Your browser doesn't support javascript.
loading
Uric acid-driven NLRP3 inflammasome activation triggers lens epithelial cell senescence and cataract formation.
Lin, Hong Liang; Wang, Sheng; Sato, Kota; Zhang, Yu Qiao; He, Bei Ting; Xu, Jing; Nakazawa, Toru; Qin, Yong Jie; Zhang, Hong Yang.
Affiliation
  • Lin HL; Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
  • Wang S; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan.
  • Sato K; Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
  • Zhang YQ; School of Medicine, South China University of Technology, Guangzhou, China.
  • He BT; Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan.
  • Xu J; Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan.
  • Nakazawa T; Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
  • Qin YJ; Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China.
  • Zhang HY; Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
Cell Death Discov ; 10(1): 126, 2024 Mar 09.
Article in En | MEDLINE | ID: mdl-38461179
ABSTRACT
Excessive uric acid (UA) is associated with age-related cataract. A previous study showed that a high UA level in the aqueous humor stimulated the senescence of lens epithelial cells (LECs), leading to cataract progression. To better understand the underlying mechanisms, we investigated UA-driven senescence in human lens tissue samples obtained during surgery, rat lens organ cultures, and in vivo experiments, using senescence-associated ß-galactosidase (SA-ß-gal) staining, electronic microscopy, Western blotting, and histological analyses. Initially, we identified markedly higher expressions of NLRP3 and caspase-1 in the lens capsules of hyper-uricemic patients compared to normo-uricemic patients. This increase was accompanied by a significant rise in the SA-ß-gal positive rate. We next built a cataract model in which rat lenses in an organ culture system were treated with an increasing dosage of UA. Notably, opacification was apparent in the lenses treated with 800 µM of UA starting on the fifth day. Mechanistically, UA treatment not only significantly induced the expression of NLRP3, caspase-1, and IL-1ß, but also upregulated the levels of SA-ß-gal and the senescence regulators p53 and p21. These effects were fully reversed, and lens opacification was ameliorated by the addition of MCC950, a selective NLRP3 antagonist. Moreover, an in vivo model showed that intravitreal UA injection rapidly induced cataract phenotypes within 21 days, an effect significantly mitigated by co-injection with MCC950. Together, our findings suggest that targeting the UA-induced NLRP3 inflammasome with MCC950 could be a promising strategy for preventing cataract formation associated with inflammageing.

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cell Death Discov Year: 2024 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Language: En Journal: Cell Death Discov Year: 2024 Document type: Article Affiliation country:
...