Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Apr 9.
Published in final edited form as: Immunol Res. 2011 Apr;49(0):124–134. doi: 10.1007/s12026-010-8176-8

The Role of Natural Regulatory T cells in Infection

Ana M Sanchez a, Yiping Yang a,b
PMCID: PMC3621134  NIHMSID: NIHMS457881  PMID: 21116872

Abstract

Naturally occurring regulatory T cells (TReg) suppress multiple cell types of the immune system to maintain dominant tolerance to protect from autoimmunity, down-modulate anti-tumor immunity and restrain allergic diseases. In addition to these functions, TReg can alter effector responses to invading pathogens, leading to a variety of outcomes affecting both the host and infecting microorganisms. Here, we review how TReg can influence the immune responses to chronic infections where pathogen-specific TReg can contribute to pathogen persistence and, in some cases, concomitant immunity, as well as control immunopathology associated with robust immune responses. We also review the data on TReg during acute infection, focusing on the questions these studies raise regarding the most appropriate model(s) to examine TReg during infection. Finally, we discuss ways in which the TReg function can be altered by invading pathogens and how these can be exploited to develop methods therapeutically to influence disease and vaccine outcomes.

Keywords: Regulatory T cells, infection, immunopathology, Foxp3

Natural regulatory T cells

More that 100 years ago, Paul Ehrlich speculated that the immune system has the potential for “horror autotoxicus” and that mechanisms must exist to avoid this deleterious fate [1]. We now know that lymphocytes of the adaptive immune system have the unique ability to discriminate between foreign and self-antigens to successfully defend against pathogens while maintaining self-tolerance. The delicate balance between autoimmunity and pathogen defense is maintained by several processes that act in both cell-intrinsic and cell-extrinsic manners. Central tolerance ensures that self-reactive T cells are deleted in the thymus in response to a high-affinity major histocompatability complex (MHC)/self-peptide and T-cell receptor (TCR) interaction [2]. Despite this process, a small number of self-reactive T cells enter the periphery and are controlled by three main mechanisms: deletion, induction of anergy and suppression [3]. Regulatory T cells (TReg) are devoted to the active suppression of the immune system to maintain self-tolerance and immune homeostasis. For the purpose of this review, we will be focusing on naturally occurring CD4+CD25+ TReg that develop in the thymus as a unique lineage of CD4+ T cells (for a review of induced TReg see [4]).

TReg develop in the thymus as a subset of CD4+ T cells that acquire their suppressive ability as the result of a unique microenvironment. While all of the signals required for this microenvironment are unknown, studies using TCR transgenic mice that co-express their cognate antigen have suggested that TReg development in the thymus requires high affinity TCR stimulation [57] and is critically dependent on B7 costimulation [8, 9]. It is believed that these and potentially other signals lead to the expression of the transcription factor forkhead box p3 (Foxp3), which is required for and defines the TReg lineage [10, 11]. In addition to the expression of Foxp3, TReg are characterized by the high level expression of the IL-2 receptor α chain (CD25) [10, 12], cytotoxic T lymphocyte antigen-4 (CTLA-4) [13, 14] and glucocorticoid-induced TNFR (GITR) [15, 16]. Because these molecules can also be found on activated non-TReg, Foxp3 is considered the most specific marker for TReg in both mice [10, 11] and humans [17, 18]. Once TReg develop in the thymus, they exit into the periphery where they represent 5–10% and 2–4% of the CD4+ T cell population in healthy mice and humans, respectively [19].

Once TReg exit into the periphery, they perform a variety of functions. Early studies of TReg focused on their role in dominant tolerance—the active, trans-acting suppression of the immune system. These studies initiated when it was shown that thymectomy in neonatal mice three days after birth resulted in the development of organ-specific autoimmunity that could prevented by transferring syngeneic T cells from the spleen or thymus [20]. Further work by Sakaguchi et al. demonstrated that the cell type responsible for preventing autoimmune disease in this model was CD4+ CD25+ TReg [10, 12]. Later studies further classified these cells as Foxp3+ [10, 11, 21].

The importance of TReg for the maintenance of tolerance is exemplified in studies where genetic mutations or other manipulations that functionally delete TReg results in aggressive and fatal autoimmune disease. Mice with the spontaneous scurfy mutation (sf), which renders Foxp3 nonfunctional, develop fatal autoimmune disease characterized by severe lymphoproliferative and lymphocytic organ-infiltration ultimately leading to death three to four weeks after birth [22, 23]. In humans, an analogous mutation leads to a condition termed ‘immune dysregulation, polyendocrinopathy, enteropathy, X-linked’ (IPEX), where patients display symptoms similar to those seen in sf mice [2426]. Disregulation of TReg has also been associated with other autoimmune diseases including multiple sclerosis, type 1 diabetes and rheumatoid arthritis [2729]. In addition to autoimmune diseases, TReg have been implicated in the development of allergic diseases such as asthma [30], in the suppression of anti-tumor immunity [31] and during pathogen infection, which will be discussed at length here.

The diverse functions of TReg are a result of their ability to suppress multiple cells of the immune system, including non-TReg CD4+ T cells, CD8+ T cells, dendritic cells (DC), B cells, natural killer (NK) cells, NK T cells, macrophage and mast cells [8, 3238]. Currently, the exact mechanisms by which TReg suppress all these cell types are not fully elucidated, and many mechanisms remain controversial. Current evidence suggests that in vivo suppression can be mediated by anti-inflammatory cytokines, such as interleukin 10 (IL-10), IL-35 and transforming growth factor β (TGF-β) and via cell-cell contact potentially through the inhibitory receptor CTLA-4 [39, 40]. Additional studies have suggested a role for granzyme or perforin-dependent killing [4143] and adenosine and cyclic AMP in TReg suppression [44, 45]. Studies using intravital two-photon laser scanning microscopy (TPLSM) have attempted to further understand the dynamics of TReg suppression in vivo. These studies have demonstrated that TReg suppress the immune response by preventing stable contact between DCs and non-TReg [46, 47] suggesting that in vivo TReg may modulate antigen presenting cells (APC) function to suppress T cells. Further studies are required to more fully appreciate how suppression occurs in vivo and what mechanisms of suppression are utilized during pathogen invasion.

Regulatory T cells in chronic infection

While initial studies with TReg focused on their role in dominant tolerance, accumulating evidence indicates an important role for TReg in the control of immune responses to pathogens [48, 49]. One of the earliest implications for this role came from a model of chronic infection with Leishmania major when it was demonstrated that IL-10, which is known to be produced by TReg, contributes to pathogen persistence [50]. Subsequent studies demonstrated that Leishmania-specific TReg found at the sites of infection suppress T cells, by both IL-10-dependent and –independent mechanisms, to prevent complete pathogen clearance, leading to the development of concomitant immunity [51, 52]. Additional studies in both experimental animal models and humans have shown that TReg can accumulate following multiple types of infections and modulate the immune response in a variety of ways that can be beneficial for both the host and the pathogen.

Studies in humans have demonstrated that TReg can expand following infection and accumulate at the sites of infection. For example, TReg have been found in patients with parasitic infections (i.e. Leishmania braziliensis [53]), viral infections (i.e. hepatitis B [54]), fungal infection (i.e. Paracoccidioides brasiliensis [55]) and bacterial infection (i.e. Helicobacter pylori [56]). Studies to examine the effect of TReg during human infection are generally limited to an in vitro suppression assay whereby responder cells (non-TReg) are cultured with TReg in the presence of a stimulus for activation (i.e. APC + anti-CD3 antibody or agonist peptide). When these cells are cultured together, TReg will suppress the proliferation of non-TReg, which can be experimentally measured. Using this method, the suppression of pathogen-specific effector cells by human TReg has been demonstrated in a variety of disease models including Plasmodium falciparum where the removal of TReg enhances in vitro immune responses against the pathogen [57]. While in vivo evidence for TReg function during infection in humans is lacking, one study comparing an ethnic group with a naturally lower susceptibility to Plasmodium falciparum malaria than sympatric ethnic groups has shed some light. It was found that the group’s resistance was not associated with classic malaria-resistance genes, but rather may be related to the fact that members of this group have a propensity for functionally deficient TReg characterized by reduced RNA for genes associated with TReg function, as well as lower serum levels of TGF-β [57]. These data suggest a strong role for TReg in malaria resistance and provide a unique human system to further understand how TReg can affect the outcome of pathogen infection.

To more fully understand the role of TReg during infection than can be garnered with human studies, murine models of chronic parasitic, bacterial, viral and fungal infections have been utilized. These experiment have shown that the presence of TReg can suppress effector cells, preventing the immune system from mounting an effective immune response, which can lead to pathogen persistence. In some extreme cases, this can be detrimental to host survival, such as infection with a lethal strain of Plasmodium yoelii where TReg vigorously block an effective immune response, and depletion of TReg can actually improve host survival [58]. In what appears to be a similar mechanism, during infection with a hypervirulent, lethal strain of Mycobacterium tuberculosis as the infection proceeds, the number of effector T cells is reduced, and this is associated with an increase in the presence of TReg [59]. Further studies are required to confirm that the TReg are responsible for the decrease in effector cells and whether this is related to the lethality of this strain.

Multiple other studies of chronic infection have demonstrated that, while the presence of TReg does not contribute to the pathogen’s lethality, depletion of TReg leads to increased effector responses, supporting pathogen clearance. For example, in a murine model of ocular infection with herpes simplex virus 1 (HSV1), depletion of TReg leads to enhanced CD4 and CD8 expansion, effector function and infiltration into the cornea [60, 61]. Similar enhancement of effector function has been observed in multiple other models of parasitic, bacterial, fungal and viral infection (for review see [62]). However, not all studies have demonstrated that TReg function to block effector activation. With Mycobacterium tuberculosis (Mtb) infection, depletion of Foxp3+ TReg prior to infection results in significantly reduced bacterial burden in the lungs, but this reduction was not associated with an increase in pathogen-specific non-TReg effector function. This seemingly paradoxical effect may be related to the low levels of antigen available for T cell activation that occurs following TReg depletion in this model [63]. However, using the same model of infection, Quinn et al. demonstrated that depleting TReg with an anti-CD25 antibody increased effector IFN-γ, production but did not alter bacterial load [64] suggesting that the effects observed during TReg depletion may be related to the method of depletion utilized, which may be critical in this particular model because pathogen load is highly limiting to T cell expansion due to the inefficiency of antigen presentation.

While the presence of TReg may lead to pathogen persistence, this can actually be beneficial for the host as it is becoming increasingly clear that this can be critical to develop protective immunity. For example, with L. major infection the downregulation of the effector response by TReg leads to concomitant immunity, which is lost by removal of TReg [52]. This has also been demonstrated with a murine model of infection with the fungus Candida albicans where the presence of TReg was required for resistance to reinfection [65]. Additionally, similar results have been observed in infection with a lose-dose of HSV1 [60].

In addition to allowing for the development of protective immunity, TReg can benefit the host by reducing immune-mediated pathology that can occur as collateral damage associated with a strong anti-pathogen immune response. This role for TReg in protecting against immune-mediated damage has been demonstrated in models of pulmonary inflammation caused by Pneumocystis pneumonia, inflammatory eye lesions with HSV, liver pathology in Schistosoma mansoni infection and stomach pathology with Candida albicans where depletion of TReg leads to enhanced pathology [60, 6568]. However, not all chronic models of infection support a role for TReg in the prevention of immunopathology. Paradoxically, in a model of Plasmodium yoelii infection, removal of TReg actually resulted in reduced immunopathology in the brain [69]. This result was thought to occur because the depletion of TReg significantly reduced both parasite burden and the recruitment of CD8+ T cells in the brain [69]. In this model of experimental cerebral malaria (ECM) it appears as if the high parasite burden localized in the brain, as well as the potential exclusion of TReg from the brain during ECM [70], may make immunopathology an unavoidable consequence to combating P. yoelii infection.

Collectively, these results indicate there is a delicate balance between TReg and non-TReg, and any changes to this equilibrium can alter the outcome of a chronic infection. If TReg suppression is too vigorous, this can lead to poor pathogen clearance and possibly even host death. On the other end of the spectrum, without adequate suppression, non-TReg can cause collateral tissue damage due to an overzealous immune response. However, if non-TReg and TReg are balanced then this can lead to protective immunity without excessive immunopathology. Studies are underway to determine whether this balance can be exploited to modulate vaccine or disease outcomes.

Regulatory T cells in acute infection

Compared to studies with chronic infection, to date very few studies have been conducted to understand the function of TReg during acute infection. The results of these studies have painted contradictory roles for TReg during acute infection and highlight how different models of TReg depletion can lead to opposing results. Utilizing an anti-CD25 antibody to deplete TReg, it was shown that TReg do not affect the immune response or the disease outcome in Pseudomonas aeruginosa lung infection in mice [71]. Using a similar method of depletion, two independent studies examined TReg during acute Trypanosoma cruzi infection in mice and found no role in one of the studies and a limited role for TReg in the other study [72, 73]. The impact of TReg was largely dependent on the dose of T. cruzi where mice receiving a lower dose displayed reduced parasitemia and mortality following TReg depletion, but these effects were no longer observed when higher numbers of parasites were used for infection [73]. The discrepancies between these two studies and between the different parasite doses may be related to the relative inefficiency of TReg depletion by anti-CD25 antibodies observed during infection and/or the depletion of effector T cells, which upregulate CD25 following activation [74, 75]. These pitfalls of depletion may be more apparent in models of acute infection because effector cells can express higher levels of CD25 during acute compared to chronic infection [75]. Because of these caveats with CD25 antibody depletion, newer mouse models that selectively deplete Foxp3+ TReg may provide a better method to study the role of TReg during infection.

To overcome the inherent problems of using anti-CD25 antibodies to study TReg during infection, Foxp3DTR mice, which express the human diphtheria toxin receptor (DTR) under control of the Foxp3 promoter, have been used to study TReg during acute genital infection with HSV-2 [76]. In these mice, treatment with DT allows for the rapid and efficient depletion of Foxp3-expressing cells [77]. The results from these experiments were quite unexpected; while TReg-depleted mice displayed increased activation of effector cells in the draining lymph nodes, they were unable to control genital HSV-2 infection [76]. This led to severe lesions and faster viral dissemination into the spinal cord resulting in hindlimb paralysis, and as a result, DT-treated Foxp3DTR mice succumbed to disease much earlier than TReg-sufficient mice [76]. This apparently paradoxical effect was related to TReg modulating chemokine levels to ensure proper recruitment of immune cells to the site of infection [76]. In TReg-depleted mice the arrival of DCs, NK cells and T cells was delayed at the site of infection compared to TReg-sufficient mice [76]. This is not the first report that has implicated a role for TReg in cellular migration; in a model of respiratory syncytial virus infection, TReg were shown to alter CD8+ T cell trafficking to the lung following infection [78].

While using Foxp3-DTR mice is a much more specific method to study the role of TReg during infection, they must be carefully controlled to ensure that they are properly interpreted. Depletion of TReg using this method leads to increased activation of T cells, an increase in the number and activation of DC and the production of inflammatory cytokines and chemokines even in the absence of infection [76, 77]. These effects may be a direct result of increased activation of autoreactive T cells, which are normally controlled by TReg, but may also reflect direct or indirect effects of TReg on other cells of the immune system, such as NK cells or DC. These changes highlight the catastrophic changes that occur in the immune system when TReg are ablated and suggest that a more fine-tuned approach may be necessary to study TReg function during an immune response to ensure that the effects observed are not related to induced autoimmunity.

These studies also raise the question as to whether suppression during infection is antigen-specific or whether it occurs as the result of a bystander effect of TReg responding to self-antigens. This is critical to determine whether depletion of the polyclonal TReg population is the best method to study TReg during infection. Previous work to classify the TReg TCR repertoire has suggested some overlap between the TCRs of TReg and non-TReg suggesting that TReg could recognize more than just self-antigens [79, 80]. Indeed, work by Suffia et al. demonstrated that TReg found at the sites of L. major infection proliferate in a pathogen-specific manner and that suppression in vitro is antigen-specific [51]. Additional studies with both mice and humans in other models of infection support these observations [81, 82]. While the antigen-specificity of suppression in vivo has been demonstrated in our laboratory in an autoimmune setting [83], further studies are necessary to demonstrate that the in vivo suppression observed during infection is antigen-specific. Additionally, studies to examine whether non-TReg and TReg recognize the same the immunodominant epitopes as this may provide a mechanism to discriminate between the two cell types in vaccine development.

Taken together, it is clear that additional studies with acute infection are necessary as multiple additional questions remain. First, it is still not entirely clear whether TReg play a role during acute infection or perhaps in just select acute infection models. These questions can be addressed utilizing different models of acute infection coupled with different approaches to deplete TReg perhaps identifying systems that do not remove the polyclonal population but rather study pathogen-specific TReg. Additionally, work will need to be done to further clarify how TReg modulate the outcome acute infection, for example, whether they simply modulate migration as with genital HSV-2 infection or influence effector function and/or immunopathology as observed in chronic infection models. Finally, studies using acute infection models can be used to determine what happens to activated TReg following the resolution of an acute infection.

Manipulating regulatory T cells to regulate disease or vaccine outcome

Because of the important function of TReg during infection, researchers are studying how to manipulate them to adjust either disease or vaccine outcome. While therapeutic modulation of TReg is a viable research area, it appears as if microorganisms have already devised mechanisms to alter TReg function to promote their survival. One mechanism for modulation by pathogens may be through recognition of pathogens by innate pathogen pattern receptors such as Toll-like receptors (TLRs) (for review see [84]). While it has been demonstrated that TReg express various TLRs, how direct TLR signaling on TReg has not been fully elucidated and remains debated. Some studies have suggested that TLR2 or 8 stimulation abrogates the suppressive capacity of TReg, while others have shown that TLR4 and 5 can act to enhance suppression [84]. These studies are not without controversy, however, as it was recently suggested that TLR2 stimulation of TReg results in increased survival, but does not affect suppressive capacity [85]. Because many of these studies have been conducted in vitro, additional studies are necessary to fully tease apart the intricacies of TLR signaling on TReg function in vivo. In addition to direct TLR stimulation on TReg, TLRs can also affect TReg through an indirect mechanism via DCs to block their ability to suppress via an IL-6-dependent mechanism [86].

Another mechanism by which pathogens may alter TReg is though the creation of chemokine microenvironments that support TReg retention at the site of infection. There is evidence that chemokine receptor and integrin expression on TReg are critical for their function [8789]. Expression of the integrin αEβ7 (CD103), which primarily binds to E-cadherin, can be used to separate TReg with “effector/memory” phenotypes that have enhanced suppressive capabilities and distinct homing properties [90, 91]. Studies have demonstrated a role for CD103+ TReg during infection. Work with L. major has demonstrated that CD103 is required for retention of TReg at the sites of infection and that L. major-infected DC can stimulate CD103 expression in TReg [92] suggesting that the pathogen manipulates DCs to support TReg retention. Further work with L. major infection has shown that the chemokine receptor CCR5 is critical for TReg migration into the skin. [93]. Similar results with CCR5 have also been shown with the dimorphic fungi Paracoccidiodes brasiliensis and Histoplasma capsulatum [94, 95]. These data suggest unique chemokine and integrin signals are utilized by TReg to migrate to sites of infection and perhaps that the expression of these receptors can be modulated by pathogens. Additional work is necessary to explore other chemokines and/or integrins that affect TReg migration during infection and the ways in which pathogens can manipulate the expression of these molecules.

By understanding the ways in which microbes modulate TReg, as well as the ways in which TReg suppress, therapies can be developed to target TReg to manipulate disease or vaccine outcome to control microorganisms, block immunopathology or support the development of protective memory. With any of these potential therapies, care will need to be taken to maintain the balance between immunity to pathogens and autoimmunity as ablation of TReg can have deleterious effects [77]. As with studies in infection, perhaps therapies aimed a pathogen-specific TReg may be an effective and specific method that could avoid potentially damaging side effects.

Concluding remarks

Our knowledge about the functions of TReg in the immune system has grown tremendously since the days of the ill-fated “suppressor cell.” This expanding knowledge has revealed unique roles for TReg during pathogen invasion, especially in models of chronic infection. To further understand how TReg modulate infection outcomes, novel model systems of TReg depletion or addition will need to be carried out, particularly with acute infections. Elucidating these roles more fully, as well as the ways in which pathogens modulate TReg function is critical to harness their potential for vaccine development or therapeutics to treat infection. Overall, research examining TReg during infection has suggested that the relationship between TReg, invading microorganisms and the resulting immune response is one filled with various ententes, which will need to be considered in any therapeutic intervention.

Acknowledgements

This work was supported by the National Institutes of Health grants CA047741, CA111807, and CA136934 (to Y.Y.), an Alliance for Cancer Gene Therapy grant (to Y.Y.)

References

  • 1.Ehrlich P. Collected Studies on Immunity. New York: J. Wiley & Sons; 1910. [Google Scholar]
  • 2.Hogquist KA, Baldwin TA, Jameson SC. Central tolerance: learning self-control in the thymus. Nature reviews. 2005;5:772–782. doi: 10.1038/nri1707. [DOI] [PubMed] [Google Scholar]
  • 3.Abbas AK, Lohr J, Knoechel B, Nagabhushanam V. T cell tolerance and autoimmunity. Autoimmun Rev. 2004;3:471–475. doi: 10.1016/j.autrev.2004.07.004. [DOI] [PubMed] [Google Scholar]
  • 4.Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity. 2009;30:626–635. doi: 10.1016/j.immuni.2009.05.002. [DOI] [PubMed] [Google Scholar]
  • 5.Kawahata K, et al. Generation of CD4(+)CD25(+) regulatory T cells from autoreactive T cells simultaneously with their negative selection in the thymus and from nonautoreactive T cells by endogenous TCR expression. J Immunol. 2002;168:4399–4405. doi: 10.4049/jimmunol.168.9.4399. [DOI] [PubMed] [Google Scholar]
  • 6.Apostolou I, Sarukhan A, Klein L, von Boehmer H. Origin of regulatory T cells with known specificity for antigen. Nature immunology. 2002;3:756–763. doi: 10.1038/ni816. [DOI] [PubMed] [Google Scholar]
  • 7.Jordan MS, et al. Thymic selection of CD4+CD25+ regulatory T cells induced by an agonist self-peptide. Nature immunology. 2001;2:301–306. doi: 10.1038/86302. [DOI] [PubMed] [Google Scholar]
  • 8.Ghiringhelli F, et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. The Journal of experimental medicine. 2005;202:1075–1085. doi: 10.1084/jem.20051511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tang Q, et al. Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells. J Immunol. 2003;171:3348–3352. doi: 10.4049/jimmunol.171.7.3348. [DOI] [PubMed] [Google Scholar]
  • 10.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science (New York, NY. 2003;299:1057–1061. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  • 11.Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature immunology. 2003;4:330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  • 12.Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol. 1995;155:1151–1164. [PubMed] [Google Scholar]
  • 13.Read S, Malmstrom V, Powrie F. Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. The Journal of experimental medicine. 2000;192:295–302. doi: 10.1084/jem.192.2.295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Takahashi T, et al. Immunologic self-tolerance maintained by CD25(+)CD4(+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. The Journal of experimental medicine. 2000;192:303–310. doi: 10.1084/jem.192.2.303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.McHugh RS, et al. CD4(+)CD25(+) immunoregulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity. 2002;16:311–323. doi: 10.1016/s1074-7613(02)00280-7. [DOI] [PubMed] [Google Scholar]
  • 16.Shimizu J, Yamazaki S, Takahashi T, Ishida Y, Sakaguchi S. Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nature immunology. 2002;3:135–142. doi: 10.1038/ni759. [DOI] [PubMed] [Google Scholar]
  • 17.Walker MR, et al. Induction of FoxP3 and acquisition of T regulatory activity by stimulated human CD4+CD25− T cells. The Journal of clinical investigation. 2003;112:1437–1443. doi: 10.1172/JCI19441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wang HY, et al. Tumor-specific human CD4+ regulatory T cells and their ligands: implications for immunotherapy. Immunity. 2004;20:107–118. doi: 10.1016/s1074-7613(03)00359-5. [DOI] [PubMed] [Google Scholar]
  • 19.Baecher-Allan C, Viglietta V, Hafler DA. Human CD4+CD25+ regulatory T cells. Seminars in immunology. 2004;16:89–98. doi: 10.1016/j.smim.2003.12.005. [DOI] [PubMed] [Google Scholar]
  • 20.Nishizuka Y, Sakakura T. Thymus and reproduction: sex-linked dysgenesia of the gonad after neonatal thymectomy in mice. Science (New York, NY. 1969;166:753–755. doi: 10.1126/science.166.3906.753. [DOI] [PubMed] [Google Scholar]
  • 21.Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nature immunology. 2003;4:337–342. doi: 10.1038/ni909. [DOI] [PubMed] [Google Scholar]
  • 22.Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. The American journal of pathology. 1991;138:1379–1387. [PMC free article] [PubMed] [Google Scholar]
  • 23.Brunkow ME, et al. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nat Genet. 2001;27:68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  • 24.Bennett CL, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet. 2001;27:20–21. doi: 10.1038/83713. [DOI] [PubMed] [Google Scholar]
  • 25.Chatila TA, et al. JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. The Journal of clinical investigation. 2000;106:R75–R81. doi: 10.1172/JCI11679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wildin RS, et al. X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nat Genet. 2001;27:18–20. doi: 10.1038/83707. [DOI] [PubMed] [Google Scholar]
  • 27.Viglietta V, Baecher-Allan C, Weiner HL, Hafler DA. Loss of functional suppression by CD4+CD25+ regulatory T cells in patients with multiple sclerosis. The Journal of experimental medicine. 2004;199:971–979. doi: 10.1084/jem.20031579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lindley S, Dayan CM, Bishop A, Roep BO, Peakman M, Tree TI. Defective suppressor function in CD4(+)CD25(+) T-cells from patients with type 1 diabetes. Diabetes. 2005;54:92–99. doi: 10.2337/diabetes.54.1.92. [DOI] [PubMed] [Google Scholar]
  • 29.Ehrenstein MR, et al. Compromised function of regulatory T cells in rheumatoid arthritis and reversal by anti-TNFalpha therapy. The Journal of experimental medicine. 2004;200:277–285. doi: 10.1084/jem.20040165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Curotto de Lafaille MA, Lafaille JJ. CD4(+) regulatory T cells in autoimmunity and allergy. Current opinion in immunology. 2002;14:771–778. doi: 10.1016/s0952-7915(02)00408-9. [DOI] [PubMed] [Google Scholar]
  • 31.Wang HY, Wang RF. Regulatory T cells and cancer. Current opinion in immunology. 2007;19:217–223. doi: 10.1016/j.coi.2007.02.004. [DOI] [PubMed] [Google Scholar]
  • 32.von Boehmer H. Mechanisms of suppression by suppressor T cells. Nature immunology. 2005;6:338–344. doi: 10.1038/ni1180. [DOI] [PubMed] [Google Scholar]
  • 33.Piccirillo CA, Shevach EM. Cutting edge: control of CD8+ T cell activation by CD4+CD25+ immunoregulatory cells. J Immunol. 2001;167:1137–1140. doi: 10.4049/jimmunol.167.3.1137. [DOI] [PubMed] [Google Scholar]
  • 34.Misra N, Bayry J, Lacroix-Desmazes S, Kazatchkine MD, Kaveri SV. Cutting edge: human CD4+CD25+ T cells restrain the maturation and antigen-presenting function of dendritic cells. J Immunol. 2004;172:4676–4680. doi: 10.4049/jimmunol.172.8.4676. [DOI] [PubMed] [Google Scholar]
  • 35.Lim HW, Hillsamer P, Banham AH, Kim CH. Cutting edge: direct suppression of B cells by CD4+ CD25+ regulatory T cells. J Immunol. 2005;175:4180–4183. doi: 10.4049/jimmunol.175.7.4180. [DOI] [PubMed] [Google Scholar]
  • 36.La Cava A, Van Kaer L, Fu Dong S. CD4+CD25+ Tregs and NKT cells: regulators regulating regulators. Trends Immunol. 2006;27:322–327. doi: 10.1016/j.it.2006.05.003. [DOI] [PubMed] [Google Scholar]
  • 37.Taams LS, et al. Modulation of monocyte/macrophage function by human CD4+CD25+ regulatory T cells. Hum Immunol. 2005;66:222–230. doi: 10.1016/j.humimm.2004.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gri G, et al. CD4+CD25+ regulatory T cells suppress mast cell degranulation and allergic responses through OX40-OX40L interaction. Immunity. 2008;29:771–781. doi: 10.1016/j.immuni.2008.08.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Miyara M, Sakaguchi S. Natural regulatory T cells: mechanisms of suppression. Trends in molecular medicine. 2007;13:108–116. doi: 10.1016/j.molmed.2007.01.003. [DOI] [PubMed] [Google Scholar]
  • 40.Rudensky AY, Campbell DJ. In vivo sites and cellular mechanisms of T reg cell-mediated suppression. The Journal of experimental medicine. 2006;203:489–492. doi: 10.1084/jem.20060214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Grossman WJ, Verbsky JW, Barchet W, Colonna M, Atkinson JP, Ley TJ. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity. 2004;21:589–601. doi: 10.1016/j.immuni.2004.09.002. [DOI] [PubMed] [Google Scholar]
  • 42.Gondek DC, Lu LF, Quezada SA, Sakaguchi S, Noelle RJ. Cutting edge: contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J Immunol. 2005;174:1783–1786. doi: 10.4049/jimmunol.174.4.1783. [DOI] [PubMed] [Google Scholar]
  • 43.Cao X, et al. Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance. Immunity. 2007;27:635–646. doi: 10.1016/j.immuni.2007.08.014. [DOI] [PubMed] [Google Scholar]
  • 44.Bopp T, et al. Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. The Journal of experimental medicine. 2007;204:1303–1310. doi: 10.1084/jem.20062129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Deaglio S, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. The Journal of experimental medicine. 2007;204:1257–1265. doi: 10.1084/jem.20062512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Tadokoro CE, et al. Regulatory T cells inhibit stable contacts between CD4+ T cells and dendritic cells in vivo. The Journal of experimental medicine. 2006;203:505–511. doi: 10.1084/jem.20050783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tang Q, et al. Visualizing regulatory T cell control of autoimmune responses in nonobese diabetic mice. Nature immunology. 2006;7:83–92. doi: 10.1038/ni1289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mills KH. Regulatory T cells: friend or foe in immunity to infection? Nat Rev Immunol. 2004;4:841–855. doi: 10.1038/nri1485. [DOI] [PubMed] [Google Scholar]
  • 49.Belkaid Y, Rouse BT. Natural regulatory T cells in infectious disease. Nat Immunol. 2005;6:353–360. doi: 10.1038/ni1181. [DOI] [PubMed] [Google Scholar]
  • 50.Belkaid Y, et al. The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure. The Journal of experimental medicine. 2001;194:1497–506. doi: 10.1084/jem.194.10.1497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Suffia IJ, Reckling SK, Piccirillo CA, Goldszmid RS, Belkaid Y. Infected site-restricted Foxp3+ natural regulatory T cells are specific for microbial antigens. The Journal of experimental medicine. 2006;203:777–788. doi: 10.1084/jem.20052056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Belkaid Y, Piccirillo CA, Mendez S, Shevach EM, Sacks DL. CD4+CD25+ regulatory T cells control Leishmania major persistence and immunity. Nature. 2002;420:502–507. doi: 10.1038/nature01152. [DOI] [PubMed] [Google Scholar]
  • 53.Campanelli AP, et al. CD4+CD25+ T cells in skin lesions of patients with cutaneous leishmaniasis exhibit phenotypic and functional characteristics of natural regulatory T cells. J Infect Dis. 2006;193:1313–1322. doi: 10.1086/502980. [DOI] [PubMed] [Google Scholar]
  • 54.Xu D, et al. Circulating and liver resident CD4+CD25+ regulatory T cells actively influence the antiviral immune response and disease progression in patients with hepatitis B. J Immunol. 2006;177:739–747. doi: 10.4049/jimmunol.177.1.739. [DOI] [PubMed] [Google Scholar]
  • 55.Cavassani KA, et al. Systemic and local characterization of regulatory T cells in a chronic fungal infection in humans. J Immunol. 2006;177:5811–5818. doi: 10.4049/jimmunol.177.9.5811. [DOI] [PubMed] [Google Scholar]
  • 56.Kandulski A, et al. Naturally occurring regulatory T cells (CD4+, CD25high, FOXP3+) in the antrum and cardia are associated with higher H. pylori colonization and increased gene expression of TGF-beta1. Helicobacter. 2008;13:295–303. doi: 10.1111/j.1523-5378.2008.00612.x. [DOI] [PubMed] [Google Scholar]
  • 57.Walther M, et al. Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity. 2005;23:287–296. doi: 10.1016/j.immuni.2005.08.006. [DOI] [PubMed] [Google Scholar]
  • 58.Hisaeda H, et al. Escape of malaria parasites from host immunity requires CD4+ CD25+ regulatory T cells. Nat Med. 2004;10:29–30. doi: 10.1038/nm975. [DOI] [PubMed] [Google Scholar]
  • 59.Ordway D, et al. The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation. J Immunol. 2007;179:522–531. doi: 10.4049/jimmunol.179.1.522. [DOI] [PubMed] [Google Scholar]
  • 60.Suvas S, Azkur AK, Kim BS, Kumaraguru U, Rouse BT. CD4+CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J Immunol. 2004;172:4123–4132. doi: 10.4049/jimmunol.172.7.4123. [DOI] [PubMed] [Google Scholar]
  • 61.Suvas S, Kumaraguru U, Pack CD, Lee S, Rouse BT. CD4+CD25+ T cells regulate virus-specific primary and memory CD8+ T cell responses. The Journal of experimental medicine. 2003;198:889–901. doi: 10.1084/jem.20030171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Belkaid Y, Tarbell K. Regulatory T cells in the control of host-microorganism interactions (*) Annual review of immunology. 2009;27:551–589. doi: 10.1146/annurev.immunol.021908.132723. [DOI] [PubMed] [Google Scholar]
  • 63.Scott-Browne JP, et al. Expansion and function of Foxp3-expressing T regulatory cells during tuberculosis. The Journal of experimental medicine. 2007;204:2159–2169. doi: 10.1084/jem.20062105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Quinn KM, et al. Inactivation of CD4+ CD25+ regulatory T cells during early mycobacterial infection increases cytokine production but does not affect pathogen load. Immunol Cell Biol. 2006;84:467–474. doi: 10.1111/j.1440-1711.2006.01460.x. [DOI] [PubMed] [Google Scholar]
  • 65.Montagnoli C, et al. B7/CD28-dependent CD4+CD25+ regulatory T cells are essential components of the memory-protective immunity to Candida albicans. J Immunol. 2002;169:6298–6308. doi: 10.4049/jimmunol.169.11.6298. [DOI] [PubMed] [Google Scholar]
  • 66.McKinley L, Logar AJ, McAllister F, Zheng M, Steele C, Kolls JK. Regulatory T cells dampen pulmonary inflammation and lung injury in an animal model of pneumocystis pneumonia. J Immunol. 2006;177:6215–6226. doi: 10.4049/jimmunol.177.9.6215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hesse M, et al. The pathogenesis of schistosomiasis is controlled by cooperating IL-10-producing innate effector and regulatory T cells. J Immunol. 2004;172:3157–3166. doi: 10.4049/jimmunol.172.5.3157. [DOI] [PubMed] [Google Scholar]
  • 68.Guilliams M, et al. African trypanosomiasis: naturally occurring regulatory T cells favor trypanotolerance by limiting pathology associated with sustained type 1 inflammation. J Immunol. 2007;179:2748–2757. doi: 10.4049/jimmunol.179.5.2748. [DOI] [PubMed] [Google Scholar]
  • 69.Amante FH, et al. A role for natural regulatory T cells in the pathogenesis of experimental cerebral malaria. The American journal of pathology. 2007;171:548–559. doi: 10.2353/ajpath.2007.061033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Steeg C, Adler G, Sparwasser T, Fleischer B, Jacobs T. Limited role of CD4+Foxp3+ regulatory T cells in the control of experimental cerebral malaria. J Immunol. 2009;183:7014–7022. doi: 10.4049/jimmunol.0901422. [DOI] [PubMed] [Google Scholar]
  • 71.Carrigan SO, et al. Depletion of natural CD4+CD25+ T regulatory cells with anti-CD25 antibody does not change the course of Pseudomonas aeruginosa-induced acute lung infection in mice. Immunobiology. 2009;214:211–222. doi: 10.1016/j.imbio.2008.07.027. [DOI] [PubMed] [Google Scholar]
  • 72.Kotner J, Tarleton R. Endogenous CD4(+) CD25(+) regulatory T cells have a limited role in the control of Trypanosoma cruzi infection in mice. Infect Immun. 2007;75:861–869. doi: 10.1128/IAI.01500-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sales PA, Jr, et al. The regulatory CD4+CD25+ T cells have a limited role on pathogenesis of infection with Trypanosoma cruzi. Microbes Infect. 2008;10:680–688. doi: 10.1016/j.micinf.2008.03.008. [DOI] [PubMed] [Google Scholar]
  • 74.Couper KN, Blount DG, de Souza JB, Suffia I, Belkaid Y, Riley EM. Incomplete depletion and rapid regeneration of Foxp3+ regulatory T cells following anti-CD25 treatment in malaria-infected mice. J Immunol. 2007;178:4136–4146. doi: 10.4049/jimmunol.178.7.4136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Couper KN, et al. Anti-CD25 antibody-mediated depletion of effector T cell populations enhances susceptibility of mice to acute but not chronic Toxoplasma gondii infection. J Immunol. 2009;182:3985–3994. doi: 10.4049/jimmunol.0803053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lund JM, Hsing L, Pham TT, Rudensky AY. Coordination of early protective immunity to viral infection by regulatory T cells. Science (New York, NY. 2008;320:1220–1224. doi: 10.1126/science.1155209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kim JM, Rasmussen JP, Rudensky AY. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nature immunology. 2007;8:191–197. doi: 10.1038/ni1428. [DOI] [PubMed] [Google Scholar]
  • 78.Fulton RB, Meyerholz DK, Varga SM. Foxp3+ CD4 Regulatory T Cells Limit Pulmonary Immunopathology by Modulating the CD8 T Cell Response during Respiratory Syncytial Virus Infection. J Immunol. doi: 10.4049/jimmunol.1000423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Pacholczyk R, Kern J, Singh N, Iwashima M, Kraj P, Ignatowicz L. Nonself-antigens are the cognate specificities of Foxp3+ regulatory T cells. Immunity. 2007;27:493–504. doi: 10.1016/j.immuni.2007.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hsieh CS, Zheng Y, Liang Y, Fontenot JD, Rudensky AY. An intersection between the self-reactive regulatory and nonregulatory T cell receptor repertoires. Nature immunology. 2006;7:401–410. doi: 10.1038/ni1318. [DOI] [PubMed] [Google Scholar]
  • 81.McKee AS, Pearce EJ. CD25+CD4+ cells contribute to Th2 polarization during helminth infection by suppressing Th1 response development. J Immunol. 2004;173:1224–1231. doi: 10.4049/jimmunol.173.2.1224. [DOI] [PubMed] [Google Scholar]
  • 82.Kinter AL, et al. CD25(+)CD4(+) regulatory T cells from the peripheral blood of asymptomatic HIV-infected individuals regulate CD4(+) and CD8(+) HIV-specific T cell immune responses in vitro and are associated with favorable clinical markers of disease status. The Journal of experimental medicine. 2004;200:331–343. doi: 10.1084/jem.20032069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Huang X, Zhu J, Yang Y. Protection against autoimmunity in nonlymphopenic hosts by CD4+ CD25+ regulatory T cells is antigen-specific and requires IL-10 and TGF-beta. J Immunol. 2005;175:4283–4291. doi: 10.4049/jimmunol.175.7.4283. [DOI] [PubMed] [Google Scholar]
  • 84.van Maren WW, Jacobs JF, de Vries IJ, Nierkens S, Adema GJ. Toll-like receptor signalling on Tregs: to suppress or not to suppress? Immunology. 2008;124:445–452. doi: 10.1111/j.1365-2567.2008.02871.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Chen Q, Davidson TS, Huter EN, Shevach EM. Engagement of TLR2 does not reverse the suppressor function of mouse regulatory T cells, but promotes their survival. J Immunol. 2009;183:4458–4466. doi: 10.4049/jimmunol.0901465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pasare C, Medzhitov R. Toll pathway-dependent blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science (New York, NY. 2003;299:1033–1036. doi: 10.1126/science.1078231. [DOI] [PubMed] [Google Scholar]
  • 87.Sather BD, et al. Altering the distribution of Foxp3(+) regulatory T cells results in tissue-specific inflammatory disease. The Journal of experimental medicine. 2007;204:1335–1347. doi: 10.1084/jem.20070081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Schneider MA, Meingassner JG, Lipp M, Moore HD, Rot A. CCR7 is required for the in vivo function of CD4+ CD25+ regulatory T cells. The Journal of experimental medicine. 2007;204:735–745. doi: 10.1084/jem.20061405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wei S, Kryczek I, Zou W. Regulatory T-cell compartmentalization and trafficking. Blood. 2006;108:426–431. doi: 10.1182/blood-2006-01-0177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Lehmann J, et al. Expression of the integrin alpha Ebeta 7 identifies unique subsets of CD25+ as well as CD25− regulatory T cells. Proc Natl Acad Sci U S A. 2002;99:13031–13036. doi: 10.1073/pnas.192162899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Huehn J, et al. Developmental stage, phenotype, and migration distinguish naive- and effector/memory-like CD4+ regulatory T cells. The Journal of experimental medicine. 2004;199:303–313. doi: 10.1084/jem.20031562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Suffia I, Reckling SK, Salay G, Belkaid Y. A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J Immunol. 2005;174:5444–5455. doi: 10.4049/jimmunol.174.9.5444. [DOI] [PubMed] [Google Scholar]
  • 93.Yurchenko E, Tritt M, Hay V, Shevach EM, Belkaid Y, Piccirillo CA. CCR5-dependent homing of naturally occurring CD4+ regulatory T cells to sites of Leishmania major infection favors pathogen persistence. The Journal of experimental medicine. 2006;203:2451–2460. doi: 10.1084/jem.20060956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Moreira AP, et al. CCR5-dependent regulatory T cell migration mediates fungal survival and severe immunosuppression. J Immunol. 2008;180:3049–3056. doi: 10.4049/jimmunol.180.5.3049. [DOI] [PubMed] [Google Scholar]
  • 95.Kroetz DN, Deepe GS., Jr CCR5 dictates the equilibrium of proinflammatory IL-17+ and regulatory Foxp3+ T cells in fungal infection. J Immunol. 184:5224–5231. doi: 10.4049/jimmunol.1000032. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES