scholarly journals In vivo equilibrium of proinflammatory IL-17+ and regulatory IL-10+ Foxp3+ RORγt+ T cells

2008 ◽  
Vol 205 (6) ◽  
pp. 1381-1393 ◽  
Author(s):  
Matthias Lochner ◽  
Lucie Peduto ◽  
Marie Cherrier ◽  
Shinichiro Sawa ◽  
Francina Langa ◽  
...  

The nuclear hormone receptor retinoic acid receptor–related orphan receptor γt (RORγt) is required for the generation of T helper 17 cells expressing the proinflammatory cytokine interleukin (IL)-17. In vivo, however, less than half of RORγt+ T cells express IL-17. We report here that RORγt+ Tαβ cells include Foxp3+ cells that coexist with IL-17–producing RORγt+ Tαβ cells in all tissues examined. The Foxp3+ RORγt+ Tαβ express IL-10 and CCL20, and function as regulatory T cells. Furthermore, the ratio of Foxp3+ to IL-17–producing RORγt+ Tαβ cells remains remarkably constant in mice enduring infection and inflammation. This equilibrium is tuned in favor of IL-10 production by Foxp3 and CCL20, and in favor of IL-17 production by IL-6 and IL-23. In the lung and skin, the largest population of RORγt+ T cells express the γδ T cell receptor and produce the highest levels of IL-17 independently of IL-6. Thus, potentially antagonistic proinflammatory IL-17–producing and regulatory Foxp3+ RORγt+ T cells coexist and are tightly controlled, suggesting that a perturbed equilibrium in RORγt+ T cells might lead to decreased immunoreactivity or, in contrast, to pathological inflammation.

2019 ◽  
Vol 10 (1) ◽  
Author(s):  
Hanane Yahia-Cherbal ◽  
Magda Rybczynska ◽  
Domenica Lovecchio ◽  
Tharshana Stephen ◽  
Chloé Lescale ◽  
...  

Abstract T helper 17 (Th17) cells have crucial functions in mucosal immunity and the pathogenesis of several chronic inflammatory diseases. The lineage-specific transcription factor, RORγt, encoded by the RORC gene modulates Th17 polarization and function, as well as thymocyte development. Here we define several regulatory elements at the human RORC locus in thymocytes and peripheral CD4+ T lymphocytes, with CRISPR/Cas9-guided deletion of these genomic segments supporting their role in RORγt expression. Mechanistically, T cell receptor stimulation induces cyclosporine A-sensitive histone modifications and P300/CBP acetylase recruitment at these elements in activated CD4+ T cells. Meanwhile, NFAT proteins bind to these regulatory elements and activate RORγt transcription in cooperation with NF-kB. Our data thus demonstrate that NFAT specifically regulate RORγt expression by binding to the RORC locus and promoting its permissive conformation.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A185-A185
Author(s):  
Michelle Fleury ◽  
Derrick McCarthy ◽  
Holly Horton ◽  
Courtney Anderson ◽  
Amy Watt ◽  
...  

BackgroundAdoptive cell therapies have shown great promise in hematological malignancies but have yielded little progress in the context of solid tumors. We have developed T cell receptor fusion construct (TRuC®) T cells, which are equipped with an engineered T cell receptor that utilizes the full complement of TCR signaling subunits and recognizes tumor-associated antigens independent of HLA. In clinical trials, mesothelin (MSLN)-targeting TRuC-T cells (TC-210 or gavo-cel) have shown unprecedented results in patients suffering from advanced mesothelioma and ovarian cancer. To potentially increase the depth of response, we evaluated strategies that can promote intra-tumoral T cell persistence and function. Among the common ??-chain cytokines, IL-15 uniquely supports the differentiation and maintenance of memory T cell subsets by limiting terminal differentiation and conferring resistance to IL-2 mediated activation-induced cell death (AICD). In the studies described here, we evaluated the potential of IL-15 as an enhancement to TRuC-T cell phenotype, persistence and function against MSLN+ targets.MethodsPrimary human T cells were activated and transduced with a lentiviral vector encoding an anti-MSLN binder fused to CD3ε alone or co-expressed with a membrane-tethered IL-15rα/IL-15 fusion protein (IL-15fu). Transduced T cells were expanded for 9 days and characterized for expression of the TRuC, IL-15rα and memory phenotype before subjecting them to in vitro functional assays to evaluate cytotoxicity, cytokine production, and persistence. In vivo efficacy was evaluated in MHC class I/II deficient NSG mice bearing human mesothelioma xenografts.ResultsIn vitro, co-expression of the IL-15fu led to similar cytotoxicity and cytokine production as TC-210, but notably enhanced T-cell expansion and persistence upon repeated stimulation with MSLN+ cell lines. Furthermore, the IL-15fu-enhanced TRuC-T cells sustained a significantly higher TCF-1+ population and retained a stem-like phenotype following activation. Moreover, the IL-15fu-enhanced TRuCs demonstrated robust in vivo expansion and intra-tumoral accumulation as measured by ex vivo analysis of TRuC+ cells in the tumor and blood, with a preferential expansion of CD8+ T cells. Finally, IL-15fu-enhanced TRuC-T cells could be observed in the blood long after the tumors were cleared.ConclusionsThese pre-clinical studies suggest that the IL-15fu can synergize with TC-210 to increase the potency and durability of response in patients with MSLN+ tumors.Ethics ApprovalAll animal studies were approved by the respective Institutional Animal Care and Use Committees.


2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Ioana Sandu ◽  
Dario Cerletti ◽  
Manfred Claassen ◽  
Annette Oxenius

Abstract Chronic viral infections are often associated with impaired CD8+ T cell function, referred to as exhaustion. Although the molecular and cellular circuits involved in CD8+ T cell exhaustion are well defined, with sustained presence of antigen being one important parameter, how much T cell receptor (TCR) signaling is actually ongoing in vivo during established chronic infection is unclear. Here, we characterize the in vivo TCR signaling of virus-specific exhausted CD8+ T cells in a mouse model, leveraging TCR signaling reporter mice in combination with transcriptomics. In vivo signaling in exhausted cells is low, in contrast to their in vitro signaling potential, and despite antigen being abundantly present. Both checkpoint blockade and adoptive transfer of naïve target cells increase TCR signaling, demonstrating that engagement of co-inhibitory receptors curtails CD8+ T cell signaling and function in vivo.


Blood ◽  
2003 ◽  
Vol 102 (5) ◽  
pp. 1764-1770 ◽  
Author(s):  
Karin Elflein ◽  
Marta Rodriguez-Palmero ◽  
Thomas Kerkau ◽  
Thomas Hünig

AbstractSlow recovery of T-cell numbers and function contributes to the high incidence of life-threatening infections after cytotoxic cancer therapies. We have tested the therapeutic potential of a novel class of superagonistic CD28–specific antibodies that induce polyclonal T-cell proliferation without T-cell receptor engagement in an experimental rat model of T lymphopenia. We show that in lethally irradiated, bone marrow–reconstituted hosts, CD28 superagonist is able to dramatically accelerate repopulation by a small inoculum of mature, allotype-marked T cells. CD28-driven recovery of CD4 cells was superior to that of CD8 T cells. CD28 superagonist– expanded CD4 T cells had maintained repertoire diversity and were functional both in vitro and in vivo, suggesting that treatment with a human CD28–specific superagonist will protect T-lymphopenic patients from opportunistic infections.


Blood ◽  
2002 ◽  
Vol 100 (10) ◽  
pp. 3656-3662 ◽  
Author(s):  
David A. Ingram ◽  
Lei Zhang ◽  
Jennifer McCarthy ◽  
Mary Jo Wenning ◽  
Lucy Fisher ◽  
...  

Ras plays an essential role in lymphocyte development and function. However, in vivo consequence(s) of regulation of Ras activity by guanosine triphosphatase (GTPase)–activating proteins (GAPs) on lymphocyte development and function are not known. In this study we demonstrate that neurofibromin, the protein encoded by theNF1 tumor suppressor gene functions as a GAP for Ras in T cells. Loss of Nf1 in T cells results in enhanced Ras activation, which is associated with thymic and splenic hyperplasia, and an increase in the absolute number of immature and mature T-cell subsets compared with control mice. Interestingly, in spite of a profound T-cell expansion and higher thymidine incorporation in unstimulated Nf1-deficient T cells, T-cell receptor and interleukin-2 receptor–mediated proliferation of thymocytes and mature T cells was substantially reduced compared with control mice. Collectively, these results identify neurofibromin as a GAP for Ras in T cells for maintaining immune homeostasis in vivo.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 66-66
Author(s):  
Jinfeng Yang ◽  
Abdulraouf Ramadan ◽  
Dawn Reichenbach ◽  
Michael Loschi ◽  
Jilu Zhang ◽  
...  

Abstract Allogeneic hematopoietic cell transplantation (allo-HCT) is an essential therapeutic modality for patients with hematological malignancies and benign blood disorders. Unfortunately, acute graft-versus-host disease (aGVHD) remains the major complication of allo-HCT and is associated with high mortality. Soluble STimulation-2 (sST2) is increased during aGVHD while regulatory T cells (Tregs) that express membrane-bound ST2 prevent GVHD through unknown mechanisms. Herein, we studied nonlymphoid tissue ST2+ Tregs in murine models of allo-HCT. Transplantation of Foxp3- T cells and Tregs sorted from different Foxp3 reporter mice indicated that ST2+Tregs isolated from aGVHD mice were of donor origin, thymus-derived, and predominantly intestine localized (data not shown). To further understand the mechanism, we performed a transcriptome analysis of sorted Foxp3GFP Tregs from aGVHD mice receiving wild type (WT) CD25-depleted total T cells with either WT or ST2-/- Foxp3GFP Tregs in both major (B6, H-2bàBALB/c, H-2d, Mhc) and minor (B6, H-2bàC3H.SW, H-2b, miH) mismatched aGVHD models. ST2-/- Foxp3GFP Tregs showed decreased expression of key molecules needed for Treg function such as Foxp3, Il2ra, Icos, Il27, Ctla4, and Ahr. Strikingly, transcription of RAR-related-orphan-receptor-c (Rorc), essential for proinflammatory IL-17+ T cells development, was the most upregulated transcript in ST2-/- Foxp3GFP Treg isolated from the gut in both models (Figure 1A). The transcriptome finding led us to investigate the frequency and function of ST2+Tregs in the gut of naive mice deficient for Rorc. Isolation and staining of intestinal T cells showed a significant increase of ST2+Treg frequency in the gut of Rorc-/- mice compared to WT mice: 55% vs. 12%, respectively (Figure 1B). Rorc-/- T cells transplantation enhanced the day 10 post-HCT frequency of intestinal ST2+Tregs (Figure 1C, left) that co-expressed TIGIT and Helios expression (data not shown), and decreased aGVHD score and mortality (Figure 1C, right). Previous studies have shown that IL-33 signaling in Tregs elicits their amphiregulin expression and increases their ability to suppress effector T cells (Teffs). IL-23, a key proinflammatory cytokine in the maintenance and amplification of Th17/Tc17, can restrain Treg responses by inhibiting IL-33 responsiveness. Using ex vivo polyclonal Tregs from donor B6 cultured with IL-2 (control Tregs) vs IL-2+IL-33 (TregIL-33) vs IL-2+IL-33+IL-23+IL-17, we found that adding IL-33 doubled the frequencies of ST2+ Tregs compared to control Tregs, and adding IL-23+IL-17 reversed this activity (Figure 2A). Markers of Tregs activation (KLRG1) (Figure 2B) and function (Helios, LAG3) (data not shown) were augmented in TregIL-33 compared to other Treg conditions. TregIL-33 also expressed more amphiregulin than Tregs cultured in the two other conditions (Figure 2C). Next, we evaluated their function in vitro and in vivo. TregIL-33 displayed better immunosuppression than other Tregs on CD4+ T cells at a physiological Tregs:Teffs ratio of 1:12; differences were not seen at higher ratios or on suppression of CD8+ T cells (Figure 3A). Then we examined the in vivo capacity of donor TregIL-33 to protect from aGVHD intestinal damage by co-adoptive transfer of Tregs cultured in the three aforementioned conditions with CD25-depleted WT T cells in Mhc and miH mismatch aGVHD models. We used a physiological ratio of 1 donor Treg per 20 donor Teffs (5% of total T cells). Mice receiving TregIL-33 showed significantly less severe aGVHD and better survival compared to those cultured either without IL-33 or with IL-33 and IL23/IL17 in both Mhc (Figure 3B) and miH (data not shown) mismatched aGVHD models. Analysis of intestinal T cells of these mice showed decreased total lymphocytes infiltration in the gut at day 10 after HCT, and decreased intestinal Tbet and RORγt expressing Teffs in both Mhc (Figure 3C) and miH (not shown) mismatched aGVHD models. Altogether, we conclude that within GVHD target organs, ST2+Tregs are predominantly intestine localized, and inversely correlated with RORgt in steady state and inflammatory conditions. Triggering the ST2/IL-33 pathway in Tregs prevents severe aGVHD, possibly through a dual mechanism: upregulation of amphiregulin and immunosuppression on effector T cells. Thus, TregIL-33 is a potential cellular therapy avenue for preventing or treating aGVHD. Disclosures Blazar: Kadmon Corporation, LLC: Consultancy, Research Funding. Paczesny:Viracor IBT Laboratories: Patents & Royalties.


1995 ◽  
Vol 181 (2) ◽  
pp. 713-721 ◽  
Author(s):  
C S Hsieh ◽  
S E Macatonia ◽  
A O'Garra ◽  
K M Murphy

A host's ability to resist certain pathogens such as Leishmania major can depend upon the phenotype of T helper (Th) subset that develops. Different murine genetic backgrounds are known to significantly alter the direction of Th subset development, although the cellular basis of this influence is poorly understood. To examine the basis of this effect we used an in vitro alpha/beta-T cell receptor (TCR) transgenic system for analysis of Th phenotype development. To control for TCR usage, we derived the DO11.10 alpha/beta-TCR transgene in several genetic backgrounds. Our findings suggest that the effects of genetic background on Th phenotype development reside within the T cell, and not the antigen-presenting cell compartment. Transgenic T cells from both the B10.D2 and BALB/c backgrounds showed development toward either the Th1 or Th2 phenotype under the strong directing influence of interleukin (IL) 12 and IL4, respectively. However, when T cells were activated in vitro under neutral conditions in which exogenous cytokines were not added, B10.D2-derived T cells acquired a significantly stronger Th1 phenotype than T cells from the BALB/c background, correspondent with in vivo Th responses to Leishmania in these strains. Importantly, these cytokine differences resulted in distinct functional properties, because B10.D2- but not BALB/c-derived T cells could induce macrophage production of nitric oxide, an important antimicrobial factor. Thus, the genetically determined default Th phenotype development observed in vitro may correspond to in vivo Th subset responses for pathogens such as Leishmania which do not initiate strong Th phenotype-directing signals.


Immunity ◽  
2011 ◽  
Vol 34 (3) ◽  
pp. 409-421 ◽  
Author(s):  
Yi Chen ◽  
Christopher J. Haines ◽  
Ilona Gutcher ◽  
Kristin Hochweller ◽  
Wendy M. Blumenschein ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document