Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Nov 1.
Published in final edited form as: Immunol Invest. 2016 Sep 12;45(8):767–775. doi: 10.1080/08820139.2016.1213278

Lymphocyte-Mediated Immune Regulation in Health and Disease: The Treg and γδ T Cell Co-Conspiracy

Kyle K Payne 1
PMCID: PMC5167356  NIHMSID: NIHMS833859  PMID: 27617588

Abstract

The significance of lymphocytes functioning to mediate immunological tolerance has garnered increasing appreciation during the last several decades. CD4+ CD25+ α/β T cells have arguably been the most extensively studied regulatory lymphocyte to date, perhaps owing to the dramatic phenotype observed mice and humans with mutated Foxp3. However, emerging studies suggest that the lineage of regulatory lymphocytes is quite robust. Most notably, while γδ T cells are more traditionally regarded as mediators of cytotoxic function, they are beginning to be regarded as potential negative regulators of immunity. While regulatory γ/δ T cells may possess a degree of transcriptional overlap with ‘classical Tregs’, there remains less clarity in regards to the mechanisms driving the suppressive potential of these cells. In this review, I will discuss the role of Tregs in establishing tolerance in the steady state as well as disease, and how their accumulation and function may be modulated by myeloid cells in the local microenvironment. I will also discuss the necessity to extend our understanding of the regulatory nature of γδ T cells, which may lead to the unearthing of novel paradigms of immunity, perhaps most notably with respect to cancer.

Introduction

Studies of regulatory immune cells of lymphoid origin over the several decades have focused on regulatory T cells (Tregs), T cells which are classically defined by their expression of CD4, CD25, and the transcription factor Fork head box P3 (Foxp3) in concert with their ability to exert immunological suppressive function. These suppressive lymphocytes were initially discovered upon the observation that mutation of the Foxp3 gene leads to dysregulated lymphoproliferation, mulitorgan leukocyte infiltration, and autoimmunity in mice and humans (Brunkow et al. 2001, Godfrey et al. 1991, Godfrey, Wilkinson & Russell 1991). Over the years, the significance of these cells in modulating anti-viral immune responses as well as their ability to drive malignant progression through a variety of mechanisms has become apparent (Chevalier, Weiss 2013, Khaitan et al. 2016, Curiel et al. 2004). In addition to the direct cell-to-cell suppressive properties of Tregs, mediated by IL-10, TGF-β, and others (Vignali 2008), emerging evidence suggests these cells may function in complex with cells of myeloid lineage to function in a global immune suppressive network.

In addition to classical Tregs, the lymphoid lineage contains additional T lymphocytes which may function as regulatory cells in the appropriate stimulatory context. Developing studies in tumor immunology have identified gamma-delta (γδ) T cells as functioning under a suppressive program in order to dampen anti-tumor immunity and facilitate malignant progression. γδ T cells outnumber conventional αβ CD4+ Tregs in many solid tumors such as breast and ovarian cancers, yet they remain underinvestigated (Rabinovich, Conejo-Garcia 2016). Therefore, extending our understanding of the mechanistic properties driving the suppressive nature of such cells may lead to enhanced prognostic and therapeutic benefit.

‘Classical’ regulatory T cells

Tregs are a heterogeneous population, composed of CD4+ thymic-derived (nTreg) and peripherally induced Treg (iTreg). Differentiation of nTreg occurs in the thymus, while iTreg are generated from conventional naïve CD4+ T cells due to the composition of their local microenvironment in peripheral tissue. The Treg-dependent transcriptional program has yet to be fully elucidated; it is, however, known that the suppressive function of murine Tregs is dependent on the activity of Foxp3 (Fontenot, Gavin & Rudensky 2003, Hori, Nomura & Sakaguchi 2003)While human Tregs do express Foxp3, its expression is not necessarily indicative of suppressive activity of CD4+ T cells (Wang et al. 2007, Miyara et al. 2009, Hori, Nomura & Sakaguchi 2003). Foxp3 binds DNA through a winged helix-forkhead DNA binding domain and functions as a transcriptional repressor via a histone acetyltransferase-deacetylase complex (Li et al. 2007). Moreover, Foxp3 has been identified to interact with, and requires finely tuned expression of the NFATs, as well as NF-kB, and Runx1/AML1 for nTreg and iTreg development and function (Vaeth et al. 2012, Sumpter, Payne & Wilkes 2008, Ono et al. 2007, Bettelli, Dastrange & Oukka 2005).

In addition to their vital role in maintaining lymphocyte homeostasis, Tregs have been shown to contribute to the pathogenicity of chronic viral infections, such as hepatitis C virus (HCV). Ren et al. have recently demonstrated that NF-kB-mediated induction of miR146a, resulting in a Th2 response in the setting of chronic HCV infection, thus driving immune tolerance and viral persistence (Ren et al. 2016).

Interestingly, in the setting of human immunodeficiency virus (HIV) infection chronic immune activation is a key factor of disease progression (Hearps et al. 2014). It has therefore been hypothesized that increasing viral load may result in such comorbidity due to a deleterious effect on Tregs. However, clarifying the role of Tregs in HIV progression has been difficult due to conflicting studies of their suppressive function, and the unreliability of the phenotypic markers used to identify these cells in HIV infection (Chevalier, Weiss 2013). It has recently been demonstrated, however, that functionally suppressive Tregs, which coexpress the transcription factors Foxp3 and Helios, have been shown to selectively increase in HIV positive patients suppressed with antiretroviral therapy, while Foxp3+ Helios-Tregs remained similar to healthy adults (Mercer et al. 2014). In a corroborating study, it was shown that T cells coexpressing Foxp3 and Helios are bona fide memory Tregs in perinatally infected children with HIV, and are positively correlated with falling percentages of CD4+ T cells and increasing HIV plasma viremia and systemic immune activation (Khaitan et al. 2016). The identification of such functionally suppressive Tregs should now drive studies to identify the role of these cells in protecting or impairing the host’s clinical state during HIV infection.

The role of Tregs in Cancer

Growing interest in the immunobiology of cancer, coupled with the recent clinical advances in the treatment of several distinct cancers using immunotherapy has promoted an interest in elucidating the functional role of Tregs in cancer. The accumulation of Tregs has been correlated with a poor prognosis in patients with a variety of solid cancers, such as ovarian, breast, and hepatocellular carcinoma, among others (Curiel et al. 2004, Bates et al. 2006, Petersen et al. 2006, Gao et al. 2007, Griffiths et al. 2007, Hiraoka et al. 2006, Perrone et al. 2008), in which their cell intrinsic suppressive mechanisms generally utilize TGF-β and IL-10, as has been discussed elsewhere (Mougiakakos et al. 2010). Additionally, Foxp3+ CD4+ regulatory T cells have been identified within the microenvironment of cutaneous squamous cell carcinoma (cSSC) (Schipmann et al. 2014, Lai et al. 2016). cSSC is an interesting tumor from an immunological point-of-view, as these tumors are generally well infiltrated by lymphocytes, yet such lymphocytes appear incapable of rejecting this neoplasm. Indeed, it has recently been demonstrated that Foxp3 expressing T cells are responsible for mediating immune suppression within this microenvironment, and are associated with tumor cell metastatic potential (Lai et al. 2016).

Myeloid cells, in addition to Tregs, are also now well appreciated to polarize to a suppressive phenotype and to blunt immunity in cancer-bearing hosts. In fact, the cell intrinsic role of alternatively activated macrophages (M2), dendritic cells (DCs), and myeloid-derived suppressor cells (MDSCs) have been well characterized as suppressing type I immunity in cancer (Manjili 2012, Marvel, Gabrilovich 2015). However, the crosstalk between myeloid and lymphoid regulatory networks is less well characterized; the major contribution of Tregs to anti-tumor immune suppression and tumor escape may be the result of their function within a larger immunosuppressive network. Huang et al. initially described the role of immature immunosuppressive myeloid cells (now commonly regarded as MDSCs) to induce the development of Foxp3+ regulatory T cells in vitro, as well as in the MCA26 murine model of colon carcinoma (Huang et al. 2006). Such MDSCs mediated development of Tregs was shown to be dependent on IL-10 and IFN-γ, but not IL-13. Subsequent studies in cancer have also outlined the significance of CD40 expressing MDSCs functioning to activate Tregs (Pan et al. 2010), the ability of MDSCs to promote tolerance in B-cell lymphoma by expanding Tregs (Serafini et al. 2008), and the role of monocytic MDSCs (CD11b+, Ly6Glo, Ly6C+) in recruiting Tregs in a CCR5-dependent manner in the murine B16 melanoma model (Schlecker et al. 2012).

Furthermore, while the role of tumor-associated neutrophils (TANs) within the tumor microenvironment remains controversial and is likely context dependent (Deryugina et al. 2014, Eruslanov et al. 2014, Fridlender, Albelda 2012, Uribe-Querol, Rosales 2015), however, recent studies have outlined a role for TANs in the promotion of Treg infiltration into tumor beds. In a study by Mishalian et al. utilizing a murine model of malignant mesothelioma, it was demonstrated that ‘N2’ TANs were the major mediators in the production of CCL17 (Mishalian et al. 2014). The generation of this chemokine gradient was directly associated with an increase in the frequency of infiltrating Foxp3+ Tregs. This was recently corroborated in work by Zhou et al. in which TANs derived from human hepatocellular carcinoma patients were determined to produce CCL2, in addition to CCL17, to promote Treg infiltration into the tumor bed (Zhou et al. 2016). Such studies outline the role of myeloid cells, particularly MDSCs and neutrophils, in framing the immunosuppressive network in cancer.

The paradox of tumor-associated Tregs

While the accumulation of Foxp3 expressing CD4+ T cells is generally associated with poor prognosis in cancer patients due to their ability to suppress antitumor immunity, it has paradoxically been demonstrated that the presence of Tregs in some tumors may promote a beneficial response for the host in human colorectal cancer (Ladoire, Martin & Ghiringhelli 2011, Salama et al. 2009). As proposed by Ladoire et al. (Ladoire, Martin & Ghiringhelli 2011), the prognostic value of Foxp3+ tumor-infiltrating may be due to the unique microenvironment of colorectal cancer. Wherein inflammation driven by Th17 cells responding to the local gut flora may facilitate malignant progression, a higher frequency of Tregs may function to suppress Th17-specific inflammatory responses and thus delay malignant progression and result in a better prognosis. In recent years, the contribution of the gut microbiota to the modulation of anti-tumor responses has become increasingly clear (Rutkowski et al. 2015, Viaud et al. 2013, Iida et al. 2013). Thus, while a plethora of evidence correlates the infiltration of Tregs into the tumor bed with poor prognosis, the functional outcome of tumor-infiltrating Tregs appears to be highly dependent on the nature of the tumor microenvironment, and warrants intensive study. Therefore a degree of caution should be utilized in strategies invoking depletion of Tregs to improve patient responses.

γδ T cells: atypical regulatory T cells

In addition to classical regulatory T cells, i.e. CD4+ αβ T cells expressing Foxp3, the emergence of additional regulatory T cell subsets has deepened our understanding of the role of lymphocytes in the maintenance of tolerance. As early as 1989 it has been suggested that non-αβ T cells may function as immune suppressors (Patel et al. 1989), and the direct suppressive function of murine and human γδ T cells has subsequently been demonstrated (Kuhl et al. 2009, Rutkowski et al. 2015, Casetti et al. 2009, Hua et al. 2013). Such suppressive potential of Vδ1 and Vδ2 regulatory γδ T cells has been linked with the expression of Foxp3 (Hua et al. 2013, Casetti et al. 2009), which appears to occur under the influence of TGF-β-dependent signaling (Kang et al. 2009).

γδ T cells account for 5% of circulating T cells, and are mainly composed of the Vδ9Vγ2 subset. Phosphorylated metabolites have been recognized to potently activate γδ T cells in humans, and recently it was discovered that a member of the butyrophilin family, butyrophilin-3A1 (BTN3A1), effectively presents phosphorylated metabolites resulting in γδ T cell activation (Vavassori et al. 2013, Harly et al. 2012). Intriguingly γδ T cells have been shown to display both cytotoxic and regulatory functions, most notably in the setting of cancer.

More recently it has been demonstrated that γδ T cells may function in a tertiary manner to promote immune suppression. Rezende et al. have described the migration of thymic γδ T cells to the periphery, where they upregulate membrane-bound TGF-β1 and acquire a regulatory phenotype (Rezende et al. 2015). The regulatory function of such cells manifests via their ability to function as antigen presenting cells, which then induce classical CD4+ Foxp3+ regulatory T cells.

Furthermore, the expression of the transcription factor, Helios, which is widely expressed in conventional CD4+ Tregs, is also expressed in a significant percentage of freshly isolated circulating γδ T cells; Peters et al. have demonstrated a direct role of Vδ2 γδ T cells to inhibit the proliferation of CD4+ αβ T cells in a contact-dependent manner, wherein ~33% of peripheral γδ T cells constitutively expressed the transcription factor Helios (Peters et al. 2014). These studies suggest a degree of transcriptional overlap amongst conventional CD4+ Tregs, and suppressive γδ T cells, however the transcriptional program regulating the suppressive function regarding both of these cell types has yet to be fully defined.

The role of γδ T cells in cancer

It has been suggested that γδ T cells may have potential applications for cancer immunotherapy (Legut, Cole & Sewell 2015), owing to their ability to target tumor antigens irrespective of HLA haplotype and thus potentiating their application as ‘off-the-shelf’ cellular immunotherapy. For instance, γδ T cells have demonstrated cytotoxic function against colon cancer and myeloma cells, (Wu et al. 2015, von Lilienfeld-Toal et al. 2006). Additionally, recent studies have suggested a cytotoxic role for Vδ9Vγ2 T cells in patients with melanoma; wherein an elevated frequency of circulating as well as tumor infiltrating Vδ9Vγ2 T cells in early stage tumors were correlated with decreased mortality and disease relapse (Toia et al. 2016). Interestingly, however, it was observed that melanoma patients with a poor prognosis possessed a dramatically increased frequency of tumor-infiltrating Vδ9Vγ2 T cells in late stage disease. While no functional studies were performed, such observations may represent the acquisition of a suppressive phenotype due to modulation of the tumor microenvironment (Toia et al. 2016).

Indeed, there is mounting evidence suggesting that tumor-associated γδ T cells function as drivers of malignancy. In fact, it has become increasingly clear that γδ T cells possess the potential to potently suppress anti-tumor immunity (Rutkowski et al. 2015). In fact, tumor-derived factors, such as IP-10, have been implicated in driving the accumulation of γδ T cells into the tumor bed in human breast cancer; conversely, inhibition of γδ trafficking to tumor beds has demonstrable enhancement of anti-tumor immunity (Ye et al. 2013). In addition, evidence presented by Rutkowski et al. implicates a role for galectin-1 secreting γδ T cells in dampening antitumor immunity, both in transplanted and autochthonous tumor models (Rutkowski et al. 2015).

Intriguingly, the study by Rutkowski et al. unveiled an immunosuppressive network in which microbially driven TLR-5-dependent IL-6 functioned to mobilize MDSCs; these MDSCs were found to drive γδ T cell-derived galectin-1 in order to facilitate malignant progression (Rutkowski et al. 2015). Likewise, a recent study by Coffelt et al., implicates a role for IL-17 producing γδ T cells in polarizing and expanding TANs in tumor-bearing mice (Coffelt et al. 2015). Such TANs were demonstrated to specifically suppress CD8+ T cells, thus promoting metastasis in a murine model of breast cancer. These studies therefore suggest that not only do γδ T cells have the ability to function as direct suppressors of anti-tumor immunity, but that they may also function in a broader suppressive network to drive malignant progression.

Concluding Remarks

The immunoregulatory axis required for maintaining immune tolerance in a variety of biological scenarios has been the subject of intense study which now serves as the backbone of immunology. While the study of regulatory lymphocytes has, in general, predominately focused on the cellular-intrinsic suppressive activity of classical CD4+ α/β T cells, emerging data suggests that these cells operate within a broad immunosuppressive network involving regulatory myeloid cells as well a regulatory γ/δ T cells. At least in the setting of cancer, the accumulation and acquisition of suppressive function by Tregs is enhanced by the activity of both myeloid cells and γ/δ T cells in the local environment. It is also clear that future studies are needed to address the paradox of tumor-infiltrating γδ T cells in order to unearth the regulation of their cytotoxic as well as their suppressive function. While strides have been made with regards to the understanding of the mechanisms regulating the activation of these cells, it remains unclear if such stimuli confer a cytotoxic or suppressive phenotype within the context of the tumor microenvironment. Achieving clarity with regards to the phenotypic regulation of these cells, as well as the immunosuppressive mechanisms they employ, has the potential to facilitate improved immunotherapeutic applications for patients with cancer.

In closing, additional efforts to delineate the cellular intrinsic and extrinsic mechanisms driving the immunosuppressive network will further facilitate our understanding of this fundamental facet of immunity, and is likely to generate novel therapeutic approaches in settings of disease.

Acknowledgments

I thank the Wistar Institute and the University of Pennsylvania. This work was supported in part by 2T32CA009140.

Footnotes

Declaration of interest: The author reports no conflict of interest. The author alone is responsible for the content and writing of the paper.

References

  1. Bates GJ, Fox SB, Han C, Leek RD, Garcia JF, Harris AL, Banham AH. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006;24(34):5373–5380. doi: 10.1200/JCO.2006.05.9584. [DOI] [PubMed] [Google Scholar]
  2. Bettelli E, Dastrange M, Oukka M. Foxp3 interacts with nuclear factor of activated T cells and NF-kappa B to repress cytokine gene expression and effector functions of T helper cells. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(14):5138–5143. doi: 10.1073/pnas.0501675102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Brunkow ME, Jeffery EW, Hjerrild KA, Paeper B, Clark LB, Yasayko SA, Wilkinson JE, Galas D, Ziegler SF, Ramsdell F. Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nature genetics. 2001;27(1):68–73. doi: 10.1038/83784. [DOI] [PubMed] [Google Scholar]
  4. Casetti R, Agrati C, Wallace M, Sacchi A, Martini F, Martino A, Rinaldi A, Malkovsky M. Cutting edge: TGF-beta1 and IL-15 Induce FOXP3+ gammadelta regulatory T cells in the presence of antigen stimulation. Journal of immunology (Baltimore, Md: 1950) 2009;183(6):3574–3577. doi: 10.4049/jimmunol.0901334. [DOI] [PubMed] [Google Scholar]
  5. Chevalier MF, Weiss L. The split personality of regulatory T cells in HIV infection. Blood. 2013;121(1):29–37. doi: 10.1182/blood-2012-07-409755. [DOI] [PubMed] [Google Scholar]
  6. Coffelt SB, Kersten K, Doornebal CW, Weiden J, Vrijland K, Hau CS, Verstegen NJ, Ciampricotti M, Hawinkels LJ, Jonkers J, de Visser KE. IL-17-producing gammadelta T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522(7556):345–348. doi: 10.1038/nature14282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nature medicine. 2004;10(9):942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  8. Deryugina EI, Zajac E, Juncker-Jensen A, Kupriyanova TA, Welter L, Quigley JP. Tissue-infiltrating neutrophils constitute the major in vivo source of angiogenesis-inducing MMP-9 in the tumor microenvironment. Neoplasia (New York, NY) 2014;16(10):771–788. doi: 10.1016/j.neo.2014.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Eruslanov EB, Bhojnagarwala PS, Quatromoni JG, Stephen TL, Ranganathan A, Deshpande C, Akimova T, Vachani A, Litzky L, Hancock WW, Conejo-Garcia JR, Feldman M, Albelda SM, Singhal S. Tumor-associated neutrophils stimulate T cell responses in early-stage human lung cancer. The Journal of clinical investigation. 2014;124(12):5466–5480. doi: 10.1172/JCI77053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature immunology. 2003;4(4):330–336. doi: 10.1038/ni904. [DOI] [PubMed] [Google Scholar]
  11. Fridlender ZG, Albelda SM. Tumor-associated neutrophils: friend or foe? Carcinogenesis. 2012;33(5):949–955. doi: 10.1093/carcin/bgs123. [DOI] [PubMed] [Google Scholar]
  12. Gao Q, Qiu SJ, Fan J, Zhou J, Wang XY, Xiao YS, Xu Y, Li YW, Tang ZY. Intratumoral balance of regulatory and cytotoxic T cells is associated with prognosis of hepatocellular carcinoma after resection. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25(18):2586–2593. doi: 10.1200/JCO.2006.09.4565. [DOI] [PubMed] [Google Scholar]
  13. Godfrey VL, Wilkinson JE, Rinchik EM, Russell LB. Fatal lymphoreticular disease in the scurfy (sf) mouse requires T cells that mature in a sf thymic environment: potential model for thymic education. Proceedings of the National Academy of Sciences of the United States of America. 1991;88(13):5528–5532. doi: 10.1073/pnas.88.13.5528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Godfrey VL, Wilkinson JE, Russell LB. X-linked lymphoreticular disease in the scurfy (sf) mutant mouse. The American journal of pathology. 1991;138(6):1379–1387. [PMC free article] [PubMed] [Google Scholar]
  15. Griffiths RW, Elkord E, Gilham DE, Ramani V, Clarke N, Stern PL, Hawkins RE. Frequency of regulatory T cells in renal cell carcinoma patients and investigation of correlation with survival. Cancer immunology, immunotherapy : CII. 2007;56(11):1743–1753. doi: 10.1007/s00262-007-0318-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Harly C, Guillaume Y, Nedellec S, Peigne CM, Monkkonen H, Monkkonen J, Li J, Kuball J, Adams EJ, Netzer S, Dechanet-Merville J, Leger A, Herrmann T, Breathnach R, Olive D, Bonneville M, Scotet E. Key implication of CD277/butyrophilin-3 (BTN3A) in cellular stress sensing by a major human gammadelta T-cell subset. Blood. 2012;120(11):2269–2279. doi: 10.1182/blood-2012-05-430470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Hearps AC, Martin GE, Rajasuriar R, Crowe SM. Inflammatory co-morbidities in HIV+ individuals: learning lessons from healthy ageing. Current HIV/AIDS reports. 2014;11(1):20–34. doi: 10.1007/s11904-013-0190-8. [DOI] [PubMed] [Google Scholar]
  18. Hiraoka N, Onozato K, Kosuge T, Hirohashi S. Prevalence of FOXP3+ regulatory T cells increases during the progression of pancreatic ductal adenocarcinoma and its premalignant lesions. Clinical cancer research : an official journal of the American Association for Cancer Research. 2006;12(18):5423–5434. doi: 10.1158/1078-0432.CCR-06-0369. [DOI] [PubMed] [Google Scholar]
  19. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science (New York, NY) 2003;299(5609):1057–1061. doi: 10.1126/science.1079490. [DOI] [PubMed] [Google Scholar]
  20. Hua F, Kang N, Gao YA, Cui LX, Ba DN, He W. Potential regulatory role of in vitro-expanded Vdelta1 T cells from human peripheral blood. Immunologic research. 2013;56(1):172–180. doi: 10.1007/s12026-013-8390-2. [DOI] [PubMed] [Google Scholar]
  21. Huang B, Pan PY, Li Q, Sato AI, Levy DE, Bromberg J, Divino CM, Chen SH. Gr-1+CD115+ immature myeloid suppressor cells mediate the development of tumor-induced T regulatory cells and T-cell anergy in tumor-bearing host. Cancer research. 2006;66(2):1123–1131. doi: 10.1158/0008-5472.CAN-05-1299. [DOI] [PubMed] [Google Scholar]
  22. Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, Dai RM, Kiu H, Cardone M, Naik S, Patri AK, Wang E, Marincola FM, Frank KM, Belkaid Y, Trinchieri G, Goldszmid RS. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science (New York, NY) 2013;342(6161):967–970. doi: 10.1126/science.1240527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Kang N, Tang L, Li X, Wu D, Li W, Chen X, Cui L, Ba D, He W. Identification and characterization of Foxp3(+) gammadelta T cells in mouse and human. Immunology letters. 2009;125(2):105–113. doi: 10.1016/j.imlet.2009.06.005. [DOI] [PubMed] [Google Scholar]
  24. Khaitan A, Kravietz A, Mwamzuka M, Marshed F, Ilmet T, Said S, Ahmed A, Borkowsky W, Unutmaz D. FOXP3+Helios+ regulatory T cells, immune activation and advancing disease in HIV infected children. Journal of acquired immune deficiency syndromes (1999) 2016 doi: 10.1097/QAI.0000000000001000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Kuhl AA, Pawlowski NN, Grollich K, Blessenohl M, Westermann J, Zeitz M, Loddenkemper C, Hoffmann JC. Human peripheral gammadelta T cells possess regulatory potential. Immunology. 2009;128(4):580–588. doi: 10.1111/j.1365-2567.2009.03162.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Ladoire S, Martin F, Ghiringhelli F. Prognostic role of FOXP3+ regulatory T cells infiltrating human carcinomas: the paradox of colorectal cancer. Cancer immunology, immunotherapy : CII. 2011;60(7):909–918. doi: 10.1007/s00262-011-1046-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Lai C, August S, Albibas A, Behar R, Cho SY, Polak ME, Theaker J, MacLeod AS, French RR, Glennie MJ, Al-Shamkhani A, Healy E. OX40+ regulatory T cells in cutaneous squamous cell carcinoma suppress effector T cell responses and associate with metastatic potential. Clinical cancer research : an official journal of the American Association for Cancer Research. 2016 doi: 10.1158/1078-0432.CCR-15-2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Legut M, Cole DK, Sewell AK. The promise of gammadelta T cells and the gammadelta T cell receptor for cancer immunotherapy. Cellular & molecular immunology. 2015;12(6):656–668. doi: 10.1038/cmi.2015.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Li B, Samanta A, Song X, Iacono KT, Bembas K, Tao R, Basu S, Riley JL, Hancock WW, Shen Y, Saouaf SJ, Greene MI. FOXP3 interactions with histone acetyltransferase and class II histone deacetylases are required for repression. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(11):4571–4576. doi: 10.1073/pnas.0700298104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Manjili MH. Phenotypic plasticity of MDSC in cancers. Immunological investigations. 2012;41(6–7):711–721. doi: 10.3109/08820139.2012.673670. [DOI] [PubMed] [Google Scholar]
  31. Marvel D, Gabrilovich DI. Myeloid-derived suppressor cells in the tumor microenvironment: expect the unexpected. The Journal of clinical investigation. 2015;125(9):3356–3364. doi: 10.1172/JCI80005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Mercer F, Khaitan A, Kozhaya L, Aberg JA, Unutmaz D. Differentiation of IL-17-producing effector and regulatory human T cells from lineage-committed naive precursors. Journal of immunology (Baltimore, Md: 1950) 2014;193(3):1047–1054. doi: 10.4049/jimmunol.1302936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Mishalian I, Bayuh R, Eruslanov E, Michaeli J, Levy L, Zolotarov L, Singhal S, Albelda SM, Granot Z, Fridlender ZG. Neutrophils recruit regulatory T-cells into tumors via secretion of CCL17--a new mechanism of impaired antitumor immunity. International journal of cancer. 2014;135(5):1178–1186. doi: 10.1002/ijc.28770. [DOI] [PubMed] [Google Scholar]
  34. Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, Parizot C, Taflin C, Heike T, Valeyre D, Mathian A, Nakahata T, Yamaguchi T, Nomura T, Ono M, Amoura Z, Gorochov G, Sakaguchi S. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity. 2009;30(6):899–911. doi: 10.1016/j.immuni.2009.03.019. [DOI] [PubMed] [Google Scholar]
  35. Mougiakakos D, Choudhury A, Lladser A, Kiessling R, Johansson CC. Regulatory T cells in cancer. Advances in Cancer Research. 2010;107:57–117. doi: 10.1016/S0065-230X(10)07003-X. [DOI] [PubMed] [Google Scholar]
  36. Ono M, Yaguchi H, Ohkura N, Kitabayashi I, Nagamura Y, Nomura T, Miyachi Y, Tsukada T, Sakaguchi S. Foxp3 controls regulatory T-cell function by interacting with AML1/Runx1. Nature. 2007;446(7136):685–689. doi: 10.1038/nature05673. [DOI] [PubMed] [Google Scholar]
  37. Pan PY, Ma G, Weber KJ, Ozao-Choy J, Wang G, Yin B, Divino CM, Chen SH. Immune stimulatory receptor CD40 is required for T-cell suppression and T regulatory cell activation mediated by myeloid-derived suppressor cells in cancer. Cancer research. 2010;70(1):99–108. doi: 10.1158/0008-5472.CAN-09-1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Patel SS, Wacholtz MC, Duby AD, Thiele DL, Lipsky PE. Analysis of the functional capabilities of CD3+CD4-CD8− and CD3+CD4+CD8+ human T cell clones. Journal of immunology (Baltimore, Md: 1950) 1989;143(4):1108–1117. [PubMed] [Google Scholar]
  39. Perrone G, Ruffini PA, Catalano V, Spino C, Santini D, Muretto P, Spoto C, Zingaretti C, Sisti V, Alessandroni P, Giordani P, Cicetti A, D’Emidio S, Morini S, Ruzzo A, Magnani M, Tonini G, Rabitti C, Graziano F. Intratumoural FOXP3-positive regulatory T cells are associated with adverse prognosis in radically resected gastric cancer. European journal of cancer (Oxford, England : 1990) 2008;44(13):1875–1882. doi: 10.1016/j.ejca.2008.05.017. [DOI] [PubMed] [Google Scholar]
  40. Peters C, Oberg HH, Kabelitz D, Wesch D. Phenotype and regulation of immunosuppressive Vdelta2-expressing gammadelta T cells. Cellular and molecular life sciences : CMLS. 2014;71(10):1943–1960. doi: 10.1007/s00018-013-1467-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Petersen RP, Campa MJ, Sperlazza J, Conlon D, Joshi MB, Harpole DH, Jr, Patz EF., Jr Tumor infiltrating Foxp3+ regulatory T-cells are associated with recurrence in pathologic stage I NSCLC patients. Cancer. 2006;107(12):2866–2872. doi: 10.1002/cncr.22282. [DOI] [PubMed] [Google Scholar]
  42. Rabinovich GA, Conejo-Garcia JR. Shaping the immune landscape in cancer by galectin-driven regulatory pathways. Journal of Molecular Biology. 2016 doi: 10.1016/j.jmb.2016.03.021. [DOI] [PubMed] [Google Scholar]
  43. Ren JP, Ying RS, Cheng YQ, Wang L, Elgazzar MA, Li GY, Ning SB, Moorman JP, Yao ZQ. HCV-induced miR146a controls SOCS1/STAT3 and cytokine expression in monocytes to promote regulatory T-cell development. Journal of viral hepatitis. 2016 doi: 10.1111/jvh.12537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Rezende RM, da Cunha AP, Kuhn C, Rubino S, M’Hamdi H, Gabriely G, Vandeventer T, Liu S, Cialic R, Pinheiro-Rosa N, Oliveira RP, Gaublomme JT, Obholzer N, Kozubek J, Pochet N, Faria AM, Weiner HL. Identification and characterization of latency-associated peptide-expressing gammadelta T cells. Nature communications. 2015;6:8726. doi: 10.1038/ncomms9726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Rutkowski MR, Stephen TL, Svoronos N, Allegrezza MJ, Tesone AJ, Perales-Puchalt A, Brencicova E, Escovar-Fadul X, Nguyen JM, Cadungog MG, Zhang R, Salatino M, Tchou J, Rabinovich GA, Conejo-Garcia JR. Microbially driven TLR5-dependent signaling governs distal malignant progression through tumor-promoting inflammation. Cancer cell. 2015;27(1):27–40. doi: 10.1016/j.ccell.2014.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Salama P, Phillips M, Grieu F, Morris M, Zeps N, Joseph D, Platell C, Iacopetta B. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2009;27(2):186–192. doi: 10.1200/JCO.2008.18.7229. [DOI] [PubMed] [Google Scholar]
  47. Schipmann S, Wermker K, Schulze HJ, Kleinheinz J, Brunner G. Cutaneous and oral squamous cell carcinoma-dual immunosuppression via recruitment of FOXP3+ regulatory T cells and endogenous tumour FOXP3 expression? Journal of cranio-maxillo-facial surgery : official publication of the European Association for Cranio-Maxillo-Facial Surgery. 2014;42(8):1827–1833. doi: 10.1016/j.jcms.2014.06.022. [DOI] [PubMed] [Google Scholar]
  48. Schlecker E, Stojanovic A, Eisen C, Quack C, Falk CS, Umansky V, Cerwenka A. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. Journal of immunology (Baltimore, Md: 1950) 2012;189(12):5602–5611. doi: 10.4049/jimmunol.1201018. [DOI] [PubMed] [Google Scholar]
  49. Serafini P, Mgebroff S, Noonan K, Borrello I. Myeloid-derived suppressor cells promote cross-tolerance in B-cell lymphoma by expanding regulatory T cells. Cancer research. 2008;68(13):5439–5449. doi: 10.1158/0008-5472.CAN-07-6621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Sumpter TL, Payne KK, Wilkes DS. Regulation of the NFAT pathway discriminates CD4+CD25+ regulatory T cells from CD4+CD25− helper T cells. Journal of leukocyte biology. 2008;83(3):708–717. doi: 10.1189/jlb.0507321. [DOI] [PubMed] [Google Scholar]
  51. Toia F, Buccheri S, Anfosso A, Moschella F, Dieli F, Meraviglia S, Cordova A. Skewed Differentiation of Circulating Vgamma9Vdelta2 T Lymphocytes in Melanoma and Impact on Clinical Outcome. PloS one. 2016;11(2):e0149570. doi: 10.1371/journal.pone.0149570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Uribe-Querol E, Rosales C. Neutrophils in Cancer: Two Sides of the Same Coin. Journal of immunology research. 2015;2015:983698. doi: 10.1155/2015/983698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Vaeth M, Schliesser U, Muller G, Reissig S, Satoh K, Tuettenberg A, Jonuleit H, Waisman A, Muller MR, Serfling E, Sawitzki BS, Berberich-Siebelt F. Dependence on nuclear factor of activated T-cells (NFAT) levels discriminates conventional T cells from Foxp3+ regulatory T cells. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(40):16258–16263. doi: 10.1073/pnas.1203870109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Vavassori S, Kumar A, Wan GS, Ramanjaneyulu GS, Cavallari M, El Daker S, Beddoe T, Theodossis A, Williams NK, Gostick E, Price DA, Soudamini DU, Voon KK, Olivo M, Rossjohn J, Mori L, De Libero G. Butyrophilin 3A1 binds phosphorylated antigens and stimulates human gammadelta T cells. Nature immunology. 2013;14(9):908–916. doi: 10.1038/ni.2665. [DOI] [PubMed] [Google Scholar]
  55. Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillere R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, Schlitzer A, Ginhoux F, Apetoh L, Chachaty E, Woerther PL, Eberl G, Berard M, Ecobichon C, Clermont D, Bizet C, Gaboriau-Routhiau V, Cerf-Bensussan N, Opolon P, Yessaad N, Vivier E, Ryffel B, Elson CO, Dore J, Kroemer G, Lepage P, Boneca IG, Ghiringhelli F, Zitvogel L. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science (New York, NY) 2013;342(6161):971–976. doi: 10.1126/science.1240537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Vignali D. How many mechanisms do regulatory T cells need? European journal of immunology. 2008;38(4):908–911. doi: 10.1002/eji.200738114. [DOI] [PubMed] [Google Scholar]
  57. von Lilienfeld-Toal M, Nattermann J, Feldmann G, Sievers E, Frank S, Strehl J, Schmidt-Wolf IG. Activated gammadelta T cells express the natural cytotoxicity receptor natural killer p 44 and show cytotoxic activity against myeloma cells. Clinical and experimental immunology. 2006;144(3):528–533. doi: 10.1111/j.1365-2249.2006.03078.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Wang J, Ioan-Facsinay A, van der Voort EI, Huizinga TW, Toes RE. Transient expression of FOXP3 in human activated nonregulatory CD4+ T cells. European journal of immunology. 2007;37(1):129–138. doi: 10.1002/eji.200636435. [DOI] [PubMed] [Google Scholar]
  59. Wu D, Wu P, Wu X, Ye J, Wang Z, Zhao S, Ni C, Hu G, Xu J, Han Y, Zhang T, Qiu F, Yan J, Huang J. expanded human circulating Vdelta1 gammadeltaT cells exhibit favorable therapeutic potential for colon cancer. Oncoimmunology. 2015;4(3):e992749. doi: 10.4161/2162402X.2014.992749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Ye J, Ma C, Wang F, Hsueh EC, Toth K, Huang Y, Mo W, Liu S, Han B, Varvares MA, Hoft DF, Peng G. Specific recruitment of gammadelta regulatory T cells in human breast cancer. Cancer research. 2013;73(20):6137–6148. doi: 10.1158/0008-5472.CAN-13-0348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Zhou SL, Zhou ZJ, Hu ZQ, Huang XW, Wang Z, Chen EB, Fan J, Cao Y, Dai Z, Zhou J. Tumor-Associated Neutrophils Recruit Macrophages and T-regulatory Cells to Promote Progression of Hepatocellular Carcinoma and Resistance to Sorafenib. Gastroenterology. 2016 doi: 10.1053/j.gastro.2016.02.040. [DOI] [PubMed] [Google Scholar]

RESOURCES