aliquot of Biomol GrzB. This will result in a new
concentration of 1.5 GrzB units/μL (see Note 11).
(b) Prepare an additional seven standard tubes; label “B to H”
(see recipe below).
Volume of Final
Volume diluenta volume
of biomol GrzB (μL) (μL) Final biomol units
5 μL stock 328.3 166.7 30 units (A)
166.7 μL of (A) 166.7 166.7 15 units (B)
166.7 μL of (B) 166.7 166.7 7.5 units (C)
166.7 μL of (C) 166.7 166.7 3.75 units (D)
166.7 μL of (D) 166.7 166.7 1.875 units (E)
166.7 μL of (E) 166.7 166.7 0.938 units (F)
166.7 μL of (F) 166.7 166.7 0.469 units (G)
0 μL 166.7 166.7 0 units (only Cell Lysis Buffer
with 2 μg/μL BSA) = blank (H)
a
Cell lysis buffer w/BSA
(c) When performing the standard serial dilutions, be sure to
vortex each tube (medium speed for ~5 s), resuspend well,
transfer appropriate volume, and change pipette tip after
every volume transfer. Also, minimize bubble formation.
After final volume transfer to standard G, draw up and dis-
card 166.7 μL which will result in the same volume of
standard in all tubes (see Note 12).
2. Pipette 20 μL in duplicate of each standard (A–H) to the
appropriate wells of microtiter plate using a micropipette (see
plate template below). Before use, vortex the tube (medium
speed for ~5 s), resuspend and directly dispense the appropriate
Cytokine and Granzyme B Assessment of Influenza Response 131
1 2 3 4 5 6 7 8 9 10 11 12
A 30 units Unknown samples 1–40 in duplicate
B 15 units
C 7.5 units
D 3.75 units
E 1.875 units
F 0.938 units
G 0.469 units
H 0 units
(b) Be sure to make up enough master mix daily to run all plates
(# of samples + # standards + minimum 20 % extra).
(c) Directly after a batch of master mix is prepared and just
before addition to the plates, thoroughly mix by very gen-
tle inversion of tube to avoid formation of bubbles.
5. Pipette 80 μL of previously prepared master mix to all wells
with a multichannel pipette (see Note 15).
6. Cover each plate with a thermoplastic seal and seal very well to
prevent moisture from entering wells.
7. Incubate for 20 h in a dark humidified chamber at 37 °C
( see Notes 1 and 16).
8. Once the plates have been placed in the incubator, immediately
proceed to the protein assay (Part B).
9. After 20 h incubation, read the plates using a single wavelength
on the plate reader. Measure the absorbency of GrzB at 405 nm
(see Note 17).
132 Janet E. McElhaney and Beth Gentleman
free for standards and begin plating the samples at column 4).
Before use, vortex the tube (medium speed for ~5 s), resus-
pend well, and directly dispense the appropriate volume to the
plate. Also, resuspend between replicates (see Note 13). Keep
samples and microtiter plate cold at all times during the plating
process in same manner as the GrzB assay.
3. Final BCA Standard Preparation:
(a) While the plates, loaded with samples, remain in the fridge,
complete preparation of the standards by adding the appro-
priate volume of BSA to tubes as per the recipe.
(b) When performing the standard dilutions be sure to vortex
each tube (between medium to high speed for ~5 s), resus-
pend well, transfer appropriate volume, and change pipette
tip after every volume transfer. Also, minimize bubble
formation.
4. Unused BSA stock should be stored at 4 °C and is good for 1
week. Pipette 10 μL in triplicate of each standard (A–H) to
the appropriate wells of microtiter plate (columns 1–3).
Before use, vortex the tube (between medium to high speed
for ~5 s), resuspend well, and directly dispense the appropri-
ate volume to the plate. Also, resuspend between replicates
(see Notes 13 and 20).
5. Determine quantity of A + B reagent needed at 200 μL per well,
then dilute A:B at 50:1, and mix by inversion/swirling until the
solution becomes clear green (for a single plate: 22.540 mL
reagent A + 460 μL reagent B → provides 20 % extra).
6. Add 200 μL of the A + B mixture to all wells with a multichannel
pipette (see Note 21).
7. Cover each plate with a thermoplastic seal and incubate in CO2
incubator at 37 °C for 30 min (see Note 22).
8. Wipe bottom of microtiter plate before inserting in plate
reader.
9. Read plate immediately at 560 nm.
10. Determine the concentration of protein in μg/mL using linear
curve fit.
3.7 Determination We use the Bio-Ples Pro color-coded, bead-based multplex assays
of INFγ and IL-10 that are designed to measure multiple cytokines in diverse solu-
Cytokine Levels tions including serum, plasma, and tissue culture supernatants.
IFNg and IL-10 levels are measured in harvested PBMC superna-
tant samples.
4 Notes
References
1. McElhaney JE, Xie D, Hager WD et al (2006) T enhanced CTL response to influenza vaccination
cell responses are better correlates of vaccine pro- in older adults. Vaccine 27:2418–2425
tection in the elderly. J Immunol 176:6333–6339 5. Shahid Z, Kleppinger A, Gentleman B, Falsey
2. Effros RB (2007) Role of T lymphocyte replicative AR et al (2010) Clinical and immunologic pre-
senescence in vaccine efficacy. Vaccine 25:599–604 dictors of influenza illness among vaccinated
3. Gijzen K, Liu WM, Visontai I et al (2010) older adults. Vaccine 28:6145–6151
Standardization and validation of assays deter- 6. Ewen CL, Rong J, Kokaji AI et al (2006)
mining cellular immune responses against influ- Evaluating antigen-specific cytotoxic T lym-
enza. Vaccine 28:3416–3422 phocyte responses by a novel mouse granzyme
4. McElhaney JE, Ewen C, Zhou X et al (2009) B ELISPOT assay. J Immunol Methods 308:
Granzyme B: correlates with protection and 156–166
Chapter 12
Abstract
Autophagy is an essential catabolic process that regulates a diverse array of functions by targeting cellular
components for degradation by lysosomes. Studies in mammalian cells have shown that the regulation of
autophagy is highly complex and optimization of experimental approaches to analyze this process needs to
be developed for each model studied. This chapter provides an overview of two of the most commonly
used ways to monitor autophagy activity in T cell. It involves description of common techniques, namely
Western blot and cell immunostaining, giving specific recommendations for working with T cells and
monitoring macroautophagy. We also discuss the analysis required for correct interpretation of the results
and quantification of macroautophagy activity.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_12, © Springer Science+Business Media New York 2015
143
144 Yair Botbol and Fernando Macian
2 Materials
2.3 Cell Lysis 1. RIPA buffer 1×: 50 mM Tris–HCl pH 7.7, 150 mM NaCl, 1 %
and Western Blot (v/v) NP40, 0.5 % Sodium deoxycholate, 0.1 % SDS. Add
fresh Complete Protease Inhibitor cocktail 1× (Roche), 2 mM
PMSF, 1 mM DTT.
2. 15 % Tris-Glycine SDS-PAGE buffers and solutions and nitro-
cellulose membrane.
3. Blocking buffer: 1× PBS, 0.1 % (v/v) Tween-20, 5 % Non-fat
dry milk.
4. Antibody Incubation Buffer (AIB): 1× PBS, 0.1 % Tween-20,
4 % BSA.
5. Wash buffer (WB): 1× PBS, 0.1 % (v/v) Tween-20.
6. Anti-mouse LC3 (MBL PM036), anti-mouse β-Actin (clone
AC-15 from Abcam).
7. HRP-conjugated secondary antibodies (rabbit and mouse).
3 Methods
3.1 CD4+ T Cell 1. Prepare plate with bound anti-mouse CD3ε using a 10 μg/mL
Isolation Ab solution in sterile 1× PBS. Add 250 μL per well in 24-well
and Activation plate and incubate for 3.5 h at 37 °C in a tissue culture incuba-
tor under 10 % CO2. Wash the wells twice with 1× PBS just
before adding the cells on step 2 (see Note 1).
2. Isolate Lymph nodes and/or spleen from the mouse and
perform CD4+ isolation using Dynabeads® Mouse CD4
(L3T4) and following the manufacturer’s protocol (see Note
2). Bring cells to a final density of 1.25 × 106 cells/mL of TCM.
Add 0.5 μg/mL of anti-mouse CD28 and seed the cells in the
plate with bound antiCD3e prepared in step 1 of
Subheading 3.1 (1 mL per well). Incubate at 37 °C in 10 %-CO2
incubator (see Note 3).
3. After 21 h of culture add ammonium chloride/Leupeptin
solution (NL) to a final concentration of 20 mM and 100 μM,
respectively, for 3 h to half of the wells used for each condition
analyzed (see Note 4).
3.2 Cell Lysate 1. From this step on keep everything on ice. Harvest T cells after
and Western Blot the 3 h treatment with NL, centrifuge 5 min at 300 × g at 4 °C,
and discard the supernatant.
2. Wash cell pellet with 1 mL of ice-cold 1× PBS (for immunos-
taining take an aliquot of these cells and go to Subheading 3.3),
pellet the cells as in step 1, and gently aspirate the
supernatant.
3. Flick the tube to break up the cell pellet and resuspend it in
RIPA buffer. Homogenize by pipetting up and down 5–10
times (see Note 5). Leave the tubes on ice for 30 min to com-
plete lysis. Flick the tubes a couple of times during the lysis.
Freeze at −80 °C for storage or proceed directly to step 4.
4. Centrifuge the crude extract at 4 °C, for 30 min at 20,000 × g.
Transfer the supernatant to a new tube and perform an addi-
tional run of centrifugation. Collect the supernatant.
5. Measure the protein concentration (see Note 6).
Assays of Macroautophagy in T Cells 149
3.3 LC3 Immuno- 1. From step 2 of Subheading 3.2, take an aliquot of cells, pellet
staining in T Cells down (5 min at 300 g) and resuspend in ice-cold 1× PBS at 106
cells/ml.
From here on all steps are performed at room temperature.
2. Load cells into a cytology funnel (~105 cells in 100 μL) attached
to a glass slide.
3. Centrifuge in cytospin for 5 min at 220 × g.
4. Immediately draw a ring in the slide around the cells using a
hydrophobic-ink pen and add fixation buffer. Incubate for
10 min.
5. Wash once with 1× PBS.
6. Add permeabilization buffer and incubate for 12 min.
7. Wash cells with 1× PBS, twice for 10 min each time.
8. Incubate cells with CBB for 45 min to block unspecific binding
of the antibodies.
9. Incubate cells with LC3 antibody dilution (1/750) in CBB for
30 min.
10. Wash the cells in WB, three times for 10 min.
11. Incubate with secondary Ab fluorophore conjugated in CBB
for 45 min.
12. Wash as in step 10.
150 Yair Botbol and Fernando Macian
4 Notes
1. Coat only half of the wells in a plate to keep the other half for
resting condition. For other plate sizes, use the same CD3ε Ab
dilution adjusting the volume in proportion to the surface area
(1 well of 24-well plate is 1.9 cm2).
Assays of Macroautophagy in T Cells 151
per gel and fixed Voltage max at 125 V. The voltage at the
beginning of the run should be around 70 V and then will
increase up to the max setup.
8. Transfer at no more than 150 mA per transfer cell (mini size of
Protean Biorad) for at least 3 h, rather than using higher
current and shorter times. This will avoid excessive heating of
the transfer buffer, producing a cleaner membrane and a more
reproducible blot.
9. Increasing LC3-Ab binding time does not usually improve
specific signal, but will increase the appearance of nonspecific
bands.
10. Follow ImageJ commands for gel densitometry. Select both
LC3-I and LC3-II bands. Even if macroautophagy flux quan-
tification considers only LC3-II accumulation, this allows to
precisely quantifying background around LC3 bands by
measuring pixel intensity between the two peaks. We have
observed that the level of LC3-II protein in the absence of
lysosome inhibitors is very low in both resting and activated
T cells, which suggest a very fast rate of macroautophagy
activity in T cells. However, with lysosome inhibitors the bands
become very intense, especially in activated cells, where macro-
autophagy is highly induced. In any case, if the signal is still
low, increase the quantity of protein loaded on gel to 12–15 μg
per lane (no more than 15 μg is recommended to avoid satura-
tion of LC3-II signal in activated cells).
11. We have checked protein level as well as mRNA level of β-Actin
and we did not observe any differences in any of the conditions
used to study macroautophagy in T cells so far, making this
protein a good housekeeping gene.
12. The ratio of the LC3-II levels between samples treated with or
without inhibitors has also been used and it provides a mea-
surement of the “speed of the process”; however subtraction
will quantify the real net change in total levels of LC3-II.
Although initially ratio of LC3-II to LC3-I was widely used, it
has become clear that the amount of LC3 primed for conjuga-
tion (LC3-I) is very cell type dependent and this ratio is not
indicative of macroautophagy activity. Furthermore, differ-
ences in the affinity of different anti-LC3 antibodies for LC3-I
and LC3-II may lead to inaccurate estimations of the real rate
of conversion of LC3-I into LC3-II.
13. This process can be done manually or using an image analysis
software such as ImageJ. The particle-counting’s functions of
ImageJ must be used very carefully and always with a shrewd
eye since many parameters still need to be defined manually
(threshold, particles size, etc.). We found that ImageJ
MaxEntropy auto-threshold method appeared to be a good
Assays of Macroautophagy in T Cells 153
Acknowledgments
References
1. Cuervo AM (2008) Autophagy and aging: 9. Bell BD, Leverrier S, Weist BM, Newton RH,
keeping that old broom working. Trends Arechiga AF, Luhrs KA et al (2008) FADD
Genet 24:604–612 and caspase-8 control the outcome of autoph-
2. Mizushima N (2009) Physiological functions agic signaling in proliferating T cells. Proc Natl
of autophagy. Curr Top Microbiol Immunol Acad Sci U S A 105:16677–16682
335:71–84 10. Pua HH, Dzhagalov I, Chuck M, Mizushima
3. Sridhar S, Botbol Y, Macian F, Cuervo AM N, He YW (2007) A critical role for the
(2012) Autophagy and disease: always two autophagy gene Atg5 in T cell survival and
sides to a problem. J Pathol 226:255–273 proliferation. J Exp Med 204:25–31
4. Deretic V (2012) Autophagy as an innate 11. Li C, Capan E, Zhao Y, Zhao J, Stolz D,
immunity paradigm: expanding the scope and Watkins SC et al (2006) Autophagy is induced
repertoire of pattern recognition receptors. in CD4+ T cells and important for the growth
Curr Opin Immunol 24:21–31 factor-withdrawal cell death. J Immunol 177:
5. Gannage M, Munz C (2009) Autophagy in MHC 5163–5168
class II presentation of endogenous antigens. Curr 12. Klionsky DJ, Abdalla FC, Abeliovich H,
Top Microbiol Immunol 335:123–140 Abraham RT, Acevedo-Arozena A, Adeli K
6. Maue AC, Yager EJ, Swain SL, Woodland DL, et al (2012) Guidelines for the use and inter-
Blackman MA, Haynes L (2009) T-cell immu- pretation of assays for monitoring autophagy.
nosenescence: lessons learned from mouse mod- Autophagy 8:445–544
els of aging. Trends Immunol 30:301–305 13. Mizushima N, Yoshimori T, Ohsumi Y (2011)
7. Phadwal K, Alegre-Abarrategui J, Watson AS, The role of Atg proteins in autophagosome
Pike L, Anbalagan S, Hammond EM et al formation. Annu Rev Cell Dev Biol 27:
(2012) A novel method for autophagy detec- 107–132
tion in primary cells: impaired levels of macro- 14. Mizushima N, Yoshimori T, Levine B (2010)
autophagy in immunosenescent T cells. Methods in mammalian autophagy research.
Autophagy 8:677–689 Cell 140:313–326
8. Hubbard VM, Valdor R, Patel B, Singh R, 15. Tanida I, Minematsu-Ikeguchi N, Ueno T,
Cuervo AM, Macian F (2010) Macroautophagy Kominami E (2005) Lysosomal turnover, but
regulates energy metabolism during effector not a cellular level, of endogenous LC3 is a
T cell activation. J Immunol 185:7349–7357 marker for autophagy. Autophagy 1:84–91
Chapter 13
Abstract
Protein oxidation and misfolding have been considered as key players for progression of aging and etiology
of various pathological conditions. However, few attempts have been made to develop sensitive and repro-
ducible assays to quantify the changes in protein oxidation and alteration in structure. Here we describe
three distinct fluorescence-based assays to quantify changes in protein oxidation, namely carbonylation and
disulfides and alteration in protein surface hydrophobicity as a reporter for protein conformation. These
techniques will provide investigators the opportunity to address important biological questions in their
experimental models.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_13, © Springer Science+Business Media New York 2015
155
156 Asish R. Chaudhuri et al.
2 Materials
2.4 Disulfide Assay 1. 10× KP buffer, pH 8.0: 200 mM potassium phosphate (KP),
Components 5 mM magnesium chloride (MgCl2), and 10 mM
ethylenediamine-tetraacetic acid (EDTA), pH 8.0. First, pre-
pare 50 mL stocks of 200 mM KP monobasic and dibasic. Add
1.36 g of KP monobasic, add up to 50 mL water, and mix. Add
1.74 g of KP dibasic, add up to 50 mL water, and mix. Next,
prepare a 50 mL stock of 10× KP buffer, pH 8.0. Add 0.24 g
of MgCl2, 0.146 g of EDTA, 47 mL of KP dibasic, and 3 mL
KP of monobasic (see Note 18). Add up to 50 mL water and
mix (the 10× KP buffer should be pH 8.0). Store at room
temperature.
2. 1× KP buffer, pH 8.0: 20 mM KP, 0.5 mM MgCl2, and
1 mM EDTA, pH 8.0 with pi. To make 50 mL stock, mix
5 mL of 10× KP buffer, pH 8.0 and 45 mL water. Store at
room temperature if protease inhibitors have not yet been
added (see Note 12).
3. 1× KP buffer, pH 8.0 with 200 mM iodoacetamide (IAM) and
pi: 20 mM KP, 0.5 mM MgCl2, and 1 mM EDTA, pH 8.0
with pi. Add IAM, dilute 10× KP buffer, pH 8.0 to 1× with
water, and mix. Store at room temperature if IAM and pi have
not yet been added (see Notes 12 and 19).
4. 1× P3 buffer, pH 8.0: 1× KP buffer, pH 8.0 with 2 % SDS,
0.5 % IGEPAL CA-630, 0.5 % Sodium Deoxycholate, and pi
(see Note 12). Prepare a 50 mL stock by adding 1 g of SDS,
750 μL of IGEPAL CA-630, 0.75 g of Sodium Deoxycholate,
and 5 mL of 10× KP buffer, pH 8.0. Add up to 50 mL of water
and mix. Store at room temperature if pi has not yet been
added (see Note 12).
5. 1× P3 buffer, pH 8.0 with 200 mM IAM and pi (see Notes 12
and 19).
6. 6 and 8 M Urea dissolved in water (see Note 20).
7. 20 % trichloroacetic acid (TCA) dissolved in water.
8. 1:1 ethanol/ethyl acetate: Prepare 50 mL of 1:1 ethanol/ethyl
acetate mixture by adding 25 mL of 200 proof ethanol and
25 mL ethyl acetate.
160 Asish R. Chaudhuri et al.
2.5 Carbonyl Assay 1. 10× KP buffer, pH 6.0: 200 mM potassium phosphate (KP),
Components 5 mM magnesium chloride (MgCl2), and 10 mM
ethylenediamine-tetraacetic acid (EDTA), pH 8.0. First, pre-
pare 50 mL stocks of 200 mM KP monobasic and dibasic. Add
1.36 g of KP monobasic, add up to 50 mL water, and mix. Add
1.74 g of KP dibasic, add up to 50 mL water, and mix. Next,
prepare a 50 mL stock of 10× KP buffer, pH 6.0. Add 0.24 g
of MgCl2, 0.146 g of EDTA, 6.6 mL of KP dibasic, and
43.4 mL KP of monobasic (see Note 18). Add up to 50 mL
water and mix (the 10× KP buffer should be pH 6.0). Store at
room temperature.
2. 1× KP buffer, pH 6.0: 20 mM KP, 0.5 mM MgCl2, and 1 mM
EDTA, pH 6.0 with pi. To make 50 mL stock, mix 5 mL of
10× KP buffer, pH 6.0 and 45 mL water. Store at room tem-
perature if pi has not yet been added (see Note 12).
3. 1× P3 buffer, pH 6.0: 1× KP buffer, pH 6.0 with 2 % SDS,
0.5 % IGEPAL CA-630, 0.5 % Sodium Deoxycholate, and pi
(see Note 12). Prepare a 50 mL stock by adding 1 g of SDS,
750 μL of IGEPAL CA-630, 0.75 g of Sodium Deoxycholate,
and 5 mL of 10× KP buffer, pH 6.0. Add up to 50 mL of water
and mix. Store at room temperature if pi has not yet been
added (see Note 12).
4. Dithiothreitol (DTT): Prepare a 1 mL stock of 200 mM
DTT. Dissolve 0.031 g of DTT, add up to 1 mL of water, and
mix. Store at 4 °C.
5. Fluorescein-5-thiosemicarbazide (FTC) fluorescent probe
(Invitrogen catalog number F-121): Prepare 100 mM aliquots
by adding 2.37 mL DMSO to the bottle and mix. Cover ali-
quots with foil and store at −20 °C (see Note 15).
6. Guanidine hydrochloride: Prepare 1 mL stock of 3 M guani-
dine hydrochloride. Add 0.287 g of guanidine hydrochloride,
add up to 1 mL with water, then mix.
7. 20 % trichloroacetic acid (TCA) dissolved in water.
Quantitative Assessment of Protein Oxidation 161
3 Methods
3.1 12 % Sodium All steps are performed at room temperature unless indicated
Dodecyl Sulfate otherwise.
Polyacrylamide Gel
1. Remove well comb from empty criterion cassette, and place
Electrophoresis cassette on a rack to keep it vertical.
(SDS-PAGE)
2. Prepare the 12 % running gel by mixing 2.5 mL of resolving
gel buffer, 5 mL of 30 % acrylamide/Bis solution, 125 μL of
10 % SDS, and 4.8 mL of water. Add 6 μL of TEMED and
63 μL of 10 % APS, then mix again (see Note 21).
3. Quickly fill empty criterion cassette with running gel mixture
up to 2–3 mm below the bottom of the well insert.
4. Gently layer with 2 mL isopropanol (see Note 22).
5. Let set for 1 h to allow for full gel polymerization.
6. Rinse isopropanol out of the cassette with water, and briefly
dry (see Note 23).
7. Prepare the 4 % stacking gel by mixing 833 μL of stacking gel
buffer, 444 μL of 30 % acrylamide/Bis solution, 33 μL of 10 %
SDS, and 2 mL of water. Add 3 μL of TEMED and 33 μL of
10 % APS, then mix again (see Note 21).
8. Quickly fill criterion cassette with stacking gel mixture to the
top (see Note 24).
9. Carefully place well comb into the criterion cassette (see
Note 25).
10. Let set for 40 min to allow for full gel polymerization.
11. Prepare samples to run in SDS-PAGE by adding appropriate
volume of 10× loading dye and mix (see Note 6).
12. Warm samples to 37 °C for 5 min.
13. Spin down samples at 1000 × g for 30 s to bring down any con-
densate or sample stuck on the walls.
14. Remove sticker strip from the bottom of the criterion cassette
(see Note 26).
15. Place criterion cassette into criterion cell.
16. Fill criterion cell with 1× Tris/glycine/SDS running buffer up
to marked fill line.
162 Asish R. Chaudhuri et al.
17. Add enough Bio-safe Coomassie stain to cover gel (see Note 17).
This will correct for protein loading.
18. Wrap plastic container with plastic wrap and let gel stain over-
night with gentle rocking.
19. Wash gel 3× in water for 5 min. There should be noticeable
blue bands visible in the gel.
20. Capture Coomassie image under visible light.
21. Analyze all images using Unscanit software (see Note 34).
22. Figure 1 represents the use of the BisANS assay in two distinct
mouse models of oxidative stress; (A) Sod1−/− and (B) f-ALS.
Fig. 1 Alteration in global protein conformation in mouse models of in vivo oxidative stress and f-ALS. BisANS
UV crosslinked skeletal muscle proteins were analyzed by SDS-PAGE in (a) 20-mo-old wild-type (WT) and
Sod1−/− mice and (b) SOD1G93A and SOD1H46R/H48Q mouse models of human f-ALS. Results are expressed as
mean ± standard error of the mean of 4–5 mice/group and analyzed by two-tailed t-test (**p < 0.01,
***p < 0.001). Panel (a) demonstrates a 22 ± 3 % reduction in BisANS labeling in old Sod1−/− mice compared
with old WT controls. Panel (b) demonstrates a 24 ± 2 % reduction in BisANS labeling in SOD1G93A mice com-
pared to the longer-lived SOD1H46R/H48Q mice. Previously, we reported that creatinine kinase (43 kDa) and glyc-
eraldehyde 3-phosphate dehydrogenase (37 kDa) are affected conformationally in response to oxidative stress
[20–22] and these proteins were also affected in the f-ALS and Sod1−/− mouse models as depicted in (a) and
(b) by (1) and (2), respectively
164 Asish R. Chaudhuri et al.
47. Add enough Bio-safe Coomassie stain to cover gel (see Note 17).
This will correct for protein loading.
48. Wrap plastic container with plastic wrap and let gel stain
overnight while gently rocking.
49. Wash gel 3× in water for 5 min. There should be noticeable
blue bands visible in the gel.
50. Capture Coomassie image under visible light.
51. Analyze all images using Unscanit software (see Note 33).
52. Figure 2 represents the use of the disulfide assay in f-ALS
mouse model.
53. Quantification for disulfides was performed utilizing Unscanit
software as described previously for BisANS (see Note 33).
4 Notes
21. Always add TEMED and APS last to the mixture, as this will
start the gel polymerization process.
22. It is normal to observe a slight rolling effect at the border of
the running gel mixture and isopropanol.
23. To dry, invert the cassette onto paper towels and lightly tap to
remove excess water.
24. Try to add stacking gel mixture carefully and avoid bubbles.
25. We try to push any bubbles to the side as we slide the well
comb in. If bubbles remain in the stacking gel mixture, sam-
ples will not run properly.
26. Failure to remove sticker strip will prevent current flow and
samples will not run down gel.
27. We completely fill the top buffer reservoir. It is crucial to have
the wells covered in 1× Tris/glycine/SDS buffer. Failure to do
so will prevent current flow and samples will not run down gel.
28. Stacking gel is fragile and can break off or bend if well comb
removal is not careful.
29. We pipette samples starting from the bottom of the well and
slowly raising pipette tip as well is filled with sample.
30. The dye front generally runs faster than the smallest molecular
weight band of the precision plus kaleidoscope protein stan-
dards, so is easy to spot when to stop electrophoresis.
31. We generally try to prepare reaction mixtures of 100 μg pro-
tein in 100 μL total volume in 1× BisANS buffer plus protease
inhibitors. A lower total volume will be difficult to uniformly
cover the entire well surface of the 96-well plate.
32. It is very important to keep the plate on ice to avoid sample
evaporation, as the plate will heat up over time while the UV
light is on.
33. Unscanit software utilizes the area under the curve for given
bands and allows you to calculate the areas under the curve for
each band which can be summed after analysis for all bands on
the gel. For all analysis, the total BisANS, disulfides, or carbon-
yls were measured as an area under the curve in arbitrary units
for all bands and normalized to the area under the curve for
the total protein loaded on the gels as measured by coomassie.
Values were converted to fold change relative to the wild-type
animals. Arrows located in Fig. 1 indicate two proteins that we
observed in our 2006 manuscript which confirmed the pro-
teins that were misfolded by mass spectrometry analysis [20].
Any alternative image analysis software will work for the
analysis.
34. For sonicating volumes of 50–100 μL, we suggest using
0.2 mL thick wall ultracentrifuge tubes.
172 Asish R. Chaudhuri et al.
35. SDS precipitates at 4 °C; make sure that all samples containing
P3 buffer are worked with at room temperature.
36. We generally try to prepare reaction mixtures of at least 200 μg
protein in 200 μL total volume if limited on sample. Ethanol/
ethyl acetate wash steps lose a lot of protein; use at maximum
500 μg protein in 500 μL total volume.
37. Add an equal volume of 20 % TCA as the total volume of
reaction mixture (e.g., 200 μL TCA to 200 μL total volume
reaction mixture).
38. Reaction mixture becoming cloudy or speckled is a good indi-
cation of protein precipitation. If working with very low
amounts of protein, TCA precipitation time can be increased.
39. Several washes are required to get rid of excess TCA and free
fluorescent probe. Failure to do so will cause the gel fluores-
cence to look streaky and lack distinguished band patterning.
40. Fluorescence imaging will also work with other imagers with
similar excitation/emission/filter capabilities.
41. The addition of guanidine hydrochloride will make pellet sam-
ples difficult to work with, so make sure to add it last to the
reaction mixture.
References
1. Christians ES, Benjamin IJ (2012) Proteostasis 7. Curtis JM, Hahn WS, Long EK, Burrill JS,
and REDOX state in the heart. Am J Physiol Arriaga EA, Bernlohr DA (2012) Protein car-
Heart Circ Physiol 302:H24–H37 bonylation and metabolic control systems.
2. Bokov A, Chaudhuri A, Richardson A (2004) Trends Endocrinol Metab 23:399–406
The role of oxidative damage and stress in 8. Bhattacharya A, Leonard S, Tardif S,
aging. Mech Ageing Dev 125:811–826 Buffenstein R, Fischer KE, Richardson A,
3. Butterfield DA, Kanski J (2001) Brain protein Austad SN, Chaudhuri AR (2011) Attenuation
oxidation in age-related neurodegenerative of liver insoluble protein carbonyls: indicator
disorders that are associated with aggregated of a longevity determinant? Aging Cell 10:
proteins. Mech Ageing Dev 122:945–962 720–723
4. Olivares-Corichi IM, Ceballos G, Medina- 9. Uchida K (2003) Histidine and lysine as tar-
Santillan R, Medina-Navarro R, Guzman- gets of oxidative modification. Amino Acids
Grenfell AM, Hicks JJ (2005) Oxidation by 25:249–257
reactive oxygen species (ROS) alters the struc- 10. Amici A, Levine RL, Tsai L, Stadtman ER
ture of human insulin and decreases the (1989) Conversion of amino acid residues in
insulin-dependent D-glucose-C14 utilization proteins and amino acid homopolymers to car-
by human adipose tissue. Front Biosci bonyl derivatives by metal-catalyzed oxidation
10:3127–3131 reactions. J Biol Chem 264:3341–3346
5. Uchida K, Toyokuni S, Nishikawa K, Kawakishi 11. Doorn JA, Petersen DR (2002) Covalent
S, Oda H, Hiai H, Stadtman ER (1994) modification of amino acid nucleophiles by the
Michael addition-type 4-hydroxy-2-nonenal lipid peroxidation products 4-hydroxy-2-non-
adducts in modified low-density lipoproteins: enal and 4-oxo-2-nonenal. Chem Res Toxicol
markers for atherosclerosis. Biochemistry 15:1445–1450
33:12487–12494 12. Berlett BS, Stadtman ER (1997) Protein oxi-
6. Nakamura A, Kawakami K, Kametani F, dation in aging, disease, and oxidative stress.
Nakamoto H, Goto S (2010) Biological J Biol Chem 272:20313–20316
significance of protein modifications in aging 13. Huggins TG, Wells-Knecht MC, Detorie NA,
and calorie restriction. Ann N Y Acad Sci Baynes JW, Thorpe SR (1993) Formation of
1197:33–39 o-tyrosine and dityrosine in proteins during
Quantitative Assessment of Protein Oxidation 173
radiolytic and metal-catalyzed oxidation. J Biol Detection and quantification of protein disul-
Chem 268:12341–12347 fides in biological tissues a fluorescence-based
14. Thomas JA, Mallis RJ (2001) Aging and proteomic approach. Methods Enzymol 473:
oxidation of reactive protein sulfhydryls. Exp 161–177
Gerontol 36:1519–1526 20. Pierce A, deWaal E, Van Remmen H, Richardson
15. Zhou JQ, Gafni A (1991) Exposure of rat A, Chaudhuri A (2006) A novel approach for
muscle phosphoglycerate kinase to a nonenzy- screening the proteome for changes in protein
matic MFO system generates the old form of conformation. Biochemistry 45:3077–3085
the enzyme. J Gerontol 46:B217–B221 21. Pierce A, Mirzaei H, Muller F, De Waal E,
16. Chaudhuri AR, Khan IA, Luduena RF (2001) Taylor AB, Leonard S, Van Remmen H,
Detection of disulfide bonds in bovine brain Regnier F, Richardson A, Chaudhuri A (2008)
tubulin and their role in protein folding and GAPDH is conformationally and functionally
microtubule assembly in vitro: a novel disul- altered in association with oxidative stress in
fide detection approach. Biochemistry 40: mouse models of amyotrophic lateral sclerosis.
8834–8841 J Mol Biol 382:1195–1210
17. Chaudhuri AR, de Waal EM, Pierce A, Van 22. Pierce AP, de Waal E, McManus LM, Shireman
Remmen H, Ward WF, Richardson A (2006) PK, Chaudhuri AR (2007) Oxidation and
Detection of protein carbonyls in aging liver structural perturbation of redox-sensitive
tissue: a fluorescence-based proteomic enzymes in injured skeletal muscle. Free Radic
approach. Mech Ageing Dev 127:849–861 Biol Med 43:1584–1593
18. Salmon AB, Leonard S, Masamsetti V, Pierce 23. Wei R, Bhattacharya A, Chintalaramulu N,
A, Podlutsky AJ, Podlutskaya N, Richardson Jernigan AL, Liu Y, Van Remmen H,
A, Austad SN, Chaudhuri AR (2009) The long Chaudhuri AR (2012) Protein misfolding,
lifespan of two bat species is correlated with mitochondrial dysfunction and muscle loss are
resistance to protein oxidation and enhanced not directly dependent on soluble and aggre-
protein homeostasis. FASEB J 23:2317–2326 gation state of mSOD1 protein in skeletal
19. Perez VI, Pierce A, de Waal EM, Ward WF, muscle of ALS. Biochem Biophys Res
Bokov A, Chaudhuri A, Richardson A (2010) Commun 417:1275–1279
Chapter 14
Abstract
Telomeres are repetitive DNA repeats that cap the ends of all eukaryotic chromosomes. Their proper
maintenance is essential for genomic stability and cellular viability. Dysfunctional telomeres could arise
through natural attrition of telomeric DNA or due to the removal of shelterin components. These
uncapped chromosomal ends are recognized as DSBs by the DDR pathway, leading to the accumulation
of DNA damage sensors at telomeres. The association of these DDR proteins with dysfunctional telo-
meres forms telomere dysfunction induced DNA damage foci (TIFs). Detection of TIFs at telomeres
provides an opportunity to quantify the extent of telomere dysfunction and monitor downstream DNA
damage signaling pathways. Here we describe a method for the detection of TIFs using a fluorescent in situ
hybridization (FISH) approach.
Key words DNA damage, Telomere dysfunction, Telomere induced foci, Telomere FISH
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_14, © Springer Science+Business Media New York 2015
175
176 Rekha Rai and Sandy Chang
2 Materials
2.2 Retroviral 1. pCL Eco, Empty pBabe, p-Babe shTRF2, Empty pQCXIP,
Production pQCXIP TPP1ΔRD [8].
and Infection 2. Polybrene Stock 6 mg/ml. Filter through a 0.2 μm filter, ali-
quot, and store at −20 °C.
3. Lipofectamine Plus Reagent (Invitrogen), Fugene 6 (Roche).
4. 0.45 μm syringe filters (Millipore).
2.3 Immunofluo- 1. Nunc Lab-Tek 8 well slide chambers (Nalgene Nunc, 12-565-1),
rescence-Telomere Microscope coverslips 18 × 18-1 (Fisher-12-548-A), Microscope
FISH Slides (Fisher 12-544-3).
Detection of Telomere Dysfunction 177
3 Methods
3.1 Retrovirus 1. Day 0: On the day before transfection, plate 0.5–1 ×1 06 293T
Production in 293T cells into a 6 cm tissue culture dish in 4.0 ml DMEM supple-
Cells mented with 10 % FBS (see Note 1). The cells are ready for
transfection after 18–20 h, or when they are about 60–70 %
confluent. Plate slightly more cells when making VSV-G pseu-
dotyped viruses.
2. Day 1: Aspirate the medium and replace the cells with 2.0 ml
DMEM without FBS or any antibiotic. Transfect 293T cells
with 2.0 μg pCL-ECO and 4.0 μg of transfer vectors (p-Babe
shTRF2, pQXCIP TPP1ΔRD) using Lipofectamine Plus Reagent
following the manufacturer’s protocol. For making VSV-G
retroviruses by triple transfection, use 0.9 μg Gag/Pol expres-
sion vector, 0.1 μg VSV-G expression vector, and 1.0 μg trans-
fer vector. For making VSV-G pseudotype viruses, use Fugene
6 following the manufacturer’s protocol.
3. Change the medium 5–7 h after transfection with 4.0 ml of
fresh medium (see Note 2).
4. Day 3: Harvest the viral supernatants at 48 h post-transfection.
Filter the viral supernatant with a 0.45 μm syringe filter. Add
10 ml of fresh DMEM medium supplemented with 10 % FBS
to the cells.
5. Day 4: Harvest the viral supernatant at 72 h of post-infection
as above.
3.3 Immunofluo- 1. Day 6: Aspirate the medium and wash the cells twice for 5 min
rescence-Telomere each with 1× PBS at room temperature (RT).
FISH 2. Fix the cells with 2 % paraformaldehyde/2 % sucrose for 10 min
at RT.
3. Wash the fixed cells twice for 5 min each with 1× PBS.
4. Permeabilize the cells with 0.5 % Nonidet-P40 for 10 min at RT.
5. Wash the cells thrice for 5 min with 1× PBS.
6. Incubate the cells with PBG for 1 h to block nonspecific
binding.
Detection of Telomere Dysfunction 179
H2O 2.85 μl
2 % BSA 1.5 μl
100 μg/ml tRNA 0.15 μl
0.6× SSC 3.0 μl
100 % Formamide 21.0 μl
10 ng/μl PNA Probe 1.5 μl
Total 30.0 μl
14. Denature the slide at 85 °C on a hot plate for 3 min; place the
slide in the dark overnight in a humidified chamber (see Note 8).
15. Day 8: Wash twice for 15 min each in Wash Solution I. For
slides use a Coplin jar, for coverslips use a 6-well plate.
16. Wash thrice for 5 min each in Wash Solution II.
17. Ethanol dehydrate the slides for 2 min each with 70, 85, and
95 % Ethanol.
18. Counterstain with DAPI and seal with nail vanish. The cover-
slip should be carefully inverted to a drop of mounting medium
on a microscope slide. For 8-well chambers, carefully remove
the gasket and place a few drops of mounting medium and
cover with coverslips (see Note 9). The slides can be viewed
immediately when the varnish is dried or can be stored in the
dark at 4 °C for up to a month.
19. Visualize and image under a fluorescent microscope.
180 Rekha Rai and Sandy Chang
4 Notes
References
1. Cosme-Blanco W, Shen MF, Lazar A et al end-joining pathways in the fusion of dysfunc-
(2007) Telomere dysfunction suppresses spon- tional telomeres. EMBO J 29:2598–2610
taneous tumorigenesis in vivo by activating 6. d'Adda di Fagagna F, Reaper PM, Clay-Farrace
p53-mediated cellular senescence. EMBO Rep L et al (2003) A DNA damage checkpoint
8:497–503 response in telomere initiated senescence.
2. de Lange T (2005) Shelterin: the protein com- Nature 426:194–198
plex that shapes and safeguards human telo- 7. Takai H, Smogorzewska A, de Lange T (2003)
meres. Genes Dev 19:2100–2110 DNA damage foci at dysfunctional telomeres.
3. Deng Y, Chan SS, Chang S (2008) Telomere Curr Biol 13:1549–1556
dysfunction and tumour suppression: the senes- 8. Liu D, Safari A, O’Connor MS et al (2004) PTOP
cence connection. Nat Rev Cancer 8:450–458 interacts with POT1 and regulates its localization
4. Deng Y, Guo X, Ferguson DO et al (2009) to telomeres. Nat Cell Biol 6:673–680
Multiple roles for MRE11 at uncapped telo- 9. Guo X, Deng Y, Lin Y et al (2007) Dysfunctional
meres. Nature 460:914–918 telomeres activate an ATM-ATR-dependent
5. Rai R, Zheng H, He H et al (2010) The func- DNA damage response to suppress tumorigenesis.
tion of classical and alternative non-homologous EMBO J 26:4709–4719
Chapter 15
Abstract
The unbiased, paired analysis of T-cell receptor (TCR) α- and β-chain usage at the single-cell level provides
a valuable window of understanding into the TCR repertoire and the nature of the immune response.
Earlier technologies for TCR repertoire analysis were often limited to examining TCR complementarity-
determining region 3 (CDR3) β expression or required in vitro cloning procedures that can artificially
skew the TCR repertoire from its in vivo state. We describe here a direct ex vivo, single-cell-based strategy
for the clonotypic analysis of TCRαβ repertoires that utilizes multiplexed panels of TCRα and TCRβ-
specific primers in a nested PCR to amplify expressed transcripts from individual, epitope-specific T cells.
This strategy yields the paired TCRαβ sequences of any given population of αβ T cells of interest.
Key words T-cell receptor repertoire, Complementarity-determining region 3, TCR alpha, TCR
beta, TCR diversity, Single-cell analysis, Unbiased repertoire, Paired analysis, Clonotype, Epitope
1 Introduction
181
182 Pradyot Dash et al.
2 Materials
2.1 Cell Sorting 1. Human peripheral blood mononuclear cells (PBMC): Fresh
Reagents or frozen PBMC isolated by density gradient (e.g., GE
and Instrument Healthcare Ficoll-Paque PLUS) centrifugation from peripher-
ally withdrawn venous whole blood collected into sodium
heparin-coated collection tubes. Frozen PBMC should be
cryopreserved in liquid nitrogen.
2. Fluorochrome-conjugated monoclonal antibodies and tetra-
mer: The following list of reagents is used for the isolation
Single-Cell Analysis of Human αβT Cell Repertoire 183
2.2 RT-PCR and Gel 1. Multichannel pipette and tips: 12-channel (5–50 μl) and
Electrophoresis 8-channel (0.5–10 μl) pipettes, 10, 20, and 200 μl DNase-
and RNase-free barrier tips.
2. Individually wrapped sterile reservoir boats.
3. Reverse transcription kit: iScript cDNA Synthesis Kit (Bio-
Rad) containing 5× iScript reaction mix and iScript reverse
transcriptase.
4. Triton X-100, molecular biology grade (Sigma).
5. PCR kit: Taq polymerase-based PCR kit (Qiagen) with 10×
PCR buffer containing 15 mM MgCl2, CoralLoad buffer
(10×) containing 15 mM MgCl2, 10 mM dNTP and Taq
DNA polymerase (5 U/μl).
6. Oligonucleotide primers (Table 1) (see Note 1):
Stock dilution: Resuspend primers to 200 μM using 1× TE
buffer (low EDTA; 10 mM Tris–HCl, 0.1 mM EDTA, pH 8.0;
USB). Store the stock dilutions in −20 °C.
184 Pradyot Dash et al.
Table 1
Primers targeting T-cell receptor alpha (TRA) and beta (TRB) genes
TRA gene(s)
targeted by
primer External primer sequence (EXT) Internal primer sequence (INT)
TRAV1 5′ AACTGCACGTACCAGACATC 3′ 5′ GCACCCACATTTCTKTCTTAC 3′
TRAV2 5′ GATGTGCACCAAGACTCC 3′ 5′ CACTCTGTGTCCAATGCTTAC 3′
TRAV3 5′ AAGATCAGGTCAACGTTGC 3′ 5′ ATGCACCTATTCAGTCTCTGG 3′
TRAV4 5′ CTCCATGGACTCATATGAAGG 3′ 5′ ATTATATCACGTGGTACCAACAG 3′
TRAV5 5′ CTTTTCCTGAGTGTCCGAG 3′ 5′ TACACAGACAGCTCCTCCAC 3′
TRAV6 5′ CACCCTGACCTGCAACTATAC 3′ 5′ TGGTACCGACAAGATCCAG 3′
TRAV7 5′ AGCTGCACGTACTCTGTCAG 3′ 5′ ACAATTTGCAGTGGTACAGG 3′
TRAV8-1 5′ CTCACTGGAGTTGGGATG 3′ 5′ GTCAACACCTTCAGCTTCTC 3′
TRAV8-2, 8-4 5′ GCCACCCTGGTTAAAGG 3′ 5′ AGAGTGAAACCTCCTTCCAC 3′
TRAV8-3 5′ CACTGTCTCTGAAGGAGCC 3′ 5′ TTTGAGGCTGAATTTAAGAGG 3′
TRAV8-6 5′ GAGCTGAGGTGCAACTACTC 3′ 5′ AACCAAGGACTCCAGCTTC 3′
TRAV8-7 5′ CTAACAGAGGCCACCCAG 3′ 5′ ATCAGAGGTTTTGAGGCTG 3′
TRAV9-1, 9-2 5′ TGGTATGTCCAATATCCTGG 3′ 5′ GAAACCACTTCTTTCCACTTG 3′
TRAV10 5′ CAAGTGGAGCAGAGTCCTC 3′ 5′ GAAAGAACTGCACTCTTCAATG 3′
TRAV12-1, 5′ CARTGTTCCAGAGGGAGC 3′ 5′ AAGATGGAAGGTTTACAGCAC 3′
12-2, 12-3
TRAV13-1 5′ CATCCTTCAACCCTGAGTG 3′ 5′ TCAGACAGTGCCTCAAACTAC 3′
TRAV13-2 5′ CAGCGCCTCAGACTACTTC 3′ 5′ CAGTGAAACATCTCTCTCTGC 3′
TRAV14 5′ AAGATAACTCAAACCCAACCAG 3′ 5′ AGGCTGTGACTCTGGACTG 3′
TRAV16 5′ AGTGGAGCTGAAGTGCAAC 3′ 5′ GTCCAGTACTCCAGACAACG 3′
TRAV17 5′ GGAGAAGAGGATCCTCAGG 3′ 5′ CCACCATGAACTGCAGTTAC 3′
TRAV18 5′ TCCAGTATCTAAACAAAGAGCC 3′ 5′ TGACAGTTCCTTCCACCTG 3′
TRAV19 5′ AGGTAACTCAAGCGCAGAC 3′ 5′ TGTGACCTTGGACTGTGTG 3′
TRAV20 5′ CACAGTCAGCGGTTTAAGAG 3′ 5′ TCTGGTATAGGCAAGATCCTG 3′
TRAV21 5′ TTCCTGCAGCTCTGAGTG 3′ 5′ AACTTGGTTCTCAACTGCAG 3′
TRAV22 5′ GTCCTCCAGACCTGATTCTC 3′ 5′ CTGACTCTGTGAACAATTTGC 3′
TRAV23 5′ TGCTTATGAGAACACTGCG 3′ 5′ TGCATTATTGATAGCCATACG 3′
TRAV24 5′ CTCAGTCACTGCATGTTCAG 3′ 5′ TGCCTTACACTGGTACAGATG 3′
TRAV25 5′ GGACTTCACCACGTACTGC 3′ 5′ TATAAGCAAAGGCCTGGTG 3′
(continued)
Single-Cell Analysis of Human αβT Cell Repertoire 185
Table 1
(continued)
TRA gene(s)
targeted by
primer External primer sequence (EXT) Internal primer sequence (INT)
TRAV26-1 5′ GCAAACCTGCCTTGTAATC 3′ 5′ CGACAGATTCACTCCCAG 3′
TRAV26-2 5′ AGCCAAATTCAATGGAGAG 3′ 5′ TTCACTTGCCTTGTAACCAC 3′
TRAV27 5′ TCAGTTTCTAAGCATCCAAGAG 3′ 5′ CTCACTGTGTACTGCAACTCC 3′
TRAV29 5′ GCAAGTTAAGCAAAATTCACC 3′ 5′ CTGCTGAAGGTCCTACATTC 3′
TRAV30 5′ CAACAACCAGTGCAGAGTC 3′ 5′ AGAAGCATGGTGAAGCAC 3′
TRAV34 5′ AGAACTGGAGCAGAGTCCTC 3′ 5′ ATCTCACCATAAACTGCACG 3′
TRAV35 5′ GGTCAACAGCTGAATCAGAG 3′ 5′ ACCTGGCTATGGTACAAGC 3′
TRAV36 5′ GAAGACAAGGTGGTACAAAGC 3′ 5′ ATCTCTGGTTGTCCACGAG 3′
TRAV38-1, 5′ GCACATATGACACCAGTGAG 3′ 5′ CAGCAGGCAGATGATTCTC 3′
38-2
TRAV39 5′ CTGTTCCTGAGCATGCAG 3′ 5′ TCAACCACTTCAGACAGACTG 3′
TRAV40 5′ GCATCTGTGACTATGAACTGC 3′ 5′ GGAGGCGGAAATATTAAAGAC 3′
TRAV41 5′ AATGAAGTGGAGCAGAGTCC 3′ 5′ TTGTTTATGCTGAGCTCAGG 3′
TRAC 5′ GACCAGCTTGACATCACAG 3′ 5′ TGTTGCTCTTGAAGTCCATAG 3′
TRB gene(s)
targeted by
primer External primer sequence (EXT) Internal primer sequence (INT)
TRBV2 5′ TCGATGATCAATTCTCAGTTG 3′ 5′ TTCACTCTGAAGATCCGGTC 3′
TRBV3-1 5′ CAAAATACCTGGTCACACAG 3′ 5′ AATCTTCACATCAATTCCCTG 3′
TRBV4-1, 5′ TCGCTTCTCACCTGAATG 3′ 5′ CCTGCAGCCAGAAGACTC 3′
4-2, 4-3
TRBV5-1, 5′ GATTCTCAGGKCKCCAGTTC 3′ 5′ CTTGGAGCTGGRSGACTC 3′
5-3, 5-4
TRBV5-5, 5′ GTACCAACAGGYCCTGGGT 3′ 5′ TCTGAGCTGAATGTGAACG 3′
5-6, 5-7,
5-8
TRBV6-1, 5′ ACTCAGACCCCAAAATTCC 3′ 5′ GTGTRCCCAGGATATGAACC 3′
6-2, 6-3,
6-5, 6-6,
6-7, 6-8,
6-9
(continued)
186 Pradyot Dash et al.
Table 1
(continued)
TRB gene(s)
targeted by
primer External primer sequence (EXT) Internal primer sequence (INT)
TRBV6-4 5′ ACTGGCAAAGGAGAAGTCC 3′ 5′ TGGTTATAGTGTCTCCAGAGC 3′
TRBV7-1, 5′ TRTGATCCAATTTCAGGTCA 3′ 5′ TCYACTCTGAMGWTCCAGCG 3′
7-2, 7-3
TRBV7-4, 5′ GSWTCTYTGCAGARAGGCC 3′ 5′ TGRMGATYCAGCGCACA 3′
7-6, 7-7,
7-8, 7-9
TRBV9 5′ GATCACAGCAACTGGACAG 3′ 5′ GTACCAACAGAGCCTGGAC 3′
TRBV10-1, 5′ TGTWCTGGTATCGACAAGACC 3′ 5′ TCCYCCTCACTCTGGAGTC 3′
10-2, 10-3
TRBV11-1, 5′ CGATTTTCTGCAGAGACGC 3′ 5′ GACTCCACTCTCAAGATCCA 3′
11-2, 11-3
TRBV12-3, 5′ ARGTGACAGARATGGGACAA 3′ 5′ CYACTCTGARGATCCAGCC 3′
12-4, 12-5
TRBV13 5′ AGCGATAAAGGAAGCATCC 3′ 5′ CATTCTGAACTGAACATGAGC 3′
TRBV14 5′ CCAACAATCGATTCTTAGCTG 3′ 5′ ATTCTACTCTGAAGGTGCAGC 3′
TRBV15 5′ AGTGACCCTGAGTTGTTCTC 3′ 5′ ATAACTTCCAATCCAGGAGG 3′
TRBV16 5′ GTCTTTGATGAAACAGGTATGC 3′ 5′ GAAAGATTTTCAGCTAAGTGCC 3′
TRBV17 5′ CAGACCCCCAGACACAAG 3′ 5′ TGTTCACTGGTACCGACAG 3′
TRBV18 5′ CATAGATGAGTCAGGAATGCC 3′ 5′ CGATTTTCTGCTGAATTTCC 3′
TRBV19 5′ AGTTGTGAACAGAATTTGAACC 3′ 5′ TTCCTCTCACTGTGACATCG 3′
TRBV20-1 5′ AAGTTTCTCATCAACCATGC 3′ 5′ACTCTGACAGTGACCAGTGC 3′
TRBV23-1 5′ GCGATTCTCATCTCAATGC 3′ 5′ GCAATCCTGTCCTCAGAAC 3′
TRBV24-1 5′ CCTACGGTTGATCTATTACTCC 3′ 5′ GATGGATACAGTGTCTCTCGA 3′
TRBV25-1 5′ ACTACACCTCATCCACTATTCC 3′ 5′ CAGAGAAGGGAGATCTTTCC 3′
TRBV27, 28 5′ TGGTATCGACAAGACCCAG 3′ 5′ TTCYCCCTGATYCTGGAGTC 3′
TRBV29-1 5′ TTCTGGTACCGTCAGCAAC 3′ 5′ TCTGACTGTGAGCAACATGAG 3′
TRBV30 5′ TCCAGCTGCTCTTCTACTCC 3′ 5′ AGAATCTCTCAGCCTCCAGAC 3′
TRBC 5′ TAGAACTGGACTTGACAGCG 3′ 5′ TTCTGATGGCTCAAACACAG 3′
Primers targeting TRAV and TRBV genes are sense. Primers targeting TRAC and TRBC genes are antisense. TRAV
T-cell receptor Vα, TRAC T-cell receptor Cα, TRBV T-cell receptor Vβ, TRBC T-cell receptor Cβ
Single-Cell Analysis of Human αβT Cell Repertoire 187
3 Methods
3.2 Single-Cell 1. Resuspend the cells in 0.5 ml of sort buffer containing RNase
Sorting inhibitor at a concentration of 200 U/ml (see Note 3).
2. Filter the cell suspension through a 40 μm cell strainer.
3. (This step describes the gating strategy for the sorting of tet-
ramer + CD8+ cells. Substitute appropriate gating strategies
for other cell populations accordingly.) Gate the lymphocytes
first on their scatter properties based on an FSC-A/SSC-A
plot. From the probable lymphocyte population, select the
live cells based on Live/Dead staining and CD14 negativity.
Then gate on CD3+CD8+ cells to enrich the tetramer+ cells
for sorting.
4. Set the parameter on MoFlo to “single cell 1” with a drop
envelope of 1 (or corresponding parameter on other cell sort-
ers) to ensure stringency for single-cell only sorting. Additional
crosschecks for single-cell deposition accuracy should be car-
ried out by the users by visualization under light microscopy
of single cells in 96-well culture plates and by confirmation
of clonality in single-cell sorted hybridomas from mixed
cultures.
5. Sort the cells of interest into each well in Columns 1–10 of a
96-well polypropylene PCR plate (Eppendorf). Leave
Columns 11 and 12 empty. Column 12 will be the negative
(no template) control. Following the sort, seal the plate with
adhesive plate seal. Make sure that the edges of the adhesive
Single-Cell Analysis of Human αβT Cell Repertoire 189
3.3 Reverse The reverse transcription of the TCRα and β mRNA is carried out
Transcription (RT) directly on the lysed cells in the 96-well plate without any RNA
extraction step. Lysis is achieved by the combination of freeze-
thaw cycle and inclusion of a detergent, Triton X-100, in the RT
mixture.
1. Remove the plate from −80 °C and thaw on ice.
2. Centrifuge the plate at 500 × g for 2 min and keep on ice.
3. Prepare master mix for RT. We have reduced the final volume
of RT mix to one-eighth of the manufacturer’s recommenda-
tion (from 20 to 2.5 μl) for single-cell reactions with a modi-
fication where we add Triton-X 100 to a final concentration of
0.1 % (see Note 4).
3.4 Nested PCR Carry out a nested PCR protocol as described below for amplifying
the TCRα and β chain from single cells. A schematic for the PCR
is shown in Fig. 1.
190 Pradyot Dash et al.
Fig. 1 Overview of single-cell multiplex clonotypic analysis of epitope-specific T cells. Single epitope-specific
CD8+ T cells are sorted on a flow cytometric cell sorter into 96-well PCR plates. RT-PCR is performed on the
individual cells. The resultant cDNA is subjected to two rounds of nested PCR. In the first round, CDR3α and
CDR3β transcript amplification is achieved with the use of a multiplexed, comprehensive panel of external
sense Vα and Vβ and antisense Cα and Cβ segment-specific primers. First-round PCR products are subjected
to two separate second-round PCRs, incorporating, respectively, a multiplexed panel of external sense Vα and
antisense Cα or external sense Vβ and antisense Cβ segment-specific primers. PCR products thus derived are
sequenced and translated to yield paired CDR3αβ repertoire data. Inset: Nucleotide products from nested PCR
performed on cDNA derived from single CMV-NLV-specific CD8+ T cells from a young adult donor, incorporat-
ing TCRα- (top two rows) and TCRβ-specific primers (bottom two rows). Reproduced with permission from
Wang et al., 2012 [12]
Single-Cell Analysis of Human αβT Cell Repertoire 191
2. Remove the plate seal carefully and add 22.5 μl of master mix
to all samples and control wells using a 12-channel pipette
(5–50 μl) (see Note 5).
3. Reseal the plate thoroughly with a new adhesive plate seal,
ensuring the edges are sealed tightly, and centrifuge the plate
to settle the contents to the bottom of wells.
4. Perform PCR in a thermocycler as follows:
Initial denaturation, 5 min at 95 °C; denaturation, 20 s at
95 °C; primer annealing, 2 0 s at 52 °C; polymerase exten-
sion, 45 s at 72 °C; repeat denaturation to extension step 34
times; final extension, 7 min at 72 °C; and hold at +4 °C. The
plates can be stored at −20 °C until the next step.
5. Second-round PCR: At this point, you will set up TCRα and
TCRβ PCRs separately. Thus, identical wells of α and β plates
will be derived from single cells. For example, well position A1
of the α plate will pair with A1 of the β plate. Prepare two reac-
tion master mixes (α and β) as follows (see Note 4):
3.5 Agarose Gel 1. Cast a 2 % agarose gel in 1× TAE buffer using the tray and
Electrophoresis combs described in Subheading 2.2.
2. Using an 8-channel micropipette (0.5–10 μl), load the gel
wells with 5 μl of PCR products. The CoralLoad buffer con-
tains gel-loading dye, thus allowing direct loading of the PCR
reactions into the wells of the agarose gel.
3. Perform electrophoresis at 175 V for 40 min to separate the
PCR products.
4. At the end of the electrophoresis, visualize and record the
image of the agarose gel using a Bio-Rad or similar gel docu-
mentation system.
5. Count the number of bands and verify the negative controls
(see Note 6).
3.6 Sequencing Following a successful PCR reaction, the PCR products can be puri-
fied and sequenced by standard procedures. For example, the PCR
products can be purified using a silica-based column purification
system (Wizard SV96 PCR purification kit, Promega) as described
[11, 12]. Recently, we have adopted an Exonuclease I and Shrimp
Single-Cell Analysis of Human αβT Cell Repertoire 193
1 plate
Per reaction (μl) (100 reactions) (μl)
Shrimp alkaline 0.2 20
phosphatase (SAP, USB)
Exonuclease I (ExoI) (USB) 0.2 20
Tris–HCl, 50 mM, pH 8 4.6 460
Total 5 500
Per plate
Primer mix Per reaction (μl) (100 reactions) (μl)
TRAC INT REV (20 μM) 0.25 25
OR TRBC INT REV
(20 μM)
Water, Nuclease free 5.75 575
Total 6 600
3.7 Sequence Data We analyze the sequence data using a custom-made Microsoft Excel
Analysis analysis spreadsheet that utilizes macros obtained from http://
www.bioc.uzh.ch/antibody and ref. 14 and a TCR web application
https://tcr.stjude.org/tcr/ [11] that queries the international
ImMunoGeneTics information system (IMGT) database [15].
194 Pradyot Dash et al.
The output returns the CDR3α and CDR3β nucleotide and amino
acid sequence information and their frequencies, paired expression
profile, TRAV, TRAJ, TRBV, TRBJ, and TRBD usage data in the
same Excel spreadsheet, thus allowing further downstream calcula-
tions and statistics. The Excel spreadsheet consists of three work-
sheets for CDR3α and three worksheets for CDR3β analysis. The
data are summarized in the data summary worksheet. The next two
worksheets integrate the TRAV, TRAJ, TRBV, TRBJ, and TRBD
assignments derived from the TCR web application, https://tcr.
stjude.org/tcr/. The next two worksheets consist of pivot tables
that summarize the pairing data for CDR3, TRAV-TRAJ and
TRBV-TRBJ. A template for the customized, macro-enabled Excel
analysis spreadsheet is available from the authors upon request.
1. Download the sequence data in .ab1 and .seq format.
2. Using MegAlign software (DNASTAR Lasergene), import the
.ab1 files.
3. Copy the alignment report and paste into Cell A1 of the
“DNA seqs-Valpha” worksheet of the Excel spreadsheet.
4. Select Cells B5–B85 and run the reverse complement macro
(“revcomp.revcomp”). Over the next seven columns (C–H),
the nucleotide sequences (64 nt) are then pruned to extract
the CDR3 region (V end-CDR3-J-C beginning) and summa-
rized in Columns K, L, and M of the worksheet.
5. In the “Parsed-Valpha” worksheet, the extracted nucleotide
sequences are converted into amino acid sequences. Select
Cells A4–A83 and run the “AA_Parse_3Letter” macro. The
64 nucleotides of the CDR3 region are then parsed into three
letter blocks in Columns B4–V4.
6. Select Cells B4–V83 and run the “AA_Convert_NtoAA”
macro to derive the amino acid sequences.
7. In the “AA seqs-Valpha” worksheet, the amino acid sequence
data of the CDR3 region are further processed to extract the
exact CDR3 sequences, and their frequencies are calculated.
8. The exact same steps are followed for the analysis of the TCRβ
sequences to extract CDR3β data in the next three
worksheets.
9. The data are then summarized to derive paired CDR3αβ
expression in the “Data Summary” worksheet.
10. To assign the TRAV-TRAJ and TRBV-TRBD-TRBJ nomen-
clature, a compressed file of the .seq files is uploaded to the
https://tcr.stjude.org/tcr/ web application written in PHP 5
and MySQL 5 [11]. This interface allows users to upload indi-
vidual FASTA files or zipped archives of multiple FASTA files
and queries them against the IMGT online database using PHP
cUrl. The resulting output containing receptor family names,
Single-Cell Analysis of Human αβT Cell Repertoire 195
4 Notes
Acknowledgments
References
1. Jorgensen JL, Esser U, de St F, Groth B, Reay Kieff E, Sculley TB, Misko IS (1994) Dominant
PA, Davis MM (1992) Mapping T-cell selection of an invariant T cell antigen receptor
receptor-peptide contacts by variant peptide in response to persistent infection by Epstein-
immunization of single-chain transgenics. Barr virus. J Exp Med 180:2335–2340
Nature 355:224–230 8. Price DA, Brenchley JM, Ruff LE, Betts MR,
2. Davis MM, Bjorkman PJ (1988) T-cell antigen Hill BJ, Roederer M, Koup RA, Migueles SA,
receptor genes and T-cell recognition. Nature Gostick E, Wooldridge L, Sewell AK, Connors
334:395–402 M, Douek DC (2005) Avidity for antigen
3. Blackwell TK, Alt FW (1989) Molecular char- shapes clonal dominance in CD8+ T cell popu-
acterization of the lymphoid V(D)J recombina- lations specific for persistent DNA viruses.
tion activity. J Biol Chem 264:10327–10330 J Exp Med 202:1349–1361
4. Valmori D, Dutoit V, Lienard D, Lejeune F, 9. Wang C, Sanders CM, Yang Q, Schroeder HW
Speiser D, Rimoldi D, Cerundolo V, Dietrich Jr, Wang E, Babrzadeh F, Gharizadeh B, Myers
PY, Cerottini JC, Romero P (2000) Tetramer- RM, Hudson JR Jr, Davis RW, Han J (2010)
guided analysis of TCR beta-chain usage High throughput sequencing reveals a com-
reveals a large repertoire of melan-A-specific plex pattern of dynamic interrelationships
CD8+ T cells in melanoma patients. J Immunol among human T cell subsets. Proc Natl Acad
165:533–538 Sci U S A 107:1518–1523
5. Pannetier C, Even J, Kourilsky P (1995) T-cell 10. Sherwood AM, Desmarais C, Livingston RJ,
repertoire diversity and clonal expansions in Andriesen J, Haussler M, Carlson CS, Robins
normal and clinical samples. Immunol Today H (2011) Deep sequencing of the human
16:176–181 TCRγ and TCRβ repertoires suggests that
6. Trautmann L, Rimbert M, Echasserieau K, TCRβ rearranges after αβ and γδ T Cell com-
Saulquin X, Neveu B, Dechanet J, Cerundolo mitment. Sci Transl Med 3:90ra61
V, Bonneville M (2005) Selection of T cell 11. Dash P, McClaren JL, Oguin TH III, Rothwell
clones expressing high-affinity public TCRs W, Todd B, Morris MY, Becksfort J, Reynolds
within Human cytomegalovirus-specific CD8 C, Brown SA, Doherty PC, Thomas PG
T cell responses. J Immunol 175:6123–6132 (2011) Paired analysis of TCRα and TCRβ
7. Argaet VP, Schmidt CW, Burrows SR, Silins SL, chains at the single-cell level in mice. J Clin
Kurilla MG, Doolan DL, Suhrbier A, Moss DJ, Invest 121:288–295
Single-Cell Analysis of Human αβT Cell Repertoire 197
12. Wang GC, Dash P, McCullers JA, Doherty 14. Ewert S, Huber T, Honegger A, Pluckthun A
PC, Thomas PG (2012) T cell receptor αβ (2003) Biophysical properties of human anti-
diversity inversely correlates with pathogen- body variable domains. J Mol Biol 325:
specific antibody levels in human cytomegalo- 531–553
virus infection. Sci Transl Med 4:128ra42 15. Lefranc MP, Giudicelli V, Ginestoux C,
13. Sun X, Saito M, Sato Y, Chikata T, Naruto T, Jabado-Michaloud J, Folch G, Bellahcene F,
Ozawa T, Kobayashi E, Kishi H, Muraguchi A, Wu Y, Gemrot E, Brochet X, Lane J, Regnier
Takiguchi M (2012) Unbiased analysis of L, Ehrenmann F, Lefranc G, Duroux P (2009)
TCRα/β chains at the single-cell level in IMGT, the international ImMunoGeneTics
human CD8+ T-cell subsets. PLoS One information system. Nucleic Acids Res
7:e40386 37:D1006–D1012
Chapter 16
Abstract
The B cell receptor (BCR) repertoire is highly diverse. Repertoire diversity is achieved centrally by somatic
recombination of immunoglobulin (Ig) genes and peripherally by somatic hypermutation and Ig heavy
chain class-switching. Throughout these processes, there is selection for functional gene rearrangements,
selection against gene combinations resulting in self-reactive BCRs, and selection for BCRs with high affin-
ity for exogenous antigens after challenge. Hence, investigation of BCR repertoires from different groups
of B cells can provide information on stages of B cell development and shed light on the etiology of B cell
pathologies. In most instances, the third complementarity determining region of the Ig heavy chain (CDR-
H3) contributes the majority of amino acids to the antibody/antigen binding interface. Although CDR-
H3 spectratype analysis provides information on the overall diversity of BCR repertoires, this fairly simple
technique analyzes the relative quantities of CDR-H3 regions of each size, within a range of approximately
10–80 bp, without sequence detail and thus is limited in scope. High-throughput sequencing (HTS) tech-
niques on the Roche 454 GS FLX Titanium system, however, can generate a wide coverage of Ig sequences
to provide more qualitative data such as V, D, and J usage as well as detailed CDR3 sequence information.
Here we present protocols in detail for CDR-H3 spectratype analysis and HTS of human BCR
repertoires.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_16, © Springer Science+Business Media New York 2015
199
200 Yu-Chang Wu et al.
2 Materials
2.2 Components PCR components listed below are used to generate products for
for Polymerase Chain CDR-H3 spectratyping and HTS, using appropriate primers as
Reaction indicated in different sections.
1. PCR-grade H2O.
2. dNTP mix: 20 mM each of dATP, dTTP, dCTP, dGTP in
PCR-grade H2O.
3. Phusion High-Fidelity polymerase (NEB, UK) diluted in
PCR-grade water.
4. 5× GC buffer (supplied with Phusion High-Fidelity poly-
merase; NEB, UK).
5. Complete primer mix: choose accordingly as indicated [6, 15].
6. cDNA (see Subheading 3.1).
7. 96-well PCR plates.
8. PCR thermal cyclers.
9. Thermal seals.
7. 15 ml Falcon tubes.
8. Disposable scalpels.
9. DarkReader 46B Transilluminator instrument (LabTech, UK).
10. QIAquick gel purification kit (Qiagen, UK).
11. Qubit 2.0 Fluorometer and Qubit dsDNA HS Assay Kits
(Invitrogen, UK).
12. QIAquick PCR purification kit (Qiagen, UK).
3 Methods
3.1 Direct cDNA 1. Assemble the components of SLyRT buffer as listed in Table 1.
Synthesis from Cells Keep SLyRT buffer on ice for immediate use or store at
−20 °C.
2. Aliquot 36 μl of the SLyRT buffer into PCR tubes.
3. Deposit 1000–10,000 B cells from the flow cytometry sorter
directly into PCR tubes containing 36 μl of SLyRT buffer.
Table 1
SLyRT buffer
Table 2
Reverse transcriptase reaction conditions
4. Invert PCR tubes several times to ensure all cells are immersed
in SLyRT buffer. Briefly centrifuge the PCR tubes at 3000 rpm
in a microfuge for 2 min.
5. Add 4 μl of SuperScript III RT enzyme at 25 U/μl, final reac-
tion concentration 2.5 U/μl.
6. Carry out the RT reaction on the thermal cycler, programmed
as in Table 2.
7. Once the RT reaction is complete, centrifuge the PCR tubes at
13,000 rpm in a microfuge for 5 min. Immediately transfer
35 μl of the reaction mix to a clean PCR tube without dis-
turbing any cell debris at the bottom of the tube.
This step reduces cellular contaminants from being carried
forward to downstream applications.
8. Add 70 μl of PCR-grade H2O to 35 μl of the reaction mix and
pipette several times to mix (see Note 1).
9. Store samples at −20 °C.
Table 3
Spectratype PCR reaction mix (25 μl per reaction)
Table 4
PCR thermal cycling conditions
4. Seal PCR plates with thermal seals and carry out PCR reaction
on the thermal cycler programmed as Table 4.
5. Briefly centrifuge the PCR plate to collect the content.
6. In a non-skirted PCR plate, add 3 μl of the PCR products to
1.5 μl GeneScan 350 TAMRA Size Standard and 13.5 μl
Hi-Di formamide per well. Pipette up and down a couple of
times to mix thoroughly (see Note 8).
206 Yu-Chang Wu et al.
3.3 HTS of Human HTS allows analysis of BCR repertoires in detail, including the dis-
Immuno tribution of CDR-H3 sizes. This section includes production of
globulin Genes immunoglobulin gene amplicons, gel purification of amplicons,
assembly of multiple amplicons (up to 12 MID barcodes per
sequencing sample), and enrichment of pooled amplicons.
Sequencing on the 454 GS FLX Titanium instrument is performed
by LGC Genomics, where each sequencing sample (containing 12
MIDS) is run on 1/16th of a chip. Take care to avoid cross contami-
nation between samples. Each research (cDNA) sample that is
required to be distinguishable from other research samples needs
to be amplified with its own MID tag. Within that MID tag it is
possible to combine three different isotypes of heavy chains, and
kappa and lambda light chains for that particular research sample,
since these can be distinguished later by their sequence (see Note 11).
1. To generate MID-tagged amplicons, perform semi-nested
PCR reactions (Fig. 2).
2. Assemble reagents for PCR1 master mix (Table 5), and vortex
to mix thoroughly. Change primer mix accordingly to pro-
duce different types of amplicons (different heavy chain classes
or kappa light chain or lambda light chains).
3. Aliquot the PCR1 master mix into a 96-well PCR1 plate,
22 μl per well.
4. Add 1:3 diluted cDNA (see Subheading 3.1) to the 96-well
PCR1 plate, 3 μl per well. Use one PCR plate for each type of
amplicon (i.e., either IgA, IgG, IgM, IgK, or IgL) and per-
form eight PCR1 reactions for each sample.
5. Seal PCR plates with thermal seals and carry out PCR1 reac-
tion on the thermal cycler programmed as in Table 6.
Fig. 2 Schematic representation of immunoglobulin gene cDNA and primer binding sites for HTS, semi-nested
PCR
Assessment of Human B Cell Repertoire 207
Table 5
HTS PCR1 reaction mix (25 μl per reaction)
Table 6
PCR1 thermal cycling conditions
Table 7
HTS PCR2 reaction mix (20 μl per reaction)
Table 8
PCR2 thermal cycling conditions
Fig. 3 Diagram to illustrate the PCR amplicon sizes of different IGH isotypes, IGL and IGK, in relation to the
Hyperladder IV DNA size marker, on a 1.5 % TAE agarose gel
12. Load the preheated samples on the gel and use 5 μl of Hyper
Ladder IV Ladder. Separate DNA fragments by electrophoresis
at 100 V for 60 min.
13. Visualize DNA fragments using the DarkReader 46B
Transilluminator instrument.
14. Excise fragments of the right size (Fig. 3), using sterile scalpels
and transfer gel slices into 15 ml Falcon tubes.
210 Yu-Chang Wu et al.
15. Extract DNA using the QIAquick gel purification kit, according
to the manufacturer’s instructions, except for the following
changes:
(a) Incubate gel slices in 5 ml of Buffer QG per amplicon type
in 15 ml Falcon tubes at 55 °C. Vortex frequently until gel
slices are melted. Expect approximately 6 ml of the solu-
tions in total per amplicon type in the 15 ml Falcon tube.
(b) Use one QIAquick spin column per amplicon type: apply
750 μl of the solution to the QIAquick spin column and
centrifuge at 13,000 rpm in a microfuge for 30 s. Collect,
rather than discard, the flow-through in a separate 15 ml
Falcon tube. Repeat this process until all mixture has been
applied to the same column, spun and recollected in a sep-
arate 15 ml Falcon tube.
(c) Apply the flow-through (750 μl at a time) back to the same
spin column, spin at 13,000 rpm, and discard the second
flow-through.
(d) Wash the spin column with 750 μl Buffer PE by centrifug-
ing at 13,000 rpm for 1 min.
(e) Add 55 μl PCR-grade H2O to the column membrane and
leave at RT for 3 min.
(f) Centrifuge the column at 13,000 rpm for 1 min and col-
lect the elution in 1.5 ml microcentrifuge tube.
(g) Add additional 55 μl of PCR-grade water to the same col-
umn membrane and leave at RT for 3 min.
(h) Spin to collect the elution in the same 1.5 ml microcentri-
fuge tube. In total, approximately 100 μl of elution will be
collected in one 1.5 ml microcentrifuge tube.
16. Measure the concentration and quality of the gel-purified
products for each amplicon type using the Qubit dsDNA HS
Assay Kits on the Qubit 2.0 Fluorometer.
17. Assemble the multiple amplicons into one sequencing sample.
When 12 MID barcodes are used, transfer 900 ng of
products per MID to a clean 1.5 ml microcentrifuge tube,
i.e., a total of 10,800 ng are pooled from 12 MID types (see
Note 14). If amplicons of multiple isotypes of heavy chain
and light chains are allocated to the same MID barcode,
divide 900 ng equally. Examples for the amount (in ng) of
gel-purified products for pooling multiple amplicon types are
listed in Table 9.
18. Enrich pooled products using the QIAquick PCR purification
kit, according to the manufacturer’s instructions. This step is
carried out in a similar way as step 9, except Buffer PBI is used
to mix with the pooled gel-purified solutions. Elute products
in two lots of 55 μl PCR-grade water as step 9.
Assessment of Human B Cell Repertoire 211
Table 9
Assembly of sequencing samples
(b) Sequences that pass this quality control are then edited to
remove any terminal adapter and MID sequences added
as part of the sequencing protocol, and each sequence is
renamed so as to capture and embed sample information
based on the MID tag. Sequences are then passed in bulk
to HighV-QUEST [16, 17]. This is a freely available
online resource (http://www.imgt.org/HighV-QUEST/
index.action) that analyzes each sequence with reference
to a database of human Ig sequences and returns a sum-
mary file cataloguing a range of outputs for each sequence
including V, D, and J gene usage and CDR-H3 sequence
and length.
(c) The V-QUEST output file is then parsed locally to further
summarize the data for each sequence regarding gene
usage. The amino acid sequence of the CDR-H3 junction
peptide is extracted and analyzed for a range of physico-
chemical properties (hydrophobicity, charge, amino acid
use, and so forth) using a locally scripted version of the
ProtParam tool that is available on the ExPASy server [18].
In addition, the DNA sequence of the CDR3 region is
used, in combination with hierarchical clustering and tree
cutting, to identify clusters (“clones”) of sequences that
share related CDR-H3 sequences. Further analysis high-
lights clones that span multiple isotypes (indicative of class
switching). Finally, a single sequence from each clone is
identified that represents the most common pattern of
gene usage as a reference sequence for further analysis,
such as regarding overall repertoire (see Note 15).
4 Notes
1. At low cell numbers this is not an issue but if high cell numbers
(over 6000 cells) are used, the subsequent PCR steps may be
more efficient if the cDNA sample synthesized using SLyRT
buffer is diluted 1 in 3 (35+ 70 μl) with PCR-grade water.
2. 100 % Triton is difficult to pipette due to high viscosity so it
may be helpful to cut the end of the pipette tip with a scalpel
to make it wider, and pipette slowly.
3. Add 200 μl each of dATP, dTTP, dCTP, dGTP (20 mM each;
Promega, UK) to 200 μl DNase/RNase-free H2O and vortex
to mix.
4. 1× TAE running buffer contained 40 mM Tris-borate and
1 mM EDTA. To make 50× TAE, 242 g Tris base, 57.1 ml
glacial acetic acid (Merck, UK), and 100 ml EDTA (pH 8.0)
Assessment of Human B Cell Repertoire 213
Table 10
Spectratyping primers
Sequence (5′–3′)
5′-end primer
FW3-FAM ACACGGCTGTGTATTACTGT
In combination with
3′-end primer
IGHA or GGAAGAAGCCCTGGACCAGGC
IGHG or CACCGTCACCGGTTCGG
IGHM CAGGAGACGAGGGGGAA
Table 11
Example of multiplex sequencing sample assembly
Patient Tissue Cell type IgM IgG IgA kappa lambda Multiplex ID
A Blood Transitional B cell × NA NA × × MID1
Blood Naive B cell × NA NA × × MID2
Blood Memory B cell × × × × × MID3
Blood Plasma cell × × × × × MID4
B Blood Transitional B cell × NA NA × × MID5
Blood Naive B cell × NA NA × × MID6
Blood Memory B cell × × × × × MID7
Blood Plasma cell × × × × × MID8
C Blood Transitional B cell × NA NA × × MID9
Blood Naive B cell × NA NA × × MID10
Blood Memory B cell × × × × × MID11
Blood Plasma cell × × × × × MID12
12. PCR1 primer mix (Table 12) contains 5′-end multiplex primers
that anneal to all families of the variable regions and 3′-end prim-
ers that anneal to the constant region. 1.25 μl of the primer mix,
containing 835 nM each of 5′-end primers and 5 μM 3′-end
primer, is used in a final reaction volume of 25 μl, to give the final
concentrations at 41.75 nM each and 250 nM, respectively.
13. PCR2 primers are comprised of gene-specific sequences and
non-gene-specific, 10-base MID sequence motifs. PCR2
primer mix contains 5′-end multiplex MID-containing prim-
ers that anneal to all families of the variable regions and 3′-end
MID-containing primers that anneal to the constant region.
1 μl of the MID-containing primer mix, containing 835 nM
each of 5′-end primers and 5 μM 3′-end primer, is used in a
final reaction volume of 20 μl, to give the final concentrations
at 41.75 nM each and 250 nM, respectively. Each primer mix
contains only one MID barcode for amplification from one
research sample and a total of 12 different MID barcodes can
be used for 12 different research samples (Table 13).
The gene-specific sequences of 5′-end primers for the heavy
and light chains and 3′-end primers for the light chains are same as
PCR1. The gene-specific sequences in the 3′-end primers of the
heavy chains anneal for PCR2 reactions (Table 14) to the template
more upstream to those used in PCR1.
For space consideration, MID1-containing primers for IgA are
shown as an example (Table 15).
14. Although it only requires 5 ng (50 ng/μl) in total of the puri-
fied pooled mixture to be sequenced as one sequencing sample
using a 1/16th microchip on the 454 GS FLX Titanium
Table 12
PCR1 primer mix
Table 13
PCR2 primer mix
Sequence (5′–3′)
MID 1 acgagtgcgt
MID 2 acgctcgaca
MID 3 agacgcactc
MID 4 agcactgtag
MID 5 atcagacacg
MID 6 atatcgcgag
MID 7 cgtgtctcta
MID 8 ctcgcgtgtc
MID 9 tagtatcagc
MID 10 tctctatgcg
MID 11 tgatacgtct
MID 12 tactgagcta
Table 14
3′ Heavy chain primers
Sequence (5′–3′)
IGHA″ GGAAGAAGCCCTGGACCAGGC
IGHG″ CACCGTCACCGGTTCGGGG
IGHM″ CAGGAGACGAGGGGGAAAAGG
Table 15
MID1-containing IgA primers
3′-end primer
5′-end primer Sequence (5′–3′) Sequence (5′–3′)
MID1:IGHV1 acgagtgcgtCCTCAGTGAA combined MID1:IGHA″
GGTCTCCTGCAAGG with acgagtgcgtGGCTCCTGG
MID1:IGHV2 acgagtgcgtTCCTGCGCTG GGGAAGAAGCC
GTGAAACCCACACA
MID1:IGHV3 acgagtgcgtGGTCCCTG
AGACTCTCCTGTGCA
MID1:IGHV4 acgagtgcgtTCGGAGAC
CCTGTCCCTCACCTGC
MID1:IGHV5 acgagtgcgtCAGTCTG
GAGCAGAGGTGAAA
MID1:IGHV6 acgagtgcgtCCTGTGCCA
TCTCCGGGGACAGTG
Assessment of Human B Cell Repertoire 217
References
1. Sanz I (1991) Multiple mechanisms participate 9. Gibson KL, Wu YC, Barnett Y, Duggan O,
in the generation of diversity of human H chain Vaughan R, Kondeatis E, Nilsson BO, Wikby
CDR3 regions. J Immunol 147:1720–1729 A, Kipling D, Dunn-Walters DK (2009) B-cell
2. Tonegawa S (1983) Somatic generation of diversity decreases in old age and is correlated
antibody diversity. Nature 302:575–581 with poor health status. Aging Cell 8:18–25
3. Raaphorst FM, Raman CS, Tami J, Fischbach 10. Tschumper RC, Asmann YW, Hossain A,
M, Sanz I (1997) Human Ig heavy chain Huddleston PM, Wu X, Dispenzieri A, Eckloff
CDR3 regions in adult bone marrow BW, Jelinek DF (2012) Comprehensive
pre-B cells display an adult phenotype of diver- assessment of potential multiple myeloma
sity: evidence for structural selection of DH immunoglobulin heavy chain V-D-J intra-
amino acid sequences. Int Immunol clonal variation using massively parallel pyrose-
9:1503–1515 quencing. Oncotarget 3:502–513
4. Volpe JM, Kepler TB (2008) Large-scale anal- 11. Warsame AA, Aasheim HC, Nustad K, Troen
ysis of human heavy chain V(D)J recombina- G, Tierens A, Wang V, Randen U, Dong HP,
tion patterns. Immunome Res 4:3 Heim S, Brech A, Delabie J (2011) Splenic
5. Larimore K, McCormick MW, Robins HS, marginal zone lymphoma with VH1-02 gene
Greenberg PD (2012) Shaping of human rearrangement expresses poly- and self-reactive
germline IgH repertoires revealed by deep antibodies with similar reactivity. Blood
sequencing. J Immunol 189:3221–3230 118:3331–3339
6. Ademokun A, Wu YC, Martin V, Mitra R, Sack 12. Luo C, Tsementzi D, Kyrpides N, Read T,
U, Baxendale H, Kipling D, Dunn-Walters DK Konstantinidis KT (2012) Direct comparisons
(2011) Vaccination-induced changes in human of Illumina vs. Roche 454 sequencing tech-
B-cell repertoire and pneumococcal IgM and nologies on the same microbial community
IgA antibody at different ages. Aging Cell DNA sample. PLoS One 7:e30087
10:922–930 13. Meyer M, Stenzel U, Myles S, Prufer K,
7. Wu YC, Kipling D, Dunn-Walters DK (2012) Hofreiter M (2007) Targeted high-through-
Age-related changes in human peripheral put sequencing of tagged nucleic acid samples.
blood IGH repertoire following vaccination. Nucleic Acids Res 35, e97
Front Immunol 3:193 14. Wu YC, Kipling D, Dunn-Walters DK (2011)
8. Cuisinier AM, Guigou V, Boubli L, Fougereau The relationship between CD27 negative and
M, Tonnelle C (1989) Preferential expression positive B cell populations in human peripheral
of VH5 and VH6 immunoglobulin genes in blood. Front Immunol 2:81
early human B-cell ontogeny. Scand J Immunol 15. Wu YC, Kipling D, Leong HS, Martin V,
30:493–497 Ademokun AA, Dunn-Walters DK (2010)
218 Yu-Chang Wu et al.
Abstract
High-throughput DNA sequencing techniques have greatly accelerated the pace of research into the
repertoires of antibody and T cell receptor gene rearrangements that confer antigen specificity to adaptive
immune responses. Studies of aging-related changes in human B cell repertoires have benefited from
the ability to detect and quantify thousands to millions of B cell clones in human samples, and study the
mutational lineages and isotype switching relationships within each clonal lineage. Correlation of reper-
toire analysis with antibody gene data from antigen-specific B cells is poised to give much greater insight
into clinically relevant B cell responses and memory storage. Here, we describe strategies for preparing and
analyzing human antibody gene libraries for studying B cell repertoires.
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_17, © Springer Science+Business Media New York 2015
219
220 Chen Wang et al.
2 Materials
3 Methods
3.1 Primer Design 1. For 454 library preparation, forward and reverse PCR primers
and Barcoding are designed with the following elements in 5′–3′ order: 454
instrument-specific primer regions (required for emulsion
PCR and initiation of sequencing in the instrument protocols),
followed by a 10-base “barcode” sequence that encodes the
sample identity and PCR replicate library identity, followed by
the IGHV or IGHJ gene-specific primer sequence. Primer
sequences and barcodes are indicated in Tables 1 and 2.
3.2 Template 1. Genomic DNA and RNA are isolated from peripheral blood
Preparation mononuclear cell samples, or purified B cell samples using the
standard protocols in the Qiagen All-Prep kit.
2. cDNA synthesis is carried out with priming by random hex-
amer primers at 20 ng/μl final concentration, following the
manufacturer’s protocol for reverse transcription (Invitrogen).
The RNase H RNA hydrolysis step following cDNA synthesis
does not appear to be necessary for subsequent Ig PCR
amplifications.
Table 1
Primer sequences for PCR amplification of gDNA library 1st reaction. The 5′-end primers consist of
adapter sequences for 454 platform (454-A), barcodes encoding sample identity, and sequences
complementary to the FR1 or FR2 regions of different V-gene families [3, 7, 17]. The 3′-end primers
consist of adapter 454-B, barcodes encoding replicate identity, and sequence complementary to the
J region
2 mM dNTP: 3 μl
AmpliTaq Gold (5 U/μl): 0.3 μl
Water to 30 μl
5. Although the AmpliTaq Gold is a hot-start polymerase, we
keep the PCR plate on ice until the thermocycler block has
reached 95 °C, and then add the plate to the thermocycler.
6. The PCR program is as follows: Denaturation 95 °C for 7 min;
35 cycles of 95 °C for 30 s, 60 °C for 45 s, and 72 °C for 90 s;
and final extension: 72 °C for 10 min. If the PCR will run
overnight, we set the thermocycler to keep the plate at 10 °C
indefinitely after the run is complete.
7. Products of PCR are analyzed by 1.5 % agarose gels with
1×TAE running buffer, using ethidium bromide for DNA
visualization under ultraviolet light (ethidium bromide is
added to running buffer as well as the TAE used to make the
gel). We usually use the NEB Quick-load 100 bp ladder for
size comparison.
Antibody Gene Repertoire Sequencing 225
Table 2
Examples of sequence barcodes in the IGHJ primer being used to indicate the sample (Sample 1–
Sample 4) from which a library was generated, while barcodes in the IGHV FR1 and FR2 primer sets
are used to label the different replicate libraries generated by PCR of distinct aliquots of genomic
DNA template from the sample (replicate libraries R1–R6 for each sample). Different replicate
libraries from the same sample share the J-barcode and are distinguished by V-barcodes
3.4 Library Pooling 1. Based on the intensities of amplified bands from each PCR
and Gel Purification reaction, we pool the barcoded products in approximately
equimolar amounts (or with greater representation of particu-
lar samples, if more reads are wanted from them).
2. Run the pooled library on a new 1.5 % agarose gel in 1×TAE
buffer (using a comb with several teeth taped together to give
a broad single well), and cut out the gel slice containing the
desired PCR bands, being generous in the cutting to include
some gel above the highest band and below the lowest band.
We exclude the tips of the band from the excised slice, as
“smiling” of the band can cause the separation to be poorer at
the ends.
3. DNA is isolated from the gel slice using a Qiagen gel purification
kit, and eluted in TE buffer. The final gel-purified library band is
run on a final analytical 1.5 % agarose 1×TAE gel (see Note 3).
4. Quantitation of the pooled gel-extracted library is conducted
with Nanodrop spectrometry or Qubit fluorimetry.
226 Chen Wang et al.
3.5 High-Throughput 1. The 454 libraries are sequenced using 454 Titanium chemistry,
DNA Sequencing with amplicon sequencing reagents, and only from the “B” side.
Using the primer design presented here, the sequencing reads
start from the J segment end of the amplicon, and the Ig gene
rearrangement is sequenced as the reverse complement of the
mRNA sequence. The advantage of sequencing starting from
the J segment is that the VDJ junctional region is measured early
in the sequencing run when the read quality is highest.
2. Illumina libraries are sequenced with the MiSeq instrument,
using paired 250 bp read reagent kits. Using the primer design
presented here, the J segment end of the amplicon is sequenced
in the first read, while the second read begins from the V
segment.
3.6 DNA Sequence 1. There are a large number of options for processing and filtering
Data Analysis DNA sequence data. Our data analysis pipeline is integrated
with a PostgreSQL database for maintaining analysis results in
a consistent and orderly format, and to facilitate comparison
between sequences obtained from different experiments and
different sequencing runs. The components of the data analy-
sis pipeline are carried out sequentially as indicated below.
2. Identifying which sequences in the 454 instrument output files
came from each sample and each replicate sequence library
from that sample is carried out by searching for exact matches
to the “barcode” sequences encoded in the forward and reverse
PCR primers for each sample and replicate library. In addition
to matching the barcode sequence perfectly, the 7 bases of
gene-specific primer that follow the barcode sequence are
required to match exactly. This criterion enables the use of
pooled libraries in which the same barcode sequence can be
used with different primer sets, such as IGH primers and T cell
receptor primers, in the same sequencing run.
3. Gene-specific primer sequences are trimmed from the sequence
reads, to avoid incorporating primer-encoded sequence into
the later steps in the data analysis. For IGH amplicons that are
to be further analyzed with the iHMMune-align algorithm, we
do not trim the IGHJ primer, as the program requires the
additional length of IGHJ sequence to function. In this par-
ticular case, downstream data analysis does not make use of
apparent hypermutation positions that occur in the IGHJ
primer-encoded parts of the sequence.
4. Alignment of Ig gene products to germline V, D, and J gene
segments can be carried out with a variety of different
programs. We have used iHMMune-align (www.ihmmune.
unsw.edu.au), IgBLAST (www.ncbi.nlm.nih.gov/igblast/),
and IMGT/V-QUEST (www.imgt.org) in the past [22–24].
Antibody Gene Repertoire Sequencing 227
4 Notes
References
1. Jung D, Alt FW (2004) Unraveling V(D)J 3. Boyd SD, Marshall EL, Merker JD, Maniar JM,
recombination; insights into gene regulation. Zhang LN, Sahaf B, Jones CD, Simen BB,
Cell 116(2):299–311 Hanczaruk B, Nguyen KD, Nadeau KC, Egholm
2. Hozumi N, Tonegawa S (1976) Evidence for M, Miklos DB, Zehnder JL, Fire AZ (2009)
somatic rearrangement of immunoglobulin Measurement and clinical monitoring of human
genes coding for variable and constant regions. lymphocyte clonality by massively parallel VDJ
Proc Natl Acad Sci U S A 73(10):3628–3632 pyrosequencing. Sci Transl Med 1(12):12–23
Antibody Gene Repertoire Sequencing 231
4. Robins HS, Campregher PV, Srivastava SK, between T cell subsets and individuals revealed
Wacher A, Turtle CJ, Kahsai O, Riddell SR, by pyrosequencing. J Immunol 186(7):4285–
Warren EH, Carlson CS (2009) Comprehensive 4294, doi: jimmunol.1003898 [pii]10.4049/
assessment of T-cell receptor beta-chain diversity jimmunol.1003898
in alphabeta T cells. Blood 114(19):4099–4107, 11. Glanville J, Zhai W, Berka J, Telman D, Huerta
doi: blood-2009-04-217604 [pii]10.1182/ G, Mehta GR, Ni I, Mei L, Sundar PD, Day
blood-2009-04-217604 GM, Cox D, Rajpal A, Pons J (2009) Precise
5. Campbell PJ, Pleasance ED, Stephens PJ, determination of the diversity of a combinato-
Dicks E, Rance R, Goodhead I, Follows GA, rial antibody library gives insight into the
Green AR, Futreal PA, Stratton MR (2008) human immunoglobulin repertoire. Proc Natl
Subclonal phylogenetic structures in cancer Acad Sci U S A 106(48):20216–20221, doi:
revealed by ultra-deep sequencing. Proc Natl 0909775106 [pii]10.1073/pnas.0909775106
Acad Sci U S A 105(35):13081–13086, doi: 12. Bentley DR, Balasubramanian S, Swerdlow
0801523105 [pii]10.1073/pnas.0801523105 HP, Smith GP, Milton J, Brown CG, Hall KP,
6. Wang C, Sanders CM, Yang Q, Schroeder HW Evers DJ, Barnes CL, Bignell HR, Boutell JM,
Jr, Wang E, Babrzadeh F, Gharizadeh B, Myers Bryant J, Carter RJ, Keira Cheetham R, Cox
RM, Hudson JR Jr, Davis RW, Han J (2010) AJ, Ellis DJ, Flatbush MR, Gormley NA,
High throughput sequencing reveals a com- Humphray SJ, Irving LJ, Karbelashvili MS,
plex pattern of dynamic interrelationships Kirk SM, Li H, Liu X, Maisinger KS, Murray
among human T cell subsets. Proc Natl LJ, Obradovic B, Ost T, Parkinson ML, Pratt
Acad Sci U S A 107(4):1518–1523, doi: MR, Rasolonjatovo IM, Reed MT, Rigatti R,
0913939107 [pii]10.1073/pnas.0913939107 Rodighiero C, Ross MT, Sabot A, Sankar SV,
7. Margulies M, Egholm M, Altman WE, Attiya Scally A, Schroth GP, Smith ME, Smith VP,
S, Bader JS, Bemben LA, Berka J, Braverman Spiridou A, Torrance PE, Tzonev SS, Vermaas
MS, Chen YJ, Chen Z, Dewell SB, Du L, EH, Walter K, Wu X, Zhang L, Alam MD,
Fierro JM, Gomes XV, Godwin BC, He W, Anastasi C, Aniebo IC, Bailey DM, Bancarz
Helgesen S, Ho CH, Irzyk GP, Jando SC, IR, Banerjee S, Barbour SG, Baybayan PA,
Alenquer ML, Jarvie TP, Jirage KB, Kim JB, Benoit VA, Benson KF, Bevis C, Black PJ,
Knight JR, Lanza JR, Leamon JH, Lefkowitz Boodhun A, Brennan JS, Bridgham JA, Brown
SM, Lei M, Li J, Lohman KL, Lu H, Makhijani RC, Brown AA, Buermann DH, Bundu AA,
VB, McDade KE, McKenna MP, Myers EW, Burrows JC, Carter NP, Castillo N, Chiara
Nickerson E, Nobile JR, Plant R, Puc BP, ECM, Chang S, Neil Cooley R, Crake NR,
Ronan MT, Roth GT, Sarkis GJ, Simons JF, Dada OO, Diakoumakos KD, Dominguez-
Simpson JW, Srinivasan M, Tartaro KR, Fernandez B, Earnshaw DJ, Egbujor UC,
Tomasz A, Vogt KA, Volkmer GA, Wang SH, Elmore DW, Etchin SS, Ewan MR, Fedurco
Wang Y, Weiner MP, Yu P, Begley RF, Rothberg M, Fraser LJ, Fuentes Fajardo KV, Scott Furey
JM (2005) Genome sequencing in microfabri- W, George D, Gietzen KJ, Goddard CP, Golda
cated high-density picolitre reactors. Nature GS, Granieri PA, Green DE, Gustafson DL,
437(7057):376–380, doi: nature03959 Hansen NF, Harnish K, Haudenschild CD,
[pii]10.1038/nature03959 Heyer NI, Hims MM, Ho JT, Horgan AM,
Hoschler K, Hurwitz S, Ivanov DV, Johnson
8. Freeman JD, Warren RL, Webb JR, Nelson MQ, James T, Huw Jones TA, Kang GD,
BH, Holt RA (2009) Profiling the T-cell recep- Kerelska TH, Kersey AD, Khrebtukova I,
tor beta-chain repertoire by massively parallel Kindwall AP, Kingsbury Z, Kokko-Gonzales
sequencing. Genome Res 19(10):1817–1824, PI, Kumar A, Laurent MA, Lawley CT, Lee
doi: gr.092924.109 [pii]10.1101/gr.092924. SE, Lee X, Liao AK, Loch JA, Lok M, Luo S,
109 Mammen RM, Martin JW, McCauley PG,
9. Wu YC, Kipling D, Leong HS, Martin V, McNitt P, Mehta P, Moon KW, Mullens JW,
Ademokun AA, Dunn-Walters DK (2010) Newington T, Ning Z, Ling Ng B, Novo SM,
High-throughput immunoglobulin repertoire O'Neill MJ, Osborne MA, Osnowski A,
analysis distinguishes between human IgM Ostadan O, Paraschos LL, Pickering L, Pike
memory and switched memory B-cell popula- AC, Chris Pinkard D, Pliskin DP, Podhasky J,
tions. Blood 116(7):1070–1078, doi: blood- Quijano VJ, Raczy C, Rae VH, Rawlings SR,
2010-03-275859 [pii]10.1182/blood-2010- Chiva Rodriguez A, Roe PM, Rogers J, Rogert
03-275859 Bacigalupo MC, Romanov N, Romieu A, Roth
10. Venturi V, Quigley MF, Greenaway HY, Ng RK, Rourke NJ, Ruediger ST, Rusman E,
PC, Ende ZS, McIntosh T, Asher TE, Almeida Sanches-Kuiper RM, Schenker MR, Seoane
JR, Levy S, Price DA, Davenport MP, Douek JM, Shaw RJ, Shiver MK, Short SW, Sizto NL,
DC (2011) A mechanism for TCR sharing Sluis JP, Smith MA, Ernest Sohna Sohna J,
232 Chen Wang et al.
Spence EJ, Stevens K, Sutton N, Szajkowski L, (2009) Broad diversity of neutralizing antibodies
Tregidgo CL, Turcatti G, Vandevondele S, isolated from memory B cells in HIV-infected
Verhovsky Y, Virk SM, Wakelin S, Walcott GC, individuals. Nature 458(7238):636–640.
Wang J, Worsley GJ, Yan J, Yau L, Zuerlein M, doi:10.1038/nature07930
Mullikin JC, Hurles ME, McCooke NJ, West JS, 17. van Dongen JJ, Langerak AW, Bruggemann
Oaks FL, Lundberg PL, Klenerman D, Durbin M, Evans PA, Hummel M, Lavender FL,
R, Smith AJ (2008) Accurate whole human Delabesse E, Davi F, Schuuring E, Garcia-Sanz
genome sequencing using reversible terminator R, van Krieken JH, Droese J, Gonzalez D,
chemistry. Nature 456(7218):53–59, doi: Bastard C, White HE, Spaargaren M, Gonzalez
nature07517 [pii]10.1038/nature07517 M, Parreira A, Smith JL, Morgan GJ, Kneba
13. Glanville J, Kuo TC, von Budingen HC, Guey M, Macintyre EA (2003) Design and standard-
L, Berka J, Sundar PD, Huerta G, Mehta GR, ization of PCR primers and protocols for
Oksenberg JR, Hauser SL, Cox DR, Rajpal A, detection of clonal immunoglobulin and T-cell
Pons J (2011) Naive antibody gene-segment receptor gene recombinations in suspect lym-
frequencies are heritable and unaltered by phoproliferations: report of the BIOMED-2
chronic lymphocyte ablation. Proc Natl Acad Concerted Action BMH4-CT98-3936.
Sci U S A 108(50):20066–20071, doi: Leukemia 17(12):2257–2317. doi:10.1038/
1107498108 [pii]10.1073/pnas.1107498108 sj.leu.2403202
14. Liao HX, Lynch R, Zhou T, Gao F, Alam SM, 18. Tobisawa Y, Maruyama T, Tanikawa T,
Boyd SD, Fire AZ, Roskin KM, Schramm CA, Nakanishi K, Kurohane K, Imai Y (2011)
Zhang Z, Zhu J, Shapiro L, Becker J, Benjamin Establishment of recombinant hybrid-IgG/IgA
B, Blakesley R, Bouffard G, Brooks S, Coleman immunoglobulin specific for Shiga toxin. Scand
H, Dekhtyar M, Gregory M, Guan X, Gupta J, J Immunol 74(6):574–584. doi:10.1111/j.
Han J, Hargrove A, Ho SL, Johnson T, Legaspi 1365-3083.2011.02617.x
R, Lovett S, Maduro Q, Masiello C, Maskeri B, 19. Weinstein JA, Jiang N, White RA 3rd, Fisher
McDowell J, Montemayor C, Mullikin J, Park DS, Quake SR (2009) High-throughput
M, Riebow N, Schandler K, Schmidt B, Sison sequencing of the zebrafish antibody repertoire.
C, Stantripop M, Thomas J, Thomas P, Science 324(5928):807–810, doi:
Vemulapalli M, Young A, Mullikin JC, 324/5928/807 [pii]10.1126/science.11700
Gnanakaran S, Hraber P, Wiehe K, Kelsoe G, 20. Wu X, Zhou T, Zhu J, Zhang B, Georgiev I,
Yang G, Xia SM, Montefiori DC, Parks R, Wang C, Chen X, Longo NS, Louder M,
Lloyd KE, Scearce RM, Soderberg KA, Cohen McKee K, O’Dell S, Perfetto S, Schmidt SD,
M, Kamanga G, Louder MK, Tran LM, Chen Shi W, Wu L, Yang Y, Yang ZY, Yang Z, Zhang
Y, Cai F, Chen S, Moquin S, Du X, Joyce MG, Z, Bonsignori M, Crump JA, Kapiga SH, Sam
Srivatsan S, Zhang B, Zheng A, Shaw GM, NE, Haynes BF, Simek M, Burton DR, Koff
Hahn BH, Kepler TB, Korber BT, Kwong PD, WC, Doria-Rose NA, Connors M, Mullikin
Mascola JR, Haynes BF (2013) Co-evolution JC, Nabel GJ, Roederer M, Shapiro L, Kwong
of a broadly neutralizing HIV-1 antibody and PD, Mascola JR (2011) Focused evolution of
founder virus. Nature 496(7446):469–476. HIV-1 neutralizing antibodies revealed by
doi:10.1038/nature12053 structures and deep sequencing. Science
15. Liao HX, Chen X, Munshaw S, Zhang R, 333(6049):1593–1602, doi: science.1207532
Marshall DJ, Vandergrift N, Whitesides JF, Lu [pii]10.1126/science.1207532
X, Yu JS, Hwang KK, Gao F, Markowitz M, 21. Robins H, Desmarais C, Matthis J, Livingston
Heath SL, Bar KJ, Goepfert PA, Montefiori R, Andriesen J, Reijonen H, Carlson C,
DC, Shaw GC, Alam SM, Margolis DM, Nepom G, Yee C, Cerosaletti K (2011) Ultra-
Denny TN, Boyd SD, Marshal E, Egholm M, sensitive detection of rare T cell clones. J
Simen BB, Hanczaruk B, Fire AZ, Voss G, Immunol Methods 375(1-2):14–19, doi:
Kelsoe G, Tomaras GD, Moody MA, Kepler S0022-1759(11)00246-8 [pii]10.1016/j.
TB, Haynes BF (2011) Initial antibodies bind- jim.2011.09.001
ing to HIV-1 gp41 in acutely infected subjects
are polyreactive and highly mutated. J Exp 22. Brochet X, Lefranc MP, Giudicelli V (2008)
Med 208(11):2237–2249. doi:10.1084/ IMGT/V-QUEST: the highly customized and
jem.20110363 integrated system for IG and TR standardized
V-J and V-D-J sequence analysis. Nucleic Acids
16. Scheid JF, Mouquet H, Feldhahn N, Seaman Res 36(Web Server issue):503–508.
MS, Velinzon K, Pietzsch J, Ott RG, Anthony doi:10.1093/nar/gkn316
RM, Zebroski H, Hurley A, Phogat A,
Chakrabarti B, Li Y, Connors M, Pereyra F, 23. Giudicelli V, Brochet X, Lefranc MP (2011)
Walker BD, Wardemann H, Ho D, Wyatt RT, IMGT/V-QUEST: IMGT standardized analy-
Mascola JR, Ravetch JV, Nussenzweig MC sis of the immunoglobulin (IG) and T cell
Antibody Gene Repertoire Sequencing 233
Abstract
The rapid development of deep sequencing technologies over the last few years and concomitant increases
in sequencing depth and cost efficiencies have opened the door to a ever-widening range of applications in
biology—from whole-genome sequencing, to ChIP-seq analysis, epigenomic and RNA transcriptome sur-
veys. Here we describe the application of deep sequencing to the discovery of novel microRNAs and
characterization of their differential expression during adulthood in Caenorhabditis elegans.
Key words Caenorhabditis elegans, microRNA, Differential gene expression, Deep sequencing
1 Introduction
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_18, © Springer Science+Business Media New York 2015
235
236 Alexandre de Lencastre and Frank Slack
2 Materials
3 Methods
3.1 RNA Isolation Routine C. elegans culture procedures are carried using standard,
from Synchronized published protocols [16].
Populations of Adult
1. Allow animals to grow for at least three generations on E. coli
C. elegans
strain OP50 without starvation. In order to obtain 15–20 μg
of RNA, a mixed population of animals (medium density pop-
ulation, unstarved, on a NGM plate) are harvested from 10 to
20 small (6 cm) NGM plates or 1–2 large (15 cm) NGM plates
using 10–15 ml of M9 buffer (see Note 1). The worm pellets
238 Alexandre de Lencastre and Frank Slack
3.2 Analysis Using 1. Collect sequencing data and ensure that it is in a format that is
miRDeep2 compatible with miRDeep2 [9]. Typically, sequencing data
from Illumina will be in fastq format, which is compatible with
miRDeep2. However, if necessary (see miRDeep2 documenta-
tion for information on compatible input formats), convert the
sequencing file into an acceptable input format using any num-
ber of appropriate tools—one option is the online web server,
Galaxy [19].
2. To facilitate sequence alignment, obtain the C. elegans genome
from Wormbase and build an index of it using Bowtie and the
Bowtie-build command included in miRDeep2: bowtie-build
cel_genome.fa cel_genome
This will generate several .ebwt files which miRDeep2
will use.
3. Create a text file, config.txt, containing the filenames of your
samples, and a chosen sample ID (three-letter format), with
one sample per line, in the general format:
Fig. 1 Analysis of deep sequencing reads of known miRNAs using miRDeep2. Frequency diagram, pileup of
reads, identification of mismatches (mm), 2° structure of precursor, and mature sequence highlighted in red
Fig. 2 Identification of novel miRNAs using miRDeep2. Frequency diagram, 2° structure of putative precursor,
and identification of potential mature and star sequences from deep sequencing data
3.3 Validation For validation by qRT-PCR, extract total RNA as described before
of miRNA Expression (Subheading 3.1, step 4). To confirm expression changes of known
miRNAs during aging, RNA should be obtained from synchro-
nized animals at relevant time-points during adulthood. To vali-
date novel miRNAs, RNA should be obtained from synchronized
wild-type N2 animals as well as alg-1(gk214) mutants in young
adulthood and/or later time points (according to its expression
pattern from deep-sequencing) (see Note 11).
1. Northern analysis of known miRNAs: For confirmation of the
expression of known miRNAs, we use ~15–20 μg of total RNA
microRNAs in Caenorhabditis elegans 243
4 Notes
Acknowledgement
References
1. Bartel DP (2009) MicroRNAs: target recognition 12. Wang L, Feng Z, Wang X, Wang X, Zhang X
and regulatory functions. Cell 136:215–233 (2010) DEGseq: an R package for identifying
2. Esquela-Kerscher A, Slack FJ (2006) differentially expressed genes from RNA-seq
Oncomirs—microRNAs with a role in cancer. data. Bioinformatics 26:136–138
Nat Rev Cancer 6:259–269 13. Anders S, Huber W (2010) Differential expres-
3. Boehm M, Slack F (2005) A developmental sion analysis for sequence count data. Genome
timing microRNA and its target regulate life Biol 11:R106
span in C. elegans. Science 310:1954–1957 14. Li J, Tibshirani R (2013) Finding consistent
4. de Lencastre A et al (2010) MicroRNAs both patterns: a nonparametric approach for identi-
promote and antagonize longevity in C. ele- fying differential expression in RNA-Seq data.
gans. Curr Biol 20:2159–2168 Stat Methods Med Res 22(5):519–536
5. Brenner JL, Jasiewicz KL, Fahley AF, Kemp 15. Hardcastle TJ, Kelly KA (2010) baySeq: empir-
BJ, Abbott AL (2010) Loss of individual ical Bayesian methods for identifying differen-
microRNAs causes mutant phenotypes in sen- tial expression in sequence count data. BMC
sitized genetic backgrounds in C. elegans. Curr Bioinformatics 11:422
Biol 20:1321–1325 16. Brenner S (1974) The genetics of Caenor
6. Alvarez-Saavedra E, Horvitz HR (2010) Many habditis elegans. Genetics 77:71–94
families of C. elegans microRNAs are not 17. Lau NC, Lim LP, Weinstein EG, Bartel DP
essential for development or viability. Curr Biol (2001) An abundant class of tiny RNAs with
20:367–373 probable regulatory roles in Caenorhabditis
7. Miska EA et al (2007) Most Caenorhabditis ele- elegans. Science 294:858–862
gans microRNAs are individually not essential for 18. Hafner M et al (2012) Barcoded cDNA library
development or viability. PLoS Genet 3, e215 preparation for small RNA profiling by
8. Kato M, Chen X, Inukai S, Zhao H, Slack FJ next-generation sequencing. Methods 58:
(2011) Age-associated changes in expression of 164–170
small, noncoding RNAs, including microR- 19. Goecks J, Nekrutenko A, Taylor J, Team G
NAs, in C. elegans. RNA 17:1804–1820 (2010) Galaxy: a comprehensive approach for
9. Friedländer MR, Mackowiak SD, Li N, Chen supporting accessible, reproducible, and trans-
W, Rajewsky N (2012) miRDeep2 accurately parent computational research in the life sci-
identifies known and hundreds of novel ences. Genome Biol 11:R86
microRNA genes in seven animal clades. 20. Lim LP et al (2003) The microRNAs of
Nucleic Acids Res 40:37–52 Caenorhabditis elegans. Genes Dev 17:
10. Robinson MD, McCarthy DJ, Smyth GK 991–1008
(2010) edgeR: a Bioconductor package for dif- 21. Kato M, de Lencastre A, Pincus Z, Slack FJ
ferential expression analysis of digital gene (2009) Dynamic expression of small non-
expression data. Bioinformatics 26:139–140 coding RNAs, including novel microRNAs
11. Hausser J et al (2009) MirZ: an integrated and piRNAs/21U-RNAs, during Caenor
microRNA expression atlas and target prediction habditis elegans development. Genome Biol
resource. Nucleic Acids Res 37:W266–W272 10:R54
248 Alexandre de Lencastre and Frank Slack
22. Hofig KP, Feller A, Merz H (2007) New appli- by high-throughput sequencing. RNA 15:
cation for the LightCycler 480 system: qPCR- 992–1002
based microRNA-profiling. Biochemica:7–9 27. Farazi TA et al (2011) MicroRNA sequence
23. Pak J, Fire A (2007) Distinct populations of and expression analysis in breast tumors by
primary and secondary effectors during RNAi deep sequencing. Cancer Res 71:4443–4453
in C. elegans. Science 315:241–244 28. Grishok A et al (2001) Genes and mechanisms
24. Hafner M et al (2008) Identification of microR- related to RNA interference regulate expression
NAs and other small regulatory RNAs using of the small temporal RNAs that control C.
cDNA library sequencing. Methods 44:3–12 elegans developmental timing. Cell 106:23–34
25. Hafner M et al (2011) RNA-ligase-dependent 29. Hoogewijs D, Houthoofd K, Matthijssens F,
biases in miRNA representation in deep- Vandesompele J, Vanfleteren JR (2008)
sequenced small RNA cDNA libraries. RNA Selection and validation of a set of reliable
17:1697–1712 reference genes for quantitative sod gene
26. Fahlgren N et al (2009) Computational and expression analysis in C. elegans. BMC Mol
analytical framework for small RNA profiling Biol 9:9
Chapter 19
Abstract
Pyrosequencing is a technique that uses a sequencing-by-synthesis system which is designed to quantify
single-nucleotide polymorphisms (SNPs). Artificial C/T SNP creation via bisulfite modification permits
measurement of DNA methylation locally and globally in real time. Alteration in DNA methylation has
been implicated in aging, as well as aging-related conditions such as cancer, as well as cardiovascular,
neurodegenerative, and autoimmune diseases. Considering its ubiquitous presence in divergent clinical
pathologies, quantitative analysis of DNA CpG methylation both globally and at individual genes helps to
elucidate the regulation of genes involved in pathophysiological conditions. The ability to detect and
quantify the methylation pattern of DNA has the potential to serve as an early detection marker and poten-
tial drug target for several diseases. Here, we provide a detailed technical protocol for pyrosequencing
supplemented by critical information about assay design and nuances of the system that provides a strong
foundation for beginners in the field.
Key words Pyrosequencing technique, DNA CpG methylation, Global methylation, Biomarker
detection, SNPs, Bisulfide conversation, Assay design
1 Introduction
*
These authors made equal contributions to this work.
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4_19, © Springer Science+Business Media New York 2015
249
250 Colin Delaney et al.
Fig. 2 Deamination of cytosine via sodium bisulfide conversion. (a) Deamination of cytosine to uracil is pre-
vented by methylation of the 5-carbon position of cytosine. (b) Methylated (above) and unmethylated (below)
CpG-containing DNA undergoes bisulfite conversion. Methylated cytosines are unchanged while unmethylated
cytosines are converted to uracil. Following PCR the cytosine is retained while uracil is converted to thymine.
*C denotes methylated cytosine. Pyrimidines involved in bisulfite conversion are bolded. NaHSO3 sodium
bisulfite, PCR polymerase chain reaction
Fig. 3 Enzyme cascade system in pyrosequencing. An ssDNA template is first hybridized with the sequencing
primer and mixed with enzymes (written in italics) and two substrates (APS and luciferin). After successful
incorporation of a nucleotide by DNA polymerase into a growing DNA strands, the released PPi reacts with APS
in the presence of ATP sulfurylase giving rise to ATP. ATP in the presence of substrate luciferin and enzyme
luciferase produces oxyluciferin that generates visible light, which can be detected by inbuilt CCD camera. Any
unincorporated nucleotides and ATP are degraded into its building blocks by enzyme apyrase prior to the next
nucleotide dispensation. Cascade reactions repeat for every dispensation. ATP adenosine triphosphate, APS
Adenosine 5′ phosphosulfate, PPi pyrophosphate
1.3 Technical Genomic DNA is bisulfite converted, and then the region of inter-
Overview est is amplified via PCR. Incorporation of a single biotinylated
of Pyrosequencing PCR primer allows separation of the two strands of the amplicon
to create a ssDNA template for annealing of a pyrosequencing
primer and extension of the complementary strand by discrete dis-
pensation of nucleotides. This platform is PCR-based, yields rapid
results (i.e., within a single day if starting with PCR), and is highly
quantitative. Many different assays can be performed simultane-
ously (i.e., in a 96-well format, 96 different assays could be per-
formed on one sample, 96 samples could be analyzed with one
assay, or anything in between) and the time required is entirely
dependent on the number of dispensations needed to cover the
region of interest (approximately 5 min + 1 min/dispensation). In
addition, pyrosequencing technology can be used to interrogate
regulatory elements of specific genes [12, 14–16] or as a means of
estimating global methylation [17–19]. However, pyrosequencing
assays can be more difficult to design and extensive optimization
of these assays is required (see Subheading 4). Also, an emerging
pitfall of the system is that Bisulfite modification cannot discrimi-
nate between 5-methylcytosine and the novel modification
5-hydroxymethylcytosine. Nevertheless, pyrosequencing is a vali-
dated means of estimating both global methylation and specific
regulatory loci in mammalian samples (see Note 6).
2 Materials
3 Methods
plate from the shaker, place the probes into the plate and watch
to make sure all fluid is sucked out of the wells. Delay will give
beads time to settle out of solution, which will lower your
recovery of amplicon.
5. Transfer probes to the tray containing 70 % EtOH. Once fluid
is observed passing through the vacuum hose, count to 10 s to
wash away residual salts and unlabeled DNA.
6. Transfer probes to the tray containing denaturation buffer and
count to 10 as above. Amplicons are denatured and the unla-
beled DNA strands are removed from the sample, leaving a
ssDNA pyrosequencing template strand.
7. Transfer probes to the tray containing 1× wash buffer and
count to 10 to let the fluid drain through. The base in the pre-
vious step is neutralized, allowing the pyrosequencing reaction
to proceed at proper conditions.
256 Colin Delaney et al.
3.4 Preparing Note that these steps can and should be performed when time permits
the Pyrosequencer in the above protocol to eliminate unwanted lag between preparation
of the sample and running the pyrosequencing reaction. It is advis-
able to input the assay into the software prior to strand separation.
1. Create a new assay within a folder in the CpG assay folder by
right-clicking the folder and selecting New Assay. Enter the
sequence to analyze in the appropriate field and click
“Generate Dispensation Order.” A theoretical program is dis-
played (see Note 3).
2. Turn on machine. It will take about 2 min to warm up and
establish communication with the computer. Wait for the info
light to begin blinking as an indicator of successful data
exchange.
3. Open PyroCpG software.
4. Under the CpG run folder, select the appropriate subfolder (if
applicable), right-click, and select “New Run.” Name the run.
5. Enter the relevant information (sample ID, assay, notes about
experiment, and type of dispensing tips used) into the PyroCpG
software for each well you are using for this run. Save often to
prevent data loss if software crashes before run commences.
6. Once all wells and assays are entered, select Volume Information
from the drop down Tools menu. Note the amount of enzyme,
substrate, and nucleotides to add to the appropriate dispensing
CpG Methylation Detection Via Pyrosequencing 257
3.5 Run 1. Once the plate has cooled to RT, open the process chamber
the Pyrosequencing door using the software, insert the plate in the correct orienta-
Reaction tion, and close the process chamber lid using the software.
2. Click “Run.” The length of the run is determined only by the
number of dispensations in the longest assay selected and is not
dependent on the number of wells used.
3.6 Cleanup 1. Immediately after all runs are finished, clean out dispensing tips
to prevent salt buildup and tip blockage. RDTs and CDTs can be
“milked”; that is, rinse and fill them with water, and then apply
pressure to squeeze water in a stream through the tip. NEVER
milk NDTs as the bore size of the tip is too small; rather, gently
258 Colin Delaney et al.
rinse the tip inside and out with water. Store tips upright in a
5 mL tube rack and avoid contact with the delicate tips, which
are easily bent/damaged.
2. Rinse out plastic trays from the vacuum prep station and allow
to dry.
3.7 Data Analysis While the pyrosequencer is running, the light trace for each well
detected by the camera is presented in real time, generating a pyro-
gram of peaks, the height of which indicate the stoichiometric
incorporation of nucleotides. Each non CpG peak becomes a
reference peak that the software uses to calculate the percent meth-
ylation of the sample (Fig. 5). However, quantitative analysis
cannot be performed until the run is finished.
Fig. 5 Representative pyrograms showing hypomethylation of the B1 element following 5-azacytidine treat-
ment. (a) Theoretical pyrogram generated by analytical software based on the input sequence to analyze for
B1 element pyrosequencing primer 2 (see Table 1). (b) Pyrograms of DNA isolated from T cells cultured in the
absence (control) or presence of cytosine analog 5-azacytidine, a known hypomethylating agent. The B1
“sequence to analyze 2” is shown bolded. Grey shaded areas indicate CpG sites, tan shaded bars indicate
bisulfite control dispensations. Percent methylation is indicated above each CpG site
CpG Methylation Detection Via Pyrosequencing 259
4 Notes
Table 1
Primers and reaction conditions for the analyses of LINE1 (human) and B1 (mouse) elements
Line 1 (Human)
PCR primer (F): TTTTGAGTTAGGTGTGTGGGATATA Pyrosequencing primer (F):
PCR primer (R): biotin-AAAATCAAAAAATTCCCTTTC AGTTAGGTGTGGGATATAGT
Reaction conditions: 95 °C for 5 min; (95 °C 30 s, 50 °C 30 s, 72 °C 30 s) × 45 cycles; 72 °C for 5 min
Sequence to analyze: TTYGTGGTGYGTYGTTTTTTAAGTYGGTTTGAAAAGYGTA
B1 element (Mouse)
PCR primer (F): TGGTGGTGGTGGTTGAGAT Pyrosequencing primer 1 (F):
TGGTGGTGGTTGAGAT
PCR primer (R): Pyrosequencing primer 1 (R):
biotin-AATAACACACACCTTTAATCCCAA TTTGTAGATTAGGTTGGTTT
Reaction conditions: 95 °C for 15 min; (95 °C 30 s, 63 °C 30 s, 72 °C 30 s) × 45 cycles; 72 °C for
10 min
Sequence to analyze 1: AGYGTTTTTTTGTGTAGTTTTGGTTATTTTGGAATTTATTTTGTAGA
TTAGGTTGGTTTYGAATTT
Sequence to analyze 2: YGAATTTAGAAATTYGTTTGTTTTTGTTTTTRTTTTYGGAG
5 Conclusion
Acknowledgement
References
1. Portela A, Esteller M (2010) Epigenetic modi- 8. Lund G, Andersson L, Lauria M et al (2004)
fications and human disease. Nat Biotechnol DNA methylation polymorphisms precede any
28:1057–1068 histological sign of atherosclerosis in mice
2. Eden A, Gaudet F, Waghmare A et al (2003) lacking apolipoprotein E. J Biol Chem 279:
Chromosomal instability and tumors promoted 29147–29154
by DNA hypomethylation. Science 300:455 9. Jones PA, Baylin SB (2007) The epigenomics
3. Vera E, Canela A, Fraga MF et al (2008) of cancer. Cell 128:683–692
Epigenetic regulation of telomeres in human 10. Warnecke PM, Bestor TH (2000) Cytosine
cancer. Oncogene 27:6817–6833 methylation and human cancer. Curr Opin
4. Jaenisch R, Bird A (2003) Epigenetic regula- Oncol 12:68–73
tion of gene expression: how the genome inte- 11. Urdinguio RG, Sanchez-Mut JV, Esteller M
grates intrinsic and environmental signals. Nat (2009) Epigenetic mechanisms in neurological
Genet 33 Suppl:245–254 diseases: genes, syndromes, and therapies.
5. Ballestar E, Esteller M, Richardson BC (2006) Lancet Neurol 8:1056–1072
The epigenetic face of systemic lupus erythe- 12. Strickland FM, Hewagama A, Lu Q et al
matosus. J Immunol 176:7143–7147 (2012) Environmental exposure, estrogen and
6. Brooks WH, Le Dantec C, Pers JO et al (2010) two X chromosomes are required for disease
Epigenetics and autoimmunity. J Autoimmun development in an epigenetic model of lupus.
34:J207–J219 J Autoimmun 38:J135–J143
7. Kim M, Long TI, Arakawa K et al (2010) DNA 13. Ronaghi M, Uhlen M, Nyren P (1998) A
methylation as a biomarker for cardiovascular sequencing method based on real-time pyro-
disease risk. PLoS One 5, e9692 phosphate. Science 281:363–365
264 Colin Delaney et al.
14. Polansky JK, Kretschmer K, Freyer J et al and function in f1 mice. J Nutr 142:
(2008) DNA methylation controls Foxp3 gene 1329–1335
expression. Eur J Immunol 38:1654–1663 20. Royo JL, Pascual MH, Salinas A et al (2006)
15. Floess S, Freyer J, Siewert C et al (2007) Pyrosequencing protocol requiring a unique
Epigenetic control of the foxp3 locus in regula- biotinylated primer. Clin Chem Lab Med 44:
tory T cells. PLoS Biol 5, e38 435–441
16. Schoenborn JR, Dorschner MO, Sekimata M 21. Yang AS, Doshi KD, Choi SW et al (2006)
et al (2007) Comprehensive epigenetic profiling DNA methylation changes after 5-aza-2′-
identifies multiple distal regulatory elements deoxycytidine therapy in patients with leukemia.
directing transcription of the gene encoding Cancer Res 66:5495–5503
interferon-gamma. Nat Immunol 8:732–742 22. Bollati V, Baccarelli A, Hou L et al (2007)
17. Yang AS, Estecio MR, Doshi K et al (2004) A Changes in DNA methylation patterns in
simple method for estimating global DNA subjects exposed to low-dose benzene. Cancer
methylation using bisulfite PCR of repetitive Res 67:876–880
DNA elements. Nucleic Acids Res 32, e38 23. Bollati V, Schwartz J, Wright R et al (2009)
18. Jeong KS, Lee S (2005) Estimating the total Decline in genomic DNA methylation through
mouse DNA methylation according to the B1 aging in a cohort of elderly subjects. Mech
repetitive elements. Biochem Biophys Res Ageing Dev 130:234–239
Commun 335:1211–1216 24. Baccarelli A, Wright RO, Bollati V et al (2009)
19. Delaney C, Hoeltzel M, Garg SK et al (2012) Rapid DNA methylation changes after expo-
Maternal micronutrient supplementation sure to traffic particles. Am J Respir Crit Care
suppresses T cell chemokine receptor expression Med 179:572–578
INDEX
Albert C. Shaw (ed.), Immunosenecence: Methods and Protocols, Methods in Molecular Biology, vol. 1343,
DOI 10.1007/978-1-4939-2963-4, © Springer Science+Business Media New York 2015
265
IMMUNOSENECENCE: METHODS AND PROTOCOLS
266 Index
CD45RA ................................................... 84, 116, 117, 119 Cryopreserved ........................ 14, 17, 88, 111, 113, 182, 188
CD45RO......................................................................... 116 CXCL12 ........................................................................... 23
CD49 ................................................................................ 22 CXCR4 ................................................................. 21–23, 29
CD56 ............................................... 9, 11, 14, 15, 16, 69, 99 Cysteine ................................................................... 155, 156
CD57 ..................................................... 9, 10, 13, 14, 15, 17 CyTOF........................................................................ 82–84
CD62L ...................................................................... 88, 116 Cytokine
CD68 ................................................................................ 22 inteferon-γ (IFN-γ) ............................... 10, 12, 116, 121
CD80 .......................................................................... 21–23 interferon-α (IFN-α) ................................................... 67
CD81 ................................................................................ 22 interleukin-6 (IL-6)............................................... 23, 72
CD85j.......................................................................... 12, 14 tumor necrosis factor-α (TNF-α) ................................ 24
CD86 .......................................................................... 21–23 Cytomegalovirus (CMV) .......................... 10, 116, 182, 183
CD94 .............................................................. 10, 12, 13, 14 Cytosine ................... 107, 249–251, 253, 254, 258, 259–262
CD95 .............................................................. 68, 73, 76, 80 Cytotoxic ........................................ 9, 10, 12, 14–16, 23, 116
CD105............................................................................... 22
CD107a ........................................................... 10, 12, 14, 17 D
CD108W .......................................................................... 24 Data dual calibration ............................................. 83, 88, 90
CD112......................................................................... 10, 85 Decorin........................................................................ 22, 24
CD115............................................................................... 22 Deep sequencing ...................... 236–237, 239–242, 244–246
CD123................................................................... 69, 72, 99 Degranulation........................................................ 10, 12, 14
CD127....................................................... 74, 116, 117, 119 Dendritic cell (DC)
CD127 (IL-7 receptor) ............................................. 74, 116 myeloid DC ........................................................... 69, 70
CD155............................................................................... 10 plasmacytoid DC ............................................. 69, 70, 78
CD163......................................................................... 21, 22 Dermis ......................................................................... 37–39
CD164............................................................................... 22 Detergent extraction ............................................................ 2
CD172a ............................................................................. 22 Detergent-resistant membrane (DRM)............................... 2
CD206............................................................................... 22 4, 4’-Dianilino-1,1’-binapthyl-5,5’-disulfonic
CD209............................................................................... 22 acid ........................................................ 156, 159
Cell aggregates ............................................................ 82, 83 Differential gene expression ............................................ 236
Cell cycle ............................................................. 74, 88, 175 Differentiation .................................... 10, 21–23, 73, 88, 249
Cell length ................................................................... 82, 83 Digital flow cytometry................................................. 53–64
Cellular stress ............................................................ 10, 143 Digital imaging.................................................................. 98
Central memory CD8+ T cell ......................................... 117 Dinitrophenyl hydrazine (DNPH) .................................. 156
Centrifugation .............................. 1–6, 68, 78, 117, 148, 182 Disulfide .................................................................. 155–172
Chelator............................................................................. 82 Diversity ............ 181, 182, 195, 199, 200, 208, 219, 220, 229
Chemokine ..................................... 1, 21–23, 25, 35, 73, 116 DNA damage response (DDR) ....................... 175–180, 235
Chloroquine .................................................................... 151 DNAM-1 .............................................................. 10, 12–14
Cisplatin ............................................................................ 82 DNA methyltransferases (DNMTs)................................ 249
Class switch recombination (CSR).......................... 107, 200 DNA polymerase.............................. 183, 191, 192, 250, 251
Clonality score ......................................................... 228, 229 Dy164................................................................................ 84
Clonotype ........................................................................ 182
Coalescence ......................................................................... 2 E
Collagen ...................................................................... 21, 29
E47 .................................................................................. 107
Collagen-Iα (Col-Iα) ........................................................ 23
Effector memory CD8+ T cell ................................ 116, 117
Compensation ................ 28, 53, 54, 61–63, 82, 86, 103, 113
Electrophoresis ............................ 5, 161–162, 171, 183–187,
Compensation matrix .................................................. 61, 63
192, 196, 201, 202, 208, 209, 222–225, 238, 253
Complementarity-determining region 3
Endotoxin ............................................................................ 3
(CDR3) .......... 181, 182, 194, 212, 220, 227, 228
Endotoxin-free .................................................................... 3
Complementarity determining region 3 heavy chain
Eotaxin .............................................................................. 22
(CDR-H3) ........................................... 181, 200,
Epidermis .................................................................... 36–39
201–206, 211, 212, 213, 220
Epigenetic ....................................................................... 249
Concanavalin A ....................................................... 124, 125
Epitope ...................................................... 89, 181, 182, 190
Contracture ....................................................................... 39
Excisional wound injury ............................ 36, 37, 40, 43–45
CpG methylation ............................................................ 260
Exhausted memory B cell ................................................ 113
CpG oligodeoxynucleotide ............................. 108–111, 113
Exonuclease ............................................................. 220, 227
IMMUNOSENECENCE: METHODS AND PROTOCOLS
Index
267
Extracellular matrix (ECM) ................. 20–22, 24, 25, 30, 36 HLA-DR .............................................................. 67, 70, 99
Extracellular signal-related kinase (Erk)............................ 22 Homologous recombination repair .................................. 175
Hyaluronan.................................................................. 22, 24
F Hydrophobicity ....................................................... 156, 212
Fcγ receptor ................................................................. 21, 22 5-Hydroxymethylcytosine ............................................... 252
Fibroblast ........................................................ 19, 21, 24, 36 Hypodermis ................................................................. 37–39
Fibroblast activation protein (FAP) ................................... 22
Fibrocyte ..................................................................... 19–30 I
Fibronectin ........................................................................ 22 Idiopathic pulmonary fibrosis ...................................... 20, 23
Fibrosis ................................................. 19, 20, 23, 24, 25, 39 IFN-γ ................................................................................ 15
Flagellin........................................................... 100, 102, 105 IFN-γ-IL-10 ratio................................................... 121, 122
Flotillin................................................................................ 6 IgA ................................................... 113, 205, 206, 211, 214
Flow cytometry...................................... 9–18, 21, 26–29, 41, IgD ...................................................................... 68, 73, 113
47, 53–82, 87, 89, 98, 99, 108, 109, 110, 113, IgG ................ 21, 25, 113, 117, 118, 205, 206, 211, 214, 215
115–119, 181, 182, 203 IgM memory B cell ......................................................... 113
Fluorescein-5-thiosemicarbazide (FTC)......... 160, 167, 169 IL-4 ..................................................................... 22, 23, 116
Fluorescence recovery after photobleaching IL-17 ......................................................................... 67, 116
(FRAP) ...................................................... 97, 98 IL-7-receptor-alpha (CD127) .............. 68, 74, 116, 117, 119
Fluorescence resonance energy transfer Imiquimod....................................................... 66, 69, 70, 72
(FRET) ...................................................... 97, 98 Immunoblotting .............................................................. 2, 6
Fluorescent in situ hybridization Immunoglobulin
(FISH) ................................... 176–177, 190–191 constant region .......................................................... 204
Fluorochrome diversity region .................................................. 199, 220
Alexa-700 .................................................................... 54 framework region....................................................... 221
APC (allophycocyanin) ......................................... 54, 60 heavy chain ........................................................ 199, 219
FITC (fluorescein isothiocyanate) ......................... 54, 56 isotype ............................................... 204, 206, 219, 220
PE (phycoerythrin) ........................................ 54, 56, 105 joining region............................................................. 200
Forward scatter ................... 14, 15, 27, 28, 30, 56, 57, 70, 82 leader sequence .......................................................... 221
Foxp3 ................................................................................. 74 light chain
G kappa ........................................................... 200, 206
lambda ......................................................... 200, 206
Gene rearrangement ................................ 220, 221, 223–226 variable region ........................................................... 214
Global methylation .................................................. 252, 262 Immunological synapse ....................................................... 2
Glutathione ..................................................................... 155 Immunome ...................................................................... 219
Glycerophospholipids .......................................................... 2 Immunoprecipitation ........................................................... 5
Gr1 .................................................................................... 21 Immunoreceptor tyrosine-based inhibitory
Gradient ......................................... 1–6, 11, 12, 89, 100, 182 motif (ITIM) ................................................... 22
Granulocyte-macrophage colony stimulating factor Immunostaining .............................................. 144, 146–150
(GM-CSF) ...................................................... 24 In115 ................................................................................. 85
Granzyme A ...................................................... 14, 130–132 Indoleamine oxidase .......................................................... 24
Granzyme B ...................................................... 14, 121–140 Influenza virus .................. 121, 122–123, 124, 127, 128, 135
Inhibitors ........................................... 2–6, 17, 109, 112, 145,
H
146, 147, 150–152, 158, 159, 171, 183, 188, 195,
Hairpins .......................................................... 240, 242, 246 201, 203, 237
γ-H2AX .......................................................... 176, 177, 179 Innate lymphoid cells .......................................................... 9
Heat shock .......................................................................... 3 Intercalator .................................................................. 82, 83
Heavy metal ions ............................................................... 81 iridium ................................................................... 92, 94
Hemagglutination inhibition assay .......................... 108, 109 Intercellular adhesion molecule-1 (ICAM-1) ............. 22, 23
Hemagglutination units................................... 122, 135, 136 Interferon-γ ............................................................... 24, 121
Heparin .......................................... 11, 17, 26, 116, 118, 182 Interferon-γ (IFNγ)................................................... 24, 134
Hepatocyte growth factor (HGF) ..................................... 24 Interleukin-2 (IL-2) .............................................. 11, 67, 78
High-throughput DNA sequencing ........................ 219–230 Interleukin-4 (IL-4) ............................................ 22, 23, 116
High throughput sequencing.......................... 182, 201–204, Interleukin-6 (IL-6) .............................................. 23, 67, 72
206–212, 213, 220, 221, 222, 228, 229, 230 Interleukin-8 (IL-8) .................................................... 23, 24
IMMUNOSENECENCE: METHODS AND PROTOCOLS
268 Index
O Q
Oxidation .................................................... 85–87, 155–157 Quantitative PCR ........................................................... 246
Oxidative stress................................................ 156, 157, 163 Quantitative RT-PCR ..................................................... 243
Quantum dots (Qdots) ...................................................... 85
P
R
Panniculus carnosus ........................................................... 37
Paranitroanalide............................................... 122, 125, 136 5’ Rapid amplification of cDNA ends (5'RACE) ............ 221
P53 binding protein-1 (53BP1)....................... 176, 177, 179 Receptor editing .............................................................. 220
PBMC. See Peripheral blood mononuclear cell (PBMC) Receptors .................................. 1, 2, 9, 10, 12, 13, 21, 22, 23,
PCR. See Polymerase chain reaction (PCR) 66, 69, 71, 73, 74, 116, 181–196, 219, 220, 226
PD-1 ................................................................................. 88 Remodeling phase ....................................................... 35, 36
Pellet .................................................. 4, 6, 12, 13, 16, 17, 26, Repertoire ......................... 181–196, 199–217, 219–230, 235
48, 69, 78, 100, 101, 129, 137, 148, 149, 164, 165, Retrovirus ........................................................................ 178
167, 168, 172, 237, 238, 244 Rhesus macaque .............................................. 66–69, 73, 74
Pentraxin ........................................................................... 22 Rheumatoid arthritis ................................................. 20, 250
Perforin........................................................................ 12, 14
S
Peripheral blood mononuclear cell (PBMC) .............. 12–17,
26–28, 30, 54, 56, 57–64, 68–70, 72, 73, 75, 78, S-adenosyl homocysteine ................................................ 250
79, 81–95, 98, 100, 101, 108, 111, 113, 117–119, S-adenosylmethionine (SAM) ........................................ 250
121, 122–125, 127, 128, 134, 182, 188, 223 Scleroderma ................................................................. 20, 23
Perlecan ............................................................................. 22 Semaphorin 7a (Sema 7a)...................................... 22, 23, 24
Phenotype .................................. 9–18, 23, 30, 115–119, 236 Senescence ........................................................... 65–80, 175
Phorbol 12-myristate 13-acetate (PMA) ........ 15, 79, 89, 90 Senescence associated mutation prone............................... 23
Phosphatidylethanolamine .............................................. 145 Shelterin .......................................................................... 176
Phosphoinositol-3-kinase (PI3 kinase) ............................. 21 Side scatter ...................................................... 14, 15, 27, 28,
Piwi-interacting RNAs (piRNAs) ................................... 244 30, 57, 70, 82, 98, 118, 177, 179
Platelet derived growth factor (PDGF) ............................. 24 Single cell analysis ............................................. 81, 181–196
Platelets ............................................................... 24, 35, 137 Single cell PCR ....................................................... 195, 221
Plexin C1........................................................................... 23 Single nucleotide polymorphisms (SNPs) ....... 250, 260, 263
Pluripotent ........................................................................ 24 α-Smooth muscle actin (α-SMA) ............................... 22, 23
Polymerase chain reaction (PCR) ............................. 50, 108, S-nitrosylated .................................................................. 155
109, 112, 182, 183, 187, 188, 189–193, 195, 196, Somatic hypermutation (SHM) ..................... 107, 200, 220,
200, 201–213, 221–230, 237, 243, 245, 246, 221, 222, 227, 228
250–257, 259–262 Spectral overlap ........................................................... 54, 61
POT1- and TIN2-organizing protein (TPP1) ........ 176, 177 Spectratyping............. 181, 200, 201, 202, 204, 205, 206, 213
Primer....... 109, 112, 182, 183–187, 190–193, 195, 202, 203, Sphingomyelin .................................................................... 2
204, 205–207, 208, 211, 213, 214–216, 221–226, Staphylococcus aureus ........................................................... 45
230, 237, 243, 250, 251–253, 254, 256, 258, 259, S-thiolated ....................................................................... 155
260, 261, 262 Stromal derived factor-1 (SDF-1) ..................................... 23
Proliferative phase ............................................................. 36 Sucrose ......................................... 3–5, 6, 125, 126, 177, 178
Proline ............................................................................. 155 Sulfenic acid .................................................................... 155
Prolyl-4-hydroxylase .......................................................... 22 Sulfinic acid ..................................................................... 155
Protection of telomeres 1 (POT1) ........................... 176, 177 Sulfonic acid .................................................................... 155
Protein .................................. 6, 21, 22, 24, 42, 48, 74, 76, 79, Supernatant ....................................... 4, 6, 12, 13, 16, 26, 48,
125, 127, 129, 131, 132, 133–134, 136, 137, 140, 63, 68–73, 75, 77, 78, 79, 92, 100, 101, 102, 111,
144–149, 151, 152, 155–172, 176, 201, 219, 227 121–123, 127, 128–129, 134, 137, 148, 162, 164,
Pseudomonas aeruginosa ................................................ 45–46 165, 167, 168, 178, 244
IMMUNOSENECENCE: METHODS AND PROTOCOLS
270 Index
Superoxide dismutase ...................................................... 157 Transforming growth factor β1 (TGF-β1) ............ 22, 23, 24
Switched memory B cells ........................................ 107–114 TRF1-interacting protein 2 (TIN2) ................................ 176
Triton X-100 .................... 4, 6, 125, 126, 129, 183, 189, 201
T Tumor necrosis factor (TNF) ...................................... 22, 24
T cell ......................................... 2, 23, 24, 66, 68, 69, 73, 74, Tumor necrosis factor-α (TNF-α) ......................... 12, 14, 15
76, 77–80, 86, 88, 89, 111, 115–119, 143–153,
U
181–196, 219, 226, 249, 258
T cell receptor (TCR) Uracil ................................................ 107, 250, 251, 253, 259
diversity region .......................................................... 181
joining region..................................................... 200, 213 V
TCR alpha .......................... 181, 187, 189, 191, 192, 195 Vaccination
TCR beta.................................... 181, 187, 190, 191, 194 influenza ............................................................ 108, 110
variable region ........................................................... 214 salmonella .................................................................. 108
Tellurium (Te) ................................................................... 85 Streptococcus pneumoniae ............................................. 108
Telomere .................................................................. 175–180 tetanus ....................................................................... 108
Telomere dysfunction induced DNA damage foci Vascular endothelial growth factor (VEGF)...................... 24
(TIFs) .................................................... 176, 177 Vβ frequency analysis ...................................................... 181
Telomeric-repeat-binding factor 1 (TRF1) ..................... 176 Versican ....................................................................... 22, 24
Telomeric-repeat-binding factor 2 (TRF2) ............. 176, 177 Viability ........................................ 12, 13, 16, 17, 82, 99, 125
Tenascin............................................................................. 22 Vimentin ..................................................................... 22, 29
Tetramer .................................................... 75, 182, 183, 188 Vinblastine .............................................................. 146, 151
T-helper 1 ........................................................................ 116
T-helper 2 ........................................................................ 116 W
T-helper 17 ...................................................................... 116
Wound closure, measurement of............................ 41, 46–47
Thiocarbazide .................................................................. 156
Wound contraction ...................................................... 23, 25
Thiol.................................................................. 83, 155, 156
Wound healing ................................................ 35–39, 43, 45
Th1 T cell .......................................................... 22, 116, 151
Wound tissue
Th2 T cell .......................................................... 22, 116, 151
flow cytometry ............................................................. 41
Th17 T cell ...................................................................... 116
formalin fixation .................................................... 42, 48
Thy1.1 ............................................................................... 22
immunofluorescence .............................................. 42, 49
Tm169 ............................................................................... 84
OCT embedding ................................................... 42, 48
Toll-like receptor (TLR) .............. 23, 55, 66, 69, 79, 98, 144
preparation and isolation ....................................... 40, 43
TLR4................................................................... 69, 105
protein isolation ..................................................... 42, 48
TLR7........................................................................... 69
RNA isolation........................................................ 41, 48
Total body surface area (TBSA) .................................. 39, 44
Trafficking ......................................................... 2, 20, 23, 25 X
Transcriptional repressor/activator protein
(RAP1) .......................................................... 176 Xylazine ................................................................. 40, 43, 44