(Methods in Molecular Biology 1323) Rémy Bosselut, Melanie S. Vacchio (Eds.) - T-Cell Development - Methods and Protocols (2016, Humana Press) PDF
(Methods in Molecular Biology 1323) Rémy Bosselut, Melanie S. Vacchio (Eds.) - T-Cell Development - Methods and Protocols (2016, Humana Press) PDF
(Methods in Molecular Biology 1323) Rémy Bosselut, Melanie S. Vacchio (Eds.) - T-Cell Development - Methods and Protocols (2016, Humana Press) PDF
Rémy Bosselut
Melanie S. Vacchio Editors
T-Cell
Development
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
Fifty and a few years since the function of the thymus was discovered, the study of T cell
development remains a fascinating part of immunology. This is certainly because T cells,
essential components of immune responses, develop in the thymus. Interactions in the thy-
mus shape the reactivity and function of T cell precursors and are essential to establish a
functional and tolerant immune system. Thus, identifying intrathymic interactions and
understanding their mechanisms are intrinsic parts of T cell immunology. In addition, the
study of T cell (and more broadly lymphocyte) development has exerted broad appeal
beyond the immunological realm. Indeed, throughout their thymic journey, T cell precur-
sors recapitulate key processes involved in metazoan development, including proliferation,
differentiation, death-survival, and migration decisions. Studies of T cell development have
brought decisive insight into critical concepts of modern biology, including somatic DNA
recombination, DNA repair, programmed cell death, and epigenetic gene silencing.
For all its accomplishments and potential, the study of T cell development enjoys the
not-so-enviable reputation of being “complicated.” While this is in part due to the sheer
complexity of T cell development, it certainly also has to do with difficulties in experi-
mental approaches. The objective of the present volume of Methods in Molecular Biology
is to overcome such practical obstacles, by giving simple and accessible experiment pro-
tocols to explore thymus biology. This book is organized in three parts. The first two
chapters offer short reviews on T cell development, for readers with little or no familiarity
with the topic. Both provide basic concepts as a preparation to parsing the protocol chap-
ters, and references for further study. The second part discusses analysis strategies and
presents basic protocols for cell preparation, flow cytometry analyses, and the study of T
cell responses. The last part includes state-of-the-art protocols to explore multiple aspects
of thymocyte biology.
In building this selection, we have tried to put emphasis on three themes. The first is
the emergence and refinement, over the last few years, of a remarkable panoply of in vitro
approaches to study T cell development. While in vivo studies, harnessing the power of
mouse genetics, remain the standard of proof, new strategies offer unprecedented possibili-
ties to investigate thymic biology in a dish. Such approaches, using derivatives of the OP9
stromal line expressing Notch ligands, have truly revolutionized the study of early thymo-
cyte development. In addition, continued refinement of organ culture systems has increased
their power to address later stages of αβ T cell development, including mechanisms of selec-
tion. The present volume includes detailed protocols for three of these approaches: fetal
thymic organ culture, reconstituted organ culture, and culture of thymic “slices.” Future
progress can be expected from attempts at generating thymic epithelial cells in vitro, includ-
ing using “reprogramming” approaches. An important asset of organ culture is to open the
door to live imaging techniques, as described in the chapter on thymic slices, which incor-
porates time and space resolution into the study of thymocyte differentiation. Last but not
least, because in vitro techniques can combine T cell precursors and stromal cells of distinct
genotypes, they offer useful alternatives to complex breeding strategies.
v
vi Preface
The second point we wanted to emphasize is the plasticity of thymocytes, which can
fine-tune intracellular signaling and gene expression to specific environmental conditions.
This is critical during the building of the TCR repertoire: by shifting the repertoire of TCR
specificities being selected, thymocyte plasticity offers the thymic organ a major potential to
compensate signaling or transcriptional changes at the single-cell level. This emphasizes the
importance of genetic approaches analyzing the development of cells carrying a defined
specificity, together with bone marrow chimera techniques allowing the study of cell devel-
opment in a competitive environment. More broadly, even though there are few truly
single-cell assays for the study of T cell development, such cell plasticity should be taken
into consideration when designing experimental approaches.
Last, we have tried to extend the range of protocols presented in this volume beyond
the “workhorse” laboratory mouse. The power of mouse genetics has been instrumental to
T cell immunology, including in defining such essential concepts as MHC restriction and
positive selection. The last decade has seen the realization that thymic functions are present
early on during evolution, actually predating the emergence of the “modern” lymphoid
system of jawed vertebrates. This strengthens the rationale to study T cell development in
other organisms, to bolster concepts generated from mouse studies, especially if they offer
unique opportunities for genetic intervention. The zebrafish embryo is one such powerful
experimental model, allowing for oligonucleotide-directed “knock-down” of gene function
and offering quick and powerful approaches for the study of T cell development. On the
other hand, it is becoming possible, and increasingly important, to use the conceptual
frameworks generated in mouse models as platforms to investigate human T cell develop-
ment, with the objective of understanding how repertoire formation and selection drives T
cell responsiveness in physiological and pathological situations. Three chapters address
these issues, providing approaches to study human thymocytes and to model T cell devel-
opment in humanized mice. New methods for in vivo gene recombination, including
improvements of the bacteria-derived CRISPR system, should increase the power of such
studies in the years to come.
It is our hope that the present volume will be useful to readers whose scientific curiosity
brings them to interrogate the thymus, whether because of its role in shaping the T cell
repertoire or of its ability to encapsulate, in a simple and accessible organ, most of the basic
processes at work in developmental biology. We are grateful to the many authors who
shared their unique expertise to make this volume possible.
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 293
Contributors
ix
x Contributors
Background Information
Chapter 1
Abstract
T lymphocytes (T cells) are essential for proper adaptive immune responses. They perform a variety of
functions in defenses against pathogens, and notably control, positively or negatively, other cells involved
in immune responses. T cells develop in the thymus from bone marrow-derived precursors. These precur-
sors (thymocytes) proliferate, rearrange the genes encoding subunits of the T cell antigen receptor, which
endow them with their unique antigen specificity, and undergo various degrees of pre-programming for
their functions in immune responses. Thus, analyzing T cell development in the thymus is essential for
understanding their functions in immune responses. In addition, the thymus constitutes an attractive
experimental model to analyze mechanisms of cell proliferation, differentiation and survival, all of which
are involved in thymocyte development. This chapter presents a quick overview of the key events charac-
terizing intrathymic T cell development, as an introduction for readers entering this field of study.
Key words T cells, T cell development, T cell receptor, TCR gene rearrangement, Positive selection,
Negative selection
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_1, © Springer Science+Business Media New York 2016
3
4 Melanie S. Vacchio et al.
a
TCR
MHC
peptide CD4
V V
C C
P
P
Zap70
TCR
P
CD4 nor CD8. Among the best characterized are natural killer
(NK) T cells, of which most recognize CD1d-bound lipids,
mucosal-associated invariant T cells (MAIT), which recognize
MR1-bound riboflavin derivatives, and intraepithelial lymphocytes
(IELs) [9–12]. The organization of the γδ T cell population is less
well understood (see Chapter 2). It includes multiple subsets differ-
ing by antigen receptor chain usage, functional properties, and
unique developmental attributes (including their appearance at
specific embryonic or fetal developmental stages). Most such cells
express neither CD4 nor CD8.
Fig. 2 Schematic outline of T cell development. T cell development can be conceptually separated into four
steps: (i) the T lineage commitment of recent thymic immigrants, which enter the thymus as uncommitted
precursors, (ii) the rearrangement of TCR genes (dependent on Rag gene expression), itself a multistep pro-
cess, of which a critical outcome is the distribution of thymocytes into the αβ or γδ lineage, and (iii) the MHC-
based selection of thymocytes expressing αβ TCRs, during which only cells with appropriate avidity for self
antigens survive and complete their intrathymic differentiation. Most of these cells exit the thymus as “naïve,”
and acquire effector (“helper” or cytotoxic) functions during antigen-induced proliferation in peripheral lym-
phoid organs (see text for details). However, most γδ thymocytes, and a small fraction of αβ lineage cells,
acquire such effector functions in the thymus. Note that there is “chronological” overlap between these con-
ceptual steps during the developmental sequence. This is most striking in the αβ lineage, in which commit-
ment to the αβ lineage follows the rearrangement of TCRβ genes but precedes that of TCRα, and termination
of the latter is mechanistically coupled to MHC-based selection
2.5 Signaling Positive selection, strictly speaking the rescue from programmed
Positive Selection cell death, results from TCR signaling and involves increased
expression of anti-apoptotic molecules Bcl2 and Mcl1 [2]. It is
associated with differentiation events that quickly follow or coin-
cide with rescue from cell death. These first include the cessation
of TCRα gene rearrangement, at least in part by silencing expres-
sion of Rag1 and Rag2 genes. Positive selection is also accompa-
nied by the up-regulation of CCR7, a receptor for CCL19 and
CCL21 chemokines, both expressed in the thymic medulla but not
in the cortex. Together with the converse change in the expression
of CXCR4, whose ligand (CXCL12, also called SDF1) is preferen-
tially expressed in the cortex, CCR7 expression results in the
migration of thymocytes from the cortex to the medulla [41].
A more distant consequence of TCR signaling is the expression or
IL-7Rα, an event essential to T cell differentiation because it
restores IL-7 responsiveness, which is essential for long-term sur-
vival of mature T cells [42]. In addition, IL-7 is important for the
proper development of CD8+-lineage T cells [43, 44].
How such changes in gene expression are related to positively
selecting TCR signals is not yet fully understood [45]. In thymo-
cytes as in T cells, TCR signaling activates a variety of intracellular
signal transduction cascades, including the Ras-Erk kinase and cal-
cium pathway, both important for positive selection. Nuclear targets
of TCR signals include members of the Egr, AP-1, Ets and NFAT
families, as well as inhibitors (Id2 and I3) of E-box binding proteins
[46]. Because of genetic redundancy, the role of many of these fac-
tors and pathways remains to be fully elucidated, and it is likely that
other, yet to be discovered, factors contribute to positive selection.
200 Million Thymocytes and I: A Beginner’s Survival Guide… 11
2.6 Central The other side of thymic selection is the elimination (or inactivation)
Tolerance of thymocytes carrying TCRs with high avidity for self ligands.
and Negative Selection There is indeed strong genetic evidence for a thymic role in estab-
lishing immune tolerance [47]. Such “central” tolerance (i.e.
established in the thymus) is now understood as having two com-
ponents. The first prevents self-reactive cells from differentiating
into mature T cells, by causing their TCR-induced cell death in the
thymus (a deletion process referred to “negative selection”). The
second causes self-reactive cells to adopt a “regulatory” (Treg)
fate, characterized by and requiring expression of the transcription
factor Foxp3, which endows the cells with suppressive functions
[48, 49]. Both mechanisms appear important for central tolerance,
although their respective contribution is not yet fully understood
[50]. Analyses in mice suggest that deletion mechanisms do not
result in complete elimination of self-reactive cells [51], although
such studies have so far been carried only in experimental models
with clonal or reduced-diversity repertoires, and future studies will
be needed to fully measure the contribution of deletion mecha-
nisms to central tolerance. The genetic evidence for the impor-
tance of Treg cells is compelling, both in mice, in which disruption
of Foxp3 causes a severe autoimmune and inflammatory disease,
and in humans in which Foxp3 mutations are the cause for a rare
but severe autoimmune disease of infancy and childhood called
IPEX syndrome (Immunodysregulation, Polyendocrinopathy, and
Enteropathy, X-linked) [52]. The importance of Treg cell genera-
tion in the thymus is independently underscored by mouse neona-
tal thymectomy experiments; when performed before the
appearance of Treg cells (i.e. by fourth day of age), thymectomy
results in multiple autoimmune manifestations that are prevented
by adoptive transfer of Treg cells [48].
Interactions between thymocytes and medullary epithelial cells
are essential for both arms of central tolerance. That is at least in
part due to the expression, by medullary epithelial cells, of tissue-
specific antigens (epitomized by insulin, whose expression is other-
wise limited to pancreatic islets) [40]. Expression of tissue-specific
antigens in medullary epithelial cells requires the transcriptional
regulator Aire (autoimmune regulator) [47], a multifunctional pro-
tein whose dysfunction causes an autoimmune disease targeting the
endocrine system (Autoimmune Polyendocrinopathy-Candidiasis-
Ectodermal Dystrophy, APECED).
2.7 Positive vs. Because both positive and negative selection depend on TCR sig-
Negative Selection naling, perhaps no question has haunted the field of T cell develop-
Signals ment more than what distinguishes signals determining either
and Mechanisms outcome. While there is no definitive or simple answer to this ques-
tion, we have tried to summarize below current concepts and
perspectives.
12 Melanie S. Vacchio et al.
2.9 High-Avidity Even though CD4 and CD8-lineage conventional thymocytes are
Cells and Acquisition functionally “pre-programmed” for helper or cytotoxic differentia-
of Effector Properties tion, they exit the thymus as resting cells, i.e. lacking effector func-
tions. They acquire such functions (e.g. expression of cytotoxic
enzymes or cytokines) only upon antigen stimulation in peripheral
lymphoid organs. In contrast, a small subset of cells become effec-
tors in the thymus [54]. This, to some extent, is the case of Treg
precursors displaying high reactivity against MHC-peptide com-
plexes, which start expressing the key effector transcription factor
Foxp3 in the thymus. However, specific thymocyte subsets acquire
actual effector properties, including NK T cells, characterized by
high-level expression of cytokines or cytotoxic enzymes, in response
to thymic ligands. As in the case of effector differentiation in the
periphery, acquisition of effector functions by thymocytes appears
to be associated with their proliferation. Unlike for conventional
thymocytes, which are selected by MHC-peptide complexes
expressed by the thymic epithelium, selection of and acquisition of
effector properties by such “innate immune” thymocytes involve
ligands (MHC-like molecules and costimulatory molecules)
expressed on DP thymocytes themselves [68].
2.10 Thymic Of all the non-T cell types of the thymus (collectively referred to as
Architecture the stroma), the epithelial component has attracted the most atten-
tion [13, 14]. Its critical role is demonstrated by the thymic aplasia
and massive disruption of T cell development in the nude mouse,
caused by a mutation in the gene encoding the transcription factor
14 Melanie S. Vacchio et al.
3.1 Early T Cell Although early thymocytes express neither CD4 nor CD8, it is
Development important to realize that they only form a fraction of DN thymo-
cytes. Thus, identifying early progenitor cells requires additional
gating strategies to eliminate the large number of “other” DN cells
(e.g. γδ T cells, iNK T cells, B cells, etc.). This is easily done by
combining antibodies reacting against each specific lineage (e.g.
TCRγδ, CD19 as a B cell marker) labeled with the same fluoro-
chrome, so that all cells expressing any of these markers can be
electronically excluded from further analyses. This strategy is of
broad applicability to analyze small size subsets as it excludes
aggregates that can form during cell preparation.
DN thymocytes are conventionally subdivided into four sub-
sets (DN1-4) based on the expression of CD44 and CD25 (Figs. 2
and 3), although CD117 (cKit) seems to be a more specific marker
than CD44. Various strategies have been proposed to dissect the
earliest (DN1) subset and identify early T progenitors (ETP)
within the so-called DN1a compartment (CD24− CD117hi) [18].
There is only imperfect matching between such flow-cytometry
“staging” and developmental checkpoints: notably, the T lineage
commitment checkpoint occurs as cells down-regulate CD117.
Similarly, β-selection occurs in DN3 cells, and thymocytes that
have undergone β-selection can usually be identified on the basis of
their greater size, expression of intracellular TCRβ molecules, or
expression of surface CD27 or CD28 (Fig. 3).
16 Melanie S. Vacchio et al.
αβ
cKit (CD117) +++ +++ +++ +/– –
CD44 + + + + –
CD25 – – +++ +++ +++
CD24 – +++ ++++ ++++ ++++
Fig. 3 Early T cell development. Early T cell development stages are schematically depicted, using expression
of CD25 and CD44 to define DN1 (CD25−CD44+), DN2 (CD25+CD44+) and DN3 (CD25+CD44−) stages. The DN2
stage is further divided into pre- and post-commitment DN2a and DN2b stages, which can be distinguished
by the reduction in CD117 (cKit) expression. At the DN3 stage, successful completion of TCRβ or TCRγ rear-
rangement distinguishes DN3b cells (not depicted), which can be distinguished from DN3a cells by several
markers (see Fig. 4)
CD4 SP
Treg
Fig. 4 αβ T cell development. The key developmental stages of the αβ lineage are schematically depicted.
Commitment to the αβ lineage occurs during β-selection, following successful rearrangement of TCRβ in DN3
cells, and is accompanied by increased cell size (evaluated by flow cytometry on Forward light Scatter [FSC]),
and changes in expression of CD27 and CD28. Subsequent developmental stages and their defining markers
are schematically depicted
Table 1
Surface markers in the study of positive selection
4 Concluding Remarks
References
1. Kisielow P, Von Boehmer H (1995) 8. Singer A, Bosselut R (2004) CD4/CD8 core-
Development and selection of T cells: facts and ceptors in thymocyte development, selection, and
puzzles. Adv Immunol 58:87–209 lineage commitment: analysis of the CD4/CD8
2. Carpenter AC, Bosselut R (2010) Decision lineage decision. Adv Immunol 83:91–131
checkpoints in the thymus. Nat Immunol 11: 9. Bendelac A, Savage PB, Teyton L (2007) The
666–673 biology of NKT cells. Annu Rev Immunol
3. Wucherpfennig KW, Gagnon E, Call MJ, 25:297–336
Huseby ES, Call ME (2010) Structural biology 10. Le Bourhis L, Guerri L, Dusseaux M, Martin E,
of the T-cell receptor: insights into receptor Soudais C, Lantz O (2011) Mucosal-associated
assembly, ligand recognition, and initiation of invariant T cells: unconventional development
signaling. Cold Spring Harb Perspect Biol and function. Trends Immunol 32:212–218
2:a005140 11. Gold MC, Lewinsohn DM (2013)
4. Samelson LE (2002) Signal transduction Co-dependents: MR1-restricted MAIT cells
mediated by the T cell antigen receptor: the and their antimicrobial function. Nat Rev
role of adapter proteins. Annu Rev Immunol Microbiol 11:14–19
20:371–394 12. van Wijk F, Cheroutre H (2009) Intestinal T
5. Reiner S (2008) Peripheral T lymphocyte cells: facing the mucosal immune dilemma
responses and function. In: Paul WE (ed) with synergy and diversity. Semin Immunol
Fundamental immunology. Lippincott 21:130–138
Williams & Wilkins, Philadelphia, pp 407–425 13. Gordon J, Manley NR (2011) Mechanisms of
6. Smith-Garvin JE, Koretzky GA, Jordan MS thymus organogenesis and morphogenesis.
(2009) T cell activation. Annu Rev Immunol Development 138:3865–3878
27:591–619 14. Nitta T, Murata S, Ueno T, Tanaka K,
7. Mazza C, Malissen B (2007) What guides Takahama Y (2008) Thymic microenviron-
MHC-restricted TCR recognition? Semin ments for T-cell repertoire formation. Adv
Immunol 19:225–235 Immunol 99:59–94
200 Million Thymocytes and I: A Beginner’s Survival Guide… 19
15. Rossi SW, Jenkinson WE, Anderson G, 29. Gonzalez-Garcia S, Garcia-Peydro M, Alcain J,
Jenkinson EJ (2006) Clonal analysis reveals a Toribio ML (2012) Notch1 and IL-7 receptor
common progenitor for thymic cortical and signalling in early T-cell development and leukae-
medullary epithelium. Nature 441:988–991 mia. Curr Top Microbiol Immunol 360:47–73
16. Bleul CC, Corbeaux T, Reuter A, Fisch P, 30. Jung D, Alt FW (2004) Unraveling V(D)J
Monting JS, Boehm T (2006) Formation of a recombination; insights into gene regulation.
functional thymus initiated by a postnatal epi- Cell 116:299–311
thelial progenitor cell. Nature 441:992–996 31. von Boehmer H, Aifantis I, Azogui O, Feinberg
17. Yang Q, Jeremiah Bell J, Bhandoola A (2010) J, Saint-Ruf C, Zober C, Garcia C, Buer J
T-cell lineage determination. Immunol Rev (1998) Crucial function of the pre-T-cell
238:12–22 receptor (TCR) in TCR beta selection, TCR
18. Bhandoola A, von Boehmer H, Petrie HT, beta allelic exclusion and alpha beta versus
Zuniga-Pflucker JC (2007) Commitment and gamma delta lineage commitment. Immunol
developmental potential of extrathymic and Rev 165:111–119
intrathymic T cell precursors: plenty to choose 32. von Boehmer H, Aifantis I, Feinberg J, Lechner
from. Immunity 26:678–689 O, Saint-Ruf C, Walter U, Buer J, Azogui O
19. Rothenberg EV (2011) T cell lineage commit- (1999) Pleiotropic changes controlled by the pre-
ment: identity and renunciation. J Immunol T-cell receptor. Curr Opin Immunol 11:135–142
186:6649–6655 33. Hayes SM, Love PE (2006) Strength of signal:
20. Zhang SL, Bhandoola A (2014) Trafficking to a fundamental mechanism for cell fate specifi-
the thymus. Curr Top Microbiol Immunol cation. Immunol Rev 209:170–175
373:87–111 34. Lauritsen JP, Haks MC, Lefebvre JM, Kappes
21. Hozumi K, Mailhos C, Negishi N, Hirano K, DJ, Wiest DL (2006) Recent insights into the
Yahata T, Ando K, Zuklys S, Hollander GA, signals that control alphabeta/gammadelta-
Shima DT, Habu S (2008) Delta-like 4 is indis- lineage fate. Immunol Rev 209:176–190
pensable in thymic environment specific for T 35. Kreslavsky T, Gleimer M, von Boehmer H
cell development. J Exp Med 205:2507–2513 (2010) Alphabeta versus gammadelta lineage
22. Koch U, Fiorini E, Benedito R, Besseyrias V, choice at the first TCR-controlled checkpoint.
Schuster-Gossler K, Pierres M, Manley NR, Curr Opin Immunol 22(2):185–192
Duarte A, Macdonald HR, Radtke F (2008) 36. Mingueneau M, Kreslavsky T, Gray D, Heng
Delta-like 4 is the essential, nonredundant T, Cruse R, Ericson J, Bendall S, Spitzer MH,
ligand for Notch1 during thymic T cell lineage Nolan GP, Kobayashi K, von Boehmer H,
commitment. J Exp Med 205:2515–2523 Mathis D, Benoist C, Best AJ, Knell J, Goldrath
23. Pui JC, Allman D, Xu L, DeRocco S, Karnell A, Jojic V, Koller D, Shay T, Regev A, Cohen
FG, Bakkour S, Lee JY, Kadesch T, Hardy RR, N, Brennan P, Brenner M, Kim F, Rao TN,
Aster JC, Pear WS (1999) Notch1 expression Wagers A, Heng T, Ericson J, Rothamel K,
in early lymphopoiesis influences B versus T Ortiz-Lopez A, Mathis D, Benoist C, Bezman
lineage determination. Immunity 11:299–308 NA, Sun JC, Min-Oo G, Kim CC, Lanier LL,
24. Radtke F, Wilson A, Stark G, Bauer M, van Miller J, Brown B, Merad M, Gautier EL,
Meerwijk J, MacDonald HR, Aguet M (1999) Jakubzick C, Randolph GJ, Monach P, Blair
Deficient T cell fate specification in mice with DA, Dustin ML, Shinton SA, Hardy RR,
an induced inactivation of Notch1. Immunity Laidlaw D, Collins J, Gazit R, Rossi DJ,
10:547–558 Malhotra N, Sylvia K, Kang J, Kreslavsky T,
Fletcher A, Elpek K, Bellemare-Pelletier A,
25. Rothenberg EV (2012) Transcriptional drivers Malhotra D, Turley S (2013) The transcrip-
of the T-cell lineage program. Curr Opin tional landscape of alphabeta T cell differentia-
Immunol 24:132–138 tion. Nat Immunol 14:619–632
26. Rothenberg EV, Moore JE, Yui MA (2008) 37. Williams JA, Hathcock KS, Klug D, Harada Y,
Launching the T-cell-lineage developmental Choudhury B, Allison JP, Abe R, Hodes RJ
programme. Nat Rev Immunol 8:9–21 (2005) Regulated costimulation in the thymus
27. Ho IC, Tai TS, Pai SY (2009) GATA3 and the is critical for T cell development: dysregulated
T-cell lineage: essential functions before and CD28 costimulation can bypass the pre-TCR
after T-helper-2-cell differentiation. Nat Rev checkpoint. J Immunol 175:4199–4207
Immunol 9:125–135 38. Taghon T, Yui MA, Pant R, Diamond RA,
28. Avram D, Califano D (2014) The multifaceted Rothenberg EV (2006) Developmental and
roles of Bcl11b in thymic and peripheral T molecular characterization of emerging beta-
cells: impact on immune diseases. J Immunol and gammadelta-selected pre-T cells in the
193(5):2059–2065 adult mouse thymus. Immunity 24:53–64
20 Melanie S. Vacchio et al.
39. Starr TK, Jameson SC, Hogquist KA (2003) 53. Daniels MA, Teixeiro E, Gill J, Hausmann B,
Positive and negative selection of T cells. Annu Roubaty D, Holmberg K, Werlen G, Hollander
Rev Immunol 21:139–176 GA, Gascoigne NR, Palmer E (2006) Thymic
40. Klein L, Kyewski B, Allen PM, Hogquist KA selection threshold defined by compartmental-
(2014) Positive and negative selection of the T ization of Ras/MAPK signalling. Nature
cell repertoire: what thymocytes see (and don’t 444:724–729
see). Nat Rev Immunol 14:377–391 54. Stritesky GL, Jameson SC, Hogquist KA
41. Nitta T, Ohigashi I, Nakagawa Y, Takahama Y (2012) Selection of self-reactive T cells in the
(2011) Cytokine crosstalk for thymic medulla thymus. Annu Rev Immunol 30:95–114
formation. Curr Opin Immunol 23:190–197 55. Fu G, Rybakin V, Brzostek J, Paster W, Acuto
42. Boyman O, Krieg C, Homann D, Sprent J O, Gascoigne NR (2014) Fine-tuning T cell
(2012) Homeostatic maintenance of T cells receptor signaling to control T cell develop-
and natural killer cells. Cell Mol Life Sci ment. Trends Immunol 35:311–318
69:1597–1608 56. McGargill MA, Ch’en IL, Katayama CD, Pages
43. McCaughtry TM, Etzensperger R, Alag A, Tai G, Pouyssegur J, Hedrick SM (2009) Cutting
X, Kurtulus S, Park JH, Grinberg A, Love P, edge: extracellular signal-related kinase is not
Feigenbaum L, Erman B, Singer A (2012) required for negative selection of developing T
Conditional deletion of cytokine receptor cells. J Immunol 183:4838–4842
chains reveals that IL-7 and IL-15 specify CD8 57. Fassett MS, Jiang W, D’Alise AM, Mathis D,
cytotoxic lineage fate in the thymus. J Exp Med Benoist C (2012) Nuclear receptor Nr4a1
209:2263–2276 modulates both regulatory T-cell (Treg) differ-
44. Tani-ichi S, Shimba A, Wagatsuma K, Miyachi entiation and clonal deletion. Proc Natl Acad
H, Kitano S, Imai K, Hara T, Ikuta K (2013) Sci U S A 109:3891–3896
Interleukin-7 receptor controls development 58. Sekiya T, Kashiwagi I, Yoshida R, Fukaya T,
and maturation of late stages of thymocyte sub- Morita R, Kimura A, Ichinose H, Metzger D,
populations. Proc Natl Acad Sci U S A 110: Chambon P, Yoshimura A (2013) Nr4a recep-
612–617 tors are essential for thymic regulatory T cell
45. Gascoigne NR, Palmer E (2011) Signaling in thy- development and immune homeostasis. Nat
mic selection. Curr Opin Immunol 23:207–212 Immunol 14:230–237
46. Wang L, Xiong Y, Bosselut R (2010) Tenuous 59. Staton TL, Lazarevic V, Jones DC, Lanser AJ,
paths in unexplored territory: from T cell Takagi T, Ishii S, Glimcher LH (2011)
receptor signaling to effector gene expression Dampening of death pathways by schnurri-2 is
during thymocyte selection. Semin Immunol essential for T-cell development. Nature
22:294–302 472:105–109
47. Mathis D, Benoist C (2009) Aire. Annu Rev 60. Gray DH, Kupresanin F, Berzins SP, Herold
Immunol 27:287–312 MJ, O’Reilly LA, Bouillet P, Strasser A (2012)
48. Sakaguchi S, Yamaguchi T, Nomura T, Ono M The BH3-only proteins Bim and Puma cooper-
(2008) Regulatory T cells and immune toler- ate to impose deletional tolerance of organ-
ance. Cell 133:775–787 specific antigens. Immunity 37:451–462
49. Josefowicz SZ, Lu LF, Rudensky AY (2012) 61. Stritesky GL, Xing Y, Erickson JR, Kalekar LA,
Regulatory T cells: mechanisms of differentia- Wang X, Mueller DL, Jameson SC, Hogquist
tion and function. Annu Rev Immunol 30: KA (2013) Murine thymic selection quantified
531–564 using a unique method to capture deleted T
cells. Proc Natl Acad Sci U S A 110:4679–4684
50. Guerau-de-Arellano M, Martinic M, Benoist
C, Mathis D (2009) Neonatal tolerance 62. Wang L, Bosselut R (2009) CD4-CD8 lineage
revisited: a perinatal window for Aire control of differentiation: Thpok-ing into the nucleus.
autoimmunity. J Exp Med 206:1245–1252 J Immunol 183:2903–2910
51. Bouneaud C, Kourilsky P, Bousso P (2000) 63. Xiong Y, Bosselut R (2012) CD4-CD8 differ-
Impact of negative selection on the T cell rep- entiation in the thymus: connecting circuits
ertoire reactive to a self-peptide: a large frac- and building memories. Curr Opin Immunol
tion of T cell clones escapes clonal deletion. 24:139–145
Immunity 13:829–840 64. Xiong Y, Bosselut R (2011) The enigma of
52. Verbsky JW, Chatila TA (2013) Immune dys- CD4-lineage specification. Eur J Immunol
regulation, polyendocrinopathy, enteropathy, 41:568–574
X-linked (IPEX) and IPEX-related disorders: 65. Cyster JG, Schwab SR (2012) Sphingosine-1-
an evolving web of heritable autoimmune dis- phosphate and lymphocyte egress from lym-
eases. Curr Opin Pediatr 25:708–714 phoid organs. Annu Rev Immunol 30:69–94
200 Million Thymocytes and I: A Beginner’s Survival Guide… 21
66. Hart GT, Hogquist KA, Jameson SC (2012) Takahama Y (2010) Thymoproteasome shapes
Kruppel-like factors in lymphocyte biology. J immunocompetent repertoire of CD8+ T cells.
Immunol 188:521–526 Immunity 32:29–40
67. Hedrick SM, Hess Michelini R, Doedens AL, 73. Shores EW, Van Ewijk W, Singer A (1991)
Goldrath AW, Stone EL (2012) FOXO tran- Disorganization and restoration of thymic
scription factors throughout T cell biology. Nat medullary epithelial cells in T cell receptor-
Rev Immunol 12:649–661 negative scid mice: evidence that receptor-
68. Cannons JL, Tangye SG, Schwartzberg PL bearing lymphocytes influence maturation of
(2011) SLAM family receptors and SAP the thymic microenvironment. Eur J Immunol
adaptors in immunity. Annu Rev Immunol 21:1657–1661
29:665–705 74. Philpott KL, Viney JL, Kay G, Rastan S,
69. Nitta T, Nitta S, Lei Y, Lipp M, Takahama Y Gardiner EM, Chae S, Hayday AC, Owen MJ
(2009) CCR7-mediated migration of develop- (1992) Lymphoid development in mice con-
ing thymocytes to the medulla is essential for genitally lacking T cell receptor alpha beta-
negative selection to tissue-restricted antigens. expressing cells. Science 256:1448–1452
Proc Natl Acad Sci U S A 106:17129–17133 75. Dudakov JA, Hanash AM, Jenq RR, Young
70. Nakagawa T, Roth W, Wong P, Nelson A, Farr LF, Ghosh A, Singer NV, West ML, Smith
A, Deussing J, Villadangos JA, Ploegh H, OM, Holland AM, Tsai JJ, Boyd RL, van den
Peters C, Rudensky AY (1998) Cathepsin L: Brink MR (2012) Interleukin-22 drives
critical role in Ii degradation and CD4 T cell endogenous thymic regeneration in mice.
selection in the thymus. Science 280:450–453 Science 336:91–95
71. Klein L, Hinterberger M, Wirnsberger G, 76. Bosselut R, Guinter TI, Sharrow SO, Singer A
Kyewski B (2009) Antigen presentation in the (2003) Unraveling a revealing paradox: why
thymus for positive selection and central toler- major histocompatibility complex I-signaled
ance induction. Nat Rev Immunol 9:833–844 thymocytes “Paradoxically” appear as CD4+8lo
72. Nitta T, Murata S, Sasaki K, Fujii H, Ripen transitional cells during positive selection of
AM, Ishimaru N, Koyasu S, Tanaka K, CD8+ T cells. J Exp Med 197:1709–1719
Chapter 2
Abstract
While the functions of αβ T cells in host resistance to pathogen infection are understood in far more detail
than those of γδ lineage T cells, γδ T cells perform critical, essential functions during immune responses
that cannot be compensated by αβ T cells. Accordingly, it is essential to understand how the development
of γδ T cells is controlled so that their generation and function might be manipulated in future for thera-
peutic benefit. This introductory chapter will cover the basic processes that underlie γδ T cell development
in the thymus, as well as the current understanding of how they are controlled.
Key words Gamma-delta T cells, Gamma-delta TCR, V gamma elements, Lineage commitment,
Effector fate
1 Introduction
There are two major T lineages marked by the T cell antigen receptor
(TCR) complexes they express, αβ and γδ. By analogy to the military,
αβ lineage T cells are like conventional soldiers found primarily in
lymphoid organs, which can be thought of as military bases where the
staging of military operations occur. This is consistent with the 7-day
delay required for αβ T cells to mount a primary response to an infec-
tion and travel to the site of the battle. In contrast, γδ T cells make up
a small proportion of T cells in the peripheral lymphoid organs, and
instead predominate in the epithelial tissues that form the inner and
outer surfaces of the body [1–3]. Accordingly, γδ cells are much more
like special-force soldiers found primarily in the “field,” patrolling epi-
thelial barriers and possessing rapid-strike capabilities that do not
require priming. Indeed, γδ cells have been implicated in stress-sur-
veillance by rapidly responding to stress-induced proteins on epithelial
cells (e.g., Rae1 and H60), which is more consistent with an innate-
like mode of function [3]. Nevertheless, γδ T cells are also capable of
staging delayed, more classical adaptive-type responses following
immunization, including the mounting of recall responses consistent
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_2, © Springer Science+Business Media New York 2016
23
24 David L. Wiest
Raulet: V 3 V 4 V 2 V 1.1
Tonegawa: V 5 V 6 V 4 V 1
T cell development
Thymic
12 14 16 18 20 Birth
Days of Embryonic Development
* Development of the V 7 (Tonegawa)/V 5 (Raulet) subset, which
homes to the gut, is thought to occur extrathymically.
Table 1
Commercially available anti-V region antibodies to study
mouse γδ T cell subsets
4 Development
Fig. 2 Models describing the basis for linkage of effector fate to Vγ usage. Many γδ T cells acquire effector
function during development in the thymus. Two models have been advanced to explain how this occurs. The
first is the “pre-determination model,” which suggests that effector fate is pre-programmed and is linked to
Vγ usage by virtue of their developmental timing, but is not influenced by TCR signaling. The second suggests
that effector fate is influenced by TCR signal strength with a gradation of signaling (schematized by blue tri-
angle) ranging from weakest, which specify the IL-17 producing effector fate to the strongest, which specify
the PLZF-expressing innate effector fate associated with co-production of IL-4 and IFNγ
30 David L. Wiest
5 Conclusion
References
1. Hayday AC (2000) [gamma][delta] cells: a 16. Lauritsen JP, Haks MC, Lefebvre JM et al
right time and a right place for a conserved (2006) Recent insights into the signals that
third way of protection. Annu Rev Immunol control alphabeta/gammadelta-lineage fate.
18:975–1026 Immunol Rev 209:176–190
2. Carding SR, Egan PJ (2002) Gammadelta T 17. Xiong N, Raulet DH (2007) Development and
cells: functional plasticity and heterogeneity. selection of gammadelta T cells. Immunol Rev
Nat Rev Immunol 2:336–345 215:15–31
3. Vantourout P, Hayday A (2013) Six-of-the-best: 18. Bonneville M, O’Brien RL, Born WK (2010)
unique contributions of gammadelta T cells to Gammadelta T cell effector functions: a blend
immunology. Nat Rev Immunol 13:88–100 of innate programming and acquired plasticity.
4. Sheridan BS, Romagnoli PA, Pham QM et al Nat Rev Immunol 10:467–478
(2013) gammadelta T cells exhibit multifunc- 19. O’Brien RL, Born WK (2010) gammadelta T
tional and protective memory in intestinal tis- cell subsets: a link between TCR and function?
sues. Immunity 39:184–195 Semin Immunol 22:193–198
5. King DP, Hyde DM, Jackson KA et al (1999) 20. Jin Y, Xia M, Saylor CM et al (2010) Cutting
Cutting edge: protective response to pulmo- edge: intrinsic programming of thymic gam-
nary injury requires gamma delta T lympho- madeltaT cells for specific peripheral tissue
cytes. J Immunol 162:5033–5036 localization. J Immunol 185:7156–7160
6. Ramsburg E, Tigelaar R, Craft J et al (2003) 21. Lee SY, Stadanlick J, Kappes DJ et al (2010)
Age-dependent requirement for gammadelta T Towards a molecular understanding of the dif-
cells in the primary but not secondary protec- ferential signals regulating alphabeta/gam-
tive immune response against an intestinal par- madelta T lineage choice. Semin Immunol
asite. J Exp Med 198:1403–1414 22:237–246
7. Girardi M, Oppenheim DE, Steele CR et al 22. Lewis JM, Girardi M, Roberts SJ et al (2006)
(2001) Regulation of cutaneous malignancy by Selection of the cutaneous intraepithelial gam-
{gamma}{delta} T cells. Science 20:20 madelta + T cell repertoire by a thymic stromal
8. Chen Y, Chou K, Fuchs E et al (2002) determinant. Nat Immunol 7:843–850
Protection of the intestinal mucosa by intraepi- 23. Pereira P, Zijlstra M, McMaster J et al (1992)
thelial gamma delta T cells. Proc Natl Acad Sci Blockade of transgenic gamma delta T cell
U S A 99:14338–14343 development in beta 2-microglobulin deficient
9. Sharp LL, Jameson JM, Cauvi G et al (2005) mice. EMBO J 11:25–31
Dendritic epidermal T cells regulate skin homeo- 24. Coffey F, Lee SY, Buus TB et al (2014) The
stasis through local production of insulin-like TCR ligand-inducible expression of CD73
growth factor 1. Nat Immunol 6:73–79 marks gammadelta lineage commitment and a
10. Nedellec S, Bonneville M, Scotet E (2010) metastable intermediate in effector specifica-
Human Vgamma9Vdelta2 T cells: from signals tion. J Exp Med 211:329–343
to functions. Semin Immunol 22:199–206 25. Ribot JC, deBarros A, Pang DJ et al (2009)
11. Meraviglia S, Eberl M, Vermijlen D et al (2010) CD27 is a thymic determinant of the balance
In vivo manipulation of Vgamma9Vdelta2 T between interferon-gamma- and interleukin
cells with zoledronate and low-dose interleukin- 17-producing gammadelta T cell subsets. Nat
2 for immunotherapy of advanced breast cancer Immunol 10:427–436
patients. Clin Exp Immunol 161:290–297 26. Kreslavsky T, Garbe AI, Krueger A et al
12. Cai Y, Fleming C, Yan J (2012) New insights of (2008) T cell receptor-instructed alphabeta
T cells in the pathogenesis of psoriasis. Cell versus gammadelta lineage commitment
Mol Immunol 9:302–309 revealed by single-cell analysis. J Exp Med
13. Wu P, Wu D, Ni C et al (2014) gammadeltaT17 205:1173–1186
cells promote the accumulation and expansion 27. Pennington DJ, Silva-Santos B, Shires J et al
of myeloid-derived suppressor cells in human (2003) The inter-relatedness and interdepen-
colorectal cancer. Immunity 40:785–800 dence of mouse T cell receptor gammadelta+
14. Born WK, Kemal Aydintug M, O’Brien RL and alphabeta+ cells. Nat Immunol 4:
(2013) Diversity of gammadelta T-cell anti- 991–998
gens. Cell Mol Immunol 10:13–20 28. Melichar HJ, Narayan K, Der SD et al (2007)
15. Rock EP, Sibbald PR, Davis MM et al (1994) Regulation of gammadelta versus alphabeta T
CDR3 length in antigen-specific immune lymphocyte differentiation by the transcription
receptors. J Exp Med 179:323–328 factor SOX13. Science 315:230–233
32 David L. Wiest
29. Roberts NA, White AJ, Jenkinson WE et al 39. Meyer C, Zeng X, Chien YH (2010) Ligand
(2012) Rank signaling links the development recognition during thymic development and
of invariant gammadelta T cell progenitors and gammadelta T cell function specification.
Aire(+) medullary epithelium. Immunity 36: Semin Immunol 22:207–213
427–437 40. Havran WL, Allison JP (1988) Developmentally
30. Silva-Santos B, Pennington DJ, Hayday AC ordered appearance of thymocytes expressing
(2005) Lymphotoxin-mediated regulation of different T-cell antigen receptors. Nature 335:
gammadelta cell differentiation by alphabeta T 443–445
cell progenitors. Science 307:925–928 41. Boyden LM, Lewis JM, Barbee SD et al (2008)
31. Petrie HT, Scollay R, Shortman K (1992) Skint1, the prototype of a newly identified
Commitment to the T cell receptor-alpha beta immunoglobulin superfamily gene cluster, pos-
or -gamma delta lineages can occur just prior to itively selects epidermal gammadelta T cells.
the onset of CD4 and CD8 expression among Nat Genet 40:656–662
immature thymocytes. Eur J Immunol 22: 42. Lauritsen JP, Wong GW, Lee SY et al (2009)
2185–2188 Marked induction of the helix-loop-helix pro-
32. Ciofani M, Knowles GC, Wiest DL et al (2006) tein Id3 promotes the gammadelta T cell fate
Stage-specific and differential notch depen- and renders their functional maturation Notch
dency at the alphabeta and gammadelta T lin- independent. Immunity 31:565–575
eage bifurcation. Immunity 25:105–116 43. Prinz I, Silva-Santos B, Pennington DJ (2013)
33. Narayan K, Kang J (2010) Disorderly conduct Functional development of gammadelta T
in gammadelta versus alphabeta T cell lineage cells. Eur J Immunol 43:1988–1994
commitment. Semin Immunol 22:222–227 44. Turchinovich G, Hayday AC (2011) Skint-1
34. Wong GW, Zuniga-Pflucker JC (2010) gam- identifies a common molecular mechanism for
madelta and alphabeta T cell lineage choice: the development of interferon-gamma-
resolution by a stronger sense of being. Semin secreting versus interleukin-17-secreting gam-
Immunol 22:228–236 madelta T cells. Immunity 35:59–68
35. Hayes SM, Love PE (2006) Strength of signal: 45. Narayan K, Sylvia KE, Malhotra N et al (2012)
a fundamental mechanism for cell fate specifica- Intrathymic programming of effector fates in
tion. Immunol Rev 209:170–175 three molecularly distinct gammadelta T cell
36. Hayes SM, Li L, Love PE (2005) TCR signal subtypes. Nat Immunol 13:511–518
strength influences alphabeta/gammadelta lin- 46. Jensen KD, Su X, Shin S et al (2008) Thymic
eage fate. Immunity 22:583–593 selection determines gammadelta T cell effec-
37. Haks MC, Lefebvre JM, Lauritsen JP et al tor fate: antigen-naive cells make interleukin-
(2005) Attenuation of gammadeltaTCR signal- 17 and antigen-experienced cells make
ing efficiently diverts thymocytes to the alpha- interferon gamma. Immunity 29:90–100
beta lineage. Immunity 22:595–606 47. Wencker M, Turchinovich G, Di Marco BR et al
38. Kreslavsky T, von Boehmer H (2010) gam- (2014) Innate-like T cells straddle innate and
madeltaTCR ligands and lineage commitment. adaptive immunity by altering antigen-receptor
Semin Immunol 22:214–221 responsiveness. Nat Immunol 15:80–87
Part II
Abstract
Genetics tools, and especially the ability to enforce, by transgenesis, or disrupt, by homologous recombination,
gene expression in a cell-specific manner, have revolutionized the study of immunology and propelled the
laboratory mouse as the main model to study immune responses. Perhaps more than any other aspect of
immunology, the study of T cell development has benefited from these technologies. This brief chapter
summarizes genetic tools specific to T cell development studies, focusing on mouse strains with lineage-
and stage-specific expression of the Cre recombinase, or expressing unique antigen receptor specificities. It
ends with a broader discussion of strategies to enforce ectopic lineage and stage-specific gene expression.
Key words Cre recombinase, Genetic strategies, Lineage-specific gene disruption, TCR transgenic
mice, Deletion reporter genes
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_3, © Springer Science+Business Media New York 2016
35
36 Thomas Ciucci et al.
Table 1
Cre-expressing strains
Promoter
or enhancer Reference Cre expression Commerciala Comments
Range Onset
Vav1 [ 3] All hematopoietic HSC Jax
cells
Tie2 [4] All hematopoietic HSC Jax Also endothelial
cells cells
Rag1 [5] Rag1-expressing
B and T cell
precursors
Il7r [6] B and T cells BM lymphoid
(IL-7Rα) progenitors
Lck, proximal [7] T lineage cellsc DN2 Taconic
promoterb
(Wilson)
Lck, proximal [8] T lineage cellsc DN3 Jax Cre expression,
promoterb lower and
(Marth) delayed relative
to Wilson strain
[9]. Potential
for incomplete
deletion
CD2 [3] T and B cellsc DN2 Jax
Ptcra [10] T lineage cells DN2a
(Pre-Tα)
Cd4 [ 7] αβ lineage onlyc DN3 to DP, Taconic
therefore
deleting in
all αβ T cell
precursors
Cd8 E(III) [11] αβ lineage only ISP-DP
+
Cd8 E(I) [12, 13] CD8 T cells CD24lo CD8 SP
only thymocytes
Lck (distal) [14] T cells T cells Higher in SP
thymocytes and
T cells
CD2 [15] T cells Mature Expression pattern
thymocytes unrelated to
to Naïve other CD2-
T cells based constructs.
Incomplete in
CD4+ T cells
(continued)
38 Thomas Ciucci et al.
Table 1
(continued)
Promoter
or enhancer Reference Cre expression Commerciala Comments
Range Onset
Table 2
MHC-I-restricted TCR transgenic strains
many more TCR transgenic lines have been generated and used in a
variety of studies of T cell function during physiological and patho-
logical responses (e.g. refs. 45–47).
An important condition to the success of this strategy is that
cells expressing the transgenic TCR do not express other TCR
chains generated by rearrangement of endogenous loci. That is
only partly the case. Expression of a rearranged TCRβ chain
(whether endogenous or transgenic) prevents further rearrange-
ment at the TCRβ loci (a phenomenon called allelic exclusion)
[48] and at the TCRγ loci (isotypic exclusion) [49]. However,
TCRβ allelic exclusion is not perfect; as a result, in many TCR
transgenic strains, small fractions of thymocytes express both the
transgenic and an endogenous TCRβ chain. In addition, there is
no allelic exclusion at the TCRα locus, so that expression of a
transgenic TCRαβ does not guarantee absence of endogenous
TCRα gene rearrangement, which has potential implications for
40
Table 3
MHC-II-restricted TCR transgenic strains
References
1. Gupta RM, Musunuru K (2014) Expanding the locations or retroviral insertion during gene
genetic editing tool kit: ZFNs, TALENs, and therapy but has no mandatory role in normal
CRISPR-Cas9. J Clin Invest 124:4154–4161 T-cell development. Mol Cell Biol 23:
2. Mali P, Esvelt KM, Church GM (2013) Cas9 9003–9013
as a versatile tool for engineering biology. Nat 6. Schlenner SM, Madan V, Busch K, Tietz A,
Methods 10:957–963 Laufle C, Costa C, Blum C, Fehling HJ,
3. de Boer J, Williams A, Skavdis G, Harker N, Rodewald HR (2010) Fate mapping reveals
Coles M, Tolaini M, Norton T, Williams K, separate origins of T cells and myeloid lineages
Roderick K, Potocnik AJ, Kioussis D (2003) in the thymus. Immunity 32:426–436
Transgenic mice with hematopoietic and lym- 7. Lee PP, Fitzpatrick DR, Beard C, Jessup HK,
phoid specific expression of Cre. Eur J Lehar S, Makar KW, Perez-Melgosa M,
Immunol 33:314–325 Sweetser MT, Schlissel MS, Nguyen S, Cherry
4. Kisanuki YY, Hammer RE, Miyazaki J, SR, Tsai JH, Tucker SM, Weaver WM, Kelso
Williams SC, Richardson JA, Yanagisawa M A, Jaenisch R, Wilson CB (2001) A critical
(2001) Tie2-Cre transgenic mice: a new model role for Dnmt1 and DNA methylation in T cell
for endothelial cell-lineage analysis in vivo. development, function, and survival. Immunity
Dev Biol 230:230–242 15:763–774
5. McCormack MP, Forster A, Drynan L, Pannell 8. Hennet T, Hagen FK, Tabak LA, Marth JD
R, Rabbitts TH (2003) The LMO2 T-cell (1995) T-cell-specific deletion of a polypeptide
oncogene is activated via chromosomal trans- N-acetylgalactosaminyl-transferase gene by
Genetic Tools to Study T Cell Development 43
site-directed recombination. Proc Natl Acad shows its essential function in T(H)1-T(H)2
Sci U S A 92:12070–12074 responses. Nat Immunol 5:1157–1165
9. Shi J, Petrie HT (2012) Activation kinetics 18. Klinger M, Kim JK, Chmura SA, Barczak A,
and off-target effects of thymus-initiated cre Erle DJ, Killeen N (2009) Thymic OX40
transgenes. PLoS One 7, e46590 expression discriminates cells undergoing
10. Luche H, Nageswara Rao T, Kumar S, strong responses to selection ligands. J
Tasdogan A, Beckel F, Blum C, Martins VC, Immunol 182:4581–4589
Rodewald HR, Fehling HJ (2013) In vivo fate 19. Jacob J, Baltimore D (1999) Modelling T-cell
mapping identifies pre-TCRalpha expression memory by genetic marking of memory T cells
as an intra- and extrathymic, but not prethy- in vivo. Nature 399:593–597
mic, marker of T lymphopoiesis. J Exp Med 20. Aghajani K, Keerthivasan S, Yu Y, Gounari F
210:699–714 (2012) Generation of CD4CreER(T(2))
11. Park JH, Adoro S, Guinter T, Erman B, Alag transgenic mice to study development of
AS, Catalfamo M, Kimura MY, Cui Y, Lucas peripheral CD4-T-cells. Genesis 50:908–913
PJ, Gress RE, Kubo M, Hennighausen L, 21. Soriano P (1999) Generalized lacZ expression
Feigenbaum L, Singer A (2010) Signaling by with the ROSA26 Cre reporter strain. Nat
intrathymic cytokines, not T cell antigen Genet 21:70–71
receptors, specifies CD8 lineage choice and 22. Schmidt-Supprian M, Rajewsky K (2007)
promotes the differentiation of cytotoxic-lin- Vagaries of conditional gene targeting. Nat
eage T cells. Nat Immunol 11:257–264 Immunol 8:665–668
12. Zou YR, Sunshine MJ, Taniuchi I, Hatam F, 23. Moon JJ, Chu HH, Pepper M, McSorley SJ,
Killeen N, Littman DR (2001) Epigenetic Jameson SC, Kedl RM, Jenkins MK (2007)
silencing of CD4 in T cells committed to the Naive CD4(+) T cell frequency varies for dif-
cytotoxic lineage. Nat Genet 29:332–336 ferent epitopes and predicts repertoire diver-
13. Maekawa Y, Minato Y, Ishifune C, Kurihara T, sity and response magnitude. Immunity
Kitamura A, Kojima H, Yagita H, Sakata- 27:203–213
Yanagimoto M, Saito T, Taniuchi I, Chiba S, 24. von Boehmer H (1990) Developmental biol-
Sone S, Yasutomo K (2008) Notch2 integrates ogy of T cells in T cell-receptor transgenic
signaling by the transcription factors RBP-J mice. Annu Rev Immunol 8:531–556
and CREB1 to promote T cell cytotoxicity.
Nat Immunol 9:1140–1147 25. Hogquist KA, Jameson SC, Heath WR,
Howard JL, Bevan MJ, Carbone FR (1994) T
14. Zhang DJ, Wang Q, Wei J, Baimukanova G, cell receptor antagonist peptides induce posi-
Buchholz F, Stewart AF, Mao X, Killeen N tive selection. Cell 76:17–27
(2005) Selective expression of the Cre recom-
binase in late-stage thymocytes using the distal 26. Daniels MA, Teixeiro E, Gill J, Hausmann B,
promoter of the Lck gene. J Immunol Roubaty D, Holmberg K, Werlen G, Hollander
174:6725–6731 GA, Gascoigne NR, Palmer E (2006) Thymic
selection threshold defined by compartmental-
15. Vacchio MS, Wang L, Bouladoux N, Carpenter ization of Ras/MAPK signalling. Nature
AC, Xiong Y, Williams LC, Wohlfert E, Song 444:724–729
KD, Belkaid Y, Love PE, Bosselut R (2014) A
ThPOK-LRF transcriptional node maintains the 27. Clarke SR, Barnden M, Kurts C, Carbone FR,
integrity and effector potential of post-thymic Miller JF, Heath WR (2000) Characterization
CD4 T cells. Nat Immunol 15(10):947–956 of the ovalbumin-specific TCR transgenic line
OT-I: MHC elements for positive and nega-
16. Mucida D, Husain MM, Muroi S, van Wijk F, tive selection. Immunol Cell Biol 78:110–117
Shinnakasu R, Naoe Y, Reis BS, Huang Y,
Lambolez F, Docherty M, Attinger A, Shui 28. Markiewicz MA, Girao C, Opferman JT, Sun
JW, Kim G, Lena CJ, Sakaguchi S, Miyamoto J, Hu Q, Agulnik AA, Bishop CE, Thompson
C, Wang P, Atarashi K, Park Y, Nakayama T, CB, Ashton-Rickardt PG (1998) Long-term T
Honda K, Ellmeier W, Kronenberg M, cell memory requires the surface expression of
Taniuchi I, Cheroutre H (2013) Transcriptional self-peptide/major histocompatibility complex
reprogramming of mature CD4(+) helper T molecules. Proc Natl Acad Sci U S A
cells generates distinct MHC class II-restricted 95:3065–3070
cytotoxic T lymphocytes. Nat Immunol 29. Teh HS, Kishi H, Scott B, Von Boehmer H
14:281–289 (1989) Deletion of autospecific T cells in T cell
17. Zhu J, Min B, Hu-Li J, Watson CJ, Grinberg receptor (TCR) transgenic mice spares cells
A, Wang Q, Killeen N, Urban JFJ, Guo L, Paul with normal TCR levels and low levels of CD8
WE (2004) Conditional deletion of Gata3 molecules. J Exp Med 169:795–806
44 Thomas Ciucci et al.
30. Kisielow P, Bluthmann H, Staerz UD, 41. Seder RA, Paul WE, Davis MM, Fazekas de St
Steinmetz M, von Boehmer H (1988) Groth B (1992) The presence of interleukin 4
Tolerance in T-cell-receptor transgenic mice during in vitro priming determines the
involves deletion of nonmature CD4+8+ thy- lymphokine-producing potential of CD4+ T
mocytes. Nature 333:742–746 cells from T cell receptor transgenic mice. J
31. Colf LA, Bankovich AJ, Hanick NA, Exp Med 176:1091–1098
Bowerman NA, Jones LL, Kranz DM, Garcia 42. Fink PJ, Matis LA, McElligott DL, Bookman
KC (2007) How a single T cell receptor recog- M, Hedrick SM (1986) Correlations between
nizes both self and foreign MHC. Cell T-cell specificity and the structure of the anti-
129:135–146 gen receptor. Nature 321:219–226
32. Sha WC, Nelson CA, Newberry RD, Kranz 43. Akkaraju S, Ho WY, Leong D, Canaan K,
DM, Russell JH, Loh DY (1988) Selective Davis MM, Goodnow CC (1997) A range of
expression of an antigen receptor on CD8- CD4 T cell tolerance: partial inactivation to
bearing T lymphocytes in transgenic mice. organ-specific antigen allows nondestructive
Nature 335:271–274 thyroiditis or insulitis. Immunity 7:255–271
33. Garcia KC, Tallquist MD, Pease LR, Brunmark 44. Ho WY, Cooke MP, Goodnow CC, Davis MM
A, Scott CA, Degano M, Stura EA, Peterson (1994) Resting and anergic B cells are defective
PA, Wilson IA, Teyton L (1997) Alphabeta T in CD28-dependent costimulation of naive
cell receptor interactions with syngeneic and CD4+ T cells. J Exp Med 179:1539–1549
allogeneic ligands: affinity measurements and 45. Apostolou I, Von Boehmer H (2004) The
crystallization. Proc Natl Acad Sci U S A TCR-HA, INS-HA transgenic model of
94:13838–13843 autoimmune diabetes: limitations and expecta-
34. Pircher H, Burki K, Lang R, Hengartner H, tions. J Autoimmun 22:111–114
Zinkernagel RM (1989) Tolerance induction in 46. Bettelli E, Pagany M, Weiner HL, Linington
double specific T-cell receptor transgenic mice C, Sobel RA, Kuchroo VK (2003) Myelin oli-
varies with antigen. Nature 342:559–561 godendrocyte glycoprotein-specific T cell
35. Mamalaki C, Elliott J, Norton T, Yannoutsos receptor transgenic mice develop spontaneous
N, Townsend AR, Chandler P, Simpson E, autoimmune optic neuritis. J Exp Med
Kioussis D (1993) Positive and negative selec- 197:1073–1081
tion in transgenic mice expressing a T-cell 47. Katz JD, Wang B, Haskins K, Benoist C,
receptor specific for influenza nucleoprotein Mathis D (1993) Following a diabetogenic T
and endogenous superantigen. Dev Immunol cell from genesis through pathogenesis. Cell
3:159–174 74:1089–1100
36. Robertson JM, Jensen PE, Evavold BD (2000) 48. Uematsu Y, Ryser S, Dembic Z, Borgulya P,
DO11.10 and OT-II T cells recognize a Krimpenfort P, Berns A, von Boehmer H,
C-terminal ovalbumin 323-339 epitope. J Steinmetz M (1988) In transgenic mice the
Immunol 164:4706–4712 introduced functional T cell receptor beta
37. Barnden MJ, Allison J, Heath WR, Carbone gene prevents expression of endogenous beta
FR (1998) Defective TCR expression in trans- genes. Cell 52:831–841
genic mice constructed using cDNA-based 49. Fenton RG, Marrack P, Kappler JW, Kanagawa
alpha- and beta-chain genes under the control O, Seidman JG (1988) Isotypic exclusion of
of heterologous regulatory elements. Immunol gamma delta T cell receptors in transgenic
Cell Biol 76:34–40 mice bearing a rearranged beta-chain gene.
38. Murphy KM, Heimberger AB, Loh DY (1990) Science 241:1089–1092
Induction by antigen of intrathymic apoptosis 50. Sarukhan A, Garcia C, Lanoue A, von Boehmer
of CD4+CD8+TCRlo thymocytes in vivo. H (1998) Allelic inclusion of T cell receptor
Science 250:1720–1723 alpha genes poses an autoimmune hazard due
39. Kaye J, Vasquez NJ, Hedrick SM (1992) to low-level expression of autospecific recep-
Involvement of the same region of the T cell tors. Immunity 8:563–570
antigen receptor in thymic selection and for- 51. Mombaerts P, Iacomini J, Johnson RS, Herrup
eign peptide recognition. J Immunol K, Tonegawa S, Papaioannou VE (1992)
148:3342–3353 RAG-1-deficient mice have no mature B and T
40. Kaye J, Hsu ML, Sauron ME, Jameson SC, lymphocytes. Cell 68:869–877
Gascoigne NR, Hedrick SM (1989) Selective 52. Shinkai Y, Rathbun G, Lam KP, Oltz EM,
development of CD4+ T cells in transgenic Stewart V, Mendelsohn M, Charron J, Datta
mice expressing a class II MHC-restricted anti- M, Young F, Stall AM et al (1992) RAG-2-
gen receptor. Nature 341:746–749 deficient mice lack mature lymphocytes owing
Genetic Tools to Study T Cell Development 45
to inability to initiate V(D)J rearrangement. 57. Bruno L, Fehling HJ, von Boehmer H (1996)
Cell 68:855–867 The alpha beta T cell receptor can replace the
53. Huesmann M, Scott B, Kisielow P, von gamma delta receptor in the development of
Boehmer H (1991) Kinetics and efficacy of gamma delta lineage cells. Immunity
positive selection in the thymus of normal and 5:343–352
T cell receptor transgenic mice. Cell 58. DiPaolo RJ, Unanue ER (2001) The level of
66:533–540 peptide-MHC complex determines the sus-
54. Bettini ML, Bettini M, Vignali DA (2012) ceptibility to autoimmune diabetes: studies in
T-cell receptor retrogenic mice: a rapid, flexi- HEL transgenic mice. Eur J Immunol 31:
ble alternative to T-cell receptor transgenic 3453–3459
mice. Immunology 136:265–272 59. Byersdorfer CA, Schweitzer GG, Unanue ER
55. Hwang S, Song KD, Lesourne R, Lee J, (2005) Diabetes is predicted by the beta cell level
Pinkhasov J, Li L, El-Khoury D, Love PE of autoantigen. J Immunol 175:4347–4354
(2012) Reduced TCR signaling potential 60. Baldwin TA, Sandau MM, Jameson SC,
impairs negative selection but does not result Hogquist KA (2005) The timing of TCR{alpha}
in autoimmune disease. J Exp Med expression critically influences T cell develop-
209:1781–1795 ment and selection. J Exp Med 202:111–121
56. Takahama Y, Shores EW, Singer A (1992) 61. Turner VM, Gardam S, Brink R (2010)
Negative selection of precursor thymocytes Lineage-specific transgene expression in hema-
before their differentiation into CD4+CD8+ topoietic cells using a Cre-regulated retroviral
cells. Science 258:653–656 vector. J Immunol Methods 360:162–166
Chapter 4
Abstract
T cell development is a complex multistep process that requires the coordinated activation of distinct
signaling responses and the regulated progression of developing cells (thymocytes) through key stages
of maturation. Although sophisticated techniques such as fetal thymus organ culture, in vitro thymocyte
culture, and multi-parameter flow cytometric analysis are now widely employed to evaluate thymocyte
maturation by experienced laboratories, defects in T cell development can usually be identified with
more simplified screening methods. Here, we provide a basic protocol for assessment of T cell develop-
ment that will enable laboratories with access to a four parameter flow cytometer to screen mouse
strains, including those generated from embryonic stem cells with targeted gene mutations, for thymo-
cyte maturation defects.
Key words T cell development, Thymocytes, T lymphocytes, Flow cytometry, Antibody staining,
Intracellular staining, αβ T cells, γδ T cells, NKT cells, Regulatory T cells
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_4, © Springer Science+Business Media New York 2016
47
48 Jan Y.M. Lee and Paul E. Love
a 104
CD4 SP DP
CD4SP
3
10
(4-9%) DP
(84-92%)
102
CD8 SP
CD8SP
(2-3%)
101
DN
(2-4%)
DN
CD4
100
0
10 101 102 103 104
CD8
b
Stage 1 Stage 2
ISP
DN DN
CD4
CD4
CD8 CD8
Stage 4 Stage 3
CD4SP
DP DP
CD8SP
ISP
CD4
CD4
CD8 CD8
Fig. 1 Major stages of thymocyte development. Thymocytes are stained with
anti-CD4 and anti-CD8α. (a) The classic “bird” profile of thymocytes stained with
anti-CD4 and anti-CD8α, showing the four major populations (DN double nega-
tive, DP double positive, CD4 SP CD4 single positive, and CD8 SP CD8 single
positive). Also shown is the expected percentage of each population for a wild-
type mouse thymus. (b) Stage 1: CD4−CD8− (DN double negative); Stage 2:
CD4−CD8+TCRβlow/− (ISP immature CD8 single positive); Stage 3: CD4+CD8+ (DP
double positive); Stage 4: CD4+CD8− (CD4 SP CD4 single positive) or CD4−CD8+
(CD8 SP CD8 single positive). Note that DP thymocytes transition through a
CD4+CD8low intermediate stage before becoming either CD4 SP or CD8 SP
Assessment of T Cell Development by Flow Cytometry 49
a
20000 20000
15000 15000
10000
# of Cells
# of Cells
10000
5000 5000
0 0
100 101 102 103 104 100 101 102 103 104
TCRβ CD5
DN 15000
TCRβ+
600
900
2000 ISP
10000 400
600
1000
300 5000 200
0 0 0 0
# of Cells
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
TCRβ
1000
400
20000 2500
800
2000 300
15000
600
1500
10000 200
400 1000
5000 100
200 500
0 0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
CD5
Fig. 2 TCRβ and CD5 surface expression on thymocytes. Thymocytes are stained with anti-CD4, anti-CD8α,
anti-TCRβ, and anti-CD5. (a) TCRβ and CD5 levels on total thymocytes. (b) TCRβ and CD5 levels on DN, DP, CD4
SP, and CD8 SP thymocyte populations. The TCRβlow/− cells in the CD8 SP population are ISPs
a b
104 104
Lineage
Lineage Mix
101 101
DN4 DN3
Negative
Cells
CD44
100 100
0 200 400 600 800 1000 100 101 102 103 104
Fig. 3 Stages of double negative (DN) thymocyte development. Thymocytes are stained with anti-CD25, anti-CD44,
and a lineage mix containing the following markers: anti-CD4, anti-CD8α, anti-TCRβ, anti-TCRγδ, anti-CD19,
anti-NK1.1, anti-DX5, anti-Gr1 (Ly6C/G), anti-CD11b, and anti-Ter119 to facilitate gating on DN cells (see Note 1).
All the antibodies used in the lineage mix are conjugated to the same fluorochrome. (a) Shown is a Lineage Mix
vs. Forward Scatter (FSC) plot of total thymocytes and the gate used to analyze DN cells. (b) Gating on thymocytes
that are negative for the lineage mix, the DN subsets are revealed by the CD44 vs. CD25 plot
a 104
b 104
CD4 SP
103 103 ISP
ISP+CD8 SP
DP
102 102
CD8 SP
ISP
+
DN 101 101
CD8
CD4
CD4
SP
CD24
100 100
100 101 102 103 104 100 101 102 103 104
CD8 CD8 TCRb
c
100
60 CD8 SP
40
20
0
100 101 102 103 104 100 101 102 103 104
CD5 TCRb
* Iso Ctrl = Isotype Control
Fig. 4 Immature CD8 single positive thymocytes (ISPs) are intermediates between the DN and DP stages.
Thymocytes are stained with anti-CD4, anti-CD8α, anti-TCRβ, and anti-CD24 (HSA) or anti-CD5. (a) Focusing
on the ISP/CD8 SP gate of the thymus “bird” profile. (b) ISPs can be distinguished from more mature CD8 SP
thymocytes by staining for CD24 and TCRβ. ISPs are CD24hiTCRβlow/− whereas CD8 SP cells are CD24int/
low
TCRβhi (Gated on CD8 SP). (c) ISPs express low surface levels of CD5 and TCRβ compared to more mature
CD8 SP thymocytes
a b
CD4 SP
DP 4 3
CD8 SP
1
ISP
DN
+
CD4
TCRβ
CD8 CD69
c 104
d 104
DP gated
103 103
4 3
DP
102 102
101 101
2
1
TCRβ
CD4
100 100
100 101 102 103 104 100 101 102 103 104
CD8 CD69
Fig. 5 Assessment of Double Positive (DP) thymocyte development. Thymocytes are stained with anti-CD4,
anti-CD8α, anti-TCRβ, and anti-CD69. (a) Focusing on the DP population in the thymus “bird” profile. (b) Gating
on the DP population, the four stages of DP development can be visualized by TCRβ vs. CD69 expression: Stage
1, TCRβlow/−CD69low/−; Stage 2, TCRβlowCD69low; Stage 3, TCRβhiCD69hi; Stage 4, TCRβhi CD69low. (c) CD4 vs. CD8
plot of total thymocytes as depicted in (a). (d) TCRβ vs. CD69 plot of DP thymocytes as depicted in (b)
a
CD4 SP
DP
CD8 SP
DN
CD4
CD8
b 104 104 104
CD4 SP CD4 SP CD8 SP
10
3
103 103 ISP
Immature
Immature
101 101 101
Mature Mature
CD24
CD4
0 0 0
10 10 10
0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10 100 101 102 103 104
CD8 TCRβ
c 4
104 104
10
CD4 SP CD4 SP CD8 SP
103 Mature 10 3 Mature
103
2 2
10 10
102
CD8 SP
Immature Immature
101 101
101
TCRβ
CD4
0 100 100
10
100 101 102 103 10
4
10
0
10
1
10
2
10
3
104
100 101 102 103 104
CD8 CD69
Fig. 6 Assessment of Single Positive (SP) thymocyte maturation. Thymocytes are stained with anti-CD4, anti-
CD8α, anti-TCRβ, and anti-CD24. (a) Focusing on the CD4 SP and CD8 SP thymocyte populations. (b) Gating
on either CD4 SP or CD8 SP population, one can differentiate between immature (TCRβhiCD24hi) or mature
(TCRβhiCD24low) SP thymocytes. Note that in the case of CD8 SP, the ISPs are also visible as TCRβlow/−CD24hi
cells. (c) CD69 can also be used as a maturation marker. Gating on CD4 SP or CD8 SP population, immature
SP cells are TCRβhiCD69hi while mature SP cells are TCRβhiCD69low/−
2 Materials
a 10
4 b 10
4
CD4 SP T cells
CD4SP 10
3
10
3
2 2
10 10
B cells
B cells,
1 1
MF, NK, CD8SP 10 CD8 10
etc. SP
CD3e
CD4
CD4
0 0
10 10
0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10
c 1500 1500
CD4SP CD8SP
1000 1000
500 500
# of Cells
0 0
0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10
TCRb
Fig. 7 T cell population in the periphery. Lymph node cells are stained with anti-CD4, anti-CD8α, anti-CD3ε,
and anti-TCRβ or anti-B220 (CD45R). (a) CD4 SP, CD8 SP, and CD4−CD8− cells in the lymph nodes. (b) Peripherial
T and B cells can be differentiated by the expression of CD3ε and B220, respectively. The majority of CD4−CD8−
cells in the lymph nodes are B cells with a very small population of NKT, macrophages, etc. (c) TCRβ levels are
high on CD4 SP and CD8 SP cell populations
a 4
10
CD4 SP
3
10
2
10
1
10 CD8
SP
CD4
0
10
0 1 2 3 4
10 10 10 10 10
CD8
b 4
CD4 SP 4
CD8 SP
10 10
Central 3 3
Naïve 10 10
Memory
2 2
10 10
Effector 1
10 1
10
Memory
CD62L
CD62L
0 0
10 10
0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10
CD44 CD44
Fig. 8 Naïve and memory CD4 SP and CD8 SP cells in the periphery. Lymph node cells are stained with anti-
CD4, anti-CD8α, anti-CD62L, and anti-CD44. (a) Focusing on CD4 SP and CD8 SP cells in the lymph nodes. (b)
Gating on either CD4 SP or CD8 SP lymph node T cells, the naïve and memory (central and effector, respec-
tively) phenotypes can be differentiated by CD62L vs. CD44
a 10
4
CD4 SP
10
3
DP
2
10
CD8 SP
1
10
DN
CD4
0
10
0 1 2 3 4
10 10 10 10 10
CD8
b 4
DN 4 DP 4 CD4 SP 4 CD8 SP
10 10 10 10
3 3 3 3
10 10 10 10
2 2 2 2
10 10 10 10
1 1 1 1
10 10 10 10
CD3e
0 0 0 0
10 10 10 10
0 1 2 3 4 0 1 2 3 4 0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10
3 3 3 3
10 10 10 10
2 2 2 2
10 10 10 10
1 1 1 1
10 10 10 10
CD3e
0 0 0 0
10 10 10 10
0 1 2 3 4 0 1 2 3 4 0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10 10
800 3 3
10 10
600
2 2
10 10
400
1 1
10 10
200
CD3e
FSC
CD3e
0 0 0
10 10
0 1 2 3 4
10 10 10 10 10 0 1 2 3 4 0 1 2 3 4
10 10 10 10 10 10 10 10 10 10
Fig. 9 Detection of Natural Killer T cells (NKT) in the thymus and periphery. Thymocytes are stained with anti-
CD4, anti-CD8α, anti-CD3ε, and α-Gal/Cer CD1d tetramer or “empty” CD1d tetramer. Splenocytes are stained
with anti-CD4, anti-CD3ε, α-Gal/Cer CD1d tetramer or “empty” CD1d tetramer, and an antibody mixture of
anti-CD8α + anti-CD19 + anti-B220. (a) Focusing on all populations of the thymus “bird” profile. (b) CD3ε vs.
α-Gal/Cer CD1d (or “empty” CD1d) tetramer surface expression on DN, DP, CD4 SP, and CD8 SP populations in
the thymus. (c) Gating on CD8, CD19, and B220 negative cells in the spleen. (d) CD3ε vs. α-Gal/Cer CD1d (or
empty CD1d) tetramer surface expression on splenocytes that are CD8/CD19/B220 negative. Expected % of
CD1d+ cells in a C57BL/6 mouse thymus: <1 % in DP and CD8 SP and ~2–3 % in DN and CD4 SP, respectively.
Expected % of CD1d+ cells in the spleen is ~3 %
Assessment of T Cell Development by Flow Cytometry 57
a CD4 SP CD4 SP
104 104 104
CD4 SP DP CD25+
CD4 SP FoxP3+
103 103 103
FoxP3+
102 102 102
CD8 SP
101 101 101
DN
CD25
CD4
CD4
100 100 100
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
b CD4 SP
CD4 SP 104
104 104
CD25+
CD4 SP
103
FoxP3+
103 103
102 102
102
Conv. CD4 SP
CD4 SP FoxP3+ 101
101 CD8 101
SP CD25
CD4
CD4
100 100
100
100 101 102 103 104 100 101 102 103 104
100 101 102 103 104
3 Methods
a 104 10
4
CD3e+ TCRgd +
10
3
103
2
102 10
Lineage Mix
101
Lineage 10
1
Negative
TCRgd
Cells 100
100
0 1 2 3 4
0 200 400 600 800 1000 10 10 10 10 10
10
3 103 CD3e+ TCRgd +
102 102
Lineage Mix
Cells 0
100 10
0 200 400 600 800 1000 100 101 102 103 104
Fig. 11 γδ T cells in the thymus and periphery. Thymocytes are stained with anti-CD3ε, anti-TCRγδ, and lin-
eage mix that contains anti-CD4, anti-CD8α, anti-TCRβ, anti-CD19, anti-NK1.1, anti-DX5, anti-Gr1 (Ly6C/G),
anti-CD11b, and anti-Ter119. Lymph node cells are stained with anti-CD3ε, anti-TCRγδ, and a lineage mix that
contains anti-CD4, anti-CD8α, and anti-TCRβ. (a) Gating on thymocytes that are lineage negative, TCRγδ+ cells
can be detected as CD3ε+TCRγδ+. Because of their low numbers, it is advantageous to use a lineage mix and
acquire multiple events on the flow cytometer. (b) Gating on lymph node cells that are lineage negative,
TCRγδ+ lymphocytes can be detected as CD3ε+TCRγδ+. Expected % of TCRγδ+ cells in a C57BL/6 mouse:
~1–2 % in the thymus and ~3 % in the lymph nodes
3.2 Cell Surface 1. Add one million cells in a sample tube (see Note 4).
Staining 2. Add 2 mL of cell staining buffer to the tube and centrifuge for
with Fluorochrome 5 min at 450 × g, 4 °C.
Conjugated Antibodies
3. Vacuum-aspirate excess buffer, leaving cell pellet undisturbed
at the bottom of the tube. Excess buffer can also be decanted.
4. Stain with antibody mix* (see below) for 30 min at 4 °C in the
dark (see Notes 5 and 6). Taking into account residue buffer
from the cell pellet, final volume of cells AND antibody mix
should be between 50 and 100 μL.
*Composition of Antibody Mix:
10 μL of FcγIII Receptor (CD16) clone 2.4G2 (titered)
10 μL of FITC labeled antibody (titered)
10 μL of PE labeled antibody (titered)
10 μL of PerCp-Cy5.5 antibody (titered)
10 μL of APC antibody (titered)
50 μL Total volume
4 Notes
a 4
b 10
4
10
7AAD+ 7AAD+
3
10
3 10
2
10
2 10
1
10
1 10
7AAD
7AAD
7AAD- 0
7AAD-
10
0 10
0 200 400 600 800 1000 0 200 400 600 800 1000
0 0 0
0 200 400 600 800 1000 0 200 400 600 800 1000 0 200 400 600 800 1000
gate
800
600
400
200
0
0 200 400 600 800 1000
References
1. Anderson G, Moore NC, Owen JJ, Jenkinson entiations. Annu Rev Cell Dev Biol 17:
EJ (1996) Cellular interactions in thymocyte 387–403
development. Annu Rev Immunol 14:73–99 4. Godfrey DI, Kennedy J, Suda T, Zlotnik A
2. Petrie HT (2003) Cell migration and the con- (1993) A developmental pathway involving
trol of post-natal T-cell lymphopoiesis in the four phenotypically and functionally distinct
thymus. Nat Rev Immunol 3(11):859–866 subsets of CD3-CD4-CD8- triple-negative
3. Weissman IL, Anderson DJ, Gage F (2001) adult mouse thymocytes defined by CD44
Stem and progenitor cells: origins, pheno- and CD25 expression. J Immunol 150(10):
types, lineage commitments, and transdiffer- 4244–4252
64 Jan Y.M. Lee and Paul E. Love
5. Shortman K, Wu L (1996) Early T lymphocyte 10. Sprent J, Surh CD (2002) T cell memory.
progenitors. Annu Rev Immunol 14:29–47 Annu Rev Immunol 20:551–579
6. Von Boehmer H (1995) Control of T-cell 11. Bendelac A, Savage PB, Teyton L (2007) The
development by the pre-T and alpha beta biology of NKT cells. Annu Rev Immunol
T-cell receptor. Ann N Y Acad Sci 766:52–61 25:297–336
7. Michie AM, Zuniga-Pflucker JC (2002) 12. Sakaguchi S (2004) Naturally arising CD4+
Regulation of thymocyte differentiation: pre- regulatory t cells for immunologic self-
TCR signals and beta-selection. Semin tolerance and negative control of immune
Immunol 14(5):311–323 responses. Annu Rev Immunol 22:
8. Jameson SC, Hogquist KA, Bevan MJ (1995) 531–562
Positive selection of thymocytes. Annu Rev 13. Hayday AC (2000) [gamma][delta] cells: a
Immunol 13:93–126 right time and a right place for a conserved
9. Sebzda E et al (1999) Selection of the T cell third way of protection. Annu Rev Immunol
repertoire. Annu Rev Immunol 17:829–874 18:975–1026
Chapter 5
Abstract
The parenchyma of the thymus is compartmentalized into the cortex and the medulla, which are
constructed by cortical thymic epithelial cells (cortical TECs, cTECs) and medullary thymic epithelial cells
(mTECs), respectively. cTECs and mTECs essentially and differentially regulate the development and
repertoire selection of T cells. Consequently, the biology of T cell development and selection includes the
study of TECs in addition to the study of developing T cells and other hematopoietic cells including den-
dritic cells. In this chapter, we describe the methods for flow cytometric analysis and sorting of TECs and
their subpopulations, including cTECs and mTECs.
Key words Thymic epithelial cell, mTEC, cTEC, Aire, β5t, Flow cytometry, Cell sorting
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_5, © Springer Science+Business Media New York 2016
65
66 Izumi Ohigashi and Yousuke Takahama
2 Materials
2.1 Dissociation 1. Euthanasia chamber.
of Stromal Cells 2. Scissors and forceps.
from Thymus
3. PBS, pH 7.2.
4. RPMI 1640 medium.
5. Collagenase solution: RPMI 1640 supplemented with 0.125 %
collagenase D and 0.01 % DNase I (see Note 1).
6. PBS + EDTA buffer: PBS supplemented with 2 mM EDTA and
2 % fetal calf serum (FCS).
7. 30-mm dish.
8. 1.5-ml microtubes.
9. 1-ml syringes.
10. 26-gauge needles.
11. 100-μm nylon mesh.
12. Water bath or block incubator.
2.3 Intracellular 1. Antibodies specific for β5t and Aire (see Note 3).
Staining for Flow 2. FACS buffer (see Subheading 2.2).
Cytometric Analysis
3. 2 % Paraformaldehyde: PBS supplemented with 2 %
paraformaldehyde.
Flow Cytometry Analysis of Thymic Epithelial Cells and Their Subpopulations 67
3 Methods
Fig. 1 Representative flow cytometry profiles of TECs. Collagenase-digested thymus cells from 3-week-old
C57BL/6 mice (as in Subheading 3.1) were stained with antibodies specific for CD326 (PECy7), CD45 (FITC),
and CD205 (APC), as well as with UEA-1 (APCeFluor780) and propidium iodide (PI) (as in Subheading 3.2). (a)
Profiles show CD45 and CD326 expression in PI− total viable cells (left) and CD205 and UEA-1 expression in
CD45−CD326+ PI− viable cells (middle) and CD45+CD326+ PI− viable cells (right) (see Note 11). Numbers in
dot plots indicate frequency within indicated area. Most CD45−CD326+ PI− viable cells consisted of either
_
UEA1–CD205+ cTECs or UEA1+CD205 mTECs. CD45+CD326+ PI− viable cells consisted of thymocyte-associ-
ated cTECs and thymocyte-associated mTECs (see Note 11) as well as many UEA1–CD205– cells. (b) Profiles
show class II MHC (I-Ab) expression (solid lines) and isotype control (shaded histograms) in CD205−UEA-1+
UEA1+CD205– PI− viable mTECs (left) and UEA1–CD205+ CD45−CD326+ PI− viable cTECs (right). The class II
MHC expression indicates that the cells indeed represent TEC subpopulations
4 Notes
References
1. Petrie HT, Zúñiga-Pflücker JC (2007) Zoned 3. Anderson G, Takahama Y (2012) Thymic epi-
out: functional mapping of stromal signaling thelial cells: working class heroes for T cell
microenvironments in the thymus. Annu Rev development and repertoire selection. Trends
Immunol 25:649–679 Immunol 33:256–263
2. Klein L, Hinterberger M, Wirnsberger G, 4. Alves NL, Goff OR, Huntington ND (2009)
Kyewski B (2009) Antigen presentation in the Characterization of the thymic IL-7 niche
thymus for positive selection and central toler- in vivo. Proc Natl Acad Sci U S A 106:
ance induction. Nat Rev Immunol 9:833–844 1512–1517
Flow Cytometry Analysis of Thymic Epithelial Cells and Their Subpopulations 73
5. Hara T, Shitara S, Imai K et al (2012) 12. Ueno T, Saito F, Gray DH et al (2004) CCR7
Identification of IL-7 producing cells in primary signals are essential for cortex-medulla migra-
and secondary lymphoid organ using IL7-GFP tion of developing thymocytes. J Exp Med
knock-in mice. J Immunol 189:1577–1584 200:493–505
6. Hozumi K, Mailhos C, Negishi N et al (2008) 13. Liu C, Saito F, Liu Z et al (2006) Coordination
Delta-like 4 is indispensable in thymic environ- between CCR7- and CCR9-mediated chemo-
ment specific for T cell development. J Exp kine signals in prevascular fetal thymus coloni-
Med 205:2507–2513 zation. Blood 108:2531–2539
7. Koch U, Fiorini E, Benedito R et al (2008) 14. Laufer TM, DeKoning J, Markowitz JS et al
Delta-like 4 is the essential, nonredundant (1996) Unopposed positive selection and
ligand for Notch1 during thymic T cell lineage autoreactivity in mice expressing class II MHC
commitment. J Exp Med 205:2515–2523 only on thymic cortex. Nature 383:81–85
8. Murata S, Sasaki K, Kishimoto T et al (2007) 15. Gray DH, Chidgey AP, Boyd RL (2002)
Regulation of CD8+ T cell development by Analysis of thymic stroma cell populations
thymus-specific proteasomes. Science using flow cytometry. J Immunol Methods
316:1349–1353 260:15–28
9. Nitta T, Murata S, Sasaki K et al (2010) 16. Williams KM, Mella H, Lucas PJ et al (2009)
Thymoproteasome shapes immunocompetent Single cell analysis of complex thymus stroma
repertoire of CD8+ T cells. Immunity 32:29–40 cell populations: rapid thymic epithelial prepa-
10. Nagamine K, Peterson P, Scott HS et al (1997) ration characterizes radiation injury. Clin
Positional cloning of the APECED gene. Nat Transl Sci 2:279–285
Genet 17:393–398 17. Nakagawa Y, Ohigashi I, Nitta T et al (2012)
11. Anderson MS, Venanzi ES, Klein L et al Thymic nurse cells provide microenvironment
(2002) Projection of an immunological self for secondary TCRα rearrangement in cortical
shadow within the thymus by the Aire protein. thymocytes. Proc Natl Acad Sci U S A 109:
Science 298:1395–1401 20572–20577
Chapter 6
Abstract
Immunofluorescence analysis of thymic tissue sections is an indispensable technique for visualizing spatial
relationships among thymocyte and stromal cell subsets. The thymus is organized into distinct microenvi-
ronmental zones in which particular thymic epithelial cell (TEC) subsets support specific stages of thymo-
cyte maturation. Conversely, thymocytes and lymphoid tissue inducer cells support functional maturation
of TECs. The composition and organization of TECs change during ontogeny to generate a maximally
functional organ in the young adult. Deterioration of thymic architecture and stromal organization occurs
with age as the thymus undergoes involution. Such changes can be monitored by immunofluorescent
staining of thymic sections obtained at different ages throughout the life-span. Here we describe methods
to generate frozen or paraffin-embedded thymic tissue sections for multicolor immunofluorescence stain-
ing using antibodies to surface and/or cytoplasmic antigens.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_6, © Springer Science+Business Media New York 2016
75
76 Virginia Bain and Ellen R. Richie
2 Materials
Fig. 1 Tissue morphology varies with fixation. Keratin 5 and keratin 8 staining of 4-week thymi. (a) Fresh-
frozen tissue is optimal for cytoplasmic markers such as keratin 5 and keratin 8 but is difficult to distinguish
nuclear morphology. (b) Fixed-frozen tissue gives better morphology but has less optimal cytoplasmic stain
and more tissue defects when sectioning. (c) Paraffin-embedded tissue gives clear nuclear morphology and
fewer tissue defects but requires antigen retrieval and has less optimal cytoplasmic or cell surface stain. Scale
bars indicate 50 μm
Fig. 2 Antibodies may require different tissue preparation. (a) Fresh-frozen tissue is required by some cell
surface markers such as the vascular marker CD31. (b) PFA fixation is required by some nuclear markers such
as the TEC-specific transcription factor Foxn1. Scale bars indicate 50 μm and tissue used is 4-week thymi
3 Methods
3.1 Tissue Dissected thymi from embryos or adult mice as well as whole
Preparation for embryos can be flash frozen for cryosectioning. Embryos and adult
Fresh-Frozen Tissue mice should be euthanized according to institutional guidelines.
1. Dab tissue gently on a Kimwipe to remove excess PBS.
2. Place the tissue in a cryomold and cover in OCT. Rotate the
tissue to make certain that it is completely surrounded by OCT
with no pockets of PBS or air.
3. Use forceps to hold the cryomold in liquid nitrogen. Remove
the mold as soon as the entire block has frozen to avoid crack-
ing (see Note 11).
4. Store frozen blocks at −80 °C until ready to cryosection.
5. Move frozen blocks to the cryostat at least 30 min before cut-
ting to allow the blocks to thaw.
6. Trim the block with a single-edged razor blade to reduce cut-
ting area.
7. Mount the blocks to specimen discs using enough OCT to
surround the base of the block on all sides and allow the addi-
tional OCT to completely freeze before sectioning.
8. Section thawed and trimmed blocks at desired thickness
(see Note 12).
9. Allow frozen sections to dry completely at room temperature
for 15 min after the final section on the slide is cut.
10. Store sectioned tissue at −80 °C until ready to stain. Avoid
repeated thawing of the slides.
11. When ready to stain, first allow slides to thaw for 15 min on
the bench top.
12. Fix slides in ice-cold acetone for 10 min (see Note 13).
13. Proceed to Subheading 3.4.
3.2 Tissue Tissue fixed for frozen embedding receives a short PFA fix and
Preparation for does not require antigen retrieval.
Fixed-Frozen Tissue
1. Collect thymi or embryos in PBS and store on ice until ready
to fix.
2. Fix tissue in cold 4 % PFA with duration depending on size of
embryo or thymi (see Note 14).
3. Wash fixed tissue three times for 5 min each in PBS
(see Note 15).
4. Dehydrate tissue in 5 % sucrose/PBS for 1 h at 4 °C. Replace
5 % sucrose/PBS with 15 % sucrose/PBS and leave at 4 °C
overnight.
5. Rinse tissue in OCT (see Note 16).
6. Place tissue in a cryomold and cover with OCT. Freeze on dry
ice (see Note 17).
80 Virginia Bain and Ellen R. Richie
3.3 Tissue Thymi or embryos collected for paraffin embedding require a lon-
Preparation for ger PFA fixation, must be thoroughly dehydrated, and require
Paraffin-Embedded antigen retrieval.
Fixed Tissue 1. Collect thymi or embryos in PBS and store on ice until ready
to fix.
2. Fix tissue in cold 4 % PFA with duration depending on the size
of embryo or thymi (see Note 18).
3. Wash fixed tissue three times for 5 min each.
4. Dehydrate tissue in 70 % ethanol, 90 % ethanol, 95 % ethanol,
and three 100 % ethanol washes at 4 °C. Dehydration time will
vary with size (see Note 19). Tissue can be stored at −20 °C in
the third 100 % ethanol wash.
5. When ready to embed, tissue must be cleared in xylenes. This
will remove fat and allow wax to permeate the tissue. Xylene
washes are also size dependent (see Note 20). Once the tissue
is translucent remove it from xylene, and place it in an embed-
ding cassette (see Note 21).
6. Cassettes containing cleared thymi or embryos are incubated
in three paraffin washes at 65 °C (see Note 22).
7. Place your specimen in a paraffin-embedding mold, fill with
warm paraffin, orient appropriately, and allow the block to
cool. Blocks may be stored at 4 °C.
8. Allow cold blocks to warm to room temperature. Cut off
excess wax from the block to form a square to allow for ribbon-
ing during sectioning.
9. Mount the block in the base of a cassette and clamp it to the
microtome for sectioning.
10. Because paraffin-embedded tissue is thoroughly dehydrated,
cells are condensed and thinner sections should be cut.
Do not exceed 8 μm of paraffin-embedded tissue for
immunofluorescence.
11. Cut sections may be kept in ribbons and floated on a warm
water bath (do not exceed 45 °C).
12. Slides must be thoroughly dried (see Note 23).
13. Dry slides can be stored in slide boxes at 4 °C indefinitely.
14. Allow slides to warm to RT. Use two sets of mailers: one set
used for dewaxing and rehydration and another set used for
antigen retrieval.
15. Fill one set of mailers 2/3rd full with AR buffer and place in a
300 mL beaker filled with 150 mL of water. Heat the water
Immunofluorescence on Thymic Sections 81
and buffer to 95 °C. Cover the beaker with foil to avoid excessive
evaporation (see Note 24).
16. Place slides in clean mailers and wash twice for 5 min each in
xylene to remove the paraffin.
17. Remove excess xylene from the tissue by washing twice for
2 min in 100 % ethanol.
18. Rehydrate tissue by 2-min washes in 90, 70, and 30 % ethanol.
Leave slides in a dH2O wash for at least 5 min.
19. Transfer slides from rehydration mailers to AR mailers (see
Note 25).
20. Tissue is antigen retrieved for 30 min at 95 °C. Keep the water
level in the beaker at the level of the slide labels (see Note 26).
21. Allow mailers of slides in hot AR buffer to cool on your bench
for 20 min with the lid open.
10. Lay slides flat on a Kimwipe and add at least 100 μL of mounting
media to the bottom edge of each slide (see Note 34).
11. Coverslip each slide and blot the edges of the slide to remove
excess liquid (see Note 35).
12. Slides may be stored in the dark at 4 °C for up to 2 weeks to
preserve fluorescence.
4 Notes
Fig. 3 Alternating slides allows for staining the same tissue with many markers. (a) When cutting through tis-
sue, adjacent sections are similar while further apart sections may have distinct features. (b) Consecutive
sections are placed on alternating slides to allow multiple stains to be performed on the same specimen while
still analyzing many parts of the specimen
84 Virginia Bain and Ellen R. Richie
(E11.5) or 30 min for E15.5. Adult thymi can be fixed for 30 min
and dissected embryonic thymi require shorter fixation times of
5 min for E12.5–E15.5 and 10 min for E16.5–E18.5. Tissue
which is insufficiently fixed will fail to stain for antibodies requir-
ing fixation while tissue which is over-fixed will autofluoresce.
15. Ten rapid successive PBS washes can be used instead of three
5-min washes to reduce autofluorescence.
16. Place a small amount of OCT in a Petri dish and coat the tissue
thoroughly to remove excess PBS. This will help to reduce
cracks when sectioning.
17. PFA-fixed tissue will crack when snap frozen in liquid nitro-
gen. Use dry ice instead.
18. Aim for the shortest fixation time that allows the tissue to survive
the embedding process. A good starting time for embryos is
30 min for E11.5 or 2 h for E15.5. PFA will not permeate the skin
of embryos older than E15.5 and it must be removed. Fix adult
thymi for 45 min. Tissue that is under-fixed will fall apart during
the embedding process or fail to stain for antibodies requiring fixa-
tion while tissue that is over-fixed will autofluoresce.
19. 20-min washes are sufficient for E11.5 embryos or similar
sized tissue. Older embryos will require longer washes. Adult
thymi should be kept at each stage for at least 1 h. Longer
dehydration washes do not harm the tissue.
20. Adult thymi require two 10-min washes, whereas fetal thymi
require two to three 5-min washes. It is best to check the tissue
frequently while clearing it by holding it to the light and look-
ing for translucency.
21. This is a critical step. Excess time in xylene can lead to brittle
tissue that causes the sections to crumble.
22. Paraffin permeabilization is size dependent. Find the time that
exposes your tissue to the least amount of heat. For E11.5
embryos three 20-min washes are sufficient. For adult thymi
three 2-h washes are necessary.
23. If you are in a hurry, slides may be sufficiently dry as soon as
4 h after sectioning. Spacing sections apart from one another
on the slide will expedite this process. It is best to dry slides
overnight to make certain that no water is left on the slide as
any water under the tissue will cause it to fall off the slide in
future steps.
24. This limits you to 20 slides—four mailers maximum. Processing
more than 20 slides can result in less than ideal antigen retrieval.
25. It is best to pour off the water before transfer. Do this step
quickly to avoid lowering the temperature of the AR buffer.
26. Both time and temperature are critical for antigen retrieval.
This is the most important step in paraffin embedding.
Immunofluorescence on Thymic Sections 85
Make certain that the beaker stays full of 95–100 °C water and
remove slides from the water bath at exactly 30 min.
27. Make your antibody mix at N + 1. (If you have 20 slides make
enough mix for 21 slides—2.1 mL of mix.)
28. Your chamber should contain the same liquid you incubate
in—in this case PBS. Cover the base of the slide box with
folded paper towels. Pour at least 25 mL of PBS in each side.
29. Antibodies that give low signal but little background give the
best results with an overnight incubation at 4 °C, while anti-
bodies with higher signal and background give better results
when incubated at RT for a shorter duration.
30. Tyramide streptavidin amplification will substantially boost the
signal but has the potential to significantly increase the back-
ground. If you use a TSA kit to amplify a primary antibody, use
a streptavidin secondary reagent for staining, step 7.
31. Good results can be obtained with highly absorbed secondary
antibodies conjugated with DyLight or Alexa Fluor fluoro-
chromes. Cy3 and Cy5 also give bright signals. FITC and
Texas Red conjugates give weaker signals due in part to the
rapid photobleaching and relatively higher background stain-
ing, respectively.
32. Longer incubations especially at 4 °C give higher background
and rarely amplify the signal.
33. DAPI can be added to the second PBS wash. DAPI concentra-
tion is determined by user preference (I use 1:15,000). Do not
add DAPI if you use an AMCA- or Alexa350-conjugated sec-
ondary reagent.
34. Do not exceed five slides at a time.
35. Lower the cover slip gradually from bottom to top to spread
the mounting media without introducing bubbles on the slide.
Alternatively, place the edge of the cover slip at the bottom of
the slide and allow adhesive forces to move the cover slip. Avoid
bubbles as attempts to remove them may damage your tissue.
References
1. van Ewijk W, Shores EW, Singer A (1994) microenvironment. Front Biosci 16:
Crosstalk in the mouse thymus. Immunol Today 2461–2477
15:214–217 5. Anderson G, Takahama Y (2012) Thymic epi-
2. Gray DH, Ueno T, Chidgey AP et al (2005) thelial cells: working class heroes for T cell
Controlling the thymic microenvironment. development and repertoire selection. Trends
Curr Opin Immunol 17:137–143 Immunol 33(6):256–263
3. Gordon J, Manley NR (2011) Mechanisms of 6. Petrie HT, Zúñiga-Pflücker JC (2007) Zoned
thymus organogenesis and morphogenesis. out: functional mapping of stromal signaling
Development 138:3865–3878 microenvironments in the thymus. Annu Rev
4. Manley NR, Richie ER, Blackburn CC et al Immunol 25:649–679
(2011) Structure and function of the thymic
Chapter 7
Abstract
Many analytical or cell culture procedures require homogeneous starting cell populations that cannot be
obtained directly from organ dissection. Here, we describe two enrichment procedures to achieve this goal
and discuss their respective advantages in specific experimental contexts. Notes in this chapter include
some tips on how to determine the appropriate level of purity (see Note 1).
Key words Single-cell suspension, T cell purification, Magnetic bead depletion, Flow cytometric
sorting
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_7, © Springer Science+Business Media New York 2016
87
88 Andrea C. Carpenter et al.
2 Materials
3 Methods
3. For the thymus, take two curved forceps and gently tease apart
the organ.
4. For the LNs and spleen, use the rubber end of the syringe
pestle to dissociate the cells from the organ tissue, and ensure
that you have dissociated any observable clumps as well. It is
helpful to use the lid of the dish to store 5 mL additional
medium for step 5. At this point, wash the used end of the
syringe pestle in this medium, by swirling it in the medium;
then discard the pestle.
5. Tilt the dish containing the cells at a 30° angle, draw up the
medium with the free cells, and wash this medium back over the
dish. Gently pipet up the medium from the dish again and run
it through the 4 cm2 100 μm filter into a 15 mL conical tube.
Before passing the medium through the filter, use the pipet tip
to push the filter half way into the top of the tube, and then pull
the pipet tip a few mm back from the filter. Pulling the pipet
back prevents the medium from directly hitting the filter and
splashing out of the tube. Leave the filter balanced in the top of
the conical tube. Next, use the additional 5 mL medium to wash
the dish and pass that through the same filter into the conical
tube. The dish and filter can now be discarded. Pellet the cells at
150 × g, for 5 min at 4 °C, and decant the supernatant.
6. For spleen cell preparation, lyse red blood cells by resuspend-
ing the cell pellet in 2 mL red cell lysing buffer for 2 min on
ice. Then immediately, dilute out the red cell lysing buffer by
adding 8 mL complete medium. Pellet the cells at 150 × g, for
5 min at 4 °C, and decant the supernatant.
7. Resuspend the organs at 10–20 × 106 cells/mL in complete
medium (see Note 5 on expected yields).
8. Count the cells using a dye that distinguishes dead cells (for
example: trypan blue). Calculate the cell concentration and
total number. Set aside an aliquot (106 cells is ideal) from each
starting cell suspension for flow cytometric analysis.
9. In unmanipulated mice, spleen and LN cells can be combined
to maximize the size of the starting population. For T cells,
when large cell numbers are not needed, LN may be a prefer-
able starting point because of the greater frequency of T cells
in LN than in spleen.
3.2 Magnetic Bead Several commercial kits are available to purify target populations
Purification of T Cell (e.g., CD4 or CD8 T cells), most of them using proprietary
Subsets by Depletion reagents and materials (magnets). The following procedure uses
Dynabeads (LifeTech) to negatively select the desired population.
Provided the beads carry the appropriate secondary reagent,
Dynabeads can also be used with user-provided antibodies (see
Note 6). Volumes and bead numbers are given for 107 starting
92 Andrea C. Carpenter et al.
3.3 Magnetic Bead A similar procedure used in Subheading 3.2 can be used to increase
Enhancement Pre-flow the frequency of rare target cells prior to flow cytometric sorting.
Cytometric Sorting This is especially useful when starting from high cell numbers
(>108) for which sorting times quickly become prohibitive. In this
case, because the objective is to remove most of the nontarget cells
rather than achieving high purity, significant savings can be achieved
by lowering bead numbers and antibody concentrations. While
pilot experiments are needed to titer down these reagents, a good
starting point is to use 1/8 of the recommended amounts of beads
and antibodies while keeping cell concentration intact and all incu-
bations at 4 °C. This “light” procedure routinely achieves >40 %
cell purity with very little loss of desired cells.
1. Pellet cells to be run through kit by spinning at 150 × g, for
5 min, at 4 °C.
2. Resuspend cells in 100 μL isolation buffer per 107 cells. If any
cell clumping is observed, refilter before proceeding.
3. Add 20 μL FBS cells and 2.5 μL kit antibody.
4. Incubate for 20 min at 2–8 °C (wash beads during this incuba-
tion and keep on ice until needed; see steps 5 and 6).
5. To wash the beads: first resuspend beads in the vial by vortex-
ing for 30 s. Pipet 25 μL beads into a new 15 mL tube and
resuspend in equal volume or at least 1 mL isolation buffer.
6. Put the tube without the lid on magnet (DynaMag 15, 50, or
equivalent) for 1 min, remove buffer by pipetting, and discard.
Remove tube from magnet and resuspend beads in the same
volume of isolation buffer as the original bead volume.
7. After antibody incubation, wash cells by adding 2 mL isolation
buffer. Pellet the cells at 150 × g, for 5 min, at 4 °C and decant
the supernatant.
8. Resuspend cells in 975 μL isolation buffer per 107 cells and add
pre-washed beads.
9. Incubate for 15 min at 4 °C with tilt and rotation on a rocker
at a speed sufficient to keep the beads in suspension. (The 4 °C
incubation reduces efficiency, but better preserves cell viability
in advance of flow cytometric sorting.)
10. Add 1 mL isolation buffer per 107 cells, and gently pipet up
and down five times with a large-bore pipet (5 mL or similar).
11. Put tube without lid on magnet for 2 min.
12. Transfer supernatant containing enhanced homogeneity to a
new tube, and wash with complete medium as soon as possible
after transferring.
13. Count cells to determine recovery and take an aliquot of 105
cells to quantify purity by flow cytometric analysis (see Notes 7
and 8).
14. Purity will be >40 % with very little loss of desired cells.
94 Andrea C. Carpenter et al.
3.4 Flow Cytometric Before sorting cells, consider doing a pre-depletion with beads (see
Sorting Subheading 3.3), especially when preparing rare cells (<5 % of the
input). Although this increases the pre-sort preparation time, it
often reduces the total cell manipulation time before downstream
procedures. When the target frequency is under 1 %, the pre-
depletion strategy should seriously be considered as the sorter
error rates can substantially decrease purity in this situation (also
see Subheading 3.4.2). The procedure outlined below assumes
basic understanding of flow cytometry procedures and does not
address operation of the cell sorter itself, which is generally left to
a professional operator.
3.4.1 Flow Cytometric 1. Plan ahead. Consult the flow cytometry facility and inquire
Sorting: General Procedure about specifics of available equipment and procedures.
Discuss the choice of fluorochromes and evaluate the time
needed for cell purification and to verify purities. It is also
important to choose a sorting nozzle of appropriate diame-
ter, as a general rule at least five times as large as the cells in
the starting population. For lymphocytes, a 70 μm nozzle
provides adequate speed without damaging the cells.
However, a 100 μm nozzle may be better suited when sorting
larger cells or when using the double-sort procedure as
described in Subheading 3.4.2. For most applications, cells
should be kept cold, and it is important to verify that the
sorter has a chillable (4 °C) sample holder.
2. Resuspend cells to be stained for flow sorting at 2 × 107 cells/mL
complete medium (use 15 mL tube for <6.0 × 107 cells, 50 mL
for >6.0 × 107 cells). Before staining for sort, a titration of the
antibodies is advisable; 0.25 μg antibody per 107 cells is a good
starting point. If using the protocol in Subheading 3.3 prior to
sorting, ensure that the antibodies used for flow cytometric
sorting were not blocked by the antibodies in the bead kit (see
Notes 7–9).
3. Do not forget to prepare cell samples for flow cytometer setup
if needed. Typically set aside 0.5 × 106 cells per compensation
tube. These will be the single-color tubes used to set up the
flow cytometric sorter or analyzer for purity checks.
4. Add appropriate antibodies, mix well, and incubate at 4 °C for
45 min in the dark.
5. Wash with complete medium using four times staining vol-
ume. Pellet the cells at 150 × g, for 5 min, at 4 °C and decant
the supernatant.
6. Resuspend cells to be flow sorted at 2 × 107 cells/mL complete
medium in 5 mL round-bottom polystyrene tubes with caps.
Purification of Thymocyte and T Cell Subsets 95
3.4.2 Secondary Protocol This procedure uses two consecutive sorts to isolate good purity
for Sorting Populations (>95 %) populations from very rare cells (less than 0.01 % of the
≥0.01 % starting population) if bead isolation procedures are unavailable or
inadequate for pre-sort enrichment (see Subheading 3.3).
1. Perform the primary sort following the procedure defined in
Subheading 3.4.1 through step 10 with the following changes:
(a) Only use a 100 μm nozzle.
(b) In step 8, use a wide gate to select the entire desired popu-
lation and maximize yields. Unwanted cells gated in error
will be removed in the second sort.
2. After step 10, pellet the cells at 150 × g, for 7 min, at 4 °C and
decant the supernatant.
3. Resuspend the cells in a small volume, ≥300 μL complete
medium.
4. Perform the secondary sort, using a tight gate that is slightly
inside the edges of the desired population. This will increase
purity to acceptable levels (>95 %).
5. Follow steps 10–13 in Subheading 3.4.1.
96 Andrea C. Carpenter et al.
4 Notes
Abstract
Transduction of lymphoid progenitors with retroviral or lentiviral vectors is a powerful experimental strategy
to tease out the role of a gene or pathway in T cell development via gain-of-function or loss-of-function
strategies. Here we discuss different approaches to use this powerful technology, and present some protocols
that we use to transduce murine HSCs, thymocytes, and lymphoid cell lines with these viral vectors.
Key words Retrovirus, Hematopoietic stem cell, Thymocytes, Bone-marrow chimera, Development
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_8, © Springer Science+Business Media New York 2016
99
100 Amie Simmons and José Alberola-Ila
2 Materials
3 Procedures
3.2 Transduction In general, the method involves plating cells in 24-well plates
with 2 ml of fresh warm viral supernatant and 4 μg/ml lipo-
fectamine or 5 μg/ml polybrene, centrifuging cells at 20 °C for
1–1.5 h at 460 × g, culturing cells for 1 h at 37 °C, and then
replacing the viral supernatant with media (see Note 15); addi-
tional details are provided below. These approaches yield good
efficiency of infection when coupled with a good virus, e.g.,
>90 % with lymphoid lines, up to 60 % with fetal liver and early
thymocytes, 5–20 % with DP thymocytes, and 15–30 % with
enriched bone marrow cells.
3.2.1 Retroviral 1. Day 0: Cells are plated in BMGM, and cultured for 24–36 h at
Transduction 2 × 106 cells/ml (the yield is approximately 2–5 × 106 cells per
Hematopoietic Cells 5-FU treated mouse).
2. Day 1: Use freshly produced virus, or thaw on ice an aliquot of
previously frozen virus. Calculate how much virus you will
need, depending on how many cells are going to be infected.
For a 6-well plate we use 2 ml viral supernatant and 2.5 × 106
cells/well. For a 24-well plate, we use 0.5 ml virus and 0.5 × 106
cells/well.
104 Amie Simmons and José Alberola-Ila
4 Notes
capacity, and that the bigger the final construct is, the lower the
titer of the virus we will be able to produce [25]. Retroviruses
bigger than 8 kb, or lentiviruses bigger than 10 kb, are very hard
to work with. Keep this in mind when thinking about SIN len-
tiviruses with a tissue-specific promoter, your gene of interest,
and IRES-GFP to follow infection. Also, systems that work well
with one transgene may not work with other transgenes.
3. Mice are injected I.P. with 250 μg/g body weight of
5-fluorouracil dissolved in PBS (10 mg/ml), and the bone
marrow cells are harvested 4–5 days later.
4. We use the BD Biosciences Mouse Hematopoietic Progenitor
(Stem) Cell Enrichment Set and magnetic depletion, but there
are other equally good commercial alternatives.
5. 293T cells are grown in a 37 °C degree incubator containing
5 % CO2. The cells should not be allowed to become over-
confluent, and should not be split more than 1:5. 293T cells
easily detach from the tissue culture dishes after approximately
30 s of treatment with trypsin at room temperature (0.05 %
trypsin/0.53 mM EDTA). It is important to freeze multiple
(50–100) vials of the 293T cells after first receiving and
expanding them. This will ensure a ready supply of backup vials
to allow for uniform virus production over several years. Go
back to an early freeze and expand again when running low on
backup vials.
6. This is the most finicky reagent in the process. It normally
takes two or three batches of 2× HBS before finding one that
works well for transfections. The ability of the 2× HBS solu-
tion to produce working CaPO4 precipitates deteriorates after
6 months to 1 year, even when the 2× HBS solution is stored
at −20 °C. Prepare and test a fresh batch before the old one is
finished or too old! Despite this, in our experience no com-
mercial reagent works as well as a good homemade batch.
7. It is recommended to grow the viral backbone in RecA-
negative strains, and at 30 °C, to minimize recombination.
8. Other cytokines may also be added to this cocktail such as Flt3
(50 ng/ml) and TPO (25 ng/ml).
9. Matrix proteins such as fibronectin mediate colocalization of
target cells and vector [26]. The highest gene transfer is
obtained with the recombinant protein CH-296 (RetroNectin).
10. This is important. Retronectin binds much better to non-tissue
culture-treated plates [27].
11. Be careful with your 293T cells. Do not use cells that have
been kept in culture for a long time, or your transfection
efficiency will be reduced. It is extremely important that the
cells are not overly clumped and are at the correct density.
It is essential that the cells are extremely healthy prior to
106 Amie Simmons and José Alberola-Ila
References
1. Hawley RG, Fong AZ, Burns BF et al (1992) 11. Kowolik CM, Hu J, Yee JK (2001) Locus
Transplantable myeloproliferative disease control region of the human CD2 gene in a
induced in mice by an interleukin 6 retrovirus. lentivirus vector confers position-independent
J Exp Med 176:1149–1163 transgene expression. J Virol 75:4641–4648
2. Swift S, Lorens J, Achacoso P et al (2001) 12. Marodon G, Mouly E, Blair EJ et al (2003)
Rapid production of retroviruses for efficient Specific transgene expression in human and
gene delivery to mammalian cells using 293T mouse CD4+ cells using lentiviral vectors with
cell-based systems. Curr Protoc Immunol regulatory sequences from the CD4 gene.
Chapter 10:Unit 10.17C Blood 101:3416–3423
3. Burns JC, Friedmann T, Driever W et al (1993) 13. Lois C, Hong EJ, Pease S et al (2002) Germline
Vesicular stomatitis virus G glycoprotein pseu- transmission and tissue-specific expression of
dotyped retroviral vectors: concentration to transgenes delivered by lentiviral vectors.
very high titer and efficient gene transfer into Science 295:868–872
mammalian and nonmammalian cells. Proc Natl 14. Romero Z, Torres S, Cobo M et al (2011)
Acad Sci U S A 90:8033–8037 A tissue-specific, activation-inducible, lenti-
4. Mostoslavsky G, Kotton DN, Fabian AJ et al viral vector regulated by human CD40L
(2005) Efficiency of transduction of highly proximal promoter sequences. Gene Ther
purified murine hematopoietic stem cells by 18:364–371
lentiviral and oncoretroviral vectors under 15. Kraunus J, Schaumann DH, Meyer J et al
conditions of minimal in vitro manipulation. (2004) Self-inactivating retroviral vectors with
Mol Ther 11:932–940 improved RNA processing. Gene Ther 11:
5. Schambach A, Bohne J, Chandra S et al (2006) 1568–1578
Equal potency of gammaretroviral and lentivi- 16. Schambach A, Mueller D, Galla M et al (2006)
ral SIN vectors for expression of Overcoming promoter competition in pack-
O6-methylguanine-DNA methyltransferase in aging cells improves production of self-
hematopoietic cells. Mol Ther 13:391–400 inactivating retroviral vectors. Gene Ther 13:
6. Barrette S, Douglas JL, Seidel NE et al (2000) 1524–1533
Lentivirus-based vectors transduce mouse 17. Schmitt TM, Zuniga-Pflucker JC (2002)
hematopoietic stem cells with similar efficiency Induction of T cell development from hemato-
to moloney murine leukemia virus-based poietic progenitor cells by delta-like-1 in vitro.
vectors. Blood 96:3385–3391 Immunity 17:749–756
7. Laurenti E, Barde I, Verp S et al (2010) 18. Lerner C, Harrison DE (1990) 5-Fluorouracil
Inducible gene and shRNA expression in resi- spares hemopoietic stem cells responsible for
dent hematopoietic stem cells in vivo. Stem long-term repopulation. Exp Hematol
Cells 28:1390–1398 18:114–118
8. Tiscornia G, Tergaonkar V, Galimi F et al 19. Harrison DE, Lerner CP (1991) Most primi-
(2004) CRE recombinase-inducible RNA tive hematopoietic stem cells are stimulated to
interference mediated by lentiviral vectors. cycle rapidly after treatment with 5-fluorouracil.
Proc Natl Acad Sci U S A 101:7347–7351 Blood 78:1237–1240
9. Heinonen JE, Mohamed AJ, Nore BF et al 20. Naviaux RK, Costanzi E, Haas M et al (1996)
(2005) Inducible H1 promoter-driven lentiviral The pCL vector system: rapid production of
siRNA expression by Stuffer reporter deletion. helper-free, high-titer, recombinant retrovi-
Oligonucleotides 15:139–144 ruses. J Virol 70:5701–5705
10. Koo BK, Stange DE, Sato T et al (2012) 21. Pear WS, Miller JP, Xu L et al (1998) Efficient
Controlled gene expression in primary Lgr5 and rapid induction of a chronic myelogenous
organoid cultures. Nat Methods 9:81–83 leukemia-like myeloproliferative disease in
108 Amie Simmons and José Alberola-Ila
mice receiving P210 bcr/abl-transduced bone 26. Moritz T, Patel VP, Williams DA (1994) Bone
marrow. Blood 92:3780–3792 marrow extracellular matrix molecules improve
22. Lee NS, Dohjima T, Bauer G et al (2002) gene transfer into human hematopoietic cells
Expression of small interfering RNAs targeted via retroviral vectors. J Clin Invest 93:
against HIV-1 rev transcripts in human cells. 1451–1457
Nat Biotechnol 20:500–555 27. Tonks A, Tonks AJ, Pearn L et al (2005)
23. Hernandez-Hoyos G, Alberola-Ila J (2005) Optimized retroviral transduction protocol
Analysis of T-cell development by using which preserves the primitive subpopulation of
short interfering RNA to knock down pro- human hematopoietic cells. Biotechnol Prog
tein expression. Methods Enzymol 392: 21:953–958
199–217 28. Bain G, Quong MW, Soloff RS et al (1999)
24. Paithankar KR, Prasad KS (1991) Precipitation Thymocyte maturation is regulated by the
of DNA by polyethylene glycol and ethanol. activity of the helix-loop-helix protein, E47.
Nucleic Acids Res 19:1346 J Exp Med 190:1605–1616
25. Kumar M, Keller B, Makalou N et al (2001) 29. Gimferrer I, Hu T, Simmons A et al (2011)
Systematic determination of the packaging Regulation of GATA-3 expression during CD4
limit of lentiviral vectors. Hum Gene Ther lineage differentiation. J Immunol 186:
12:1893–1905 3892–3898
Chapter 9
Abstract
Radiation chimeras are prepared by subjecting recipient mice to sublethal or lethal dose of irradiation and
injecting them with hematopoietic stem cells (HSC) from untreated donor mice. HSC can be obtained
from bone marrow or fetal liver. This technique is a powerful tool when coupled with gene targeting
strategies to investigate function of HSCs, thymocyte development, and T cell function. This protocol
describes how to produce bone marrow or fetal liver chimeras.
Key words Bone marrow, Transplantation, Hematopoietic stem cells, Immune reconstitution, T cell
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_9, © Springer Science+Business Media New York 2016
109
110 Francis A. Flomerfelt and Ronald E. Gress
2 Materials
2.1 Host Mice (See 1. C57BL/6 CD45.2 congenic mice 6–12 weeks old, weight at
Note 1) least 20 g (see Notes 2 and 3).
2. Drinking water acidified with hydrochloric acid to pH 2.5–3.0.
3. Antibiotics: Amoxicillin or other.
2.2 Bone Marrow 1. C57BL/6 CD45.1 congenic mice 6–12 weeks old.
Preparation 2. 10 ml syringes tipped with 25 g needles.
3. Flushing media: RPMI 1640, 2 % fetal calf serum, 1 % penicil-
lin/streptomycin, 1 % HEPES, and 1 % l-glutamine.
4. ACK lysing buffer.
3 Methods
3.1 Prepare 1. One week before irradiation, the recipient mice are given acidi-
Host Mice fied drinking water (to prevent growth of Pseudomonas spe-
cies) supplemented with antibiotics such as amoxicillin
(0.5 mg/ml).
3.3 Optional T Cell T cell depletion of marrow eliminates resident T cells in the graft
Depletion of Marrow which allows analysis of T cell function of cells derived from
engrafted HSC and prevents graft-versus-host disease (GVHD) if
MHC mismatches are present (see Note 2).
1. Place counted bone marrow cells in 50 ml tube and fill with wash-
ing media, spin down cells, and remove media (see Note 8).
2. Prepare bone marrow cell suspension at 2.0 × 107 cells/ml in
RPMI/PSG without FCS.
3. Add antibodies at predetermined concentrations and mix gently
(see Note 4).
4. Place on ice for 15 min.
5. Wash bone marrow cells with RPMI/PSG without FCS.
Do not resuspend pellet.
6. Add diluted guinea pig complement to bring cell number to
2.0 × 107/ml (see Note 9). Add DNase to 40 μg/ml of cell
suspension. Mix well.
7. Place in 37 °C water bath × 40 min.
8. Wash with RPMI/PSG and resuspend cells in 50 % volume
used in step 1. Count cells and resuspend at 5.0 × 107/ml in
sterile PBS (see Note 10).
3.4 Prepare Liver 1. Harvest E14–E15 embryos and place in flushing media in a
Cell Suspension 15 cm tissue culture dish on ice (see Note 11).
2. Once cooled, rinse embryo in flushing media, place on a piece
of gauze, and euthanize the following institutional guidelines.
Remove a piece of tissue (tail, foot) and freeze for genotyping
if needed (see Note 12).
3. Dissect liver and place in sterile Eppendorf tube with 1 ml
flushing media. Be sure that liver is numbered the same as
genotyping biopsy.
4. Prepare cell suspension by mashing and pipetting the liver up
and down first with a P1000 tip and then with a P200 tip.
5. Allow cells to settle for 5 min on ice. Transfer cell suspension
to a new sterile Eppendorf tube taking care to avoid debris that
has settled to the bottom of the tube (see Note 13).
6. Strain cells with a 40 μM filter. Count viable cells and adjust to
0.25–1 × 107 cells/ml in sterile PBS for injection.
3.5 Inject HSC 1. At least 4–6 h before injection, irradiate recipient mice using
into Mice 9.5 Gy (see Note 14). It is advisable to wait ~2 h after the last
irradiation dose before injecting cells.
2. Inject 1.0 × 107 depleted BM cells in 200 μl sterile PBS in the
tail vein of the irradiated recipient mouse. Alternatively inject
0.5–2 × 106 fetal liver cells in 200 μl sterile PBS (see Note 15).
Bone Marrow and Fetal Liver Radiation Chimeras 113
4 Notes
Acknowledgement
References
1. de la Morena MT, Gatti RA (2011) A history of in two strains of genetically engineered mice.
bone marrow transplantation. Hematol Oncol Lab Anim (NY) 38:305–310.
Clin North Am 25:1–15 4. Gudmundsson KO, Stull SW, Keller JR (2012)
2. Hakim F, Fowler DH, Shearer GM, Gress RE Transplantation of mouse fetal liver cells for
(2001) Animal models of acute and chronic analyzing the function of hematopoietic stem
graft-versus-host disease. Curr Protoc and progenitor cells. Methods Mol Biol 879:
Immunol Chapter 4:Unit 4 3 123–133
3. Mader SL, Libal NL, Pritchett-Corning K, Yang R,
Murphy SJ (2009) Refining timed pregnancies
Chapter 10
Abstract
In vitro culture is an important complement, or substitute, to in vivo approaches in order to study T cell
effector differentiation. Here, we describe culture conditions that generate specific effector cell types by
exposing naïve T cells to appropriate cytokine signals.
Key words T cell differentiation, Tc, Th0, ThN, Th1, Th2, Th17, iTreg
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_10, © Springer Science+Business Media New York 2016
117
118 Elizabeth A. Wohlfert et al.
Table 1
Effector differentiation in vitro
Table 2
Effector differentiation in vivo
2 Materials
2.3 Beads 1. Mouse pan T (CD90.2) depletion magnetic beads (see Note 1).
3 Methods
3.1.1 Isolation of Splenic For naïve T cells conditioned in Th0 Th1, Th2, Th17, or Tc condi-
CD11c+ DC tions: use a ratio of 25–50 T cells for every DC. iTregs Conditioning
mix should include a 5–25 ratio of T cells for every DC cultured
( see Note 12). The following procedure is for one spleen and
volumes can be scaled proportionally as needed. All work should be
performed under sterile conditions in a tissue culture cabinet.
1. Place 5 ml of Digestion medium into a 60 mm petri dish.
Surgically remove the spleen from a euthanized mouse and
place directly into Digestion medium.
2. While holding the spleen with forceps, inject spleen with the
Digestion medium using a tuberculin syringe. Then, using
scissors, cut spleen into five to ten smaller pieces.
3. Place in 37 °C incubator with 5 % CO2 for 20 min.
4. Add a final concentration of 5 mM EDTA to the dish and incu-
bate five additional minutes to stop the enzymic activity of the
Digestion medium.
5. Transfer spleen pieces and medium to a 50 ml conical tube and
pass through a 70 μm cell strainer. Use cell scraper to remove
any adherent cells and rinse the petri dish thoroughly with
5 ml Staining medium.
In Vitro Analyses of T Cell Effector Differentiation 123
2x Conditioning mix
APCs in
100 µL 100 µL
200 µL
96 well plate
Fig. 1 Schematic of procedure
6. Lyse with 1 ml Red cell lysis buffer for 2 min on ice, wash by
filling tube to 10 ml with Staining medium.
7. Resuspend in 5 ml Staining medium and count cells.
8. Refer to Chapter 7 on “Isolating T cell subsets” for staining
cells with fluorescently labeled antibodies and sorting
CD11c+MHC II+.
9. Prepare DC suspensions in Culture medium at a concentration
of 1–2 × 103 cells/100 μl/well for Th0, Th1, Th2, Th17, or Tc
cultures, or of 2–10 × 103 cells/100 μl/well for iTreg cultures
(see introduction to this section and Fig. 1 for more informa-
tion). Refrain from aliquoting cells into wells at this point as
the Conditioning mix will be added to this master mix. Keep
DCs on ice until ready to prepare Conditioning mixes.
10. See Subheading 3.3 for preparing Conditioning mixes.
3.1.2 Irradiated T Carry out all procedures at 4 °C unless otherwise specified and
Cell-Depleted Splenocytes under sterile conditions in a tissue culture cabinet. The ideal ratio
for this type of APC is 5 for every one T cell.
1. See Chapter 7 on “Isolating T cell subsets” for procedure on
isolating a single-cell suspension from a mouse spleen.
2. Immunomagnetic cell separation for depletion of pan T cells.
The following protocol is for a 1 × 108 total splenocyte suspen-
sion, and can be scaled up or down as necessary. While this
protocol utilizes immunomagnetic beads from Dynabead,
users may employ immunomagnetic beads of their choice with
the appropriate protocol.
3. Preparation of Dynabeads: Resuspend beads fully (either vortex
whole vial or place on a tilt rotator for at least 5 min).
124 Elizabeth A. Wohlfert et al.
3.3 Conditioning To the APCs prepared in Subheading 3.1, add the following reagents
Mixes to prepare the corresponding 2× Conditioning mixes. These should
be prepared immediately before setting up cultures and kept on ice
until ready to aliquot for culture. Note that all concentrations
given below refer to the concentration in the conditioning mix,
therefore twice as high as in the final culture (see Table 1).
In Vitro Analyses of T Cell Effector Differentiation 125
3.3.1 ThN or CD8+ Tc 1. Add: 2 μg/ml anti-CD3, 6 μg/ml anti-CD28, 20 ng/ml IL-2.
Conditioning Mix
3.3.2 Th17 Substituting IMDM for RPMI increases the frequency of IL-17
Conditioning Mix producing cells (see Note 7). The Th17 Conditioning mix does
not contain IL-2.
1. Add: 2 μg/ml anti-CD3, 6 μg/ml anti-CD28, 20 ng/ml IL-6,
5.0 ng/ml TGF-β, 20 μg/ml anti-IL-4, 20 μg/ml anti-IFN-γ,
20 μg/ml anti-IL-12.
3.3.6 iTreg 1. Add: 2 μg/ml anti-CD3, 5.0 ng/ml TGF-β, 20 μg/ml anti-
Conditioning Mix IL-4, 20 μg/ml anti-IFNγ (see Note 16). (Retinoic acid can
be added to the culture to enhance the induction of Foxp3,
see Note 17.)
2. At 48 h, add 10 ng/ml IL-2 (final concentration) and incubate
for an additional 24 h. This is earlier than the day 3 culture split
recommended for non-iTreg cultures (see Subheading 3.5).
3.3.7 Culture Set Up 1. Add 100 μl/well of each APC-containing conditioning mix to
wells in a 96 well round bottom tissue culture plate.
2. Add 100 μl T cell suspension (5 × 104 cells) to each APC-
containing well. The final volume should now be 200 μl
per well.
3. Place in a 37 °C incubator with 5 % CO2 for 3 days
(see Subheading 3.5) (see Note 18).
3.4.2 Preparation Prepare 2× cell-free conditioning mixes for each stimulation condi-
of APC-Free Conditioning tion by supplementing APC-free Culture medium with cytokines
Mixes and anti-cytokine antibodies (but not soluble anti-CD3 and soluble
anti-CD28) using concentrations indicated in Subheading 3.3 above
(see also Table 1).
3.4.3 Culture Set Up 1. Per well in a 96 well flat bottom tissue culture plate, add
100 μl/well of APC-free conditioning mix and 100 μl T cell
suspension (5 × 104 cells, prepared as in Subheading 3.2), for a
final volume of 200 μl per well.
2. Place in a 37 °C incubator with 5 % CO2 for 72 h (see
Subheading 3.5).
3.5 Split the Culture 1. Incubate for 72 h and then split each 200 μl well into two
at Day 3 100 μl wells and add 100 μl of Culture medium to each
well.
2. Incubate an additional 24 h.
3. Harvest cells for downstream applications.
4 Notes
References
1. Seder RA, Paul WE (1994) Acquisition of pathways of T cell activation. J Exp Med 159:
lymphokine-producing phenotype by CD4+ 881–905
T cells. Annu Rev Immunol 12:635–673 7. Veldhoen M, Hirota K, Christensen J, O’Garra
2. Kupper T, Horowitz M, Lee F, Robb R, Flood A, Stockinger B (2009) Natural agonists for
PM (1987) Autocrine growth of T cells inde- aryl hydrocarbon receptor in culture medium
pendent of interleukin 2: identification of are essential for optimal differentiation of Th17
interleukin 4 (IL 4, BSF-1) as an autocrine T cells. J Exp Med 206:43–49
growth factor for a cloned antigen-specific 8. Mucida D, Park Y, Kim G, Turovskaya O, Scott
helper T cell. J Immunol 138:4280–4287 I, Kronenberg M, Cheroutre H (2007)
3. Swain SL, Weinberg AD, English M, Huston G Reciprocal TH17 and regulatory T cell differ-
(1990) IL-4 directs the development of entiation mediated by retinoic acid. Science
Th2-like helper effectors. J Immunol 145: 317:256–260
3796–3806 9. Coombes JL, Siddiqui KR, Arancibia-Cárcamo
4. Andrus L, Granelli Piperno A, Reich E (1984) CV, Hall J, Sun CM, Belkaid Y, Powrie F (2007)
Cytotoxic T cells both produce and respond to A functionally specialized population of mucosal
interleukin 2. J Exp Med 159:647–652 CD103+ DCs induces Foxp3+ regulatory T cells
5. Zhu J, Yamane H, Paul WE (2010) via a TGF-beta and retinoic acid-dependent
Differentiation of effector CD4 T cell popula- mechanism. J Exp Med 204:1757–1764
tions. Annu Rev Immunol 28:445–489 10. Sun CM, Hall JA, Blank RB, Bouladoux N,
6. Ashwell JD, Defranco AL, Paul WE, Schwartz Oukka M, Mora JR, Belkaid Y (2007) Small
RE et al (1984) Antigen presentation by rest- intestine lamina propria dendritic cells promote
ing B cells. Radiosensitivity of the antigen- de novo generation of Foxp3 T reg cells via
presentation function and two distinct retinoic acid. J Exp Med 204:1775–1785
Part III
Specific Techniques
Chapter 11
Abstract
Recently, tissue slices have been adapted to study both mouse and human T cell development. Thymic slices
combine and complement the strengths of existing organotypic culture systems to study thymocyte differ-
entiation. Specifically, the thymic slice system allows for high throughput experiments and the ability to
introduce homogenous developmental intermediate populations into an environment with a well-established
cortex and medulla. These qualities make thymic slices a highly versatile and technically accessible model
to study thymocyte development. Here we describe methods to prepare, embed, and slice thymic lobes to
study T cell development in situ.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_11, © Springer Science+Business Media New York 2016
131
132 Jenny O. Ross et al.
2 Materials
3 Methods
Fig. 1 Embedding and preparing thymic slices. (a) Using blunt forceps, grasp connective tissue and transfer
individual thymic lobes into a tissue mold filled half way with 4 % agarose solution. (b) Gently push the lobe
into position within the agarose before the agarose solution sets. (c) Once the lobe is positioned, place the
mold into an ice water bath to set. (d) Example of three lobes embedded in one tissue mold. (e) Trim the excess
agarose around the embedded lobes. Left, thymic lobe embedded horizontally. Right, example of a thymic lobe
embedded vertically. (f) Up to three blocks of embedded tissue can be glued to the vibratome stage and sliced
simultaneously. Use a bent spatula to transfer the thymic slices after each cut as they float free of the vibra-
tome blade. (g) Use a pipet tip to gently push the thymic slice onto a cell culture insert within a 6-well plate.
(h) Use a pipet to remove any liquid in the transwell and around the edges of the thymic slice
3.3 Overlay 1. Remove all excess liquid from the insert (any accumulated PBS
of Thymocytes onto and/or medium) surrounding the thymic slices by pipetting
Thymic Slices carefully to prevent tearing or damaging the agarose surrounding
the tissue (see Note 16) (Fig. 1h).
2. Pipet 10 μL of labeled (either congenic or dye, etc.) thymo-
cytes of interest on top of each slice (see Note 17) carefully
without touching the tissue or the surrounding agarose
(Fig. 2d–f).
3. Incubate slices at 37 °C for 2 h to allow overlaid cells to migrate
into the thymic slices.
4. Rinse the slices with 1 mL/well PBS to wash off any cells that
have not yet migrated into the slice.
5. Continue to incubate slices at 37 °C for the desired length of
time (see Notes 18 and 19).
T Cell Development in Thymic Slices 137
Fig. 2 Quality control and overlay of thymic slices. (a) Example slices from a vertically (top) versus horizontally
(bottom) embedded thymic lobe. (b) Examples of poor quality slices that will cause overlaid cells to run off. The
top slice has a tear in the agarose and the bottom slice has a chunk of agarose torn free. (c) Example of a tear
in the thymic tissue (top slice) versus a good, intact slice (bottom). After pipetting off excess liquid, inspection
of the thymic tissue itself, as well as the borders of the agarose surrounding the tissue will reveal any slices
with defects. These should be discarded and replaced with good quality slices with clean, clear edges of aga-
rose for optimal overlay. (d) Add 10–20 μL of cell suspension per slice by pipetting the volume so that it forms
a suspended drop. (e) Gently touch the drop of resuspended cells to the center of the thymic slice, without
touching the pipet tip itself to the slice. (f) The overlaid cells will remain on top of the slice
4 Notes
10. Ensure that the thymic lobe touches the bottom of the mold
while it is being embedded in agarose to provide a 0.5 cm buf-
fer of agarose below the tissue. This will allow for the entire
tissue to be vibratome-sliced.
11. There is approximately a 30 s window after the molds are
placed into the ice water bath during which the placement of
the lobes can be adjusted within the agarose. Use the blunt
forceps to move the thymus into the position desired (Fig. 1b).
12. Vibratome blades can be reused for a couple of experiments.
Discard after they have been used for trimming the blocks.
13. Up to three tissue blocks aligned side-by-side parallel to the
vibratome blade can be sliced simultaneously. Ensure that they
are roughly equivalent in height for best results (Fig. 1f).
14. 400–500 μm thick slices work well for tissue culture and
500–1000 μm thick slices work well for imaging.
15. Place as many slices as will fit per insert without touching each
other or the insert wall. Ensure that there are no air bubbles
below the insert.
16. If the slices are not dried properly, the overlaid cells may spill off.
Any tears or holes in the agarose may also lead to loss of overlaid
cells/fluid volume (Fig. 2a–c). Finally, properly drying the slices
allows for visual inspection of the slice quality. It is a good idea
to cut extra slices and replace any as needed.
17. Overlaying 1 × 106 cells/slice for 2 h prior to washing typically
results in the cells of interest making up 0.5–2 % of the total
thymocytes within the slice. Longer incubations result in a
greater proportion of cells migrating into the tissue slice.
18. If a heterogeneous population of thymocytes is overlaid, har-
vesting an early time-point will establish a baseline of the cells
that migrated into the slices. The quality of the slices deterio-
rates significantly after just a day or two, but development
seems to proceed for several days.
19. Over time, thymocytes leak out of the slices due to the absence
of a capsule.
Acknowledgements
References
1. Markert ML, Watson TJ, Kaplan I et al (1997) thymocyte migration in situ. J Clin Invest
The human thymic microenvironment during 123:2131–2142
organ culture. Clin Immunol Immunopathol 6. Dzhagalov IL, Chen KG, Herzmark P et al
82:26–36 (2013) Elimination of self-reactive T cells in the
2. Bhakta NR, Oh DY, Lewis RS (2005) Calcium thymus: a timeline for negative selection. PLoS
oscillations regulate thymocyte motility during Biol. doi:10.1371/journal.pbio.1001566
positive selection in the three-dimensional thy- 7. Anderson G, Partington KM, Jenkinson EJ
mic environment. Nat Immunol 6:143–151 (1998) Differential effects of peptide diversity
3. Ehrlich LI, Oh DY, Weissman IL et al (2009) and stromal cell type in positive and negative
Differential contribution of chemotaxis and sub- selection in the thymus. J Immunol
strate restriction to segregation of immature and 161:6599–6603
mature thymocytes. Immunity 31:986–998 8. Jenkinson EJ, Anderson G, Owen JJ (1992)
4. Le Borgne M, Ladi E, Dzhagalov I et al (2009) Studies on T cell maturation on defined thymic
The impact of negative selection on thymocyte stromal cell populations in vitro. J Exp Med
migration in the medulla. Nat Immunol 176:845–853
10:823–830 9. Schaefer BC, Schaefer ML, Kappler JW et al
5. Halkias J, Melichar HJ, Taylor KT et al (2013) (2001) Observation of antigen-dependent
Opposing chemokine gradients control human CD8þ T-cell/dendritic cell interactions in vivo.
Cell Immunol 214:110–122
Chapter 12
Abstract
Potentially harmful T cell precursors are removed from the conventional T cell pool by negative selection.
This process can involve the induction of apoptosis, anergy, receptor editing or deviation into a regulatory
T cell lineage. As such this process is essential for the health of an organism through its contribution to
central and peripheral tolerance. While a great deal is known about the process, the precise mechanisms
that regulate negative selection are not clear. Furthermore, the signals that distinguish the different forms
of negative selection are not fully understood. Numerous models exist with the potential to address these
questions in vitro and in vivo. This chapter describes methods of fetal thymic organ culture designed to
analyze the signals that determine these unique cell fates.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_12, © Springer Science+Business Media New York 2016
141
142 Cody A. Cunningham et al.
2 Materials
3 Methods
3.1 Timed Matings The days of the week represent an example of the scheduling procedure
used in our lab.
1. Tuesday: Prepare time mating cages by placing a cage divider
with feeders and water for both sides in each cage.
2. Wednesday, morning: Place two to three females on one side
of the divider and one male on the opposite side of the divider
(see Note 7).
3. Friday, late afternoon/evening: Remove cage divider.
4. Saturday, morning (before 10 a.m.): Remove male.
OPTIONAL: check for and record mucous plugs (see Note 8).
5. Saturday (or the day of the plug) is considered day 0.5
(although technically it could be anywhere from 0 to 0.5 days)
(see Note 9).
6. Two weeks from the following Monday (Day 15.5), harvest
fetuses (see below).
3.2 Preparing 1. Boil cellulose filters three times in dH2O. The final time cover
the Culture Dishes with foil and, when cool, transfer into the hood. Place the fil-
ters in medium just before use.
2. Prepare medium.
3. Remove a strip of gel-foam and cut it into three squares (KEEP
STERILE), placing each square in one well. Using one straight
and one curved forceps soak the gel-foam and tease the air
bubbles out.
4. Place one filter (from step 1) on each gel-foam square.
5. Place the plate in a FTOC chamber (see Note 4). Pre-warm in
the incubator.
3.4 Feeding We use two methods of feeding. Both are described here.
1. Replace the media in the cultures every day.
(a) To do this, carefully suction off the media with a sterile
Pasteur pipette.
(b) Add 2 ml fresh warm media, carefully basting thymic lobes.
2. Replace the media in the cultures every 2–5 days.
(a) Carefully baste thymic lobes daily.
(b) To replace media, carefully suction off the media with a
sterile Pasteur pipette.
(c) Add 2 ml fresh warm media, carefully basting thymic lobes.
3. Seven days of culture is sufficient for good thymic develop-
ment from an E15.5 lobe. However, they remain viable up to
14 days.
In vitro Analysis of Thymocyte Signaling 147
4 Notes
1. This brand of filters float nicely in the media (shiny side up)
such that expensive (and sometimes difficult to obtain) surgi-
cal gel-foam is not needed.
2. We often test our FCS in FTOC in advance to minimize
serum-based artifacts of selection.
3. If you wish to add, antibodies, peptides, etc., add them to
media prior to plating out thymi.
4. It is critically important to avoid drying of the thymic lobe. To
do this we use a covered Pyrex baking dish (available from the
local grocery store). We have small holes drilled in the cover
(>5 mm). We place cover of a 6- or 12-well plate in the bot-
tom of the baking dish as a pedestal, and fill the bottom of the
dish with autoclaved dH2O. The culture dish containing the
FTOC are placed on top of the pedestal and placed in the
incubator. See also Jenkinson et al J. Vis. Exp. (2008) for an
alternative apparatus.
5. With practice day 15.5 embryos are easily removed without
the aid of a microscope.
6. It is critically important to have a selection outcome control
for each test condition. For CD8 T cells, we always save a lobe
or partial lobe (minimum of 1 × 104 cells) and stain for TCR,
CD4, CD8α, and CD8β. This is then analyzed by flow cytom-
etry for the following:
(a) The generation of CD8β SP thymocytes is the test of posi-
tive selection.
(b) The lack of the generation of CD8α and CD8β SP thymo-
cytes and the maintenance of a DP population indicates
no selection.
(c) The lack of the generation of CD8β SP thymocytes with
the concomitant loss of DP indicates negative selection.
148 Cody A. Cunningham et al.
Acknowledgements
The authors thank the members of the Daniels and Teixeiro labs
for assistance in compiling these protocols. Funding provided by
grants from the Missouri Mission Enhancement Fund and
University of Missouri Research Board is greatly acknowledged.
References
1. Palmer E (2003) Negative selection–clearing 8. Petrie HT, Zuniga-Pflucker JC (2007) Zoned
out the bad apples from the T-cell repertoire. out: functional mapping of stromal signaling
Nat Rev Immunol 3(5):383–391 microenvironments in the thymus. Annu Rev
2. Starr TK, Jameson SC, Hogquist KA (2003) Immunol 25:649–679
Positive and negative selection of T cells. Annu 9. Borgne ML, Ladi E, Dzhagalov I, Herzmark
Rev Immunol 21:139–176 P, Liao YF, Chakraborty AK, Robey EA (2009)
3. Stritesky GL, Jameson SC, Hogquist KA The impact of negative selection on thymocyte
(2012) Selection of self-reactive T cells in the migration in the medulla. Nat Immunol
thymus. Annu Rev Immunol 30:95–114 10(8):823–830
4. Baldwin TA, Hogquist KA, Jameson SC 10. Dzhagalov IL, Chen KG, Herzmark P, Robey
(2004) The fourth way? Harnessing aggressive EA (2013) Elimination of self-reactive T cells
tendencies in the thymus. J Immunol 173(11): in the thymus: a timeline for negative selec-
6515–6520 tion. PLoS Biol 11(5), e1001566
5. Daniels MA, Devine L, Miller JD, Moser JM, 11. Daniels MA, Teixeiro E, Gill J, Hausmann B,
Lukacher AE, Altman JD, Kavathas P, Roubaty D, Holmberg K, Werlen G, Holländer
Hogquist KA, Jameson SC (2001) CD8 bind- GA, Gascoigne NR, Palmer E (2006) Thymic
ing to MHC class I molecules is influenced by selection threshold defined by compartmental-
T cell maturation and glycosylation. Immunity ization of Ras/MAPK signalling. Nature
15(6):1051–1061 444(7120):724–729
6. Davey GM, Schober SL, Endrizzi BT, Dutcher 12. Daniels MA, Teixeiro E (2010) The persis-
AK, Jameson SC, Hogquist KA (1998) tence of T cell memory. Cell Mol Life Sci
Preselection thymocytes are more sensitive to 67(17):2863–2878
T cell receptor stimulation than mature T cells. 13. Cunningham CA, Knudson KM, Peng BJ,
J Exp Med 188(10):1867–1874 Teixeiro E, Daniels MA (2013) The POSH/
7. Lucas B, Stefanová I, Yasutomo K, Dautigny JIP-1 scaffold network regulates TCR-mediated
N, Germain RN (1999) Divergent changes in JNK1 signals and effector function in CD8 T
the sensitivity of maturing T cells to structurally cells. Eur J Immunol 43(12):3361–3371
related ligands underlies formation of a useful 14. Gil D, Schrum A, Alarcon B, Palmer E (2005)
T cell repertoire. Immunity 10(3):367–376 T cell receptor engagement by peptide – MHC
In vitro Analysis of Thymocyte Signaling 149
ligands induces a conformational change in the thymocyte dulling assay. Methods Mol Biol
CD3 complex of thymocytes. J Exp Med 156:219–232
201(4):517–522 18. Hogquist KA, Jameson SC, Bevan MJ (1994)
15. Schrum AG, Gil D, Dopfer EP, Wiest DL, The ligand for positive selection of T lympho-
Turka LA, Schamel WW, Palmer E (2007) cytes in the thymus. Curr Opin Immunol
High-sensitivity detection and quantitative 6(2):273–278
analysis of native protein-protein interactions 19. Travers H, Anderson G, Gentle D, Jenkinson
and multiprotein complexes by flow cytome- E, Girdlestone J (2001) Protocols for high
try. Sci STKE 2007(389):pl2 efficiency, stage-specific retroviral transduc-
16. Anderson G, Jenkinson EJ, Moore NC, Owen tion of murine fetal thymocytes and thymic
JJ (1993) MHC class II-positive epithelium epithelial cells. J Immunol Methods 253(1–2):
and mesenchyme cells are both required for 209–222
T-cell development in the thymus. Nature 20. Lambolez F, Kronenberg M, Cheroutre H
362(6415):70–73 (2007) Thymic differentiation of TCR alpha
17. Hogquist KA (2001) Assays of thymic selec- beta(+) CD8 alpha alpha(+) IELs. Immunol
tion. Fetal thymus organ culture and in vitro Rev 215:178–188
Chapter 13
Abstract
Reconstituted thymus organ culture is based on fetal thymus organ culture (FTOC). Purified thymocyte
populations, from genetically modified mice or even from other species, are cultured in vitro with thymic
lobes depleted of their endogenous thymocytes (by 2′-deoxyguanosine treatment) to form a new thymus.
This potent and timesaving method is distinct from FTOC, which assesses development of unmodified
thymic lobes, and reaggregate thymic organ culture, in which epithelial cells are separately purified before
being aggregated with thymocytes.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_13, © Springer Science+Business Media New York 2016
151
152 Zimu Deng et al.
2 Materials
2.1 Reagents All reagents must be purchased sterile, prepared with sterile
reagents or be sterile-filtered.
1. 1× PBS (Ca2+/Mg2+-free).
2. RPMI-1640 medium: supplemented with 2 mM L-Glutamine,
100 U/ml Penicillin, 0.1 mg/ml Streptomycin, 1× Non-
Essential Amino Acids and 10 mM Hepes-pH 7.4. Add
2-Mercaptoethanol in the medium to a concentration of
0.05 mM before use. Referred to as RPMI medium through-
out. The concentration of FBS varies from 10 to 20 % as indi-
cated in each step.
3. 2′-Deoxyguanosine (dGuo) (e.g. Sigma-Aldrich, D7145) 10×
stock solution: 13.5 mM in 1× PBS (dissolve at 37 °C). Keep
the stock solution at −20 °C.
4. IL7 (e.g. Peprotech, 96-217-17-2): 105 U/ml 1000× stock in
1× PBS, store at −20 °C.
Reconstituted Thymus Organ Culture 153
3 Methods
3.1 Preparation We usually put two female mice and one male in one cage at day
of Timed Pregnancies end (5:00 p.m.). Every female is examined for the presence of a
vaginal plug at 8 am every subsequent morning. The day a plug
detected is counted as Day 1 of gestation. The pregnant mice will
be kept in a new cage till use. See Notes 1–4.
Fig. 2 Fetal thymus lobes before and after treatment with 2′-deoxyguanosine. (a) Thymus lobes from E15.5
feta mice. (b) An isolated thymus lobe. (c) A lobe cultured with dGuo for 6 days
3. Cut open the uterus and collect the embryos. Swirl the Petri
dish to wash off the blood from the embryos and transfer
them into a 60-mm Petri dish containing 4 ml PBS under the
stereo-dissecting microscope.
4. Hold one embryo in a face-up position with thin forceps on
the head. Using the other thin forceps, pull each foreleg in a
head-to-toe direction, until the chest skin comes off. Two thy-
mus lobes (below the first pair of ribs and above the heart)
should be seen. If not, use thin forceps to snip off the first pair
of ribs under the armpit and open the chest to reveal the lobes
completely.
5. Carefully remove the lobes together using thin forceps and
transfer to a new 60-mm Petri dish containing 4 ml RPMI-
1640 medium with 10 % FBS. See Note 6.
6. Gently swirl the dish to wash off any blood. Use thin forceps
to tear off any connective tissue between the two lobes and
separate them (Fig. 2).
5. Keep the plate on ice and carefully detach the lobes from filters
under a dissecting microscope (Fig. 2). Scoop up the lobes and
transfer them into a new 10-cm dish containing 10 ml fresh
10 % FBS-supplemented RPMI-1640. Swirl the plate and put it
in the incubator for 20 min to diffuse away dGuo. Change the
medium every 20 min, three times. The lobes are ready for the
next step Hanging-Drop reconstitution with T cell precursors.
7. Transfer the lobes onto the filters of the culture well set up in
step 5 above.
8. Incubate the plate in the humidified box in tissue culture incu-
bator for 7–10 days. The introduced thymocytes then can be
assessed by FACS analysis (Fig. 3).
3.6 Optional Step: 1. Seed 1 × 106 Plat E cells [12] in a 60-mm Petri dish containing
Retroviral Transfection 3.5 ml DMEM supplemented with 10 % FBS. Place for
of Thymocytes 18–24 h in tissue culture incubator. See Notes 10 and 11.
2. Transfect 20 μg of retroviral plasmid DNA (e.g. pMCS-IRES-
GFP) using adhesion-assisted lipofection or calcium phos-
phate precipitation method.
3. Seven hours after transfection, remove the supernatants and
add 2 ml fresh DMEM supplemented with 10 % FBS. Collect
culture (retroviral) supernatants 48–72 h after the transfection
and use for infection [13]. Replace with 2 ml fresh DMEM
supplemented with 10 % FBS. See Note 12.
4. Pre-warm up the centrifuge to 32 °C.
5. Filter 500 μl retroviral supernatants with a 0.2-μm filter and
add it in a well of a 24-well plate. Dilute polybrene stock 1:500
into the retroviral supernatants to a final concentration of
20 μg/ml. Add 100–10,000 thymocytes (prepared in
Subheading 3.4) into each well and seal the plate with parafilm.
Spin the plate at 1000 × g in pre-warmed centrifuge for 90 min.
6. After transduction, take an aliquot of thymocytes, culture
overnight and assess cell viability and transduction efficiency
by flow cytometry (Fig. 4).
7. Use the rest of the cells in hanging-drop culture as described
in Subheading 3.5.
Reconstituted Thymus Organ Culture 157
Fig. 4 A typical result of RTOC. The pMCS-IRES-GFP retrovirus-infected thymocytes were reconstituted with
dGuo-treated lobes and cultured for 8 days. Cells were isolated and then stained with anti-CD4 and anti-CD8
antibodies
4 Notes
Acknowledgements
References
1. Owen JJ, Ritter MA (1969) Tissue interaction fetal liver progenitor cells. J Immunol 156:
in the development of thymus lymphocytes. 3233–3242
J Exp Med 129:431–442 8. Rui J, Liu H, Zhu X, Cui Y, Liu X (2012)
2. Mandel T, Russell PJ (1971) Differentation of Epigenetic silencing of CD8 genes by ThPOK-
foetal mouse thymus. Ultrastructure of organ mediated deacetylation during CD4 T cell dif-
cultures and of subcapsular grafts. Immunology ferentiation. J Immunol 189:1380–1390
21:659–674 9. Anderson G, Jenkinson EJ (2007) Fetal thy-
3. Jenkinson EJ, Van Ewijk W, Owen JJ (1981) mus organ culture. CSH Protoc 2007:pdb
Major histocompatibility complex antigen prot4808
expression on the epithelium of the developing 10. Jenkinson EJ, Franchi LL, Kingston R, Owen
thymus in normal and nude mice. J Exp Med JJ (1982) Effect of deoxyguanosine on lym-
153:280–292 phopoiesis in the developing thymus rudiment
4. Takahama Y (2000) Differentiation of mouse in vitro: application in the production of chi-
thymocytes in fetal thymus organ culture. meric thymus rudiments. Eur J Immunol 12:
Methods Mol Biol 134:37–46 583–587
5. Jenkinson EJ, Anderson G, Owen JJ (1992) 11. Ueno T, Liu C, Nitta T, Takahama Y (2005)
Studies on T cell maturation on defined thy- Development of T-lymphocytes in mouse fetal
mic stromal cell populations in vitro. J Exp thymus organ culture. Methods Mol Biol
Med 176:845–853 290:117–133
6. Watanabe Y, Katsura Y (1993) Development 12. Morita S, Kojima T, Kitamura T (2000) Plat-E:
of T cell receptor alpha beta-bearing T cells in an efficient and stable system for transient pack-
the submersion organ culture of murine fetal aging of retroviruses. Gene Ther 7:1063–1066
thymus at high oxygen concentration. Eur J 13. Travers H, Anderson G, Gentle D, Jenkinson
Immunol 23:200–205 E, Girdlestone J (2001) Protocols for high
7. Tsuda S, Rieke S, Hashimoto Y, Nakauchi H, efficiency, stage-specific retroviral transduction
Takahama Y (1996) Il-7 supports D-J but of murine fetal thymocytes and thymic epithe-
not V-DJ rearrangement of TCR-beta gene in lial cells. J Immunol Methods 253:209–222
Chapter 14
Abstract
Recreating the thymic microenvironment in vitro poses a great challenge to immunologists. Until recently,
the only approach was to utilize the thymic tissue in its three-dimensional form and to transfer the hema-
topoietic progenitors into this tissue to generate de novo T cells. With the advent of OP9-DL cells (bone
marrow-derived cells that are transduced to express Notch ligand, Delta-like), hematopoietic stem cells
(HSC) could be induced to differentiate into T cells in culture for the first time outside of the thymic tissue
on a monolayer. We, as well as others, asked whether the ability to support T cell development in vitro in
a monolayer is unique to BM-derived OP9 cells, and showed that provision of Delta-like expression to
thymic epithelial cells and fibroblasts also allowed for T cell development. This provides the opportunity
to design an autologous coculture system where the supportive stromal and the hematopoietic compo-
nents are both derived from the same individual, which has obvious clinical implications. In this chapter,
we describe methods for establishing a primary murine dermal fibroblast cell population that is transduced
to express Delta-like 4, and describe the conditions for its coculture with HSCs to support T cell lineage
initiation and expansion, while comparing it to the now classic OP9-DL coculture.
Key words T cell development, Lymphopoiesis, Notch, Delta-like, Fibroblasts, Hematopoietic stem
cells, Stromal cells, IL-7, Stem cell factor
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_14, © Springer Science+Business Media New York 2016
159
160 M. Mohtashami et al.
from HSCs [4]. The OP9-DL1 cells could mimic the thymus in its
ability to support early T cell development as well as the generation
of mature CD8 single positive (SP) cells [5, 6]. The detailed meth-
ods for generating T cells from mouse and human HSCs using
OP9-DL cells have been published earlier [7]. We also demon-
strated that rather than Dll1, Dll4, which is the Notch ligand phys-
iologically expressed in the thymus, is more efficient at steering
HSCs toward T cell lineage when ectopically expressed at physio-
logical levels on OP9 cells (OP9-DL4) [8].
There are several advantages to working with OP9-DL (OP9
cells expressing Dll1 or Dll4) cells, namely the freeing of the reli-
ance on availability of thymic tissue, the ability to perform clonal
assays and the ease of expansion of T lineage cell populations [9].
However, one limitation is the ability to test genetic differences
within stromal cells supporting T cell development, or to match
the genetic background of stem/progenitor cells to that of the
supporting stromal cells. To circumvent these issues, we and others
recently showed that T cell development can also be supported by
thymic epithelial cells [10, 11] and primary murine dermal fibro-
blasts expressing Dll4 (mFibro-DL4) [12, 13]. Dermal fibroblasts
are easily accessible, and though not as efficient as OP9 cells, these
cells are nevertheless effective in supporting T cell development,
and importantly can be used to establish an autologous T cell
coculture system where both the supporting stromal and the T
cells are derived from the same individual or genetic background.
Here, we present the protocol for (a) establishing a mouse pri-
mary fibroblast, (b) its transduction to express Dll1 or Dll4, (c)
purification of mouse HSCs, and (d) maintenance/expansion of
cocultures and comparison to OP9-DL coculture, also demonstrat-
ing that mFibro-DL1 can also support T cell development (Fig. 1).
2 Materials
2.1 Cell Lines 1. Primary dermal fibroblasts derived from mouse ear biopsies.
and Media 2. Fetal bovine serum (FBS) (see Note 1). Heat-inactivate at
56 °C for 30 min. Store at 4 °C.
3. Phosphate-buffered saline (PBS) without Ca2+/Mg2+.
4. Trypsin 2.5 % (Life Technologies). Dilute with PBS to 0.25 %
solution.
5. Fibroblast media: DMEM supplemented with 10 % (v/v) FBS,
2 mM GlutaMAX, 2 mM 2-mercaptoethanol, Penicillin
100 U/mL–Streptomycin 100 μg/mL.
6. Sterilized 22 mm × 22 mm glass coverslips.
7. Sterilized scissors and forceps.
8. Tissue culture ware (10-cm dishes, 6-well plates), tissue
culture-treated, serological pipettes.
Induction of T Cell Development In Vitro by Delta-Like (Dll)-Expressing Stromal Cells 161
Fig. 1 Cocultures of OP9-DL1, mFibro-GFP, and mFibro-DL1 cell with hematopoietic stem cells. Different
hematopoietic cell lineage differentiation outcomes are obtained from E15 fetal liver-derived HSCs cultured
with OP9-DL1, mFibro-GFP, and mFibro-DL1 cell lines. Cocultures were set up with 103 mouse fetal liver-
derived Lineage− CD117+ Sca1+ HSCs were cultured with the different stromal cell lines as indicated. (a) At
day 6 of coculture, flow cytometric analysis was performed for the expression of CD25, CD44 for T lineage-
specified cells (upper panel) and CD19, CD11b for B and myeloid cells, respectively (lower panels). (b) A frac-
tion of the cocultures were transferred to fresh plates with stromal cells, and at day 12 of coculture, in addition
to CD25 and CD44 (upper panel), the expression of CD4 and CD8 (lower panel) was also examined. Cells were
electronically gated on live (DAPI−) and CD45+ hematopoietic cells. Data are representative of at least three
independent experiments
2.3 HSC Isolation 15. OP9-DL cells: OP9 cells (Riken repository, Tsukuba, Japan,
and Coculture http://www.rtc.riken.go.jp) retrovirally transduced to express
the coding region for Dll1 or Dll4 cDNA, as previously
reported [8, 14].
16. 40 μm cell strainers.
17. Anti-CD24 mAb: supernatant from J11D.2 hybridoma clone
or purified anti-CD24 mAb.
18. Rabbit complement.
19. Lympholyte®-M Cell Separation Media.
162 M. Mohtashami et al.
3 Methods
3.1 Primary Mouse Care must be taken to practice sterile culture techniques, especially
Fibroblast Culture for the isolation of primary mouse dermal fibroblasts, as the skin is
a principal source of contamination. The cultures can be prepared
as below [15]:
1. Ear tips are removed from mouse (see Note 4) that is eutha-
nized, in accordance to the institution’s Animal User Protocol
approval, using sterile scissors and forceps. The tissue samples
are then washed with 10 mL 70 % ethanol for 5 min and trans-
ferred to a laminar flow clean bench and rinsed in 10 mL
PBS. The samples are minced (~5 mm2) using sterilized surgi-
cal scissors under PBS and treated with trypsin (0.25 %) for at
least 1 h at 37 ° C.
2. Using fine sterile forceps, the apposed layers of ear epidermis
are pulled away from each other, exposing the dermis. The
peeled away tissues are placed under sterile coverslips in wells
of a 6-well per plate with 2.5 mL of Fibroblast media. The
coverslips are pressed down against the tissue with the hair side
against the coverslip using a sterile pipette tip. This has the
double effect of forcing out air bubbles and of increasing sur-
face interaction between the dermal tissue and the tissue cul-
ture plate. Several wells may be prepared this way.
3. Each well is monitored, and cells are given time to grow out
into the well for 7–10 days. Fibroblast cells migrate from the
tissue to the plate.
4. To establish a cell culture of primary fibroblasts, the media is
removed and 2 mL PBS is used to wash off any remaining
Induction of T Cell Development In Vitro by Delta-Like (Dll)-Expressing Stromal Cells 163
3.2 Mouse 1. Retroviral vector pMIG containing the Dll1 or Dll4 cDNA
Fibroblasts coding region tagged with hemagglutinin (HA) peptide [8]
Transduced to Express and an IRES-eGFP (pMIG-DL) are transfected into HEK-
Dll1/ Dll4 293T cells, using CaPO4 transfection, along with plasmids
encoding the VSV-Env and GAG/Pol. Briefly, pMIG-DL
(10 ug), VSV-env (0.3 ug) and GAG/Pol (1 ug) are mixed in
450 uL of TE and 50 uL of CaCl2 before their addition to
500 uL of 2× BES solution, while vigorously mixing by mak-
ing bubbles. The mixture is then incubated at room tempera-
ture for 20 min followed by their addition to a 10 cm plate of
HEK293Ts.
2. After overnight incubation, the medium containing the trans-
fection material is discarded. Cells are washed very gently with
5 mL PBS, and replaced with 7–10 mL fibroblast medium.
3. Within 8 h after replacement with fresh fibroblast medium,
supernatant containing the retroviral particles produced and
released into the medium by HEK293T cells is collected. To
exclude possible HEK293Ts, the supernatant is filtered using
0.2 μm filters. The filtered supernatant is then placed on
50 % confluent GP+E-86 viral packaging cells. The infection
164 M. Mohtashami et al.
3.3 Isolation of HSCs All experimental procedures related to the use of mice require
from Mouse d15 Fetal approval and are performed according to the guidelines specified
Liver by local institution’s animal user protocol guidelines.
1. CD1 (or desired mouse strain) timed pregnant mice are com-
mercially available from Charles River Laboratories.
Alternatively, timed paired matings may be set up in-house
in local animal facility.
2. At day 14–15 p.c., fetal mice are obtained and the livers dis-
sected using sterilized scissors and forceps.
3. Using the sterile rubber end of a 3 or 5 mL syringe plunger,
day 14–15 fetal livers are then crushed against a 40 μm cell
strainer that fits into a 50 mL conical tube. The strainer is
washed several times using coculture medium.
4. The cells are centrifuged at 480 × g for 5 min at 4 °C. The super-
natant is discarded and the cells in the pellet are resuspended in
4 mL of coculture medium per up to ten fetal livers.
Induction of T Cell Development In Vitro by Delta-Like (Dll)-Expressing Stromal Cells 165
3.4 Initiation Cells may be seeded in 6-well plates or 10 cm plates or other ves-
and Maintenance sels. Here we describe coculture in 6-well plates.
of Coculture
1. Seed 1 × 103 HSCs in 3 mL of coculture medium per 1 well of
~90 % confluent OP9-DL or mFibro-DL cells, and add 5 ng/
mL Flt-3L and 1 ng/mL IL-7 and 25 ng/mL SCF (Note, the
addition of SCF is not necessary when setting up OP9-DL1
cocultures). Place the culture plates in the incubator.
166 M. Mohtashami et al.
4 Notes
References
1. Jenkinson EJ, Owen JJ (1990) T-cell differen- levels shows differential lymphomyeloid lin-
tiation in thymus organ cultures. Semin eage commitment outcomes. J Immunol
Immunol 2:51 185:867
2. Takahama Y (2000) Differentiation of mouse 9. Zuniga-Pflucker JC (2004) T-cell development
thymocytes in fetal thymus organ culture. made simple. Nat Rev Immunol 4:67
Methods Mol Biol 134:37 10. Beaudette-Zlatanova BC et al (2011) A human
3. Deftos ML, Bevan MJ (2000) Notch signaling thymic epithelial cell culture system for the
in T cell development. Curr Opin Immunol promotion of lymphopoiesis from hematopoi-
12:166 etic stem cells. Exp Hematol 39:570
4. Schmitt TM, Zuniga-Pflucker JC (2002) 11. Mohtashami M, Zuniga-Pflucker JC (2006)
Induction of T cell development from hemato- Three-dimensional architecture of the thymus
poietic progenitor cells by delta-like-1 in vitro. is required to maintain delta-like expression
Immunity 17:749 necessary for inducing T cell development.
5. Dervovic DD, Ciofani M, Kianizad K, Zuniga- J Immunol 176:730
Pflucker JC (2012) Comparative and func- 12. Mohtashami M, Shah DK, Kianizad K, Awong
tional evaluation of in vitro generated to G, Zuniga-Pflucker JC (2013) Induction of
ex vivo CD8 T cells. J Immunol 189:3411 T-cell development by Delta-like 4-expressing
6. Dervovic DD et al (2013) Cellular and molec- fibroblasts. Int Immunol 25:601
ular requirements for the selection of in vitro- 13. Smeets MF et al (2013) Removal of myeloid
generated CD8 T cells reveal a role for Notch. cytokines from the cellular environment
J Immunol 191:1704 enhances T-cell development in vitro. Int
7. Holmes R, Zuniga-Pflucker JC (2009) The Immunol 25:589
OP9-DL1 system: generation of T-lymphocytes 14. Schmitt TM, Zuniga-Pflucker JC (2006) T-cell
from embryonic or hematopoietic stem cells development, doing it in a dish. Immunol Rev
in vitro. Cold Spring Harb Protoc 2009:pdb. 209:95
prot5156 15. Takashima A (2001) Establishment of fibro-
8. Mohtashami M et al (2010) Direct comparison blast cultures. Curr Protoc Cell Biol Chapter
of Dll1- and Dll4-mediated Notch activation 2:Unit 2.1
Chapter 15
Abstract
From the moment a developing thymocyte expresses a TCR, it is subjected to numerous interactions with
self-peptide/MHC complexes that determine its ultimate fate. These include death by neglect, negative
selection (apoptosis and lineage deviation), positive selection, and lineage commitment. The identification
of signals that govern these unique cell fates requires the ability to assess the activity, level of expression,
subcellular location, and the molecular associations of numerous proteins within the developing T cell.
Thus, this chapter describes methods designed to analyze thymocyte signaling under various types of
peptide-based stimulation in vitro.
Key words T cell development, Positive selection, Negative selection, TCR signaling, Death by neglect
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_15, © Springer Science+Business Media New York 2016
169
170 Cody A. Cunningham et al.
2 Materials
3 Methods
3.1 Harvesting 1. Prepare sterile tools and small Petri dishes with 1 mL of com-
Thymi (See Note 5) plete media and 1 piece of sterile nylon mesh.
2. Place Petri dish on ice.
3. Harvest thymi from mice that have been euthanized according
to your institutional guidelines. Place in the Petri dish on ice
(one thymus per Petri dish).
4. Place another piece of mesh over the thymus and press with a
plunger from a 3 mL syringe to create a single cell
suspension.
5. Pass cells to a clean 15 mL conical tube that contains 3 mL of
complete media.
6. Rinse the mesh with 1 mL of fresh complete media and trans-
fer to the conical tube.
7. Spin cells at 483 × g (450–500 × g) for 5 min in centrifuge (4 °C).
8. Resuspend cells in fresh complete media (1–2 mL/thymus).
9. Transfer the cells to a new conical (50 mL) through cell
strainer (or nylon mesh) to remove large clumps of cells and
thymic debris.
10. Count with a hemocytometer (or electronic counter) and
dilute cells to a concentration 10 × 106/mL. Proceed to stimu-
lation of your choice.
In vitro Analysis of Thymocyte Signaling 173
3.2 Stimulation 1. Prepare media that contains the peptide at the desired concen-
with APC tration (see Note 6).
2. Resuspend APC at 20 × 106/mL in the peptide solution (from
3.2.1 Peptide-
step 1).
Loaded APC
3. Incubate at 37 °C in the culture incubator from a minimum of
30 min.
4. Wash cells with complete media, spin at 483 × g (450–500 × g)
for 5 min and discard supernatant.
5. Resuspend cells at 10 × 106 cells/mL of complete media.
3.3 Confocal 1. Wash cells from stimulation (above) and resuspend pellet
Microscopy (10 × 106 cells/mL) by gently tapping the microfuge tube (see
Note 8).
3.3.1 Mounting Cells
2. Place 100 μL of stimulated cells on the appropriate coverslip.
TAKE NOTE OF PLATE LAYOUT.
3. To allow the cells to settle onto the coverslip, carefully place
covered 6-well plates in refrigerator for minimum of 10 min.
During this time prepare the staining buffers. Fixation:
(see Note 9).
174 Cody A. Cunningham et al.
3.3.2 Permeablization/ 1. Gently, but quickly add 100 μL of ice cold 0.2 % triton drop-
Blocking wise and incubate for 2 min. (For the best results, work with
six wells at a time. Start the timer after adding the triton to last
coverslip.)
2. Aspirate triton and add ice cold 3 % BSA/PBS.
3. Repeat steps 5 and 6 until all coverslips have been treated.
4. Place plates at 4 °C overnight. Alternatively, block for a mini-
mum of 1 h then proceed to staining procedure.
3.3.4 Mounting 1. Using a forceps to pick up coverslip, dab the edge of the cov-
the Coverslip erslip with a lint-free tissue to remove excess stain. Then dip
the coverslip in PBS and dab the edge of the coverslip with
tissue three times, then dip in water and dab once. Place the
coverslip CELL SIDE DOWN, onto a microscope slide that
has one drop of mounting media (see Note 10).
2. Cover the mounted coverslip/slides and let dry overnight at
RT, then paint the edges of each coverslip with clear nail polish
(to make a seal) and place in slide box and place at 4 °C. NOTE:
The slides are then ready for analysis by confocal microscopy.
While in theory the stains remain stable for up to a year, we
suggest they be analyzed within 3 months of mounting.
3.4.2 Sample Lysis 1. Lyse 10 × 106 thymocytes in 100 μL Lysis Buffer (ice-cold) in
a 1.5 mL microcentrifuge tube for 20 min on ice.
2. Following lysis spin crude lysate for 50 min at ~16,000 × g at
4 °C.
3. Pass supernatant to a fresh 1.5 mL microcentrifuge tube
(see Note 11).
176 Cody A. Cunningham et al.
3.4.3 Staining of Beads 1. Wash CML beads that have been incubated with lysate three
for FCM times in ice-cold Lysis Buffer being sure to discard as much
buffer as possible without disturbing the CML bead pellet.
2. Resuspend CML beads in FCM staining buffer and pass beads
to 96-well round-bottom plate.
(a) Split the CML beads into enough wells to assess the pro-
teins of interest plus isotype control(s).
3. Add fluorochrome-conjugated mAbs (or primary Abs) to the
samples and incubate for 30 min on ice in darkness. α-POSH,
clone M-290, α-JIP-1 clone M-300 and α-JNK1 clone C-17
used for demonstration (Fig. 2) [5].
4. Wash beads two times with FCM Buffer, spinning at ~500 × g
for 5 min at 25 °C.
5. If unlabeled primary Abs are used, add fluorochrome-
conjugated secondary Ab and incubate for 30 min on ice in
darkness. If directly conjugated Abs are used, skip this step.
6. Resuspend IP beads in 200 μL FCM Staining Buffer and pro-
ceed to analysis on the flow cytometer.
Fig. 2 IP-FCM analysis of protein interactions in pre-selection DP thymocytes. (a) Gating on singlet beads
based on FSC and SSC properties. (b) Proper calibration of fluorescence voltage such that the isotype control
falls between 100 and 101 while a “bright” positive control falls on scale. (c) Representative experiment where
α-POSH CML beads are used on lysates of pre-selection DP thymocytes. Histograms represent the presence
of JIP-1 and JNK1 bound to POSH. Red histogram represents the “loading control” to quantify how much POSH
was precipitated
In vitro Analysis of Thymocyte Signaling 177
3.4.4 Acquisition of Data 1. Increase the FSC and SSC of the cytometer such that bead
by FCM singlets are clearly visible (see Note 13).
2. Draw a gate around the population of bead singlets (Fig. 2a).
3. Apply this gate and visualize the fluorescence data on a log
scale for each of the fluorescence channels being used.
4. Pass the isotype control sample and confirm that the generated
histogram falls between 100 and 101 (see Note 14).
5. Pass the control for a very bright sample (see Note 15).
6. Pass experimental samples (Fig. 2c).
7. Acquire >1000 bead events. Analyze as described [5, 7].
4 Notes
Acknowledgements
The authors thank the members of the Daniels and Teixeiro labs
for assistance in compiling these protocols. Funding provided by
grants from the Missouri Mission Enhancement Fund and
University of Missouri Research Board is greatly acknowledged.
References
1. Starr TK, Jameson SC, Hogquist KA (2003) 5. Cunningham CA, Knudson KM, Peng BJ,
Positive and negative selection of T cells. Annu Teixeiro E, Daniels MA (2013) The POSH/
Rev Immunol 21:139–176 JIP-1 scaffold network regulates TCR-mediated
2. Stritesky GL, Jameson SC, Hogquist KA JNK1 signals and effector function in CD8 T
(2012) Selection of self-reactive T cells in the cells. Eur J Immunol 43(12):3361–3371
thymus. Annual Rev Immunol 30:95–114 6. Gil D, Schrum A, Alarcon B, Palmer E (2005)
3. Daniels MA, Teixeiro E, Gill J, Hausmann B, T cell receptor engagement by peptide – MHC
Roubaty D, Holmberg K, Werlen G, Holländer ligands induces a conformational change in the
GA, Gascoigne NR, Palmer E (2006) Thymic CD3 complex of thymocytes. J Exp Med
selection threshold defined by compartmental- 201(4):517–522
ization of Ras/MAPK signalling. Nature 7. Schrum AG, Gil D, Dopfer EP, Wiest DL,
444(7120):724–729 Turka LA, Schamel WW, Palmer E (2007)
4. Daniels MA, Teixeiro E (2010) The persistence High-sensitivity detection and quantitative
of T cell memory. Cell Mol Life Sci 67(17): analysis of native protein-protein interactions
2863–2878 and multiprotein complexes by flow cytometry.
Sci STKE 2007(389):pl2
Chapter 16
Abstract
PCR on genomic DNA isolated from lymphocyte populations is an invaluable technique to analyze T cell
receptor (TCR) α and β gene rearrangements. Although this approach is powerful, it also has limitations
that must be accounted for in experimental design and data interpretation. Here, we provide background
required for understanding these limitations, and then outline standard PCR methods that can be used for
analysis of TCRα and β gene rearrangements in mice.
Key words VDJ recombination, T cell receptor (TCR) α and β genes, Variable (V), diversity (D), and
joining (J) gene segments, Thymocytes, αβ T cells
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_16, © Springer Science+Business Media New York 2016
179
180 Levi J. Rupp et al.
1.2 Structure The genomic structure of TCRβ loci is conserved across commonly
and Recombination used lab mouse strains. The TCRβ locus spans 685 kb on chromo-
of TCRβ Loci some 6. It is comprised of 23 functional Vβ segments and two
Dβ-Jβ-Cβ clusters; each Dβ-Jβ-Cβ cluster spans ~6 kb and contains
a single Dβ segment (Dβ1 or Dβ2), six functional Jβ segments
(Jβ1.1-1.7 or Jβ2.1-2.7 where Jβ1.7 and Jβ2.6 are not functional),
and two sets of Cβ region exons (Cβ1 or Cβ2) (Fig. 1). All but two
of the 23 functional Vβs reside within a 234 kb Vβ cluster that ends
250 kb upstream of Dβ1. One of the other two Vβs resides 156 kb
upstream of this Vβ cluster and the second lies 10 kb downstream
of Cβ2. In addition to these 23 functional Vβs, the Vβ cluster con-
tains 12 non-functional Vβs that are pseudogenes or lack compe-
tent RSSs. Except for the Vβ downstream of Cβ2, all Vβs reside in
the same transcriptional orientation as the Dβ-Jβ-Cβ clusters.
Historically, Vβ segments were named in order numerically as
they were identified. Presently, the IMGT (www.imgt.org) and
NCBI name Vβ segments based on their genomic location with the
Vβ furthest upstream of Dβ1 called TRBV1 and the other func-
tional and non-functional Vβs designated by number in ascending
order moving along the locus (with the exception of the TRBV12
and TRBV13 families that are interspersed). Dβ and Jβ segments
Fig. 1 Schematic of TCRβ locus. Gene segments are listed below each region, from distal to proximal, and are
identified by NCBI and IMGT nomenclature. Gray lettering indicates that segments are non-functional. Locus is
not to scale, and double hash marks indicate large intergenic distances. Arrow denotes that TRBV31 is in
opposite transcriptional orientation to the rest of the locus
Molecular Analysis of Mouse T Cell Receptor α and β Gene Rearrangements 181
are also numbered in ascending order and labeled TRBD and TRBJ
in the current system. Peptides encoded by Vβ segments, however,
are still referred to by their historical names. For example, the
TRBV1 segment was historically called Vβ2, and TCRβ proteins
that contain this Vβ are still referred to as Vβ2+ TCRβ chains. In
Table 1, we summarize the differences between historical and
IMGT/NCBI nomenclature. For this chapter, we use the IMGT/
NCBI nomenclature for Vβ gene segments and encoded peptides.
Table 1
Current NCBI and historical nomenclature for TCRβ locus variable (V)
region segments
(continued)
182 Levi J. Rupp et al.
Table 1
(continued)
1.3 Structure The TCRα locus is larger than the TCRβ locus and contains many
and Recombination more gene segments. Moreover, it is polymorphic and structurally
of TCRα Loci distinct in different strains of inbred mice (Fig. 2). Five different
Vα haplotypes were described based on restriction fragment length
polymorphisms [5]. Detailed sequence analysis is available for
strains 129 (which has the same haplotype as BALB/c, AKR,
CBA/J, C3H and A/J) and C57BL/6. The 129 TCRα locus spans
about 1.6 Mb on chromosome 14, whereas the C57BL/6 locus
spans about 1.8 Mb, with the major structural difference mapping
to the array of V gene segments. The 129 locus contains 104 V
gene segments, of which 88 are likely functional, whereas the
C57BL/6 locus contains 138 V gene segments, of which 117 are
likely functional. A complicating feature of the locus is the repeti-
tive nature of the V gene array. In strain 129 mice, there is a 400 kb
duplication in the central portion of the array, with nearly identical
V gene paralogues in each repeat. C57BL/6 mice have copies of
these repeats plus an additional repeat sandwiched between them,
creating even greater complexity. Both strains have unique V gene
segments in non-duplicated regions that flank the repeat region.
The repeat structure makes locus analysis rather challenging.
Downstream of the V gene segments are Dδ, Jδ and Cδ gene
segments that are used to assemble TCRδ genes. Downstream of
Cδ is a single V gene segment in opposite transcriptional orienta-
tion to other gene segments in the locus; this is followed by an
array of 60 Jα gene segments (of which 43 are likely functional as
defined by their ability to undergo recombination and splicing to
create mature TCRα transcripts) and Cα. TCRα rearrangement is
Vα-to-Jα, whereas TCRδ rearrangement is Vδ-to-Dδ-to-Jδ.
TCRα locus nomenclature has been particularly challenging
because there have been several versions; for example, Vα19 was
called AV19 by Arden [6] and is now referred to as TRAV1 by
IMGT and NCBI, which name the Vα gene segment families in
linear order of their first occurrence, from distal to proximal.
184 Levi J. Rupp et al.
Fig. 2 Schematic of TCRα locus. The structures of the 129 and C57BL/6 TCRα loci are presented. The repeat
structure of the V array is noted and the portions shared by 129 and C57BL/6 as well as the portion unique to
C57BL/6 are indicated. Gene segments are listed below each region, from distal to proximal, and are identified
by NCBI and IMGT nomenclature. Gray lettering indicates that genes are non-functional based on the presence
of a stop codon or their inability to rearrange or undergo appropriate mRNA splicing. These designations
diverge somewhat from those in IMGT based on Jα repertoire studies conducted in our laboratory [15–18] as
well as deep sequencing of the peripheral Vα-Jα repertoire [14]. Those V gene segments that are recognized
by IMGT to serve as both a Vα and a Vδ have a dual designation (e.g., 15-1/DV6-1, short for TRAV15-1/
TRDV6-1); those that are recognized to serve exclusively as a Vδ have a single designation (e.g., DV1, short for
TRDV1). Data on TRAV haplotypes were previously reported [5]
Fig. 3 Diagram of αβ T cell development with markers relevant for cell sorting. Critical checkpoints in αβ T cell
development are shown, including surface markers that will differentiate pre- and post-selection DN thymo-
cytes, and pre- and post-selection DP thymocytes
186 Levi J. Rupp et al.
1.5 Analysis PCR is the most commonly used technique for analysis of antigen
of TCRα and β Gene receptor gene rearrangements because it generates large amounts
Rearrangements of information in a short amount of time. Both conventional (see
Subheading 3.1) and real-time (see Subheading 3.2) PCR applica-
tions may be applied to characterize TCRα and TCRβ rearrange-
ments. Additionally, PCR may be coupled with a Southern blot
and probe approach (see Subheading 3.3) to increase sensitivity;
this application is particularly relevant for analysis of TCRα rear-
rangements but can be utilized at either locus.
The general PCR strategy is to amplify TCRα and β gene
rearrangements from genomic DNA using a forward primer that
anneals in a V segment or upstream (5′) of a D segment and a
reverse primer that anneals in or downstream (3′) of a J segment. In
most cases, these primers anneal to genomic sequences too far apart
to amplify products from germline (un-rearranged) alleles, the
exceptions being PCR reactions with 5′Dβ and 3′Jβ primers since
Dβ segments reside ~700 bp upstream of the 5′ Jβ segment in each
cluster. Although PCR reactions on serial dilutions of input DNA
are commonly used to determine the linear range of amplification
and provide semi-quantitative analyses of V(D)J recombination,
Molecular Analysis of Mouse T Cell Receptor α and β Gene Rearrangements 187
Table 2
Primers for TCRβ join PCRs including oligos 5′ of Dβ and 3′ of Jβ segments
Table 3
Current NCBI and historical nomenclature for representative TCRα locus
variable (V) region segments
Current NCBI and historical nomenclature for TCRα locus joining (J) region segments
NCBI Historical
nomenclature nomenclature Join PCR primers Southern blot probe
TRAJ61 Jα61 ATGAGTCTTCCAGTCATGGC (R)
TRAJ58 Jα58 GACTCACTGTGAGCTTTGCC (R,C) AGCTTAGACCCAGTGCCTTG
(R,C)
Levi J. Rupp et al.
Table 5
Cell surface markers for isolation of pre- and post-selection DN or DP
thymocytes
2 Materials
2.1 Conventional All PCRs should use molecular biology grade H2O, while standard
PCR tap water is acceptable for running buffers.
1. diH2O—molecular biology grade.
2. Cells of interest (see Note 1).
3. Rapid Lysis Buffer: 100 mM Tris-base pH 7.4, 0.2 % sodium
dodecyl sulfate, 5 mM EDTA, 200 mM NaCl.
4. Proteinase K. Re-suspended at 20 mg/mL in diH2O.
5. Isopropanol.
6. TE buffer pH 8.0: 10 mM Tris-base pH 8.0, 1 mM EDTA in
diH2O.
7. 10× PCR reaction buffer (as supplied by DNA polymerase
manufacturer).
8. DNA polymerase (see Note 2).
9. 10 mM dNTP mixture (2.5 mM each dATP, dTTP, dCTP,
dGTP), available commercially.
10. Primers of interest (see Tables 1, 2, 3, and 4). Re-suspend
primers at 5 pmol/μL using diH2O.
11. CD14 control primers. Sense primer: 5′-GCTCAAACTTTCA
GAATCTAC-3′; Antisense primer: 5′-AGTCAGTTCGTGGA
GGCCGGAAATC-3′; if following with Southern blot
(Subheading 3.3), probe: 5′-AGCAGATCTGGGGCAGTT
CACTGA-3′.
12. Standard thermocycler.
13. 1× TBE buffer: 89 mM Tris-base, 89 mM boric acid, 2 mM
EDTA in H2O. To make a 5× stock solution: Dissolve 1080 g
Tris-base, 550 g boric acid, and 400 mL 0.5 M EDTA in 5 L
H2O. Adjust final volume to 20 L, and dilute to 1× in H2O
prior to use. Adjustment to precise pH is not necessary at any
point.
14. Agarose.
15. Ethidium bromide (10 mg/mL stock solution).
16. 6× Gel Loading Dye.
17. 1 kb DNA ladder.
2.3 Southern Blot Molecular biology grade diH2O should be utilized for steps involv-
ing DNA while standard diH2O is suitable for running buffer,
transfer buffer, hybridization solution, and washing buffers.
1. Join PCR product from Subheading 3.1.
2. 1× TAE buffer: 40 mM Tris-base, 0.11 % glacial acetic acid
(v/v), 1 mM EDTA in H2O. To make a 50× TAE stock solu-
tion: Dissolve 968 g Tris-base in 2 L H2O. Add 228.4 mL gla-
cial acetic acid and 400 mL 0.5 M EDTA solution. Adjust final
volume to 4 L in H2O. Dilute to 1× in H2O prior to use.
Adjustment to precise pH is not necessary at any point.
3. Agarose.
4. Ethidium bromide (10 mg/mL stock solution).
5. DNA ladder (see Note 3).
6. UV source (gel box or wand).
7. Denaturing solution: 0.5 M NaOH, 0.6 M NaCl in diH2O.
8. Tupperware (see Note 4).
9. Orbital shaker such as Stovall Belly Dancer™.
10. Sponges (see Note 5).
11. Blotting paper (see Note 6).
12. 10× SSC: 1.5 M NaCl, 150 mM trisodium citrate in diH2O. To
make a 20× SSC stock solution: Dissolve 3506 g NaCl and
1764 g sodium citrate in a final volume of 20 L H2O. Dilute
as necessary in H2O prior to use. Adjustment to precise pH is
not necessary at any point.
13. Nylon membrane (see Note 7).
14. Paper towels.
15. Parafilm.
16. Disposable pipette.
17. Gel box and electrophoresis power source.
18. Drying oven.
19. 19× SSCPE: 2.85 M NaCl, 285 mM trisodium citrate,
247 mM KH2PO4, and 19 mM EDTA in H2O. Adjustment to
precise pH is not necessary.
20. Oligo pre-hybridization solution: 6× SSCPE, 5× Denhardt’s
solution, 1 % SDS in H2O. Prepare 100 mL by combining the
following: 31.5 mL 19× SSCPE, 10 mL 50× Denhardt’s solu-
tion, 10 mL 10 % SDS solution, and 48.5 mL diH2O. Adjustment
to precise pH is not necessary.
21. Oligo hybridization solution: 6× SSCPE, 3× Denhardt’s solu-
tion, 1 % SDS, 100 μg/mL yeast tRNA in H2O. Prepare
100 mL by combining the following: 31.5 mL 19× SSCPE,
6 mL 50× Denhardt’s solution, 10 mL 10 % SDS solution,
Molecular Analysis of Mouse T Cell Receptor α and β Gene Rearrangements 195
3 Methods
3.1 Conventional 1. Isolate genomic DNA by method of choice. We utilize the lysis
PCR and isopropanol precipitation protocol described in steps 2–5.
2. Pellet 104–107 cells of interest in 1.7 mL eppendorf tube.
Re-suspend cells in 500 μL rapid lysis buffer (see Note 10)
with 5 μL Proteinase K (20 mg/mL stock). Incubate over-
night at 56 °C.
3. Add 800 μL isopropanol to Eppendorf tube containing lysed
cells. Mix thoroughly by shaking.
4. Centrifuge 10 min at maximum speed in bench-top centrifuge
to pellet DNA.
5. Aspirate supernatant. Re-suspend DNA in appropriate volume
TE buffer or diH2O (see Note 11) and place at 56 °C for 2 h.
6. Determine DNA concentration by method of choice. Dilute
an aliquot to 50 ng/μL in diH2O.
196 Levi J. Rupp et al.
3.3 Southern Blot Unless otherwise described, all procedures may be carried out at
room temperature. All solutions may be stored at room tempera-
ture, but we store the oligo pre-hybridization solution, oligo
hybridization solution, and wash buffer at 42 °C for ease of use. All
steps involving radiation should be performed behind a radiation
shield and according to relevant institutional guidelines. We utilize
a dedicated sink, 37 °C heating block, and heated shaker for radia-
tion purposes.
1. Prepare running gel by dissolving 4 g agarose in 500 mL 1×
TAE with ethidium bromide (5 μL of stock 10 mg/mL solu-
tion). Combs should be sized to permit 50 μL loading volume
(see Note 14).
2. Load samples on running gel, leaving the outer two lanes on
each side empty, as these will be lost in transfer. Load one lane
of DNA ladder (see Note 15).
3. Run gel overnight (18–24 h) at 23 V.
4. Remove gel from gel box and examine by UV to verify sam-
ples have amplified (see Note 16).
5. Activate the gel by treating for 30 s with short-wave UV irra-
diation (~254 nm) using wand or gel box, with appropriate
shielding.
6. Fill a Tupperware container with 1 L of denaturing solution.
Place gel in Tupperware. Shake gently on orbital shaker for
1 h at room temperature (see Note 17).
7. Prepare for transfer. Place a series of sponges (large enough to
fully support the gel) side-by-side in large Tupperware con-
tainer. Lay one sheet of blotting paper over the sponges,
198 Levi J. Rupp et al.
4 Notes
Acknowledgements
References
1. Schatz DG, Swanson PC (2011) V(D)J recom- 7. Williams JA, Hathcock KS, Klug D et al (2005)
bination: mechanisms of initiation. Annu Rev Regulated costimulation in the thymus is criti-
Genet 45:167–202 cal for T cell development: dysregulated CD28
2. Bassing CH, Swat W, Alt FW (2002) The costimulation can bypass the pre-TCR check-
mechanism and regulation of chromosomal point. J Immunol 175:4199–4207
V(D)J recombination. Cell 109:S44–S55 8. Taghon T, Yui MA, Pant R et al (2006)
3. Sleckman BP, Gorman JR, Alt FW (1996) Developmental and molecular characterization
Accessibility control of antigen-receptor of emerging β- and γδ-selected pre-T cells in
variable-region gene assembly: role of cis-acting the adult mouse thymus. Immunity 24:53–64
elements. Annu Rev Immunol 14:459–481 9. Khor B, Sleckman BP (2002) Allelic exclusion
4. Krangel MS (2009) Mechanics of T cell recep- at the TCRβ locus. Curr Opin Immunol
tor gene rearrangement. Curr Opin Immunol 14:230–234
21:133–139 10. Malissen M, Trucy J, Jouvin-Marche E et al
5. Jouvin-Marche E, Morgado MG, Trede N (1992) Regulation of TCRα and β gene allelic
et al (1989) Complexity, polymorphism, and exclusion during T-cell development. Immunol
recombination of mouse T-cell receptor α gene Today 13:315–322
families. Immunogenetics 30:99–104 11. Gascoigne NR, Alam SM (1999) Allelic exclu-
6. Arden B, Clark SP, Kabelitz D, Mak TW (1995) sion of the T cell receptor α-chain: develop-
Mouse T-cell receptor variable gene segment mental regulation of a post-translational event.
families. Immunogenetics 42:501–530 Semin Immunol 11:337–347
202 Levi J. Rupp et al.
Intrathymic Injection
Sugata Manna and Avinash Bhandoola
Abstract
Intrathymic injection is used in several T cell-associated immunological studies to deliver cells or other
substances directly into the thymus. Here, we describe the intrathymic injection procedure involving surgi-
cal incision of the mouse with or without a thoracotomy. Though this procedure can result in poor recov-
ery, postsurgical complications, and distress to the animal, it is actually a simple procedure that can be
carried out relatively easily and quickly with experience.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_17, © Springer Science+Business Media New York 2016
203
204 Sugata Manna and Avinash Bhandoola
2 Materials
1. Sterile drape.
2. Sterile latex surgical gloves.
3. Sterile gauge sponge or swab.
4. 1 cc insulin syringe, with 29 1/2 G ultrafine needle.
5. Betadine.
6. Surgical board.
7. Rubber bands.
8. Eye moisture salve.
9. Forceps, scissors appropriate for incision.
10. Wound clip.
11. 10 μL syringe (Hamilton Co; Reno, NV), attached with a
27 G needle (no dead volume).
12. Anesthetics: Ketamine and xylazine (see Notes 1 and 2).
Ketamine and xylazine should be diluted in 1× PBS. Ketamine/
xylazine anesthetic should be administered at a dose of
0.1 mg/g mouse weight and 0.02 mg/g mouse weight for
ketamine and xylazine, respectively.
3 Procedures
3.1 Intrathymic 1. Anesthetize the mice with ketamine and xylazine adminis-
Injection with tration at a dose/volume of 0.01 mL/g mouse weight
Thoracotomy (~200 μL for a 20 g mouse) via intraperitoneal injection
(use 1 cc insulin syringe, with 29 1/2 G ultrafine needle)
(see Notes 3 and 4).
2. Rinse the work area with betadine and lay down a sterile drape.
3. Place the anesthetized mouse on its back on a surgical board
and immobilize it by strapping its feet with rubber bands
(Fig. 1a) (see Note 5).
Intrathymic Injection 205
Fig. 1 (a) Anesthetized mouse placed and immobilized with rubber bands on surgical board. (b) Midline longi-
tudinal incision, with mouse’s rib cage cut for visualization of thymus. (c) Intrathymic injection, mouse chest
held open with forceps and needle with bevel up, is positioned and inserted into the thymus
16. Return mouse to the cage; check to ensure that tongue is still
out. Place subsequent mice next to previous ones to conserve
heat until they recover from anesthesia (may take up to 2 h for
complete recovery) (see Notes 12 and 13).
Fig. 2 (a) Small incision made perpendicular to the sternum and across the midline of the upper thoracic
region. (b) Intercostal intrathymic injection, the operator inserts the needle between third and fourth ribs
208 Sugata Manna and Avinash Bhandoola
4 Notes
Acknowledgments
References
1. Rezzani R, Nardo L, Favero G et al (2014) cellular traffic from the thymus to the periph-
Thymus and aging: morphological, radiologi- ery in mice. Eur J Immunol 10:210–218
cal, and functional overview. Age 36:313–351 12. Weber BN, Chi AW, Chavez A et al (2011) A
2. Chen W, Issazadeh S, Sayegh MH et al (1997) critical role for TCF-1 in T-lineage specifica-
In vivo mechanisms of acquired thymic toler- tion and differentiation. Nature 476:63–68
ance. Cell Immunol 179:165–173 13. Guidos CJ, Weissman IL, Adkins B (1989)
3. Chen W, Sayegh MH, Khoury SJ (1998) Intrathymic maturation of murine T lympho-
Mechanisms of acquired thymic tolerance cytes from CD8+ precursors. Proc Natl Acad
in vivo: intrathymic injection of antigen Sci U S A 86:7542–7546
induces apoptosis of thymocytes and periph- 14. Jerabek L, Weissman IL (2002) Intrathymic
eral T cell anergy. J Immunol 160:1504–1508 injection for analysis of T-cell progenitor
4. DeMatteo RP, Chu G, Ahn M et al (1997) activity. In: Klug CA, Jordan CT (eds)
Long-lasting adenovirus transgene expression Hematopoietic stem cell protocols, vol 63,
in mice through neonatal intrathymic toler- Methods Mol Med. Humana Press, Totowa,
ance induction without the use of immuno- NJ, pp 161–165
suppression. J Virol 71:5330–5335 15. Hardy RR, Shinton SA (2004) Characterization
5. Muto M, Kubo E, Sado T (1987) Development of B lymphopoiesis in mouse bone marrow
of prelymphoma cells committed to thymic and spleen. In: Gu H, Rajewsky K (eds) B cell
lymphomas during radiation-induced thymic protocols, Methods in molecular biology.
lymphomagenesis in B10 mice. Cancer Res Humana Press, Totowa, NJ, pp 1–24
47:3469–3472 16. Rooke R, Benoist C, Mathis D (2000)
6. Goldschneider I, Komschlies KL, Greiner DL Intrathymic delivery of MHC genes using
(1986) Studies of thymocytopoiesis in rats and recombinant Adenoviruses. In: Kearse KP (ed)
mice. I. Kinetics of appearance of thymocytes T cell protocols: development and activation,
using a direct intrathymic adoptive transfer Methods in molecular biology. Humana Press,
assay for thymocyte precursors. J Exp Med Totowa, NJ, pp 69–79
163:1–17 17. Liu LL, Du XM, Wang Z et al (2012) A sim-
7. Adjali O, Marodon G, Steinberg M et al plified intrathymic injection technique for
(2005) In vivo correction of ZAP-70 immu- mice. Biotech Histochem 87:140–147
nodeficiency by intrathymic gene transfer. 18. de la Cueva T, Naranjo A, de la Cueva E et al
J Clin Invest 115:2287–2295 (2007) Refinement of intrathymic injection in
8. Seggewiss R, Dunbar CE (2005) A new direc- mice. Lab Anim (NY) 36:27–32
tion for gene therapy: intrathymic T cell- 19. Adjali O, Montel-Hagen A, Swainson L et al
specific lentiviral gene transfer. J Clin Invest (2009) In vivo and ex vivo gene transfer in
115:2064–2067 thymocytes and thymocyte precursors. In:
9. De Obaldia ME, Bell JJ, Bhandoola A (2013) Baum C (ed) Genetic modification of hemato-
Early T-cell progenitors are the major granulo- poietic stem cells, vol 506, Methods Mol Biol.,
cyte precursors in the adult mouse thymus. pp 171–190
Blood 121:64–71 20. Handon RB, Mueller K, Miller SH (2010) An
10. De Obaldia ME, Bell JJ, Wang X et al (2013) alternative method for intrathymic injections
T cell development requires constraint of the in mice. Lab Anim (NY) 39:248–252
myeloid regulator C/EBP-α by the Notch tar- 21. Tuckett AZ, Thornton RH, Shono Y et al
get and transcriptional repressor Hes1. Nat (2014) Image-guided intrathymic injection of
Immunol 14:1277–1284 multipotent stem cells supports lifelong T-cell
11. Scollay RG, Butcher EC, Weissman IL (1980) immunity and facilitates targeted immunother-
Thymus cell migration: quantitative aspects of apy. Blood 123:2797–2805
Chapter 18
Abstract
Determination of cellular proliferation and population turnover is an important tool for research on lym-
phoid cell function. Historically this has been done using radiolabeled nucleotides or nucleoside analogs,
such as BrdU (5-bromo-2-deoxyuridine), that are incorporated into nascent DNA during S-phase.
Recently, a new procedure was developed to label nascent DNA using EdU (5-Ethynyl-2-deoxyuridine).
This new method overcomes limitations imposed by the procedure used to detect BrdU because EdU
detection is based on an easily performed chemical reaction that does not require DNA denaturation, is
quick and reproducible, and has a superior signal-to-noise ratio. This technique offers a wide range of
opportunities to analyze cellular proliferation, population homeostasis, and cell marking procedures.
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_18, © Springer Science+Business Media New York 2016
211
212 Francis A. Flomerfelt and Ronald E. Gress
EdU to form a stable triazole ring [8]. Because of the small size of
the click detection reagents, no harsh DNA denaturation steps are
required. As a result, EdU detection uses a simple protocol that
takes less than 30 min with more reproducible results and greater
signal-to-noise ratio. A variety of fluorochromes for EdU detection
facilitate analysis of specific populations defined antibody based
phenotypic profiling by specific proteins. It is likely that the advent
of new and improved techniques using EdU to label nascent DNA
will usher in a new wave of creative scientific exploration and excit-
ing findings.
2 Materials
2.3 Stain Cell 1. FACS buffer (1× PBS, 0.5–1.0 % BSA, 0.1 % (W/V) sodium
Surface Markers azide) (see Note 2).
2. Tubes compatible with flow cytometer to be used.
3. Fluorescently labeled antibodies.
4. Anti CD16 (2.4G2) antibody use at 5 μg/ml.
2.6 Optional 1. Cells that have been fixed, permeabilized, and stained for EdU
Additional Antibody with the Click-it reaction.
Staining for 2. Fluorescently labeled antibodies.
Intracellular
3. 1× Click-iT saponin-based permeabilization and wash buffer.
Antigens or with RPE,
PE-Tandem, or Qdot
Antibody Conjugates
3 Methods
3.1 In Vitro EdU 1. Establish culture conditions where cells are proliferating.
Labeling 2. Add 4 ml of DMSO or PBS to vial containing 10 mg EdU to
make a 10 mM solution (see Note 1).
3. Add EdU to culture at 2–10 μM. EdU can be dissolved in
DMSO or in aqueous solutions. EdU stock solution is 10 mM
(see Note 3). Include cultures treated with vehicle alone for
controls.
4. Culture for period to label nascent DNA (see Note 4). Harvest
cells, count, and wash with PBS. Resuspend in FACS buffer at
1 × 107 cells/ml and keep on ice.
3.2 In Vivo EdU 1. Prepare EdU at 1 mg/ml in sterile phosphate buffered saline
Labeling (PBS).
2. For short term or pulse labeling of adult mice, perform an
intraperitoneal (IP) injection of 100–200 μl dissolved EdU
(see Note 5). Harvest organs/cells at desired timepoints
(see Note 6).
3. For long term labeling, perform IP injection with 100 μl of
1 mg/ml EdU in PBS. Mice are then provided with drinking
water ad libitum containing 0.3 mg/ml EdU (see Note 7).
Replace drinking water with freshly prepared EdU every 2–3
days (see Notes 1 and 8).
4. Harvest lymphoid organs of interest and process into single
cell suspensions. Count cells and wash with PBS. Resuspend in
FACS buffer at 1 × 107 cells/ml and keep on ice.
5. If cells will not be stained with antibodies, place 100 μl of
the cell suspension (1 × 106 cells) into FACS tubes, add 3 ml
FACS buffer, pellet cells, remove supernatant and proceed to
Subheading 3.4.
Analysis of Cell Proliferation and Homeostasis Using EdU Labeling 215
3.3 Stain Cell 1. Prepare a chart for the staining strategy listing fluorochromes,
Surface Markers EdU detection and DNA stain. Antibodies using PE, PE-
tandem, or Qdot(R) conjugates should not be used before the
EdU detection step as their signal is reduced by the Click it reac-
tion conditions. Currently, Life Technologies offers azides with
Alexa Fluor 488, Alexa Fluor 647, and Pacific Blue. Also plan
for additional samples for controls including unstained cells and
single colors of each fluorochrome for compensation. When
analyzing rare populations, it is useful to use additional control
samples containing “all stains except one” to set the gates for
the population identified by the missing fluorochromes.
2. Place 100 μl of the cell suspension (1 × 106 cells) into FACS
tubes. Add 0.5 μg anti CD16/CD32 antibody in tubes to be
stained with antibodies to block antibody binding by the Fc
receptor and incubate on ice 10 min.
3. Set up single stain controls for each antibody. Reserve tubes
with cells for EdU detection reagents and DNA stains to be
added later. Incubate tubes on ice 15 min, add 3 ml FACS
buffer, pellet cells and remove supernatant. Proceed to
Subheading 3.4.
4. While control cells are staining, make a mixture with predeter-
mined amounts of all desired antibodies and add an aliquot to
samples to be stained. This procedure facilitates dispensing the
antibodies and reduces tube-to-tube variability. Incubate tubes
on ice 15–30 min, add 3 ml FACS buffer, pellet cells and
remove supernatant. Proceed to Subheading 3.4.
3.4 Fix and 1. All samples and controls should be fixed and permeabilized to
Permeabilize Cells ensure uniform characteristics when run on the flow cytome-
ter. Prepare needed amount of 1× Click-iT saponin-based per-
meabilization and wash reagent by diluting the provided 10×
stock (Component E) with water (see Note 9). A total of
3.6 ml per sample is needed for the standard reaction. If intra-
cellular staining will be done, a total of 6.7 ml per sample is
needed.
2. Gently drag tubes across a rack or use other method to break
up the cell pellets from Subheadings 3.2, step 5 and 3.3, steps
3 and 4.
3. Add 100 μl of Click-iT fixative (Component D) to each tube
and mix well. Incubate the cells for 15 min at room tempera-
ture in the dark.
4. Add 3 ml of FACS buffer, pellet cells, and remove the super-
natant (see Note 10).
5. Gently break up the cell pellet again and add 100 μl of 1×
Click-iT saponin-based permeabilization and wash reagent.
Mix well and proceed directly to Subheading 3.5 (see Note 11).
216 Francis A. Flomerfelt and Ronald E. Gress
3.5 EdU Detection 1. Allow kit components to come to room temperature before
opening.
2. Prepare a working solution of fluorescent azide for EdU detec-
tion by adding 130 μl of DMSO to Component B of the kit
(Alexa Fluor 488, Alexa Fluor 647, or Pacific Blue) and mix
well. Any remaining working solution will be stable for a year
if stored at ≤−20 °C.
3. Add 2 ml of deionized water to the vial containing the Click-iT
EdU buffer additive (Component G) to make a 10× stock
solution and gently mix until fully dissolved. Any remaining
stock solution should be dispensed into single use aliquots and
is stable for a year when stored at ≤−20 °C.
4. Prepare a master mix of the components required for the
Click-it reaction. For each tube mix together 438 μl PBS or
TBS, 10 μl CuSO4 (Component F), 2.5 μl fluorescent azide
(prepared in Subheading 3.5, step 1), 50 μl 10× reaction buf-
fer additive (prepared in Subheading 3.5, step 2). Scale up the
mixture for the number of samples to be treated and add
500 μl to each tube. It is important to use the cocktail within
15 min of preparation. It is good practice to include a control
sample of cells not exposed to EdU. In addition, these cells are
needed for single staining compensation controls for intracel-
lular antigens or antigens stained with RPE, PE-tandem, or
Qdot antibody conjugates.
5. Incubate the mixture for 30 min at room temperature in the
dark.
6. Add 3 ml of the Click-iT permeabilization and wash buffer
(prepared in Subheading 3.4, step 1), pellet the cells and
remove the supernatant.
7. Gently dislodge the cell pellet. If intracellular antibody stain-
ing is desired, add 100 μl Click-iT permeabilization and wash
buffer and proceed to Subheading 3.6. Otherwise add 500 μl
of the Click-iT permeabilization and wash buffer and go to
Subheading 3.7 for DNA staining or Subheading 3.8 for flow
cytometry.
3.6 Optional Staining 1. Add predetermined amounts of antibodies to the cells and mix
for Intracellular well. Incubate on ice for 30 min protected from light.
Antigens or with PE, 2. Add 3 ml of the Click-iT permeabilization and wash buffer,
PE-Tandem, or Qdot pellet the cells, and remove the supernatant.
Antibody Conjugates 3. Gently dislodge the cell pellet and add 500 μl of the Click-iT
permeabilization and wash buffer and go to Subheading 3.7
for DNA staining.
Analysis of Cell Proliferation and Homeostasis Using EdU Labeling 217
3.7 Cell Cycle Stain 1. Dilute 5 mg/ml DAPI stock 1:100 with diH2O and add
12.5 μl to each tube (3 μM final concentration) and incubate
for 15 min at room temperature (see Note 12). Ensure that a
tube of fixed and permeabilized cells is stained with only DAPI
for compensation control. After incubation proceed to
Subheading 3.8.
3.8 Flow Cytometry 1. Use unlabeled and single color controls to set up compensa-
Analysis tion on cytometer and run samples. Use “all but one” controls
to set gates if needed (see Note 13).
2. Gate on the cell population of interest. Collect the fluorescent
signal from DAPI or other DNA content dyes using linear
amplification; all other fluorescent signals should be collected
with logarithmic amplification. When measuring cellular DNA
content on most flow cytometers, use a low flow rate (<500
events per second) during data collection. Use the same col-
lection rate for all samples in the experiment (see Note 14).
3. Cell clumping is often observed when processing samples for
flow cytometry. When analyzing DNA content, it is important
to distinguish between G1 doublets from a G2-M single event.
To discriminate the doublet from the singlet, plot the width
(W) versus area (A) for the channel used in a dot plot graph.
A population of single cells will form a diagonal. W increases
with the diameter of the clump, while the A of a G1 doublet
and the G2/M single cell is the same. Therefore, discrimina-
tion of G1 doublets and clumped cells from G2/M single cells
can be made by gating out events that deviate with greater
width from the diagonal as shown in Fig. 1a [10].
Fig. 1 Human epithelial cells were cultured with EdU for 2 h then stained with Pacific Blue azide and 7-aad. (a)
7-aad fluorescence was collected using the FL3 channel and plotted area (FL3-A) versus width (FL3-W) to
allow discrimination between clumped doublets and single cells shown within the rectangular gate. (b) Cells
were gated based on size and the percentage of EdU labeled cells is shown as a histogram. (c) Bivariate analy-
sis of EdU versus 7-aad staining is shown for cells gated as in (a). Numbers shown denote percentages.
Clockwise from the top are gates that show cells in S-phase (59 %), G2/M (13 %) and G0/G1 (20 %)
218 Francis A. Flomerfelt and Ronald E. Gress
4. Using the desired phenotypic gate, the percent cells that syn-
thesized DNA and incorporated EdU during the pulse period
can be determined with a simple histogram (Fig. 1b). Plotting
EdU versus DAPI provides more information about the cell
cycle (Fig. 1c). In this example, human epithelial cells were
pulsed with EdU for 2 h and stained with 7-aad and Pacific
Blue azide. The bivariate analysis shows that 59 % of these cells
synthesized DNA during the pulse and represent cells in
S-phase (Fig. 1c). Twenty percent of the cells were quiescent
and remained in G1 phase while 13 % did not synthesize DNA
and were in G2-M phase (Fig. 1c). A population of stable qui-
escent cells can be identified by lack of EdU incorporation
over a longer labeling period, while a population of cycling
cells will become uniformly labeled.
4 Notes
Acknowledgement
References
1. Darzynkiewicz Z, Traganos F, Zhao H, mature lymphocytes. Eur J Immunol
Halicka HD, Li J (2011) Cytometry of DNA 20:1697–1708
replication and RNA synthesis: Historical per- 7. Cavanagh BL, Walker T, Norazit A, Meedeniya
spective and recent advances based on “click AC (2011) Thymidine analogues for tracking
chemistry”. Cytometry A 79:328–337 DNA synthesis. Molecules 16:7980–7993
2. Dolbeare F, Gratzner H, Pallavicini MG, Gray 8. Salic A, Mitchison TJ (2008) A chemical
JW (1983) Flow cytometric measurement of method for fast and sensitive detection of
total DNA content and incorporated bromo- DNA synthesis in vivo. Proc Natl Acad Sci
deoxyuridine. Proc Natl Acad Sci USA 80: USA 105:2415–2420
5573–5577 9. Buck SB, Bradford J, Gee KR, Agnew BJ,
3. Gray JW, Dolbeare F, Pallavicini MG, Beisker Clarke ST, Salic A (2008) Detection of S-phase
W, Waldman F (1986) Cell cycle analysis using cell cycle progression using 5-ethynyl-2’-de-
flow cytometry. Int J Radiat Biol Relat Stud oxyuridine incorporation with click chemistry,
Phys Chem Med 49:237–255 an alternative to using 5-bromo-2’-deoxyuri-
4. Tough DF, Sprent J (1994) Turnover of naive- dine antibodies. Biotechniques 44:
and memory-phenotype T cells. J Exp Med 927–929
179:1127–1135 10. Nunez R (2001) DNA measurement and cell
5. Parretta E, Cassese G, Santoni A, Guardiola J, cycle analysis by flow cytometry. Curr Issues
Vecchio A, Di Rosa F (2008) Kinetics of in Mol Biol 3:67–70
vivo proliferation and death of memory and 11. Clarke LE, Young KM, Hamilton NB, Li H,
naive CD8 T cells: parameter estimation based Richardson WD, Attwell D (2012) Properties
on 5-bromo-2’-deoxyuridine incorporation in and fate of oligodendrocyte progenitor cells in
spleen, lymph nodes, and bone marrow. the corpus callosum, motor cortex, and piri-
J Immunol 180:7230–7239. form cortex of the mouse. J Neurosci 32:
6. Rocha B, Penit C, Baron C, Vasseur F, 8173–8185
Dautigny N, Freitas AA (1990) Accumulation 12. Tough DF, Sprent J, Stephens GL (2007)
of bromodeoxyuridine-labeled cells in central Measurement of T and B cell turnover with
and peripheral lymphoid organs: minimal esti- bromodeoxyuridine. Curr Protoc Immunol
mates of production and turnover rates of Chapter 4:Unit 4 7
Chapter 19
Abstract
During their development, human T cells undergo similar genomic changes and pass through the same
developmental checkpoints as developing thymocytes in the mouse. The difference between both species,
however, is that some of these developmental stages are characterized by different phenotypic markers and
as a result, evidence emerges that the molecular regulation of human T cell development subtly differs
from the mouse [1–4]. In this chapter, we describe in detail how the different stages of human T cell
development can be characterized and isolated using specific surface markers.
Key words Human, T cell development, Thymocytes, Isolation of T cell progenitors, Fluorescent
activated cell sorting, Magnetic activated cell sorting
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_19, © Springer Science+Business Media New York 2016
221
222 Inge Van de Walle et al.
CD56 +
NKT CD3 +
TCR-αβ+
Bone Thymus
marrow
Foxp3 +
CD25 +
Treg
CD4 +
Commitment β-selection +/- selection CD3 +
TCR-αβ+
CD1a+ CD1a-
CD4 + CD8 CD3 +
CD34 + CD34 + CD34 +/- CD1a+ CD1a+ TCR-αβ+
CD8αβ +/- SP
CD10 + CD7 + CD1a+ CD4 + CD4 + CD3 + CD45RA +
CD7 + CD1a + CD4+ CD8αα+/- CD8αβ+/- TCR-αβ+ CD45RO -
CD1a- CD4 - CD8αα+/- CD28+ CD3 - CD69 -
CD1a-
CD4
HPC T-S T-C ISP4 β DP αβ CD3 +
SP TCR-αβ+
CD45RA +
CD45RO -
CD45RA +
CD38 - CD3 +
CD34 + TCR-γδ+
HPC γδ γδ
CD10 + CD1 -
CD7 -/low
CD34 + CD3 +
CD10 + TCR-γδ+
CD7 -/low TCR rearrangements CD1 +
δ α
γ
β
Fig. 1 Schematic overview of the different developmental stages that characterize human T cell development.
HSC hematopoietic stem cell, T-S T-lineage specified progenitor, T-C T cell committed progenitor, ISP4 CD4
immature single positive, β rearranged T cell receptor-β chain, DP double positive, αβ T cell receptor-αβ
positive cell, NKT natural killer T cell, Treg regulatory T cell, CD8 SP CD8 single positive, CD4 SP CD4 single
positive, γδ T cell receptor-γδ positive cell, CD cluster of differentiation
2 Materials
Table 1
Overview of monoclonal antibodies to sort different T cell subsets
3 Methods
3.1 Human 1. Transfer thymus tissue, obtained from cardiac surgeries (see
Thymocyte Cell Note 5), to a 140 mm petri dish with 25 ml IMDM supple-
Suspension mented with penicillin (100 U/ml), streptomycin (100 μg/
ml), L-glutamine (2 mM) and 10 % heat-inactivated FBS. Mince
thymus tissue into 4–5 mm pieces with a sterile scalpel.
2. Gently smash thymus fragments with the end of a plunger
from a 10 ml syringe to release the thymocytes. The medium
will become turbid.
226 Inge Van de Walle et al.
3.2 Purification 1. Dilute cord blood 1:3 with PBS. Dilute bone marrow 1:5 with
of Viable Cells by PBS (see Note 8).
Lymphoprep Density 2. Transfer maximum 30 ml of diluted cord blood or thymus
Gradient suspension in a 50 ml polypropylene conical tube. Gently pipet
15 ml Lymphoprep™ below diluted cord blood or thymus
suspension. For bone marrow, 15 ml Lymphoprep™ is trans-
ferred in a 50 ml polypropylene conical tube. Gently pipet
30 ml of diluted bone marrow on top of the Lymphoprep™.
3. Centrifuge at 940 × g for 20 min at room temperature (accel-
eration and brake at 30 % of maximum).
4. Aspirate the interface (Fig. 7), containing mononuclear cells
from cord blood sample, bone marrow (see Note 9) or thymus
suspension, using a Pasteur pipette (see Note 10).
5. Pool all interface samples from cord blood, bone marrow or
thymus suspension in a 50 ml polypropylene conical tube and
wash twice in 50 ml cold PBS for cord blood and bone mar-
row, or cold DPBS for thymus suspension (centrifuge at
400 × g for 10 min at 4 °C).
6. Resuspend cell pellet in 50 ml MACS buffer and determine
cell number using Turk’s solution.
7. Centrifuge at 400 × g for 10 min at 4 °C and remove
supernatant.
8. The mononuclear cell fraction is now ready for further purifi-
cation of specific subsets. Optional: cells can be frozen and
stored in liquid nitrogen at this stage for later use (see Note 6).
Characterization and Isolation of Human T Cell Progenitors 227
3.3 Isolation Extrathymic CD34 positive progenitor cells can be isolated from
of Extrathymic CD34 the mononuclear fraction obtained after Lymphoprep density gra-
Positive Cells Using dient of bone marrow or cord blood.
Magnetic Activated 1. Determine cell number (see Note 11), centrifuge cell suspen-
Cell Sorting (MACS) sion at 500 × g for 10 min, aspirate supernatant completely.
2. Resuspend cell pellet in 300 μl MACS buffer for up to 108
total cells, add 100 μl of FcR blocking Reagent, add 100 μl of
CD34 MicroBeads (see Note 12), mix well and incubate for
30 min in the refrigerator (2–8 °C, see Note 13).
3. Wash labeled cells twice by adding 15 ml MACS buffer and
centrifuge at 500 × g for 10 min, aspirate supernatant.
4. Resuspend cell pellet in 3 ml MACS buffer and proceed to
magnetic separation.
5. Place the LS separation column in the magnetic field of the
MACS Separator, prepare LS column by rinsing three times
with 3 ml MACS buffer.
6. Apply cell suspension onto the column (see Note 14), discard
flow-through containing unlabeled cells. Wash the column
three times with 3 ml MACS buffer; perform washing steps by
adding buffer aliquots only when the column reservoir is
empty.
7. Remove the column from the separator and place it in a 15 ml
polypropylene conical tube. Pipette 3 ml MACS buffer onto
the column and immediately flush out the magnetically labeled
cells by firmly pushing the supplied plunger into the column.
Optional: Cells can be passed on a second LS column if a
higher purity is desired without the need for cell sorting as
described below. Repeat steps 4–7.
8. Count the cells and check CD34+ purity on 10,000 cells by
adding a fluorescent labeled anti-human CD34 monoclonal
antibody.
9. Incubate for 30 min at 2–8 °C.
10. Wash cells by adding 2 ml of human wash buffer, centrifuge at
500 × g for 6 min, discard supernatant by decanting 5 ml tube.
11. Run sample on a flow cytometer (Fig. 2).
12. Purity of CD34+ cells obtained from cord blood is between 60
and 80 %.
Optional: the CD34+ enriched cell fraction can be frozen and
stored in liquid nitrogen (see Note 6).
3.4 Purification of Enriched CD34+ cells, either freshly isolated or frozen (see Chapter
CD34+ Precursors by 20 for thawing cells), are preferentially further purified to >99 %
Fluorescence-Activated purity by cell sorting to avoid outgrowth of contaminating mature
Cell Sorting (FACS) T cells.
228 Inge Van de Walle et al.
a b c
Fig. 2 Gating strategy to sort extrathymic CD34+Lin− progenitor cells derived from cord blood. Within the lym-
phocyte gate (a), a viable (b) population of CD34+ lin− progenitor cells can be gated (c). These cells are negative
for the monocyte marker CD14, the B cell marker CD19, the NK cell marker CD56 and the T cell marker CD3
a b
Fig. 3 Gating strategy to sort CD34+ thymocytes. CD4 expression is used to dis-
criminate between the CD34+CD4− cells and the more differentiated CD34+CD4+
that immediately precedes the immature single positive cells (a). In the
CD34+CD4− population two populations can be discriminated based on the
expression of CD1 (b). Upregulation of CD1 correlates with commitment to the
T-cell lineage in postnatal human thymocytes
230 Inge Van de Walle et al.
a b
Fig. 4 Gating strategy to sort CD4 immature single positive thymocytes (4ISP)
after CD3 and CD8 depletion of total thymocytes. Two populations can be dis-
criminated in the CD4+CD3−CD8−CD34− cell fraction based on the expression of
CD28. The differential expression of CD28 discriminates between pre- and post
(CD28+) β-selected cells
3.7 Isolation 1. Due to their limited life span, CD4+CD8+ DP cells need to be
of Double Positive (DP) isolated directly from freshly prepared thymus suspension.
and Single Positive Both CD3− and CD3+ thymocytes can be isolated. Using the
(SP) Thymocytes same labeling, mature SP CD4+CD3+ and CD8+CD3+ thymo-
cytes can be sorted at the same time, if the available cell sorter
has the capacity to sort 4 populations simultaneously.
2. Put the desired cell number (see Note 21) into a 15 ml poly-
propylene conical tube, centrifuge at 500 × g for 6 min.
3. Resuspend in 100 μl MACS buffer and stain cells for CD4,
CD8, and CD3 for 30–45 min at 2–8 °C. Use the desired
antibodies in appropriate concentrations as recommended by
the manufacturers (see Note 20).
4. Follow steps 14–18 from Subheading 3.4 but pay attention
to Note 22.
5. See Fig. 5 for representative FACS plots.
232 Inge Van de Walle et al.
a b
c d
Fig. 5 Gating strategy to sort DP and SP thymocytes. CD4+CD8+ double positive cells as shown in (a) can be
separated in CD3− and CD3+ subsets as shown in panel (b). Single positive cells CD4+CD8− and CD8+CD4−
thymocytes (a) express high levels of CD3 (c, d) that can be incorporated as a marker to sort for CD4 and CD8
SP cells
3.8 Isolation of TCR 1. Freshly prepared thymocytes (see Subheading 3.1) are used as
γδ Cells starting material.
2. Determine the cell number (see Note 23), centrifuge the cells
at 500 × g for 6 min and aspirate the supernatant.
3. Resuspend the cell pellet in 20 μl MACS buffer per 107 total
cells (see Note 24).
4. Label thymocytes with anti-TCR γδ haptenated (see Note 3)
antibodies (10 μl per 107 cells). The amount of antibody added
is based on 1:4 of the total cell number (see Note 25).
5. Mix well and incubate for 10 min at 4–8 °C.
6. Add 15 μl MACS buffer per 107 total cells and add MACS
Anti-Hapten MicroBeads-FITC (20 μl per 107 cells) based on
1:4 of the total cell number.
7. Mix well and incubate for 15 min at 4–8 °C.
8. Proceed with MACS protocol (see Subheading 3.3, steps 3–7).
Characterization and Isolation of Human T Cell Progenitors 233
a b
Fig. 6 Gating strategy to sort immature and mature TCR-γδ thymocytes. Two
populations can be obtained in the TCR-γδ population based on the expression
of CD1. Downregulation of CD1 correlates with a more mature phenotype
4 Notes
/
dead cell fraction
Fig. 7 Lymphoprep density gradient. The different fractions are shown following
centrifugation
References
1. Taghon T, Waegemans E, Van de Walle I 7. Dik WA, Pike-Overzet K, Weerkamp F et al
(2012) Notch signaling during human T cell (2005) New insights on human T cell develop-
development. Curr Top Micobiol Immunol ment by quantitative T cell receptor gene rear-
360:75–97 rangement studies and gene expression
2. Haddad R, Guimiot F, Sic E et al (2006) profiling. J Exp Med 201:1715–1723
Dynamics of thymus-colonizing cells dur- 8. Carrasco YR, Trigueros C, Ramiro AR et al
ing human development. Immunity 24: (1999) Beta-selection is associated with the onset
217–230 of CD8beta chain expression on CD4(+)
3. Hao QL, George AA, Zhu J et al (2008) CD8alphaalpha(+) pre-T cells during human
Human intrathymic lineage commitment is intrathymic development. Blood 94:3491–3498
marked by differential CD7 expression: identi- 9. Joachims ML, Chain JL, Hooker SW et al
fication of CD7- lympho-myeloid thymic pro- (2006) Human alpha beta and gamma delta
genitors. Blood 111:1318–1326 thymocyte development: TCR gene rearrange-
4. Taghon T, Rothenberg EV (2008) Molecular ments, intracellular TCR beta expression, and
mechanisms that control mouse and human gamma delta developmental potential–differ-
TCR-αβ and TCR-γδ T cell development. ences between men and mice. J Immunol
Semin Immunopathol 30:383–398 176:1543–1552
5. Weerkamp F, Baert MR, Brugman MH et al 10. Taghon T, Van de Walle I, De Smet G et al
(2006) Human thymus contains multipotent (2009) Notch signaling is required for prolifera-
progenitors with T/B lymphoid, myeloid, tion but not for differentiation at a well-defined
and erythroid lineage potential. Blood 107: beta-selection checkpoint during human T-cell
3131–3137 development. Blood 113:3254–3263
6. Ratajczak MZ (2008) Phenotypic and func- 11. Plum J, De Smedt M, Leclercq G et al (2008)
tional characterization of hematopoietic stem Human intrathymic development: a selective
cells. Curr Opin Hematol 15:293–300 approach. Semin Immunopathol 30:411–423
Chapter 20
Abstract
Not only is human T cell development characterized by unique changes in surface marker expression, but
it also requires specific growth factors and conditions to mimic and study T cell development in vitro. In
this chapter, we provide an overview of the specific aspects that need attention when performing T cell
differentiation cultures with human progenitors.
Key words Human, T cell development, OP9 coculture, Viral transduction, Gene perturbation,
In vitro, Hematopoietic stem cell, Lineage differentiation
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_20, © Springer Science+Business Media New York 2016
239
240 Anne-Catherine Dolens et al.
2 Materials
2.5 Analysis of OP9 1. Flow cytometer capable of multi-color analysis (e.g., FACS
Coculture Experiments LSRII).
2. Anti-human FcR Blocking Reagent.
242 Anne-Catherine Dolens et al.
3 Methods
2. Passage the OP9 stromal cells that are not being used for
cocultures each time when the confluence reaches about
80–95 %. Do not let them reach confluency! To split OP9 cells:
(a) Carefully aspirate the medium from the cell culture dish
and wash the monolayer with 5 ml PBS to wash out the
serum.
(b) Add 2 ml 0.25 % Trypsin–EDTA, incubate for 5 min at
37 °C.
(c) Verify that all OP9 stromal cells have detached. If not,
incubate for another 5 min at 37 °C.
(d) Neutralize hydrolysis reaction by adding 4 ml of OP9
medium and transfer the cells from the culture dish into a
15 ml polypropylene conical tube.
(e) Spin at 500 × g for 6 min at 4 °C and aspirate
supernatant.
(f) Resuspend cells in 1 ml OP9 medium and determine cell
number with Trypan Blue.
3. Maintain OP9 stromal cells by seeding 200 × 103–250 × 103
cells per 100 mm × 20 mm cell culture dish in a total volume
of 10 ml OP9 medium. Split OP9 cells every 2–3 days.
4. For coculture experiments, prepare a single layer of OP9 stro-
mal cells 24 or 48 h before initiating the coculture. Therefore,
plate 20 × 103 or 10 × 103 cells, respectively, in a total volume
of 500 μl OP9 medium in a 24-well flat bottom tissue culture
plate (see Note 10).
3.4 Initiation 1. Verify using a microscope that the OP9 cells have generated a
and Maintenance confluent layer of cells and remove medium from the wells.
of OP9 Coculture 2. Resuspend the purified progenitor cells in the appropriate vol-
Experiments ume of “OP9 coculture medium” (see Note 11), supplemented
with the required cytokines. The medium volume depends on
the number of progenitor cells that will be used to initiate the
culture (see Table 1). Supplemented cytokines depend on the
hematopoietic lineage being studied (see Table 2).
In vitro T Cell Development 245
Table 1
Maximum number of cells from different progenitor populations for
initiation of cocultures onto different well types
Table 2
Different cytokine cocktails for the generation of different cell types
3.5 Analysis of OP9 1. Before labeling, centrifuge the cells for 6 min at 500 × g.
Coculture Experiments 2. Discard the supernatant.
3. Resuspend the cells in 100 μl human wash buffer and pre-
incubate on ice for 5 min with 1 μl anti-human FcR Blocking
Reagent and 2 μl anti-mouse FcRγII/III 20 μg/ml (clone
2.4.G2) to avoid nonspecific binding.
Fig. 1 Kinetic analysis of human T cell development from CD34+Lin− cord blood progenitors on OP9-DLL4low
vs. OP9-DLL4high cells. Dot plots also show effects of low (OP9-DLL4low) versus high (OP9-DLL4high) levels
of Notch activation on human T cell development as discussed in more detail elsewhere [2]. Early specification
towards the T cell lineage is favored by high Notch activity and can be tracked by the upregulation of CD7 while
CD34+ expression is maintained. Further differentiation is characterized by the upregulation of CD5 and down-
regulation of CD34. Addition of HLA-DR to the panel is important to discriminate T cell progenitors from mono-
cytic/dendritic cells. While CD34+ cells coexpress HLA-DR, also following upregulation of CD7 (green cells),
CD7+CD5+ T cell progenitors will downregulate CD34+ as well as HLA-DR (blue cells), while CD5+ cells also
contain HLA-DR+CD4+ monocytic/dendritic cells (red cells). Thus, CD5 or CD7 are insufficient as sole markers
for early T cell progenitors. Further differentiation into CD4+CD8β+ double positive thymocytes and TCR-αβ+
CD3+ T cells is more efficient when Notch activity is reduced (OP9-DLL4low) but requires over 50 days
of coculture to develop. TCR-γδ T cells preferentially develop in conditions of high Notch signaling activity
(OP9-DLL4high) (color figure online)
In vitro T Cell Development 247
Table 3
Antibody panels to study different early stage of human T cell development in vitro (see Note 14)
Early T cells Double positive cells TCRαβ/TCRγδ T cells Myeloid cells B cells
PI PI PI PI PI
CD45 CD45 CD45 CD45 CD45
(HI30) (HI30) (HI30) (HI30) (HI30)
CD34 CD8b TCRαβ CD4 CD19
(AC136) (2ST8.5H7) (BW242/412) (RPA-T4) (LT19)
CD7 CD8 TCRγδ HLA-DR HLA-DR
(M-T701) (SK1) (11F2) (LN3) (LN3)
CD5 CD4 CD3 CD14
(UCHT2) (RPA-T4) (UCHT1) (TUK4)
CD1a CD3 CD4 CD15
(HI149) (UCHT1) (RPA-T4) (VIMC6)
CD4 HLA-DR CD8b CD123
(RPA-T4) (LN3) (2ST8.5H7) (6H6)
HLA-DR CD1a CD303
(LN3) (HI149) (AC144)
Antibody clones are indicated in italicized parenthesis
4 Notes
References
1. Weinacht KG, Brauer PM, Felgentreff K et al control human TCR-αβ/γδ development by
(2012) The role of induced pluripotent inducing differential Notch signal strength. J Exp
stem cells in research and therapy of primary Med 210:683–697
immunodeficiencies. Curr Opin Immunol 4. Verhasselt B, Naessens E, Stove V (2003)
24:617–624 Generation of transgenic T cells from human
2. Taghon T, Waegemans E, Van de Walle I CD34+ cord blood cells. Methods Mol Biol
(2012) Notch signaling during human T cell 215:327–339
development. Curr Top Micobiol Immunol 5. Van de Walle I, De Smet G, Gärtner M et al
360:75–97 (2011) Jagged2 acts as a Delta-like Notch
3. Van de Walle I, Waegemans E, De Medts J et al ligand during early hematopoietic cell fate deci-
(2013) Specific Notch receptor-ligand interactions sions. Blood 117:4449–4459
Chapter 21
Abstract
While in vitro models exist to study human T cell development, they still lack the precise environmental
stimuli, such as the exact combination and levels of cytokines and chemokines, that are present in vivo.
Moreover, studying the homing of hematopoietic stem (HSC) and progenitor (HPC) cells to the thymus
can only be done using in vivo models. Although species-specific differences exist, “humanized” models
are generated to circumvent these issues.
In this chapter, we focus on the humanized mouse models that can be used to study early T cell devel-
opment. Models that study solely mature T cells, such as the SCID-PBL (Tary-Lehmann et al., Immunol
Today 16:529–533) are therefore not discussed here, but have recently been reviewed (Shultz et al., Nat
Rev Immunol 12:786–798).
Key words Immunodeficient mice, Humanized mice, In vivo mouse model, T cell development, BLT
mouse model, SCID-Hu mouse model
1 Introduction
1.1 Mouse Strains Engraftment of human hematopoietic cells and fetal hematopoietic
Used to Study In Vivo tissues was initially described in severe combined immunodeficient
T Cell Development (SCID) mice [1–3]. Due to the Prkdcscid (protein kinase, DNA
activated, catalytic polypeptide) mutation, these mice lack func-
tional T- and B-lymphocytes, allowing higher and more diverse
engraftment than was described earlier in nude and beige-nude-
Xid (bnx) mice, which support T cell but not B cell development
[4–6]. However, engraftment of human cells in SCID mice was
still suboptimal, owing to high levels of host natural killer (NK)
cell activity and innate immunity. Another way to delete T and B
cells in the murine host, is to target one of the recombination acti-
vating gene 1 and 2 (Rag1 and Rag2) genes, crucial for rearrange-
ments of both T and B cell receptor genes during T and B cell
development [7, 8]. However, these mice also retain high NK cell
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_21, © Springer Science+Business Media New York 2016
253
254 Sarah Bonte et al.
activity that leads to low human cell engraftment. The scid mutation,
as compared to the Rag mutation, also results in defective DNA
repair (as the Prkdc gene encodes for a polypeptide important in
repair of double-stranded DNA breaks) and, consequently, higher
radiosensitivity [9].
The development of nonobese diabetic (NOD)-scid mice, by
crossing the scid mutation onto the NOD background, led to the
next breakthrough in the development of immunodeficient mouse
models. Additional defects in innate immunity in NOD-scid mice,
lacking hemolytic complement and showing functional defects
in both macrophages and NK cells, led to higher engraftment
of human hematopoietic cells, compared to scid mice [10–12].
However, the residual activity of both NK cells and macrophages
in NOD-scid mice still limited human hematopoietic engraftment,
and especially T cell development was very inefficient. To address
this problem, existing models were adapted, e.g., by injecting mice
with TM-β1, an IL-2 receptor beta chain (IL2Rβ) blocking anti-
body [13–15], further decreasing NK cell activity in these mice,
allowing the engraftment of T cell precursors in the murine thymus
with in vivo T cell development in about 50 % of the cases. The
major drawback of this model was that large amounts of antibody
and several intraperitoneal injections of the mice were necessary.
Later, new mouse strains lacking a functional IL-2 receptor gamma
chain (Il2rgnull) were developed such as NOD/Shi-scid Il2rgnull
(NOG) [16], NOD/LtSz-scid Il2rgnull (NSG) [17], BALB/c--
Rag2null Il2rgnull (BRG) [18], NOD/Rag1null Il2rgnull (NRG) [19].
Since the IL2Rγ chain is an important component of the receptors
for IL-2, IL-4, IL-7, IL-9, IL-15, and IL-21, these mice com-
pletely lack NK-cell development, as well as functional T- and
B-lymphocytes, features that lead to superior human hematopoi-
etic engraftment and allow for T cell development in the murine
thymus in a near to 100 % frequency. An important disadvantage
of the NOD-scid strain is the relatively short life span of these mice
due to thymoma development [20], thereby not allowing evalua-
tion of the injected and/or in vivo generated cells for an extended
period of time. Il2rgnull mice, however, have a longer median life
span (more than 90 weeks for NSG mice compared with 37 weeks
for NOD-scid mice) and thymoma development is very rare to
nonexistent [21]. Also in mice lacking beta2-microglobulin
(B2m−/−) (NOD/SCID/B2m−/−) [22] B, T, and NK cell develop-
ment and functions are disrupted, as β2m is necessary for major
histocompatibility complex (MHC) class I-mediated immunity.
These and other immunodeficient mouse strains are extensively
reviewed in ref. 21.
The thymic microenvironment and especially the thymic epi-
thelial cells support positive and negative selection processes through
interaction of MHC molecules on the epithelial cells with the TCR
Humanized Mice to Study Human T Cell Development 255
(a) Basic
T cell HSC
Mouse strain Alternative name Abbreviation Background Genetic defect development engraftment Refs
NODShi.Cg-PrkdcscidIl2rgtm1Sug NOD/Shi-scid Il2rgnull NOG NOD/SCID IL-2Rγ chain lacks +++ ++ [16]
IC domain
NOD.Cg-PrkdcscidIl2rgtm1Wjl NOD/LtSz-scid Il2rgnull NSG NOD/SCID Null mutation of +++ +++ [17]
IL-2Rγ
NOD.Cg-Rag1tmMomIl2rgtmWjl NOD/Rag1null Il2rgnull NRG NOD Null mutation of +++ +++ [19]
Rag1 and IL-2Rγ
C.Cg-Rag2tmFwaIl2rgtmSug BALB/c-Rag2nullIl2rgnull BRG Balb/c Null mutation of ++ ++ [18]
Rag2 and IL-2Rγ
chain lacks IC
domain
t
C.Cg-Rag1tm1MomIl2rg mWjl BALB/c-Rag1nullIl2rgnull BRG Balb/c Null mutation of ++ ++ [19]
Rag1 and IL-2Rγ
chain lacks IC
domain
C;129S4-Rag2tmFlvIl2rgtm1Flv/J / BRG or Mixed (Balb/c x (Null mutation of ++ ++ [29]
C;129RG 129Rag2null) Rag2) Null
mutation of
IL-2Rγ chain
(b) Supplementally engineered mice to enhance T cell development
Supplemental
Background Additional genetic effects on T cell
Mouse strain Abbreviations (see ref. 30) defect development Refs
NOD.Cg-PrkdcscidIl2rgtm1WjlTg(HLA-A/H2-D/B2M)1Dvs NSG Tg(HLA-A2/ NSG Transgene HLA-A2 Supports [31]
B2M) and human development of [32]
NSG-A2/B2M β2-microglobulin human HLA-A2
restricted T cells
NOD.Cg-PrkdcscidIl2rgtm1WjlTg(HLA-A2.1)1Enge NSG Tg(HLA-A2) NSG Transgene HLA-A2 Supports [23]
NSG-A2 development of
human HLA-A2
restricted T cells
NOD.Cg-Rag1tm1MomIL2rgtmWjlTg(HLA-DRB1)31Dmz NSG Tg NSG Transgene Supports [33]
(HLA-DR4) HLA-DR4 development of
NSG-DR4 human HLA-DR
restricted T cells,
enhanced
immune function
NOD.Cg-PrkdcscidH2-Ab1tm1DolIl2rgtm1SugTg(HLA-DRA, NOG NOG Transgene Supports [24]
HLA-DRB1*0405)1Jic/Jic Tg(HLA-DR4) HLA-DR4 development of
NOG-DR4 human HLA-DR
restricted T cells,
enhanced
immune function
NOD.Cg-PrkdcscidIl2rgtm1Wjl/Sz Tg(HLA-A2*0201/ NSG Tg(HLA-A2/ NSG Transgene HLA-A2 Supports [25]
HLA-DRA*0101, HLA-DRB1*0101) DR1) and HLA-DR1 development of
NSG-A2/DR1 human HLA-A2
and HLA-DR
restricted T cells,
enhanced
immune function
Abbreviation: IC intracytoplasmic; nc not compared
Adapted from [26]
258 Sarah Bonte et al.
BLT SCID-Hu
under kidney
intravenous
capsule
femur liver
kidney
Hu-HSC
Fig. 1 Schematic overview of humanized mouse models
doses up to 400 cGy [17] but mostly 200–300 cGy is used [46–49].
For adult BRG mice, radiation doses of 500–600 cGy are used
[50]. The only mouse strain that seems to overcome the need for
irradiation, and therefore might provide a more physiological envi-
ronment, is the newborn NSG mouse expressing membrane-bound
SCF [27] and the adult Rag2−/−Il2rg−/−KitWv/Wv mouse [51], in
which the mutant Kit receptor opens up the stem cell niches across
species barriers [52]. As human lymphoid development in the
Rag2−/−Il2rg−/−KitWv/Wv mouse has not yet been published, this
mouse strain is not discussed above.
An alternative method that substitutes irradiation is injection
of the mice with the chemotherapeutic agent Busulfan. Choi et al.
[53] showed that this leads to a higher survival rate and enhanced
T cell development.
Other engraftment-enabling methods that have previously
been used are injection of the mice with TM-β1, before injection
of human cells [13], a strategy which has been made redundant by
the newer strains, lacking IL2Rγ or β2m, and in vivo injection of
various recombinant human cytokines, such as IL-15, IL-7, and
SCF, after injection of human cells [54–56].
Also, intrafemoral injection of HSC can be considered as a
method to enable engraftment, as injection of HSC directly into
the bone marrow circumvents the need for homing of these cells to
the bone marrow and therefore allows for immediate engraftment.
2 Materials
2.2 CD34+ Isolation 1. Hematopoietic stem cell (HSC) source (umbilical cord blood,
mobilized peripheral blood, bone marrow, or fetal liver)
(see Note 3).
2. Lymphoprep or Ficoll, stored at room temperature.
3. Method to isolate CD34+ cells (see Note 4).
2.7 Subcapsular 1. SCID mice (preferably male, 6–8 weeks old) or NOD.
Kidney Transplant Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NOD/LtSz-scid Il2rg−/−) mice
(male or female, 6–10 weeks old) (see Note 5).
2. Human fetal liver and fetal thymus of 14–23 weeks of gesta-
tional age (preferably HLA-typed, see Note 7) (see Note 8).
3. Sterile PBS supplemented with 50 μg/ml gentamicin (Gibco,
Invitrogen, Merelbeke, Belgium).
4. Sterile RPMI-1640 medium (Gibco, Invitrogen, Merelbeke,
Belgium) supplemented with 50 μg/ml gentamicin.
5. Anesthetic and/or anesthesia equipment (according to institu-
tional guidelines).
6. Special injection device/transplantation needle (see Note 9).
7. Surgical instruments: scalpel, microdissecting scissors, Dumont
tweezers, microdissecting forceps, microdissecting scissors,
wound closure staple and clip and/or absorbable suture, nee-
dle holder and suture scissors, scissors and toothed tweezers
(see Note 6).
Humanized Mice to Study Human T Cell Development 263
3 Methods
3.1 CD34+ Isolation 1. Isolate mononuclear cells from HSC source using Lymphoprep
or Ficoll density gradient centrifugation (see Chapter 19 by
K. Davids et al. for details).
2. From the obtained mononuclear cells, isolate CD34+ cells
(see Note 4 and Chapter 19 by K. Davids et al.).
3. Resuspend the isolated CD34+ cells in sterile PBS at the desired
cell density (volumes and cell numbers typically used are discussed
in Note 10) and keep on ice until transplanted (see Note 11).
3.2 Mice Irradiation 1. Place mice in sterile autoclaved housing device for irradiation
(see Notes 12 and 13 for additional information on handling
newborn mice).
2. Irradiate mice with the appropriate amount of total-body irra-
diation (see Notes 1 and 14).
3.3 Intrahepatic 1. Monitor breeding pairs for the birth of new litters (this can be
Injection in Newborn estimated using timed matings). The procedure should be per-
Mice formed on 1- (not earlier to avoid rejection by the mother) to
4- (not later as engraftment efficiency will decrease) day old
newborn pups.
2. Isolate CD34+ as described in Subheading 3.1.
3. Irradiate pups as described in Subheading 3.2. Pups are placed
back with their mother afterwards.
4. Wait for at least 2 h and not more than 24 h before starting
with injection procedure.
5. Place irradiated pups on a sterile gauze pad in a sterile petri
dish on ice for 5–10 min until anesthetized (when gross move-
ment ceases).
6. Load the CD34+ cell suspension into a 1-cc tuberculin syringe
with 25-G needle or a 1-cc insulin syringe with 29-G needle.
7. Swab abdomen of mice with sterile alcohol swab.
264 Sarah Bonte et al.
NSG mice
RAG mice
Fig. 2 T cell reconstitution after intrahepatic injection of cord blood CD34+ cells in newborn NSG and Rag −/−
mice at 12 weeks after injection
3.6 Fetal Thymus 1. Place the fetal thymus in a petri dish and rinse twice with PBS/
Preparation gentamicin.
for Subcapsular 2. Transfer the thymus to a petri dish filled with cold RPMI
Kidney Transplant medium.
3. Remove the connective tissue from the thymus.
4. Cut the thymus into small pieces (1 × 1 × 1 mm) using micro-
dissecting scissors (see Note 21). Keep on ice.
3.7 Fetal Liver 1. Place the fetal liver in a petri dish and rinse twice with PBS/
Preparation gentamicin.
for Subcapsular 2. Transfer the liver to a petri dish filled with cold RPMI medium.
Kidney Transplant
3. Cut the liver into small pieces (1 × 1 × 1 mm) using microdis-
secting scissors (see Note 21). Keep on ice.
4. Optional (BLT model): Digest small tissue pieces using RPMI-
1640 supplemented with collagenase and DNase I at 37 °C for
1 h. Gently disrupt the tissues by mixing every 15 min. Filter
the cell suspension through a sterile 70 μm cell strainer. Isolate
CD34+ cells as stated above. If for the experiment cells arising
from injected FL CD34+ and transplanted FL pieces need to be
discriminated, CD34+ cells can be labeled (see Note 22).
4 Notes
13. Pups should have milk in their stomach (seen as a white spot
under the relatively transparent skin) before irradiation proce-
dures can be started.
14. Depending on the immunodeficient mice strain used, a differ-
ent amount of irradiation should be given (see Subheading 1.3).
Newborn NSG or NOG mice are typically irradiated with
100 cGy, newborn BRG mice with 400 cGy.
15. The liver can be seen as a dark spot under the relatively trans-
parent skin of the newborn pup and is situated above the white
stomach (filled with milk).
16. Mice are best restrained by one investigator with one hand at
their scruff and another holding hind legs and tale, and injected
by another investigator. When performing the injection with-
out a second investigator, mice can be held with two fingers
(thumb and forefinger) at the scruff and little finger at the tail
and injecting can be performed using the other hand.
17. Make sure not to overheat mice, the temperature should not
exceed 30–32 °C (85–90 °F).
18. The topical nasal decongestant applied to the snout of the mother
masks the foreign odors left by the investigators and improves
the chance that the mother will accept her pups back into the
cage after completion of the procedure (see also Note 12).
19. Staples are preferred as they are less easily removed by the
mice.
20. Mice should be monitored regularly for wound healing and
possible signs of distress. Wound staples should be removed
after 7–10 days.
21. The diameter of the fetal thymus at 14–23 weeks of gestational
age is typically 1–1.5 cm. The length of the fetal liver at 14–23
weeks of gestational age is typically 15–35 mm.
22. CD34+ cells can be labeled with an intracellular dye (e.g.,
CFSE, PKH, or cell trace) or by transduction with a fluores-
cent marker (e.g., EGFP), depending on the experiment.
References
1. Lapidot T, Pflumio F, Doedens M et al (1992) 3. Vormoor J, Lapidot T, Pflumio F et al (1994)
Cytokine stimulation of multilineage hemato- SCID mice as an in vivo model of human cord
poiesis from immature human cells engrafted blood hematopoiesis. Blood Cells 20:316–320
in SCID mice. Science 255:1137–1141 4. Kamel-Reid S, Dick JE (1988) Engraftment of
2. Vormoor J, Lapidot T, Pflumio F et al (1994) immune-deficient mice with human hemato-
Immature human cord blood progenitors poietic stem cells. Science 242:1706–1709
engraft and proliferate to high levels in severe 5. Dao MA, Nolta JA (1998) Use of the bnx/hu
combined immunodeficient mice. Blood xenograft model of human hematopoiesis to
83:2489–2497 optimize methods for retroviral-mediated
270 Sarah Bonte et al.
stem cell transduction. Int J Mol Med 93: engrafted with mobilized human hematopoietic
2414–2419 stem cells. J Immunol 174:6477–6489
6. Nolta JA, Dao MA, Wells S et al (1996) 18. Traggiai E, Chicha L, Mazzucchelli L et al
Transduction of pluripotent human hemato- (2004) Development of a human adaptive
poietic stem cells demonstrated by clonal anal- immune system in cord blood cell-transplanted
ysis after engraftment in immune-deficient mice. Science 304:104–107
mice. PNAS 93:2414–2419 19. Brehm MA, Cuthbert A, Yang C et al (2010)
7. Mombaerts P, Iacomini J, Johnson RS et al Parameters for establishing humanized mouse
(1992) RAG-1-deficient mice have no mature models to study human immunity: analysis of
B and T lymphocytes. Cell 68:869–877 human hematopoietic stem cell engraftment in
8. Shinkai Y, Rathbun G, Lam KP et al (1992) three immunodeficient strains of mice bearing
RAG-2-deficient mice lack mature lympho- the IL2rgamma(null) mutation. Clin Immunol
cytes owing to inability to initiate V(D)J rear- 135:84–98
rangement. Cell 68:855–867 20. Cashman JD, Eaves CJ (1999) Human growth
9. Fulop GM, Philips RA (1990) The scid muta- factor-enhanced regeneration of transplantable
tion in mice causes a general defect in DNA human hematopoietic stem cells in nonobese
repair. Nature 347:479–482 diabetic/severe combined immunodeficient
10. Conneally E, Cashman J, Petzer A et al (1997) mice. Blood 93:481–487
Expansion in vitro of transplantable human 21. Shultz LD, Ishikawa F, Greiner DL (2007)
cord blood stem cells demonstrated using Humanized mice in translational biomedical
a quantitative assay of their lympho-myeloid research. Nat Rev Immunol 7:118–130
repopulating activity in nonobese diabetic- 22. Christianson SW, Greiner DL, Hesselton RA
scid/scid mice. PNAS 94:9836–9841 et al (1997) Enhanced human CD4+ cell
11. Larochelle A, Vormoor J, Hanenberg H et al engraftment in beta2-microglobulin-deficient
(1996) Identification of primitive human NOD-scid mice. J Immunol 158:3578–3586
hematopoietic cells capable of repopulating 23. Strowig T, Gurer C, Ploss A et al (2009)
NOD/SCID Mouse bone marrow: implica- Priming of protective T cell responses against
tions for gene therapy. Nat Med 2:1329–1337 virus-induced tumors in mice with human
12. Hogan CJ, Shpall EJ, McNulty O et al (1997) immune system components. J Exp Med 206:
Engraftment and development of human 1423–1434
CD34(+)-enriched cells from umbilical cord 24. Suzuki M, Takahashi T, Katano I et al (2012)
blood in NOD/LtSz-scid/scid mice. Blood Induction of human humoral immune res-
90:85–96 ponses in a novel HLA-DR-expressing trans-
13. Kerre TC, De Smet G, De Smedt M et al genic NOD/Shi-scid/gammacnull mouse. Int
(2002) Adapted NOD/SCID model supports Immunol 24:243–252
development of phenotypically and function- 25. Billerbeck E, Horwitz JA, Labitt RN et al
ally mature T cells from human umbilical cord (2013) Characterization of human antiviral
blood CD34(+) cells. Blood 99:1620–1626 adaptive immune responses during hepato-
14. McKenzie JL, Gan OI, Doedens M et al (2005) tropic virus infection in HLA-transgenic
Human short-term repopulating stem cells are human immune system mice. J Immunol
efficiently detected following intrafemoral 191:1753–1764
transplantation into NOD/SCID recipients 26. Shultz LD, Brehm MA, Garcia-Martinez JV
depleted of CD122+ cells. Blood et al (2012) Humanized mice for immune
106:1259–1261 system investigation: progress, promise and
15. Shultz LD, Banuelos SJ, Leif J et al (2003) challenges. Nat Rev Immunol 12:786–798
Regulation of human short-term repopulating 27. Brehm MA, Racki WJ, Leif J et al (2012)
cell (STRC) engraftment in NOD/SCID mice by Engraftment of human HSCs in nonirradiated
host CD122+ cells. Exp Hematol 31:551–558 newborn NOD-scid IL2rgamma null mice is
16. Ito M, Hiramatsu H, Kobayashi K et al (2002) enhanced by transgenic expression of membrane-
NOD/SCID/gamma(c)(null) mouse: an excel- bound human SCF. Blood 119:2778–2788
lent recipient mouse model for engraftment of 28. O’Connell RM, Balazs AB, Rao DS et al
human cells. Blood 100:3175–3182 (2010) Lentiviral vector delivery of human
17. Shultz LD, Lyons BL, Burzenski LM et al (2005) interleukin-7 (hIL-7) to human immune sys-
Human lymphoid and myeloid cell development tem (HIS) mice expands T lymphocyte popula-
in NOD/LtSz-scid IL2R gamma null mice tions. PLoS One 5, e12009
Humanized Mice to Study Human T Cell Development 271
29. Song J, Willinger T, Rongvaux A et al (2010) tive and innate immune responses to EBV and
A mouse model for the human pathogen TSST-1. Nat Med 12:1316–1322
Salmonella typhi. Cell Host Microbe 8: 42. Denton PW, Garcia JV (2009) Novel human-
369–376 ized murine models for HIV research. Curr
30. Tary-Lehmann M, Saxon A, Lehmann PV HIV/AIDS Rep 6:13–19
(1995) The human immune system in hu-PBL- 43. McDermott SP, Eppert K, Lechman ER et al
SCID mice. Immunol Today 16:529–533 (2010) Comparison of human cord blood
31. Shultz LD, Saito Y, Najima Y et al (2010) engraftment between immunocompromised
Generation of functional human T-cell subsets mouse strains. Blood 116:193–200
with HLA-restricted immune responses in 44. Notta F, Doulatov S, Dick JE (2010)
HLA class I expressing NOD/SCID/IL2r Engraftment of human hematopoietic stem
gamma(null) humanized mice. PNAS 107: cells is more efficient in female NOD/
13022–13027 SCID/IL-2Rgc-null recipients. Blood 115:
32. Jaiswal S, Pazoles P, Woda M et al (2012) 3704–3707
Enhanced humoral and HLA-A2-restricted 45. Pearson T, Greiner DL, Shultz LD (2008)
dengue virus-specific T-cell responses in Creation of “humanized” mice to study human
humanized BLT NSG mice. Immunology immunity. Curr Protoc Immunol Chapter 15,
136:334–343 unit 15.21
33. Danner R, Chaudhari SN, Rosenberger J et al 46. King MA, Covassin L, Brehm MA et al (2009)
(2011) Expression of HLA class II molecules Human peripheral blood leucocyte non-obese
in humanized NOD.Rag1KO.IL2RgcKO mice diabetic-severe combined immunodeficiency
is critical for development and function of interleukin-2 receptor gamma chain gene
human T and B cells. PLoS One 6, e19826 mouse model of xenogeneic graft-versus-host-
34. Holyoake TL, Nicolini FE, Eaves CJ (1999) like disease and the role of host major histo-
Functional differences between transplantable compatibility complex. Clin Exp Immunol
human hematopoietic stem cells from fetal 157:104–118
liver, cord blood, and adult bone marrow. Exp 47. Bruck F, Belle L, Lechanteur C et al (2013)
Hematol 27:1418–1427 Impact of bone marrow-derived mesenchymal
35. Ishikawa F, Yasukawa M, Lyons B et al (2005) stromal cells on experimental xenogeneic graft-
Development of functional human cord blood versus-host disease. Cytotherapy 15:267–279
and immune systems in NOD/SCID/IL2 48. Ali N, Flutter B, Sanchez RR et al (2012)
receptor {gamma} chain(null) mice. Blood Xenogeneic graft-versus-host-disease in NOD-
106:1565–1573 scid IL-2Rgammanull mice display a T-effector
36. McCune JM, Namikawa R, Kaneshima H et al memory phenotype. PLoS One 7, e44219
(1988) The SCID-hu mouse: murine model 49. Cai S, Wang H, Bailey B et al (2011)
for the analysis of human hematolymphoid Differential secondary reconstitution of
differentiation and function. Science 241: in vivo-selected human SCID-repopulating
1632–1639 cells in NOD/SCID versus NOD/SCID/
37. Kyoizumi S, Baum CM, Kaneshima H et al gamma chain mice. Bone Marrow Res
(1992) Implantation and maintenance of func- 2011:252953
tional human bone marrow in SCID-hu mice. 50. Kwant-Mitchell A, Pek EA, Rosenthal KL et al
Blood 79:1704–1711 (2009) Development of functional human NK
38. Baum CM, Weissman IL, Tsukamoto AS et al cells in an immunodeficient mouse model with
(1992) Isolation of a candidate human hema- the ability to provide protection against tumor
topoietic stem-cell population. PNAS 89: challenge. PLoS One 4, e8379
2804–2808 51. Waskow C, Madan V, Bartels S et al (2009)
39. Chen BP, Galy A, Kyoizumi S et al (1994) Hematopoietic stem cell transplantation with-
Engraftment of human hematopoietic precur- out irradiation. Nat Methods 6:267–269
sor cells with secondary transfer potential in 52. Waskow C, Cosgun N, Rahmig S (2013) Stable
SCID-hu mice. Blood 84:2497–2505 human hematopoietic stem cell engraftment
40. Shih CC, Hu MC, Hu J et al (1999) Long- supports continuous de novo generation of
term ex vivo maintenance and expansion of mature human blood cells in mice. Exp
transplantable human hematopoietic stem cells. Hematol 41:S11
Blood 94:1623–1636 53. Choi B, Chun E, Kim M et al (2011) Human
41. Melkus MW, Estes JD, Padgett-Thomas A et al B cell development and antibody production in
(2006) Humanized mice mount specific adap- humanized NOD/SCID/IL-2Rgamma(null)
272 Sarah Bonte et al.
Abstract
While zebrafish have for some time been regarded as a powerful model organism with which to study early
events in hematopoiesis, recent evidence suggests that it also ideal for unraveling the molecular requi-
rements for T cell development in the thymus. Like mammals, zebrafish possess an adaptive immune
system, comprising B lymphocytes as well as both the γδ and αβ lineages of T cells, which develop in the
thymus. Moreover, the molecular processes underlying T cell development in zebrafish appear to be
remarkably conserved. Thus, findings in the zebrafish model will be of high relevance to the equivalent
processes in mammals. Finally, molecular processes can be interrogated in zebrafish far more rapidly than
is possible in mammals because the zebrafish possesses many unique advantages. These unique attributes,
and the methods by which they can be exploited to investigate the role of novel genes in T cell develop-
ment, are described here.
Key words Zebrafish, T cell development, Morpholino, Functional rescue, In situ hybridization
1 Introduction
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5_22, © Springer Science+Business Media New York 2016
273
274 Yong Zhang and David L. Wiest
Table 1
Transgenic lines for studying thymocyte development in zebrafish
2 Materials
2.2 Zebrafish Lines Wild-type and transgenic zebrafish lines are available from Zebrafish
International Resource Center (ZIRC, Eugene, OR). For examples
described in this chapter, we employed the following fish lines: (1)
AB wild-type fish; and (2) Tg(lck:EGFP)cz1 [17].
276 Yong Zhang and David L. Wiest
Fig. 1 Microinjection and dechorionation for zebrafish embryos. (a) Injection mold. (b) Injection chamber/plate.
(c) Schematic of morpholino or plasmid injection. For morpholino knockdown, we inject the liquid (red spot) to
the interface between cell and yolk. For plasmid overexpression, we directly inject the solution to the cyto-
plasm (green spot) at 1-cell stage. (d) Dechorionation process. The arrows indicate the chorions around the
24hpf embryos. (e) Alignment of the one-cell stage embryos in the trenches of the injection plate. (f) Schematic
of the vertical section of the injection plate. (g) Lateral view of a 5dpf Tg(lck:EGFP)cz1 embryo. Arrow indicates
the EGFP+ T cells in the thymus
2.5 Transient Rescue 1. Plasmid DNA preparation kit for highly purified DNA.
Experiments 2. Water bath.
3. Construct: For heat shock mediated inducible expression, clone
cDNA encoding the gene of interest into the heat-inducible
pSGH2 vector [6, 20]. The pSGH2 vector contains two I-SceI
restriction sites.
4. I-SceI meganuclease: (New England Biolabs) Aliquot 2 μl each
upon arrival and store at −80 °C) along with I-SceI buffer
(New England Biolabs), supplied with the NEBuffer I-SceI
pack (see Note 3).
2.6 Western Blot [21] 1. Deyolking buffer: 55 mM NaCl, 1.8 mM KCl, 1.25 mM
NaHCO3.
2. Wash buffer: 10 mM Tris–HCl pH 8.5, 110 mM NaCl,
3.5 mM KCl, 2.7 mM CaCl2.
3. 200 μl Precision tips.
4. Protease inhibitor cocktail (Roche).
5. PMSF.
278 Yong Zhang and David L. Wiest
2.7 Whole-Mount 1. In Situ Cell Death Detection Kit, TMR red (Roche).
TUNEL Assays 2. Acetone.
3. Permeabilization solution (0.1 % v/v Triton X-100 and 0.1 %
w/v sodium citrate in PBS): 100 μl Triton X-100 and 1 ml
10 % sodium citrate in 98 ml of pH 7.4 PBS, freshly
prepared.
3 Methods
3.1 Identifying Many mammalian genes have zebrafish orthologs. We use both
Zebrafish Ortholog homology and synteny-based approaches to identify the zebrafish
orthologs of mammalian genes. Conserved synteny (i.e., contigu-
ous genes or ESTs with conserved map order on the chromosomes
of different species) is a prominent feature of vertebrate genomes.
Analysis of zebrafish genomic mapping data has revealed conserved
rearrangements and homology segments between zebrafish and
human genomes [5]. Thus, the homology based syntenic analysis
can be used to effectively predict and find orthologous genes
between zebrafish and human.
1. Go to http://www.ensembl.org/Homo_sapiens/Info/index.
html and enter the name of the human gene (e.g., RPL22) in
the SEARCH section.
2. Click on ContigView, then the gene name, to get the amino
acid sequence as well as genomic position.
3. Go to http://www.ncbi.nlm.nih.gov/, select tBLASTn and
paste the amino acid sequence in the dialog box. Select
“Nucleotide selection (nr/nt)” in the “Choose search set-
Database” window, and “zebrafish” in the “Organism” win-
dow, and click on BLAST, followed by clicking on “Formatting”
in a new window to get the search summary.
4. Record the zebrafish gene name (e.g., zgc:123327, rpl22), then
go to http://www.genome.ucsc.edu/ to identify the genomic
position of this gene.
Studying T Cell Development in Zebrafish 281
5. Repeat these steps for other genes located near the gene
of interest until it is clear that there is a good syntenic
relationship.
6. Next align the amino acid sequences of the human, mouse and
zebrafish orthologs of your gene of interest. The Clustal W2
algorithm is an effective tool for this task (http://www.ebi.
ac.uk/Tools/msa/clustalw2/).
3.4 Dechorionation The chorion is the membrane around the developing zebrafish
embryo (see Fig. 1d, arrows). Embryos should be removed from
their protective chorions to facilitate further observation, fixation,
or other manipulations. Removal of the chorion, or dechorion-
ation, is accomplished by pronase digestion of embryos older than
18hpf. We generally dechorionate at 24hpf.
1. Dilute 10 mg/ml pronase stock with egg water to 2 mg/ml
working solution.
2. Transfer the embryos to a petri dish, removing as much egg
water as possible. Add 2 ml of the diluted pronase solution to
the dish and swirl the embryos. Incubate at room temperature
for 10 min.
3. Use a Pasteur pipette to gently transfer the embryos to a fresh
petri dish full of egg water. Most of chorions will have been
removed at this time (see Fig. 1d).
4. Rinse the embryos thoroughly (at least three times) with egg
water to remove the pronase.
3.5 FACS Sorting 1. Transgenic zebrafish embryos produced as above from appro-
of Lineage Marked priate lines [Table 1; e.g., Tg(lck:EGFP)cz1] are dechorionated
T Cell Progenitors by pronase treatment. For FACS sorting of fluorescently
labeled progenitors, we use about 100 transgenic embryos
(5dpf, see Fig. 1e) that have been grown in the 28.5 °C
incubator.
2. To anesthetize the embryos, transfer them to a petri dish con-
taining Tricaine in egg water at 0.2 mg/ml and incubate until
they are no longer swimming.
3. Transfer anesthetized embryos to deyolking buffer (keep on
ice) and pass them through a 200 μl pipette tip several times to
remove their yolk.
4. Dissect and macerate deyolked whole embryos in ice-cold
0.9 × PBS plus 5 % FBS using a scalpel blade to facilitate
digestion.
5. Transfer embryos to a 35 mm culture dish containing 1× trypsin–
EDTA solution and incubate for 30–60 min at 32 °C. During
incubation, gently agitate by pipetting every 10 min.
6. Terminate the digestion by adding CaCl2 to a final concentra-
tion of 1 mM and fetal calf serum to 10 %.
7. Pellet the cells for 5 min at 400 × g, then wash twice with
0.9 × PBS at 400 × g.
8. Filter the suspension through 40 μm nylon mesh to eliminate
debris (Keep cold).
9. Pellet and resuspend the cells in 0.9 × PBS/5 % FBS. Stain with
1 μg/ml propidium iodide (PI) 30 min on ice to identify dead
cells and debris.
Studying T Cell Development in Zebrafish 283
3.6 Morpholino 1. Before injection, heat the morpholino (MO; antisense oligo-
Microinjection nucleotide) in a thermocycler for 10 min at 65 °C to dissolve
any precipitates that can clog the microinjection needle.
2. Assemble the needle to the microinjection arm.
3. Turn on air the compressor pump and PM1000 microinjector.
4. Press the “Timer” and “BALN” buttons to activate balance
pressure. Press the “VENT” button.
5. Use the “Fill” button to load the needle with the MO target-
ing your gene of interest.
6. Use the “Inject” knob on the injector panel to adjust the injec-
tion air pressure and set the injection time. For the PM1000
cell microinjector, we use 60 ms injection time and 10-psi
injection pressure as the starting point. Use the “Balance”
knob to adjust the balance pressure between 0 and 0.4 psi to
prevent both liquid from leaking out of the needle and medium
from flowing back into the needle.
7. Place a drop of mineral oil on the microscope stage microme-
ter. Submerge the tip of the injection needle into the mineral
oil and press the foot pedal to finish the injection. The released
droplet will form a perfect sphere in the oil. Measure the diam-
eter of the droplet and calculate the volume according
to 4/3πR3. Adjust the injection pressure and injection time to
produce the desired injection volume and dose of morpholino
(see Table 2).
8. Transfer embryos into the trenches of the injection plate using
a Pasteur pipette (Fig. 1e).
9. Use the fine probe to align the embryos, with the animal pole
facing you (see Fig. 1e, f).
10. Manipulate the injection arm to advance the needle, gently
piercing the chorion of the embryos. Inject the morpholino
into the interface between cytoplasm and yolk of one-cell stage
embryos (see Fig. 1c, lower panel and Note 4).
11. Move the injection plate and repeat until all of the embryos on
the plate have been injected, then transfer the embryos to
a clean dish containing egg water, and place in 28.5 °C
incubator.
284 Yong Zhang and David L. Wiest
Table 2
Morpholino (MO) dose per microinjection
Fig. 2 Using zebrafish model to investigate the role of Rpl22 in T cell development. (a and b) Targeted knock-
down of Rpl22 in zebrafish embryos. (a) For Rpl22, the MO is designed to specifically block the initiation of
translation at the start codon in exon 1. The short black line and blue arrow indicate the positions of sequences
targeted. (b) The effect of 1 ng Rpl22-MO knockdown on expression of endogenous Rpl22 is assessed by
immunoblotting on 5dpf embryos. (c) Rpl22-MO injected (1 ng), but not MM (5-base pair mismatch) control
injected, Tg(lck:EGFP) fish exhibit a loss of EGFP-marked T cells at 5 dpf (lateral view, red circles; dorsal view,
yellow rectangles). Numbers refer to fraction of morphants with the depicted phenotypes. (d) Injection of 1 ng
Rpl22-MO causes apoptosis in the thymus at 3.5 dpf, while injection of the same quantity of 5-base pair mis-
match control morpholino (Rpl22-MM) caused no change in the thymus (green dashed rectangles). The white
dashed line delimits the eye. Numbers refer to fraction of morphants with the depicted phenotypes. (e–h) The
use of a heat-shock inducible, bidirectional expression construct to restore T cell development in rpl22 mor-
phants. (e) Upper panels depict schematics of the heat-inducible expression plasmid pSGH2-mRpl22. (f) The
heat shock-inducible expression plasmid associated with I-sceI mediated transient overexpression is injected
into one-cell stage embryos. At 3dpf, the embryos are heat shocked at 37 °C for 1 h and then effects on T cell
development are analyzed at 5 dpf using WISH for lck, a marker of thymic progenitors. (g) After heat-shock,
GFP+ embryos are selected for subsequent WISH analysis (pink arrowheads). (h) Heat shock induction of
mouse Rpl22 expression rescues T cell development in rpl22 morphants (5dpf lck WISH staining, red rectan-
gles). Numbers refer to fraction of morphants with the depicted phenotypes
Studying T Cell Development in Zebrafish 287
4 Notes
Acknowledgments
We are grateful to the support and help from Dr. Jennifer Rhodes,
Allison Ulrich and Alison N. Bilbee for the zebrafish work.
We gratefully acknowledge the assistance of the following core
facilities of the Fox Chase Cancer Center: Flow Cytometry, DNA
Sequencing, Imaging and Laboratory Animal/Zebrafish. This work
was supported by NIH grants AI081814, AI073920, NIH core
grant P01CA06927, Center grant P30-DK-50306. Y.Z. is a W.J.
Avery Postdoctoral Fellow of Fox Chase Cancer Center.
References
1. Amatruda JF, Zon LI (1999) Dissecting established by the Notch-Runx pathway. Genes
hematopoiesis and disease using the zebrafish. Dev 19(19):2331–2342
Dev Biol 216(1):1–15 5. Rhodes J, Hagen A, Hsu K, Deng M, Liu TX,
2. Lieschke GJ, Currie PD (2007) Animal models Look AT, Kanki JP (2005) Interplay of pu.1
of human disease: zebrafish swim into view. and gata1 determines myelo-erythroid progen-
Nat Rev Genet 8(5):353–367 itor cell fate in zebrafish. Dev Cell 8(1):
3. Trede NS, Langenau DM, Traver D, Look AT, 97–108
Zon LI (2004) The use of zebrafish to under- 6. Bajoghli B, Aghaallaei N, Hess I, Rode I,
stand immunity. Immunity 20(4):367–379 Netuschil N, Tay BH, Venkatesh B, Yu JK,
4. Burns CE, Traver D, Mayhall E, Shepard JL, Kaltenbach SL, Holland ND, Diekhoff D,
Zon LI (2005) Hematopoietic stem cell fate is Happe C, Schorpp M, Boehm T (2009)
292 Yong Zhang and David L. Wiest
Evolution of genetic networks underlying the 15. Hess I, Boehm T (2012) Intravital imaging
emergence of thymopoiesis in vertebrates. Cell of thymopoiesis reveals dynamic lympho-
138(1):186–197 epithelial interactions. Immunity 36(2):
7. Murayama E, Kissa K, Zapata A, Mordelet E, 298–309
Briolat V, Lin HF, Handin RI, Herbomel P 16. Langenau DM, Feng H, Berghmans S, Kanki
(2006) Tracing hematopoietic precursor mig- JP, Kutok JL, Look AT (2005) Cre/lox-
ration to successive hematopoietic organs regulated transgenic zebrafish model with con-
during zebrafish development. Immunity 25(6): ditional myc-induced T cell acute lymphoblastic
963–975 leukemia. Proc Natl Acad Sci U S A 102(17):
8. Schorpp M, Bialecki M, Diekhoff D, Walderich 6068–6073
B, Odenthal J, Maischein HM, Zapata AG, 17. Langenau DM, Ferrando AA, Traver D, Kutok
Boehm T (2006) Conserved functions of Ikaros JL, Hezel JP, Kanki JP, Zon LI, Look AT,
in vertebrate lymphocyte development: genetic Trede NS (2004) In vivo tracking of T cell
evidence for distinct larval and adult phases of development, ablation, and engraftment in
T cell development and two lineages of B cells transgenic zebrafish. Proc Natl Acad Sci U S A
in zebrafish. J Immunol 177(4):2463–2476 101(19):7369–7374
9. Willett CE, Cherry JJ, Steiner LA (1997) 18. Nasevicius A, Ekker SC (2000) Effective tar-
Characterization and expression of the recom- geted gene ‘knockdown’ in zebrafish. Nat
bination activating genes (rag1 and rag2) of Genet 26(2):216–220
zebrafish. Immunogenetics 45(6):394–404 19. Kimmel CB, Ballard WW, Kimmel SR, Ullmann
10. Langenau DM, Zon LI (2005) The zebrafish: B, Schilling TF (1995) Stages of embryonic
a new model of T-cell and thymic develop- development of the zebrafish. Dev Dyn 203(3):
ment. Nat Rev Immunol 5(4):307–317 253–310
11. Ma D, Wei Y, Liu F (2013) Regulatory mecha- 20. Bajoghli B, Aghaallaei N, Heimbucher T,
nisms of thymus and T cell development. Dev Czerny T (2004) An artificial promoter con-
Comp Immunol 39(1-2):91–102 struct for heat-inducible misexpression during
12. Zhang Y, Duc AC, Rao S, Sun XL, Bilbee AN, fish embryogenesis. Dev Biol 271(2):416–430
Rhodes M, Li Q, Kappes DJ, Rhodes J, Wiest DL 21. Link V, Shevchenko A, Heisenberg CP (2006)
(2013) Control of hematopoietic stem cell emer- Proteomics of early zebrafish embryos. BMC
gence by antagonistic functions of ribosomal pro- Dev Biol 6:1
tein paralogs. Dev Cell 24(4):411–425 22. Anderson SJ, Lauritsen JP, Hartman MG,
13. Kissa K, Murayama E, Zapata A, Cortes A, Foushee AM, Lefebvre JM, Shinton SA,
Perret E, Machu C, Herbomel P (2008) Live Gerhardt B, Hardy RR, Oravecz T, Wiest DL
imaging of emerging hematopoietic stem cells (2007) Ablation of ribosomal protein L22
and early thymus colonization. Blood selectively impairs alphabeta T cell develop-
111(3):1147–1156 ment by activation of a p53-dependent check-
14. Bertrand JY, Kim AD, Teng S, Traver D (2008) point. Immunity 26(6):759–772
CD41+ cmyb+ precursors colonize the zebraf- 23. Gupta S, Zhu H, Zon LI, Evans T (2006)
ish pronephros by a novel migration route to BMP signaling restricts hemato-vascular devel-
initiate adult hematopoiesis. Development opment from lateral mesoderm during somito-
135(10):1853–1862 genesis. Development 133(11):2177–2187
INDEX
A CD249............................................................................... 71
CD326............................................................. 66, 68–70, 72
Aire................................................. 11, 14, 27, 42, 65, 66, 70 CD1a ....................................................... 222, 229, 233, 248
Allelic exclusion......................................................... 39, 186 CD16/CD32 antibody ............................ 111, 113, 213, 215
7-Aminoactinomycin D (7-AAD) ........ 55, 59–62, 217–219 CD1d tetramers............................................ 6, 51, 55, 56, 60
alternatives ........................................................... 62, 219 CD51, mouse .................................................................. 250
Antibiotics ....................................................... 110, 111, 113 CD4 SP ................................ 14, 15, 48–51, 53–57, 222, 232
Antigen-presenting cells (APCs) ........................... 5, 14, 69, CD8 SP .......................... 15, 16, 37, 48–51, 53–56, 222, 232
117, 118, 120, 122–127, 142, 171, 173, 177, 255 Cell cycle .................................... 62, 101, 212, 214, 217, 218
Apoptosis.......................... 141, 142, 148, 169, 186, 285, 286 Central memory ................................................................ 51
agonist induced .......................................................... 142 Changes in repertoire ...................................................... 189
Chimeras
B
bone marrow ........................... 36, 41, 101, 104, 109–115
Bcl2 ......................................................................... 8, 10, 12 fetal liver ............................................................ 109–115
Bim .................................................................................... 12 Click reaction .................................................................. 212
BLAST............................................................................ 280 Collagenase ................................ 66, 67, 69, 71, 72, 263, 266
Bone marrow............................................... 6, 7, 14, 36, 41, 47, 75, Collection tubes, polystyrene ....................................... 95, 97
100, 101, 103–106, 109–115, 159, 171, 221, 225–227, Complement
229, 234, 246, 250, 258, 260, 261, 264, 265 guinea pig .................................................. 110, 112, 114
T cell depletion .......................................... 112–114, 171 rabbit ................................................. 110, 114, 161, 165
Bone marrow growth medium (BMGM)................ 102–104 Conditioning mix ............................................ 118, 122–126
Bone marrow transplantation (BMT) ..................... 109, 113 Confocal microscopy ................................. 82, 143, 170–175
Bone wax ................................................................. 262, 265 Continuous labelling ....................................................... 212
5-Bromo-2-deoxyuridine (BrdU) ............ 211, 212, 218, 219 Corticomedullary junction (CMJ) ..................................... 75
Busulfan........................................................................... 261 Cre recombinase ................................................................ 36
Cytotoxic T (Tc) ...................... 118–120, 122, 123, 125, 223
C
D
CCR7 ........................................................... 7, 10, 14, 17, 65
CD3 .................... 4, 8, 50, 118, 222, 224–225, 228–233, 248 Danieau buffer ................................................................. 276
CD7 ........................................................ 221, 222, 246–248 Death by neglect .............................................. 142, 143, 169
CD8 .............................................. 4–6, 9, 10, 12, 13, 15–18, Dechorionation ................................ 277, 282, 284, 287, 289
25–27, 37–39, 41, 47–56, 75, 76, 88, 91, 92, 96, 118, Defects in V(D)J recombination.............................. 189, 192
120, 125, 127, 144, 147, 160, 161, 170, 185, 192, Deletion reporters.............................................................. 36
222–226, 229–232, 236, 248 Dendritic epidermal T cells (DETC) .................... 25–28, 30
CD25 ........................... 15, 16, 49–51, 57, 62, 161, 192, 222 2′-Deoxyguanosine (dGuo) .............. 151, 152, 154, 155, 157
CD27 ......................................................... 9, 15, 17, 26, 185 Dermal fibroblast..................................................... 160, 162
CD28 .............................................. 9, 15, 17, 117, 118, 125, Developmental arrest ............................................... 289–290
185, 222, 223, 225, 231 4′,6-Diamidino-2-phenylindole
CD34+..................................... 221, 222, 225, 227–229, 234, (DAPI) ................................................... 62, 78, 85,
235, 242, 243, 246, 248, 250, 261, 263–269 97, 161, 171,214, 217–220
CD38 ...................................................................... 225, 228 Dimethyl sulfoxide (DMSO) ......................... 115, 162, 213,
CD44 .......................... 15, 16, 49, 50, 55, 120, 161, 192, 222 214, 216, 218, 223, 240, 242, 249
CD73 .......................................................................... 27, 29 Dispase ...................................................................... 71, 263
CD80 ................................................................................ 71 DN1 ...................................................................... 15, 16, 49
CD117 (cKit) ...................................................... 15, 16, 165 DN3 ................................................... 15–17, 37, 49, 50, 142
CD205................................................................... 66, 69–71 DNA content analysis ..................................................... 212
Rémy Bosselut and Melanie S. Vacchio (eds.), T-Cell Development: Methods and Protocols, Methods in Molecular Biology,
vol. 1323, DOI 10.1007/978-1-4939-2809-5, © Springer Science+Business Media New York 2016
293
T-CELL DEVELOPMENT: METHODS AND PROTOCOLS
294 Index
IP followed by flow cytometry (IP-FCM) ............. 143, 170, Mucosal-associated invariant T cells (MAIT) ..................... 6
172, 175–177 Murine dermal fibroblasts expressing Dll4
IRF1 .................................................................................. 13 (mFibro-DL4) ..................................... 160, 164–166
Irradiation....................................................... 109, 111, 112, Murine stem cell virus (MSCV) ............... 99, 100, 102, 249
115, 197, 259, 261, 263, 268, 269 Mutagen .................................................................. 218, 219
I-sceI meganuclease ......................................... 277, 289, 290
ISP.................................................. 48–51, 53, 223, 225, 235 N
Natural killer NK cells .................................... 221, 228, 245,
K
253, 254, 260
Ketamine/xylazine ................................................... 204, 208 Natural killer T (NKT)..................................... 6, 51, 54, 56,
222, 223, 253
L Negative selection ................................ 11–12, 14, 27, 40–42,
lck...................................... 4, 37, 38, 274, 275, 286, 289, 290 50, 65, 87–89, 131, 132, 141–148, 169, 170, 223, 254
Lentiviral vectors ..................................................... 100, 104 Nomenclature
Lentivirus ......................................... 100–102, 105, 240, 249 historical ............................................ 181–182, 189–191
Lineage commitment ..................................... 7, 9, 15, 27, 28 IMGT/NCBI .................................... 180, 181, 183, 184
instructional ................................................................. 28 TCRα .......................................................... 25, 184, 189
stochastic ..................................................................... 28 Non-homologous end-joining (NHEJ) ........................... 179
Lineage deviation .................................................... 148, 169 Notch ........................................................ 7–9, 14, 159, 160,
Lineage markers ...................................... 101, 228, 282–283 206, 239, 241, 246, 250
Lineage mix ........................................................... 50, 58, 60 Notch ligand Delta-like....................................... 7, 159–167
Lipofectamine ......................................................... 102–104 Nozzle, 100mm ..................................................... 72, 94, 95
Live gate ...................................................................... 61, 62 Nucleoside analogs .......................................................... 212
Lymph nodes (LN) Nude mouse....................................................................... 13
mesenteric.................................................................... 96 Nur77 ................................................................................ 12
peripheral............................................................... 57, 96 Nylon mesh ......................................... 54, 57, 58, 66–68, 70,
133, 147, 153, 155, 162, 166, 167, 172, 235, 280, 282
M
O
Macrophages ............................................... 14, 54, 241, 254
Magnetic beads, antibody coated .......................... 87, 90–93, OP9 co-culture system .................... 240–242, 244–248, 250
114, 120, 121, 230 OP9-DL.......................................... 159–161, 164–166, 246
Major histocompatibility complex (MHC) .............. 5, 7–13, OP9 stromal cells expressing human DLL4
169, 254, 255, 258, 260 (OP9-hDLL4)..................................... 241, 243, 250
MAP kinases, selection...................................................... 12 Ortholog .......................................................... 274, 280–281
Methylene blue ........................................................ 275, 290
P
MHC Class I..................................................... 24, 254, 255
MHC Class II ................................................................. 255 Paraffin embedding ................................... 76–78, 80–81, 84
MHC, non-classical .................................................... 24, 30 Paraffin permeabilization................................................... 84
β2 Microglobulin ............................................... 41, 254, 257 Paraformaldehyde (PFA) ...................................... 63, 66, 68,
Microinjection .................................................. 38, 276–277, 76–80, 82, 84, 278, 285
281, 283–284, 289, 290 Plat-E cells .............................................................. 156, 158
mobilized peripheral blood .............................. 258, 260, 261 PLZF ................................................................................ 29
Morpholinos (MO) .......... 274, 276, 277, 283–286, 290, 291 Polybrene .......................................... 102–104, 106, 153, 156
Mouse model Polymerase chain reaction (PCR)
BALB/c-Rag2null (BRG) ................... 115, 183, 254–256, conventional ....................... 187, 189, 193, 195–196, 283
259, 261, 269 quantitative real-time......................... 167, 187, 193–197
BLT ............................................ 255, 258, 260, 263, 267 Positive selection.......................................... 9, 10, 12, 17, 40,
Hu-HSC ................................................... 255, 258, 260 50, 65, 87, 89, 131, 132, 142, 143, 147, 169, 186
NOD/LtSz-scid Il2rgnull (NSG)................. 254, 256, 262 Pre-T cell receptor (pTα)........................................... 27, 223
NOD/Rag1null IL2rgnull (NRG).......................... 254, 256 Primary mouse fibroblast ......................................... 162–163
NOD-scid ................................................................. 254 Prkdc ................................................. 253, 254, 256, 257, 262
NOD/Shi-scid Il2rgnull (NOG) .......................... 254, 256 Propidium iodide (PI) ..................................... 62, 66, 68, 69,
SCID HU.................................................. 255, 258, 260 219, 248, 250, 282
SCID PBL ........................................................ 258–261 Pseudotyping .................................... 100, 106, 111, 180, 186
T-CELL DEVELOPMENT: METHODS AND PROTOCOLS
296 Index
pSGH2 vector ......................................................... 277, 290 T cell depletion, complement ......................... 110, 112–114,
Pulse labelling.......................................................... 214, 219 120, 123–124, 127, 171
Puma ................................................................................. 12 T cells
CD4+ ..................................... 5, 9, 12, 13, 16, 25, 27, 37,
R 38, 41, 47–49, 76, 88, 96, 120, 127, 185, 192, 222,
RANK ............................................................................... 14 223, 225, 226, 229–232, 235, 236, 246
Recombination CD8+ ........................................... 5, 9, 10, 12, 13, 16, 25,
TCRα ............................................ 8, 179, 183–187, 189, 27, 37, 38, 41, 47–49, 76, 88, 96, 118, 120, 125, 127,
192, 200, 223, 273 185, 192, 236
TCRβ ............................. 8, 180–183, 185, 187, 192, 273 γδ .............................. 4, 6–8, 15, 23–30, 52, 58, 222, 273
Recombination activating gene 1 (Rag1)................ 8, 10, 37, naïve ................................................ 7, 29, 37, 38, 51, 55,
41, 179, 253, 254, 256, 257, 273, 274 117, 119, 120, 122
Recombination activating gene 2 (Rag2)................ 8, 10, 41, regulatory............................................. 11, 13, 51, 57, 62,
115, 179, 253, 254, 256, 261, 273, 274 65, 118, 141, 222, 223
Recombination signal sequences (RSSs).......... 179, 180, 182 TCR-α .......................................... 4, 5, 7–10, 12, 38–42, 50,
Reconstitution ........................................... 41, 101, 106, 109, 179–180, 183–192, 196, 223, 273, 274
110, 113, 115, 151–158, 162, 165, 255, 260, 264, 265 TCR-β............................................. 4, 5, 7–9, 15–17, 39–42,
Restriction fragment length polymorphisms ................... 183 49–54, 180–183, 185–188, 192, 196, 273, 274
Retinoic acid .................................................... 119, 125, 127 TCR-δ............................................... 4, 8, 52, 183, 184, 186,
Retronectin ............................... 102, 104–106, 240, 243, 249 222, 223, 273, 274
Retroviral transduction TCR-γ ....................................................... 4, 8, 9, 16, 39, 52,
hematopoietic cells ......................... 41, 99, 103–104, 161 221, 233, 273, 274
lymphoid cell lines ............................. 102, 104, 106, 109 TCR signal, nuclear targets of .......................... 4, 10–12, 16,
primary thymocytes ................................................... 104 28–30, 118, 223
Retrovirus Terasaki plates ......................................................... 153, 155
host range ............................................................ 99, 100 Teratogen......................................................................... 218
vectors, FIV-based ..................................................... 100 Th0 .......................................................... 118, 122, 123, 125
vectors, HIV-1 based ................................................. 100 Th1 ................................................... 118–120, 122, 123, 125
vectors, MSCV .................................... 99, 100, 102, 249 Th2 ................................................... 118–120, 122, 123, 125
RORγt ...................................................... 9, 29, 30, 101, 119 Th17 ......................................... 118–120, 122, 123, 125, 126
Runx ...................................................................... 9, 13, 274 Th17, Iscove's modified Dulbecco's medium
(IMDM) ............................................120, 125, 126,
S 223–226, 228, 240, 242, 243, 249
ThN......................................................................... 118, 125
Schnurri-2 ......................................................................... 12
Thoracotomy ........................................................... 204–208
SCID mice ...................................................... 253–260, 262
Thpok .......................................................................... 13, 38
Self-inactivating viruses (SIN) ................................ 100, 105
Thrombopoietin (TPO), human ............................. 240, 241
Serum testing, bovine ...................................... 223, 233, 249
Thymic epithelial cell (TEC) ............................... 6, 7, 9, 13,
Silanized slides .................................................................. 82
14, 27, 65–72, 75–77, 109, 142, 143, 169, 254, 260
Skint1 .......................................................................... 28, 30
cortical (cTEC) ................................... 65, 66, 69–72, 76
Southern blot............................ 186–188, 190, 193, 197–200
medullary (mTEC)......................... 27, 65, 66, 69–72, 76
Sphingosine 1 phosphate (S1P)......................................... 13
Thymic epithelium
Splice-blocking morpholinos................................... 276, 291
cortex ........................................ 6, 9, 10, 14, 65, 138, 142
Stat5 .................................................................................. 13
medulla ................................... 10–12, 14, 27, 42, 65, 142
Stem cell factor, human .................................. 102, 162, 165,
Thymic nurse cells (TNC) ................................................ 72
240, 241, 243, 245, 255, 261
Thymic organ culture
Stromal cells, thymic ...................................... 66–68, 71, 75,
fetal ...................................................... 41, 131, 142, 166
76, 131, 151, 152, 159–167, 241, 244, 250
reaggregate................................................................. 131
Surgery, cardiac........................................ 225, 234, 260, 267
reconstituted ................................................ 41, 151–158
T Thymic slices ........................................................... 131–139
Thymocytes
β5t ......................................................................... 65, 66, 70 average lifespan of DP ............................................... 186
TAP transporters ............................................................... 41 ISP............................................ 37, 48–51, 223, 225, 235
T-CELL DEVELOPMENT: METHODS AND PROTOCOLS
Index
297