35 research outputs found

    SGA性低身長症における成長ホルモンの体格および脂質代謝に及ぼす影響

    Get PDF
    東医療センター小児科開局50 周年記念論文

    Low-dose Warfarin Functions as an Immunomodulator to Prevent Cyclophosphamide-induced NOD Diabetes

    Get PDF
    Warfarin has been used as an anticoagulant for a long time. Recently, the pleiotropic effect of warfarin has been investigated. As low-dose warfarin has been reported to have anti-inflammatory effect through suppression of IL-6 secretion and inhibit the immune-associated signal between Tyro3 and its ligand, Gas6, the effect of low-dose warfarin on autoimmune diabetes in NOD mice was examined. To investigate the anti-inflammatory effect of warfarin, IL-6 secretion by splenocytes was examined in the presence of various concentrations of warfarin. Low concentration of warfarin inhibited IL-6 secretion. mRNA expression of Rse, one of the Tyro3 receptor family members, and Gas6 were analyzed in NOD mice. It was detected in islets, splenocytes and bone-marrow derived dendritic cells. 0.25 mg/l or 0.50 mg/l of warfarin was orally administered to NOD mice as a cyclophosphamide-induced diabetes model. Oral administration of warfarin at much lower doses than those clinically used as an anticoagulant significantly reduced the degree of insulitis and diabetes incidence in this model. We previously demonstrated that anti-FasL Ab-treatment led to complete prevention of autoimmune diabetes in NOD mice. As Fas/FasL signaling is reported to be essential for cyclophosphamide-induced diabetes model, we extracted RNA from lymphocytes of the inguinal lymph nodes of anti-FasL Ab-treated NOD mice and performed real-time PCR to determine expression of Rse gene. Interestingly, the expression of Rse gene related to the blockade of Fas/FasL signaling was reduced to less than half the level of untreated mice. In conclusion, low-dose warfarin is a potential immunomodulator which can prevent autoimmune diabetes. Type 1 diabetes is a chronic autoimmune disease caused by autoreactive T cells promoting the specific destruction of insulin-producing β cells of the pancreatic islets (1,6). Nonobese diabetic (NOD) mouse is an animal model of human autoimmune diabetes (19). In the NOD mouse, diabetes develops as the result of a chronic inflammation that starts with leukocytic infiltration of islets from 3-5 weeks of age and gradually exacerbates until hyperglycemia develops after 16 weeks of age in a high percentage of female mice. Warfarin has been widely used for a long time as an oral anticoagulant agent. In addition, Kater et al. reported the pleiotropic effect of low-dose warfarin related with inflammation, demonstrating that low-dose warfarin inhibited inflammatory signal transduction through suppression of TNF-α induced IL-6 secretion from murine macrophages (12)

    The Dual Role of Scavenger Receptor Class A in Development of Diabetes in Autoimmune NOD Mice

    No full text
    <div><p>Human type 1 diabetes is an autoimmune disease that results from the autoreactive destruction of pancreatic β cells by T cells. Antigen presenting cells including dendritic cells and macrophages are required to activate and suppress antigen-specific T cells. It has been suggested that antigen uptake from live cells by dendritic cells via scavenger receptor class A (SR-A) may be important. However, the role of SR-A in autoimmune disease is unknown. In this study, SR-A<sup>−/−</sup> nonobese diabetic (NOD) mice showed significant attenuation of insulitis, lower levels of insulin autoantibodies, and suppression of diabetes development compared with NOD mice. We also found that diabetes progression in SR-A<sup>−/−</sup> NOD mice treated with low-dose polyinosinic-polycytidylic acid (poly(I∶C)) was significantly accelerated compared with that in disease-resistant NOD mice treated with low-dose poly(I∶C). In addition, injection of high-dose poly(I∶C) to mimic an acute RNA virus infection significantly accelerated diabetes development in young SR-A<sup>−/−</sup> NOD mice compared with untreated SR-A<sup>−/−</sup> NOD mice. Pathogenic cells including CD4<sup>+</sup>CD25<sup>+</sup> activated T cells were increased more in SR-A<sup>−/−</sup> NOD mice treated with poly(I∶C) than in untreated SR-A<sup>−/−</sup> NOD mice. These results suggested that viral infection might accelerate diabetes development even in diabetes-resistant subjects. In conclusion, our studies demonstrated that diabetes progression was suppressed in SR-A<sup>−/−</sup> NOD mice and that acceleration of diabetes development could be induced in young mice by poly(I∶C) treatment even in SR-A<sup>−/−</sup> NOD mice. These results suggest that SR-A on antigen presenting cells such as dendritic cells may play an unfavorable role in the steady state and a protective role in a mild infection. Our findings imply that SR-A may be an important target for improving therapeutic strategies for type 1 diabetes.</p></div

    Disruption of SR-A confirmed by flow cytometry and by PCR analysis in SR-A<sup>−/−</sup> NOD mice.

    No full text
    <p>DCs (A) and macrophages (B) were analyzed by flow cytometry. Bone marrow cells isolated from 7–9-week-old SR-A<sup>−/−</sup> NOD mice or NOD mice were cultured with granulocyte-macrophage colony-stimulating factor and IL-4 for 7 days and DCs were isolated using magnetic beads. Macrophages were obtained from the abdominal cavity of 8-week-old mice. Representative data for the double staining of CD11c and CD204 (SR-A) or CD11b and CD204 are shown for SR-A<sup>−/−</sup> NOD mice. (C) Neo gene–containing PCR products (1.5 kb and 1.2 kb) from genomic DNA of tails and mRNA from spleen cells and BMDCs were detected in SR-A<sup>−/−</sup> NOD mice, but not in NOD mice. PCR analysis confirmed that SR-A gene was indeed deficient in SR-A<sup>−/−</sup> NOD mice.</p
    corecore