RNase L contributes to experimentally induced type 1 diabetes onset in mice

    1. Aimin Zhou1,2,3
    1. 1Clinical Chemistry Program, Department of Chemistry, Cleveland State University, SI 424, Cleveland, Ohio 44115, USA
      2Center for Gene Regulation in Health and Diseases, Cleveland State University, Cleveland, Ohio 44115, USA
      3Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
      4Barbara Davis Center of Childhood Diabetes, University of Colorado Health Science Center, Denver, Colorado 80045, USA
      5Central Laboratory, the Eighth Hospital of Xi'an, 2 East Zhangba Road, Xi'an 710061, China
      6Department of Biological Sciences, University of Toledo, Toledo, Ohio 43606, USA
      7Department of Biological Sciences, Case Western Reserve University School of Dental Medicine, Cleveland, Ohio 44106, USA
    1. Correspondence should be addressed to A Zhou; Email: a.zhou{at}csuohio.edu

    Abstract

    The cause of type 1 diabetes continues to be a focus of investigation. Studies have revealed that interferon α (IFNα) in pancreatic islets after viral infection or treatment with double-stranded RNA (dsRNA), a mimic of viral infection, is associated with the onset of type 1 diabetes. However, how IFNα contributes to the onset of type 1 diabetes is obscure. In this study, we found that 2-5A-dependent RNase L (RNase L), an IFNα-inducible enzyme that functions in the antiviral and antiproliferative activities of IFN, played an important role in dsRNA-induced onset of type 1 diabetes. Using RNase L-deficient, rat insulin promoter-B7.1 transgenic mice, which are more vulnerable to harmful environmental factors such as viral infection, we demonstrated that deficiency of RNase L in mice resulted in a significant delay of diabetes onset induced by polyinosinic:polycytidylic acid (poly I:C), a type of synthetic dsRNA, and streptozotocin, a drug which can artificially induce type 1-like diabetes in experimental animals. Immunohistochemical staining results indicated that the population of infiltrated CD8+T cells was remarkably reduced in the islets of RNase L-deficient mice, indicating that RNase L may contribute to type 1 diabetes onset through regulating immune responses. Furthermore, RNase L was responsible for the expression of certain proinflammatory genes in the pancreas under induced conditions. Our findings provide new insights into the molecular mechanism underlying β-cell destruction and may indicate novel therapeutic strategies for treatment and prevention of the disease based on the selective regulation and inhibition of RNase L.

    Keywords
    • Received in final form 30 September 2014
    • Accepted 6 October 2014
    • Made available online as an Accepted Preprint 6 October 2014
    | Table of Contents