Your browser doesn't support javascript.
loading
REDD1-dependent GSK3ß dephosphorylation promotes NF-κB activation and macrophage infiltration in the retina of diabetic mice.
Sunilkumar, Siddharth; VanCleave, Ashley M; McCurry, Christopher M; Toro, Allyson L; Stevens, Shaunaci A; Kimball, Scot R; Dennis, Michael D.
Affiliation
  • Sunilkumar S; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • VanCleave AM; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • McCurry CM; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • Toro AL; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • Stevens SA; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • Kimball SR; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA.
  • Dennis MD; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA; Department of Ophthalmology, Penn State College of Medicine, Hershey, Pennsylvania, USA. Electronic address: mdennis@psu.edu.
J Biol Chem ; 299(8): 104991, 2023 08.
Article in En | MEDLINE | ID: mdl-37392853
ABSTRACT
Increasing evidence supports a role for inflammation in the early development and progression of retinal complications caused by diabetes. We recently demonstrated that the stress response protein regulated in development and DNA damage response 1 (REDD1) promotes diabetes-induced retinal inflammation by sustaining canonical activation of nuclear transcription factor, NF-κB. The studies here were designed to identify signaling events whereby REDD1 promotes NF-κB activation in the retina of diabetic mice. We observed increased REDD1 expression in the retina of mice after 16 weeks of streptozotocin (STZ)-induced diabetes and found that REDD1 was essential for diabetes to suppress inhibitory phosphorylation of glycogen synthase kinase 3ß (GSK3ß) at S9. In human retinal MIO-M1 Müller cell cultures, REDD1 deletion prevented dephosphorylation of GSK3ß and increased NF-κB activation in response to hyperglycemic conditions. Expression of a constitutively active GSK3ß variant restored NF-κB activation in cells deficient for REDD1. In cells exposed to hyperglycemic conditions, GSK3ß knockdown inhibited NF-κB activation and proinflammatory cytokine expression by preventing inhibitor of κB kinase complex autophosphorylation and inhibitor of κB degradation. In both the retina of STZ-diabetic mice and in Müller cells exposed to hyperglycemic conditions, GSK3 inhibition reduced NF-κB activity and prevented an increase in proinflammatory cytokine expression. In contrast with STZ-diabetic mice receiving a vehicle control, macrophage infiltration was not observed in the retina of STZ-diabetic mice treated with GSK3 inhibitor. Collectively, the findings support a model wherein diabetes enhances REDD1-dependent activation of GSK3ß to promote canonical NF-κB signaling and the development of retinal inflammation.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Diabetes Mellitus, Experimental / Hyperglycemia Type of study: Prognostic_studies Limits: Animals / Humans / Male Language: En Journal: J Biol Chem Year: 2023 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Diabetes Mellitus, Experimental / Hyperglycemia Type of study: Prognostic_studies Limits: Animals / Humans / Male Language: En Journal: J Biol Chem Year: 2023 Document type: Article Affiliation country: United States