Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Aug 1.
Published in final edited form as: Curr Opin Organ Transplant. 2014 Aug;19(4):357–362. doi: 10.1097/MOT.0000000000000096

HOMEOSTATIC EXPANSION AS A BARRIER TO LYMPHOCYTE DEPLETION STRATEGIES

Nicholas A Zwang 1, Laurence A Turka 2
PMCID: PMC4476295  NIHMSID: NIHMS634455  PMID: 24926701

Introduction

Purpose of review

Following lymphodepletion, lymphocytes repopulate the immune space both through enhanced thymopoiesis and proliferation of residual non-depleted peripheral lymphocytes. The term homeostatic proliferation (alternatively homeostatic expansion or lymphopenia-induced proliferation) refers to the latter process. Homeostatic proliferation is especially relevant to reconstitution of the lymphocyte compartment following immunodepletion therapy in transplantation. Repopulating lymphocytes can skew toward an effector memory type capable of inducing graft rejection, autoimmunity, or, in the case of allogeneic bone marrow transplantation, graft versus host disease. Here we review recent studies exploring the biologic mechanisms underlying homeostatic proliferation and explore implications for therapy in transplantation.

Recent findings

Two immune-depleting agents, alemtuzumab and rabbit antithymocyte globulin, have been well-characterized in their abilities to induce an effector-memory phenotype in repopulating lymphocytes. Additionally, we have gained new understandings of the mechanisms by which the cytokines Interleukin-7 (IL-7) and Interleukin-15 (IL-15) regulate this process. Recent studies have also explored the functions of non-cytokine and signaling molecules in lymphopnenia-induced proliferation. Finally, we have seen the promise and limitations of several therapeutic approaches, including recombinant IL-7 therapy, CD8+-targeted antibodies, and peri-transplant cyclophosphamide, to treat post-transplant lymphopenia and reduce the risks of immune dysregulation following homeostatic proliferation.

Summary

Immune dysfunction following homeostatic proliferation is a special challenge in transplantation. A deeper understanding of the underlying biology has led to a number of promising new therapies to overcome this problem.

Keywords: Homeostatic proliferation, transplantation, lymphodepletion, autoimmunity

I. Introduction

Following lymphodepletion, lymphocytes repopulate the immune space both through enhanced thymopoiesis and proliferation of residual non-depleted peripheral lymphocytes. The term homeostatic proliferation (alternatively homeostatic expansion or lymphopenia-induced proliferation) refers to the latter process. Early studies of homeostatic proliferation showed that T cells surviving lymphodepletion divided, developed memory phenotype and function, and then acted in a dominant fashion to render animals resistant to cardiac or renal allograft tolerance via costimulatory blockade.1,2 In line with these findings, recent studies have shown that lymphopenia itself is enough to break stable costimulatory blockade-based peripheral tolerance.3 In a mouse model of MHC-mimatched cardiac transplantation, lymphopenia (achieved either by irradiation or anti-CD4+/CD8+ monoclonal antibodies) induced acute T and B cell-mediated rejection, accompanied by a T cell shift toward a CD44hi effector-memory (EM) phenotype and the appearance of donor-specific antibodies. The process of homeostatic proliferation can be divided into “slow” (one cell division per 24–36 hours) or “fast” (one division per 6–8h) kinetics. While slow proliferation occurs in response to a “sensing of empty space”, rapid proliferation is primarily a gut antigen-driven process.4 Slow homeostatic proliferation predominates in homeostatic proliferation following lymphodepletion in mouse models. Furthermore, both T and B cells can undergo homeostatic proliferation. In this review, we will focus on recent developments in the slow homeostatic proliferation of T cells following lymphodepletion, which we believe is most relevant to understanding rejection and autoreactivity following transplantation.

II. Pharmacologic lymphocyte depletion promotes autoimmunity and the expansion of alloreactive lymphocytes

Alemtuzumab (anti-CD52) is a potent lymphocyte depletional agent that has been used as induction therapy for transplantation and for treatment of multiple sclerosis. CD4+ cells and, to a lesser extent, naïve CD8+ cells, are most susceptible to alemtuzumab-induced lymphodepletion.5,6,7,8 A larger population of naïve T cells may remain undeleted, however, as peripheral lymph nodes may be a reservoir for these cells following alemtuzumab induction.9 Alemtuzumab therapy leads to skewing toward memory CD4+ and CD8+ phenotypes in renal transplant recipients; those with evidence of rejection (by biopsy, new or donor-specific antibodies) following alemtuzumab therapy have an increased proportion of CD8+ effector memory cells (CD45ROCD62L).10 These same patients further have decreased frequencies of regulatory T cells (Tregs) among CD4+ cells. While other work, in contrast, has suggested an increased frequency of Foxp3+ cells following alemtuzumab induction.11 It is possible that in this instance Foxp3 expression may be only a transient marker of T cell activation.12,13, 14 Among patients with multiple sclerosis, homeostatic proliferation following alemtuzumab therapy leads to recovery of a highly activated, proliferative, oligoclonal, and memory-like population of CD4+ and CD8+ cells.15 In particular, the CD8 pool is dominated by a terminally-differentiated, effector memory CD28CD57+CD8 population expressing perforin and Granzyme B. Such as population is known to be associated with autoimmunity, and indeed in this study of 87 patients, two thirds developed (primarily thyroid) autoimmunity.

Rabbit antihymocyte globulin (rATG) depletes naïve but not central memory or effector memory T cell subsets in kidney transplant patients.16 Early studies of immune monitoring of CD4+ cells following ATG induction has indeed revealed a residual population of CD25+CD45RO+CD45RA (i.e., EM) cells but also transient expression of Foxp3,17 which, again, may only be a marker of CD4+ activation. Nonetheless, administration of mouse ATG in a mouse model of Treg-dependent autoimmune diabetes increased the prevalence of Foxp3+ and CTLA-4+ cells while delaying the onset of autoimmunity.18 Perhaps Treg induction (transiently) counterbalances memory T cell expansion following ATG induction. Recent examination of the kinetics of lymphocyte depletion following rATG given as induction therapy in renal transplantation found that rATG durably depletes the T cell compartment to counts below 250 CD3+ cells/uL at six months, compared to minimal T cell depletion following basiliximab or no induction therapy.19 In contrast to prior studies, this recent investigation found no increase in thymopoiesis (i.e., CD31+ cells among CD4+ or CD8+ cells) one month following rATG induction. Rather, peripheral cytokine-mediated signaling by IL-7 and IL-15 via Stat5 increased in the first month following rATG therapy, particularly among memory T cell subsets. These studies indicate that T cell recovery following ATG comes from peripheral T cell pools rather than heightened thymopoiesis.

III. New insights into the immunobiology of homeostatic proliferation: IL-7, IL-15, and non-cytokine regulators

In humans, unlike mice, the majority of proliferating T cells derives from the periphery rather than the thymus.20 Therefore, peripheral cytokine signaling is essential to maintain the lymphoreplete state and repopulate the T cell compartment in lymphopenia. IL-7 is the primary cytokine responsible for T cell homeostatic proliferation. In young thymectomized and elderly adults, circulating IL-7 levels are higher than those of healthy controls.21 IL-7 in these patients with low or no thymic function appears to stimulate T cell proliferation via STAT5 signaling. IL-7 itself has been described as a “rheostat” to maintain the T cell compartment.22 In lymphopenia, excess IL-7 stimulates T cell proliferation. Proliferating T cells consume IL-7, and levels fall to the basal state as the T cell compartment repopulates. This mechanism prevents excess proliferation and preserves T cell homeostasis. A recent study found that IL-7-induced proliferation requires intermittent (rather than continuous) signaling and that TCR engagement provides this interruption.23 T cells with inadequate affinity for peripheral (self) TCR ligands die following prolonged IL-7 signaling; this mechanism maintains a population of T cells with appropriate affinity for self ligands.

In addition to IL-7, IL-15 signaling is important for CD8+ T cell survival and proliferation.24,25,26 While IL-15 enhances homeostatic proliferation of memory CD8+ cells, IL-15 alone is not enough for homeostatic proliferation of naïve CD8 T cells. 27 In naïve CD8+ cells, MHC I engagement is also necessary for homeostatic proliferation.28 Emerging data show that memory CD4+ may also be responsive to IL-15.29,30,31 Finally, TGB-β may attenuate IL-15 signaling and act as a brake on homeostatic proliferation-driven autoimmunity.32,33, 34,35,36,37

In the past year, a number of studies have shown the importance of cell-intrinsic signaling, non-cytokine TCR regulation, and trafficking molecules in regulating homeostatic proliferation.

The protein tyrosine phosphatase gene product PTPN2, which dampens TCR signaling in CD4+ and CD8+ cells, is implicated in human autoimmunity.38,39 T cell knockout of PTPN2 in a mouse model resulted in more rapid lymphopenia-induced CD8+ proliferation compared to control animals. Adoptive transfer of PTPN2-deleted CD8+ cells into congenic hosts resulted in effector/memory differentiation and autoimmunity compared to adoptive transfer of control CD8+ cells..40 This response was IL-7-independent. miRNA-181a enhances TCR signaling, in part by suppressing expression of other protein phosphatases.41 Thus, miRNA-181 or another miRNA might inhibit PTPN2 expression and thereby dampen lymphopenia-induced proliferation.

It has been suggested that transcription factors may regulate the ability of hematopoietic stem cells to repopulate the lymphocyte compartment. For example, Hoxb4 signaling may promote a hematopoietic stem cell CD4+ central memory (CD44hiCD62L+) phenotype in response to lymphopenia.42 In competitive adoptive transfer experiments, Hoxb4-overexpressing central memory cells contributed less than wild-type central memory cells to reconstitution of lymphoid organs.

Finally, the integrin CD18 (lymphocyte function-associated antigen-1, or LFA-1) functions in naïve T cell trafficking between the gut and secondary lymphoid organs43,44 and is implicated in gut autoimmunity.45 Adoptive transfer of CD4+ CD18−/− cells into Rag−/− hosts has shown the requirement of CD18 both for fast and slow lymphopenia-induced proliferation.46 The above studies have illustrated the importance of non-cytokine regulators of homeostatic proliferation that skew toward an effector memory phenotype in homeostatic proliferation.

IV. Clinical implications for autoimmunity after treatment of lymphopenia and strategies to prevent autoreactivity

Recombinant IL-7 has come into use to promote T cell recovery in lymphopenia. The therapeutic goal has been to expand the population of mature lymphocytes rather than to induce thymopoiesis.47,48 While studies of T cell subsets suggest that IL-7 administration to humans with refractory malignancies increases naïve T cell fractions,49 these results need to be interpreted with caution. IL-7 administration in lymphopenic hosts may have markedly different effects. A recent Phase I trial of IL-7 administration in allo-HSCT skewed repopulating CD4+ and CD8+ cells toward an effector-memory (CD45RACCR7) phenotype.50 Recent studies of IL-7 administration to mice have suggested that CD4+ single-positive (miR181a -expressing) thymocytes are more sensitive to IL-7-induced proliferation than are peripheral CD4+ cells.51 In these animals, peripheral lymphopenic CD4+ proliferation in response to IL-7 had a strong TCR co-stimulation requirement; these proliferating cells were primarily slow rather than fast-proliferating cells. These data suggest that thymopoiesis provides an important contribution to resolving lymphopenia and that IL-7 therapy affects slowly rather than quickly-proliferating cells under conditions of homeostatic proliferation. Prior studies have suggested that PI3K signaling is essential for proliferation of these CD4+ cells, particularly in CD31+ recent thymic emigrants.52,53

Another potential approach to overcoming homeostatic proliferation as a barrier to transplantation is to delete potentially pathologic CD8+ cells specifically in transplant recipients. Yamada et al employed this approach with the use of anti-CD8 mAbs at the time of lymphodepletion in a mixed chimerism model of MHC mismatched renal transplantation in nonhuman primates;54 their findings of decreased Tmem responses in CD8-depleted antimals are encouarging. The same group subsequently studied alefacept, a fusion protein of the extracellular CD2-binding portion of the human leukocyte function antigen-3 (LFA-3) adhesion molecule.55 This agent is thought to interrupt cytotoxic effector memory T cell proliferation by blocking the interaction between effector-memory CD2+ cells and LFA-3. Alefacept therapy for psoriasis preferentially depleted CD4+CD45RO+ effector memory cells, which correlated with clinical improvement in skin lesions.56 Alefacept preferentially and reversibly depleted CD8+ effector memory (CD28CD95+) cells in a nonhuman primate transplantation model57; CD28 cells in this model were CD2hi, helping to explain alefacept’s ability to preferentially deplete CD8+ cells.

Post-transplant cyclophosphamide administration is an attractive approach to prevent GVHD by depleting alloreactive CD8+ cells that might otherwise survive induction therapy.58,59 Recent data suggest that post-transplant cyclophosphamide administration primarily targets rapidly-dividing allo-specific cells, relatively sparing naïve cells essential to maintenance of immunocompetence following HSCT.60 CD4+Foxp3+ Tregs appear resistant to cyclophosphamide and recover quickly following cyclophosphamide induction for allogeneic bone marrow transplantation.61 Sparing of Tregs may partly underlie the mechanism by which cyclophosphamide prevents GVHD.

Finally, in contrast to Tconv cells, Tregs require IL-2 is essential for Treg survival and proliferation, both in lymphopenia and the lymphoreplete state.62 This observation has led to low-dose IL-2 as therapy to promote Treg proliferation in hepatitis C-associated vasculitis63 and GVHD following stem cell transplantation.64 IL-2 therapy for GVHD restores pStat5 signaling and proliferation of Tregs without affecting Tconvs.65 Thus, Treg-specific therapy may ultimately find a place in transplant immunosuppression protocols alongside more generalized lymphodepletion strategies and targeted effector-memory cell deletion.

V. Conclusion

Lymphodepletion for induction transplant therapy is double-edged sword. On the one hand, highly effective agents such as alemtuzumab and rATG are able to prevent early T cell mediated rejection. On the other hand, repopulated T cells skew toward allo-specific, effector-memory phenotypes following lymphodepletion. An interesting concept in tumor immunobiology turns the concept of homeostatic proliferation on its head. The conditions of homeostatic proliferation could be used to cultivate CD8+ cells specific to tumor antigens, allowing for specific immunotherapy.66,67 This type of approach remains experimental in mouse models. But exploring the spectrum of homeostatic proliferation can improve our understanding of this important phenomenon, not only as it relates to transplantation but also to HIV therapy and autoimmunity. The use of recombinant IL-7 in transplantation treatment of lymphopenia may be a tight rope to walk if this therapy skews repopulating T cells toward an effector memory phenotype capable of autoreactivity. Alternatively, targeted deletion of effector memory CD8+ cells upon transplantation—with depleting antibodies or cyclophosphamide—is an intriguing way to solve this problem, so long as these approaches do not increase the risks of infection unacceptably. Finally, the role of Tregs in controlling autoimmunity or alloreactivity following lyphodepletion merits further investigation. IL-2 therapy may not adequately enhance Treg function to overcome the problems of homeostatic proliferation. A deeper understanding of Treg biology, however, may ultimately lead to Treg-specific therapies that ameliorate the EM-skewing induced by lymphodepletion.

Key Points.

  • Repopulating lymphocytes can skew toward an effector memory type capable of inducing graft rejection, autoimmunity, or, in the case of allogeneic bone marrow transplantation, graft versus host disease.

  • In the past year, we have gained new understandings of the mechanisms by which the cytokines Interleukin-7 (IL-7) and Interleukin-15 (IL-15) regulate this process. Recent studies have also explored the functions of non-cytokine and signaling molecules in lymphopnenia-induced proliferation.

  • Additionally, we have seen the promise and limitations of several therapeutic approaches, including recombinant IL-7 therapy, CD8+-targeted antibodies, and peri-transplant cyclophosphamide, to treat post-transplant lymphopenia and reduce the risks of immune dysregulation following homeostatic proliferation.

Acknowledgments

N.A.Z would like to thank the Brigham and Women’s Hospital/Massachusetts General Hospital Joint Nephrology Fellowship Program and its leadership for their ongoing support.

Abbreviations

EM

Effector memory

HP

homeostatic proliferation

GVHD

Graft versus host disease

IL-2

Interleukin-2

IL-7

Interleukin-7

IL-15

Interleukin-15

Tmem

memory T cell

rATG or ATG

rabbit antithymocyte globulin

Treg

regulatory T cell

TCR

T cell receptor

Footnotes

Conflict of Interest

Laurence Turka owns equity in and has a family member employed by Novartis.

References

  • 1.Wu Z, Bensinger SJ, Zhang J, et al. Homeostatic proliferation is a barrier to transplantation tolerance. Nat Med. 2004;10:97–92. doi: 10.1038/nm965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Moxham VF, Karegli J, Phillips RE, et al. Homeostatic proliferation of lymphocytes results in augmented memory-like function and accelerated allograft rejection. J Immunol. 2008;180:3910–3918. doi: 10.4049/jimmunol.180.6.3910. [DOI] [PubMed] [Google Scholar]
  • 3*.Iida S, Suzuki T, Tanabe K, et al. Transient lymphopenia breaks costimulatory blockade-based peripheral tolerance and initiates cardiac allograft rejection. American J Transplant. 2013;13:2268–2279. doi: 10.1111/ajt.12342. This study demonstrated an important limitation for costimulation blockade. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Tchao NK, Turka LA. Lymphodepletion and homeostatic proliferation: implications for transplantation. Am J Transplant. 2012;12:1079–1090. doi: 10.1111/j.1600-6143.2012.04008.x. [DOI] [PubMed] [Google Scholar]
  • 5.Lowenstein H, Shah A, Chant A, Khan A. Different mechanisms of Campath-1H mediated depletion for CD4 and CD8 T cells in peripheral blood. Transplant Int. 2006;19:927–936. doi: 10.1111/j.1432-2277.2006.00382.x. [DOI] [PubMed] [Google Scholar]
  • 6.Pearl JP, Parris J, Hale DA, et al. Immunocompetent T-cells with a memory-like phenotype are the dominant cell type following antibody-mediated T-cell depletion. Am J Transplant. 2005;5:465–474. doi: 10.1111/j.1600-6143.2005.00759.x. [DOI] [PubMed] [Google Scholar]
  • 7.Trzonkowski P, Zilvetti M, Chapman S, et al. Homeostatic repopulation by CD28−CD8+ T cells in alemtuzumab-depleted kidney transplant recipients treated with reduced immunosuppression. Am J Transplant. 2008;8:338–347. doi: 10.1111/j.1600-6143.2007.02078.x. [DOI] [PubMed] [Google Scholar]
  • 8.van der Windt DJ, Smetanka C, Macedo C, et al. Investigation of lymphocyte depletion and repopulation using alemtuzumab (Campath-1H) in cynomolgus monkeys. Am J Transplant. 2010;10:773–783. doi: 10.1111/j.1600-6143.2010.03050.x. [DOI] [PubMed] [Google Scholar]
  • 9.Marco MRL, Dons EM, van der Windt DJ, et al. Post-transplant repopulation of naïve and memory T cells in blood and lymphoid tissue after alemtuzumab-mediated depletion in heart-transplanted cynomolgus monkeys. Transplant Immunol. 2013;29:88–98. doi: 10.1016/j.trim.2013.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Macedo C, Walters JT, Orkis EA, et al. Long-term effects of alemtuzumab on regulatory and memory T-cell subsets in kidney transplantation. Transplantation. 2012;93:813–821. doi: 10.1097/TP.0b013e318247a717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Bloom DD, Chang Z, Fechner JH, et al. CD4+CD25+Foxp3+ regulatory T cells increase de novo in kidney transplant patients after immunodepletion with Campath-1H. Am J Transplant. 2008;8:792–802. doi: 10.1111/j.1600-6143.2007.02134.x. [DOI] [PubMed] [Google Scholar]
  • 12.Wang J, Ioan-Facsinay A, van der Voort EI, et al. Transient epression of FOXP3 in human activated nonregulatory CD4+ T cells. Eur J Immunol. 2007;37:129–138. doi: 10.1002/eji.200636435. [DOI] [PubMed] [Google Scholar]
  • 13.Tran DQ, Ramsey H, Shevach EM. Induction of FOXP3 expression in naïve human CD4+Foxp3 T cells by T-cell receptor stimulation is transforming growth factor-beta dependent but does not confer a regulatory phenotype. Blood. 2007;110:2983–2990. doi: 10.1182/blood-2007-06-094656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Broady R, Yu J, Levings MK. ATG-induced expression of FOXP3 in human CD4+ T cells is associated with T-cell activation and not the induction of FOXP3+ T regulatory cells. Blood. 2009;114:5003–5006. doi: 10.1182/blood-2009-04-214437. [DOI] [PubMed] [Google Scholar]
  • 15**.Jones JL, Thompson SAJ, Loh P, et al. Human autoimmunity after lymphocyte depletion is caused by homeostatic T-cell proliferation. PNAS. 2013;110:20200–20205. doi: 10.1073/pnas.1313654110. This unique study demonstrated the possibility that alemtuzumab therapy can promote autoimmunity. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gurkan S, Luan Y, Dhillon N, et al. Immune reconstitution following rabbit antithymocyte globulin. Am J Transplant. 2010;10:2132–2141. doi: 10.1111/j.1600-6143.2010.03210.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Serban G, Whittaker V, Fan J, et al. Significance of immune cell function monitoring in renal transplantation after thymoglobin induction therapy. Human Immunol. 2009;70:882–890. doi: 10.1016/j.humimm.2009.07.027. [DOI] [PubMed] [Google Scholar]
  • 18.Lu Y, Suzuki J, Guillioli M, et al. Induction of self-antigen –specific Foxp3+ regulatory T cells in the periphery by lymphodepletion treatment with anti-mouse thymocyte globulin in mice. Immunology. 2001;134:50–59. doi: 10.1111/j.1365-2567.2011.03466.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19*.Bouvy AP, Kho MML, Klepper M, et al. Kinetics of homeostatic proliferation and thymopoiesis after rATG induction therapy in kidney transplant patients. Transplantation. 2013;96:904–913. doi: 10.1097/TP.0b013e3182a203e4. A relevant examination of the immunophenotypic conesequences of rATG lymphodepletion and T cell repopulation. [DOI] [PubMed] [Google Scholar]
  • 20.Den Braber I, Mugwagwa T, Vrisekoop N, et al. Maintenance of peripheral naïve T cells is sustained by thymus output in mice but not humans. Immunity. 2012;36:288–297. doi: 10.1016/j.immuni.2012.02.006. [DOI] [PubMed] [Google Scholar]
  • 21.Sauce D, Larsen M, Fastenackels S, et al. Lymphopenia-driven homeostatic regulation of naïve T cells in elderly and thymectomized young adults. J Immunol. 2012;189:5541–5548. doi: 10.4049/jimmunol.1201235. [DOI] [PubMed] [Google Scholar]
  • 22.Tchao NK, Turka LA. Lymphodepletion and homeostatic proliferation: implications for transplantation. Am J Transplant. 2012;12:1079–1090. doi: 10.1111/j.1600-6143.2012.04008.x. [DOI] [PubMed] [Google Scholar]
  • 23**.Kimura M, Pobezinsky LA, Guinter TI, et al. IL-7 signaling must be intermittent, not continuous, during CD8+ T cell homeostasis to promote cell survival instead of cell death. Nature Immunol. 2013;14:143–152. doi: 10.1038/ni.2494. A very interesting insight into the nature of IL-7 signaling and its implications for maintaining appropriate affinity to self-ligands. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Berger C, Jensen MC, Lansdorp PM, et al. Adoptive transfer of effector CD8 T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118:294–305. doi: 10.1172/JCI32103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Schluns KS, Williams K, Ma A, et al. Cutting edge: requirement for IL-15 in the generation of primary and memory antigen-specific CD8 T cells. J Immunol. 2002;168:4827–4831. doi: 10.4049/jimmunol.168.10.4827. [DOI] [PubMed] [Google Scholar]
  • 26.Becker TC, Wherry EJ, Boone D, et al. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp Med. 2002;195:1541–1548. doi: 10.1084/jem.20020369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Goldrath AW, Sivakumar PV, Glaccum M, et al. Cytokine requirements for acute and basal homeostatic proliferation of naïve and memory CD8+ T cells. J Exp Med. 2002;195:1515–1522. doi: 10.1084/jem.20020033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Stoklasek TA, Colpitts SL, Smilowitz HM, Lefrançois L. MHC class I and TCR avidity control the CD8 T cell response to IL-15/IL15Rα complex. J Immunol. 2010;185:6857–6865. doi: 10.4049/jimmunol.1001601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Purton JF, Tan JT, Rubinstein MP, et al. Antiviral CD4+ memory T cells are IL-15 dependent. J Exp Med. 2007;204:951–961. doi: 10.1084/jem.20061805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Huntington ND, Alves NL, Legrand N, et al. IL-15 transpresentation promotes both human T-cell reconstitution and T-cell-dependent antibody responses in vivo. Proc Natl Acad Sci USA. 2011;108:6217–6222. doi: 10.1073/pnas.1019167108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Van Belle TL, Dooms H, Boonefaes T, et al. IL-15 augments TCR-induced CD4+ T cell expansion in vitro by inhibiting the suppressive function of CD25high CD4+ T cells. PLOS ONE. 2012;7:e45299. doi: 10.1371/journal.pone.0045299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lucas PJ, Kim SJ, Mackall CL, et al. Dysregulation of IL-15 mediated T-cell homeostasis in TGF-beta dominant negative receptor transgenic mice. Blood. 2006;108:2789–2795. doi: 10.1182/blood-2006-05-025676. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sanjabi S, Mosaheb MM, Flavell RA. Opposing effects of TGF-beta and IL-15 cytokines control the number of short-lived effector CD8+ T cells. Immunity. 2009;31:131–144. doi: 10.1016/j.immuni.2009.04.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T-cell dependent and -independent mechanisms. Immunity. 2006;25:455–471. doi: 10.1016/j.immuni.2006.07.011. [DOI] [PubMed] [Google Scholar]
  • 35.Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-beta receptor. Immunity. 2006;25:441–454. doi: 10.1016/j.immuni.2006.07.012. [DOI] [PubMed] [Google Scholar]
  • 36.Zhang N, Bevan MJ, et al. TGF-β signaling to T cells inhibits autoimmunity during lymphopenia-driven proliferation. Nature Immunol. 2012;13:667–674. doi: 10.1038/ni.2319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Johnson LDS, Jameson SC. TGF-β sensitivity restrains CD8+ T cell homeostatic proliferation by enforcing sensitivity to IL-7 and IL-15. PLoS ONE. 2012;7:e42268. doi: 10.1371/journal.pone.0042268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Espino-Paisan L, de la Calle H, Fernández-Arguero M, et al. A polymorphism in PTPN2 gene is associated with an earlier onset of type 1 diabetes. Immunogenetics. 2011;63:255–58. doi: 10.1007/s00251-010-0500-x. [DOI] [PubMed] [Google Scholar]
  • 39.Festen EA, Goyette P, Green T, et al. A meta-analysis of genome-wide association scans identifies IL18RAP, PTPN2, TAGAP, and PUS10 as shared risk loci for Cronh’s disease and celiac disease. PLoS Genet. 2011;7:e1001283. doi: 10.1371/journal.pgen.1001283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40**.Wiede F, La Gruta NL, Tiganis T. PTPN2 attenuates T-cell lymphopenia-induced proliferation. Nature Communications. 2014;5:3073. doi: 10.1038/ncomms4073. The relationship between miRNA modulation of phosphatase activity and downstream signaling is an important and emerging topic. [DOI] [PubMed] [Google Scholar]
  • 41.Li QJ, Chau J, Ebert PJ, et al. miR-181a is an intrinsic modulator of T cell sensitivity and selection. Cell. 2007;129:147–161. doi: 10.1016/j.cell.2007.03.008. [DOI] [PubMed] [Google Scholar]
  • 42*.Frison H, Giono G, Thébault P, et al. Hoxb4 overexpression in CD4 memory phenotype T cells increases the central memory population upon homeostatic proliferation. PLOS ONE. 2013;8:e81573. doi: 10.1371/journal.pone.0081573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Abraham C, Griffith J, Miller J. The dependence for leukocyte function-associated antigen-1/ICAM-1 interactions in T cell activation cannot be overcome by expression of high density TCR ligand. J Immunol. 1999;162:4399–4405. [PubMed] [Google Scholar]
  • 44.Kandula S, Abraham C. LFA-1 on CD4+ T cells is required for optimal antigen-dependent activation in vivo. J Immunol. 2004;173:4443–4451. doi: 10.4049/jimmunol.173.7.4443. [DOI] [PubMed] [Google Scholar]
  • 45.Uzel G, Kleiner DE, Kuhns DB, Holland SM. Dysfunctional LAD-1 neutophils and colitis. Gastroenterology. 2001;121:958–964. doi: 10.1053/gast.2001.28022. [DOI] [PubMed] [Google Scholar]
  • 46.Sarin R, Abraham C. CD18 is required for optimal lymphopenia-induced proliferation of mouse T cells. Am J Physiol Gastrointest Liver Physiol. 2012;303:G851–G860. doi: 10.1152/ajpgi.00520.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Chu YW, Memon SA, Sharrow SO, et al. Exogenous IL-7 increases recent thymic emigrants in peripheral lymphoid tissue without enhanced thymic function. Blood. 2004;104:1110–1119. doi: 10.1182/blood-2003-10-3635. [DOI] [PubMed] [Google Scholar]
  • 48.Rosenberg SA, Sportès C, Ahmazadeh M, et al. IL-7 administration to humans leads to expansion of CD8+ and CD4+ cells but a relative decrease of CD4+ T-regulatory cells. J Immunother. 2006;29:313–319. doi: 10.1097/01.cji.0000210386.55951.c2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sportès C, Hakim FT, Memon SA, et al. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naïve T cell subsets. J Exp Med. 2008;205:1701–1714. doi: 10.1084/jem.20071681. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Perales MA, Goldberg JD, Yuan J, et al. Recombinant human interleukin-7 (CYT107) promotes T-cell recovery after allogenic stem cell transplantation. Blood. 2012;120:4882–4891. doi: 10.1182/blood-2012-06-437236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51*.Hennion-Tscheltzoff O, Leboeuf D, Gauthier SD, et al. TCR triggering modulates the responsiveness and homeostatic proliferation of CD4+ thymic emigrants to IL-7 therapy. Blood. 2013;121:4684–4693. doi: 10.1182/blood-2012-09-458174. Another important contribution to the literature on use of IL-7 therapy in humans. [DOI] [PubMed] [Google Scholar]
  • 52.Swainson L, Kinet S, Mongellaz C, et al. IL-7-induced proliferation of recent thymic emigrants requires activation of the PI3K pathway. Blood. 2007;109:1034–1042. doi: 10.1182/blood-2006-06-027912. [DOI] [PubMed] [Google Scholar]
  • 53.Azevedo RI, Soares MV, Barata JT, et al. IL-7 sustains CD31 expression in human naïve CD4+ T cells and preferentially expands the CD31+ subset in a PI3K-dependent manner. Blood. 2009;113:2999–3007. doi: 10.1182/blood-2008-07-166223. [DOI] [PubMed] [Google Scholar]
  • 54.Yamada Y, Boskovic S, Aoyama A, et al. Overcoming memory T-cell responses for induction of delayed tolerance in nonhuman primates. Am J Transplant. 2012;12:330–340. doi: 10.1111/j.1600-6143.2011.03795.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Lee S, Yamada Y, Tonsho M, et al. Alefacept promotes immunosuppression-free renal allograft survival in nonhuman primates via depletion of recipient memory T cells. Am J Transplant. 2013;13:3223–3229. doi: 10.1111/ajt.12500. This study adds to the growing literature on the use of alefacept as immunotherapy and, here, in solid organ transplantation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ellis CN, Krueger GG Alefacept Clinical Study Group. Treatment of chronic plaque psoriasis by selective targeting of memory effector T lymphocytes. N Engl J Med. 2001;345:248–255. doi: 10.1056/NEJM200107263450403. [DOI] [PubMed] [Google Scholar]
  • 57.Weaver TA, Charafedding AH, Agarwal A, et al. Alefacept promotes co-stimulation blockade based allograft survival in nonhuman primates. Nat Med. 2009;15:746–749. doi: 10.1038/nm.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ciurea SO, Mulanovich V, Saliba RM, et al. Improved early outcomes using a T cell replete graft compared with T cell depleted haploidentical hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2012;18:1835–1844. doi: 10.1016/j.bbmt.2012.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Luznik L, Bolaños-Meade J, Zahurak M, et al. High-dose cyclophosphamide as single-agent, short-course prophylaxis of graft-versus-host disease. Blood. 2010;115:3224–3230. doi: 10.1182/blood-2009-11-251595. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60*.Ross D, Jones M, Komanduri K, Levy RB. Antigen and lyphopenia-driven donor T cells are differentially diminished by post-transplantation administration of cyclophosphamide after hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2013;19:1403–1438. doi: 10.1016/j.bbmt.2013.06.019. A study in mice suggesting that cyclophosphamide may reduce graft versus host disease but preserve immune competence following bone marrow transplantation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61**.Kanakry CG, Ganguly S, Zahurak M, et al. Aldehyde dehydrogenase expression drives human regulatory T cell resistant to posttransplantation cyclophosphamide. Sci Transl Med. 2013;5:211ra157. doi: 10.1126/scitranslmed.3006960. An in-depth study of the mechanism to explain the clinical observation that cyclophosphamide appears to spare Tregs in bone marrow transplantation. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Stouvenel L, Auffray C, Martin B, et al. IL-2 and IL-7 determine the homeostatic balance between the regulatory and conventional CD4+ cell compartments during peripheral T cell reconstitution. J Immunol. 2012;189:3339–3346. doi: 10.4049/jimmunol.1103152. [DOI] [PubMed] [Google Scholar]
  • 63.Saadoun D, Rosenzwaig M, Joly F, et al. Regulatory T-cell responses to low-dose interleukin-2 in HCV-induced vasculitis. N Engl J Med. 2011;365:2067–2077. doi: 10.1056/NEJMoa1105143. [DOI] [PubMed] [Google Scholar]
  • 64.Koreth J, Matsuoka K, Kim HT, et al. Interleukin-2 and regulatory T cells in graft-versus-host disease. N Engl J Med. 2011;365:2055–2066. doi: 10.1056/NEJMoa1108188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65**.Matsuoka K, Koreth J, Kim HT, et al. Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease. Sci Transl Med. 2013;5:179ra43. doi: 10.1126/scitranslmed.3005265. An important advance in the clinical use of IL-2 therapy to stimulate regulatory T cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Brown IE, Blank C, Kline J, et al. Homeostatic proliferation as an isolated variable reverses CD8+ T cell anergy and promotes tumor rejection. J Immunol. 2006;177:4521–4529. doi: 10.4049/jimmunol.177.7.4521. [DOI] [PubMed] [Google Scholar]
  • 67**.Kaiser AD, Gadiot J, Guislain A, Blank CU. Mimicking homeostatic proliferation in vitro generates T cells with high anti-tumor function in non-lymphopenic hosts. Cancer Immunol Immunother. 2013;62:503–515. doi: 10.1007/s00262-012-1350-1. Part of the growing literature on immunologic therapy to treat malignancies, this approach actually takes advantage of the effector memory skewing see in homeostatic proliferation to target tumor cells. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES