scholarly journals Regulatory T Cells: Molecular Actions on Effector Cells in Immune Regulation

2016 ◽  
Vol 2016 ◽  
pp. 1-12 ◽  
Author(s):  
Asiel Arce-Sillas ◽  
Diana Denisse Álvarez-Luquín ◽  
Beatriz Tamaya-Domínguez ◽  
Sandra Gomez-Fuentes ◽  
Abel Trejo-García ◽  
...  

T regulatory cells play a key role in the control of the immune response, both in health and during illness. While the mechanisms through which T regulatory cells exert their function have been extensively described, their molecular effects on effector cells have received little attention. Thus, this revision is aimed at summarizing our current knowledge on those regulation mechanisms on the target cells from a molecular perspective.

Author(s):  
Anne M. Pesenacker ◽  
Lucy R. Wedderburn

In recent years, there have been many new developments in the field of regulatory T cells (Treg), challenging the consensus on their behaviour, classification and role(s) in disease. The role Treg might play in autoimmune disease appears to be more complex than previously thought. Here, we discuss the current knowledge of regulatory T cells through animal and human research and illustrate the recent developments in childhood autoimmune arthritis (juvenile idiopathic arthritis (JIA)). Furthermore, this review summarises our understanding of the fields and assesses current and future implications for Treg in the treatment of JIA.


Circulation ◽  
2015 ◽  
Vol 132 (suppl_3) ◽  
Author(s):  
Vimal Ramjee ◽  
Manvendra K Singh ◽  
Feiyan Liu ◽  
Kurt A Engleka ◽  
Lauren J Manderfield ◽  
...  

Ischemic heart disease constitutes the most prevalent type of heart disease in the US. Remodeling post-myocardial infarction (MI) is a multifaceted process driven by a strong immune response. Regulatory T-cells, a subset of CD4+ T-cells, have been shown to suppress the innate and adaptive immune response following myocardial injury to allow for less deleterious remodeling. To date, the precise mechanism by which injured myocardium recruits these suppressive immune cells remains unknown. Here, we show a novel role for the epicardium in suppressing the post-infarct inflammation response through recruitment of T-regulatory cells. The Hippo pathway is a signal transduction pathway, which has gained importance in determining organ size and is implicated as a critical regulator of tissue regeneration. Mice deficient in epicardial YAP/TAZ, two core effectors of the Hippo signaling pathway, develop profound pericardial inflammation, thoracic adhesions and myocardial fibrosis post-MI, with resultant cardiomyopathy and death. These mice demonstrate fewer suppressive T-regulatory cells in the injured myocardium, due to a deficiency of interferon-gamma, a known inducer of these T-cells. Collectively, these results suggest a novel role for Hippo signaling in immune regulation. In addition, our data suggest that the epicardium plays an important role in homing suppressive T-regulatory cells to injured myocardium so that the inflammation response may be attenuated following MI.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A914-A914
Author(s):  
Andrew Rankin ◽  
Edwina Naik

BackgroundThe clinical success of PD-1- and CTLA-4- immune checkpoint inhibitors highlights the key contribution of immunosuppression to limiting effective anti-tumor responses. However, as many patients do not respond to anti-PD1 or CTLA4 therapy1-3 novel therapeutics that target additional immune-suppressive mechanisms are needed. Regulatory T cells (Tregs) inhibit immune responses in the tumor microenvironment via multiple suppressive mechanisms.4 5 Existing Treg-targeting agents lack specificity for intratumoral Tregs and can also deplete effector cells, a property that has likely contributed to the lack of clinical activity observed to date. CCR8 (C-C chemokine receptor 8) is selectively expressed on highly activated intratumoral Tregs, its high expression correlates with poor prognosis in multiple human tumor types6 7 and depletion of CCR8+ Tregs in preclinical models elicited potent anti-tumor activity. These observations provided rationale for the development of a CCR8-specific human depleting antibody.MethodsHuman FOXP3 and CCR8 expression was correlated across multiple tumor types using TCGA datasets and expression of CCR8 evaluated in primary tumor explants and PBMCs by flow cytometry. The efficacy of anti-CCR8 antibody treatment was evaluated in the MC38 and CT26 murine tumor models. The depletion of Tregs following anti-CCR8 treatment was assessed by flow cytometry. Flow cytometric-based binding assays were performed using cell lines expressing human or cynomolgus CCR8. Purified human NK cells were co-cultured with CCR8+ target cells and flow cytometry used to evaluate antibody-dependent killing activity.ResultsCCR8 expression was highly correlated with FoxP3 across multiple cancer subtypes and was low to absent on effector T cells. Importantly, CCR8 was not detected on any peripheral human leukocyte subset. In murine tumor models, anti-CCR8 antibody treatment reduced tumor growth in a dose- and Fc-gamma-receptor-dependent manner and resulted in complete regressions and the development of memory. Tumor shrinkage was associated with a reduction in intratumoral Tregs and increased representation of intratumoral CD8 T cells. FPA157 is a highly specific human and cynomolgus crossreactive CCR8 antibody that does not bind closely related chemokine receptors. FPA157 was engineered to enhance antibody-dependent cell-mediated cytotoxicity (eADCC) and elicited potent NK-mediated killing of target cells expressing CCR8 at levels observed on human intratumoralTregs.ConclusionsFPA157 is a CCR8-specific monoclonal antibody with eADCC activity that is being developed for the treatment of cancer. Depletion of CCR8+ Tregs induced substantial anti-tumor activity in pre-clinical models, thus supporting the clinical evaluation of FPA157 as a novel approach to alleviate immune suppression in the microenvironment of human solid tumors.ReferencesHellmann MD, Ciuleanu TE, Pluzanski A, Lee JS, Otterson GA, Audigier-Valette C, Minenza E, Linardou H, Burgers S, Salman P, Borghaei H, Ramalingam SS, Brahmer J, Reck M, O’Byrne KJ, Geese WJ, Green G, Chang H, Szustakowski J, Bhagavatheeswaran P, Healey D, Fu Y, Nathan F, Paz-Ares L. Nivolumab plus Ipilimumab in lung cancer with a high tumor mutational burden. N Engl J Med 2018;378(22):2093-2104.Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob JJ, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, Ferrucci PF, Smylie M, Dummer R, Hill A, Hogg D, Haanen J, Carlino MS, Bechter O, Maio M, Marquez-Rodas I, Guidoboni M, McArthur G, Lebbé C, Ascierto PA, Long GV, Cebon J, Sosman J, Postow MA, Callahan MK, Walker D, Rollin L, Bhore R, Hodi FS, Larkin J. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377(14):1345-1356.Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, Powles T, Donskov F, Neiman V, Kollmannsberger CK, Salman P, Gurney H, Hawkins R, Ravaud A, Grimm MO, Bracarda S, Barrios CH, Tomita Y, Castellano D, Rini BI, Chen AC, Mekan S, McHenry MB, Wind-Rotolo M, Doan J, Sharma P, Hammers HJ, Escudier B; CheckMate 214 Investigators. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med 2018 Apr 5;378(14):1277-1290.Teng MW, Ngiow SF, von Scheidt B, McLaughlin N, Sparwasser T, Smyth MJ. Conditional regulatory T-cell depletion releases adaptive immunity preventing carcinogenesis and suppressing established tumor growth [published correction appears in Cancer Res. 2010; 70(23):10014]. Cancer Res 2010;70(20):7800-7809.Simpson TR, Li F, Montalvo-Ortiz W, Sepulveda MA, Bergerhoff K, Arce F, Roddie C, Henry JY, Yagita H, Wolchok JD, Peggs KS, Ravetch JV, Allison JP, Quezada SA. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J Exp Med 2013;210(9):1695-710.Plitas G, Konopacki C, Wu K, Bos PD, Morrow M, Putintseva EV, Chudakov DM, Rudensky AY. Regulatory T cells exhibit distinct features in human breast cancer. Immunity 2016;45(5):1122-1134.De Simone M, Arrigoni A, Rossetti G, Gruarin P, Ranzani V, Politano C, Bonnal RJP, Provasi E, Sarnicola ML, Panzeri I, Moro M, Crosti M, Mazzara S, Vaira V, Bosari S, Palleschi A, Santambrogio L, Bovo G, Zucchini N, Totis M, Gianotti L, Cesana G, Perego RA, Maroni N, Pisani Ceretti A, Opocher E, De Francesco R, Geginat J, Stunnenberg HG, Abrignani S, Pagani M. Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity 2016;45(5):1135-1147.


Cells ◽  
2021 ◽  
Vol 10 (10) ◽  
pp. 2655
Author(s):  
Inesa Navasardyan ◽  
Benjamin Bonavida

The T cell-mediated immune response is primarily involved in the fight against infectious diseases and cancer and its underlying mechanisms are complex. The anti-tumor T cell response is regulated by various T cell subsets and other cells and tissues in the tumor microenvironment (TME). Various mechanisms are involved in the regulation of these various effector cells. One mechanism is the iNOS/.NO that has been reported to be intimately involved in the regulation and differentiation of the various cells that regulate the anti-tumor CD8 T cells. Both endogenous and exogenous .NO are implicated in this regulation. Importantly, the exposure of T cells to .NO had different effects on the immune response, depending on the .NO concentration and time of exposure. For instance, iNOS in T cells regulates activation-induced cell death and inhibits Treg induction. Effector CD8 T cells exposed to .NO result in the upregulation of death receptors and enhance their anti-tumor cytotoxic activity. .NO-Tregs suppress CD4 Th17 cells and their differentiation. Myeloid-derived suppressor cells (MDSCs) expressing iNOS inhibit T cell functions via .NO and inhibit anti-tumor CD8 T cells. Therefore, both .NO donors and .NO inhibitors are potential therapeutics tailored to specific target cells that regulate the T cell effector anti-tumor response.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 4092-4092 ◽  
Author(s):  
Clodagh Keohane ◽  
Shahram Y Kordasti ◽  
Thomas Seidl ◽  
Pilar Perez Abellan ◽  
Nicholas Shaun B. Thomas ◽  
...  

Abstract Background The myeloproliferative neoplasms (MPN), in particular myelofibrosis, are associated with elevated levels of inflammatory cytokines and constitutional symptoms. Treatment with JAK inhibitors (JAKi) have lead to marked improvement in symptoms and splenomegaly. Signaling through the JAK pathway is critical for T cell development and differentiation. However the baseline immune signature remains largely undescribed in MPN as does the effect of JAK inhibition on the immune subsets in this disease. Materials and Methods The % and absolute number of CD4+ T cell subsets (TH1, TH2 and TH17 and Foxp3+ T regulatory cells) in peripheral blood (PB) were investigated by flow cytometry. T cells were stimulated and stained intracellularly for IFNg, IL-4, IL-17 & TNFα. Tregs were defined as CD4+ CD25highCD27+FOXp3+. The serum level of 30 cytokines was also measured by Luminex. Patients received either ruxolitinib (n=21) or SAR302503 (n=13) as JAKi. Results We analysed 50 MPN patients (30 Myelofibrosis, 15 Polycythemia Vera, 5 Essential Thrombocythemia) and 14 healthy donors (HD). 34 patients were treated with JAKi and sequential PB samples were obtained at 1, 3, 6 and 12 month intervals (median follow up 6 months). Tregs are significantly lower in MPN patients compared to HD and drop further following treatment (p<0.0001 and p=0.0049 respectively). There was no difference at baseline in the T effector subsets between the groups including TH1, TH2 and TH17 secreting cells but there was a significant increase in TH17 following JAKi therapy (fig 1a). JAKi resulted in a significant decrease (p=0.03) in CD4 T cells secreting pro-inflammatory cytokines at 3 months follow up although this was less evident at 6 months follow up and occurred irrespective of disease response to treatment. This silencing was confirmed by both intracellular staining and luminex assay of supernatants including a significant decrease in Interleukin-2 receptor (IL-2r) p=0.0007, Interferon gamma induced protein (IP-10) p=0.0006, monokine induced by gamma interferon (MIG) p=0.0008 and hepatocyte growth factor (HGF) p=0.0009. This finding was reproduced in-vitro in healthy peripheral blood mononuclear cells (PBMCs). PBMCs were treated with the JAKi ruxolitinib (100-300NM) in the presence or absence of plate bound anti-CD3/28 stimulation and cultured for 5 days. Tregs were reduced in number and there was a considerable increase in the percentage of “cytokine negative” or “silent” T effector cells by FACS analysis compared to untreated or vehicle treated cells (median of 42 % of CD4 to 91% of CD4) (fig 1b). This finding was reproduced by Luminex cytokine assay of supernatants. Western blot demonstrated a reduction in pSTAT3 in ruxolitinib treated cells. To assess the effect of JAKi on Treg function, healthy isolated Tregs were treated with ruxolitinib and co cultured with CFSE labeled autologous T effector cells. Short term JAKi treated Tregs were unable to suppress the proliferation of T effector cells compared to Tregs treated with vehicle alone. Similarly, proliferation rate and function of Tregs was reduced following 4 weeks expansion in the presence of ruxolitinib compared to expanded Tregs in the presence of ATRA and rapamycin as a control. Conclusions Tregs are significantly lower in MPN patients compared to healthy controls in keeping with the inflammatory environment of MPN and decrease further with JAKi. Surprisingly, T effector numbers were not significantly different to healthy controls at baseline and TH1 and TH2 subsets did not change with therapy. However, secretion of proinflammatory cytokines from these cells was blocked with JAKi both invivo and invitro resulting in a functional silencing of T effectors. Interestingly, TH17 subsets increase with treatment possibly representing a polarization from a Treg phenotype to a TH17 phenotype, suggesting the re-establishment of immune-surveillance against the malignant clone. Further investigation is required to confirm the hypothesis that these expanded TH17 cells originate from Tregs or previously “silenced” CD4 T cells. Disclosures: Harrison: Novartis: Honoraria; Sanofi : Honoraria.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1513-1513
Author(s):  
Sary El Daker ◽  
Qi Gao ◽  
Mikhail Roshal ◽  
Ahmet Dogan

Physiologically T-regulatory cells (Treg) suppress immune responses against self-antigens preventing autoimmunity. However, in cancer they are believed to suppress anti-tumor immune response, and the presence of Treg in the tumor microenvironment has been associated with adverse outcome in most cancers. Treg are integral part of tumor microenvironment in lymphoma, and can use different mechanisms to inhibit both adaptive and immune response (including CD4, CD8, DC, macrophages, B-cells and NK cells) modulating therefore the interaction between lymphoma and the host microenvironment. Increased numbers of Treg have been associated with adverse clinical outcome in follicular lymphoma (FL) but a more favorable outcome in classical Hodgkin lymphoma (CHL). In this study we examined Treg phenotype in detail using multiparameter flow cytometry in lymph node specimens with normal histology (NLN) and involved by FL and CHL. We report that both CD4 positive T-cells and Treg subset show distinct phenotypes in different disease entities suggesting different biological functions. Single cell suspensions were prepared from NLN (n=10) and lymph node biopsies involved by FL (n=10) and CHL (n=10), and high dimensional multiparameter flow cytometric immunophenotyping was performed using antibodies against immune checkpoints (TIGIT, TIM3, PD-1, CD96, LAG3, CTLA4, CD73). To characterize the CD4 T-cell compartments we used a dimensionality reduction algorithm for non-linear data representations (tSNE). We studied the CD4+FoxP3-CD25- and Treg characterized by CD4+FoxP3+CD25+ phenotype separately and in both the compartments we identified and characterized subpopulations specific for each of the disease cohorts. tSNE representations CD4 positive T-cells and Treg showed different distributions in NLN, FL and HL. In NLN, CD4 cells broader heterogeneity without distinct clusters whereas in FL and CHL CD4 positive T-cells and Treg (Figure 2 A-B) showed highly polarized phenotypes which were distinct from each other and nearly absent within normal Treg compartment. We observed a strong expression of the immune checkpoint regulators TIM3, LAG3, CTL4, Tbet and PD1 in CD4 T-cells derived from HL tissues. In contrast FL CD4 T-cells were mainly characterized by an up regulation of PD1, CTLA4 and TIGIT. The TIGIT+ cells in FL samples are mainly CXCR5+PD1bright and they up regulate CTLA-4 in the FoxP3+ compartment. In all the neoplastic tissues the FoxP3+CD25+ T-Reg express mainly an activated/memory phenotype (CD45RO+CTLA4+), but while in HL microenvironment they show a TH1 phenotype (CXCR3+Tbet+), in FL they mainly express PD1+CXCR5+ (Figure 1B). Additionally, the dominant population of regulatory T-cells derived from HL samples down modulate PD1 expression, show high level of expression of TH1-associated transcription factor Tbet and have high proliferation index, while in FL, PD1 is brightly expressed, Tbet is not expressed and proliferation index is low in the dominant population (Figure 1A). The distinct phenotypic differences of Treg in FL lymphoma and CHL may account for the better prognosis seen in CHL with increased Treg which has TH1 like phenotype, therefore predicted to have anti-tumor activity. The differences seen in the expression immune checkpoint regulators both on CD4 positive T-cells and Treg subset may explain different rate responses seen in FL compared to CHL with checkpoint therapy. Disclosures Roshal: Physicians' Education Resource: Other: Provision of services; Celgene: Other: Provision of Services; Auron Therapeutics: Equity Ownership, Other: Provision of services. Dogan:Corvus Pharmaceuticals: Consultancy; Novartis: Consultancy; Takeda: Consultancy; Roche: Consultancy, Research Funding; Celgene: Consultancy; Seattle Genetics: Consultancy.


2020 ◽  
Author(s):  
Lital Remez ◽  
Esther Ganelin Cohen ◽  
Dina Safina ◽  
Mark Hellman ◽  
Itay Lotan ◽  
...  

Abstract Background: Alemtuzumab (ALM) effectively prevents multiple sclerosis (MS)​ relapses. It causes lymphocytic depletion with subsequent enhancement of T-regulatory cell population. Direct administration of ALM to T-cells causes cytolysis. However, the T-cells may be indirectly affected by myeloid cells, which are resistant to ALM cytotoxicity. Does ALM modulate monocytes? Does the cross-talk between exposed monocytes and lymphocytes result in anti-inflammatory effects?Methods: CD14​+​ monocytes of 10 healthy controls and 10 MS (treatment naïve) patients were isolated from peripheral blood mononuclear cells (PBMCs), exposed to ALM and reintroduced to PBMCs depleted from CD14​+​ cells. After treatment, macrophage profile and T-cells markers were measured. Results: ALM promoted M2 anti-inflammatory phenotype, noted by increased​ percentage of CD23​+​, CD83​+ ​and CD163+​ cells. CD23​+​ cells were the most prominently upregulated (7-fold, p=0.0002). Observed effect was larger in MS patients compared to healthy subjects. The exposed macrophages increased the proportion of T-regulatory cells, without affecting T-effector cells. ​ Neutralization of​ monocytic CD23 reversed the effect on T-regulatory cells. Conclusions: ALM enabled monocytes’ conversion towards anti-inflammatory​ macrophages, which in turn promoted T-regulatory enhancement, in CD23 dependent manner. These findings suggest an ALM mechanism of action, which may explain some aspects of the MS pathogenesis.


2020 ◽  
Vol 8 (3) ◽  
pp. 144-156
Author(s):  
Şule KARATAŞ ◽  
Fatma SAVRAN OĞUZ

Introduction: Peptides obtained by processing intracellular and extracellular antigens are presented to T cells to stimulate the immune response. This presentation is made by peptide receptors called major histocompatibility complex (MHC) molecules. The regulation mechanisms of MHC molecules, which have similar roles in the immune response, especially at the gene level, have significant differences according to their class. Objective: Class I and class II MHC molecules encoded by MHC genes on the short arm of the sixth chromosome are peptide receptors that stimulate T cell response. These peptides, which will enable the recognition of the antigen from which they originate, are loaded into MHC molecules and presented to T cells. Although the principles of loading and delivering peptides are similar for both molecules, the peptide sources and peptide loading mechanisms are different. In addition, class I molecules are expressed in all nucleated cells while class II molecules are expressed only in Antigen Presentation Cells (APC). These differences; It shows that MHC class I is not expressed by exactly the same transcriptional mechanisms as MHC class II. In our article, we aimed to compare the gene expressions of both classes and reveal their similarities and differences. Discussion and Conclusion: A better understanding of the transcriptional mechanisms of MHC molecules will reveal the role of these molecules in diseases more clearly. In our review, we discussed MHC gene regulation mechanisms with presence of existing informations, which is specific to the MHC class, for contribute to future research. Keywords: MHC class I, MHC class II, MHC gene regulation, promoter, SXY module, transcription


Sign in / Sign up

Export Citation Format

Share Document