scholarly journals Extracellular Vesicles isolated from Mesenchymal Stromal Cells Modulate CD4+ T Lymphocytes Toward a Regulatory Profile

Cells ◽  
2020 ◽  
Vol 9 (4) ◽  
pp. 1059 ◽  
Author(s):  
Flavia Franco da Cunha ◽  
Vinicius Andrade-Oliveira ◽  
Danilo Candido de Almeida ◽  
Tamiris Borges da Silva ◽  
Cristiane Naffah de Souza Breda ◽  
...  

Mesenchymal stromal cells (MSCs) can generate immunological tolerance due to their regulatory activity in many immune cells. Extracellular vesicles (EVs) release is a pivotal mechanism by which MSCs exert their actions. In this study, we evaluate whether mesenchymal stromal cell extracellular vesicles (MSC-EVs) can modulate T cell response. MSCs were expanded and EVs were obtained by differential ultracentrifugation of the supernatant. The incorporation of MSC-EVs by T cells was detected by confocal microscopy. Expression of surface markers was detected by flow cytometry or CytoFLEX and cytokines were detected by RT-PCR, FACS and confocal microscopy and a miRNA PCR array was performed. We demonstrated that MSC-EVs were incorporated by lymphocytes in vitro and decreased T cell proliferation and Th1 differentiation. Interestingly, in Th1 polarization, MSC-EVs increased Foxp3 expression and generated a subpopulation of IFN-γ+/Foxp3+T cells with suppressive capacity. A differential expression profile of miRNAs in MSC-EVs-treated Th1 cells was seen, and also a modulation of one of their target genes, TGFbR2. MSC-EVs altered the metabolism of Th1-differentiated T cells, suggesting the involvement of the TGF-β pathway in this metabolic modulation. The addition of MSC-EVs in vivo, in an OVA immunization model, generated cells Foxp3+. Thus, our findings suggest that MSC-EVs are able to specifically modulate activated T cells at an alternative regulatory profile by miRNAs and metabolism shifting.

2017 ◽  
Vol 2017 ◽  
pp. 1-15 ◽  
Author(s):  
Yury Rubtsov ◽  
Кirill Goryunov ◽  
Аndrey Romanov ◽  
Yulia Suzdaltseva ◽  
George Sharonov ◽  
...  

Mesenchymal stromal cells (MSC) control excessive inflammation and create a microenvironment for tissue repair protecting from chronic inflammation and tissue fibrosis. We examined the molecular mechanisms of MSC immunomodulatory function in mixed cultures of human adipose-derived MSC with lymphocytes. Our data show that MSC promote unstimulated lymphocyte survival potentially by an increase in antigen presentation. Under inflammatory conditions, mimicked by stimulation of TCR in lymphocytes, MSC suppress activation and proliferation of stimulated T cells. Immunosuppression is accompanied by downregulation of IL-2Rαthat negatively affects the survival of activated T cells. MSC upregulate transcription of indolamine-2,3-dioxygenase (IDO) and inducible NO synthase (iNOS), which generate products negatively affecting T cell function. Both MSC and lymphocytes dramatically increase the surface ICAM-1 level in mixed cultures. Antibody-mediated blockage of surface ICAM-1 partially releases MSC-mediated immune suppression in vitro. Our data suggest that MSC have cell-intrinsic molecular programs depending on the inflammatory microenvironment. We speculate that MSC sense soluble factors and respond by surface ICAM-1 upregulation. ICAM-1 is involved in the control of T cell activation leading to immunosuppression or modest stimulation depending on the T cell status. Immunomodulation by MSC ranging from support of naive T cell survival to immunosuppression of activated T cells may affect the tissue microenvironment protecting from aberrant regeneration.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 3674-3674
Author(s):  
Rodrigo A Panepucci ◽  
Felipe Saldanha-Araujo ◽  
Kelen C R Malmegrim ◽  
Fabio M Oliveira ◽  
Patricia V B Palma ◽  
...  

Abstract Abstract 3674 Poster Board III-610 HLA-G is a nonclassic human leukocyte antigen which is characterized by its limited variability, its highly tissue-specific expression and for its very distinct immunological role. Instead of triggering immune responses, HLA-G is exclusively inhibitory, suppressing immune cell functions. For instance, HLA-G is expressed by cells in sites considered immunologically privileged, such as the fetal cytotrophoblast, at the fetal–maternal interface, were it acts protecting the fetal tissue from the mother's immune response. HLA-G is also pathologically expressed in a diverse set of tumors, acting as an immunescape mechanism. Trough splicing mechanisms, HLA-G can be expressed as a membrane-bound or as a soluble isoform. Interestingly, the transference of membrane fragments between cells, a process called trogocytosis, can spread HLA-G inhibitory function beyond the reach of HLA-G-expressing cells. For instance, upon trogocytosis, effector CD4+ T cells stop proliferating, stop responding to stimulation, and behave as regulatory T cells. Recently, mesenchymal stromal cells (MSC) were shown to secrete soluble HLA-G, adding to the existing set of secreted molecules, by which MSC can modulate cells of the immune system. Despite the importance of secreted factors, cell-to-cell contacts have an important role in the immunological effects exerted by MSC. Based in these facts, we hypothesized that one of the mechanisms used by MSC to immunomodulate T cells, could involve trogocytosis mediated HLA-G transference. To test this, CD3+ T cell were immunomagnetically selected from peripheral blood mononuclear cells (PBMC) and pre-activated for 72hs using anti-CD2/CD3/CD28 beads. Activated T-cells were then incubated for 30 min, alone or with MSC. After this period, cells were recovered and the transfer of membrane bound molecules from MSC to T-cells was analyzed by flow cytometry (3 experiments) using antibodies against HLA-G. Confocal microscopy was carried in an additional experiment, using antibodies against HLA-G, CD140B (a specific MSC marker) and CD3. DAPI was used for nuclear staining. Flow cytometry analysis revealed that activated CD3+ T-cells did not express HLA-G, while MSC expressed HLA-G intracytoplasmatic. After 30 min of co-incubation, membrane bound HLA-G was detected in a variable percentage of CD3+ cells, ranging from 3 to up to 16%, characterizing the transfer of HLA-G from MSC to CD3+ T-cells. Confocal microscopy revealed that activated T-cells cultured alone did not stained for HLA-G or CD140B, while, about 12% of the CD3+ cells stained for membrane HLA-G, following the 30 min incubation with MSC. Additionally, CD140B was also transferred from MSC to CD3+ T-cells. These data are the first demonstration of trogocytosis mediated transfer of HLA-G from MSC to activated T lymphocytes. Given the significance of HLA-G expression and trogocytosis in normal and pathological situations, this newly described immunological mechanism should be considered in the development and application of MSC-based therapies. Supported by CNPq and FAPESP. Disclosures: No relevant conflicts of interest to declare.


2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii44-iii44
Author(s):  
R Pedrosa ◽  
J M Kros ◽  
B Schrijver ◽  
R Marques ◽  
P Leenen ◽  
...  

Abstract BACKGROUND In previous work we showed the prominence of the T-cell response in the formation of brain metastases of primary ER negative breast cancers (Mustafa et al, Acta Neuropathol 2018). We also showed that breast cancer cells co-cultured with stimulated T lymphocytes overexpress Guanylate-binding protein 1 (GBP1) accompanying increased trespassing ability through an in vitro blood-brain barrier (BBB) model. In addition, we demonstrated a predilection for metastasizing to brain of breast cancer cells that were co-cultured with activated T cells in a mouse model. We now scrutinize the importance of the IFNγ pathway for tresspassing of the tumor cells through the BBB following T cell contact. MATERIAL AND METHODS Anti-hIFN-γ-IgA antibodies were used to neutralize the IFNγ effects on the tumor cells. The effects on the tumor cells is only due to native IFNγ produced by activated T cells, not by recombinant IFNγ. Since the IFNγ expression itself enhances its expression by the T cells, we blocked IFNγ receptors prior to adding CD3+ T cell conditioned media to the breast cancer cells. The receptor blocking was achieved by antibodies to the IFNγα and IFNγβ subunits. Activation of the STAT1 pathway was monitored by GBP1 expression. For functional read-out the in vitro BBB model was used. RESULTS The presence of T-lymphocyte-secreted IFNγ in the primary breast cancer microenvironment activates the STAT1-dependent IFNγ pathway in breast cancer cells, endowing them with an increased ability to trespass the in vitro BBB. Moreover, direct inhibition of soluble IFNγ, or blocking of the IFNγ-specific receptor in breast cancer cells significantly decreases their ability to cross the BBB. CONCLUSION The results illustrate the specific action of T lymphocytes in the formation of cerebral metastasis involves the IFNγ signaling pathway as one of the crucial entangled pathways Subsequent studies should aim at the interference with the IFNγ pathway to develop preventive strategies against the formation of cerebral metastases of breast cancer.


2019 ◽  
Vol 2019 ◽  
pp. 1-13 ◽  
Author(s):  
Ryutaro Oba ◽  
Motomichi Isomura ◽  
Akira Igarashi ◽  
Kinya Nagata

Extracellular vesicles (EVs) are known to contain unique proteins that reflect the cells of origins. Activated T cells are reported to secrete various EVs. To establish T cell subset-specific biomarkers, we performed proteomic analysis with Th1- and Th2-derived EVs and identified HLA-DR as a Th1-dominated EV membrane protein. We designed a measurement system for CD3+CD4+, CD3+CD8+, and CD3+HLA-DR+EVs to specifically determine EV subpopulations derived from CD4+, CD8+, and Th1-type T cells, respectively.In vitroanalysis showed that CD3+CD4+EVs and CD3+CD8+EVs were selectively secreted from activated CD4+and CD8+T cells, respectively, and that CD3+HLA-DR+EVs were actively secreted from not only Th1 but also activated CD8+T (probably mostly Tc1) cells. To evaluate the clinical usefulness of these EVs, we measured the serum levels in patients with inflammatory diseases, including Epstein-Barr virus (EBV,n=13) infection, atopic dermatitis (AD,n=10), rheumatoid arthritis (RA,n=20), and osteoarthritis (OA,n=20) and compared the levels with those of healthy adults (n=20). CD3+CD4+EVs were significantly higher in all of EBV infection, AD, RA, and OA while CD3+CD8+EVs were higher in EBV infection, lower in RA, and not different in AD and OA relative to the control. The levels of CD3+HLA-DR+EVs were markedly higher in EBV infection and significantly lower in AD. The results suggest that these EV subpopulations reflectin vivoactivation status of total CD4+, total CD8+, and Th1/Tc1-type T cells, respectively, and may be helpful in T cell-related clinical settings, such as cancer immunotherapy and treatment of chronic infection, autoimmune diseases, and graft-versus-host disease.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Do Hee Kwon ◽  
Jae Berm Park ◽  
Joo Sang Lee ◽  
Sung Joo Kim ◽  
Bongkum Choi ◽  
...  

AbstractHuman delta-like 1 (hDlk1) is known to be able to regulate cell fate decisions during hematopoiesis. Mesenchymal stromal cells (MSCs) are known to exhibit potent immunomodulatory roles in a variety of diseases. Herein, we investigated in vivo functions of hDlk1-hMSCs and hDlk1+hMSCs in T cell development and T cell response to viral infection in humanized NOD/SCID/IL-2Rγnull (NSG) mice. Co-injection of hDlk1-hMSC with hCD34+ cord blood (CB) cells into the liver of NSG mice markedly suppressed the development of human T cells. In contrast, co-injection of hDlk1+hMSC with hCD34+ CB cells into the liver of NSG dramatically promoted the development of human T cells. Human T cells developed in humanized NSG mice represent markedly diverse, functionally active, TCR V$$\upbeta $$ β usages, and the restriction to human MHC molecules. Upon challenge with Epstein-Barr virus (EBV), EBV-specific hCD8+ T cells in humanized NSG mice were effectively mounted with phenotypically activated T cells presented as hCD45+hCD3+hCD8+hCD45RO+hHLA-DR+ T cells, suggesting that antigen-specific T cell response was induced in the humanized NSG mice. Taken together, our data suggest that the hDlk1-expressing MSCs can effectively promote the development of human T cells and immune response to exogenous antigen in humanized NSG mice. Thus, the humanized NSG model might have potential advantages for the development of therapeutics targeting infectious diseases in the future.


2021 ◽  
Vol 12 ◽  
Author(s):  
Haiping He ◽  
Atsuko Takahashi ◽  
Takeo Mukai ◽  
Akiko Hori ◽  
Miwako Narita ◽  
...  

Mesenchymal stromal cells (MSCs) are known to have immunosuppressive ability and have been used in clinical treatment of acute graft-versus-host disease, one of severe complications of the hematopoietic stem cell transplantation. However, MSCs are activated to suppress the immune system only after encountering an inflammatory stimulation. Thus, it will be ideal if MSCs are primed to be activated and ready to suppress the immune reaction before being administered. Triptolide (TPL) is a diterpene triepoxide purified from a Chinese herb-Tripterygium wilfordii Hook.f. It has been shown to possess anti-inflammatory and immunosuppressive properties in vitro. In this study, we aimed to use TPL to prime umbilical cord-derived MSCs (TPL-primed UC-MSCs) to enter a stronger immunosuppressive status. UC-MSCs were primed with TPL, which was washed out thoroughly, and the TPL-primed UC-MSCs were resuspended in fresh medium. Although TPL inhibited the proliferation of UC-MSCs, 0.01 μM TPL for 24 h was tolerable. The surface markers of TPL-primed UC-MSCs were identical to those of non-primed UC-MSCs. TPL-primed UC-MSCs exhibited stronger anti-proliferative effect for activated CD4+ and CD8+ T cells in the allogeneic mixed lymphocyte reaction assay than the non-primed UC-MSCs. TPL-primed UC-MSCs promoted the expression of IDO-1 in the presence of IFN-γ, but TPL alone was not sufficient. Furthermore, TPL-primed UC-MSCs showed increased expression of PD-L1. Conclusively, upregulation of IDO-1 in the presence of IFN-γ and induction of PD-L1 enhances the immunosuppressive potency of TPL-primed UC-MSCs on the proliferation of activated T cells. Thus, TPL- primed MSCs may provide a novel immunosuppressive cell therapy.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2417-2417
Author(s):  
Felipe Saldanha Araujo ◽  
Rodrigo Alexandre Panepucci ◽  
Kelen Cristina Farias ◽  
Amelia G Araujo ◽  
Maristela Delgado Orellana ◽  
...  

Abstract Mesenchymal stromal cells (MSCs) exert an immune regulatory function and suppress T-cell proliferation in vitro and in vivo. One of the ways by which MSCs may modulate immune responses is by the induction of CD4+CD25+FOXP3+ regulatory T cells (Treg), which suppress alloreactivity in vitro and prevent or attenuate GVHD in animal models. TGF-b (TGFB1 gene), one of the factors secreted by MSC, is known to induce the expression of FOXP3 and to drive the generation of Tregs from CD4+CD25− T cells. T cell activation is characterized by the expression of many surface molecules, and CD69 is one of the earliest markers, transiently expressed following activation. CD69 is selectively expressed at sites of chronic inflammation and recent in vivo and in vitro results indicate that this receptor may modulate the inflammatory response, by inducing TGF-b production. Despite the importance of secreted factors, cell to cell contacts promote increased lymphocyte immune modulation. Interestingly, TGF-b is known to induce the expression of b-IG-H3 (TGFBI gene), a secreted extracellular matrix adaptor protein, whose expression is higher on hematopoietic stem cells adherent to MSC. Given the potential role of CD69 as a regulatory molecule, we explored the effects of MSC on the expression of CD69, TGFB1, FOXP3 and TGFBI on co-cultured T-cells. Peripheral blood mononuclear cells (PBMC) from 6 individuals were co-activated by anti-CD3/CD28 beads and cultured either in the presence or in the absence of MSC (5:1) adhered to the bottom of culture wells. IL2 (20U/ml) was added in the 3rd day for full activation. Cultured PBMC were collected 1, 3 and 5 days after activation. Percentage of CD69+ cells and proliferation of activated lymphocytes (cell division tracking by CFSE) were evaluated by flow cytometry on gated CD3+ lymphocytes. Transcripts levels of TGFB1, TGFBI, FOXP3 and IL10 were determined by real time PCR and normalized using ACTB as an endogenous control. Relative expression levels were calculated in comparison with activated PBMC cultivated alone at the 5th day. Proliferation of lymphocytes co-cultured with MSC was significantly inhibited. As expected, activation of lymphocytes cultured alone was evidenced by the expression of CD69 in 5% of cells in the first day (mean percentage), followed by a decrease in the subsequent days (4% and 3%, respectively). Interestingly, lymphocytes co-cultured with MSC displayed a completely different pattern, with a similar initial activation (7%) which was followed by significant increase in the 3rd day (16%), maintained in the 5th day (14%). Furthermore, TGFBI and IL10 were both expressed at significantly higher levels on PBMC co-cultured with MSC, compared to PBMC alone, in all days evaluated. In addition, their transcript levels decreased faster on PBMC cultured alone. In the other hand, TGFB1 and FOXP3 levels in PBMC cultured alone or in the presence of MSC were similarly higher in the 1st day, but decreased till the 5th day, when their levels were slightly, but significantly higher on PBMC co-cultured with MSC, compared to PBMC cultured alone. We demonstrate for the first time that co-culture with MSC causes a sustained increase of the CD69 marker on CD3+ lymphocytes, which is accompanied by increased levels of TGFB1, TGFBI, FOXP3 and IL10 on total PBMC. Our results are in line with the proposed immunoregulatory role of CD69. In addition, higher TGFBI levels on PBMC may increase lymphocyte adherence to MSC, thus favoring immune modulation. This work was supported by FINEP, CNPq and FAPESP.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 953-953
Author(s):  
Shan He ◽  
Fang Xie ◽  
Qing Tong ◽  
Kazuhiro Mochizuki ◽  
Yongnian Liu ◽  
...  

Abstract Abstract 953 Adoptive T cell therapy has the potential to enhance antitumor immunity and improve vaccine efficacy, of which a key challenge is to generate sufficient numbers of T cells that can persist in vivo after transfer. Cellular metabolism plays important roles in regulating T cell proliferation and survival. T cells responding to antigen activation dramatically upregulate both glycolysis and oxidative phosphorylation (OXPHOS), leading to increased production of adenosine triphosphate (ATP) and metabolic intermediates that are required for cell growth and proliferation. Without sufficient support for their demands, activated T cells may be deleted or become quiescent. Thus, better understanding of the mechanism that regulates cellular metabolism in T cell response will lead to new strategies to improve the efficacy of adoptive T cell therapy. Here we explore the functional impact of an epigenetic pathway in cellular metabolism in antigen-driven T cells and tumor immunity. Using genetic approaches and experimental mouse models, we demonstrate that Ezh2, which is a histone methyltransferase that represses the transcription of cohorts of developmental regulators, promotes the survival and expansion of antigen-driven T cells through regulating bioenergetic metabolism. Conditional deletion of Ezh2 caused selective apoptosis in T cells upon activation with alloantigens in vivo and in vitro or with T cell receptor (TCR)-ligation in vitro. Ezh2 deficiency resulted in markedly increased expression of proapoptotic gene Bim, but had no significant impact on the expression of other Bcl-2 family members (e.g., anti-apoptotic genes Bcl-2 and Bcl-xL,). Genetic inactivation of Bim only slightly improved the survival of alloantigen-activated Ezh2-deficient T cells, suggesting that Ezh2 may control T-cell immunity largely through a Bim-independent mechanism. This differs from our recent observations showing that Bim is required for increased apoptosis in activated T cells treated with a pharmacologic inhibitor of Ezh2 and histone methylation 3-Deazaneplanocin A (Blood, 2012). Our prior studies and others suggest that impaired cellular metabolism may lead to increased apoptosis of antigen-activated T cells. We observed that upon TCR-ligation Ezh2 null T cells were incapable to upregulate OXPHOS as compared to wild-type (WT) T cells, which was accompanied with reduced ATP levels and increased reactive oxygen species (ROS). Neutralization of ROS by N-acetylcysteine significantly improved the survival of TCR-activated Ezh2 null T cells. Interestingly, overexpression of WT Ezh2 in TCR-activated Ezh2 null T cells, but not enzymatically inactive H689A Ezh2 mutant or nuclear localization-inactive Ezh2 mutant, restored the ability of Ezh2-deficient T cells to upregulate OXPHOS, reduced ROS levels, and rescued their survival capability in vitro. These results suggest that Ezh2 is important for regulating bioenergetic metabolism in activated T cells. Furthermore, the nuclear but not cytoplasmic Ezh2 is required to regulate bioenergetic metabolism in activated T cells during clonal expansion phase, although Ezh2 in the cell cytoplasm could be involved in regulating actin polymerization. In mouse models of graft-versus-host disease (GVHD) and leukemia, transfer of donor T cells lacking Ezh2 failed to mediate GVHD and anti-leukemia activity in mice receiving allogeneic bone marrow transplantation. In addition, Ezh2 deficiency also ablated the ability of adoptively transferred antigen-specific CD8 T cells to control tumor growth in mice with established melanoma. Importantly, the absence of Ezh2 did not impair the development of effector T cells producing IFN-γ, granzyme B, Fas ligand and Trail, ruling out the possibility that impaired T-cell immunity of Ezh2 null T cells results from defective effector differentiation. Our findings identify the critical role of Ezh2 in regulating bioenergetic metabolism in antigen-driven T cells, therefore for the first time linking the epigenetic pathway to cellular metabolism in T cell response. Thus, Ezh2 and its-regulated bioenergetic metabolism may represent novel targets to improve the efficacy of adoptive T-cell immunotherapy. Modulation of Ezh2 and its activity may have broad implications in the treatment of many other inflammatory disorders, such as graft rejection after organ transplantation, GVHD and autoimmune diseases. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 22 (13) ◽  
pp. 6837
Author(s):  
Pauline Rozier ◽  
Marie Maumus ◽  
Claire Bony ◽  
Alexandre Thibault Jacques Maria ◽  
Florence Sabatier ◽  
...  

Systemic sclerosis (SSc) is a complex disorder resulting from dysregulated interactions between the three main pathophysiological axes: fibrosis, immune dysfunction, and vasculopathy, with no specific treatment available to date. Adipose tissue-derived mesenchymal stromal cells (ASCs) and their extracellular vesicles (EVs) have proved efficacy in pre-clinical murine models of SSc. However, their precise action mechanism is still not fully understood. Because of the lack of availability of fibroblasts isolated from SSc patients (SSc-Fb), our aim was to determine whether a TGFβ1-induced model of human myofibroblasts (Tβ-Fb) could reproduce the characteristics of SSc-Fb and be used to evaluate the anti-fibrotic function of ASCs and their EVs. We found out that Tβ-Fb displayed the main morphological and molecular features of SSc-Fb, including the enlarged hypertrophic morphology and expression of several markers associated with the myofibroblastic phenotype. Using this model, we showed that ASCs were able to regulate the expression of most myofibroblastic markers on Tβ-Fb and SSc-Fb, but only when pre-stimulated with TGFβ1. Of interest, ASC-derived EVs were more effective than parental cells for improving the myofibroblastic phenotype. In conclusion, we provided evidence that Tβ-Fb are a relevant model to mimic the main characteristics of SSc fibroblasts and investigate the mechanism of action of ASCs. We further reported that ASC-EVs are more effective than parental cells suggesting that the TGFβ1-induced pro-fibrotic environment may alter the function of ASCs.


Sign in / Sign up

Export Citation Format

Share Document