scholarly journals Human Adipose Tissue-Derived Mesenchymal Stromal Cells Inhibit CD4+ T Cell Proliferation and Induce Regulatory T Cells as Well as CD127 Expression on CD4+CD25+ T Cells

Cells ◽  
2021 ◽  
Vol 10 (1) ◽  
pp. 58
Author(s):  
Agnese Fiori ◽  
Stefanie Uhlig ◽  
Harald Klüter ◽  
Karen Bieback

Mesenchymal stromal cells (MSC) exert their immunomodulatory potential on several cell types of the immune system, affecting and influencing the immune response. MSC efficiently inhibit T cell proliferation, reduce the secretion of pro-inflammatory cytokines, limit the differentiation of pro-inflammatory Th subtypes and promote the induction of regulatory T cells (Treg). In this study, we analyzed the immunomodulatory potential of human adipose tissue-derived MSC (ASC), on CD4+ T cells, addressing potential cell-contact dependency in relation to T cell receptor stimulation of whole human peripheral blood mononuclear cells (PBMC). ASC were cultured with not stimulated or anti-CD3/CD28-stimulated PBMC in direct and transwell cocultures; PBMC alone were used as controls. After 7 days, cocultures were harvested and we analyzed: (1) the inhibitory potential of ASC on CD4+ cell proliferation and (2) phenotypic changes in CD4+ cells in respect of Treg marker (CD25, CD127 and FoxP3) expression. We confirmed the inhibitory potential of ASC on CD4+ cell proliferation, which occurs upon PBMC stimulation and is mediated by indoleamine 2,3-dioxygenase. Importantly, ASC reduce both pro- and anti-inflammatory cytokine secretion, without indications on specific Th differentiation. We found that stimulation induces CD25 expression on CD4+ cells and that, despite inhibiting overall CD4+ cell proliferation, ASC can specifically induce the proliferation of CD4+CD25+ cells. We observed that ASC induce Treg (CD4+CD25+CD127−FoxP3+) only in not stimulated cocultures and that ASC increase the ratio of CD4+CD25+CD127+FoxP3− cells at the expense of CD4+CD25+CD127−FoxP3− cells. Our study provides new insights on the interplay between ASC and CD4+ T cells, proposing that ASC-dependent induction of Treg depends on PBMC activation which affects the balance between the different subpopulations of CD4+CD25+ cells expressing CD127 and/or FoxP3.

1990 ◽  
Vol 172 (4) ◽  
pp. 1185-1192 ◽  
Author(s):  
Y Nojima ◽  
M J Humphries ◽  
A P Mould ◽  
A Komoriya ◽  
K M Yamada ◽  
...  

We previously showed that fibronectin (FN) synergized with anti-CD3 in induction of CD4+ T cell proliferation, and that VLA-5 acted as a functional FN receptor in a serum-free culture system. In the present study, we showed that VLA-4 is also involved in this CD3-dependent CD4 cell activation through its interaction with the alternatively spliced CS1 domain of FN. When highly purified CD4 cells were cultured on plates coated with anti-CD3 plus synthetic CS1 peptide-IgG conjugate, significant proliferation could be observed. Neither CS1 alone nor anti-CD3 alone induced this activation. This proliferation was completely blocked by anti-VLA beta 1 (4B4) and anti-VLA-4 (8F2), while anti-VLA-5 (monoclonal antibody [mAb] 16 and 2H6) had no effect. These data indicate that VLA-4 mediates CD3-dependent CD4 cell proliferation via the CS1 domain of FN. Anti-VLA-4 also partially (10-40%) inhibited CD4 cell proliferation induced by native FN plus anti-CD3, implying that the CS1 domain is active in the native plasma FN. However, this native FN-dependent proliferation was entirely abolished by addition of anti-VLA-5 alone. Moreover, when native FN-coated plates were pretreated with anti-FN (mAb 333), which blocks RGDS sites but not CS1 sites, no CD4 cell activation could be observed. These results strongly suggest that CD4 cell activation induced by plasma FN/anti-CD3 may be dependent on both VLA4/CS1 and VLA5/RGDS interactions, although the latter interaction may be required for function of the former.


2016 ◽  
Vol 2016 ◽  
pp. 1-17 ◽  
Author(s):  
Sebastien Hagmann ◽  
Claudia Rimmele ◽  
Florin Bucur ◽  
Thomas Dreher ◽  
Felix Zeifang ◽  
...  

Introduction. The participation of an inflammatory joint milieu has been described in osteoarthritis (OA) pathogenesis. Mesenchymal stromal cells (MSCs) play an important role in modulating inflammatory processes. Based on previous studies in an allogeneic T-cell coculture model, we aimed at further determining the role of synovial MSCs in OA pathogenesis.Methods. Bone-marrow (BM) and synovial membrane (SM) MSCs from hip joints of late stage OA patients and CD4+ T-cells from healthy donors were analysed regarding surface marker expression before and after coculture. Proliferation upon CD3/CD28 stimulation and cytokine analyses were compared between MSCs.Results. SM-MSCs differed from BM-MSCs in several surface markers and their osteogenic differentiation potential. Cocultures of both MSCs with CD4+ T-cells resulted in recruitment of CD45RA+ FoxP3+ regulatory T-cells. Upon stimulation, only SM-MSCs suppressed CD4+ T-cell proliferation, while both SM-MSCs and BM-MSCs modified cytokine profiles through suppressing IL-2 and TNF-αas well as increasing IL-6 secretion.Conclusions. Synovial MSCs from OA joints are a unique fraction that can be distinguished from their bone-marrow derived counterparts. Their unique ability to suppress CD3/CD28 induced CD4+ T-cell proliferation makes them a potential target for future therapeutic approaches.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4865-4865
Author(s):  
Rodrigo Haddad ◽  
Felipe Saldanha-araujo ◽  
Amélia Araujo ◽  
Dimas T Covas ◽  
Marco A. Zago ◽  
...  

Abstract Abstract 4865 Introduction: Mesenquimal Stromal Cells (MSCs) possess immunosuppressive properties, becoming these cells a promising subject of study for future approaches in cell-based therapy. During co-culture with activated lymphocytes and probably through T-cell derived cytokines, MSCs are activated in order to become suppressive. Once MSCs are triggered, they acquire an inhibitory profile increasing the release of immunoregulatory factors, such as IDO, IFNγ, PGE2, NO and TGF-β, responsible for T-cell inhibition. In addition, expression of adhesion molecules, such as ICAM-1, and generation of regulatory T cells subsets, such as CD69+ T cells, are important in immunosuppressive property of human MSCs. However, the exact mechanisms underlying the immunomodulatory functions of MSCs remain largely unknown. NF-kB comprises a family of inducible transcription factors that serve as important regulators of the host immune and inflammatory responses. The NF-kB signals are activated via canonical (mediated mainly by RelA-p50) and/or non-canonical (mediated by RelB-p52) pathways in response to diverse stimuli. Given the immunomodulatory properties of MSCs and the possible involvement of NF-Bk pathway in this effect, here, it was explored the role of non-canonical NF-KB signaling in the immunomodulatory capacity of MSCs cocultured with activated T cells. Methods: siRNAs targeting RelB were transfected into MSC with lipofectamine. siRNAs C- were used as negative control and siRNA-FITC as transfection control. After 24 hours, immunomagnetically purified CD3+ T cells were stained with CFSE, activated by anti-CD2/CD3/CD28 beads and cultured in the presence of MSCs. After 3 days, flow cytometry was performed to observe the transfection efficiency, T cell proliferation and percentage of CD69+ T regulatory cells. Additionally, RNA from MSCs was extracted and RelB and ICAM-1 mRNA expression were quantified by Real-time PCR. Results: The transfection efficiency was around 75% and RelB mRNA level was reduced by 80% in MSCs transfected with siRNA RelB compared to siRNAs C- transfected cells. Compared to MSCs previously transfected with siRNA C- and co-cultured with activated T cells, MSCs transfected with siRNA RelB resulted increasing of 22% in T cell proliferation and decreasing of 9,2% and 30% in the CD69+ regulatory T cells generation and ICAM-1 expression respectively. Conclusion: Non-canonical NF-kB pathway, mediated by RelB, may be partially involved in acquisition of the inhibitory profile by decreasing T cells proliferation, and increasing the expression of ICAM-1 and the generation of CD69+ regulatory T cells. Disclosures: No relevant conflicts of interest to declare.


2020 ◽  
Vol 9 (4) ◽  
pp. 1052 ◽  
Author(s):  
Ramona Sturm ◽  
Lara Xanthopoulos ◽  
David Heftrig ◽  
Elsie Oppermann ◽  
Teodora Vrdoljak ◽  
...  

Objective: Severely injured patients frequently develop an immunological imbalance following the traumatic insult, which might result in infectious complications evoked by a persisting immunosuppression. Regulatory T cells (Tregs) maintain the immune homeostasis by suppressing proinflammatory responses, however, their functionality after trauma is unclear. Here, we characterized the role of Tregs in regulating the proliferation of CD4+ lymphocytes in traumatized patients (TP). Methods: Peripheral blood was obtained daily from 29 severely injured TP (Injury Severity Score, ISS ≥16) for ten days following admission to the emergency department (ED). Ten healthy volunteers (HV) served as controls. The frequency and activity of Tregs were assessed by flow cytometry. Proliferation of CD4+ cells was analyzed either in presence or absence of Tregs, or after blocking of either IL-10 or IL-10R1. Results: The frequencies of CD4+CD25high and CD4+CD25+CD127− Tregs were significantly decreased immediately upon admission of TP to the ED and during the following 10 post-injury days. Compared with HV CD4+ T cell proliferation in TP increased significantly upon their admission and on the following days. As expected, CD4+CD25+CD127− Tregs reduced the proliferation of CD4+ cells in HV, nevertheless, CD4+ proliferation in TP was increased by Tregs. Neutralization of IL-10 as well as blocking the IL-10R1 increased further CD4+ T cell proliferation in Tregs-depleted cultures, thereby confirming an IL-10-mediated mechanism of IL-10-regulated CD4+ T cell proliferation. Neutralization of IL-10 in TP decreased CD4+ T cell proliferation in Tregs-depleted cultures, whereas blocking of the IL-10R1 receptor had no significant effects. Conclusions: The frequency of Tregs in the CD4+ T lymphocyte population is reduced after trauma; however, their inductiveness is increased. The mechanisms of deregulated influence of Tregs on CD4+ T cell proliferation are mediated via IL-10 but not via the IL-10R1.


1993 ◽  
Vol 22 (2-3) ◽  
pp. 80-85
Author(s):  
A. Malley ◽  
N. Pangares ◽  
S.K. Mayo ◽  
M. Zeleny‐Pooley ◽  
J.V. Torres ◽  
...  

1997 ◽  
Vol 186 (10) ◽  
pp. 1787-1791 ◽  
Author(s):  
Pan Zheng ◽  
Yang Liu

It has been proposed that some bystander T cell activation may in fact be due to T cell antigen receptor (TCR) cross-reactivity that is too low to be detected by the effector cytotoxic T lymphocyte (CTL). However, this hypothesis is not supported by direct evidence since no TCR ligand is known to induce T cell proliferation and differentiation without being recognized by the effector CTL. Here we report that transgenic T cells expressing a T cell receptor to influenza virus A/NT/68 nucleoprotein (NP) 366-374:Db complexes clonally expand and become effector CTLs in response to homologous peptides from either A/PR8/34 (H1N1), A/AA/60 (H2N2), or A/NT/68 (H3N2). However, the effector T cells induced by each of the three peptides kill target cells pulsed with NP peptides from the H3N2 and H2N2 viruses, but not from the H1N1 virus. Thus, NP366–374 from influenza virus H1N1 is the first TCR ligand that can induce T cell proliferation and differentiation without being recognized by CTLs. Since induction of T cell proliferation was mediated by antigen-presenting cells that express costimulatory molecules such as B7, we investigated if cytolysis of H1N1 NP peptide–pulsed targets can be restored by expressing B7-1 on the target cells. Our results revealed that this is the case. These data demonstrated that costimulatory molecule B7 modulates antigen specificity of CTLs, and provides a missing link that explains some of the bystander T cell activation.


2005 ◽  
Vol 153 (4) ◽  
pp. 750-757 ◽  
Author(s):  
M. Vukmanovic-Stejic ◽  
A. McQuaid ◽  
K.E. Birch ◽  
J.R. Reed ◽  
C. Macgregor ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document