88 research outputs found

    VALPROIC ACID INDUCES APOPTOSIS AND INCREASES CXCR7 EXPRESSION IN EPITHELIAL OVARIAN CANCER CELL LINE SKOV-3.

    Get PDF
    Background: The chemokine receptor, CXCR7 is described to play a biologically relevant role in tumor growth and spread. Recently, it was reported that CXCR7 overexpression is associated with an unfavorable prognosis and metastatis of epithelial ovarian cancer (EOC). Aware that, several reports indicated that Histone deacetylases (HDACs) regulate the expression and activity of many proteins involved in both cancer initiation and progression, the aim of this work, was to study the effect of the HDAC inhibitor valproic acid (VPA) on the expression of CXCR7 as well as its impact on survival function in the epithelial ovarian cell line (SKOV-3). Methods: cells were cultured with varying concentrations of VPA (1, 2, 3, 4, 5 and 10 mM) for different durations (0, 12 h, 24 h and 48 h). Cell survival was assessed by Neutral red assay and by colony counting which being stained with crystal violet. CXCR7 expression was determined at mRNA level using quantitative real-time PCR (qRT-PCR) or at the protein level using western blotting. Results: VPA reduces cell survival of SKOV-3 cancer cells. The inhibition effect of VPA was dose and time-dependent. Exposure to VPA at concentrations above 2 mM at 24 h resulted in an increase expression of CXCR7 at both the mRNA and protein levels . Conclusion: These observations provide, for the first time, a better insight into the epigenetic mechanisms involved in regulating CXCR7 expression in EOC and will open new avenues for evaluating drugs that specifically stimulate the apoptosis of EOC with minimal unwanted side effect

    VALPROIC ACID INDUCES APOPTOSIS AND INCREASES CXCR7 EXPRESSION IN EPITHELIAL OVARIAN CANCER CELL LINE SKOV-3.

    Get PDF
    Background: The chemokine receptor, CXCR7 is described to play a biologically relevant role in tumor growth and spread. Recently, it was reported that CXCR7 overexpression is associated with an unfavorable prognosis and metastatis of epithelial ovarian cancer (EOC). Aware that, several reports indicated that Histone deacetylases (HDACs) regulate the expression and activity of many proteins involved in both cancer initiation and progression, the aim of this work, was to study the effect of the HDAC inhibitor valproic acid (VPA) on the expression of CXCR7 as well as its impact on survival function in the epithelial ovarian cell line (SKOV-3). Methods: cells were cultured with varying concentrations of VPA (1, 2, 3, 4, 5 and 10 mM) for different durations (0, 12 h, 24 h and 48 h). Cell survival was assessed by Neutral red assay and by colony counting which being stained with crystal violet. CXCR7 expression was determined at mRNA level using quantitative real-time PCR (qRT-PCR) or at the protein level using western blotting. Results: VPA reduces cell survival of SKOV-3 cancer cells. The inhibition effect of VPA was dose and time-dependent. Exposure to VPA at concentrations above 2 mM at 24 h resulted in an increase expression of CXCR7 at both the mRNA and protein levels . Conclusion: These observations provide, for the first time, a better insight into the epigenetic mechanisms involved in regulating CXCR7 expression in EOC and will open new avenues for evaluating drugs that specifically stimulate the apoptosis of EOC with minimal unwanted side effect

    Differentiation associated regulation of microRNA expression in vivo in human CD8+ T cell subsets

    Get PDF
    BACKGROUND: The differentiation of CD8+ T lymphocytes following priming of naïve cells is central in the establishment of the adaptive immune response. Yet, the molecular events underlying this process are not fully understood. MicroRNAs have been recently shown to play a key role in the regulation of haematopoiesis in mouse, but their implication in peripheral lymphocyte differentiation in humans remains largely unknown. METHODS: In order to explore the potential implication of microRNAs in CD8+ T cell differentiation in humans, microRNA expression profiles were analysed using microarrays and quantitative PCR in several human CD8+ T cell subsets defining the major steps of the T cell differentiation pathway. RESULTS: We found expression of a limited set of microRNAs, including the miR-17~92 cluster. Moreover, we reveal the existence of differentiation-associated regulation of specific microRNAs. When compared to naive cells, miR-21 and miR-155 were indeed found upregulated upon differentiation to effector cells, while expression of the miR-17~92 cluster tended to concomitantly decrease. CONCLUSIONS: This study establishes for the first time in a large panel of individuals the existence of differentiation associated regulation of microRNA expression in human CD8+ T lymphocytes in vivo, which is likely to impact on specific cellular functions

    A microRNA profile of human CD8(+) regulatory T cells and characterization of the effects of microRNAs on Treg cell-associated genes.

    Get PDF
    Recently, regulatory T (Treg) cells have gained interest in the fields of immunopathology, transplantation and oncoimmunology. Here, we investigated the microRNA expression profile of human natural CD8(+)CD25(+) Treg cells and the impact of microRNAs on molecules associated with immune regulation. We purified human natural CD8(+) Treg cells and assessed the expression of FOXP3 and CTLA-4 by flow cytometry. We have also tested the ex vivo suppressive capacity of these cells in mixed leukocyte reactions. Using TaqMan low-density arrays and microRNA qPCR for validation, we could identify a microRNA 'signature' for CD8(+)CD25(+)FOXP3(+)CTLA-4(+) natural Treg cells. We used the 'TargetScan' and 'miRBase' bioinformatics programs to identify potential target sites for these microRNAs in the 3'-UTR of important Treg cell-associated genes. The human CD8(+)CD25(+) natural Treg cell microRNA signature includes 10 differentially expressed microRNAs. We demonstrated an impact of this signature on Treg cell biology by showing specific regulation of FOXP3, CTLA-4 and GARP gene expression by microRNA using site-directed mutagenesis and a dual-luciferase reporter assay. Furthermore, we used microRNA transduction experiments to demonstrate that these microRNAs impacted their target genes in human primary Treg cells ex vivo. We are examining the biological relevance of this 'signature' by studying its impact on other important Treg cell-associated genes. These efforts could result in a better understanding of the regulation of Treg cell function and might reveal new targets for immunotherapy in immune disorders and cancer

    Investigation of the molecular mechanisms controlling the function of human natural regulatory T cells

    No full text
    Regulatory T cells (Tregs) are a subpopulation of T cells with immuno-suppressive properties. Tregs play a key role in immune response regulation and tolerance to antigens, thereby preventing autoimmunity, and may be partly responsible for the lack of an appropriate immune response against tumor cells. However, a human microRNA (miR) Treg signature has not been described yet. We investigated human natural Tregs and identified a signature composed of five microRNAs (-21, -31, -125a, -181c and -374). Among those, two were considerably under-expressed (miR-31 and miR-125a). We identified a functional target sequence for miR-31 in the 3’ untranslated region (3’ UTR) of FOXP3 mRNA. Using lentiviral transduction of fresh cord blood T cells, we demonstrated that miR-31 and miR-21 had opposite effects on FOXP3 expression. We showed that miR-31 negatively regulates FOXP3 expression by binding directly to its potential target site in the 3’ UTR of FOXP3 mRNA. We next demonstrated that miR-21 acted as a positive, though indirect, regulator of FOXP3 expression.Recent reports have shown that histone deacetylase inhibitors increased FOXP3 expression in T cells. We therefore decided to investigate in non-Treg CD4-positive cells, the mechanisms by which an aspecific opening of the chromatin could lead to an increased FOXP3 expression. We focused on the binding of potentially activating transcription factors to the promoter region of FOXP3 and on modifications in the five miRs constituting the Treg signature. Valproate treatment induced binding of Ets-1 and Ets-2 transcription factors to the FOXP3 promoter and acted positively on its expression, by increasing the acetylation of histone H4 lysines. Valproate treatment also induced the acquisition of the miRs of Treg signature. To elucidate whether the changes in the miRs expression could be due to the increased FOXP3expression, we transduced these non-Tregs with a FOXP3 lentiviral expression vector, and found no changes in miRs expression. Therefore, the modification in their miR expression profile is not due to an increased expression of FOXP3 but directly results from histone deacetylase inhibition. Rather, the increased FOXP3 expression results from the additive effects of Ets factors binding and the change in the expression level of miR-21 and miR-31. These data, by allowing a better understanding of the molecular phenomena underlying the number and function of Tregs, could open the door to novel therapeutic approaches in cancer immunotherapy and treatment of autoimmune disorders.Doctorat en Sciencesinfo:eu-repo/semantics/nonPublishe

    The potential of mesenchymal stromal cells in immunotherapy.

    No full text
    SCOPUS: re.jinfo:eu-repo/semantics/publishe

    Identification and Evaluation of New Immunoregulatory Genes in Mesenchymal Stromal Cells of Different Origins: Comparison of Normal and Inflammatory Conditions.

    No full text
    BACKGROUND Mesenchymal stromal cells (MSCs) possess potent immunomodulatory properties that increase their value as a cell-based therapeutic tool for managing various immune-based disorders. Over the past years, accumulated results from trials using MSCs-based therapy have shown substantial contradictions. Although the reasons underlying these discrepancies are still not completely understood, it is well known that the immunomodulatory activities mediated by distinct MSCs differ in a manner dependent on their tissue origin and adequate response to inflammation priming. Thus, characterization of new molecular pathway(s) through which distinct MSC populations can exert their immunomodulatory effects, particularly during inflammation, will undoubtedly enhance their therapeutic potential. MATERIAL AND METHODS After confirming their compliance with ISCT criteria, quantitative real time-PCR (qRT-PCR) was used to screen new immunoregulatory genes in MSCs, derived from adipose tissue, foreskin, Wharton's jelly or the bone-marrow, after being cultivated under normal and inflammatory conditions. RESULTS FGL2, GAL, SEMA4D, SEMA7A, and IDO1 genes appeared to be differentially transcribed in the different MSC populations. Moreover, these genes were not similarly modulated following MSCs-exposure to inflammatory signals. CONCLUSIONS Our observations suggest that these identified immunoregulatory genes may be considered as potential candidates to be targeted in order to enhance the immunomodulatory properties of MSCs towards more efficient clinical use.SCOPUS: ar.jinfo:eu-repo/semantics/publishe

    Data on nitric oxide production by human bone marrow-derived mesenchymal stromal cells

    No full text
    Due to its anti-inflammatory and immunosuppressive potential, Nitric oxide (NO), a gaseous radical, is of special importance during graft-versus-host diseases (GVHD) and feoto-maternal tolerance. NO is a major mediator of murine mesenchymal stromal cells (MSCs)-immunosuppressive capacity. In this data article, we characterized NO production by human bone marrow-derived MSCs (hBMSCs). MSCs, isolated from healthy donors (n=5), were defined according to the International Society for cellular Therapy (ISCT) guidelines. Based on a fluorometric detection system, and upon using Nitrite (NO2−)/Nitrate ( NO3−) Assay Kit, the amounts of NO metabolites ( NO2− and NO3−) produced by hBMSCs, being grown in a culture medium either lacking (constitutive condition) or containing IL-4, IL-10 or a pro-inflammatory cytokine cocktail made of IL-1β, TNF-α, IFN-α and IFN-γ, were assessed. All assays were carried out in triplicates and the mean values are reported. The data from this study supports and corroborates the discussion associated with our previously published work entitled “The Immunomodulatory Potential of Mesenchymal Stromal Cells: A Story of a Regulatory Network” (Najar et al., 2016) [1]

    Study of the microRNA expression profile of foreskin derived mesenchymal stromal cells following inflammation priming

    No full text
    Background: Due to their self-renewal capacity, multi-lineage potential, and immunomodulatory properties, mesenchymal stromal cells (MSCs) are an attractive tool for different therapeutic strategies. Foreskin (FSK), considered as a biological waste material, has already been shown to be a valuable source of MSCs. Besides their typical fibroblast like morphology and International Society for cellular Therapy compliant phenotype, foreskin-MSCs (FSK-MSCs) are clonogenic, and highly proliferative cells with multi-lineage and strong immunomodulatory capacities. Of importance, FSK-MSCs properly adjust their fate following exposure to inflammatory signals. Being potent regulators of gene expression, miRNAs are involved in modulating nearly all cellular processes and in orchestrating the roles of different immune cells. In this study, we characterized the miRNome of FSK-MSCs by determining the expression profile of 380 different miRNAs in inflammation primed vs. control non-primed cells. Methods: TaqMan low density array (TLDA) was performed to identify dysregulated miRNAs after exposing FSK-MSCs to inflammatory signals. Quantitative real-time RT-PCR was carried out to validate the observations. DIANA-miRPath analysis web server was used to identify potential pathways that could be targeted by the dysregulated miRNAs. Results: Sixteen miRNAs were differentially expressed in inflammation-primed vs. non-primed FSK-MSCs. The expression level of miR-27a, -145, -149, -194, -199a, -221, -328, -345, -423-5p, -485-3p, -485-5p, -615-5p and -758 was downregulated whilst that of miR-155, -363 and -886-3p was upregulated. Target pathway prediction of those differentially expressed miRNAs identified different inflammation linked pathways. Conclusions: After determining their miRNome, we identified a striking effect of inflammatory signals on the miRNAs' expression levels in FSK-MSCs. Our results highlight a potential role of miRNAs in modulating the transcription programs of FSK-MSCs in response to inflammatory signals. Further, we propose that specific miRNAs could serve as interesting targets to manipulate some functions of FSK-MSCs, thus ameliorating their therapeutic potential.SCOPUS: ar.jinfo:eu-repo/semantics/publishe
    corecore