Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 May 1.
Published in final edited form as: J Immunol. 2012 May 1;188(9):4135–4140. doi: 10.4049/jimmunol.1102661

The role of naïve T cell precursor frequency and recruitment in dictating immune response magnitude

Marc K Jenkins *, James J Moon
PMCID: PMC3334329  NIHMSID: NIHMS362591  PMID: 22517866

Abstract

Recent advances in technology have lead to the realization that the populations of naïve T cells specific for different foreign peptide:MHC (p:MHC) ligands vary in size. This variability is due in part to the fact that certain peptides contain amino acids that engage in particularly favorable interactions with TCRs. In addition, deletion of clones with cross-reactivity for self p:MHC ligands may reduce the size of some naïve populations. In many cases, the magnitude of the immune response to individual p:MHC epitopes correlates with the size of the corresponding naïve populations. However, this simple relationship may be complicated by variability in the efficiency of T cell recruitment into the immune response. The knowledge that naïve population size can predict immune response magnitude may create opportunities for production of more effective subunit vaccines.


It is well established that the magnitude of the primary T cell response is influenced by the amount and duration of presentation of the relevant peptide:MHC (p:MHC) ligands by APC in secondary lymphoid organs (1-4). Peptides that are derived from abundant proteins, or are processed efficiently, or bind strongly or stably to MHC are more likely to be displayed in larger amounts and for longer periods of time on APC than peptides that lack these properties (5, 6). Abundantly presented p:MHC complexes will then trigger more intense TCR signaling in cognate T cells than less abundant complexes, which will promote effector and memory cell formation. It is also possible, however, that responses to certain p:MHC ligands are strong because the cognate naïve T cell population is larger than average.

The latter possibility has been difficult to test because the frequency of T cells specific for individual p:MHC ligands is so low (7). This infrequency is a direct consequence of the vast number of αβ TCRs that can be produced by random joining of Tcra and Tcrb V (D) J segments and non-templated N-region additions (8). Thus, within the vast pool of TCRs displayed by individual cells in the naïve T cell repertoire, only a few are likely by chance to have high affinity for any individual p:MHC ligand. We now know that the frequency of such cells is at most about 100 cells per million naive T cells (Tables 1 and 2) (9). This low frequency together with the stringent activation requirements of naïve T cells explains why conventional 96 well plate proliferation assays containing ~106 T cells per well are incapable of detecting p:MHC-specific T cell populations in individuals who were not previously immunized (10).

Table 1.

Foreign p:MHC-specific naïve T cell frequencies in mice determined by tetramer-based cell enrichment

Source Epitope Total cells
per mouse1
Cells per
million
naive
CD4+ or
CD8+ T
cells2
Reference
MHCI
Ovalbumin OVA-257:Kb 600 36 (50)
170 10 (51)
130 8 (39)
70 4 (44)
LCMV gp-33:Db 449 27 (40)
287 17 (39)
NP-396:Db 151 9 (39)
117 7 (40)
L-2062:Kb 90 5 (40)
NP-205:Kb 57 3 (40)
gp-118:Kb 43 3 (40)
L-156:Kb 24 1 (40)
L-338:Db 15 1 (40)
MCMV M45:Db 1500 89 (52)
603 36 (39)
M57:Kb 900 54 (52)
m139:Kb 300 18 (52)
IE3:Kb 90 5 (52)
M38:Kb 80 5 (52)
Influenza virus NS2-114:Kb 282 17 (45)
PB1-F2-62:Db 225 13 (45)
PA-224:Db 120 7 (39)
68 4 (45)
64 4 (36)
NP-366:Db 36 2 (45)
18 1 (36)
Vaccinia virus B8R:Kb 1400 83 (50)
1100 65 (52)
1070 64 (51)
B8-20:Kb 320 19 (44)
F2-26:Ld 150 9 (44)
A47-138:Kb 120 7 (44)
K3-6:Db 40 2 (44)
RSV M2-82:Kd 419 25 (46)
M-187:Db 370 22 (46)
HSV 1gB:Kb 490 29 (51)
VSV N:Kb 166 10 (39)
MHCII
2W peptide 2W:I-Ab 384 15 (23)
307 12 (53)
300 12 (54)
225 9 (27)
190 8 (7)
183 7 (24)
L. monocytogenes LLO-190:I-Ab 80 3 (25)
MHV M133:I-Ab 62 2 (55)
S. typhimurium FliC-427:I-Ab 32 1 (26)
27 1 (24)
24 1 (23)
20 0.8 (7)
Ovalbumin OVA-329:I-Ab 43 2 (23)
1

Numbers represent total cells detected in pooled spleen and macroscopic lymph nodes.

2

Frequencies calculated from total cell numbers reported divided by approximate total numbers of naive T cells in pooled spleen and lymph nodes (1.7 × 107 CD8+ T cells or 2.5 × 107 CD4+ T cells).

Table 2.

Foreign p:MHC-specific naïve T cell frequencies in humans determined by tetramer- based cell enrichment

Source Epitope Frequency per
million naive CD4+
or CD8+ T cells1
Reference
MHCI
HCV NS3-1073:A2 60 (56)
NS3-1406:A2 29 (42)
11 (57)
Core-132:A2 1 (57)
1 (42)
NS5B-2594:A2 1 (42)
HIV Gag-77:A2 6 (56)
4 (57)
CMV pp65-495:A2 6 (56)
1 (57)
MHCII
B. anthracis PA-713:DR1 10 (32)
PA-401:DR1 2 (32)
PA-505:DR1 0.2 (32)
1

In cases where data was only reported as a proportion of total CD4+ or CD8+ T cells, frequencies were calculated assuming that 50% of these cells were naive phenotype.

Sallusto and colleagues recently devised a clever “T cell library” approach to solve this problem (11). These investigators cultured 384,000 naïve human CD4+ T cells in 192 wells at 2,000 cells/well with the phytohemagglutinin mitogen, allogeneic blood cells, and IL-2. These conditions led to 1,000-fold expansion of all the naïve cells in each well and converted them into hardy effector cells that could be restimulated with a foreign antigen plus autologous blood cells as APC. If 1 of the 192 original wells contained cells that proliferated in response to the foreign antigen, then it could be deduced that the frequency of naïve T cells specific for p:MHCII ligands derived from the antigen was 1/384,000. Using this approach, it was determined that naïve CD4+ T cells specific for p:MHCII ligands derived from keyhole limpet hemocyanin exist at frequencies of 10-70 cells per million naive CD4+ T cells, and for Bacillus anthracis protective antigen at 10-26 cells per million. If there are 10 different p:MHCII epitopes derived from keyhole limpet hemocyanin and each epitope is recognized by a naïve population of the same size, then each naïve population would exist at a frequency of 1-7 cells per million naive CD4+ T cells. An advantage of this technique is that it does not require knowledge of the subject’s MHC molecules and yields a total frequency that is the sum of the frequencies of the populations specific for all the relevant p:MHCII epitopes from the protein. A disadvantage of the technique is that it does not reveal the frequency of T cells specific for individual p:MHCII epitopes.

Recently, however, the combined use of fluorochrome-labeled p:MHC tetramers and magnetic particle-based cell enrichment has solved this problem. Fluorochrome-labeled p:MHC tetramers bind to the TCRs on specific T cells, marking them for detection (12). The difficulty, however, has been the limited capacity of flow cytometers to analyze only about 106 cells at a time, while the rare naïve T cells of interest are mixed in with ~2 × 108 total nucleated cells from the secondary lymphoid organs of a mouse or 100 ml of human peripheral blood. This problem was solved by staining all of the cells in the secondary lymphoid organs with a p:MHC tetramer and then with magnetic beads coupled to antibodies specific for the fluorochrome component of the tetramer (13-22). The sample could then be passed over a magnetic column to capture all of the tetramer-bound cells plus about 106 contaminants. The total number of cells in this bound fraction was therefore small enough that it could be analyzed in its entirety by flow cytometry. The tetramer-bound cells could be distinguished from contaminants by staining with a mixture of fluorochrome-labeled antibodies specific for T cell-and non-T cell-specific surface proteins.

This approach was used to identify naïve CD4+ T cells specific for different foreign p:MHCII ligands in C57BL/6 (B6) mice (7, 23-27). The tetramers used in these studies contained the I-Ab MHCII molecule bound to the 2W variant of peptide 52-68 from the I-E MHCII alpha chain (28), peptide 427-441 from the FliC protein of Salmonella typhimurium (29), peptide 329-337 from chicken ovalbumin (OVA) (30), or peptide 190-201 from the listeriolysin O protein of Listeria monocytogenes (31). The 2W:I-Ab- and LLO190-201:I-Ab-binding CD4+ T cell populations consisted of about 260 (7, 23-25, 27) and 80 (25) cells per mouse, respectively, while the FliC:I-Ab-binding (7, 23, 24, 26) and OVA:I-Ab-binding (23) populations contained about 20 and 40 cells per mouse. Assuming that mice contain 2.5 × 107 CD4+ T cells with a naive phenotype (9), the frequencies of naïve T cells specific for these individual p:MHCII complexes ranged from 0.8-10 cells per million naive CD4+ T cells.

Recently, Kwok and colleagues (32) used p:MHCII tetramer-based cell enrichment to enumerate human naïve CD4+ T cells specific for 3 different B. anthracis protective antigen peptide:HLA-DR1 epitopes in unvaccinated individuals. The frequencies of these naïve populations were 0.2, 2, and10 cells per million naïve CD4+ T cells (Table 2). This range of frequencies is remarkably similar to the 0.8-10 cells per million range reported for mouse T cells specific for individual p:MHCII epitopes determined by the same method (Tables 1 and 2). This congruence is surprising because the TCR diversity of humans has been estimated to be 10 times greater than that of mice (33, 34),

The fact that Kwok et al. (32) and Geiger et al. (11) both studied B. anthracis protective antigen allows for a comparison between the p:MHCII tetramer-based cell enrichment and T cell library methods. The sum of the frequencies of the three peptide:HLA-DR1-specific naïve populations measured by p:MHCII tetramer-based cell enrichment was ~12 cells per million naïve CD4+ cells. The collective frequency of T cells specific for all the p:MHCII epitopes in the protein measured by the T cell library approach was 10-26 cells per million. The similarity of the frequencies derived from p:MHCII tetramer staining and the T cell library approach that relies on antigen-driven T cell proliferation suggests that the former method may not greatly underestimate the number of T cells that are capable of responding to antigen as recently suggested (35).

p:MHCI tetramer-based cell enrichment has also been performed by several laboratories to enumerate the pre-immune frequencies for many different foreign p:MHCI-specific CD8+ T cell populations (Tables 1 and 2). The number of naïve phenotype CD8+ T cells in B6 mice ranged from 15 Lymphocytic choriomeningitis virus (LCMV) L338-346:Db-specific cells to 1,100 Vaccinia virus (VACV) B8R:Kb-specific cells per mouse (Table 1). These numbers translate to frequencies of 1-89 cells per million naive CD8+ T cells, assuming a total of 2 × 107 CD8+ naïve T cells per mouse (9). Again, a similar range of frequencies has been reported for human CD8+ T cells (Table 2).

An important conclusion from these studies is that naïve T cell populations specific for different p:MHC vary in size in a predictable fashion in individuals that express the same MHC molecules. Our analysis of relatively small and large naïve CD4+ T cell populations specific for different peptides bound to the same MHCII molecule revealed that the smaller population underwent more clonal deletion on cross-reactive self p:MHCII ligands than the larger population (24). Similarly, Day et al. (36) found that the reduction in the number of influenza acid polymerase 224–233:H-2Db-specific CD8+ naïve T cells in mice that expressed H-2Db and H-2Kk compared to mice that expressed H-2Db alone was explained in part by the deletion of influenza acid polymerase 224–233:H-2Db-specific cells due to cross-reactivity with H-2Kk molecules. An abnormally low amount of deletion of T cells that cross-react on self p:MHC may explain the extraordinary capacity of CD8+ T cells specific for HIV peptide:HLA-B57 complexes to control infection (37). HLA-B57 molecules bind a less diverse set of self peptides than other MHCI molecules, and thus may mediate less extensive negative selection of the CD8+ T cell repertoire. As a result, foreign peptide:HLA-B57-specific T cell populations may be larger and more promiscuous than average, thereby providing more effective immune responses to HIV.

In our studies, we also found that a peptide recognized by a relatively large naive cell population contained tryptophan residues as TCR contacts, which when changed to other amino acids reduced the size of the population (23). Thus, foreign p:MHCII ligands that tend to be recognized by large naive populations may have chemical properties that allow favorable interactions with many different TCRs. The finding of Turner and colleagues that CD8+ T cell populations with more TCR diversity recognize peptides containing TCR contact amino acids with large side chains than peptides with small side chains (38) is consistent with this possibility.

Variation in naïve T cell population size is of more than academic interest because it can determine the magnitude of the T cell response. Several studies demonstrated that naïve T cells expand in proportion to their starting frequency following exposure to the relevant p:MHC ligand (7, 39). Work by Sette and colleagues (40) showed that variation in naïve T cell population size is a partial explanation for the phenomenon of immunodominance. Lymphocytic choriomeningitis virus (LCMV) infection of B6 mice activates CD8+ T cells specific for one of at least 28 different pMHCI combinations. However, three of these p:MHCI ligands account for about one third of the total response. This dominance was not completely explained by MHCI binding affinity because several of the other peptides bound to MHCI in the same affinity range as the dominant peptides. Rather, the best predictor of response magnitude was the size of the naïve T cell population. The populations specific for the dominant peptides were about 10 times larger than those specific for the less dominant peptides. This correlation between naive population size and immunodominance was also seen in studies of influenza-infected humanized HLA-transgenic mice (41) or hepatitis C virus-specific human peripheral T cells (42), and B. anthracis-vaccinated humans (32). Therefore, although antigen abundance, efficiency of peptide generation by antigen processing, MHC binding affinity, and stability of p:MHC complexes certainly influence immunodominance (5, 6), naïve T cell population size is also an important factor. Large naïve T cell populations are advantageous to the host because they can generate a fixed number of microbicidal effector cells more quickly than smaller populations. For example, it took 4 days to generate 10,000 effector cells from 200 naïve phenotype 2W:I-Ab-specific CD4+ T cells, but 8 days from 20 naïve phenotype FliC:I-Ab-specific cells (7). Large naïve T cell populations may be particularly important to aged individuals because small populations are susceptible to extinction as the total pre-immune repertoire contracts during aging (43).

While the correlation between naive T cell population size and immune response magnitude has been substantiated by several studies (7, 32, 39-42, 44), this relationship was not observed in some recent comparisons of p:MHCI-specific CD8+ T cell populations during virus infection (45, 46). A disconnect between naïve population size and response magnitude could occur if only a fraction of the population is recruited into the response. This situation could result if the number of APC displaying the relevant p:MHC ligand is very low such that some T cells by chance interact with a p:MHC+ APC while other cells in the populations do not. Alternatively, low numbers of p:MHC ligands per APC could result in the response of only those T cells in the naïve population with highest affinity TCRs. In either case, a large naïve population would produce a smaller than expected response.

Schumacher and colleagues introduced unique molecular tags into each cell of a monoclonal naive CD8+ T cell population and then tracked the efficiency of naïve T cell recruitment into the primary response (47). Following adoptive transfer, it was found that essentially all of the cells in the population were efficiently recruited into the primary response regardless of the dose of antigen or type of infection. Therefore, naïve CD8+ T cells with an identical affinity for a p:MHCI ligand were recruited into the response in an all or none fashion. This finding raises the possibility that at a very low dose of antigen, no members of a large T cell population would respond, while at a slightly higher antigen dose, all of the members of the population would respond. To imagine how this situation could result in a disconnect between naïve T cell population size and primary response magnitude, consider two p:MHC complexes, pA:MHC and pB:MHC, derived from the same virus and recognized by naïve T cell populations containing 1,000 or 100 members, respectively. If the amount of pA:MHC complexes produced during infection is very small and the number of pB:MHC complexes is large, then it is possible that the 1,000 pA:MHC-specific cells will remain naïve while the 100 pB:MHC-specific cells will proliferate to produce 100,000 progeny.

Differential T cell proliferation following initial recruitment could also produce a lack of correlation between naïve T cell population size and primary response magnitude. This contention is supported by work from Zehn and colleagues who used altered peptide ligands with variations in TCR binding affinity for a monoclonal TCR to model what would happen to clones within a polyclonal p:MHCI-specific CD8+ T cell population with varying affinities for the p:MHCI complex in question (48). The monoclonal T cells began to proliferate in response to each of the peptides. However, the high-affinity peptides induced larger expanded T cell populations by inducing longer periods of proliferation following initial activation. These findings are largely consistent with TCR repertoire analyses showing that TCR affinity-based immunodominance patterns are not apparent immediately after the first few rounds of cell division, but emerge later on in the primary immune response (49). Taken together, these studies indicate that the initial recruitment of naive T cells from a pMHC-specific population into a primary immune response is fairly uniform across all TCR affinities, but sustained proliferation and eventual effector cell population size depends on TCR signal strength. Again consider two p:MHC complexes, pA:MHC and pB:MHC derived from the same virus and recognized by naïve T cell populations containing 1,000 or 100 members, respectively. If the pB:MHC-specific population contains a greater proportion of high affinity clones than the pA:MHC-specific population, then despite the initial response from all clones from both populations, the 100 naive cells from the pB:MHC-specific population will eventually undergo more extensive proliferation than the 1,000 naive cells from the pA:MHC-specific population, resulting in 100,000 versus 10,000 effector progeny and creating a disconnect between naïve population size and response magnitude. In contrast, if the two populations are represented by similar proportions of high-affinity clones and they see similar levels of p:MHC complexes, then they will proliferate largely in proportion to their starting numbers, thereby creating a situation where the larger pA:MHC-specific naïve population will produce more effector cells than the smaller pB:MHC-specific population. Under these conditions, naïve T cell population size will be the major determinant of primary immune response magnitude.

CONCLUSION

We speculate that an understanding of the rules that govern the size of naive T cell populations could lead to bioinformatic methods for assessing a person’s immune potential. Advances in microbial genomics, HLA typing, and peptide-HLA binding prediction algorithms suggest that it may soon be possible to scan all the peptides from a microbe that will bind to any of a person’s MHC molecules. In addition, some of the studies mentioned above hint that it will eventually be possible to predict the number of naive T cells specific for each MHC-binding peptide based on the nature of the TCR contacts in the peptide. Together, this information could be used to predict the magnitude of a person’s T cell response to all epitopes from a given microbe. This knowledge could inform the composition of subunit vaccines and identify people who are particularly susceptible to certain infections.

REFERENCES

  • 1.Henrickson SE, Mempel TR, Mazo IB, Liu B, Artyomov MN, Zheng H, Peixoto A, Flynn MP, Senman B, Junt T, Wong HC, Chakraborty AK, von Andrian UH. T cell sensing of antigen dose governs interactive behavior with dendritic cells and sets a threshold for T cell activation. Nat. Immunol. 2008;9:282–291. doi: 10.1038/ni1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Obst R, van Santen HM, Mathis D, Benoist C. Antigen persistence is required throughout the expansion phase of a CD4(+) T cell response. J. Exp. Med. 2005;201:1555–1565. doi: 10.1084/jem.20042521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Prlic M, Hernandez-Hoyos G, Bevan MJ. Duration of the initial TCR stimulus controls the magnitude but not functionality of the CD8+ T cell response. J. Exp. Med. 2006;203:2135–2143. doi: 10.1084/jem.20060928. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Itano AA, McSorley SJ, Reinhardt RL, Ehst BD, Ingulli E, Rudensky AY, Jenkins MK. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity. 2003;19:47–57. doi: 10.1016/s1074-7613(03)00175-4. [DOI] [PubMed] [Google Scholar]
  • 5.Sant AJ, Chaves FA, Jenks SA, Richards KA, Menges P, Weaver JM, Lazarski CA. The relationship between immunodominance, DM editing, and the kinetic stability of MHC class II:peptide complexes. Immunol. Rev. 2005;207:261–278. doi: 10.1111/j.0105-2896.2005.00307.x. [DOI] [PubMed] [Google Scholar]
  • 6.Yewdell JW. Confronting complexity: real-world immunodominance in antiviral CD8+ T cell responses. Immunity. 2006;25:533–543. doi: 10.1016/j.immuni.2006.09.005. [DOI] [PubMed] [Google Scholar]
  • 7.Moon JJ, Chu HH, Pepper M, McSorley SJ, Jameson SC, Kedl RM, Jenkins MK. Naive CD4(+) T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity. 2007;27:203–213. doi: 10.1016/j.immuni.2007.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Nikolich-Zugich J, Slifka MK, Messaoudi I. The many important facets of T-cell repertoire diversity. Nat. Rev. Immunol. 2004;4:123–132. doi: 10.1038/nri1292. [DOI] [PubMed] [Google Scholar]
  • 9.Jenkins MK, Chu HH, McLachlan JB, Moon JJ. On the composition of the preimmune repertoire of T cells specific for Peptide-major histocompatibility complex ligands. Annu. Rev. Immunol. 2010;28:275–294. doi: 10.1146/annurev-immunol-030409-101253. [DOI] [PubMed] [Google Scholar]
  • 10.Corradin G, Etlinger HM, Chiller JM. Lymphocyte specificity to protein antigens. I. Characterization of the antigen-induced in vitro T cell-dependent proliferative response with lymph node cells from primed mice. J. Immunol. 1977;119:1048–1053. [PubMed] [Google Scholar]
  • 11.Geiger R, Duhen T, Lanzavecchia A, Sallusto F. Human naive and memory CD4+ T cell repertoires specific for naturally processed antigens analyzed using libraries of amplified T cells. J. Exp. Med. 2009;206:1525–1534. doi: 10.1084/jem.20090504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Altman JD, Moss PAH, Goulder JR, Barouch DH, McHeyzer-Williams MG, Bell JI, McMichael AJ, Davis MM. Phenotypic analysis of antigen-specific T lymphocytes. Science. 1996;274:94–96. [PubMed] [Google Scholar]
  • 13.Bodinier M, Peyrat MA, Tournay C, Davodeau F, Romagne F, Bonneville M, Lang F. Efficient detection and immunomagnetic sorting of specific T cells using multimers of MHC class I and peptide with reduced CD8 binding. Nat. Med. 2000;6:707–710. doi: 10.1038/76292. [DOI] [PubMed] [Google Scholar]
  • 14.McDermott AB, Spiegel HM, Irsch J, Ogg GS, Nixon DF. A simple and rapid magnetic bead separation technique for the isolation of tetramer-positive virus-specific CD8 T cells. AIDS. 2001;15:810–812. doi: 10.1097/00002030-200104130-00024. [DOI] [PubMed] [Google Scholar]
  • 15.Keenan RD, Ainsworth J, Khan N, Bruton R, Cobbold M, Assenmacher M, Milligan DW, Moss PA. Purification of cytomegalovirus-specific CD8 T cells from peripheral blood using HLA-peptide tetramers. Br. J. Haematol. 2001;115:428–434. doi: 10.1046/j.1365-2141.2001.03106.x. [DOI] [PubMed] [Google Scholar]
  • 16.Jager E, Hohn H, Necker A, Forster R, Karbach J, Freitag K, Neukirch C, Castelli C, Salter RD, Knuth A, Maeurer MJ. Peptide-specific CD8+ T-cell evolution in vivo: response to peptide vaccination with Melan-A/MART-1. Int. J. Cancer. 2002;98:376–388. doi: 10.1002/ijc.10165. [DOI] [PubMed] [Google Scholar]
  • 17.Jang MH, Seth NP, Wucherpfennig KW. Ex vivo analysis of thymic CD4 T cells in nonobese diabetic mice with tetramers generated from I-A(g7)/class II-associated invariant chain peptide precursors. J. Immunol. 2003;171:4175–4186. doi: 10.4049/jimmunol.171.8.4175. [DOI] [PubMed] [Google Scholar]
  • 18.Day CL, Seth NP, Lucas M, Appel H, Gauthier L, Lauer GM, Robbins GK, Szczepiorkowski ZM, Casson DR, Chung RT, Bell S, Harcourt G, Walker BD, Klenerman P, Wucherpfennig KW. Ex vivo analysis of human memory CD4 T cells specific for hepatitis C virus using MHC class II tetramers. J. Clin. Invest. 2003;112:831–842. doi: 10.1172/JCI18509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lemaitre F, Viguier M, Cho MS, Fourneau JM, Maillere B, Kourilsky P, Van Endert PM, Ferradini L. Detection of low-frequency human antigen-specific CD4(+) T cells using MHC class II multimer bead sorting and immunoscope analysis. Eur. J. Immunol. 2004;34:2941–2949. doi: 10.1002/eji.200425281. [DOI] [PubMed] [Google Scholar]
  • 20.Barnes E, Ward SM, Kasprowicz VO, Dusheiko G, Klenerman P, Lucas M. Ultra-sensitive class I tetramer analysis reveals previously undetectable populations of antiviral CD8+ T cells. Eur. J. Immunol. 2004;34:1570–1577. doi: 10.1002/eji.200424898. [DOI] [PubMed] [Google Scholar]
  • 21.Lucas M, Day CL, Wyer JR, Cunliffe SL, Loughry A, McMichael AJ, Klenerman P. Ex vivo phenotype and frequency of influenza virus-specific CD4 memory T cells. J. Virol. 2004;78:7284–7287. doi: 10.1128/JVI.78.13.7284-7287.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Scriba TJ, Purbhoo M, Day CL, Robinson N, Fidler S, Fox J, Weber JN, Klenerman P, Sewell AK, Phillips RE. Ultrasensitive detection and phenotyping of CD4+ T cells with optimized HLA class II tetramer staining. J. Immunol. 2005;175:6334–6343. doi: 10.4049/jimmunol.175.10.6334. [DOI] [PubMed] [Google Scholar]
  • 23.Chu HH, Moon JJ, Kruse AC, Pepper M, Jenkins MK. Negative selection and peptide chemistry determine the size of naive foreign peptide-MHC class II-specific CD4+ T cell populations. J. Immunol. 2010;185:4705–4713. doi: 10.4049/jimmunol.1002276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chu HH, Moon JJ, Takada K, Pepper M, Molitor JA, Schacker TW, Hogquist KA, Jameson SC, Jenkins MK. Positive selection optimizes the number and function of MHCII-restricted CD4+ T cell clones in the naive polyclonal repertoire. Proc. Natl. Acad. Sci. U S A. 2009;106:11241–11245. doi: 10.1073/pnas.0902015106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Pepper M, Pagan AJ, Igyarto BZ, Taylor JJ, Jenkins MK. Opposing signals from the bcl6 transcription factor and the interleukin-2 receptor generate T helper 1 central and effector memory cells. Immunity. 2011;35:583–595. doi: 10.1016/j.immuni.2011.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Lee SJ, McLachlan JB, Kurtz JR, Fan D, Winter SE, Baumler AJ, Jenkins MK, McSorley SJ. Temporal expression of bacterial proteins instructs host CD4 T cell expansion and Th17 development. PLoS Pathog. 2012;8:e1002499. doi: 10.1371/journal.ppat.1002499. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dileepan T, Linehan JL, Moon JJ, Pepper M, Jenkins MK, Cleary PP. Robust antigen specific Th17 T cell response to group A Streptococcus is dependent on IL-6 and intranasal route of infection. PLoS Pathog. 2011;7:e1002252. doi: 10.1371/journal.ppat.1002252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Rees W, Bender J, Teague TK, Kedl RM, Crawford F, Marrack P, Kappler J. An inverse relationship between T cell receptor affinity and antigen dose during CD4(+) T cell responses in vivo and in vitro. Proc. Natl. Acad. Sci. U S A. 1999;96:9781–9786. doi: 10.1073/pnas.96.17.9781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.McSorley SJ, Cookson BT, Jenkins MK. Characterization of CD4+ T cell responses during natural infection with Salmonella typhimurium. J. Immunol. 2000;164:986–993. doi: 10.4049/jimmunol.164.2.986. [DOI] [PubMed] [Google Scholar]
  • 30.Robertson JM, Jensen PE, Evavold BD. DO11.10 and OT-II T cells recognize a C-terminal ovalbumin 323-339 epitope. J. Immunol. 2000;164:4706–4712. doi: 10.4049/jimmunol.164.9.4706. [DOI] [PubMed] [Google Scholar]
  • 31.Geginat G, Schenk S, Skoberne M, Goebel W, Hof H. A novel approach of direct ex vivo epitope mapping identifies dominant and subdominant CD4 and CD8 T cell epitopes from Listeria monocytogenes. J. Immunol. 2001;166:1877–1884. doi: 10.4049/jimmunol.166.3.1877. [DOI] [PubMed] [Google Scholar]
  • 32.Kwok WW, Tan V, Gillette L, Littell CT, Soltis MA, Lafond RB, Yang J, James EA, Delong JH. Frequency of epitope-specific naive CD4+ T cells correlates with immunodominance in the human memory repertoire. J. Immunol. 2012 doi: 10.4049/jimmunol.1102190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Arstila TP, Casrouge A, Baron V, Even J, Kanellopoulos J, Kourilsky P. A direct estimate of the human alphabeta T cell receptor diversity. Science. 1999;286:958–961. doi: 10.1126/science.286.5441.958. [DOI] [PubMed] [Google Scholar]
  • 34.Casrouge A, Beaudoing E, Dalle S, Pannetier C, Kanellopoulos J, Kourilsky P. Size estimate of the alpha beta TCR repertoire of naive mouse splenocytes. J. Immunol. 2000;164:5782–5787. doi: 10.4049/jimmunol.164.11.5782. [DOI] [PubMed] [Google Scholar]
  • 35.Sabatino J, J. J, Huang J, Zhu C, Evavold BD. High prevalence of low affinity peptide–MHC II tetramer–negative effectors during polyclonal CD4+ T cell responses. J. Exp. Med. 2011;208:81–90. doi: 10.1084/jem.20101574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Day EB, Charlton KL, La Gruta NL, Doherty PC, Turner SJ. Effect of MHC class I diversification on influenza epitope-specific CD8+ T cell precursor frequency and subsequent effector function. J. Immunol. 2011;186:6319–6328. doi: 10.4049/jimmunol.1000883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kosmrlj A, Read EL, Qi Y, Allen TM, Altfeld M, Deeks SG, Pereyra F, Carrington M, Walker BD, Chakraborty AK. Effects of thymic selection of the T-cell repertoire on HLA class I-associated control of HIV infection. Nature. 2010;465:350–354. doi: 10.1038/nature08997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Turner SJ, Kedzierska K, Komodromou H, La Gruta NL, Dunstone MA, Webb AI, Webby R, Walden H, Xie W, McCluskey J, Purcell AW, Rossjohn J, Doherty PC. Lack of prominent peptide-major histocompatibility complex features limits repertoire diversity in virus-specific CD8+ T cell populations. Nat. Immunol. 2005;6:382–389. doi: 10.1038/ni1175. [DOI] [PubMed] [Google Scholar]
  • 39.Obar JJ, Khanna KM, Lefrancois L. Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity. 2008;28:859–869. doi: 10.1016/j.immuni.2008.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kotturi MF, Scott I, Wolfe T, Peters B, Sidney J, Cheroutre H, von Herrath MG, Buchmeier MJ, Grey H, Sette A. Naive precursor frequencies and MHC binding rather than the degree of epitope diversity shape CD8+ T cell immunodominance. J. Immunol. 2008;181:2124–2133. doi: 10.4049/jimmunol.181.3.2124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Tan AC, La Gruta NL, Zeng W, Jackson DC. Precursor frequency and competition dictate the HLA-A2-restricted CD8+ T cell responses to influenza A infection and vaccination in HLA-A2.1 transgenic mice. J. Immunol. 2011;187:1895–1902. doi: 10.4049/jimmunol.1100664. [DOI] [PubMed] [Google Scholar]
  • 42.Schmidt J, Neumann-Haefelin C, Altay T, Gostick E, Price DA, Lohmann V, Blum HE, Thimme R. Immunodominance of HLA-A2-restricted hepatitis C virus-specific CD8+ T cell responses is linked to naive-precursor frequency. J. Virol. 2011;85:5232–5236. doi: 10.1128/JVI.00093-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yager EJ, Ahmed M, Lanzer K, Randall TD, Woodland DL, Blackman MA. Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus. J. Exp. Med. 2008;205:711–723. doi: 10.1084/jem.20071140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Flesch IE, Woo WP, Wang Y, Panchanathan V, Wong YC, La Gruta NL, Cukalac T, Tscharke DC. Altered CD8(+) T cell immunodominance after vaccinia virus infection and the naive repertoire in inbred and F(1) mice. J. Immunol. 2010;184:45–55. doi: 10.4049/jimmunol.0900999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.La Gruta NL, Rothwell WT, Cukalac T, Swan NG, Valkenburg SA, Kedzierska K, Thomas PG, Doherty PC, Turner SJ. Primary CTL response magnitude in mice is determined by the extent of naive T cell recruitment and subsequent clonal expansion. J. Clin. Invest. 2010;120:1885–1894. doi: 10.1172/JCI41538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ruckwardt TJ, Malloy AM, Gostick E, Price DA, Dash P, McClaren JL, Thomas PG, Graham BS. Neonatal CD8 T-cell Hierarchy Is Distinct from Adults and Is Influenced by Intrinsic T cell Properties in Respiratory Syncytial Virus Infected Mice. PLoS Pathog. 2011;7:e1002377. doi: 10.1371/journal.ppat.1002377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.van Heijst JW, Gerlach C, Swart E, Sie D, Nunes-Alves C, Kerkhoven RM, Arens R, Correia-Neves M, Schepers K, Schumacher TN. Recruitment of antigen-specific CD8+ T cells in response to infection is markedly efficient. Science. 2009;325:1265–1269. doi: 10.1126/science.1175455. [DOI] [PubMed] [Google Scholar]
  • 48.Zehn D, Lee SY, Bevan MJ. Complete but curtailed T-cell response to very low-affinity antigen. Nature. 2009;458:211–214. doi: 10.1038/nature07657. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Malherbe L, Hausl C, Teyton L, McHeyzer-Williams MG. Clonal selection of helper T cells is determined by an affinity threshold with no further skewing of TCR binding properties. Immunity. 2004;21:669–679. doi: 10.1016/j.immuni.2004.09.008. [DOI] [PubMed] [Google Scholar]
  • 50.Hamilton SE, Schenkel JM, Akue AD, Jameson SC. IL-2 complex treatment can protect naive mice from bacterial and viral infection. J. Immunol. 2010;185:6584–6590. doi: 10.4049/jimmunol.1001215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Haluszczak C, Akue AD, Hamilton SE, Johnson LD, Pujanauski L, Teodorovic L, Jameson SC, Kedl RM. The antigen-specific CD8+ T cell repertoire in unimmunized mice includes memory phenotype cells bearing markers of homeostatic expansion. J. Exp. Med. 2009;206:435–448. doi: 10.1084/jem.20081829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Akue AD, Lee JY, Jameson SC. Derivation and maintenance of virtual memory CD8 T cells. J. Immunol. 2012 doi: 10.4049/jimmunol.1102213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Moon JJ, Dash P, Oguin TH, 3rd, McClaren JL, Chu HH, Thomas PG, Jenkins MK. Quantitative impact of thymic selection on Foxp3+ and Foxp3− subsets of self-peptide/MHC class II-specific CD4+ T cells. Proc. Natl. Acad. Sci. U S A. 2011;108:14602–14607. doi: 10.1073/pnas.1109806108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Pepper M, Linehan JL, Pagan AJ, Zell T, Dileepan T, Cleary PP, Jenkins MK. Different routes of bacterial infection induce long-lived TH1 memory cells and short-lived TH17 cells. Nat. Immunol. 2010;11:83–89. doi: 10.1038/ni.1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhao J, Zhao J, Fett C, Trandem K, Fleming E, Perlman S. IFN-gamma- and IL-10-expressing virus epitope-specific Foxp3(+) T reg cells in the central nervous system during encephalomyelitis. J. Exp. Med. 2011;208:1571–1577. doi: 10.1084/jem.20110236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Legoux F, Debeaupuis E, Echasserieau K, De La Salle H, Saulquin X, Bonneville M. Impact of TCR reactivity and HLA phenotype on naive CD8 T cell frequency in humans. J. Immunol. 2010;184:6731–6738. doi: 10.4049/jimmunol.1000295. [DOI] [PubMed] [Google Scholar]
  • 57.Alanio C, Lemaitre F, Law HK, Hasan M, Albert ML. Enumeration of human antigen-specific naive CD8+ T cells reveals conserved precursor frequencies. Blood. 2010;115:3718–3725. doi: 10.1182/blood-2009-10-251124. [DOI] [PubMed] [Google Scholar]

RESOURCES