scholarly journals Multiple Environmental Signaling Pathways Control the Differentiation of RORγt-Expressing Regulatory T Cells

2020 ◽  
Vol 10 ◽  
Author(s):  
Hind Hussein ◽  
Sébastien Denanglaire ◽  
Frédéric Van Gool ◽  
Abdulkader Azouz ◽  
Yousra Ajouaou ◽  
...  
2014 ◽  
Author(s):  
Juanjuan Zhao ◽  
Yongju Li ◽  
Yan Hu ◽  
Chao Chen ◽  
Ya Zhou ◽  
...  

Backgroud: CCR6+ CD4+ regulatory T cells (CCR6+Tregs), a distinct Tregs subset, played an important role in various immune diseases. Recent evidence showed that microRNAs (miRNAs) are vital regulators in the function of immune cells. However, the potential role of miRNAs in the function of CCR6+Tregs remains largely unknown. In this study, we detected the expression profile of miRNAs in CCR6+ Tregs. Materials and Methods: The expression profile of miRNAs as well as genes in CCR6+Tregs or CCR6-Tregs from Balb/c mice were detected by microarray. The signaling pathways were analyzed using Keggs pathway library. Results: We found that there were 58 miRNAs significantly upregulated and 62 downregulated up to 2 fold in CCR6+Tregs compared with CCR6-Tregs. Moreover, 1391 genes were observed with 3 fold change and 20 signaling pathways were enriched using Keggs pathway library. Conclusion: The present data firstly showed CCR6+Tregs expressed specific miRNAs pattern, which provide an insight into the role of miRNAs in the biological function of distinct Tregs subsets.


2014 ◽  
Author(s):  
Juanjuan Zhao ◽  
Yongju Li ◽  
Yan Hu ◽  
Chao Chen ◽  
Ya Zhou ◽  
...  

Backgroud: CCR6+ CD4+ regulatory T cells (CCR6+Tregs), a distinct Tregs subset, played an important role in various immune diseases. Recent evidence showed that microRNAs (miRNAs) are vital regulators in the function of immune cells. However, the potential role of miRNAs in the function of CCR6+Tregs remains largely unknown. In this study, we detected the expression profile of miRNAs in CCR6+ Tregs. Materials and Methods: The expression profile of miRNAs as well as genes in CCR6+Tregs or CCR6-Tregs from Balb/c mice were detected by microarray. The signaling pathways were analyzed using Keggs pathway library. Results: We found that there were 58 miRNAs significantly upregulated and 62 downregulated up to 2 fold in CCR6+Tregs compared with CCR6-Tregs. Moreover, 1391 genes were observed with 3 fold change and 20 signaling pathways were enriched using Keggs pathway library. Conclusion: The present data firstly showed CCR6+Tregs expressed specific miRNAs pattern, which provide an insight into the role of miRNAs in the biological function of distinct Tregs subsets.


2011 ◽  
Vol 186 (10) ◽  
pp. 5533-5537 ◽  
Author(s):  
Scott J. Patterson ◽  
Jonathan M. Han ◽  
Rosa Garcia ◽  
Kiran Assi ◽  
Tianyan Gao ◽  
...  

2015 ◽  
Vol 2015 ◽  
pp. 1-10 ◽  
Author(s):  
Shannon M. Ruppert ◽  
Ben A. Falk ◽  
S. Alice Long ◽  
Paul L. Bollyky

Cyclosporine A (CSA) is an immunosuppressive agent that specifically targets T cells and also increases the percentage of pro-tolerogenic CD4+Foxp3+ regulatory T cells (Treg) through unknown mechanisms. We previously reported that CD44, a receptor for the extracellular matrix glycosaminoglycan hyaluronan (HA), promotes Treg stability in IL-2-low environments. Here, we asked whether CD44 signaling also promotes Treg resistance to CSA. We found that CD44 cross-linking promoted Foxp3 expression and Treg viability in the setting of CSA treatment. This effect was IL-2 independent but could be suppressed using sc-355979, an inhibitor of Stat5-phosphorylation. Moreover, we found that inhibition of HA synthesis impairs Treg homeostasis but that this effect could be overcome with exogenous IL-2 or CD44-cross-linking. Together, these data support a model whereby CD44 cross-linking by HA promotes IL-2-independent Foxp3 expression and Treg survival in the face of CSA.


2017 ◽  
Vol 6 (8) ◽  
pp. e1338238 ◽  
Author(s):  
Dhifaf Sarhan ◽  
Caroline Leijonhufvud ◽  
Shannon Murray ◽  
Kristina Witt ◽  
Christina Seitz ◽  
...  

Shock ◽  
2011 ◽  
Vol 35 (3) ◽  
pp. 252-257 ◽  
Author(s):  
Marc Hanschen ◽  
Goro Tajima ◽  
Fionnuala O'Leary ◽  
Kimiko Ikeda ◽  
James A. Lederer

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 4722-4722
Author(s):  
Xueqiang Wu ◽  
Yan Chen ◽  
Chunsheng Han ◽  
Yiwen Gong ◽  
Dingfang Bu ◽  
...  

Abstract Abstract 4722 TNF is a pleiotropic cytokine with biphasic proinflammatory and immunosuppressive effects. Previous work clearly demonstrated that TNF has the capacity to preferentially activate and promote the proliferative expansion of CD4+FoxP3+ regulatory T cells (Tregs), which represent a basis for the paradoxical anti-inflammatory action of TNF. Our studies also indicate that TNFR2, one of the TNF receptor which is preferentially expressed by Tregs, mediates the Treg-activating effect of TNF. However, the molecular mechanism and signaling pathways mediated Treg-activating effect of TNF remain to be understood. In this study, we first further verified that TNFR2 transduces the activating signal of TNF on Tregs, based on the evidence that 1) human TNF, known to only bind to mouse TNFR1, did not activate mouse Tregs; 2) a blocking anti mouse TNFR2 Ab, but not TNFR1 Ab, dose-dependently inhibited TNF-mediated proliferation of mouse Tregs at concentrations of 2.5–500 ng/ml. We next examined the signaling pathways of TNFR2 leading to the proliferation of Tregs, by using specific small-molecule inhibitors. It is well established that the activation of IKK-NFkB is a major consequence of activation of TNFR2. However, small molecule inhibitors of NFkB signaling pathway, namely BAY11–7082 and Sulfasalazine, did not block TNF-mediated proliferation of Tregs. In contrast, small molecule inhibitors of MAPK signaling pathway, SB203580 (P38 MAPK inhibitor), SP600125 (JNK inhibitor) and PD98059 (Erk1/2 inhibitor), potently suppressed TNF-induced replication of Tregs in a dose-dependent manner. Our results indicate that TNF selectively stimulates the expansion of FoxP3+ Tregs through TNFR2. Activation of MAPK (ERK1/2,P38 and JNK), rather than NFkB, is responsible for this activity of TNF. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 3353-3353 ◽  
Author(s):  
Jaebok Choi ◽  
Matthew L Cooper ◽  
Kiran R. Vij ◽  
Bing Wang ◽  
Julie Ritchey ◽  
...  

Abstract The therapeutic benefits of allogeneic hematopoietic stem cell transplantation (allo-HSCT) for hematologic malignancies are primarily derived from an anti-leukemia effect that is mediated by T cells in donor grafts. Unfortunately, these T cells also mediate graft-versus-host disease (GvHD), the major complication of allo-HSCT. We have previously published that in vivo administration of JAK1/JAK2 inhibitors to murine allo-HSCT recipients of interferon gamma receptor deficient (IFNγR-/-) T cells results in 100% survival in a fully MHC-mismatched B6 to Balb/c allo-HSCT model (Choi et al., 2014, PLoS ONE). Since the infusion of IFNγR-/- T cells alone is associated with only ~70% survival, we hypothesize that JAK1/JAK2 inhibitors have either additional off-target effects or are inhibiting other non-IFNγR signaling pathways which are themselves dependent on JAK1/JAK2. The major other cytokine receptor signaling pathway mediated via both JAK1 and JAK2 is the interleukin 6 receptor (IL6R) signaling pathway. Thus, it is possible that JAK1/JAK2 inhibitors also block signaling through IL6R in addition to IFNγR. In addition, Alam et al. have recently reported that single nucleotide polymorphisms of donor IFNγ and IL6 are closely linked to gastrointestinal GvHD in patients (2015, BMT). Therefore, we examined if blockade of both IFNγR and IL6R signaling results in complete elimination of GvHD after a fully MHC-mismatched allo-HSCT in which B6 (H-2b) T cell-depleted bone marrow cells (5x106) along with B6 pan T cells (5x105) are intravenously injected into lethally irradiated Balb/c mice (H-2d). As shown in Fig. 1, we have found that blocking both IFNγR (IFNγR-/- T cells) and IL6R (α-IL6R Ab) signaling dramatically reduces GvHD and results in >95% survival. In addition, we found that blocking both IFNγR and IL6R signaling significantly increased regulatory T cells (Tregs) in peripheral blood (23.2% Foxp3+ in CD4+ T cells (n=17) vs 2.5% in WT T cell control (n=16) at day 27 after allo-HSCT), suggesting that increase in Tregs might be a potential mechanism underlying the reduced GvHD after dual blockade of IFNγR and IL6R signaling. Baricitinib is a potent and balanced JAK1/JAK2 inhibitor currently being clinically developed by Eli Lilly for the treatment of inflammatory diseases. We hypothesize that baricitinib will optimally block both IFNγR and IL6R signaling pathways and prevent GvHD. We found that that baricitinib is a potent suppressor of GvHD in B6 to Balb/c allo-HSCT models (100% survival), superior to ruxolitinib and similar to blockade of both IFNγR and IL6R signaling (Fig. 2A). Baricitinib increases Tregs in vivo (Fig. 2B) and reduces the ratio of IL5 (Th2 cytokine) to IL2 (cytokine for Treg induction) in plasma (p=0.0046), a potential diagnostic marker for GvHD (Fujii et al., 2006, Int J Mol Med), significantly better than ruxolitinib. Lastly, we found that baricitinib inhibits the expression of T-bet (Fig. 2C), which is the master transcription factor of Th1 cells, that are primary effector T cells in inducing GvHD. These data suggest that the suppression of GvHD by baricitinib results from increased Tregs and decreased Th1 and Th2 cells. We next examined if the prevention of GvHD by baricitinib is dependent on natural regulatory T cells (Tregs) in donor grafts. Tregs were depleted from donor pan T cells before allo-HSCT (B6 to Balb/c). We found that in vivo administration of baricitinib resulted in 70% survival (0% control, p<0.0001; 100% Treg-replete T cells + baricitinib. In addition, based on clinical GvHD score when recipients of Treg-replete T cells were compared with those of Treg-deplete T cells, the beneficial effect of Tregs in the donor grafts for the prevention of GvHD was observed only for the first two weeks after allo-HSCT (p≤0.01). Lastly, we examined whether baricitinib can cure ongoing GvHD by administering baricitinib starting at day 10 after allo-HSCT when GvHD is established (B6 to Balb/c). We found that baricitinib treatment results in a significant reduction of GvHD and 100% survival (10% control, p<0.0001). All of these data suggest that pharmacologic co-blockade of IFNγR and IL6R pathways is a promising therapeutic strategy to prevent and effectively treat established GvHD. The inhibitory effect of baricitinib, ruxolitinib, and blockade of IFNγR and IL6R on JAK-STAT signaling using JAK/STAT phosphorylation antibody arrays is currently being investigated and will be presented. Disclosures DiPersio: Incyte Corporation: Research Funding.


Sign in / Sign up

Export Citation Format

Share Document