scholarly journals Tissue environment, not ontogeny, defines intestinal intraepithelial T lymphocytes

2021 ◽  
Author(s):  
Alejandro J Brenes ◽  
Maud Vandereyken ◽  
Olivia J James ◽  
Jens Hukelmann ◽  
Laura Spinelli ◽  
...  

Tissue-resident intestinal intraepithelial T lymphocytes (T-IEL) patrol the gut and have important roles in regulating intestinal homeostasis. T-IEL include both induced T-IEL, derived from systemic antigen-experienced lymphocytes, and natural IEL, which are developmentally targeted to the intestine. While the processes driving T-IEL development have been elucidated, the precise roles of the different subsets and the processes driving activation and regulation of these cells remain unclear. To gain functional insights into these enigmatic cells, we used high-resolution, quantitative mass spectrometry to investigate the proteomic landscape of the main T-IEL populations in the gut. Comparing the proteomes of induced T-IEL and natural T-IEL subsets, with naive CD8+ T cells from lymph nodes exposes the dominant effect of the gut environment over ontogeny on T-IEL phenotypes. Analyses of protein copy numbers of >7000 proteins in T-IEL reveal skewing of the cell surface repertoire towards epithelial interactions and checkpoint receptors; strong suppression of the metabolic machinery indicating a high energy barrier to functional activation; and changes in T cell antigen receptor signalling pathways reminiscent of chronically activated T cells. These novel findings illustrate how multiple input signals need to be integrated to regulate T-IEL function.

eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Alejandro Brenes ◽  
Maud Vandereyken ◽  
Olivia J James ◽  
Harriet Watt ◽  
Jens Hukelmann ◽  
...  

Tissue-resident intestinal intraepithelial T lymphocytes (T-IEL) patrol the gut and have important roles in regulating intestinal homeostasis. T-IEL include both induced T-IEL, derived from systemic antigen-experienced lymphocytes, and natural IEL, which are developmentally targeted to the intestine. While the processes driving T-IEL development have been elucidated, the precise roles of the different subsets and the processes driving activation and regulation of these cells remain unclear. To gain functional insights into these enigmatic cells, we used high-resolution, quantitative mass spectrometry to compare the proteomes of induced T-IEL and natural T-IEL subsets, with naive CD8+ T cells from lymph nodes. This data exposes the dominant effect of the gut environment over ontogeny on T-IEL phenotypes. Analyses of protein copy numbers of >7000 proteins in T-IEL reveal skewing of the cell surface repertoire towards epithelial interactions and checkpoint receptors; strong suppression of the metabolic machinery indicating a high energy barrier to functional activation; upregulated cholesterol and lipid metabolic pathways, leading to high cholesterol levels in T-IEL; suppression of T cell antigen receptor signalling and expression of the transcription factor TOX, reminiscent of chronically activated T cells. These novel findings illustrate how T-IEL integrate multiple tissue-specific signals to maintain their homeostasis and potentially function.


1986 ◽  
Vol 163 (5) ◽  
pp. 1037-1050 ◽  
Author(s):  
J H Kehrl ◽  
L M Wakefield ◽  
A B Roberts ◽  
S Jakowlew ◽  
M Alvarez-Mon ◽  
...  

This study examines the potential role of transforming growth factor beta (TGF-beta) in the regulation of human T lymphocyte proliferation, and proposes that TGF-beta is an important autoregulatory lymphokine that limits T lymphocyte clonal expansion, and that TGF-beta production by T lymphocytes is important in T cell interactions with other cell types. TGF-beta was shown to inhibit IL-2-dependent T cell proliferation. The addition of picograms amounts of TGF-beta to cultures of IL-2-stimulated human T lymphocytes suppressed DNA synthesis by 60-80%. A potential mechanism of this inhibition was found. TGF-beta inhibited IL-2-induced upregulation of the IL-2 and transferrin receptors. Specific high-affinity receptors for TGF-beta were found both on resting and activated T cells. Cellular activation was shown to result in a five- to sixfold increase in the number of TGF-beta receptors on a per cell basis, without a change in the affinity of the receptor. Finally, the observations that activated T cells produce TGF-beta mRNA and that TGF-beta biologic activity is present in supernatants conditioned by activated T cells is strong evidence that T cells themselves are a source of TGF-beta. Resting T cells were found to have low to undetectable levels of TGF-beta mRNA, while PHA activation resulted in a rapid increase in TGF-beta mRNA levels (within 2 h). Both T4 and T8 lymphocytes were found to make mRNA for TGF-beta upon activation. Using both a soft agar assay and a competitive binding assay, TGF-beta biologic activity was found in supernatants conditioned by T cells; T cell activation resulted in a 10-50-fold increase in TGF-beta production. Thus, TGF-beta may be an important antigen-nonspecific regulator of human T cell proliferation, and important in T cell interaction with other cell types whose cellular functions are modulated by TGF-beta.


eLife ◽  
2017 ◽  
Vol 6 ◽  
Author(s):  
Alexandria C Wells ◽  
Keith A Daniels ◽  
Constance C Angelou ◽  
Eric Fagerberg ◽  
Amy S Burnside ◽  
...  

The differentiation of naive CD8 T cells into effector cytotoxic T lymphocytes upon antigen stimulation is necessary for successful antiviral, and antitumor immune responses. Here, using a mouse model, we describe a dual role for the let-7 microRNAs in the regulation of CD8 T cell responses, where maintenance of the naive phenotype in CD8 T cells requires high levels of let-7 expression, while generation of cytotoxic T lymphocytes depends upon T cell receptor-mediated let-7 downregulation. Decrease of let-7 expression in activated T cells enhances clonal expansion and the acquisition of effector function through derepression of the let-7 targets, including Myc and Eomesodermin. Ultimately, we have identified a novel let-7-mediated mechanism, which acts as a molecular brake controlling the magnitude of CD8 T cell responses.


Circulation ◽  
2015 ◽  
Vol 132 (suppl_3) ◽  
Author(s):  
Giovanni Cimmino ◽  
Giovanni Ciccarelli ◽  
Stefano Conte ◽  
Grazia Pellegrino ◽  
Luigi Insabato ◽  
...  

Background: Activation of T-cells plays an important role in the pathophysiology of acute coronary syndromes (ACS). We have previously shown that plaques from ACS patients are characterized by a selective oligoclonal expansion of T-cells, indicating a specific, antigen-mediated recruitment of T-cells within the unstable lesions. We have also previously reported that activated T-cells in vitro express functional Tissue Factor (TF) on their surface. At the moment, however it is not known whether expression of TF by T-cells may contribute to thrombus formation in vivo. Methods: Blood was collected from the aorta and the coronary sinus of 13 NSTEMI and 10 stable CAD patients. CD3+ cells were selectively isolated and TF gene expression (real time PCR)and protein levels (western blot) were evaluated. In additional 7 STEMI patients, thrombotic formation material was obtained from the occluded coronary artery by catheter aspiration during primary PCI. TF expression in CD3+ cells was evaluated by immunohistochemistry and confocal microscopy. Results: Transcardiac TF expression in CD3+ cells was significantly higher in NSTEMI patients as compared to CD3+ cells obtained from stable CAD patients. Interestingly, thrombi aspirated from STEMI patients resulted enriched in CD3+cells, which expressed TF on their surface as shown in the figure. Conclusions: Our data demonstrate that in patients with ACS, T-lymphocytes may express surface TF, thus contributing to the process of thrombus formation. This finding may shed new light into the pathophysiologyof ACS.


2002 ◽  
Vol 76 (18) ◽  
pp. 9103-9111 ◽  
Author(s):  
Odile Ducrey-Rundquist ◽  
Mireille Guyader ◽  
Didier Trono

ABSTRACT The metabolic and cell cycle status of primary T lymphocytes conditions their susceptibility to human immunodeficiency virus (HIV) and HIV-derived vectors. While in fully quiescent T lymphocytes the reverse transcription and nuclear import of these retroelements are impaired, leading to an abortive infection, various stimuli can induce a state of virus permissiveness. Here, we studied the modalities by which interleukin-7 (IL-7), an important controller of T-cell homeostasis, exerts this effect. IL-7-exposed cord blood T lymphocytes proliferated and were efficiently transduced by HIV-derived vectors. In contrast, similarly treated adult peripheral blood (PB) T lymphocytes failed to divide, and only a subset of these cells became infectible. HIV-resistant and -sensitive subsets of IL-7-treated PB T lymphocytes differed in cell cycle status but not in naïve, memory, or activation phenotypes. Nuclear factor of activated T cells was not induced by IL-7, and cyclosporine did not prevent HIV-mediated gene transfer. Furthermore, the phosphatidylinositol 3-kinase (PI3K) inhibitor wortmannin blocked IL-7-induced cell survival and Bcl-2 synthesis but had no effect on the acquisition of HIV susceptibility, suggesting that IL-7-induced HIV type 1 permissiveness is not mediated by the PI-3 K pathway and that, perhaps, the Jak/STAT5 pathway, the other known mediator of IL-7-triggered signaling in T cells, governs this process.


1981 ◽  
Vol 153 (1) ◽  
pp. 213-218 ◽  
Author(s):  
A Moretta ◽  
M C Mingari ◽  
B F Haynes ◽  
R P Sekaly ◽  
L Moretta ◽  
...  

Mixed lymphocyte reaction (MLR)-activated T cells were analyzed according to the expression of various cell surface markers by the specific cytotoxic T lymphocytes (CTL) generated in the MLR. CTL were found exclusively in a population of MLR-activated T cells that lacked detectable Fc gamma R but that expressed a surface antigen recognized by the 4F2 monoclonal antibody. In contrast, CTL were found in both the Ia-positive and Ia-negative cells after MLR activation. Thus, the specific CTL generated in the allogeneic MLR can be identified and isolated by virtue of the expression of a particular cell surface marker.


1992 ◽  
Vol 35 (1) ◽  
pp. 50-59 ◽  
Author(s):  
Assia Eljaafari ◽  
Isabelle Dorval ◽  
Dominique Zeliszewski ◽  
Sylvie Le Gac ◽  
Ghislaine Sterkers

Blood ◽  
1997 ◽  
Vol 90 (9) ◽  
pp. 3629-3639 ◽  
Author(s):  
Laurent Genestier ◽  
Romain Paillot ◽  
Nathalie Bonnefoy-Berard ◽  
Geneviéve Meffre ◽  
Monique Flacher ◽  
...  

Abstract In addition to their major function in antigen presentation and natural killer cell activity regulation, HLA class I molecules may modulate T-cell activation and proliferation. Monoclonal antibodies (MoAbs) that recognize distinct epitopes of HLA class I molecules were reported to interfere with T-cell proliferation. We show here that two MoAbs (mouse MoAb90 and rat YTH862) that bind to an epitope of the α1 domain of HLA class I heavy chain induce apoptotic cell death of activated, but not resting, peripheral T lymphocytes. Other reference anti-HLA class I antibodies specific for distinct epitopes of the α1 (B9.12.1), α2 (W6/32), or α3 (TP25.99) domains of the heavy chain decreased T-cell proliferation but had little or no apoptotic effect. Apoptosis shown by DNA fragmentation, phosphatidylserine externalization, and decrease of mitochondrial transmembrane potential was observed whatever the type of T-cell activator. Apoptosis did not result from Fas/Fas-L interaction and distinct though partly overlapping populations of activated T cells were susceptible to Fas– and HLA class I–mediated apoptosis, respectively. Induction of apoptosis did not require HLA class I cross-linking inasmuch as it could be observed with monovalent Fab′ fragments. The data indicate that MoAb90 and YTH862 directed against the α1 domain of HLA class I trigger apoptosis of activated T lymphocytes by a pathway which does not involve Fas-ligand.


2019 ◽  
Vol 20 (1) ◽  
Author(s):  
Wei Chen ◽  
Xianying Fang ◽  
Yuan Gao ◽  
Ke Shi ◽  
Lijun Sun ◽  
...  

Abstract Background T lymphocytes play an important role in contact hypersensitivity. This study aims to explore the immunosuppressive activity of SBF-1, an analog of saponin OSW-1, against T lymphocytes in vitro and in vivo. Methods Proliferation of T lymphocytes from lymph nodes of mice was determined by MTT assay. Flow cytometry analysis was performed to assess T cell activation and apoptosis. Levels of cytokines were determined by PCR and ELISA. BALB/c mice were sensitized and challenged with picryl chloride and thickness of left and right ears were measured. Results SBF-1 effectively inhibited T lymphocytes proliferation induced by concanavalin A (Con A) or anti-CD3 plus anti-CD28 at a very low dose (10 nM) but exhibited little toxicity in non-activated T lymphocytes at concentrations up to 10 μM. In addition, SBF-1 inhibited the expression of CD25 and CD69, as well as he phosphorylation of AKT in Con A-activated T cells. SBF-1 also induced apoptosis of activated T cells. In addition, SBF-1 also downregulated the induction of the T cell cytokines, IL-2 and IFN-γ in a dose-dependent manner. Furthermore, SBF-1 significantly suppressed ear swelling and inflammation in a mouse model of picryl chloride-induced contact hypersensitivity. Conclusions Our findings suggest that SBF-1 has an unique immunosuppressive activity both in vitro and in vivo mainly through inhibiting T cell proliferation and activation. Its mechanism appears to be related to the blockage of AKT signaling pathway.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4088-4088
Author(s):  
Serena Kimi Perna ◽  
Biagio De Angelis ◽  
Daria Pagliara ◽  
Lan Zhan ◽  
Cliona M Rooney ◽  
...  

Abstract Abstract 4088 Poster Board III-1023 Although adoptive transfer of antigen-specific CTLs is generally safe and can be clinically effective for the treatment of several malignancies, the administration of stimulatory cytokines may be required to sustain their long-term growth and persistence in vivo. IL2, a γ-chain T-cell growth cytokine, has been used clinically, but is associated with significant toxicities. In addition, IL2 supports the expansion and function of Tregs, counterbalancing its stimulatory effects on CTLs and favoring the establishment of an immune-protected microenvironment for cancer. IL15, like IL2, is a γ-chain cytokine capable of sustaining the expansion and function of effector T cells. We have explored whether this cytokine also shares with IL2 an unwanted stimulatory effect on Tregs. Naturally occurring Tregs (CD4+CD25bright) were isolated from buffy coat preparations from healthy volunteers (mean of Treg recovery: 0.7% ± 0.05% of the starting population of mononuclear cells). The suppressive function of isolated Tregs was confirmed by their ability to inhibit the proliferation of activated T lymphocytes labeled with carboxyfluorescin diacetate succinimidyl ester (CFSE) using FACS analysis to measure CFSE dilution after 5-6 days of culture (activated T cell:Treg ratio 1:1). The proliferation of activated T cells in the presence of Tregs was significantly reduced (28%±5%) as compared to activated T cells cultured in the presence of control CD4+CD25– T cells (59%±5%) (p<0.05). Following addition of IL15 (2.5 ng/mL), however, proliferation of activated T cells continued even in the presence of Tregs (83%±5% plus IL15 without Tregs vs. 80%±5% plus IL15 and Tregs) (p=0.9), suggesting that this cytokine mitigates the immunosuppressive effects of Tregs. We then analyzed whether Tregs affected the anti-tumor activity of antigen-specific CTLs. We used our Epstein-Barr-Virus-(EBV)-specific CTLs as tumor model. EBV-CTLs were co-cultured with EBV-infected cells (LCLs) (CTL:LCL ratio 1:2). Residual tumor cells were enumerated by FACS analysis after 5-7 days of culture. In the absence of exogenous IL-15, EBV-CTLs failed to eliminate EBV-infected cells (residual LCLs: 37%±8%), while the addition of IL15 (2.5 ng/mL) increased the anti-tumor effect of CTLs, so that only 4%±1% tumor cells were detectable at the end of the culture. We then explored the effects of adding Treg to the cultures (Treg:CTL ratio 1:1). The percentage of tumor cells increased rather than decreased by day 5-7 when CTLs were cultured with Tregs in the absence of IL15 (residual tumor cells from 37%±8% in the absence of Tregs to 53%±9% in the presence of Tregs) (p<0.05). When IL15 was added, Treg were more limited in their ability to inhibit T effector cells, so that residual tumor cells were 4%±1% and 11%±3% % in the absence or in the presence of Tregs, respectively. To discover if IL15 has a direct effect on Tregs, we analyzed STAT5 phosphorylation after exposing Tregs to the cytokine. We found that this molecule was phosphorylated in 47%±18% of Tregs 15 minutes after exposure to IL15 (2.5 ng/mL). This effect was mediated by the specific interaction of the cytokine with its own receptor, as no phosphorylation occurred when Treg cells were pre-incubated with an IL-15Rα blocking antibody. This action on Tregs notwithstanding, IL15 stimulation did not modulate Treg inhibitory function, since these cells, even after exposure to IL15 (2.5 ng/mL) for 3-5 days, continued to significantly inhibit the proliferation of T lymphocytes activated in the absence of IL-15 (74%±17% inhibition). Hence, IL15 enhance the proliferative and anti-tumor effects of antigen-specific CTLs, and these effects are not impaired by the presence of Tregs. Administration of IL15 may therefore benefit patients receiving adoptive T cell therapies. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document