Mesenchymal Stem Cells and Fibroblasts Have Similar Immunoregulatory Properties In Vitro but Distinct Gene Expression Profiles: Implications for Cellular Therapy.

Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1930-1930 ◽  
Author(s):  
Muzlifah A. Haniffa ◽  
Xiao N. Wang ◽  
Udo Holtick ◽  
Daniel C. Swan ◽  
Sarah Bullock ◽  
...  

Abstract Bone marrow mesenchymal stem cells (MSC) have potent immunosuppressive properties and are being evaluated in human trials of graft versus host disease (GVHD). The nature of their suppressive capacity is not well understood but attributed to their stem cell function. Evidence that adult stromal cells such as fibroblasts (Fb) also modulate T cell functions has important implications for immunoregulation and cellular therapy. We have investigated the phenomenon of MSC-mediated immunosuppression by comparing MSC with Fb of different origins in in vitro assays of T cell suppression and modulation. We have then isolated RNA from paired samples of dermal Fb and MSC from 6 healthy volunteers for comparative gene expression studies. Adherent Fb were isolated from digested dermis, synovium and lung. MSC were obtained from BM aspirate. Fb from the dermis, lung and synovium possess potent immunomodulatory properties. Fb suppress allogeneic T cell activation by autologously derived cutaneous antigen presenting cells and other stimulators. Fb-mediated suppression through soluble factors is dependent on IFNγ from activated T cells. IFNγ induces indoleamine-2, 3, dioxygenase in Fb with accelerated tryptophan metabolism being partly responsible for suppressing T cell proliferation. T cell suppression is reversible and exposure to stromal cells during activation reprogrammes T cells, increasing secretion of interleukin-4 2.3 fold, interleukin-10 4.3 fold and interleukin-13 15 fold (means of 4 experiments) upon restimulation. Increased Th2 polarization by stromal cells is associated with amelioration of pathological changes in an in vitro human GVHD model. Our findings also show that Fb from different sources are indistinguishable from MSC with respect to morphology, phenotype, growth and differentiation capacity in vitro. Clonogenicity (ratio of CFU to CD73+CD45- cells) of Fb and MSC are similar (range 0.2 to 0.46 CFU/cell) proving that the immunosuppressive effects of Fb from adult tissues are not due to the expansion of rare ‘stem’ cells. Using paired isolates of dermal Fb and MSC to control for inter-individual variation, we were able to define consistent differences in gene expression. Microarray assays were performed using a Human Genome Focus Affymetrix array and analysed with GeneSpring GX. 143 of 9600 probesets showed reproducible differences in transcript levels between dermal Fb and MSC. Probesets upregulated in MSC include genes encoding immunomodulatory mediators: vascular endothelial growth factor (7 fold), hydroxysteroid 17β dehydrogenase (10 fold) and jagged1 (5 fold); extracellular adhesion molecules: proteoglycan1 (264 fold), vascular cell adhesion molecule (175 fold), transglutaminase (67 fold) and procollagen (8 fold); and developmental regulators in the Hedgehog and Wnt signalling pathways. Our findings are further evidence that immunosuppression is a generic property of Fb isolated from several sources and not restricted to MSC. We have for the first time identified a differential expression profile of MSC compared with Fb. These differences may not confer unique in vivo immunosuppressive properties and the potential of Fb as an alternative source of cellular therapy remains untested.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2156-2156
Author(s):  
Michael Gutknecht ◽  
Simone Joas ◽  
Lisa Güttler ◽  
Lothar Kanz ◽  
Helmut R Salih ◽  
...  

Abstract Abstract 2156 Multiple approaches for treatment of malignant disease presently aim to combine targeted therapy with tyrosine kinase inhibitors (TKI) with immunotherapy. Dendritic cells (DC) are frequently used in such strategies due to their unique ability to initiate potent T cell anti-tumor immunity. Unfortunately, DC may also activate suppressive CD25+FOXP3+ regulatory T cells (Treg), which depends on the stimuli that influence DC in immature state and/or during development from precursor cells. High frequencies of Treg have been described in several types of tumors within the tumor microenvironment, which is associated with poor prognosis and reduced survival. DC development and function are moreover governed by various tyrosine kinases of which some are also inhibited by clinically used TKI. TKI thus may cause immunoinhibitory side effects, and we previously demonstrated that exposure of monocyte-derived DC to the BCR-ABL inhibitor imatinib causes up-regulation of the immunosuppressive type I transmembrane glycoprotein osteoactivin (GPNMB, DC-HIL) and reduces expression of activating surface antigens as well as T cell-stimulatory capacity of DC in vitro (Schwarzbich et al., 2012). Other investigators reported that imatinib induces functionally Treg in CML patients, but the underlying mechanisms are so far unknown. (Bachy et al., 2011). On the other hand, TKI may inhibit proliferation and suppressive capacity of regulatory T cells in vitro (Chen et al., 2007). Here we tried to solve this apparent discrepancy by analyzing the influence of TKI on DC-Treg interaction. Monocyte-derived DC (moDC) were generated over 7 days by exposing blood monocytes to GM-CSF and IL-4. TNF was added on day 6 of culture in case of maturation, and imatinib or nilotinib (3μM each) were added to the culture medium every second day starting from the first day of culture. Induction and functionality of Treg was determined by FACS and so called effector T cell suppression assays upon culture of moDC with autologous PBMC. We found that exposure of moDC to imatinib or nilotinib only slightly increased the frequency of Treg as compared to controls. However, these Treg strongly inhibited autologous T cell proliferation as assessed by T cell suppression assays. This was mediated by direct cellular interaction, as culture supernatants of TKI-treated DC did not alter Treg function and also did not contain elevated levels of the immunosuppressive (and Treg inducing) cytokines TGF-β and IL-10. Thus, our data indicate that the seemingly contradictory results of the in vivo and in vitro studies described above may be explained by the effects caused by exposure of moDC to BCR-ABL TKI which results in the induction of functionally active Treg. These findings are of special importance for future combinatory approaches using TKI and DC-based immunotherapy. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 3079-3079
Author(s):  
Rachel A. Burga ◽  
Mitchell Thorn ◽  
Cang T. Nguyen ◽  
Lauren Licata ◽  
N. Joseph Espat ◽  
...  

3079 Background: Immunotherapy for colorectal cancer liver metastases (CRCLM) is limited by the intrahepatic immunosuppressive environment mediated in part by myeloid derived suppressor cells (MDSC), which expand in response to tumor. T cell suppression can be mediated by programmed death ligand-1 (PD-L1, CD274) on MDSC binding to programmed death-1 (PD-1, CD279) on T cells. We hypothesize blocking PD-L1 will improve adoptive cellular therapy efficacy for CRCLM through inhibition of MDSC-mediated T cell suppression. Methods: “Designer” T cells (dTc) were produced from activated murine splenocytes transduced with chimeric antigen receptor (CAR) specific for CEA. C57BL/6 mice were injected with CEA+ MC38 tumor cells via spleen, and liver MDSC (CD11b+Gr1+) were purified with immunomagnetic beads after two weeks. MDSC were co-cultured with stimulated dTc with or without in vitro PD-L1 blockade. Results: MDSC expanded 2.4-fold in response to CRCLM, and expressed high levels of PD-L1 (63.8% PD-L1+). PD-L1 was equally expressed on both monocytic (CD11b+Ly6G-Ly6C+) and granulocytic (CD11b+Ly6G+) MDSC subsets (43.6% PD-L1+ and 27.9% PD-L1+, respectively). Expression of related ligand, PD-L2 was found to be negligible in both subsets. The cognate inhibitory receptor, PD-1, was expressed on dTc (23.8% PD-1+) and native T cells (37.3% PD-1+). Increasing endogenous T cell expression of PD-1 significantly correlated with MDSC expansion (r=0.9774, p<0.0001) in response to CRCLM. Co-culture of dTc with MDSC demonstrated the suppressive effect of MDSC on dTc proliferation which was abrogated with in vitro targeting of PD-L1. The percentage of dTc proliferating in the presence of CEA+ tumor decreased from 72.2% to 29.3% (p<0.001) with the addition of MDSC, and immunosuppression was reversed with blockade of PD-L1, which resulted in a 1.6-fold increase in dTc proliferation (p=0.01 ). Conclusions: Liver MDSC expand in the presence of CRCLM and mediate suppression of anti-CEA dTc via PD-L1. Our results indicate that blockade of PD-L1:PD-1 engagement is a viable strategy for enhancing the efficacy of adoptive cell therapy for liver metastases.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Min Cao ◽  
Huihui Liu ◽  
Yujun Dong ◽  
Wei Liu ◽  
Zhengyu Yu ◽  
...  

Abstract Background Idiopathic pneumonia syndrome (IPS) is a non-infectious fatal complication characterized by a massive infiltration of leukocytes in lungs and diffuse pulmonary injury after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Conventional immunosuppressive treatments for IPS have poor therapeutic effects. Safe and effective treatments are not yet available and under explorations. Our previous study demonstrated that mesenchymal stem cells (MSCs) can alleviate IPS, but the mechanisms remain unclear. Methods Co-cultured pre-activated T cells and MSCs in vitro to observe the changes in the CCR2-CCL2 axis. By establishing an IPS mouse model and administering MSCs to further verify the results of in vitro experiments. Results Co-culture of pre-activated T cells with MSCs in vitro modulated the CCR2-CCL2 axis, resulting in quiescent T cells and polarization toward CCR2+CD4+ T cell subsets. Blocking CCR2-CCL2 interaction abolished the immunoregulatory effect of MSCs, leading to re-activation of T cells and partial reversion of polarizing toward CCR2+CD4+ T cells. In IPS mouse model, application of MSCs prolonged the survival and reduced the pathological damage and T cell infiltration into lung tissue. Activation of CCR2-CCL2 axis and production of CCR2+CD4+ T cells were observed in the lungs treated with MSCs. The prophylactic effect of MSCs on IPS was significantly attenuated by the administration of CCR2 or CCL2 antagonist in MSC-treated mice. Conclusions We demonstrated an important role of CCR2-CCL2 axis in modulating T cell function which is one of the mechanisms of the prophylactic effect of MSCs on IPS.


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.


2005 ◽  
Vol 33 (7) ◽  
pp. 819-827 ◽  
Author(s):  
Andrea Bacigalupo ◽  
Marisa Valle ◽  
Marina Podestà ◽  
Anna Pitto ◽  
Elena Zocchi ◽  
...  

2019 ◽  
Author(s):  
Alexandra A. Vita ◽  
Hend Aljobaily ◽  
David O. Lyons ◽  
Nicholas A. Pullen

ABSTRACTPrevious evidence suggests that berberine (BBR), a clinically relevant plant-derived alkaloid, alleviates symptoms of clinically apparent collagen induced arthritis (CIA), and may have a prophylactic role from in vitro studies. Thus, we used a CIA model to determine if BBR merits further exploration as a prophylactic treatment for rheumatoid arthritis. Mice were treated with either 1 mg/kg/day of BBR or a vehicle (PBS) control via IP injections from day 0 to day 28, were left untreated (CIA control), or were in a non-arthritic control group. Incidence of arthritis in BBR mice was 40%, compared to 90% in the CIA and 80% in the PBS controls. Populations of B cells and T cells from the spleens and draining lymph nodes were examined from mice across treatment groups on day 14 and from the remaining mice on day 28 when arthritic signs and symptoms were expected to be apparent. BBR-treated mice had significantly reduced populations of CD4+ T cells, CXCR5+ Tfhcells, and an increased proportion of Treg at both day 14 and day 28 endpoints, as well as decreased CD28+ and CD154+ CD4+ T cells at day 14. BBR-treated mice also experienced a significant reduction of CD19+ B cells in LNs at day 28. Additionally, BBR treatment resulted in significantly lower anti-collagen type II-specific (anti-CII) IgG2a and anti-CII total IgG serum concentrations. These results indicate a potential role for BBR as a prophylactic supplement, and that its effect may be mediated through T cell suppression, which indirectly affects B cell activity.


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.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Ashton C. Trotman-Grant ◽  
Mahmood Mohtashami ◽  
Joshua De Sousa Casal ◽  
Elisa C. Martinez ◽  
Dylan Lee ◽  
...  

AbstractT cells are pivotal effectors of the immune system and can be harnessed as therapeutics for regenerative medicine and cancer immunotherapy. An unmet challenge in the field is the development of a clinically relevant system that is readily scalable to generate large numbers of T-lineage cells from hematopoietic stem/progenitor cells (HSPCs). Here, we report a stromal cell-free, microbead-based approach that supports the efficient in vitro development of both human progenitor T (proT) cells and T-lineage cells from CD34+cells sourced from cord blood, GCSF-mobilized peripheral blood, and pluripotent stem cells (PSCs). DL4-μbeads, along with lymphopoietic cytokines, induce an ordered sequence of differentiation from CD34+ cells to CD34+CD7+CD5+ proT cells to CD3+αβ T cells. Single-cell RNA sequencing of human PSC-derived proT cells reveals a transcriptional profile similar to the earliest thymocytes found in the embryonic and fetal thymus. Furthermore, the adoptive transfer of CD34+CD7+ proT cells into immunodeficient mice demonstrates efficient thymic engraftment and functional maturation of peripheral T cells. DL4-μbeads provide a simple and robust platform to both study human T cell development and facilitate the development of engineered T cell therapies from renewable sources.


2021 ◽  
Vol 12 (3) ◽  
Author(s):  
Qiuli Liu ◽  
Xiaoyong Chen ◽  
Chang Liu ◽  
Lijie Pan ◽  
Xinmei Kang ◽  
...  

AbstractLiver diseases with different pathogenesis share common pathways of immune-mediated injury. Chitinase-3-like protein 1 (CHI3L1) was induced in both acute and chronic liver injuries, and recent studies reported that it possesses an immunosuppressive ability. CHI3L1 was also expressed in mesenchymal stem cells (MSCs), thus we investigates the role of CHI3L1 in MSC-based therapy for immune-mediated liver injury here. We found that CHI3L1 was highly expressed in human umbilical cord MSCs (hUC-MSCs). Downregulating CHI3L1 mitigated the ability of hUC-MSCs to inhibit T cell activation, proliferation and inflammatory cytokine secretion in vitro. Using Concanavalin A (Con A)-induced liver injury mouse model, we found that silencing CHI3L1 significantly abrogated the hUC-MSCs-mediated alleviation of liver injury, accompanying by weakened suppressive effects on infiltration and activation of hepatic T cells, and secretion of pro-inflammatory cytokines. In addition, recombinant CHI3L1 (rCHI3L1) administration inhibited the proliferation and function of activated T cells, and alleviated the Con A-induced liver injury in mice. Mechanistically, gene set enrichment analysis showed that JAK/STAT signalling pathway was one of the most significantly enriched gene pathways in T cells co-cultured with hUC-MSCs with CHI3L1 knockdown, and further study revealed that CHI3L1 secreted by hUC-MSCs inhibited the STAT1/3 signalling in T cells by upregulating peroxisome proliferator-activated receptor δ (PPARδ). Collectively, our data showed that CHI3L1 was a novel MSC-secreted immunosuppressive factor and provided new insights into therapeutic treatment of immune-mediated liver injury.


Sign in / Sign up

Export Citation Format

Share Document