scholarly journals Mechanisms behind the Immunoregulatory Dialogue between Mesenchymal Stem Cells and Th17 Cells

Cells ◽  
2020 ◽  
Vol 9 (7) ◽  
pp. 1660 ◽  
Author(s):  
Claudia Terraza-Aguirre ◽  
Mauricio Campos-Mora ◽  
Roberto Elizondo-Vega ◽  
Rafael A. Contreras-López ◽  
Patricia Luz-Crawford ◽  
...  

Mesenchymal stem cells (MSCs) exhibit potent immunoregulatory abilities by interacting with cells of the adaptive and innate immune system. In vitro, MSCs inhibit the differentiation of T cells into T helper 17 (Th17) cells and repress their proliferation. In vivo, the administration of MSCs to treat various experimental inflammatory and autoimmune diseases, such as rheumatoid arthritis, type 1 diabetes, multiple sclerosis, systemic lupus erythematosus, and bowel disease showed promising therapeutic results. These therapeutic properties mediated by MSCs are associated with an attenuated immune response characterized by a reduced frequency of Th17 cells and the generation of regulatory T cells. In this manuscript, we review how MSC and Th17 cells interact, communicate, and exchange information through different ways such as cell-to-cell contact, secretion of soluble factors, and organelle transfer. Moreover, we discuss the consequences of this dynamic dialogue between MSC and Th17 well described by their phenotypic and functional plasticity.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ji-wen Cheng ◽  
Li-xia Duan ◽  
Yang Yu ◽  
Pu Wang ◽  
Jia-le Feng ◽  
...  

Abstract Background Mesenchymal stem cells (MSCs) play a crucial role in cancer development and tumor resistance to therapy in prostate cancer, but the influence of MSCs on the stemness potential of PCa cells by cell–cell contact remains unclear. In this study, we investigated the effect of direct contact of PCa cells with MSCs on the stemness of PCa and its mechanisms. Methods First, the flow cytometry, colony formation, and sphere formation were performed to determine the stemness of PCaMSCs, and the expression of stemness-related molecules (Sox2, Oct4, and Nanog) was investigated by western blot analysis. Then, we used western blot and qPCR to determine the activity levels of two candidate pathways and their downstream stemness-associated pathway. Finally, we verified the role of the significantly changed pathway by assessing the key factors in this pathway via in vitro and in vivo experiments. Results We established that MSCs promoted the stemness of PCa cells by cell–cell contact. We here established that the enhanced stemness of PCaMSCs was independent of the CCL5/CCR5 pathway. We also found that PCaMSCs up-regulated the expression of Notch signaling-related genes, and inhibition of Jagged1-Notch1 signaling in PCaMSCs cells significantly inhibited MSCs-induced stemness and tumorigenesis in vitro and in vivo. Conclusions Our results reveal a novel interaction between MSCs and PCa cells in promoting tumorigenesis through activation of the Jagged1/Notch1 pathway, providing a new therapeutic target for the treatment of PCa.


Author(s):  
Quan Shi ◽  
Qi He ◽  
Weiming Chen ◽  
Jianwen Long ◽  
Bo Zhang

IntroductionOleuropein (OLP) is polyphenol obtained from olive oil; it is proved in Chinese traditional medicine for its use in disorders including autoimmune and inflammatory disorders. Psoriasis (PSR) is an autoimmune and inflammatory disorder triggered by T-helper-17 (Th17) cells.Material and methodsWe developed an imiquimod (IMQ)-mediated PSR model in mice to study the anti-inflammatory role of OLP in psoriasis. The mice were given 50 mg/kg and 100 mg/kg dose of OLP. Histology was done to assess the inflammation of lesions. Western blot analysis was done for JAK3/STAT3 in isolated T cells, expression of RORgt was done by RT-PCR. The In silico molecular docking studies were done for interaction of OLP with target protein STAT3 and JAK3.ResultsTreatment of OLP attenuated proliferation in IMQ-mediated keratinocytes, improved infiltration of CD3+ cells in the skin lesions and in CD4+ and CD8+ T cells and also ameliorated the levels of cytokines. In in vitro studies in isolated T cells, OLP blocked the differentiation of Th17 cells and also the levels of IL-17 and the JAK3/STAT3 pathway. The in silico docking showed that OLP had potential binding affinity with JAK3 and STAT3 which was parallel to in vivo and in vitro findings.ConclusionsOLP ameliorates psoriasis skin lesions by blocking Th17-mediated inflammation. OLP may be an interesting molecule for treating autoimmunity in psoriasis.


2019 ◽  
Vol 28 (12) ◽  
pp. 1490-1506 ◽  
Author(s):  
Yu You ◽  
Di-guang Wen ◽  
Jian-ping Gong ◽  
Zuo-jin Liu

Liver transplantation has been deemed the best choice for end-stage liver disease patients but immune rejection after surgery is still a serious problem. Patients have to take immunosuppressive drugs for a long time after liver transplantation, and this often leads to many side effects. Mesenchymal stem cells (MSCs) gradually became of interest to researchers because of their powerful immunomodulatory effects. In the past, a large number of in vitro and in vivo studies have demonstrated the great potential of MSCs for participation in posttransplant immunomodulation. In addition, MSCs also have properties that may potentially benefit patients undergoing liver transplantation. This article aims to provide an overview of the current understanding of the immunomodulation achieved by the application of MSCs in liver transplantation, to discuss the problems that may be encountered when using MSCs in clinical practice, and to describe some of the underlying capabilities of MSCs in liver transplantation. Cell–cell contact, soluble molecules, and exosomes have been suggested to be critical approaches to MSCs’ immunoregulation in vitro; however, the exact mechanism, especially in vivo, is still unclear. In recent years, the clinical safety of MSCs has been proven by a series of clinical trials. The obstacles to the clinical application of MSCs are decreasing, but large sample clinical trials involving MSCs are still needed to further study their clinical effects.


2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Hong Kyung Lee ◽  
Eun Young Kim ◽  
Hyung Sook Kim ◽  
Eun Jae Park ◽  
Hye Jin Lee ◽  
...  

Systemic lupus erythematosus (SLE) is an autoimmune disease, which is characterized by hyperactivation of T and B cells. Human mesenchymal stem cells (hMSCs) ameliorate the progression of SLE in preclinical studies using lupus-prone MRL.Faslpr mice. However, whether hMSCs inhibit the functions of xenogeneic mouse T and B cells is not clear. To address this issue, we examined the in vitro effects of hMSCs on T and B cells isolated from MRL.Faslpr mice. Naïve hMSCs inhibited the functions of T cells but not B cells. hMSCs preconditioned with IFN-γ (i) inhibited the proliferation of and IgM production by B cells, (ii) attracted B cells for cell–cell interactions in a CXCL10-dependent manner, and (iii) inhibited B cells by producing indoleamine 2,3-dioxygenase. In summary, our data demonstrate that hMSCs exert therapeutic activity in mice in three steps: first, naïve hMSCs inhibit the functions of T cells, hMSCs are then activated by IFN-γ, and finally, they inhibit B cells.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2326-2326
Author(s):  
Evangelia Yannaki ◽  
Anastasia Papadopoulou ◽  
Minas Yiangou ◽  
Evangelia Athanasiou ◽  
Argyrw Paraskeva ◽  
...  

Abstract The recently recognized potential of mesenchymal stem cells (MSCs) to differentiate into a broad spectrum of tissues and to act as immune regulators beyond the barriers of embryonic germ layers and major histocombatibility comlex (MHC) restriction, has emerged intense research interest on their possible use in a broad spectrum of clinical entities. Although the immunoregulatory potential of MSCs has been shown to effectively control GvHD in several preclinical and clinical studies, their role in autoimmune diseases has not been extensively explored in animal models. The goal of this study was to investigate the in vitro effect of rat bone marrow-derived MSCs on cultured fibrobIast-like synoviocytes (FLS) and T-cells from the spleen after induction of adjuvant arthritis (AA) by FCA as well as their in vivo effect in a rat model of AA resembling human rheumatoid arthritis. MSCs were isolated from bone marrow and were characterized by CD45 negativity and CD54, CD29 positivity in FCM analysis. Differentiation assays were performed to confirm their adipogenic, osteogenic and chondrogenic potential. Culture of AA-FLS in the presence of supernatant from syngeneic (syng) or allogeneic (allo) MSCs at passage 2–3, reduced the AA-FLS (p<0.022) and the ConA-stimulated AA-T-cell (p=0.04) proliferation in a dose-dependent manner, as compared to AA-FLS or AA-T-cell proliferation in the absence of supernatant. Cell-to-cell contact by coculture of activated T-cells with syng or allo MSCs produced a stronger inhibition over the supernatant (p<0.0001), in all tested MSCs dilutions and even at the lowest MSCs :T-cell ratio of 0.05:1. The inhibitory effect of allo as compared to syng MSCs in activated AA T-cells, was stronger both by secreted agents (p=0.017) or by cell to cell contact (p=0.0001). In vivo, low doses of syng MSCs (0.5-5x10^5cell/recipient) administered iv, intrasplenic or intrabone marrow, at single or multiple infusions, didn’t significantly reduce the disease score of MSC-treated as compared to control rats. In contrast, repeated, higher dose (6x10^6cell/recipient), iv infusions of syng or allogeneic MSCs from male donors (Y+MSCs) to female recipients, before the onset of AA (d4 and d9 post AA induction) resulted in significantly lower arthritic scores when compared to control animals. MSC-treated animals preserved a rather normal joint architecture with focal synovial hyperplasia, limited pannus formation and without bone destruction or chondroplasia. In contrast, the joints of arthritic control rats, appeared with a thickened synovial membrane, erosive pannus and dense inflammatory cell infiltration, chondroplasia and osteoplasia. Reduced presence of CD3+, CD11b+, NF-kb+ cells and less intense angiogenesis (FVIII+cells) was demonstrated by immunohistochemistry in the synovium of transplanted rats as compared to the control group. No Y+MSCs were detected in the spleen, bone marrow or in cultured FLS from the synovial membrane at day30 post AA induction, by PCR (sry gene), immunohistochemistry (sry protein) or FISH (Y chromosome), suggesting that the observed benefit was mostly a result of immunomodulation not derived by MSCs homing to target tissues, or migration of MSCs to target tissues may have occured earlier. On the other hand, when the same cell dose was injected after the onset of arthritis (d13 and d20 post AA induction) no clinical benefit could be observed. Our data suggest that MSCs may represent a new therapeutic approach for autoimmune arthritis, however, due to dose and timing limitations in their use, further studies are needed to clinically exploit this potential.


2018 ◽  
Vol 46 (6) ◽  
pp. 2624-2635 ◽  
Author(s):  
Bo Tang ◽  
Xue Li ◽  
Yuanlin Liu ◽  
Xiuhui Chen ◽  
Ximei Li ◽  
...  

Background/Aims: Mesenchymal stem cells (MSCs) do not readily migrate to appropriate sites, and this creates a major obstacle for their use in the treatment of graft-versus-host disease (GVHD). Intercellular adhesion molecule-1 (ICAM-1) can guide the homing of various immune cells to the proper anatomical location within secondary lymphoid organs (SLOs), which are the major niches for generating immune responses or tolerance. MSCs rarely migrate to SLOs after intravenous infusion, and are constitutively low expression of ICAM-1. So in our previous work, ICAM-1 was engineered into a murine MSC line C3H10T1/2 by retrovirus transfection system (ICAM-1MSCs). Here, we hypothesized that ICAM-1highMSCs may significantly improve their immunomodulatory effect. Methods: We used different co-culture methods combined with real-time PCR and flow cytometry to evaluate ICAM-1highMSCs immunomodulatory effect on dendritic cells (DCs) and T cells in vitro and in vivo. MSCs were labeled with carboxyfluorescein diacetate succinimidylester (CFSE) to detect its distribution in mouse model. Results: Our in vitro analyses revealed ICAM-1 MSCs could suppress DCs maturation according to co-culture methods and suppress the T cell immune response according to the mixed lymphocyte response (MLR) and lymphoblast transformation test (LTT) tests. We found that infusion of ICAM-1highMSCs potently prolonged the survival of GVHD mouse model. The infused ICAM-1highMSCs migrate to SLOs in vivo, and suppressed DCs maturation, suppressed CD4+ T cell differentiation to Th1 cells, and increased the ratios of Treg cells. Conclusions: Taken together, these data demonstrate that ICAM-1highMSCs had an enhanced immunosuppressive effect on DCs and T cells, which may help explain the protective effect in a GVHD model. This exciting therapeutic strategy may improve the clinical efficacy of MSC-based therapy for GVHD.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1287-1287
Author(s):  
Kirsten J. Beggs ◽  
Elisabeth H. Javazon ◽  
Jessica C. Tebbets ◽  
Alan W. Flake

Abstract The immunologic properties of Mesenchymal Stem Cells (MSCs) are of therapeutic interest. Previous work shows MSCs do not elicit an alloreactive T cell response in vitro due to suppressive mechanisms. These results suggested that allogeneic MSCs could be used in vivo without inducing an immune response. We further explored this hypothesis using a well-characterized population of Adult Murine Mesenchymal Stem Cells (AmMSCs). These AmMSC are free of hematopoietic contamination and have the following immunologic phenotype: Class I +, Class II-, CD40-, CD80+, and CD86−. Upon treatment with interferon gamma, upregulation of Class I, Class II, CD80, and CD86 was observed. T cell proliferation assays were performed using CFDA SE tracking dye and T cell subsets were analyzed using dual color flow cytometry. The AmMSCs were capable of suppressing CD4+ and CD8+ T cell proliferation in response to alloantigen or polyclonal mitogen, independent of MHC matching, when cultured in direct contact with the T cells. Intracellular cytokine staining of CD4+ and CD8+ T cells revealed that interferon gamma and IL-10 production increased in co-cultures with AmMSC. This suggested that AmMSC are recognized by T cells, but are suppressing proliferation due to other mechanisms in vitro. In order to determine if donor AmMSCs are detected by an immunocompetent host in vivo, we conducted the following study. C57/B6 mice received an intraperitoneal injection of either one million C57/B6 GFP AmMSC (congenic, n=5), one million Balb/c AmMSC (allogeneic, n=5) , or 5 million Balb/c splenocytes (allogeneic control, n=5) at time point zero, and then were given an additional injection of the same cells at 4 weeks. Mice were bled at 0,2,4,5, and 6 weeks after the first injection. Serum was collected and assayed for alloantibody production. Alloantibody results revealed IgG and IgM responses against donor alloantigen at titers of greater than 1:100 in all 5 animals injected with Balb/c MSC. This was significantly increased over titers observed in the 5 mice injected with congenic C57/B6 GFP AmMSC, and similar to the titers seen in mice injected with allogeneic splenocytes. At six weeks post injection animals were sacrificed and a mixed lymphocyte reaction (MLR) was performed using host splenocytes as responders and irradiated donor splenocytes as stimulators. Splenocytes were stained using CFDA SE tracking dye and stained for CD4+ and CD8+ positive T cells. Proliferation of CD4+ and CD8+ T cells was measured using dual color flow cytometry. No increase in proliferation compared to a naïve MLR was noted in the animals injected with the congenic C57/B6 GFP AmMSCs. However all animals injected with allogeneic AmMSCs or allogeneic splenocytes showed increased CD4+ and CD8+ proliferation. It has been suggested primarily based on their in vitro properties, that MSCs may evade the immune response or induce tolerance after allogeneic transplantation. Our results document immune recognition of AmMSCs in vitro and in vivo. While we observed suppression of T-cell proliferation in vitro, our results after in vivo administration support a specific allogeneic immunization response equivalent to that induced by allogeneic splenocytes. These results argue against the theory that allogeneic MSCs may induce tolerance after transplantation, or that they can escape immune surveillance.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 3098-3098 ◽  
Author(s):  
Rosa M. Yañez ◽  
Maria L. Lamana ◽  
Javier Garcia-Castro ◽  
Manuel Ramirez ◽  
Juan A. Bueren

Abstract Previous studies have shown the immunomodulatory properties of bone marrow mesenchymal stem cells (BM-MSCs), opening the possibility of using these cells for the treatment of graft-versus-host disease (GVHD) in patients transplanted with allogeneic hematopoietic grafts. Additionally, Phase I studies in patients with Crohn’s disease suggested the efficacy of adipose tissue-derived mesenchymal stem cells (Ad-MSCs) for the healing of Crohn’s fistulas. In the present study we have investigated in vitro and in vivo, the immunomodulatory effects of Ad-MSCs, compared to BM-MSCs. We observed that both BM-MSCs and Ad-MSCs were negative for CD34, CD45, CD14, CD31 and MHC class I expression, while positive for CD29, CD44, CD90 and CD105. When studying the immunomodulatory effects of these cells in vitro, we found that - as happened with BM-MSCs - Ad-MSCs did not induce proliferation of allogeneic lymphocytes and were not lysed by cytotoxic T cells or alloreactive natural killer cells, indicating that Ad-MSCs are non-immunogenic. Additionally, the presence of Ad-MSCs inhibited in a dose-dependent fashion, both the mixed lymphocyte reaction (MLR) and the T cell proliferation induced by mitogens. To determine whether cell-to-cell contact between Ad-MSCs and PBMNCs was required for immunosuppression, transwell experiments were conducted. Phytohaemagglutinin (PHA)-stimulated lymphocytes were cultured in the upper chamber of a transwell, while irradiated Ad-MSCs remained in the lower chamber. As observed with BM-MSCs, Ad-MSCs were also capable of suppressing the lymphocytes proliferation in this transwell assay. When conditioned medium from Ad-MSCs was added to the MLR, the immuno-suppressive effect persisted, although at a lower level than that observed in a cell-to-cell contact system. Next we studied whether our in vitro findings were of significance in an in vivo mouse model of haploidentical transplantation. In these experiments irradiated F1(C57Bl/Balbc) recipient mice received 1x107 bone marrow cells from C57Bl mice, together with 2x107 splenocytes from the donor, to induce GVHD. One cohort of recipient mice received additional i.v. infusions of 5x105 mouse Ad-MSCs, administered at periodic intervals for up to 28 days post-transplant. When compared to the control group, the severity of the GVHD was significantly reduced in mice receiving Ad-MSCs. Our results suggest that Ad-MSCs obtained from adipose tissue may constitute a new and readily available source of immunomodulatory cells for the prophylaxis and/or treatment of GVHD in patients transplanted with allogeneic grafts.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2286-2286
Author(s):  
Haiping He ◽  
Tokiko Nagamura-Inoue ◽  
Yuki Yamamoto ◽  
Mori Yuka ◽  
Hajime Tsunoda ◽  
...  

Abstract Background Human mesenchymal stem cells (MSCs) can be isolated from several tissues, including bone marrow, umbilical cord, placenta, and adipose tissue. In recent reports, MSCs have the ability to migrate to inflammatory tissues and suppress adverse immune reactions. In fact, BM-derived MSCs are already applied for the patients with acute graft versus host disease (aGVHD) with promising efficacy. To avoid the potential risk of BM harvest, we intended to use umbilical cord Wharton's jelly (WJ) as an alternative source for MSCs. However, the mechanism by which WJ-MSCs exert their immunosuppressive effects is not completely understood. Methods Umbilical cord was collected after informed consent from the mother. WJ-MSCs were isolated by explants method and in part, continued to culture for further expansion by passage 3 (P3). Otherwise, the remaining WJ-MSCs were cryopreserved in P1. Mixed lymphocyte reaction (MLR) was performed on WJ-MSCs or control. CFSE-labeled human peripheral blood (PB) or cord blood(CB) derived mononuclear cells (MNCs) as the responder cells were mixed with irradiated (50Gy) human CB-derived dendrite cells as stimulator cells at a 10:1 ratio, in the presence of low-dose anti-CD3 antibody. CFSE-labeled responder cells were analyzed by FACS. Results WJ-MSCs corresponded to the minimal criteria of MSCs defined by the International Society for Cell Therapy. Thus WJ-MSCs showed spindle-shaped plastic adherent with positive CD105, CD73, CD90, and negative for CD45, CD34, CD14, CD19, and HLA -DR. They also have the ability of differentiation into osteoblasts, adipocytes, chondroblasts in vitro. In MLR, WJ-MSCs efficiently inhibited the responder T cells derived from the same donor of WJ-MSCs, triggered by autologous or allogeneic dendritic cells (DC)(Autologous and allogeneic MLR). Moreover, 3rd party-derived WJ-MSCs also strongly suppressed allogeneic responder T cells proliferation triggered by allogeneic DC(3rd party MLR) (n=3). And WJ-MSCs could inhibit the proliferation of T cells upon PHA-L stimulation. In order to study whether the inhibitory effects of WJ-MSCs on MLR requires the cell to cell contact or not, we next performed MLR on separated WJ-MSCs using by transwell. We observed the inhibitory effect of WJ-MSCs on MLR, although the immunosuppressive effect of the separated WJ-MSCs was much less than that of cell to cell contact. The supernatants of WJ-MSCs, which were derived from 80% confluent WJ-MSCs indicated the mild inhibitory effects on MLR. These inhibitory effects of the WJ-MSCs on MLR were reversed by the addition of indoleamine 2,3-dioxygenase (IDO) inhibitor, 1-methyltryptophan (1-MT) , in a dose dependent manner as shown in Figure 1 (n=3). We also studied the influence of the passage of WJ-MSCs on MLR. The inhibitory effect was comparable among the early passage (∼P3) and long-term passage (P10), although the long-term passage did not show the osteogenic differentiation. Discussion We demonstrated WJ-MSCs have the immunosuppressive effect on the stimulated T cells in autologous, allogeneic and 3rd party MLR setting, indicating that immunosuppressive effect of WJ-MSCs is not restricted by MHC. Soluble IDO may play an important role in immunosuppressive effect of MLR, although we need to detect the additional factors especially in cell to cell contact between WJ-MSCs and the responder T cells. Conclusively, WJ-MSCs may be a feasible alternative non-invasive source of BM-MSCs for GVHD treatment and WJ-MSCs banking can be considered for the prompt clinical applications. Disclosures: No relevant conflicts of interest to declare


2020 ◽  
Author(s):  
Dongdong Wang ◽  
Yi Fu ◽  
Junfen Fan ◽  
Chao Li ◽  
Yi Xu ◽  
...  

Abstract Background: Given their low immunogenicity and multiple differentiation capacities, mesenchymal stem cells (MSCs) have the potential to be used for “off-the-shelf” cell therapy. However, MSC allorejection indicates that they are not fully immune privileged. In this study, we investigated the immunogenicity of human adipose-derived MSCs (Ad-MSCs) and identified potential immunogenic molecules.Methods: To evaluate the immunogenicity of human Ad-MSCs in vivo, cells were transplanted into humanized mice (hu-mice), and T cell infiltration and clearance of human Ad-MSCs were observed by immunofluorescence and bioluminescence imaging. One-way mixed lymphocyte reaction (MLR) and flow cytometry were performed to evaluate the immunogenicity of human Ad-MSCs in vitro. High-throughput TCR repertoire sequencing and mass spectrometry were applied to identify potential immunogenic molecules.Results: Allogeneic human Ad-MSCs recruited T cells during transplantation and caused faster clearance in hu-mice than NOD/ShiLtJGpt-Prkdcem26Cd52Il2rgem26Cd22/Gpt (NSG) mice. The proliferation and activation of T cells was significantly enhanced by human Ad-MSCs, and the expression level of HLA-II on human Ad-MSCs was dramatically increased after coculture with human peripheral blood mononuclear cells (PBMCs) in vitro. In addition, upregulated expression of alpha-enolase (ENO1) on the surface of human Ad-MSCs increased their immunogenicity, and ENO1 inhibitor treatment decreased the human Ad-MSC triggered proliferation of T cells in vitro.Conclusions: We further confirmed the immunogenicity of human Ad-MSCs during allogeneic transplantation and provided a potential target, ENO1, for the safe clinical application of allogeneic human Ad-MSC therapy.


Sign in / Sign up

Export Citation Format

Share Document