Phytomedicine. 2025 Dec 11;150:157694. doi: 10.1016/j.phymed.2025.157694. Online ahead of print.
ABSTRACT
BACKGROUND: Myocardial ischemia-reperfusion injury (MIRI) drives adverse cardiac remodeling and ventricular dysfunction, posing a major therapeutic challenge and substantially contributing to global mortality. Despite therapeutic advances, effective MIRI treatments remain limited. Ginsenoside Rd (GSRd), a bioactive constituent from traditional Chinese herbs, has been widely recognized to have cardioprotective effects. However, the role of GSRd in MIRI remains unclear.
PURPOSE: To elucidate the therapeutic efficacy and the underlying molecular mechanisms of GSRd against MIRI.
METHODS: A MIRI model was established in male C57BL/6J mice via left anterior descending coronary artery (LAD) ligation followed by reperfusion. Post-surgery, mice received daily intraperitoneal injections of vehicle, dapagliflozin (1 mg/kg), or GSRd (5, 10, 20 mg/kg) for 28 days. Cardiac function was evaluated by echocardiography. Histopathological changes were assessed using hematoxylin and eosin (H&E), Masson’s trichrome, TUNEL, and immunofluorescence staining. In vitro, isolated adult mouse CMs were subjected to H/R injury and GSRd-containing serum treatment to assess proliferation. Cardiomyocyte proliferation was assessed by Ki-67 immunofluorescence and BrdU flow cytometry. Integrated cardiac untargeted metabolomics (UPLC-MS/MS) and transcriptomics (RNA-seq) were conducted to identify differential metabolites and genes following GSRd intervention. Key targets were validated by RT-qPCR and western blotting. Adeno-associated virus9 (AAV9) with cardiac-specific 3-hydroxy-3-methylglutaryl-CoA synthase 2 (Hmgcs2) knockdown and overexpression were employed to confirm the therapeutic targets of GSRd against MIRI. In addition, transcription factor prediction was performed by multi-platform database analysis, with subsequent chromatin immunoprecipitation-qPCR (ChIP-qPCR) providing mechanistic validation. Furthermore, molecular docking, dynamics simulation, and surface plasmon resonance (SPR) were implemented to evaluate the binding capacity between GSRd and peroxisome proliferator-activated receptor gamma (PPARG). Additionally, GW9662, a PPARG inhibitor, was used to determine the dependence of GSRd-mediated cardioprotection against MIRI on the PPARG/HMGCS2 signaling pathway.
RESULTS: The results revealed that GSRd intervention substantially improved cardiac dysfunction, attenuated ventricular remodeling, ameliorated myocardial pathology, and suppressed inflammatory cytokine and oxidative stress levels in MIRI mice. Mechanistically, multi-omics analysis demonstrated enrichment in ketone body synthesis, carnitine and lipid metabolism, and PPAR signaling after GSRd treatment. Cardiac untargeted metabolomics indicated that GSRd alleviated metabolic dysregulation, concomitant with increased cardiac β-hydroxybutyrate (β-OHB). Transcriptomics identified upregulated ketogenic enzyme gene Hmgcs2 following GSRd intervention. Critically within the infarct and border zones, GSRd concurrently upregulated HMGCS2 expression and β-OHB levels while enhancing cardiomyocyte proliferation. Furthermore, cardiac-specific Hmgcs2 knockdown significantly impaired cardiomyocyte regeneration and attenuated the cardioprotective effects of GSRd in MIRI mice. Conversely, cardiac-specific Hmgcs2 overexpression promoted cardiomyocyte proliferation and recapitulated GSRd’s cardioprotective effects. Transcription factor prediction and ChIP-qPCR analyses verified direct binding of PPARG to the promoter region of Hmgcs2. Molecular docking, dynamics simulation, and SPR confirmed high-affinity binding between GSRd and PPARG. In addition, PPARG inhibition by GW9662 markedly inhibited HMGCS2 expression, suppressed cardiac regeneration, and counteracted the cardioprotective benefits of GSRd, establishing the essential role of the PPARG/HMGCS2 axis.
CONCLUSION: Collectively, this study demonstrates that GSRd ameliorates MIRI by facilitating cardiac regeneration via PPARG/HMGCS2-driven ketone body metabolic reprogramming. Thus, these findings may offer clinicians a novel therapeutic perspective in the management of MIRI.
PMID:41442992 | DOI:10.1016/j.phymed.2025.157694
From ketogenic via this RSS feed


