All ETDs from UAB

Advisory Committee Chair

Hubert M Tse

Advisory Committee Members

Robinna G Lorenz

Victor M Darley-Usmar

Elena I Frolova

Chad Steele

Document Type

Dissertation

Date of Award

2017

Degree Name by School

Doctor of Philosophy (PhD) Heersink School of Medicine

Abstract

Type 1 diabetes (T1D) is a chronic inflammatory autoimmune disease in which autoreactive T cells selectively destroy insulin-producing pancreatic β-cells. While genetic predisposition underlies susceptibility, environmental factors are proposed to play the important role of triggering the activation of autoimmunity in T1D individuals. Viral infections, particularly by Coxsackie B viruses, are highly suspected as a precipitating event. However, the exact molecular mechanism for how viral infections elicit β-cell-specific autoimmunity remains unclear. Rodent models of virus-induced T1D suggest it is through bystander activation, during which innate antiviral responses to pancreas-tropic viral infections create an inflammatory milieu that breaks the peripheral tolerance normally keeping autoreactive T cells in check. Using the non-obese diabetic (NOD) mouse model, we have previously shown that oxidative stress and redox-regulated innate immune signaling driven by the production of NADPH oxidase 2 (NOX2)-derived superoxide is critical in T1D pathogenesis. Macrophages highly express NOX2 and are absolutely essential in T1D. Analysis of NOD macrophages lacking a functional NOX2 (NOD.Ncf1m1J) revealed that antiviral inflammatory responses to TLR3 stimulation was significantly redox-dependent. Combining these results with the proposed bystander activation model for virus-induced T1D, we hypothesized that production of NOX2-derived superoxide following Coxsackie B virus infection leads to an excessive macrophage inflammatory response that ultimately triggers T1D. Our study revealed that absence of superoxide production lead to protection against Coxsackie B3-accelerated disease, as well as continued protection against spontaneous onset. No impact on pancreatic viral load or clearance was observed. Instead, protection was preceded by dampened inflammatory TNF-α production by pancreas-infiltrating macrophages and a decreased pancreas antiviral transcriptional profile. In vitro mechanistic analyses revealed novel redox-sensitive upregulation of virus-sensing molecules. This study also revealed an early superoxide-dependent upregulation and activation of STAT1, suggesting that superoxide acts as a potent signaling molecule for establishing an inflammatory M1 macrophage phenotype. This result corroborates our study showing that NOD.Ncf1m1J macrophages throughout spontaneous T1D progression maintain an increased non-inflammatory and decreased M1 phenotype. Altogether, we provide evidence that NOX2-derived superoxide can control the antiviral response of macrophages through multiple signaling pathways, which culminates in a diabetogenic inflammatory environment following CB3 infection.

Share

COinS
 
 

To view the content in your browser, please download Adobe Reader or, alternately,
you may Download the file to your hard drive.

NOTE: The latest versions of Adobe Reader do not support viewing PDF files within Firefox on Mac OS and if you are using a modern (Intel) Mac, there is no official plugin for viewing PDF files within the browser window.