Your browser doesn't support javascript.
loading
Pgam5 aggravates hyperglycemia-induced myocardial dysfunction through disrupting Phb2-dependent mitochondrial dynamics.
Chen, Yingzhen; Huang, Jungang; Zhou, Hao; Lin, Jianguo; Tao, Jun.
Affiliation
  • Chen Y; Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
  • Huang J; Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
  • Zhou H; Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, China.
  • Lin J; Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, China.
  • Tao J; Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
Int J Med Sci ; 21(7): 1194-1203, 2024.
Article in En | MEDLINE | ID: mdl-38818468
ABSTRACT
This study aims to elucidate the roles of Phosphoglycerate Mutase Family Member 5 (Pgam5) and Prohibitin 2 (Phb2) in the context of hyperglycemia-induced myocardial dysfunction, a critical aspect of diabetic cardiomyopathy. The research employed primary cardiomyocytes, which were then subjected to hyperglycemia treatment to mimic diabetic conditions. We used siRNA transfection to knock down Pgam5 and overexpressed Phb2 using adenovirus transfection to assess their individual and combined effects on cardiomyocyte health. Mitochondrial function was evaluated through measurements of mitochondrial membrane potential using the JC-1 probe, and levels of mitochondrial reactive oxygen species (ROS) were assessed. Additionally, the study involved qPCR analysis to quantify the transcriptional changes in genes related to mitochondrial fission and mitophagy. Our findings indicate that hyperglycemia significantly reduces cardiomyocyte viability and impairs mitochondrial function, as evidenced by decreased mitochondrial membrane potential and increased ROS levels. Pgam5 knockdown was observed to mitigate these adverse effects, preserving mitochondrial function and cardiomyocyte viability. On the molecular level, Pgam5 was found to regulate genes associated with mitochondrial fission (such as Drp1, Mff, and Fis1) and mitophagy (including Parkin, Bnip3, and Fundc1). Furthermore, overexpression of Phb2 countered the hyperglycemia-induced mitochondrial dysfunction and normalized the levels of key mitochondrial antioxidant enzymes. The combined data suggest a protective role for both Pgam5 knockdown and Phb2 overexpression against hyperglycemia-induced cellular and mitochondrial damage. The study elucidates the critical roles of Pgam5 and Phb2 in regulating mitochondrial dynamics in the setting of hyperglycemia-induced myocardial dysfunction. By modulating mitochondrial fission and mitophagy, Pgam5 and Phb2 emerge as key players in preserving mitochondrial integrity and cardiomyocyte health under diabetic conditions. These findings contribute significantly to our understanding of the molecular mechanisms underlying diabetic cardiomyopathy and suggest potential therapeutic targets for mitigating myocardial dysfunction in diabetes.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Reactive Oxygen Species / Myocytes, Cardiac / Membrane Potential, Mitochondrial / Diabetic Cardiomyopathies / Mitochondrial Dynamics / Prohibitins / Hyperglycemia Limits: Animals / Humans Language: En Journal: Int J Med Sci Journal subject: MEDICINA Year: 2024 Document type: Article Affiliation country: China

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Reactive Oxygen Species / Myocytes, Cardiac / Membrane Potential, Mitochondrial / Diabetic Cardiomyopathies / Mitochondrial Dynamics / Prohibitins / Hyperglycemia Limits: Animals / Humans Language: En Journal: Int J Med Sci Journal subject: MEDICINA Year: 2024 Document type: Article Affiliation country: China