the shaping of the t cell repertoire - harvard university · the shaping of the t cell repertoire...

12
Immunity, Vol. 14, 21–32, January, 2001, Copyright 2001 by Cell Press The Shaping of the T Cell Repertoire segments. Variability is concentrated in the CDRs; CDR1 Margarida Correia-Neves,* Caroline Waltzinger,* and CDR2 are encoded within the V segments them- Diane Mathis,* § and Christophe Benoist* § selves, while CDR3’s much greater variability stems * Institut de Ge ´ ne ´ tique et de Biologie Mole ´ culaire from its position at the junction of the V(D)J elements, et Cellulaire (CNRS/INSERM/ULP) compounded by their imprecise juxtaposition, with vari- 1 rue Laurent Fries able deletion and random addition of a few nucleotides 67404 Strasbourg at the joined ends. Given the number of V and J seg- France ments that randomly combine and the impact of nucleo- tide deletions and additions, the theoretical diversity of the mouse or human ab T cell repertoire has been Summary estimated at about 10 14 (Davis and Bjorkman, 1988). This number represents a “raw” repertoire, before its By combining a TCRb transgene with a TCRa mini- streamlining by selection for self-MHC compatibility and locus comprised of a single V and two J gene seg- self-tolerance. The postselection diversity of the periph- ments, we engineered a mouse line exhibiting ample eral repertoire is what conditions the individual’s ability but focused TCR diversity, restricted to CDR3a. Using to mount appropriate immune responses or, conversely, single-cell PCR and high-throughput sequencing, we to succumb to autoimmunity. Thus, an appreciation of have exploited this system to scrutinize T cell repertoire the true extent of this diversity is important. selection and evolution. Some striking observations Defining the T cell repertoire is a daunting task be- emerged: (1) thymic selection produces a repertoire that cause of its magnitude and of the complexities inherent is very “bumpy,” with marked overrepresentation of a in such a combinatorial structure. All TCR variable re- subset of sequences; (2) MHC class I– and class II– gions can be used by both MHC class I– and class restricted TCRs can be distinguished by minute, sin- II–reactive TCRs, even though there are preferential as- gle-residue changes in CDR3a; and (3) homeostatic sociations (Kappler et al., 1987; Jameson et al., 1990; expansion and survival in the periphery can markedly Sim et al., 1996). TCR transgenic (tg) approaches have remold the postselection repertoire, likely reflecting been very powerful for monitoring the fate of cells ex- variability in the potential of cells displaying different pressing TCRs of defined restriction and specificity, TCRs to respond to homeostatic cues. thereby allowing us to visualize selection processes (von Boehmer, 1990), but such strategies generally permit Introduction the assessment of only a single receptor at a time. PCR- based analyses that resolve TCRs on the basis of CDR3 Essentially all cells in multicellular organisms exchange lengths have addressed the bulk repertoire and have signals, typically monomolecular and structurally invari- been particularly useful in highlighting recurrent amplifi- ant. T lymphocytes differ, however, in that the principal cations in the context of immune responses (Pannetier cues governing their differentiation and homeostasis are et al., 1995). Some brute-force sequencing studies have received through a somatically variable receptor that attempted to describe the diversity of the VDJ joins of recognizes variable ligands composed of a diversity of single Vb regions in thymocytes or in peripheral CD4 1 small peptides complexed with polymorphic MHC mole- and CD8 1 T cells (Candeias et al., 1991; Arstila et al., cules. This mode of cell:cell communication imposes 1999), but the results have revealed only subtle corre- important constraints that predicate some critical fea- lates, probably because of the compensating influences tures of T cell behavior: MHC restriction (or the matching of Va elements. The difficulty has been side stepped of an individual’s complements of T cells and MHC mole- with single-chain tg mice expressing the b chain of an cules) and positive selection (or the learning process antigen-specific TCR (Sant’Angelo et al., 1997, 1998). A by which T cells maturing in the thymus acquire this significant proportion of T cells in these animals is reac- matched repertoire) (Fink and Bevan, 1978; Zinkernagel tive to the cognate peptide, and these cells frequently and Doherty, 1997). Furthermore, MHC molecules may use TCRa chains that are the same as or similar to the also mold the T cell repertoire postthymically, as these parental clone. In addition, the presence or absence of molecules seem necessary for the survival and homeo- the parental sequence motif can be used as a marker stasis of naive cells in the peripheral lymphoid organs in differentiating thymocytes. (Goldrath and Bevan, 1999a). In the present report, we have attempted to obtain a The TCR is composed of an a and a b chain, each broad but manageable view of the selection and evolu- encoded by variable, randomly rearranged V and (D)J tion of the T cell repertoire by engineering a mouse line in which T lymphocytes are polyclonal but TCR diversity To whom correspondence should be addressed (e-mail: cb@igbmc. is focused. This was achieved with a TCRa minilocus u-strasbg.fr [C. B.], [email protected] [D. M.]). transgene composed of a single Va region and two Ja Present address: Instituto de Biologia Molecular e Celular, Porto, elements separated by an artificial rearrangement sub- 4150, Portugal. strate. This transgene was combined with a mutation § Present address: Section on Immunology and Immunogenetics, that inactivates the endogenous TCRa locus and with Joslin Diabetes Center, Department of Medicine, Brigham and Wom- a TCRb transgene that, due to the dictates of allelic en’s Hospital, Harvard Medical School, One Joslin Place, Boston, Massachusetts 02215. exclusion, prevents the rearrangement of endogenous

Upload: others

Post on 21-Jun-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity, Vol. 14, 21–32, January, 2001, Copyright 2001 by Cell Press

The Shaping of the T Cell Repertoire

segments. Variability is concentrated in the CDRs; CDR1Margarida Correia-Neves,*‡ Caroline Waltzinger,*and CDR2 are encoded within the V segments them-Diane Mathis,*§ and Christophe Benoist*†§

selves, while CDR3’s much greater variability stems* Institut de Genetique et de Biologie Moleculairefrom its position at the junction of the V(D)J elements,et Cellulaire (CNRS/INSERM/ULP)compounded by their imprecise juxtaposition, with vari-1 rue Laurent Friesable deletion and random addition of a few nucleotides67404 Strasbourgat the joined ends. Given the number of V and J seg-Francements that randomly combine and the impact of nucleo-tide deletions and additions, the theoretical diversityof the mouse or human ab T cell repertoire has beenSummaryestimated at about 1014 (Davis and Bjorkman, 1988).This number represents a “raw” repertoire, before itsBy combining a TCRb transgene with a TCRa mini-streamlining by selection for self-MHC compatibility andlocus comprised of a single V and two J gene seg-self-tolerance. The postselection diversity of the periph-ments, we engineered a mouse line exhibiting ampleeral repertoire is what conditions the individual’s abilitybut focused TCR diversity, restricted to CDR3a. Usingto mount appropriate immune responses or, conversely,single-cell PCR and high-throughput sequencing, weto succumb to autoimmunity. Thus, an appreciation ofhave exploited this system to scrutinize T cell repertoirethe true extent of this diversity is important.selection and evolution. Some striking observations

Defining the T cell repertoire is a daunting task be-emerged: (1) thymic selection produces a repertoire thatcause of its magnitude and of the complexities inherentis very “bumpy,” with marked overrepresentation of ain such a combinatorial structure. All TCR variable re-subset of sequences; (2) MHC class I– and class II–gions can be used by both MHC class I– and classrestricted TCRs can be distinguished by minute, sin-II–reactive TCRs, even though there are preferential as-gle-residue changes in CDR3a; and (3) homeostaticsociations (Kappler et al., 1987; Jameson et al., 1990;expansion and survival in the periphery can markedlySim et al., 1996). TCR transgenic (tg) approaches haveremold the postselection repertoire, likely reflectingbeen very powerful for monitoring the fate of cells ex-variability in the potential of cells displaying differentpressing TCRs of defined restriction and specificity,TCRs to respond to homeostatic cues.thereby allowing us to visualize selection processes (vonBoehmer, 1990), but such strategies generally permitIntroductionthe assessment of only a single receptor at a time. PCR-based analyses that resolve TCRs on the basis of CDR3Essentially all cells in multicellular organisms exchangelengths have addressed the bulk repertoire and havesignals, typically monomolecular and structurally invari-been particularly useful in highlighting recurrent amplifi-ant. T lymphocytes differ, however, in that the principalcations in the context of immune responses (Pannetiercues governing their differentiation and homeostasis areet al., 1995). Some brute-force sequencing studies havereceived through a somatically variable receptor thatattempted to describe the diversity of the VDJ joins ofrecognizes variable ligands composed of a diversity ofsingle Vb regions in thymocytes or in peripheral CD41

small peptides complexed with polymorphic MHC mole-and CD81 T cells (Candeias et al., 1991; Arstila et al.,cules. This mode of cell:cell communication imposes1999), but the results have revealed only subtle corre-important constraints that predicate some critical fea-lates, probably because of the compensating influences

tures of T cell behavior: MHC restriction (or the matchingof Va elements. The difficulty has been side stepped

of an individual’s complements of T cells and MHC mole-with single-chain tg mice expressing the b chain of an

cules) and positive selection (or the learning process antigen-specific TCR (Sant’Angelo et al., 1997, 1998). Aby which T cells maturing in the thymus acquire this significant proportion of T cells in these animals is reac-matched repertoire) (Fink and Bevan, 1978; Zinkernagel tive to the cognate peptide, and these cells frequentlyand Doherty, 1997). Furthermore, MHC molecules may use TCRa chains that are the same as or similar to thealso mold the T cell repertoire postthymically, as these parental clone. In addition, the presence or absence ofmolecules seem necessary for the survival and homeo- the parental sequence motif can be used as a markerstasis of naive cells in the peripheral lymphoid organs in differentiating thymocytes.(Goldrath and Bevan, 1999a). In the present report, we have attempted to obtain a

The TCR is composed of an a and a b chain, each broad but manageable view of the selection and evolu-encoded by variable, randomly rearranged V and (D)J tion of the T cell repertoire by engineering a mouse line

in which T lymphocytes are polyclonal but TCR diversity† To whom correspondence should be addressed (e-mail: cb@igbmc. is focused. This was achieved with a TCRa minilocusu-strasbg.fr [C. B.], [email protected] [D. M.]). transgene composed of a single Va region and two Ja‡ Present address: Instituto de Biologia Molecular e Celular, Porto, elements separated by an artificial rearrangement sub-4150, Portugal.

strate. This transgene was combined with a mutation§ Present address: Section on Immunology and Immunogenetics,that inactivates the endogenous TCRa locus and withJoslin Diabetes Center, Department of Medicine, Brigham and Wom-a TCRb transgene that, due to the dictates of allelicen’s Hospital, Harvard Medical School, One Joslin Place, Boston,

Massachusetts 02215. exclusion, prevents the rearrangement of endogenous

Page 2: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity22

Figure 1. The TCRa Minilocus Used to Generate Mice with Limited TCR Diversity

(A) Va2Var construct. The artificial rearrangement substrate is composed of the Va2.3 and Ja26 elements from the OT-1 TCRa chain, separatedby a stuffer sequence flanked by RSS signals (see Experimental Procedures). This fragment is placed between the XmaI and SacII sites ofthe pTCRaCass vector (Kouskoff et al., 1993). Rearrangement will lead to multiple CDR3a variants of the same VJ combination.(B) Total thymic RNA from two independent tg founder lines was amplified by RT–PCR with Va2.3- and Ja26-specific primers, and the productswere electrophoresed with or without prior digestion by PvuII, which distinguishes tg from endogenous transcripts; the band corresponds toamplification of the rearranged VJ segment.(C) The transgenes present in the Limited mice, combining the Va2Var transgene, the OT-1 TCRb chain transgene (Correia-Neves et al., 1999),and the TCRa knockout mutation.

TCRb components. We expected that in the resulting arrangement motifs (RSS) separated by a 550 bp spacer(Figure 1A). This construct does not encode a viable TCR“Limited” mice a repertoire of joining region sequences

would be generated that was broad enough that T cells as such but, after VJ rearrangement, should generate anumber of variants differing in the junctional CDR3 re-with different MHC restrictions and diverse antigen

specificities would be produced but that was con- gion. The sequences were chosen such that a directrearrangement would be out of frame, and so, nucleotidestrained enough that influences on repertoire selection

would not be too obscured. What proportion of a:b TCR additions or deletions are necessary to generate a func-tional product. (Regenerating the OT-1 TCRa itselfpairs can actually be selected? How much is the reper-

toire shaped by selection in the thymus versus preferen- would require a five-base deletion of the J segment.) Asilent mutation introduced a novel PvuII site into the Jatial amplification and maintenance in the periphery?

Starting from an MHC class I–restricted TCR, what alter- region to permit us to distinguish transcripts emanatingfrom the normal germline elements. This DNA fragmentations would permit it to recognize class II molecules?was placed into the TCRaCass vector that drives ex-pression in trangenic mice under the normal TCRa regu-Resultslatory elements and provides an additional Ja2 segment(Kouskoff et al., 1993). This other Ja segment was in-Our approach to investigating the T lymphocyte reper-

toire was to generate tg mice in which T cells expressed tended as a control to guard against the results beingoverly biased by preferential regeneration of the originala diversity of TCRs but with only focused variability. The

structure of the diverse TCRs could then be correlated OT-1 VJ combination.Several tg founder lines were obtained after injectionwith MHC class specificity, MHC restriction, and anti-

genic peptide specificity. We started from the well-stud- of this construct into B63SJL F2 embryos, identified bySouthern blot analysis (data not shown). A few linesied OT-1 TCR, Kb restricted, specific for the OVA257-

264 peptide, and composed of Va2.3Ja26 and Vb5Jb2.6 that harbored only a single transgene integrant wereselected to avoid later problems with the cells havingchains (Kelly et al., 1993; Hogquist et al., 1994). The

TCRa gene sequence was converted into an artificial rearranged several independent genes. The ability ofthe transgene to rearrange and be expressed in T cellsrearrangement substrate by inserting canonical re-

Page 3: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Thymic and Peripheral TCR Selection23

Figure 2. The Va2Var Minilocus Is Functional and Compensates a TCRa Deficiency

(A) Thymic expression. In this representative experiment, thymi from three mice were compared by four-color flow cytometry: a homozygousTCRa knockout (Cao/o), a Va2Var transgenic (also Cao/o homozygous), and a wild-type reference mouse. The CD3 histogram (left panels)identifies the CD3hi cells, whose CD4/CD8 profile is shown (middle panels); the staining of gated CD41CD82 and CD42CD81 SPs (whenpresent) for the transgene-encoded Va2 is shown in the right panel. (Staining in the wild-type sample corresponds to expression of the normalVa2 family members.)(B) Presence of T cells of both CD41 and CD81 phenotypes in lymph nodes from Va2Var/Ca0/0 mice.(C) CD4/CD8 profiles of CD31 lymph node cells from Va2Var/Ca0/0 mice, with or without the Vb5 OT-1 transgene, compared with a tg mouseexpressing both OT-1 a and b chain genes.(D) Evolution of the frequency of CD41 and CD81 cells in lymph nodes of young Limited (Va2Var/Ca0/0Vb51) mice.

was evaluated by RT–PCR analysis of thymus RNA, us- genes, whose expression was undetectable by directstaining (data not shown). The experimental strategying primers in the Va2.3 and Ca regions. Digestion of the

amplification products by PvuII revealed the diagnostic was to then examine cells from different T cell popula-tions by single-cell sorting, RT–PCR amplification, andshortening of the fragment in some founder mice but

not in negative littermates (Figure 1B). This indicated high-throughput sequencing of the CDR3a stretch.Large databases of such sequences were compiled andthat the artificial rearrangement substrate was indeed

functioning. Animals with this Va2Var transgene were analyzed.We first asked where transgene expression could bethen bred appropriately to introduce the OT-1 TCRb

chain transgene (Correia-Neves et al., 1999) and the detected and what types of T cells could be selected.As previously described (Philpott et al., 1992), no matureTCRa knockout mutation (Cao; Philpott et al., 1992). The

resulting animals (hereafter referred to as Limited mice) CD4182 or CD4281 ab thymocytes were selected in thethymus of Ca0/0 controls, in the absence of a functionalhave T cells whose TCRs exhibit only limited diversity:

(1) the TCRa chains derive only from the transgene, endogenous TCRa locus (Figure 2A, top). Introductionof the Va2Var transgene promoted selection of distinctwith its single Va and two Ja segments, and therefore

diversity is restricted to CDR3; and (2) the transgene- populations of CD41CD82CD31 and CD42CD81CD31

cells (Figure 2A). As expected, all expressed Va2 (Figureencoded OT-1 TCRb chain is expressed in .95% ofcells, preventing rearrangement of any endogenous b 2A, right). The number of mature CD4 and CD8 lineage

Page 4: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity24

cells averaged 0.5% and 0.2% of total thymocytes, re- mRNA. The remaining 266 sequences represented 212individual entities with few repeats, most of these beingspectively, or z1/20th the number observed in wild-type

control littermates (Figure 2A, bottom). This frequency intrapool and thus likely to correspond to duplicate clon-ing of the same sequence. The in-frame sequences en-is quite similar to that of T cells expressing any given

single Va. coded 164 distinct CDR3a motifs, none of which werepresent in more than 3% of the sequences. The OT-1-On the other hand, consistent with the fact that the

Va2Var transgene requires rearrangement for expres- derived Ja26 segment was used slightly less frequentlythan the Ja2 segment. Variable numbers of deletionssion, we did not detect the aberrant levels of TCR in

CD42CD82 cells that are usually observed with prere- and N nucleotide insertions (on both the V and J side)were observed (Figure 3B, bottom), with a distributionarranged TCR transgenes. Activation and maturation

markers (CD69, heat shock antigen, and peanut aggluti- quite similar to what is seen in naturally rearranged TCRachain genes (average deletion of two to three bases onnin) were also normal in the populations undergoing

positive selection (data not shown). either the V or J segment; average addition of three Nnucleotides). A few sequences repeated between differ-Since mature cells of both the CD4 and CD8 single-

positive (SP) phenotype were found, the TCRs gener- ent mice were present, but these represented a minority.ated from the artificial substrate could apparently asso-ciate with both MHC class I and II molecules. These The Postselection Repertoirecells were exported to peripheral lymphoid organs (Fig- To investigate the impact of thymic selection and periph-ure 2B), resulting in sizeable T cell populations of both eral homeostasis, we next determined the sequencesphenotypes, with a rough equivalence of CD41 and of TCR CDR3s expressed by sorted postselection TCD81 cells. This ratio was altered somewhat when the cells, analyzing large numbers of cells to permit a validOT-1 TCRb chain transgene was introduced, completing view of the repertoire. This analysis was performed bythe Limited mouse (Va2Var, Vb5, and Cao traits; Figure single-cell sorting and amplification in order to obtain2C), with an increase in the relative proportion of CD81 reliable information on repeat frequencies (which is lesscells. This increase was expected, as the OT-1 TCR is accurately determined by sorting of and cDNA amplifica-class I restricted. Interestingly, the proportion of CD41 tion from pools of cells, where repeats can be artifactual)cells changed with the age of the Limited mice (Figure and to avoid scrambled sequences generated by “chi-2D). Thus, the combination of transgenes in the Limited meric PCR” amplifications from bulk samples. The fourmouse seemed operational, generating TCRs with suffi- populations studied were (1) mature CD42CD81 and (2)cient variability to allow positive selection in the thymus CD41CD82 thymocytes and (3) CD41 and (4) CD81 lym-and maintenance in the periphery, directed by either phocytes from pooled inguinal and axillary lymph nodes.MHC class I or class II molecules. All cells were sorted with strict gates on Vb5hi cells in

order to eliminate cells that might be expressing anadditional Vb chain through escape from allelic exclu-The Preselection Repertoire

To analyze the TCR diversity generated in preselection sion. Limited mice were analyzed at 5–6 weeks of age,a time when the CD41 and CD81 populations appear topopulations, we determined the CDR3a sequences of

TCRs from immature thymocytes of Limited mice. Small, have stabilized. This early time was also chosen to avoidcomplications due to the mtv-8/mtv-9 superantigens,resting CD41CD81 (double-positive, DP) cells express-

ing a low but detectable level of surface CD3 (Figure which affect Vb5-expressing cells in older mice (Dungeret al., 1996). Single cells from three mice were sorted3A) were examined in order to ensure that the analysis

focused on products of the Va2Var transgene able to into wells of 96-well plates, and Va2 transcripts wereamplified from each cell. PCR products were purifiedpair with the Vb5 chain but without biases induced by

positive selection. Cells from three mice were sorted and sequenced directly on high-throughput sequencers.The frequency of positive RT–PCR reactions from singleinto six independent pools. (In this analysis, RT–PCR

and sequencing were performed on independent pools cells was quite high (30%–80% positive wells in variousexperiments), such that the data sets are not likely toof sorted DPs, rather than on single cells as in all of the

subsequent experiments, because the relative rarity of represent preferential amplification of rare sequencesand do give an accurate representation of the repertoireVa2Var rearrangements in DP cells prevented us from

obtaining large numbers of sequences by the single- in these mice. The complete sequence data from z200cells of each population are shown as supplementalcell approach.) RNA was prepared from each pool, the

sequences derived from the Va2Var transgene amplified data and are presented in compiled form in Figure 4A.Several points can be made from this set of se-with primers located in the Va2 and Ca segments, and

the products cloned into pBS plasmid for sequencing. quences. First, the repertoire of these selected and fullymature populations is markedly less diverse than thatTwo hundred and eighty-three independent clones were

sequenced, of which an illustrative 95 derived from two of preselection DPs. Of the 133 sequences seen in theDP sample, only 23 are also detected in postselectionindependent pools of a single mouse are presented in

Figure 3B; the complete set of sequences can be found cells. Among the 785 sequences obtained in selectedcells, there are 216 distinct CDR3a motifs, whose distri-as supplemental data at http://www.immunity.com/cgi/

content/full/14/1/21/DC1. bution is clearly nonrandom, 33 sequences being foundfive times or more in the compiled data sets. SomeClearly, rearrangement of the Va2Var transgene was

capable of generating a diversity of sequences. Only sequences are represented 40 times or more; certain ofthese were also found in immature DPs but only at a17/283 were out of frame; this low number was most

likely attributable to the reduced stability of untranslated low frequency, indicating that they do not correspond

Page 5: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Thymic and Peripheral TCR Selection25

Figure 3. Rearrangement of the TCRa Minilocus Generates CDR3a Diversity in Thymic Precursor Cells

Preselection DP thymocytes from Limited mice were sorted as expressing low levels of CD3 (two independent pools from three differentmice). The sequence of the CDR3a joining region was determined after RT–PCR and cloning (283 sequences). All sequences from a singlemouse are illustrated here (for complete sequences, see supplemental data), separated into those utilizing Ja26 and Ja2. In parentheses arethe number of times individual sequences were observed in the data set, denoted “a” when observed in two independent pools from thesame mouse and “b” when present in different animals. o-f, out of frame. Histograms at the bottom indicate the numbers of nucleotidedeletions on the V segment, N additions, and deletions on the J segment as a result of the join.

to favored rearrangements (supplemental data; M. C.-N. only one of the lineages, consistent with restriction byeither MHC class I or II molecules. This point is illustratedet al., submitted). These frequent sequences are “pub-

lic,” (i.e., they were found with surprisingly reproducible in the graph of Figure 4B, which represents the fre-quency of the CDR3a sequences most commonly ob-frequencies in all three mice examined) (Table 1). Sec-

ond, there are clear distinctions among the repeated served in the different populations. The sequencesfound at the top are essentially exclusive to the CD41sequences. They are not distributed evenly between the

CD41 and CD81 compartments, most being found in lineage—those on the bottom to the CD81 lineage. How-

Page 6: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity26

Page 7: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Thymic and Peripheral TCR Selection27

Table 1. Intermouse Reproducibility of Repeat SequenceFrequencies

CD41 CD82 CD42 CD81

Mouse number 57 63 91 57 63 91

CAAMDDNYQL 8 9 28 0 1 0CAALDDNYQL 6 5 16 0 0 0CAASKGDNYQL 1 0 10 0 0 0CAAHDNYQL 1 1 3 0 0 0CAALMDDNYQL 0 0 0 1 1 11CAASLNTGGLSGKL 0 0 1 1 2 7CAALNTGGLSGKL 0 0 0 6 5 4CAASRGDNYQL 0 1 2 2 1 5Total sequenced 41 33 113 31 43 98

ever, a few of the sequences occur in both compart-ments. The third point that comes out clearly from Figure4B is that extensive changes in frequencies have oc-curred between the thymocyte populations and theirperipheral counterparts. For example, the G-Ja26 andS-Ja26 sequences make up, together, 54% of the pe-ripheral repertoire of CD81 cells but are found in only 8%of thymocytes. Conversely, some sequences abundant

Figure 5. Activation Markers in Lymph Node Cells of Limited Miceamong mature thymocytes are essentially absent fromHistograms of gated CD41 and CD81 lymph node cells from a Lim-the peripheral cells (for example, MD-Ja26 or several ofited or a control mouse, stained for CD44 and CD62L late activationthe sequences specific to the CD81 lineage).markers and for CD69, an early marker.

These marked divergences between the thymic andperipheral repertoires could be due to expansion of par-ticular clonotypes induced by an immune response or transgene were associated with a wide range of Vb ele-to favored homeostatic expansion and survival as a con- ments. As shown in Figure 6, the correlations describedsequence of interaction with self-MHC:peptide com- above were largely absent in this case. Interestingly,plexes (Goldrath and Bevan, 1999a). If the latter, one several of the sequences found prominently in Vb51

might expect to observe the peculiar phenotypic change mice were again observed in this data set but with dif-in late activation markers that has been described for fering lineage specificity. These results indicate that theT cell populations homeostatically expanding (Goldrath MHC class distinctions imparted by CDR3a are condi-and Bevan, 1999b). This was indeed the case (Figure tioned by the Vb contribution.5); CD81 cells from Limited mice exhibited an upregula-tion of CD44 without the concomitant downregulationof CD62L normally observed with activated cells. On DiscussionCD41 cells, however, both markers were altered, andCD69 was also expressed, suggesting that homeostatic The data presented above provide a broad view of the

generation and evolution of the T lymphocyte repertoire,expansion may not have the same phenotypic conse-quences in CD41 and CD81 cells. encompassing T cell selection in the thymus and coloni-

zation of the peripheral lymphoid organs. We have takenAs discussed in detail below, the sequences in Figure4 that reflect recognition of MHC class I versus II mole- as an approach a modification of the “single-chain TCR

transgenic” strategy that has already proven so powerfulcules are only subtly different—single amino acid re-placements being enough to switch the class specificity. (Jorgensen et al., 1992a; Sant’Angelo et al., 1998), fur-

ther constraining diversity by limiting the variability ofWe wondered whether these were intrinsic propertiesdominantly imparted by the CDR3a sequence and what the second TCR chain. The combination of transgenes

in the Limited mouse performed as had been hoped for,could be the impact of TCRb chain elements. Therefore,we analyzed CDR3a sequences from mice lacking the yielding a naive repertoire broad enough for analytical

significance but of a manageable size such that impor-OT-1 Vb5 transgene, in which products of the Va2Var

Figure 4. CDR3a Sequences in Positively Selected Cells in Limited Mice

(A) Single cells from thymus and lymph node (TH and LN) of Limited mice were sorted (as Vb5hi) and their CDR3a sequence determined. Thedata from 785 cells are grouped according to CDR3a size, with the number of occurrences in each population shown. For clarity, not allamino acids are written out; those shared by most sequences are shown at the top. Sequences that were found repeatedly in different miceand compartments (.12 occurrences in the combined data set) are highlighted. Red, sequences specific to the CD41 lineage; green, sequencesspecific to the CD81 lineage; blue, sequences found for both; and purple, sequences exclusively found in peripheral CD41 lymphocytes andnever in the thymus. The sequences also found (single occurrences) in preselection thymic DPs are shown.(B) Schematic representation of the frequencies of repeated sequences in the four subpopulations. For clarity, only the distinguishing aminoacids are listed (e.g., Y-Ja26 denotes a CAAYDNYQL sequence); refer to Figure 4A or supplemental data for complete sequence. Color codingas in (A).

Page 8: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity28

pairing and reconstitution of the parental OT-1 TCR. Theexperimental strategy relied on TCRa gene transcriptamplification and sequencing from sorted single cells;the frequencies of individual sequences can thus berelied on. The observed frequency distribution, withmany repeated sequences in the postselection reper-toire, allows one to follow the fate of T cells expressinga number of different sequences in a manner that beginsto be significant. This point is bolstered by the fact thatthese frequent sequences are shared by all three miceanalyzed (Table 1) and are thus public specificities. Thisbroad view of T cell repertoire formation and flux leadsto novel conclusions along three different lines.

Repertoire Selection in the ThymusCombined positive and negative selection in the thymusresults in a T cell repertoire that is very different fromthe “raw” repertoire generated by random TCR generearrangements. We have demonstrated that, startingfrom a rather “flat” collection in which very few se-quences are repeated, these selection processes resultin a repertoire of mature thymocytes expressing a num-ber of unique sequences but dominated by many in-stances of multiply reiterated sequences. These reitera-tions make up an impressive proportion of the maturethymocyte repertoire in the Limited mouse; 63% ofCD41CD82 and 69% of CD42CD81 cells express one ofthese sequences, defined as occurring 12 times or morein the data set (supplemental data); the single mostabundant accounts for 25% of CD41CD82 thymocytes.This dominance is even more striking when one realizesthat many of these repeats are very closely related insequence (Figure 4B) (for example, SLNT-Ja2, STNT-Ja2, SGNT-Ja2, and SMNT-Ja2). These repeats are notdue to favored molecular rearrangements, as they arenot particularly frequent in the preselection repertoireof DP cells and because they are encoded by differentdegenerate nucleic acid sequences (supplemental data).They are also very unlikely to reflect trivial PCR contamina-tion artifacts, as drastic measures were taken duringsingle-cell sorting and PCR to prevent and diagnosecontamination problems.

Rough estimates suggest that these repeated se-quences are present at frequencies in the 1/106 range—amarked departure from the estimated frequency of1/1014 for any randomly generated TCR sequence com-bination. This highly nonuniform repertoire is consistent

Figure 6. CDRa Sequences in Va2Var Mice Devoid of the TCRb with and enhances previous observations (Sant’AngeloTransgene

et al., 1998). Employing a single-chain TCRb tg mouseSequences from CD41 and CD81 lymph node cells of three Va2Var/ line, it was found that the “parental” CDR3a sequenceCa0/0 mice, from 175 sorted single cells. Sequences and their relative

occurred at high frequency in a sequence set derived bydistribution are represented as in Figure 4A. Those sequences pre-amplifying cDNA from sorted CD41 cells for the Va2Ja11viously identified in the analysis of Limited mice are highlighted.segments of the original T cell clone. Our findings aredifferent in that most of the repeated sequences wefound diverge from the TCR of origin; they use two differ-tant influences could be discerned. A wide array of se-

quences was generated by the rearrangement process ent J regions, and the TCRs they encode engage differ-ent MHC molecules. Thus, the caveat of a speciallyfinalized in DP thymocytes, but only a limited subset

was maintained in the mature thymocyte pools. The favored selection of the parental sequences does notapply.combination of Va2 and Vb5 regions encoded by the

transgenes allowed sufficient flexibility for recognition We cannot formally rule out that these repeats reflectpreferential proliferative amplification of a subset of ma-of either MHC class I or II molecules. Both Ja segments

were utilized, and the conclusions apply to sequences ture thymocytes in the Limited tg mice. However, BrdUincorporation experiments have shown that the prolifer-using either of them, ruling out biases due to preferential

Page 9: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Thymic and Peripheral TCR Selection29

ation of CD41CD82 and CD42CD81 cells from Limited ment by MHC molecules to survive in the periphery aswell as for homeostatic expansion (Goldrath and Bevan,and normal mice is about the same (data not shown).1999a). Since selection in the thymus is strongly influ-Since cell division in normal mice seems to correspondenced by abundant self-peptides and since the relativeto short rounds of cycling immediately prior to exportcontribution of these peptides is likely to be somewhat(Ernst et al., 1995; Penit and Vasseur, 1997), it would bedifferent in cells of the peripheral organs, one wouldunlikely to account for the numbers of repeated se-expect the peripheral peptidic self to exert new pres-quences we observe here. Furthermore, elevated num-sures on the repertoire. Thus, the profound changes webers of these repeated sequences are found in the earli-observed between the mature thymic repertoire and itsest transitional populations in the thymus (CD3hi DPsperipheral descendant probably reflect secondary mold-and CD41CD8int) (M. C.-N. et al., submitted). Thus, theing by an altered complement of self-peptides (Ernst etdata suggest that it is the positive selection processal., 1999), differentially modulating the expansion and sur-itself that generates this “bumpy” repertoire. DP cellsvival of individual T cells.expressing particular CDR3a sequences might have

It is unlikely that these changes are due to amplifica-quite different probabilities of being selected rather thantion during background-level immune responses (1) be-having a simple yes/no choice, and these probabilitiescause they were detected in three different mice sam-may result in a spectrum of frequencies in the repertoire.pled at spaced times, (2) because some correspond toExtrapolating from the particular context of the Lim-sequences already observed in the context of an OVA-ited mouse, the implication is that a repertoire selectedspecific immune response in a mouse colony located inin the thymus contains, for any VaJaVbJb combination,another hemisphere (Kelly et al., 1993), and (3) becausetwo categories of sequences: some present at low fre-most of those repeats that were expanded in the periph-quencies and others present in many cells. We proposeery correspond to sequences already overselected inthat this stems from a probabilistic aspect of positivethe thymus. In other words, the peripheral interactionsselection. Depending on the affinity/avidity for self-continue but remold selective processes begun in theMHC, some TCRs lead to highly probable selection,thymus. It is also unlikely that the results are influencedwhile others confer only a limited probability. These over-by superantigens, as the reported effects of mtv-8 andselected TCRs may reflect positive selection on abun-mtv-9 on Vb51 cells are only apparent in mice betweendant MHC:peptide complexes—a prediction consistent15 and 30 weeks of age (Dunger et al., 1996).with the loss of “canonical” responses when moth cyto-

Limited mice have a low thymocyte output becausechrome c–specific T cells were selected on experimen-T cells in these animals rely on the rearrangement of atally introduced neopeptides displayed on thymic epi-single Va substrate. Hence, there is more homeostaticthelial cells (Nakano et al., 1997). It would be interestingexpansion than would normally be observed, and a goodto test this notion using the various “single-peptide”proportion of the CD81 cells exhibit the particular “ho-mice that have been derived over the last few years.meostatic expansion” phenotype described by GoldrathThe dominant selection of a relatively limited numberand Bevan (1999b), with disjuncted expression of theof TCRs cannot be without consequences on immuneCD44 and CD62L late activation markers. The implica-function. It is tempting to speculate that preferentiallytion from our data is that the degree of homeostaticselected repeats underlie the canonical motifs that oftenexpansion can vary widely for cells with different TCRs.characterize the responses to particular peptides, suchThese data extend recent reports (Ernst et al., 1999;as the moth cytochrome c 88–103 and myelin basicFerreira et al., 2000) showing that cells expressing differ-protein 1–11 peptides (Acha-Orbea et al., 1988; Jorgen-ent tg TCRs were variably responsive to homeostaticsen et al., 1992b). The presence of grossly overrepre-cues. It is not known at present how much of a rolesented TCRs in the repertoire might also explain thehomeostatic expansion or differential survival plays inimmunodominance of particular peptides in complexa normal immune system, although it is suggestive that

proteins (Bousso et al., 1998; Wallace et al., 2000)—one of the amplified sequences is precisely that of OT-1

immunodominance that cannot always be explained byitself (S-Ja26), already found repeatedly in normal mice.

processing considerations or by enhanced binding to It will be important to determine how much of the reper-MHC molecules (e.g., Vitiello et al., 1996). toire remolding we observed also occurs in the context

of a “full” immune system.Repertoire Molding in the Periphery A special mention must be made of three peculiarOnce shaped by selection in the thymus, the T cell reper- sequences found repetitively in the peripheral CD41

toire is subject to further molding in the peripheral compartment of all mice (Figure 4). They are very similarlymphoid organs, graphically illustrated in Figure 4B. to each other and have uncharacteristically long CDR3sThe dominant sequences still occur, but their relative with very conserved nucleotide sequences (nine N addi-prevalence changes considerably—some are virtually tions identical or highly similar in all cases and in theabsent from the peripheral pool; others become mark- different mice; see supplemental data). They are con-edly more frequent. These general patterns were ob- spicuously absent from the over 1000 sequences de-served reproducibly in three individual mice (Table 1; rived from various thymic populations analyzed in thesupplemental data). It is possible that some of the thy- course of this project (this paper and M. C.-N. et al.,mus/periphery differences are due to preferential export submitted). It is tempting to speculate that these se-from the thymus, but it seems more likely that they repre- quences reflect extrathymic differentiation, perhaps re-sent differential homeostatic expansion and mainte- lying on an unusual form of rearrangement related to thenance in the periphery. It has become clear in the past DIR motifs observed in immunoglobulin rearrangements

(Tuaillon and Capra, 1998).few years that T lymphocytes require continued engage-

Page 10: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity30

In conclusion, the analyses performed on the Limited peptides presented by MHC molecules during positiveselection in the thymus, rather than by contacts withmouse line clearly demonstrate that the repertoire ofthe MHC molecules themselves. A determining influencenaive T cells available to participate in an immune re-for peptide interactions is also consistent with resultssponse is far less diverse and flat in frequency thanfrom studies on Va families used preferentially by CD41previously thought and that it is molded in a similarlyor CD81 cells; the positions found to be crucial for theseimportant manner by thymic selection and peripheralbiases, 51 for Va3 and 30 for Va2 (Sim et al., 1996;expansion/maintenance forces. In the future, these miceCorreia-Neves et al., 1999), participate in CDR1 andshould prove valuable for investigating the fine specific-CDR2 but are centrally located in the crystal structuresity of TCR/MHC:peptide interactions.and are likely to engage primarily in peptide contacts.

These data also support the notion that MHC classMHC Class Specificity of the TCRspecificity is not solely imparted by the CDR1 and CDR2The cardinal observation is that when diversity is limitedregions of the TCR, as results from some past studiesto such an extent there are very clear differences be-(Sim et al., 1996, 1998) are often taken to indicate, buttween sequences selected into the CD41 versus CD81

that CDR3 also plays a major role (Correia-Neves et al.,lineages—by inference, primarily restricted by MHC1999). In addition, the very fact that the Va2Vb5 TCR ofclass II versus class I molecules (these assignmentsthe Limited mice allows balanced selection into bothare confirmed in M. C.-N. et al., submitted). Consistentthe CD41 and CD81 compartments indicates that withpatterns emerge from the data compiled in Figure 4A.a fixed CDR1 and 2 there is still great leeway for CDR3For example, all CDR3s of LX-Ja26 type are class IIto fix MHC class preference. It should be kept in mindrestricted, irrespective of the nature of “x”. There arethat the experiments of Sim et al. (1996, 1998) focusedalso CDR3 length biases: eight-residue CDR3s are moreon CDR1 and 2 differences and thus could only identifyfrequent in CD41 cells, and ten-residue CDR3s are moreinfluences within these regions; in the present experi-frequent in CD81 cells. However, no overriding rulements, we have concentrated on CDR3, as CDR1 andemerges, and minute sequence differences can switch2 are fixed; all three CDRs must have a role to play.MHC class preference. The clearest example can be

A few sequences were found in both CD41 and CD81found in short CDR3s of the form X-Ja26. Bulkier Y, H,

cells from Limited mice in the thymus or periphery (egL, and K residues at the X position are only found inSKG-Ja26 or LNT-Ja2). In hindsight, it may not be tooCD41 cells, while the smaller S, G, I, C, D, or V residuessurprising that such sequences exist. There must be aoccur in CD81 cells. There are many other examples indegree of evolutionary selection for general congruencethe data of Figure 4 of single amino acid differencesbetween the TCR and MHC molecules, and several au-distinguishing MHC class I– and class II–restrictedthors (Blackman et al., 1986; Gilfillan et al., 1994; GavinTCRs. Thus, much as TCR/MHC:peptide interactions areand Bevan, 1995) have demonstrated that the unse-exquisitely sensitive to variations in peptide sequence,lected repertoire has a high level of overall compatibilitysimilar discrimination applies to TCR recognition of MHCwith any given MHC molecule; in some estimates, up toclass. Generalizing from their crystal structure of the30% of unselected TCR sequences can interact with an

Ak-restricted D10 TCR, Reinherz and coworkers haveMHC molecule encountered in the thymus. Thus, one

proposed that the TCR binds MHC class II:peptide com-might expect in a Limited mouse expressing Ab and

plexes in a spatially distinct more orthogonal orientationKb/Db that up to 10% of sequences could engage both

than they bind MHC class I:peptide complexes (Reinherz MHC classes, at least before negative selection takeset al., 1999). Such a dichotomy is difficult to reconcile its toll. In fact, the proportion of such cells we observedwith the overlapping patterns and the effects of minor was rather lower, and most of the shared sequencesdifferences observed here. Our results are more in line showed distinct class bias. However, this number maywith Wilson’s proposal that the D10 structure represents be an underestimate, since we have found that, whenone extreme in a spectrum of subtly different conforma- the Limited mouse is rendered MHC class II deficient,tions (Wilson, 1999). This view is also consistent with some of the sequences normally picked up only in CD41

the great diversity of TCR/MHC contacts that has been cells are also detected in CD81 cells, e.g., the H-Ja26observed with the various structures (Garboczi et al., sequence (M. C.-N. et al., submitted).1996; Garcia et al., 1996; Ding et al., 1998; Teng et al., In the several crystal structures of TCR/MHC:peptide1998; Reinherz et al., 1999). It seems then that MHC complexes that have been published to date, the TCRclass specificity is imparted by a variable combination Va region has the most reproducible configuration andof contacts leading to congruence, rather than by differ- footprint, while there is far more divergence in the overallential docking and conserved TCR/MHC contacts. configuration of Vb, which in some structures appeared

Interestingly, the amino acid positions that condition to angle away from the MHC:peptide surface or to makeMHC class specificity in our system are those that inter- very few contacts (reviewed in Wilson, 1999). One mightact far more with peptide than with the MHC molecule have predicted then that the CDR3a sequence/MHCa helices, at least in the crystal structures that have class preference correlations we observed in the T cellbeen solved so far (Garboczi et al., 1996; Garcia et al., repertoire of the Limited mouse, with its fixed Vb5 chain1996; Ding et al., 1998; Teng et al., 1998; Reinherz et (Figure 4), might also have been seen, at least partially,al., 1999). For example, in the corresponding positions in the repertoire of mice expressing the panoply of Vbof the crystal structure of Reinherz et al. (1999), amino regions produced by rearrangement of endogenousacids 99–101 engage in 16 contacts with peptide but TCRb genes. But, this turned out not to be the case.only three with MHC molecule atoms. The implication Some of the more frequently repeated sequences were

found again, but this time in the other mature T cellis that MHC class specificity can be determined by the

Page 11: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Thymic and Peripheral TCR Selection31

and the resulting colonies were screened by hybridization with ancompartment—for example, S-Ja26 was present onlyoligo specific for the Va2 segments (GGAAGATGGACGATT).in CD81 cells displaying the OT-I Vb5 region but only

in CD41 cells when found in combination with diverseSingle-Cell Sorting and RT-PCRendogenously encoded Vbs.Thymocytes and lymphocytes were stained with anti-CD8a, anti-CD4 (Caltag Laboratories, Inc.), and MR9-4, specific for the Vb5Experimental Proceduresfamily (Bill et al., 1990). Individual cells were deposited into wellsof 96-well PCR plates containing 10 ml of RT buffer (50 mM Tris-DNA Construct and TransgenesisHCl, 75 mM KCl, and 3 mM MgCl2) with 2% Triton X-100, 1 mg BSA,The construct for generating TCRa chains with CDR3 variability500 mM dNTP, 50 ng of oligo(dT)12-18 (Pharmacia), 8 U RNasin,allowed recombination between Va2.3 and Ja26 as well as Va2.3and 30 U of Moloney murine leukemia virus reverse transcriptaseand Ja2 gene segments. The template containing Va2.3 and Ja26(Life Technologies, Grand Island, NY). The plates were incubatedwas made by PCR and introduced into the TCRa cassette vectorfor 90 min at 378C. For the first PCR round, 10 ml of the resulting(Kouskoff et al., 1993). This cassette contains the regulatory ele-cDNA were amplified by adding 40 ml of Taq buffer (50 mM KCl, 10ments for expression of the a chain, the constant region of the TCRamM Tris-HCl [pH 9.3], and 2.5 mM MgCl2) containing 2.5 U Taqchain as well as the Ja2 gene segment, downstream of the cloningpolymerase, 500 mM dNTP, 400 ng sense primer specific for thesites for the variable TCR genes. The Va2.3 and Ja26 gene segmentsVa2 family (CAGCAGCAGGTGAGACAAAGT), and antisense primerwere amplified from a plasmid containing the original OT-1 TCRaspecific for the TCRa constant region (GTTTTGTCAGTGATGAchain (clone 149.42 in Kelly et al., 1993). The Va2.3-Ja26 junctionalACGT) (3 min at 938C; 35 cycles with 508C annealing temperature).region from the original OT-1 TCRa chain was replaced by a re-For the second PCR round, 2 ml of the first amplification productarrangement substrate containing an RSS downstream of Va2.3were amplified under the same conditions in 50 ml of Taq buffer(CACAGTG.CTCTCCAGGCACCTGTAGCCCAT.ACCCAAACC) (Oka-containing 1U of Taq polymerase, 500 mM dNTP, 200 ng of sensezaki and Sakano, 1988) and the natural RSS from the Ja26 segment,AAGGCCCGGGTCTCTGACAGTCTGGGAAGGA, and AATCTGCAGseparated by the 547 bp sequence naturally found upstream of Ja26CGGCACATTGATTTGGGA nested primers (22 cycles). The PCR(Koop et al., 1992). A silent mutation was engineered in the Ja26products were purified by PEG (PEG 6000 20% and NaCl 2.5 M)segment to create a novel PvuII site (TATCAGCTGATCTGG). Theprecipitation followed by two washes in 75% ethanol. The PCRrearrangement substrate was assembled by PCR ligation with over-products were sequenced directly using one of the primers for thelapping primers (Va2.3-VaRSS-intersignal DNA-JaRSS-Ja26) andsecond round of PCR (AATCTGCAGCGGCACATTGATTTGGGA).cloned into the TCRa chain expression cassette (Kouskoff et al.,

Painstaking precautions were taken against PCR contamination,1993) using the unique XmaI and SacII sites.with 16 control wells per 96-well PCR plate in which a mock dropletThe construct was excised from plasmid DNA and injected intoof PBS was sorted. Plates showing any trace of positive amplifica-fertilized B63SJL F2 eggs. Screening of tg founders was performedtion in these control wells were discarded.on Southern blots with a 2.8 kb probe originating from the TCRa

cassette. Limited mice were generated by breeding mice carryingAcknowledgmentsthe transgene-encoded variable a chain with mice tg for the OT-1

TCRb chain (Correia-Neves et al., 1999) and again with knockoutWe thank Drs. F. Carbone, S. Chan, and D. Wiley for discussions andmice deficient for the expression of the normal TCRa chain (Philpottreagents, S. Vicaire, P. Brottier, and J. Weissenbach (Genoscope;et al., 1992), with subsequent intercrossing of progeny. All miceCentre National de Sequencage) for sequencing, F. Jeanmougin forused in this work were homozygous for the H-2b haplotype.help with data analysis, and P. Gerber, P. Marchal, J. Hergueux,All animals were maintained in the conventional facility of theand C. Ebel for technical assistance. This work was supported byInstitut de Genetique et de Biologie Moleculaire et Cellulaireinstitute funds from the INSERM, the CNRS, the Hopital Universitaire(IGBMC) under Ministere de l’Agriculture (Agrement 67227) and Eu-de Strasbourg, and by a grant from the Association pour la Re-ropean Economic Community guidelines.cherche contre le Cancer. M. C.-N. received fellowships from thePrograma Gulbenkian de Doutoramento em Biologia e Medicina andPCR Amplification to Test for the Rearrangementthe Fundacao Para a Ciencia e a Tecnologia.of the Variable a Chain

RT–PCR from lymph node RNA was performed with primers thatspecifically amplified cDNA containing a rearranged Va2 gene— Received August 31, 2000; revised December 11, 2000.sense primer (Va2), GTTCCCGGGATCGAGGAATGGACAAGATTCTG; reverse primer (TCRa constant), GGCCCCATTGCTCTTGGA

ReferencesATC. A second PCR round was performed using nested primersthat amplify Va2 gene segments (sense primer, TCCATACGTTCAG

Acha-Orbea, H., Mitchell, D.J., Timmermann, L., Wraith, D.C.,TGTCCGATAAA) recombined with Ja26 (antisense primer, GGCTausch, G.S., Waldor, M.K., Zamvil, S.S., McDevitt, H.O., andTTTATAATTAGCTTGGTCCCAGAG). The resulting PCR productsSteinman, L. (1988). Limited heterogeneity of T cell receptors from(200 bp) were digested with PvuII.lymphocytes mediating autoimmune encephalomyelitis allows spe-cific immune intervention. Cell 54, 263–273.PCR Amplification and Cloning of Va21 TCRa ChainsArstila, T.P., Casrouge, A., Baron, V., Even, J., Kanellopoulos, J.,from DP Thymocytesand Kourilsky, P. (1999). A direct estimate of the human ab T cellThymocytes were stained with anti-CD8a, anti-CD4 (Caltag Labora-receptor diversity. Science 286, 958–961.tories, Inc., Burlingame, CA), and KT3, specific for CD3 (Tomonari,

1988). Two independent pools containing 2 3 105 CD41CD81CD3low Bill, J., Kanagawa, O., Linten, J., Utsunomiya, Y., and Palmer, E.thymocytes were purified from each of three individual mice. HeLa (1990). Class I and class II MHC gene products differentially affectcells (106) were added to each sample as carrier, and RNA was the fate of Vb5 bearing thymocytes. J. Mol. Cell. Immunol. 4,prepared by NP-40 lysis. cDNA was synthesized using half of the 269–280.RNA from each sample in 40 ml of reverse transcription buffer (50 Blackman, M., Yague, J., Kubo, R., Gay, D., Coleclough, C., Palmer,mM Tris-HCl, 75 mM KCl, and 3 mM MgCl2). 10 ml of this reaction E., Kappler, J., and Marrack, P. (1986). The T cell repertoire may bewas used for the first round of PCR amplification. The PCR condi- biased in favor of MHC recognition. Cell 47, 349–357.tions used were the same as described previously (Correia-Neves

Bousso, P., Casrouge, A., Altman, J.D., Haury, M., Kanellopoulos,et al., 1999), except that only 20 cycles were performed for eachJ., Abastado, J.P., and Kourilsky, P. (1998). Individual variations inround, and the antisense primer for the second PCR round wasthe murine T cell response to a specific peptide reflect variabilitydistinct (TCTCGAATTCAGGCAGAGGGTGCTGTCC). The PCR prod-in naive repertoires. Immunity 9, 169–178.ucts were purified by phenol/chloroform extraction, digested with

XmaI and EcoRI restriction enzymes, purified on agarose gel, and Candeias, S., Waltzinger, C., Benoist, C., and Mathis, D. (1991). TheV beta 171 T cell repertoire: skewed J beta usage after thymiccloned into pBS. The ligation was transfected into XL1-blue bacteria,

Page 12: The Shaping of the T Cell Repertoire - Harvard University · The Shaping of the T Cell Repertoire ... (CNRS/INSERM/ULP) from its position at the junction of the V(D)J elements, 1

Immunity32

selection; dissimilar CDR3s in CD41 versus CD81 cells. J. Exp. Kouskoff, V., Fehling, H.J., Lemeur, M., Benoist, C., and Mathis, D.(1993). A vector driving the expression of foreign cDNAs in the MHCMed. 174, 989–1000.class II-positive cells of transgenic mice. J. Immunol. Methods 166,Correia-Neves, M., Waltzinger, C., Wurtz, J.M., Benoist, C., and287–291.Mathis, D. (1999). Amino acids specifying MHC class preference inNakano, N., Rooke, R., Benoist, C., and Mathis, D. (1997). PositiveTCR V alpha 2 regions. J. Immunol. 163, 5471–5477.selection of T cells induced by viral delivery of neopeptides to theDavis, M.M., and Bjorkman, P.J. (1988). T-cell antigen receptorthymus. Science 275, 678–683.genes and T-cell recognition. Nature 334, 395–402.Okazaki, K., and Sakano, H. (1988). Thymocyte circular DNA excisedDing, Y.H., Smith, K.J., Garboczi, D.N., Utz, U., Biddison, W.E., andfrom T cell receptor alpha-delta gene complex. EMBO J. 7, 1669–Wiley, D.C. (1998). Two human T cell receptors bind in a similar1674.diagonal mode to the HLA-A2/Tax peptide complex using differentPannetier, C., Even, J., and Kourilsky, P. (1995). T-cell repertoireTCR amino acids. Immunity. 8, 403–411.diversity and clonal expansions in normal and clinical samples. Im-Dunger, A., Cunningham, J.M., Delaney, C.A., Lowe, J.E., Green,munol. Today 16, 176–181.M.H.L., Bone, A.J., and Green, I.C. (1996). Tumor necrosis factor-aPenit, C., and Vasseur, F. (1997). Expansion of mature thymocyteand interferon-g inhibit insulin secretion and cause DNA damage insubsets before emigration to the periphery. J. Immunol. 159, 4848–unweaned-rat islets. Diabetes 45, 183–189.4856.Ernst, B., Surh, C.D., and Sprent, J. (1995). Thymic selection andPhilpott, K.L., Viney, J.L., Kay, G., Rastan, S., Gardiner, E.M., Chae,cell division. J. Exp. Med. 182, 961–971.S., Hayday, A.C., and Owen, M.J. (1992). Lymphoid development in

Ernst, B., Lee, D.S., Chang, J.M., Sprent, J., and Surh, C.D. (1999).mice congenitally lacking T cell receptor ab-expressing cells. Sci-

The peptide ligands mediating positive selection in the thymus con-ence 256, 1448–1452.

trol T cell survival and homeostatic proliferation in the periphery.Reinherz, E.L., Tan, K., Tang, L., Kern, P., Liu, J., Xiong, J., Hussey,Immunity 11, 173–181.R.E., Smolyar, A., Hare, B., Zhang, R., et al. (1999). The crystal

Ferreira, C., Barthlott, T., Garcia, S., Zamoyska, R., and Stockinger,structure of a T cell receptor in complex with peptide and MHC

B. (2000). Differential survival of naive CD4 and CD8 T cells. J.class II. Science 286, 1913–1921.

Immunol. 165, 3689–3694.Sant’Angelo, D.B., Waterbury, P.G., Cohen, B.E., Martin, W.D., van

Fink, P.J., and Bevan, M.J. (1978). H-2 antigens of the thymus deter- Kaer, L., Hayday, A.C., and Janeway, C.A., Jr. (1997). The imprintmine lymphocyte specificity. J. Exp. Med. 148, 766–774. of intrathymic self-peptides on the mature T cell receptor repertoire.Garboczi, D.N., Ghosh, P., Utz, U., Fan, Q.R., Biddison, W.E., and Immunity 7, 517–524.Wiley, D.C. (1996). Structure of the complex between human T-cell Sant’Angelo, D.B., Lucas, B., Waterbury, P.G., Cohen, B., Brabb,receptor, viral peptide and HLA-A2. Nature 384, 134–141. T., Goverman, J., Germain, R.N., and Janeway, C.A., Jr. (1998). AGarcia, K.C., Degano, M., Stanfiled, R.L., Brunmark, A., Jackson, molecular map of T cell development. Immunity 9, 179–186.M.R., Peterson, P.A., Teyton, T., and Wilson, I.A. (1996). An ab T Sim, B.C., Zerva, L., Greene, M.I., and Gascoigne, N.R.J. (1996).cell receptor structure at 2.5 A and its orientation in the TCR-MHC Control of MHC restriction by TCR Valpha CDR1 and CDR2. Sciencecomplex. Science 274, 209–219. 273, 963–966.Gavin, M.A., and Bevan, M.J. (1995). Increased peptide promiscuity Sim, B.C., Lo, D., and Gascoigne, N.R. (1998). Preferential expres-provides a rationale for the lack of N regions in the neonatal T cell sion of TCR V alpha regions in CD4/CD8 subsets: class discrimina-repertoire. Immunity 3, 793–800. tion or co-receptor recognition? Immunol. Today 19, 276–282.Gilfillan, S., Waltzinger, C., Benoist, C., and Mathis, D. (1994). More Teng, M.K., Smolyar, A., Tse, A.G., Liu, J.H., Liu, J., Hussey, R.E.,efficient positive selection of thymocytes in mice lacking terminal Nathenson, S.G., Chang, H.C., Reinherz, E.L., and Wang, J.H. (1998).deoxynucleotidyl transferase. Int. Immunol. 6, 1681–1686. Identification of a common docking topology with substantial varia-

tion among different TCR-peptide-MHC complexes. Curr. Biol. 8,Goldrath, A.W., and Bevan, M.J. (1999a). Selecting and maintaining409–412.a diverse T-cell repertoire. Nature 402, 255–262.

Tomonari, K. (1988). A rat antibody against a structure functionallyGoldrath, A.W., and Bevan, M.J. (1999b). Low-affinity ligands for therelated to the mouse T-cell receptor/T3 complex. Immunogen. 28,TCR drive proliferation of mature CD81 T cells in lymphopenic hosts.455–458.Immunity 11, 183–190.

Tuaillon, N., and Capra, J.D. (1998). Use of D gene segments withHogquist, K.A., Jameson, S.C., Heath, W.R., Howard, J.L., Bevan,irregular spacers in terminal deoxynucleotideyltransferase (TdT)1/1M.J., and Carbone, F.R. (1994). T cell receptor antagonist peptidesand TdT-/- mice carrying a human Ig heavy chain transgenic minilo-induce positive selection. Cell 76, 17–27.cus. Proc. Natl. Acad. Sci. USA 95, 1703–1708.Jameson, S.C., Kaye, J., and Gascoigne, N.R. (1990). A T cell recep-Vitiello, A., Yuan, L., Chesnut, R.W., Sidney, J., Southwood, S., Far-tor V alpha region selectively expressed in CD41 cells. J. Immunol.ness, P., Jackson, M.R., Peterson, P.A., and Sette, A. (1996). Immu-145, 1324–1331.nodominance analysis of CTL responses to influenza PR8 virus re-Jorgensen, J.L., Esser, U., Fazekas de St. Groth, B., Reay, P.A., andveals two new dominant and subdominant Kb-restricted epitopes.Davis, M.M. (1992a). Mapping T-cell receptor-peptide contacts byJ. Immunol. 157, 5555–5562.variant peptide immunization of single-chain transgenics. Naturevon Boehmer, H. (1990). Developmental biology of T cells in T-cell355, 224–230.receptor transgenic mice. Annu. Rev. Immunol. 8, 531–556.

Jorgensen, J.L., Reay, P.A., Ehrich, E.W., and Davis, M.M. (1992b).Wallace, M.E., Bryden, M., Cose, S.C., Coles, R.M., Schumacher,Molecular components of T-cell recognition. Annu. Rev. Immunol.T.N., Brooks, A., and Carbone, F.R. (2000). Junctional biases in the10, 835–873.naive TCR repertoire control the CTL response to an immunodomi-

Kappler, J.W., Roehm, N., and Marrack, P. (1987). T cell tolerance nant determinant of HSV-1. Immunity 12, 547–556.by clonal elimination in the thymus. Cell 49, 273–280.

Wilson, I.A. (1999). Perspectives: protein structure. Class-consciousKelly, J.M., Sterry, S.J., Cose, S., Turner, S.J., Fecondo, J., Rodda, TCR? Science 286, 1867–1868.S., Fink, P.J., and Carbone, F.R. (1993). Identification of conserved

Zinkernagel, R.M., and Doherty, P.C. (1997). The discovery of MHCT cell receptor CDR3 residues contacting known exposed peptiderestriction. Immunol. Today 18, 14–17.side chains from a major histocompatibility complex class I-bound

determinant. Eur. J. Immunol. 23, 3318–3326.

Koop, B.F., Wilson, R.K., Wang, K., Vernooij, B., Zallwer, D., Kuo,C.L., Seto, D., Toda, M., and Hood, L. (1992). Organization, structure,and function of 95kb of DNA spanning the murine T-cell receptor Calpha/C delta region. Genomics 13, 1209–1230.