8 research outputs found

    Real-time PCR analysis of cytokine and chemokine profiles of EAEASCs and WtASCs.

    No full text
    <p>Both cell types were cultured overnight, trypsinized, and analyzed by real-time PCR to determine the expression levels of various cytokines and chemokines TNFα, IL-6, MCP-1, MIP-1α, RANTES, KC, MIP-2α and VEGF. # means <i>P</i><0.05 EAEASCs vs WtASCs (t-test, n = 3).</p

    Cell surface marker profiles of EAEASCs and WtASCs.

    No full text
    <p>A) Cells were analyzed by flow cytometry for MSC surface markers CD29, Sca1; hematopoietic markers CD34 and CD45; phagocytic lineage marker CD11b; and endothelial marker CD31. Gray filled: isotype control; gray line: WtASCs; black line: EAEASCs. B) Cell size based on forward scatter signal of flow cytometry (<i>P</i> = 0.26, t-test, n = 7).</p

    Pro-inflammatory cytokine protein levels in mouse serum.

    No full text
    <p>Sera from 5 mice per group at sacrifice were pooled and analyzed by ELISA to detect levels of TNF-α, IL-12, and IL-17. # means <i>P</i><0.05 vs HBSS-treated EAE group (post-hoc, n = 5); • means <i>P</i><0.05 vs EAEASCs-treated EAE group (post-hoc, n = 5); & means <i>P</i><0.05 vs normal naïve mouse group (post-hoc, n = 5).</p

    Clinical scoring for disease onset and progression.

    No full text
    <p>The clinical scores for the HBSS-treated (n = 20), EAEASC-treated (n = 20) and WtASC-treated (n = 20) EAE mouse groups were recorded daily for the duration of the study. WtASCs significantly reduced the clinical symptoms from PDI13 where the EAEASCs failed to mediate any therapeutic efficacy or improvement. # means <i>P</i><0.01 vs HBSS-treated EAE group (post-hoc, n = 20); • means <i>P</i><0.01 vs EAEASCs-treated EAE group (post-hoc, n = 20).</p

    Lesion and cell infiltration analysis on the spinal cord.

    No full text
    <p>Spinal cords were collected at sacrifice from 3 mice per group. Each spinal cord was sectioned, mounted, and stained with anti-CD3, CD11b, and CD45 followed by Hematoxylin counterstain for lesion size, number and cell infiltration analysis. # means P<0.05 vs EAEASC-treated EAE group (post-hoc, n = 9); & means P<0.05 vs HBSS-treated EAE group (post-hoc, n = 9).</p

    Colony forming unit assays for EAEASCs and WtASCs.

    No full text
    <p>A total of 100 cells were plated on 56.7<sup>2</sup> Nunc cell culture plates and incubated for 14 days. Cells were stained with 3% crystal violet, and colonies 2 mm or larger in diameter were counted. # means <i>P</i><0.05 vs WtASCs (t-test, n = 5).</p

    Design, Synthesis, and Osteogenic Activity of Daidzein Analogs on Human Mesenchymal Stem Cells

    No full text
    Osteoporosis is caused by an overstimulation of osteoclast activity and the destruction of the bone extracellular matrix. Without the normal architecture, osteoblast cells are unable to rebuild phenotypically normal bone. Hormone replacement therapy with estrogen has been effective in increasing osteoblast activity but also has resulted in the increased incidence of breast and uterine cancer. In this study we designed and synthesized a series of daidzein analogs to investigate their osteogenic induction potentials. Human bone marrow derived mesenchymal stem cells (MSCs) from three different donors were treated with daidzein analogs and demonstrated enhanced osteogenesis when compared to daidzein treatment. The enhanced osteogenic potential of these daidzein analogs resulted in increased osterix (Sp7), alkaline phosphatase (ALP), osteopontin (OPN), and insulin-like growth factor 1 (IGF-1), which are osteogenic transcription factors that regulate the maturation of osteogenic progenitor cells into mature osteoblast cells

    Differences in Gastric Carcinoma Microenvironment Stratify According to EBV Infection Intensity: Implications for Possible Immune Adjuvant Therapy

    Get PDF
    <div><p>Epstein-Barr virus (EBV) is associated with roughly 10% of gastric carcinomas worldwide (EBVaGC). Although previous investigations provide a strong link between EBV and gastric carcinomas, these studies were performed using selected EBV gene probes. Using a cohort of gastric carcinoma RNA-seq data sets from The Cancer Genome Atlas (TCGA), we performed a quantitative and global assessment of EBV gene expression in gastric carcinomas and assessed EBV associated cellular pathway alterations. EBV transcripts were detected in 17% of samples but these samples varied significantly in EBV coverage depth. In four samples with the highest EBV coverage (hiEBVaGC – high EBV associated gastric carcinoma), transcripts from the BamHI A region comprised the majority of EBV reads. Expression of LMP2, and to a lesser extent, LMP1 were also observed as was evidence of abortive lytic replication. Analysis of cellular gene expression indicated significant immune cell infiltration and a predominant IFNG response in samples expressing high levels of EBV transcripts relative to samples expressing low or no EBV transcripts. Despite the apparent immune cell infiltration, high levels of the cytotoxic T-cell (CTL) and natural killer (NK) cell inhibitor, IDO1, was observed in the hiEBVaGCs samples suggesting an active tolerance inducing pathway in this subgroup. These results were confirmed in a separate cohort of 21 Vietnamese gastric carcinoma samples using qRT-PCR and on tissue samples using in situ hybridization and immunohistochemistry. Lastly, a panel of tumor suppressors and candidate oncogenes were expressed at lower levels in hiEBVaGC versus EBV-low and EBV-negative gastric cancers suggesting the direct regulation of tumor pathways by EBV.</p> </div
    corecore