IMR Press / FBL / Volume 28 / Issue 7 / DOI: 10.31083/j.fbl2807140
Open Access Original Research
A Metabolomics-Based Study on NMDAR-Mediated Mitochondrial Damage through Calcium Overload and ROS Accumulation in Myocardial Infarction
Show Less
1 Department of Cardiovascular Surgery, West China Hospital, Sichuan University, 610007 Chengdu, Sichuan, China
2 Cardiovascular Surgery Research Laboratory, West China Hospital, Sichuan University, 610007 Chengdu, Sichuan, China
3 West China - Washington Mitochondria and Metabolism Center, West China Hospital, Sichuan University, 610007 Chengdu, Sichuan, China
*Correspondence: qinchaoyi@wchscu.cn (Chaoyi Qin)
Front. Biosci. (Landmark Ed) 2023, 28(7), 140; https://doi.org/10.31083/j.fbl2807140
Submitted: 12 December 2022 | Revised: 16 March 2023 | Accepted: 21 March 2023 | Published: 19 July 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Coronary artery disease is a leading public health problem. However, the mechanisms underlying mitochondrial damage remain unclear. The present study verified and explored the novel mechanisms underlying ischemic injury based on a metabolomic analysis. Methods: Mouse models of acute myocardial infarction were established, and serum samples were collected for targeted liquid chromatography with tandem mass spectrometry analysis. Based on metabolomic analyses, the N-methyl-d-aspartic acid receptor (NMDAR)–related calcium transporting signaling pathway was selected. Primary cardiomyocyte cultures were used, and N-methyl-d-aspartic acid (NMDA) was used as an agonist to confirm the role of NMDAR in ischemic injury. In addition, Bax, Bcl-2, mitochondrial calcium, potential, and mitochondrial reactive oxygen species accumulation were used to explore the role of NMDAR in mitochondrial damage–induced apoptosis. Results: Glutamate-related metabolism was significantly altered following in acute myocardial infarction. NMDA induces apoptosis under hypoxic conditions NMDAR was translocated to the mitochondrial-related membrane after activation, and its mitochondrial expression was significantly increased (p < 0.05). Mitochondrial damage–induced apoptosis was significantly inhibited by a selective NDMAR antagonist (p < 0.05), while Bax expression was remarkably decreased and Bcl-2 expression was increased (p < 0.05). To further explore the mechanism of NMDAR, mitochondrial calcium, membrane potential, and reactive oxygen species were detected. With NMDAR inhibition under hypoxic conditions, mitochondrial morphology and function were preserved (p < 0.05). Conclusions: Our metabolomic study identified NMDAR as a promising target. In conclusion, our study provides solid data for further studies of the role of NMDAR in cardiovascular diseases and a promising target to interfere with apoptosis in acute myocardial infarction.

Keywords
metabolomics
myocardial infarction
NMDAR
calcium overload
mitochondrial damage
Funding
81900311/National Natural Science Foundation of China
2022YFS0364/Science and Technology Agency Foundation of Sichuan province
Figures
Fig. 1.
Share
Back to top