Skip to main content

Main menu

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
      • Neuroimmunology: To Sense and Protect
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons

User menu

  • Subscribe
  • My alerts
  • Log in
  • Log out

Search

  • Advanced search
The Journal of Immunology
  • Other Publications
    • American Association of Immunologists
    • ImmunoHorizons
  • Subscribe
  • My alerts
  • Log in
  • Log out
The Journal of Immunology

Advanced Search

  • Home
  • Articles
    • Current Issue
    • Next in The JI
    • Archive
    • Brief Reviews
    • Pillars of Immunology
    • Translating Immunology
    • Most Read
    • Top Downloads
    • Annual Meeting Abstracts
  • COVID-19/SARS/MERS Articles
  • Info
    • About the Journal
    • For Authors
    • Journal Policies
    • Influence Statement
    • For Advertisers
  • Editors
  • Submit
    • Submit a Manuscript
    • Instructions for Authors
    • Journal Policies
  • Subscribe
    • Journal Subscriptions
    • Email Alerts
    • RSS Feeds
    • ImmunoCasts
  • More
    • Most Read
    • Most Cited
    • ImmunoCasts
    • AAI Disclaimer
    • Feedback
    • Help
    • Accessibility Statement
  • Follow The Journal of Immunology on Twitter
  • Follow The Journal of Immunology on RSS

Homeostatic Expansion Occurs Independently of Costimulatory Signals

Martin Prlic, Bruce R. Blazar, Alexander Khoruts, Traci Zell and Stephen C. Jameson
J Immunol November 15, 2001, 167 (10) 5664-5668; DOI: https://doi.org/10.4049/jimmunol.167.10.5664
Martin Prlic
*Laboratory Medicine and Pathology,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Bruce R. Blazar
†Pediatrics,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Alexander Khoruts
‡Medicine, and
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Traci Zell
§Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Stephen C. Jameson
*Laboratory Medicine and Pathology,
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Naive T cells undergo homeostatic proliferation in lymphopenic mice, a process that involves TCR recognition of specific self peptide/MHC complexes. Since costimulation signals regulate the T cell response to foreign Ags, we asked whether they also regulate homeostatic expansion. We report in this study that homeostatic expansion of CD4 and CD8 T cells occurs independently of costimulation signals mediated through CD28/B7, CD40L/CD40, or 4-1BB/4-1BBL interactions. Using DO11.10 TCR transgenic T cells, we confirmed that CD28 expression was dispensable for homeostatic expansion, and showed that the presence of endogenous CD4+CD25+ regulatory cells did not detectably influence homeostatic expansion. The implications of these findings with respect to regulation of T cell homeostasis and autoimmunity are discussed.

Lymphocyte numbers in the periphery are tightly controlled through various homeostatic mechanisms. The size of the T and B cell subsets is regulated independently, as are naive and memory T cell pools (1, 2). Recent data from a number of groups indicated that a reduction in the number of circulating T cells is compensated by proliferation of remaining lymphocytes, a process called homeostatic expansion (3, 4, 5, 6, 7, 8). Since T cell numbers may be reduced during life as a result of both infections and diminished thymic output, homeostatic expansion may play an important role in maintaining peripheral T cell numbers.

T cell recognition of specific self peptide/MHC ligands is important for optimal homeostatic expansion, and cells undergoing this process become phenotypically and functionally indistinguishable from memory T cells (5, 6, 9). Together, these data suggest that homeostatic expansion involves a TCR signal that induces functional differentiation. As is well established, T proliferative responses toward conventional foreign peptide/MHC ligands require a second signal, referred to as costimulation, to avoid T cell anergy or death. Costimulation is mediated by a variety of cell surface molecules of which CD28/B7 and CD40L/CD40 interactions are the best characterized (reviewed in Refs. 10 and 11), although several other molecules may synergize with or even substitute for these classic costimulators (reviewed in Refs. 12, 13, 14). Prominent among these is the 4-1BB/4-1BBL interaction that can both augment and substitute for CD28/B7 interactions in both CD4 and CD8 T cell responses (12, 15, 16, 17, 18). However, the role of costimulation in homeostatic expansion has not been well defined. Unpublished data from two groups suggest that CD28 is not essential for homeostatic expansion (6, 8), a result that might reflect true independence of homeostatic expansion from costimulation, or could instead point to functional redundancy among costimulatory pathways. In this study, we show that homeostatic expansion does not require CD28 on the T cell, nor does it require CD40 or 4-1BBL expression on host APCs. This was true even when combinations of costimulator deficiencies were studied (i.e., CD28−/− T cells transferred into CD40−/− or 4-1BBL−/− hosts). Indeed, in contrast to the critical role of CD28 in responses to foreign Ags, we show that CD28-deficient cells proliferate as well (or even slightly better) than wild-type cells during homeostatic expansion of DO11.10 TCR transgenic T cells. This unexpected result did not appear to correlate with the presence or absence of the CD4+CD25+ regulatory T cell subset, as shown by depletion experiments. The implications of these data on the discrimination between conventional and homeostatic proliferation pathways are discussed.

Materials and Methods

Mice

C57BL/6 (B6) background CD40−/− mice were initially obtained from D. Parker (Portland, OR), and additional CD40−/− and B6 background CD28−/− mice were obtained from The Jackson Laboratory (Bar Harbor, ME). The 4-1BBL−/− mice (19) were obtained in collaboration with J. Peschon (Immunex, Seattle, WA). BALB/c, BALB/c Rag−/−, B6, and CD45.1 congenic B6 mice (carried under the strain name B6.Ly-5.2) were obtained from the National Cancer Institute, The Jackson Laboratory, or Taconic Farms. Normal and CD28−/− DO11.10 TCR transgenic mice have been described previously (20, 21) and were maintained at the University of Minnesota. All mice were maintained under specific pathogen-free conditions and used at 6–12 wk of age. Recipient mice were maintained on antibiotic water (polymixin B sulfate and neomycin sulfate) throughout the course of the experiment.

Adoptive transfer

Cells were purified by negative selection utilizing magnetic cell sorting using MACS microbeads (Miltenyi Biotec, Auburn, CA). To purify CD44low T cell, lymph node cells were depleted from adherent cells (90-min incubation on tissue culture-treated flasks at 37°C) and labeled with FITC-coupled Abs to B220 (clone RA3-6B2), I-Ab (AF6-120.1), and CD44 (IM-7) (0.0125 μg anti-B220 and anti-I-Ab per 1 × 106 cells; 0.004 μg anti-CD44 per 1 × 106 cells) (all from BD PharMingen, San Diego, CA). In some experiments, CD4 cells were also labeled with FITC anti-CD4 (GK1.5; BD PharMingen) (0.0125 μg per 1 × 106 cells). Following staining, cells were subject to depletion using anti-FITC MACS microbeads. Flow through cells were >90% pure (established by flow cytometric analysis).

CD4+ cells from DO11.10 animals were purified utilizing a similar protocol using biotinylated (bio)3 anti-CD8 (53.6-7) plus bio-anti-B220, followed by streptavidin (SA) microbeads. In some cases, CD25+ cells were depleted at the same time using bio-anti-CD25 (7D4) (if applicable) (PharMingen). The percentage of CD4+CD25+ T cells before and after depletion was assessed by staining an aliquot of the cells with both CD25-PE Ab and SA-PE (to reveal cells bound by the depleting bio-CD25 Ab) and counterstaining for CD4 with CD4-APC or CD4-PerCP.

After purification, cells were stained with CFSE (Molecular Probes, Eugene, OR), as described previously (7, 22). Labeled donor cells were suspended in PBS, and 1–3 million cells were injected via the tail vein of recipient mice. In some cases, host mice were sublethally irradiated (700 cGy) 1 day before cell transfer. In some experiments, recipient mice were primed i.v. with 2 mg OVA (whole protein) plus 25 μg LPS (Sigma, St. Louis, MO) 1 day cell posttransfer. Cells were analyzed on day 3 postpriming.

Flow cytometry

Recipient mice were sacrificed at the time points indicated, and single cell suspensions were prepared separately from spleen and a pool of major lymph nodes. Lymph node and spleen cells were then stained with the fluorescently conjugated Abs to CD4, CD8, CD44, and CD45RB. Biotinylated Abs to CD25, CD45.1, CD45.2, CD28, and KJ1-26 (specific for the DO11.10 TCR) were used, staining being revealed using SA-TriColor, SA-PerCP, or SA-PE (all reagents from BD PharMingen). Cells were analyzed by using a FACSCalibur (BD Biosciences, San Jose, CA) and analyzed using FLOWJO (TreeStar, San Carlos, CA) software.

Results

Experimental system

To test the role of CD28/B7 interactions in regulating homeostatic expansion, we studied proliferation of CFSE-labeled normal and CD28−/− donor T cells after transfer into normal or lymphopenic B6 hosts. To minimize mouse to mouse variation, we employed a cotransfer system in which we premixed CD45.1+ wild-type and CD45.2+ CD28−/− T cells before transfer and distinguished the two donor populations during analysis with CD45 allele-specific Abs. Similar transfers were performed using CD40−/− or 4-1BBL−/− hosts to determine the effect of disrupting CD40L/CD40 and 4-1BB/4-1BBL interactions (alone or in combination with CD28 deficiency) on homeostatic expansion.

No requirement for CD28, CD40, or 4-1BBL in T cell homeostatic expansion

Using the transfer scheme outlined above, we tested whether CD28 expression on T cells and CD40 or 4-1BBL expression on host APCs was required for T cell homeostatic expansion. As expected, neither wild-type nor CD28−/− T cells proliferate after transfer into unmanipulated syngeneic B6 hosts (Fig. 1⇓, a–d). Also as expected, normal CD8 and CD4 T cells undergo one to four rounds of proliferation 14 days after transfer into irradiated syngeneic hosts (Fig. 1⇓, e and g, respectively). Interestingly, CD28−/− T cells proliferated to the same extent as wild-type cells when transferred into irradiated wild-type hosts (Fig. 1⇓, f and h). This suggests that CD28 is not required for homeostatic expansion of either CD8 or CD4 subsets.

FIGURE 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 1.

Role of CD28 and CD40 interactions in homeostatic expansion of cotransferred CD4 and CD8 T cells. One million CD44low T cells from CD28−/− and from CD45.1 congenic B6 animals were mixed at a 1:1 ratio, CFSE labeled, and injected i.v. into the indicated host mice. In some cases (irrad.), recipient mice were sublethally irradiated (700 cGy) 1 day before transfer. Recipient mice were sacrificed 14 days posttransfer, and lymph node cells were analyzed by flow cytometry using Abs to CD4, CD8, and CD45.1 to distinguish the different subsets of cells. The CFSE profile of CD4 and CD8 subsets and of CD28−/− and wild-type populations is shown separately, but comes from the same mice in each row. Because the recipient mice are all CD45.1−, host cells appear in the analysis of the CD28−/− subsets as a CFSE-negative population. Data are representative of two to three mice per group, and comparable results were obtained in at least three experiments.

It was possible that CD40L/CD40 or 4-1BB/4-1BBL costimulation could partially or completely compensate for CD28 deficiency. Indeed, 4-1BB and CD40L interactions can synergize with CD28 or even replace it in some T cell responses (17, 19, 23, 24). To test this, we transferred both wild-type and CD28−/− T cells into CD40-deficient hosts (Fig. 1⇑, i–l) or into 4-1BBL-deficient hosts (Fig. 1⇑, m–p). Once again, homeostatic proliferation of both CD4 and CD8 T cell subsets was similar regardless of CD28 expression on the donor cells and CD40 or 4-1BBL expression in the host.

In all of these experiments, we observed similar donor T cell recoveries for both the normal and CD28−/− donors after expansion in the various irradiated host strains (data not shown), which argues against a survival role for these costimulator molecules in homeostatic expansion, at least in the short term.

CD28−/− DO11.10 TCR transgenic T cells undergo greater homeostatic expansion, but less Ag-driven proliferation than their CD28+ counterparts

The experiments above utilized polyclonal T cells and, although the overall degree of homeostatic expansion was similar with or without CD28, it was possible that different populations of T cells were responding, perhaps based on TCR specificity dictating CD28 dependence. Furthermore, we had to date looked exclusively in the C57BL/6 system, whereas there is some evidence that the impact of CD28/B7 deficiency is more extreme in certain BALB/c responses (25). Accordingly, we also studied homeostatic expansion of T cells from DO11.10 TCR transgenic mice, which are maintained on the BALB/c background. Data from several groups have shown that CD28 deficiency impairs the peptide/MHC (OVA/I-Ad) response of these T cells, both in vitro and in vivo (21, 26).

Thus, we transferred DO11.10 and DO11.10/CD28−/− cells into irradiated BALB/c hosts and analyzed their homeostatic expansion. As shown in Fig. 2⇓, not only did we fail to see a requirement for CD28 in DO11.10 homeostatic expansion, there was evidence that CD28−/− DO11.10 cells proliferate moderately better than their wild-type counterparts. The degree of this improved response was somewhat variable between individual animals, as exemplified in Fig. 2⇓. Thus, at a minimum we find that more CD28-deficient DO11 cells commit to homeostatic expansion than normal DO11 cells (see proportions of cells undergoing zero vs three divisions, Fig. 2⇓a), and that in some cases this was seen as an extra round of proliferation by the CD28−/− DO11.10 cells (Fig. 2⇓b).

FIGURE 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 2.

Homeostatic expansion of normal vs CD28−/− DO11.10 cells. One million CFSE-labeled DO11.10 or CD28−/− DO11.10 T cells were transferred into sublethally irradiated BALB/c hosts and analyzed 21 days after transfer. The CFSE profiles shown were gated on CD4+ KJ1-26+ cells. The number of rounds of proliferation (derived from comparison with CFSE levels in cells transferred into unirradiated BALB/c hosts) are indicated over the traces. Normal DO11.10 cells are represented by the shaded histogram; CD28−/− DO11.10 cells are shown as the unfilled solid line. The data for a and b come from separate host mice in the same experiment and represent the range seen among three separate experiments involving three mice per group.

Given this surprising result, it was important to test that, in our hands, CD28 deficiency had the predicted effect of blunting the DO11.10 Ag-specific response, as reported by others (21, 26). Thus, we transferred CFSE-labeled wild-type or CD28−/− DO11.10 T cells into irradiated or unirradiated BALB/c hosts. The unirradiated hosts were then immunized with OVA/LPS, and the proliferative response of the DO11.10 population was monitored. As expected, CD28 deficiency severely impaired the Ag-specific DO11.10 response (Fig. 3⇓, c vs b). However, as in Fig. 2⇑, homeostatic proliferation of the same CD28−/− DO11.10 donor population was slightly greater than that of wild-type transgenic cells (Fig. 3⇓, g vs f).

FIGURE 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 3.

Impact of CD28 expression and CD4+CD25+ T cell depletion (Depl.) on foreign Ag reactivity and homeostatic expansion of DO11.10 cells in irradiated hosts. Normal, CD28−/−, and CD25 T cell-depleted DO11.10 T cells were transferred into normal (a–e) or irradiated (f–h) BALB/c hosts. The percentages of CD4+CD25+ cells in the normal, CD28−/−, and CD25-depleted populations were 4.2%, 1.2%, and 0.5%, respectively, in the experiment shown. The unirradiated hosts were challenged with OVA/LPS 1 day after transfer and sacrificed 3 days later. Irradiated (f–h) and control unirradiated hosts (e) were not immunized and were analyzed 21 days after transfer. CFSE profiles shown are gated on CD4+ KJ1-26+ cells, except in a, which was gated on CD4+ KJ1-26− cells as a control for Ag nonspecific proliferation. Data are representative of three mice in each group.

CD4+CD25+ cells do not contribute to the difference seen between normal and CD28−/− DO11.10 T cells

Looking for an explanation for the slightly enhanced proliferation of DO11.10 CD28−/− cells, we considered the contribution of CD4+CD25+ regulatory cells (27, 28). This subset, which can suppress autoimmune T cell responses, is underrepresented in CD28−/− animals (29). This is also true in mice expressing the DO11.10 TCR transgene, in which CD28−/− DO11.10 mice have fewer CD4+CD25+ cells compared with wild-type DO11.10 animals (3–5% vs 5–10%, respectively; data not shown). Thus, we tested the impact of depleting the donor DO11.10 cells of CD25+ cells. If the CD25+ subset were restraining normal DO11.10 homeostatic expansion, then removal of these cells should lead to even greater homeostatic expansion than seen for the CD28−/− DO11.10 group. CD25+ regulatory cell depletion did not improve the robust Ag-specific T cell response in vivo, perhaps reflecting the fact that this response was already optimal (Fig. 3⇑d). Interestingly, however, CD25+ subset depletion failed to impact DO11.10 homeostatic expansion (Fig. 3⇑, compare h with f).

Since sublethal irradiation does not deplete all T cells in the host, it was possible that endogenous CD4+CD25+ cells could influence homeostatic expansion of the transferred cells. Hence, we also transferred these same DO11.10 populations into BALB/c Rag−/− hosts. Similar to the results in the irradiated hosts, proliferation of CD25-depleted DO11.10 cells did not differ significantly from that of normal DO11.10 cells, whereas there was once again a subtle, but consistent increase in proliferation by the CD28−/− DO11.10 population (Fig. 4⇓). Intriguingly, after expansion of DO11.10 cells transferred into Rag-deficient hosts, a sizeable proportion of the cells had proliferated sufficiently to dilute out the CFSE dye, which was not observed in irradiated hosts (compare Fig. 4⇓ with Figs. 2⇑ and 3⇑). This may relate to the fact that sublethally irradiated hosts recover endogenous T cell numbers during the course of the experiment (therefore limiting T cell space), whereas this will not occur in Rag−/− hosts. We also noticed a slight increase in the total number of donor T cells recovered after transfer of CD25-depleted DO11.10 cells compared with wild-type or CD28−/− cells, which was also not observed in irradiated hosts (data not shown).

FIGURE 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
FIGURE 4.

Impact of CD28 expression and CD4+CD25+ T cell depletion (Depl.) on homeostatic expansion in Rag−/− hosts. Three million CFSE-labeled normal, CD28−/−, or CD25 T cell-depleted DO11.10 cells were transferred into BALB/c Rag-2−/− hosts. The percentages of CD4+CD25+ cells in the normal, CD28−/−, and CD25-depleted populations were 4.1, 2.8, and 0.6%, respectively. Recipient mice were harvested 22 days after transfer, and the data shown are representative of three mice in each group.

Thus, these results do not support a role for the DO11.10 CD25+ subset in restraining homeostatic expansion of DO11.10 T cells.

Discussion

Since optimal homeostatic expansion involves reactivity to self peptide/MHC TCR ligands, this process has the potential to favor accumulation of autoreactive T cells, as has been proposed (30). Restraining such autoaggression may entail both differences in the nature of the TCR signal and the contribution of other T cell activation signals. Indeed, data from our group and others indicate that homeostatic expansion does not require an IL-2 signal, but is dependent on IL-7, in contrast to the requirements for these two cytokines in naive T cell responses toward foreign Ag (8, 22, 31, 32). Nonetheless, both conventional and homeostatic responses can be augmented by inflammatory cytokines, as revealed by the impact of IL-12 on CD8 T cell proliferation (22), suggesting that regulation of these responses is at least partially overlapping.

In this study, we investigated the requirement for costimulation in homeostatic expansion. The majority of our work focused on CD28, which is expressed on naive T cells and is strongly implicated in costimulating both CD4 and CD8 T cell responses (10). CD40L and 4-1BB are expressed after activation of T cells through the TCR (at least to some extent, independently of CD28 signals) (11, 12, 17, 18). Although the expression of either CD40L or 4-1BB on cells undergoing homeostatic expansion has not been addressed, there is strong evidence that this process involves TCR-mediated signals and that T cells become at least partially activated, changing expression of surface markers and changing in effector potential (9). This raises the possibility that 4-1BB and CD40L may be functionally expressed at some point during homeostatic expansion. However, in contrast to the well-documented need for costimulation in conventional naive T cell proliferative response, homeostatic expansion appears undiminished by the absence of classic costimulatory signals, mediated by CD28/B7, CD40L/CD40, or 4-1BB/4-1BBL interactions. Importantly, our results with CD28-deficient DO11.10 populations suggest that T cells can undergo homeostatic expansion regardless of whether these T cells could participate properly in an immune response to foreign Ag.

We found that CD28 was dispensable for homeostatic expansion by polyclonal or TCR transgenic T cells in irradiated or Rag−/− hosts. Using these different experimental systems minimizes the possibility that a role for CD28 is masked by either the method of host lymphocyte depletion or the TCR diversity of the donor T cell population. While we have not specifically studied whether costimulation may play a role when the degree of host lymphopenia is less severe (which might potentially raise the stringency for commitment to homeostatic expansion), it is worth noting that expansion in sublethally irradiated recipients is limited by recovery of the host T cell population; indeed, this may account for the enhanced proliferation of DO11.10 cells in Rag−/− vs irradiated hosts. Thus, the fact that we did not observe reduced expansion of CD28−/− cells in irradiated hosts argues against a role for this costimulatory pathway even in the competition for limited lymphocyte space. In any event, our data argue against a critical role for costimulation through any of these major pathways in driving homeostatic expansion.

In fact, our data suggested that lack of CD28 expression mildly enhanced DO11.10 T cell homeostatic expansion in both sublethally irradiated and Rag−/− hosts. It is important to note that this effect was subtle and somewhat variable in its magnitude (as exemplified in Fig. 2⇑). Furthermore, we could not see consistent evidence for enhanced proliferation of CD28−/− polyclonal T cells (Fig. 1⇑ and data not shown): subtle differences in the CD28−/− population may be obscured if there is variability in the degree of homeostatic proliferation among polyclonal T cells (as is most likely based on analysis of numerous TCR transgenic systems) (9, 33) or may imply that the enhancing effect of CD28 deficiency is unique to DO11.10 T cells. Further investigation will be needed to explore the significance of this finding.

While our data rule out a requirement for CD28 in homeostatic expansion, it is still possible that such responses are regulated by CTLA-4, which negatively regulates T cell proliferation in response to foreign Ag. Indeed, since CD28/B7 interactions are involved (but not essential) (34, 35) for CTLA-4 expression itself, cells proliferating during homeostatic expansion may not express CTLA-4 at sufficient levels to inhibit the expansion process. This in turn may allow these cells to evade normal regulatory control.

One potential explanation for the mild positive effect of CD28 deficiency on homeostatic expansion could be the decreased representation of CD4+CD25+ regulatory cells in CD28−/− animals (29). However, elimination of this population in the DO11.10 system did not result in enhanced homeostatic expansion in either irradiated or Rag−/− recipients. These data do not rule out the possibility that CD4+CD25+ cells may be able to inhibit homeostatic expansion if present in sufficient numbers or if activated appropriately (an apparent requirement for their suppressive effect) (36, 37). Rather, our data argue that the endogenous CD4+CD25+ population does not inhibit homeostatic expansion, in contrast to the proposed role of such cells in preventing autoimmune or alloreactive responses (27, 28, 37, 38, 39). Interestingly, this might be expected if inhibition by CD4+CD25+ regulatory cells was mediated through disruption of IL-2 production and/or IL-2Rα up-regulation (as has been proposed; 28), since IL-2 appears not to be important in homeostatic expansion and IL-2Rα is not expressed during this response (7, 8, 40, 41). Thus, these data further emphasize the differences in regulation that control homeostatic expansion compared with other T cell responses.

Note added in proof.

While this manuscript was in press, Gudmundsdottir et al. (42) reported that CTLA4Ig blocked homeostatic expansion of a subset of DO11.10 Rag−/− T cells in Rag−/− recipients. Specifically, CTLA4Ig administration caused a selective loss of the rapidly proliferating subset and this inhibition was influenced by the number of donor T cells transferred. These data suggest B7 interactions might influence homeostatic proliferation of certain subsets under specific conditions, while our data argue that CD28 interactions are not obligatory for this response. The basis for these discordant results presumably lies in the different experimental approaches used.

Acknowledgments

We thank David Parker and Jacques Peschon for knockout mice, Marc Jenkins for providing other mouse strains and suggestions, Kristen Thorstenson and Brent Koehn for excellent technical assistance, and Kris Hogquist and members of the Hogquist and Jameson labs for critical input.

Footnotes

  • ↵1 This work was supported by American Cancer Society Grant RPG 99-264 (to S.C.J.) and National Institutes of Health Grants R01-AI38903 (to S.C.J.), R01-AI34495, R37-HL56067, and P01-AI-35225 (to B.R.B.).

  • ↵2 Address correspondence and reprint requests to Dr. Stephen C. Jameson, Department of Laboratory Medicine and Pathology, Center for Immunology, MMC 334, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455. E-mail address: james024{at}umn.edu

  • ↵3 Abbreviations used in this paper: bio, biotinylated; SA, streptavidin.

  • Received August 22, 2001.
  • Accepted September 19, 2001.
  • Copyright © 2001 by The American Association of Immunologists

References

  1. ↵
    Freitas, A. A., B. Rocha. 2000. Population biology of lymphocytes: the flight for survival. Annu. Rev. Immunol. 18: 83
    OpenUrlCrossRefPubMed
  2. ↵
    Marrack, P., J. Bender, D. Hildeman, M. Jordan, T. Mitchell, M. Murakami, A. Sakamoto, B. C. Schaefer, B. Swanson, J. Kappler. 2000. Homeostasis of αβ TCR+ T cells. Nat. Immun. 1: 107
    OpenUrlCrossRef
  3. ↵
    Viret, C., F. S. Wong, C. A. J. Janeway. 1999. Designing and maintaining the mature TCR repertoire: the continuum of self-peptide:self-MHC complex recognition. Immunity 10: 559
    OpenUrlCrossRefPubMed
  4. ↵
    Bender, J., T. Mitchell, J. Kappler, P. Marrack. 1999. CD4(+) T cell division in irradiated mice requires peptides distinct from those responsible for thymic selection. J. Exp. Med. 190: 367
    OpenUrlAbstract/FREE Full Text
  5. ↵
    Goldrath, A. W., M. J. Bevan. 1999. Low-affinity ligands for the TCR drive proliferation of mature CD8+ T cells in lymphopenic hosts. Immunity 11: 183
    OpenUrlCrossRefPubMed
  6. ↵
    Ernst, B., D.-S. Lee, J. M. Chang, J. Sprent, C. D. Surh. 1999. The peptide ligands mediating positive selection in the thymus control T cell survival and homeostatic proliferation in the periphery. Immunity 11: 173
    OpenUrlCrossRefPubMed
  7. ↵
    Kieper, W. C., S. C. Jameson. 1999. Homeostatic expansion and phenotypic conversion of naive T cells in response to self peptide/MHC ligands. Proc. Natl. Acad. Sci. USA 96: 13306
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Cho, B. K., V. P. Rao, Q. Ge, H. N. Eisen, J. Chen. 2000. Homeostasis-stimulated proliferation drives naive T cells to differentiate directly into memory T cells. J. Exp. Med. 192: 549
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Surh, C. D., J. Sprent. 2000. Homeostatic T cell proliferation: how far can T cells be activated to self-ligands?. J. Exp. Med. 192: F9
    OpenUrlFREE Full Text
  10. ↵
    Salomon, B., J. A. Bluestone. 2001. Complexities of CD28/B7: CTLA-4 costimulatory pathways in autoimmunity and transplantation. Annu. Rev. Immunol. 19: 225
    OpenUrlCrossRefPubMed
  11. ↵
    van Kooten, C., J. Banchereau. 2000. CD40-CD40 ligand. J. Leukocyte Biol. 67: 2
    OpenUrlAbstract
  12. ↵
    Watts, T. H., M. A. DeBenedette. 1999. T cell co-stimulatory molecules other than CD28. Curr. Opin. Immunol. 11: 286
    OpenUrlCrossRefPubMed
  13. ↵
    Mueller, D. L.. 2000. T cells: a proliferation of costimulatory molecules. Curr. Biol. 10: R227
    OpenUrlCrossRefPubMed
  14. ↵
    Coyle, A. J., J. C. Gutierrez-Ramos. 2001. The expanding B7 superfamily: increasing complexity in costimulatory signals regulating T cell function. Nat. Immun. 2: 203
    OpenUrlCrossRefPubMed
  15. ↵
    Tan, J. T., J. K. Whitmire, K. Murali-Krishna, R. Ahmed, J. D. Altman, R. S. Mittler, A. Sette, T. C. Pearson, C. P. Larsen. 2000. 4-1BB costimulation is required for protective anti-viral immunity after peptide vaccination. J. Immunol. 164: 2320
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Gramaglia, I., D. Cooper, K. T. Miner, B. S. Kwon, M. Croft. 2000. Co-stimulation of antigen-specific CD4 T cells by 4-1BB ligand. Eur. J. Immunol. 30: 392
    OpenUrlCrossRefPubMed
  17. ↵
    Blazar, B. R., B. S. Kwon, A. Panoskaltsis-Mortari, K. B. Kwak, J. J. Peschon, P. A. Taylor. 2001. Ligation of 4-1BB (CDw137) regulates graft-versus-host disease, graft-versus-leukemia, and graft rejection in allogeneic bone marrow transplant recipients. J. Immunol. 166: 3174
    OpenUrlAbstract/FREE Full Text
  18. ↵
    Cannons, J. L., P. Lau, B. Ghumman, M. A. DeBenedette, H. Yagita, K. Okumura, T. H. Watts. 2001. 4-1Bb ligand induces cell division, sustains survival, and enhances effector function of CD4 and CD8 T cells with similar efficacy. J. Immunol. 167: 1313
    OpenUrlAbstract/FREE Full Text
  19. ↵
    DeBenedette, M. A., T. Wen, M. F. Bachmann, P. S. Ohashi, B. H. Barber, K. L. Stocking, J. J. Peschon, T. H. Watts. 1999. Analysis of 4-1BB ligand (4-1BBL)-deficient mice and of mice lacking both 4-1BBL and CD28 reveals a role for 4-1BBL in skin allograft rejection and in the cytotoxic T cell response to influenza virus. J. Immunol. 163: 4833
    OpenUrlAbstract/FREE Full Text
  20. ↵
    Murphy, K. M., A. B. Heimberger, D. Y. Loh. 1990. Induction by antigen of intrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo. Science 250: 1720
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Khoruts, A., A. Mondino, K. A. Pape, S. L. Reiner, M. K. Jenkins. 1998. A natural immunological adjuvant enhances T cell clonal expansion through a CD28-dependent, interleukin (IL)-2-independent mechanism. J. Exp. Med. 187: 225
    OpenUrlAbstract/FREE Full Text
  22. ↵
    Kieper, W. C., M. Prlic, C. S. Schmidt, M. F. Mescher, S. C. Jameson. 2001. IL-12 enhances CD8 T cell homeostatic expansion. J. Immunol. 166: 5515
    OpenUrlAbstract/FREE Full Text
  23. ↵
    DeBenedette, M. A., A. Shahinian, T. W. Mak, T. H. Watts. 1997. Costimulation of CD28− T lymphocytes by 4-1BB ligand. J. Immunol. 158: 551
    OpenUrlAbstract
  24. ↵
    Tan, J. T., J. K. Whitmire, R. Ahmed, T. C. Pearson, C. P. Larsen. 1999. 4-1BB ligand, a member of the TNF family, is important for the generation of antiviral CD8 T cell responses. J. Immunol. 163: 4859
    OpenUrlAbstract/FREE Full Text
  25. ↵
    McAdam, A. J., B. E. Gewurz, E. A. Farkash, A. H. Sharpe. 2000. Either B7 costimulation or IL-2 can elicit generation of primary alloreactive CTL. J. Immunol. 165: 3088
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Howland, K. C., L. J. Ausubel, C. A. London, A. K. Abbas. 2000. The roles of CD28 and CD40 ligand in T cell activation and tolerance. J. Immunol. 164: 4465
    OpenUrlAbstract/FREE Full Text
  27. ↵
    Mason, D., F. Powrie. 1998. Control of immune pathology by regulatory T cells. Curr. Opin. Immunol. 10: 649
    OpenUrlCrossRefPubMed
  28. ↵
    Shevach, E. M.. 2001. Certified professionals: CD4+CD25+ suppressor T cells. J. Exp. Med. 193: F41
    OpenUrlFREE Full Text
  29. ↵
    Salomon, B., D. J. Lenschow, L. Rhee, N. Ashourian, B. Singh, A. Sharpe, J. A. Bluestone. 2000. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity 12: 431
    OpenUrlCrossRefPubMed
  30. ↵
    Goronzy, J. J., C. M. Weyand. 2001. Thymic function and peripheral T-cell homeostasis in rheumatoid arthritis. Trends Immunol. 22: 251
    OpenUrlCrossRefPubMed
  31. ↵
    Schluns, K. S., W. C. Kieper, S. C. Jameson, L. Lefrancois. 2000. Interleukin-7 mediates the homeostasis of naive and memory CD8 T cells in vivo. Nat. Immun. 1: 426
    OpenUrlCrossRefPubMed
  32. ↵
    Tan, J. T., E. Dudl, E. LeRoy, R. Murray, J. Sprent, K. I. Weinberg, C. D. Surh. 2001. IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc. Natl. Acad. Sci. USA 10: 10
    OpenUrlCrossRef
  33. ↵
    Goldrath, A. W., M. J. Bevan. 1999. Selecting and maintaining a diverse T-cell repertoire. Nature 402: 255
    OpenUrlCrossRefPubMed
  34. ↵
    Lin, H., J. C. Rathmell, G. S. Gray, C. B. Thompson, J. M. Leiden, M. L. Alegre. 1998. Cytotoxic T lymphocyte antigen 4 (CTLA4) blockade accelerates the acute rejection of cardiac allografts in CD28-deficient mice: CTLA4 can function independently of CD28. J. Exp. Med. 188: 199
    OpenUrlAbstract/FREE Full Text
  35. ↵
    Blazar, B. R., P. A. Taylor, A. Panoskaltsis-Mortari, A. H. Sharpe, D. A. Vallera. 1999. Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. J. Immunol. 162: 6368
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Thornton, A. M., E. M. Shevach. 1998. CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. J. Exp. Med. 188: 287
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Thornton, A. M., E. M. Shevach. 2000. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J. Immunol. 164: 183
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Taylor, P. A., R. J. Noelle, B. R. Blazar. 2001. CD4(+)CD25(+) immune regulatory cells are required for induction of tolerance to alloantigen via costimulatory blockade. J. Exp. Med. 193: 1311
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Jordan, M. S., A. Boesteanu, A. J. Reed, A. L. Petrone, A. E. Holenbeck, M. A. Lerman, A. Naji, A. J. Caton. 2001. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nat. Immun. 2: 301
    OpenUrlCrossRef
  40. ↵
    Goldrath, A. W., L. Y. Bogatzki, M. J. Bevan. 2000. Naive T cells transiently acquire a memory-like phenotype during homeostasis-driven proliferation. J. Exp. Med. 192: 557
    OpenUrlAbstract/FREE Full Text
  41. ↵
    Murali-Krishna, K., R. Ahmed. 2000. Cutting edge: naive T cells masquerading as memory cells. J. Immunol. 165: 1733
    OpenUrlAbstract/FREE Full Text
  42. ↵
    Gudmundsdottir, H., L. A. Turka. 2001. A closer look at homeostatic proliferation of CD4+ T cells: costimulatory requirements and role in memory formation. J. Immunol. 167: 3699
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top

In this issue

The Journal of Immunology: 167 (10)
The Journal of Immunology
Vol. 167, Issue 10
15 Nov 2001
  • Table of Contents
  • About the Cover
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word about The Journal of Immunology.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
Homeostatic Expansion Occurs Independently of Costimulatory Signals
(Your Name) has forwarded a page to you from The Journal of Immunology
(Your Name) thought you would like to see this page from the The Journal of Immunology web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Homeostatic Expansion Occurs Independently of Costimulatory Signals
Martin Prlic, Bruce R. Blazar, Alexander Khoruts, Traci Zell, Stephen C. Jameson
The Journal of Immunology November 15, 2001, 167 (10) 5664-5668; DOI: 10.4049/jimmunol.167.10.5664

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Homeostatic Expansion Occurs Independently of Costimulatory Signals
Martin Prlic, Bruce R. Blazar, Alexander Khoruts, Traci Zell, Stephen C. Jameson
The Journal of Immunology November 15, 2001, 167 (10) 5664-5668; DOI: 10.4049/jimmunol.167.10.5664
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

Cited By...

More in this TOC Section

  • Innate Immunity Together with Duration of Antigen Persistence Regulate Effector T Cell Induction
  • Regulatory Roles of IL-2 and IL-4 in H4/Inducible Costimulator Expression on Activated CD4+ T Cells During Th Cell Development
  • Induction of CD4+ T Cell Apoptosis as a Consequence of Impaired Cytoskeletal Rearrangement in UVB-Irradiated Dendritic Cells
Show more CELLULAR IMMUNOLOGY AND IMMUNE REGULATION

Similar Articles

Navigate

  • Home
  • Current Issue
  • Next in The JI
  • Archive
  • Brief Reviews
  • Pillars of Immunology
  • Translating Immunology

For Authors

  • Submit a Manuscript
  • Instructions for Authors
  • About the Journal
  • Journal Policies
  • Editors

General Information

  • Advertisers
  • Subscribers
  • Rights and Permissions
  • Accessibility Statement
  • Public Access
  • Privacy Policy
  • Disclaimer

Journal Services

  • Email Alerts
  • RSS Feeds
  • ImmunoCasts
  • Twitter

Copyright © 2021 by The American Association of Immunologists, Inc.

Print ISSN 0022-1767        Online ISSN 1550-6606