Your browser doesn't support javascript.
loading
Role of aldose reductase in diabetes-induced retinal microglia activation.
Chang, Kun-Che; Shieh, Biehuoy; Petrash, J Mark.
Affiliation
  • Chang KC; Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
  • Shieh B; Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA.
  • Petrash JM; Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA; Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA. Electronic address: mark.petrash@ucdenver.edu.
Chem Biol Interact ; 302: 46-52, 2019 Apr 01.
Article in En | MEDLINE | ID: mdl-30682331
ABSTRACT
Diabetes-induced hyperglycemia plays a key pathogenic role in degenerative retinal diseases. In diabetic hyperglycemia, aldose reductase (AR) is elevated and linked to the pathogenesis of diabetic retinopathy (DR) and cataract. Retinal microglia (RMG), the resident immune cells in the retina, are thought to contribute to the proinflammatory phenotype in the diabetic eye. However, we have a limited understanding of the potential role of AR expressed in RMG as a mediator of inflammation in the diabetic retina. Glycated proteins accumulate in diabetes, including Amadori-glycated albumin (AGA) which has been shown to induce a proinflammatory phenotype in various tissues. In this study, we investigated the ability of AGA to stimulate inflammatory changes to RMG and macrophages, and whether AR plays a role in this process. In macrophages, treatment with an AR inhibitor (Sorbinil) or genetic knockdown of AR lowered AGA-induced TNF-α secretion (56% and 40%, respectively) as well as cell migration. In a mouse RMG model, AR inhibition attenuated AGA-induced TNF-α secretion and cell migration (67% and 40%, respectively). To further mimic the diabetic milieu in retina, we cultured RMG under conditions of hypoxia and observed the induction of TNF-α and VEGF protein expression. Downregulation of AR in either a pharmacological or genetic manner prevented hypoxia-induced TNF-α and VEGF expression. In our animal study, increased numbers of RMG observed in streptozotocin (STZ)-induced diabetic retina was substantially lower when diabetes was induced in AR knockout mice. Thus, in vitro and in vivo studies demonstrated that AR is involved in diabetes-induced RMG activation, providing a rationale for targeting AR as a therapeutic strategy for DR.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Hydroxyprostaglandin Dehydrogenases / Diabetes Mellitus, Experimental Type of study: Prognostic_studies Limits: Animals Language: En Journal: Chem Biol Interact Year: 2019 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Hydroxyprostaglandin Dehydrogenases / Diabetes Mellitus, Experimental Type of study: Prognostic_studies Limits: Animals Language: En Journal: Chem Biol Interact Year: 2019 Document type: Article Affiliation country: