1. Academic Validation
  2. HMGB1 and Caveolin-1 related to RPE cell senescence in age-related macular degeneration

HMGB1 and Caveolin-1 related to RPE cell senescence in age-related macular degeneration

  • Aging (Albany NY). 2019 Jul 7;11(13):4323-4337. doi: 10.18632/aging.102039.
Shuo Sun 1 Bincui Cai 1 Yao Li 2 3 Wenqi Su 1 Xuzheng Zhao 4 Boteng Gong 1 Zhiqing Li 1 Xiaomin Zhang 1 Yalin Wu 5 Chao Chen 5 Stephen H Tsang 2 3 Jin Yang 1 Xiaorong Li 1
Affiliations

Affiliations

  • 1 Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China.
  • 2 Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.
  • 3 Departments of Ophthalmology, Columbia University, New York, NY 10027, USA.
  • 4 Tangshan Eye Hospital, Tangshan, People's Republic of China.
  • 5 Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, College of Medicine, Xiamen University, Xiamen City, People's Republic of China.
Abstract

Accumulation of lipofuscin in the retinal pigment epithelium (RPE) is considered a major cause of RPE dysfunction and senescence in age-related macular degeneration (AMD), and N-retinylidene-N-retinylethanolamine (A2E) is the main fluorophore identified in lipofuscin from aged human eyes. Here, human-induced pluripotent stem cell (iPSC)-RPE was generated from healthy individuals to reveal proteomic changes associated with A2E-related RPE cell senescence. A novel RPE cell senescence-related protein, high-mobility group box 1 (HMGB1), was identified based on proteomic mass spectrometry measurements on iPSC-RPE with A2E treatment. Furthermore, HMGB1 upregulated Caveolin-1, which also was related RPE cell senescence. To investigate whether changes in HMGB1 and Caveolin-1 expression under A2E exposure contribute to RPE cell senescence, human ARPE-19 cells were stimulated with A2E; expression of HMGB1, Caveolin-1, tight junction proteins and senescent phenotypes were verified. HMGB1 inhibition alleviated A2E induced cell senescence. Migration of RPE cells was evaluated. Notably, A2E less than or equal to 10μM induced both HMGB1 and Caveolin-1 protein upregulation and HMGB1 translocation, while Caveolin-1 expression was downregulated when there was more than 10μM A2E. Our data indicate that A2E-induced upregulation of HMGB1、Caveolin-1 and HMGB1 release may relate to RPE cell senescence and play a role in the pathogenesis of AMD.

Keywords

A2E; AMD; Caveolin-1; HMGB1; RPE cell senescence.

Figures
Products