Project description:The aim of this study is to assess the Fecal Microbiota Transplantation (FMT) efficacy in the prevention of allogeneic hematopoietic stem cell transplantation (allo-HSCT) complications and particularly Graft versus Host Disease (GvHD).
The hypothesis of this study is that allogeneic FMT may improve outcomes of these patients.
Project description:This study reports the evolution of the donor CD8 effector T cell transcriptomes at multiple sites and time points within the same host following allo-SCT. Donor derived CD8+ T cells were flow sorted from multiple organs in mice developing GVHD in two clinically relevant murine models of H-2b MHC-matched minor antigen-mismatched BMT: (1) B6>129 model – transfer of C57BL/6 polyclonal CD4+ and CD8+ T cells into 129/Sv BMT recipients; (2) F>M – transfer of monoclonal HY-specific TCR-transgenic MataHari CD8+ T cells into C57BL/6 male BMT recipients.
Project description:This study reports the evolution of the donor CD8 effector T cell transcriptomes at multiple sites and time points within the same host following allo-SCT. Donor derived CD8+ T cells were flow sorted from multiple organs in mice developing GVHD in two clinically relevant murine models of H-2b MHC-matched minor antigen-mismatched BMT: (1) B6>129 model – transfer of C57BL/6 polyclonal CD4+ and CD8+ T cells into 129/Sv BMT recipients; (2) F>M – transfer of monoclonal HY-specific TCR-transgenic MataHari CD8+ T cells into C57BL/6 male BMT recipients.
Project description:Bronchiolitis obliterans (BO) is a pulmonary chronic graft-versus-host disease (cGVHD), which is a noninfectious, irreversible, and poor prognostic complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Fibroblast-myofibroblast transition (FMT)-derived myofibroblasts (MFB) are the main effector cells involved in the development of BO. Through multiple regulations, the anti-fibrotic potential of mesenchymal stem cells (MSC) has been widely studied and was implied with potential clinical value. However, the MSC-mediated regulation of FMT and underlying mechanisms remained largely undefined. By identifying TGF-β1 hypertension as the pivotal landmark during the profibrotic FMT, TGF-β1-induced MFB and uMSC co-culture models were established and utilized to investigate regulations by uMSC on FMT in vitro. Methods including RNA sequencing (RNA-seq), Western blot, qPCR and flow cytometry were used. Our results revealed that uMSC reversibly dedifferentiated MFB into a group of FB-like cells by modulating the TGF-β-SMAD2/3 signaling. Importantly, these proliferation-boosted FB-like cells remained sensitive to TGF-β1 and could be re-induced into MFB. Our findings highlighted the reversibility of uMSC-mediated inhibitions on FMT through TGF-β-SMAD2/3 signaling, which may explain MSC's inconsistent clinical efficacies in treating BO and other fibrotic diseases. These dedifferentiated FB-like cells are still sensitive to TGF-β1 and may further deteriorate MFB phenotypes unless the profibrotic environment is corrected.
Project description:This study reports the evolution of the donor CD8 effector T cell transcriptomes at multiple sites and time points within the same host following allo-SCT. Donor derived CD8+ T cells were flow sorted from multiple organs in mice developing GVHD in two clinically relevant murine models of H-2b MHC-matched minor antigen-mismatched BMT: (1) B6>129 model – transfer of C57BL/6 polyclonal CD4+ and CD8+ T cells into 129/Sv BMT recipients; (2) F>M – transfer of monoclonal HY-specific TCR-transgenic MataHari CD8+ T cells into C57BL/6 male BMT recipients. To test the effect of early lymph node egress on CD8 effector T cell transcription profile, BMT recipients were treatment with FTY720 (1.0mg/kg/day, from D+3 to D+7).
Project description:We analysed the DNA methylation patterns of inguinal white adipose tissue in M-SCT floxed and M-SCT KO, the results indicated a significant widepread hypermethylation modification in M-SCT floxed which contributed to the increased obesity susceptibility of M-SCT floxed
Project description:Background & Aims: Late-stage primary biliary cholangitis (PBC) is defined by bile duct loss, ductular reaction, peribiliary inflammation and fibrosis. Loss of anion exchanger 2 (AE2), the ‘bicarbonate umbrella’ and ductulo-canalicular junctions (DCJ) are hypothesized to promote PBC progression. The secretin (Sct)/secretin receptor (SR) axis controls cystic fibrosis transmembrane receptor (CFTR) activation and AE2 opening, and controls choleresis. We aimed to define the impact of Sct/SR signaling on biliary secretory processes and subsequent injury in late-stage PBC mouse model and human samples. Methods: Female and male wild-type (WT) and dominant-negative transforming growth factor beta receptor II (dnTGFβRII, late-stage PBC model) at 32 wks of age were treated with Sct for 1 wk or 8 wks. Pathways mediated by Sct were identified by RNA-seq in isolated cholangiocytes. Sct/SR/CFTR/AE2 expression and MUC1 levels were evaluated in human control and late-stage PBC. Results: Biliary Sct, SR, CFTR and AE2 expression is reduced in late-stage PBC mouse models and human samples. Sct treatment decreased bile duct loss, ductular reaction, inflammation and fibrosis in late-stage PBC models, as well as hepatic bile acid release and DCJ formation. RNA-seq identified that Sct promoted mature epithelial cell marker expression, specifically anterior grade 2 (Agr2, expressed only in cholangiocytes). Late-stage PBC models and human samples had reduced biliary MUC1, which was enhanced with Sct treatment. Conclusion: Loss of the Sct/SR pathway in late-stage PBC results in a faulty ‘bicarbonate umbrella’ and reduced Agr2 and mucin production. Sct restores cholangiocyte secretory processes and DCJ formation through enhanced mature cholangiocyte phenotypes and healthy bile duct growth. The Sct/SR axis may be a therapeutic target for late-stage PBC patients.
Project description:Background Treatment strategies for Crohn’s disease (CD) suppress diverse inflammatory pathways but many patients remain refractory to treatment. Autologous hematopoietic stem cell transplantation (SCT) is an emerging therapy for medically-refractory CD though the mechanisms through which it circumvents refractory pathophysiology are unknown. Objective The objective of this study is to understand how the immune system reconstitutes post-SCT and whether SCT may function as a cellular therapy restoring appropriately responsive immune cell populations from hematopoietic stem cells (HSCs). Design Adults with CD with active clinical and endoscopic disease who failed available medical therapies were enrolled in a Phase II study of SCT for refractory CD (n=19). Blood and intestinal samples were collected longitudinally and analyzed using CyTOF, scRNA-seq and TCRβ-seq. Stem cell autografts were functionally assayed in mouse xenograft models. Results scRNA-seq and CyTOF analyses reveal that SCT predominantly affected the intestinal myeloid lineage with loss of inflammatory populations and return of macrophages capable of supporting mucosal healing. Xenograft models using patient HSCs suggested HSCs support the early reconstitution of the myeloid lineage and reveal an impairment of short and long-term HSC engraftment that may determine SCT outcomes. Conclusions This study suggests SCT functions as a myeloid-directed cellular therapy reinforcing the critical role of macrophages in refractory CD pathophysiology and as a target for cellular therapies. Furthermore, we report an unrecognized functional heterogeneity among HSC subpopulations in CD that may be relevant to our understanding of CD treatment and pathophysiology.
Project description:We introduced single-chain trimer (SCT) technologies into a high throughput platform for pMHC library generation that can be used for screening antigen specific CD8+ T cells. We compared the diversity of T cell receptor repertoire captured by SCT and folded peptide-MHC multimer presenting HLA-A02:01 restricted CMV peptide. We then constructed SCT libraries designed to capture SARS-CoV-2 spike specific CD8+ T cells from COVID-19 participants and healthy donors. TCR sequencing with antigen specificity was analyzed. The immunogenicity of these epitopes was validated by functional assays of T cells with cloned TCRs captured using SCT libraries.
Project description:The aim of the study was to identify the miRNA expression profile of CD4+CD25high Tregs in SCT patients with and without GvHD early and late after transplantation compared to healthy donors. pool of n = 3 patients without GvHD early (< d100) and late (> d100) after SCT, n=3 patients with GvHD early (< d100) and late (> d100) after SCT and n = 3 sex- and age matched healthy donors for every timepoint