1
|
Luo Y, Yu Y, Zeng F, Yi Y, Lu Z, Lin B, Chen L, Zeng Z, Luo D, Liu A. Acetylation of FABP3 alleviates radioimmunotherapy-induced cardiomyocyte senescence by modulating long-chain polyunsaturated fatty acid metabolism. Int Immunopharmacol 2025; 160:114912. [PMID: 40449275 DOI: 10.1016/j.intimp.2025.114912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 05/06/2025] [Accepted: 05/17/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND The combination of thoracic radiotherapy and immunotherapy (radioimmunotherapy) has shown significant antitumor efficacy but is associated with increased cardiotoxicity, the mechanisms of which remain poorly understood. METHODS A total of 72 male C57BL/6 J mice were employed to establish the radioimmunotherapy-induced cardiac injury model, with 18 mice allocated to each of four groups, including the IR group (single-dose 16 Gy cardiac irradiation), ICI group (PD-1 inhibitor 200 μg every 3 days), iRT group (16 Gy cardiac irradiation combined with PD-1 inhibitor), and Control group (IgG). Cardiac function and myocardial senescence were assessed at 28 days, 3 months, and 5 months post-intervention. Additionally, myocardial tissue transcriptomics, non-targeted metabolomics, and acetylated proteomics were performed at 28 days post-intervention, integrated with molecular experiments to investigate the mechanisms of cardiomyocyte senescence. H9C2 cardiomyocytes with FABP3 K45 acetylation-mimetic (K45Q), empty vector (EV), and non-acetylatable (K45R) mutant were used for functional validation. RESULTS Combined radioimmunotherapy significantly exacerbated cardiac dysfunction and cardiomyocyte senescence in murine models, manifested with elevated serum levels of cardiac injury biomarkers of cTnI and NT-proBNP, reduced LVEF and LVFS, aggravated myocardial histopathological changes characterized by enhanced inflammatory infiltration, interstitial edema, and myocardium structure disorder in iRT group compared to the other three groups. Concomitantly, compared with other groups, the senescence-associated markers (p16, p21, and SASP factors) in the myocardial tissues of the iRT group were markedly upregulated from 28 days to 5 months. By integrating transcriptomic and non-targeted metabolomics analyses, as well as molecular experiments, we revealed that radioimmunotherapy resulted in dysregulated myocardial metabolism by suppressing ATP production, promoting lipid droplet accumulation, mitochondrial dysfunction, and fatty acid metabolism alterations, particularly involving long-chain polyunsaturated fatty acid (PUFAs) metabolism. Acetylome profiling identified a significant increase in FABP3 K45 acetylation (log2FC = 8.73, P < 0.05) in iRT vs. Control group, with acute-phase elevation (28 days, P < 0.001) and chronic-phase reduction (3 months, P < 0.001). Functional validation in H9C2 cardiomyocytes demonstrated that, compared to EV and K45R groups, FABP3 K45Q attenuated cellular senescence, enhanced mitochondrial oxidative phosphorylation, fatty acid metabolism, and ATP production, while attenuated ROS generation, lipid droplet accumulation, and glycolysis. Metabolomic analysis also revealed the acetylation of FABP3 K45 was significantly associated with the synthesis or accumulation of PUFAs, such as arachidonic acid and linoleic acid, which may alleviate cardiomyocyte senescence by enhancing energy supply and blocking the synthesis of inflammatory mediators. CONCLUSION FABP3 K45 acetylation mitigates radioimmunotherapy-induced cardiomyocyte senescence and metabolic dysfunction, revealing a novel regulatory mechanism that links post-translational modifications to cardiac cellular homeostasis under combined radioimmunotherapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Ying Yu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Fujuan Zeng
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Yali Yi
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Zhiqin Lu
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Bilin Lin
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China
| | - Zhimin Zeng
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China; Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province 330006, China.
| |
Collapse
|
2
|
Han X, Ashraf M, Shi H, Nkembo AT, Tipparaju SM, Xuan W. Combined Endurance and Resistance Exercise Mitigates Age-Associated Cardiac Dysfunction. Adv Biol (Weinh) 2025; 9:e2400137. [PMID: 38773896 PMCID: PMC11579252 DOI: 10.1002/adbi.202400137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/07/2024] [Indexed: 05/24/2024]
Abstract
Aging is associated with a decline in cardiac function. Exercise has been shown to effectively reduce the risks of cardiovascular diseases. Here whether a combination of endurance and resistance exercises can improve cardiac function in aged mice during late life is investigated. Through transcriptome analysis, several signaling pathways activated in the hearts of 22-month-old mice after combined exercise, including cardiac muscle contraction, mitophagy, and longevity regulation are identified. Combined exercise training mitigated age-associated pathological cardiac hypertrophy, reduced oxidative stress, cardiac senescence, and enhanced cardiac function. Upstream stimulatory factor 2 (Usf2) is upregulated in the aged mouse hearts with combined exercise compared to sedentary mice. In the human cardiomyocytes senescent model, overexpression of Usf2 led to anti-senescence effects, while knockdown of Usf2 exacerbated cellular senescence. The results suggest that a combination of endurance and resistance exercises, such as swimming and resistance running, can mitigate age-related pathological cardiac remodeling and cardiac dysfunction in late life. These cardioprotective effects are likely due to the activation of Usf2 and its anti-senescence effect. Therefore, Usf2 can potentially be a novel therapeutic target for mitigating age-related cardiac dysfunction.
Collapse
Affiliation(s)
- Xiaowei Han
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Muhammad Ashraf
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Hong Shi
- Division of Rheumatology, Department of Internal Medicine, Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Augustine T. Nkembo
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Srinivas M. Tipparaju
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Wanling Xuan
- Department of Pharmaceutical Sciences, USF Health Taneja College of Pharmacy, University of South Florida, Tampa, FL, United States
| |
Collapse
|
3
|
Qi J, Lu B, Jin CW, Shang YY, Pan H, Li H, Tong ZJ, Zhang W, Han L, Zhong M. FP receptor inhibits autophagy to aggravate aging-related cardiac fibrosis through PI3K/AKT/mTOR signaling pathway. Arch Gerontol Geriatr 2025; 133:105824. [PMID: 40096796 DOI: 10.1016/j.archger.2025.105824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND F-prostanoid receptor (FP receptor), a receptor for Prostaglandin F2α(PGF2α), is involved in the process of tissue fibrosis, but its exact role in the aging heart remains unclear. METHODS We investigated cardiac function, myocardial fibrosis levels, autophagy levels and related mechanistic pathways in different groups of mice using gene silencing. At the cellular level, we simulated the senescence process of cardiac fibroblasts and investigated the related mechanisms using relevant inhibitors. RESULTS In aging mice, FP receptor and PI3K/AKT/mTOR pathways are increased and autophagy levels are decreased, ultimately leading to cardiac fibrosis. FP receptor gene silencing slows down the above process. We found similar changes at the cellular level. CONCLUSION FP receptor could activate PI3K/AKT/mTOR pathway and inhibit cardiac autophagy, which resulted in aging-related cardiac fibrosis. Thus, the inhibition of FP receptor could improve aging-related cardiac remodeling, implicating its potential therapeutic application to treat cardiovascular diseases associated with aging.
Collapse
Affiliation(s)
- Jia Qi
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China; Department of Cardiology, Zibo Central Hospital, Zibo, Shandong,255000, PR China
| | - Bin Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Cheng-Wei Jin
- Department of Cardiology, Zibo Central Hospital, Zibo, Shandong,255000, PR China
| | - Yuan-Yuan Shang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Hui Pan
- Department of Geriatric Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong key Laboratory of Cardiovascular Proteomics, Jinan, Shandong, PR China
| | - Hao Li
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Zhou-Jie Tong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Wei Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China
| | - Lu Han
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China; Department of General Practice, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Ming Zhong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, PR China.
| |
Collapse
|
4
|
Wang X, Guo D, He C, Wang X, Wei Y, Zhang F, Wang L, Yang Y. Clinical application of mesenchymal stem cells in immunosenescence: a qualitative review of their potential and challenges. Stem Cell Res Ther 2025; 16:265. [PMID: 40437519 PMCID: PMC12121065 DOI: 10.1186/s13287-025-04360-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Aging leads to a gradual decline in immune function, termed immunosenescence, which significantly elevates the susceptibility to infections, cancers, and other aging-related diseases. Recent advancements have shed light on the molecular underpinnings of immune aging and pioneered novel therapeutic interventions to counteract its effects. Mesenchymal stem cells (MSCs)-a type of multipotent stromal cells with regenerative potential, low immunogenicity, and strong immunomodulatory properties-are increasingly recognized as a promising therapeutic option to reverse or alleviate immunosenescence-related dysfunction. This review systematically summarizes recent discoveries on how MSCs counteract immune aging, particularly their ability to rejuvenate aged immune cells and restore immune homeostasis. It also addresses key challenges, such as variations in MSC sources, donor variability, and the lack of standardized protocols, while proposing future directions to enhance therapeutic precision. Although preclinical and clinical studies highlight the potential of MSC-based strategies for delaying immunosenescence, critical issues remain unresolved, including long-term safety and efficacy, optimizing cell delivery systems, and elucidating context-specific mechanisms. Addressing these challenges will accelerate the development of MSC-based therapies to combat aging-associated immune decline.
Collapse
Affiliation(s)
- Xu Wang
- Clinical Biobank, Department Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Dan Guo
- Clinical Biobank, Department Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chengmei He
- Department of Ultrasound, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, People's Republic of China
| | - Xiaoxi Wang
- Clinical Biobank, Department Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China
| | - Fengchun Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| | - Li Wang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, National Clinical Research Center for Dermatologic and Immunologic Diseases, Beijing, China.
| | - Yanlei Yang
- Clinical Biobank, Department Medical Research Central, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
5
|
Sen I, Trzaskalski NA, Hsiao YT, Liu PP, Shimizu I, Derumeaux GA. Aging at the Crossroads of Organ Interactions: Implications for the Heart. Circ Res 2025; 136:1286-1305. [PMID: 40403108 DOI: 10.1161/circresaha.125.325637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/24/2025]
Abstract
Aging processes underlie common chronic cardiometabolic diseases such as heart failure and diabetes. Cross-organ/tissue interactions can accelerate aging through cellular senescence, tissue wasting, accelerated atherosclerosis, increased vascular stiffness, and reduction in blood flow, leading to organ remodeling and premature failure. This interorgan/tissue crosstalk can accelerate aging-related dysfunction through inflammation, senescence-associated secretome, and metabolic and mitochondrial changes resulting in increased oxidative stress, microvascular dysfunction, cellular reprogramming, and tissue fibrosis. This may also underscore the rising incidence and co-occurrence of multiorgan dysfunction in cardiometabolic aging in the population. Examples include interactions between the heart and the lungs, kidneys, liver, muscles, and brain, among others. However, this phenomenon can also present new translational opportunities for identifying diagnostic biomarkers to define early risks of multiorgan dysfunction, gain mechanistic insights, and help to design precision-directed therapeutic interventions. Indeed, this opens new opportunities for therapeutic development in targeting multiple organs simultaneously to disrupt the crosstalk-driven process of mutual disease acceleration. New therapeutic targets could provide synergistic benefits across multiple organ systems in the same at-risk patient. Ultimately, these approaches may together slow the aging process itself throughout the body. In the future, with patient-centered multisystem coordinated approaches, we can initiate a new paradigm of multiorgan early risk prediction and tailored intervention. With emerging tools including artificial intelligence-assisted risk profiling and novel preventive strategies (eg, RNA-based therapeutics), we may be able to mitigate multiorgan cardiometabolic dysfunction much earlier and, perhaps, even slow the aging process itself.
Collapse
Affiliation(s)
- Ilke Sen
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| | - Natasha A Trzaskalski
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Yung-Ting Hsiao
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
| | - Peter P Liu
- University of Ottawa Heart Institute, Brain-Heart Interconnectome, University of Ottawa, Ontario, Canada (N.A.T., P.P.L.)
| | - Ippei Shimizu
- Department of Cardiovascular Aging, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (Y.-T.H., I. Shimizu)
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (I. Shimizu)
| | - Geneviève A Derumeaux
- Department of Physiology, INSERM U955 (Institut national de la santé et de la recherche médicale, Unité 955), Assistance Publique-Hôpitaux de Paris (AP-HP), Henri Mondor Hospital, Fédération Hospitalo-Universitaire (FHU SENCODE), Ecole Universitaire de Recherche LIVE (EUR LIVE), Université Paris-Est Créteil, France (I. Sen, G.A.D.)
| |
Collapse
|
6
|
Jiang X, Chao L, Liu K, Zhao Y, Wu J, Liu C, Chen W, Zhang D, Tian H. Cationic microbubbles loading shFOXO4/SDF1 rejuvenate the aged heart and alleviate myocardial ischemia-reperfusion injury in the elderly. Free Radic Biol Med 2025:S0891-5849(25)00699-9. [PMID: 40414465 DOI: 10.1016/j.freeradbiomed.2025.05.416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Aging is a significant risk factor for cardiovascular diseases, with ischemic heart disease (IHD) being the leading cause of cardiovascular-related mortality. Inhibition of FOXO4, which selectively eliminates senescent cells, offers protective effects on the aging myocardium. However, the removal of senescent cells may lead to a reduction in tissue cell density, thereby exacerbating tissue space formation and perivascular fibrosis. Therefore, selectively eliminating senescent cells in the aging heart, while simultaneously replenishing therapeutic bone marrow-derived mesenchymal stem cells (BMSCs), holds substantial therapeutic potential for synergistically combating cardiac aging. This study proposes a promising cardiac rejuvenation strategy using ultrasound-targeted microbubble destruction (UTMD)-mediated delivery of shFOXO4/SDF1 to eliminate cellular senescence and enhance BMSC homing. Transcriptomic analysis identified FOXO4 as a pivotal transcription factor in cardiac aging, with FOXO4 protein predominantly expressed in cardiac fibroblasts (CFs) and vascular endothelial cells in the myocardium of aged rats. Knockdown of FOXO4 in aging CFs reversed cellular senescence, and co-culturing these rejuvenated CFs with BMSCs further enhanced the reversal of senescence and bolstered resistance to oxidative stress. The use of UTMD for delivering shFOXO4/SDF1 in dual-gene therapy significantly enhanced BMSC homing, ameliorating cardiac aging, oxidative stress, and inflammation. In an ischemia-reperfusion injury (MIRI) model, pretreatment with shFOXO4/SDF1 effectively reduced cardiomyocyte apoptosis, promoted neovascularization, reduced infarct size, and improved cardiac function. The combined removal of senescent cells and enhanced BMSC homing synergistically ameliorated cardiac aging and improved post-MIRI prognosis in aging hearts. These findings provide novel insights and potential therapeutic strategies for addressing cardiac aging and age-related heart diseases.
Collapse
Affiliation(s)
- Xingpei Jiang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Limeng Chao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Kexun Liu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Yuanzhong Zhao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Jie Wu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Department of Medical Genetics, School of Basic Medical Sciences, Harbin Medical University, Harbin,China
| | - Chang Liu
- Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Wei Chen
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China
| | - Dongyang Zhang
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Hai Tian
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; Future Medical Laboratory, The Second Affiliated Hospital of Harbin Medical University, Harbin , China.
| |
Collapse
|
7
|
Dai Z, Ding H, Zhang Q, Fu L, Tai S. Spatial Insights in Cardiovascular Aging. Aging Dis 2025:AD.2025.0272. [PMID: 40423633 DOI: 10.14336/ad.2025.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Spatial omics provides unprecedented insights into how the cardiovascular system is spatially organized and how cellular phenotypes are distributed. Researchers have been able to clarify the complex spatial architecture of the cardiovascular system and how cellular phenotypes are distributed during the aging process by integrating data from spatial omics. In addition, this new technology has revealed previously hidden patterns of gene expression and cellular communication that were not detected using traditional bulk omics approaches. In this review, we explore the contribution of spatial omics in clarifying the molecular mechanisms that influence cardiovascular aging, highlighting the importance and application of spatial omics in unraveling the spatial heterogeneity within the aging cardiovascular system. This will help us understand the molecular mechanisms implicated in age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Zhongling Dai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Huiqin Ding
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Quan Zhang
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Liyao Fu
- Department of Blood Transfusion, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shi Tai
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
8
|
Li Q, Song C, Wei Z, Zhou H, Wang S, Li H, Yang H, Luo Q, Li J, Chen M. Age-associated methionine sulfoxide reductase A protects against valvular interstitial cell senescence and valvular calcification. GeroScience 2025:10.1007/s11357-025-01675-w. [PMID: 40341600 DOI: 10.1007/s11357-025-01675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/22/2025] [Indexed: 05/10/2025] Open
Abstract
Calcific aortic valve disease (CAVD) is a cardiovascular disease prevalent in the aging population, resulting in high morbidity and mortality rates. However, the molecular mechanisms underlying CAVD remain unclear. We initially conducted an RNA sequencing analysis of aortic valve leaflets from rats of different ages to identify key genes involved in valvular aging and calcification. Bioinformatics analysis demonstrated that methionine sulfoxide reductase A (MSRA) was crucial to valvular calcification and senescence. To further investigate whether and how MSRA influences CAVD pathogenesis, we utilized two in vitro models: a human valvular interstitial cell (VIC) calcification model induced by osteogenic medium, and a VIC senescence model induced by hydrogen peroxide. Western blotting, immunofluorescence, flow cytometry, and alkaline phosphatase staining were conducted to evaluate the changes in calcific nodule formation and senescent markers. In vivo, ApoE-/- mice were treated either a normal chow or a high-cholesterol chow to determine the effects of MSRA overexpression on aortic valve calcification and senescence. MSRA silencing increased the osteogenic differentiation and senescence of VIC, whereas its overexpression produced the opposite effects. Similarly, we found that MSRA overexpression reduced calcium deposition and decreased the levels of senescent markers in ApoE-/- mice. Further mechanism experiments showed that MSRA suppressed osteoblastic differentiation via inhibiting the toll-like receptor (TLR2)/nuclear factor-κB (NF-κB) pathway. Our findings demonstrate that MSRA ameliorates valvular calcification and senescence by inhibiting TLR2/NF-κB pathway, highlighting MSRA as a promising target for treating age-associated CAVD.
Collapse
Affiliation(s)
- Qing Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- National Clinical Research Center for Geriatrics, The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zisong Wei
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hao Zhou
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuoding Wang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Cardiac Structure and Function Research Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
9
|
Zhao Y, Zhang C, Zhang C, Zheng X, Qi Y, Kong B, Hou Y, Ti Y, Bu P. SIRT3 suppresses vascular endothelial senescence via DHRS2 and contributes to the anti-vascular aging effect of Bazi Bushen capsule. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156571. [PMID: 40049100 DOI: 10.1016/j.phymed.2025.156571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
AIMS Vascular and endothelial aging are significant causes of chronic diseases among the elderly. This study investigated the specific mechanism by which sirtuin 3 (SIRT3) regulates vascular endothelial senescence and the beneficial role of Bazi Bushen capsule (BZBS) in preventing vascular aging. METHODS Human umbilical vein endothelial cells and mouse aortic endothelial cells were cultured with D-galactose (D-gal) to induce aging and evaluate the beneficial effects of the SIRT3-dehydrogenase/reductase member 2 (DHRS2) axis on the inhibition of vascular endothelial aging. d-Gal was injected intraperitoneally into wild-type and Sirt3 knockout mice, while BZBS was administered orally. Histochemical staining, immunohistochemistry, and western blotting assays were used to explore the beneficial effects of BZBS against aging-associated vascular remodelling. Endothelial cell function assays were used to evaluate the role of BZBS in suppressing endothelial aging in vitro. RESULTS SIRT3 deacetylated DHRS2 and modulated the translation of DHRS2. The SIRT3-DHRS2 axis played an important role in preserving mitochondrial homeostasis and reducing reactive oxygen species generation through suppressing endothelial nitric oxide synthase (eNOS) translocating to mitochondria and eNOS-Thr495 phosphorylation mediated by protein kinase C δ (PKCδ). BZBS mitigated vascular remodelling and relieved endothelial oxidative stress via the SIRT3-DHRS2 axis. CONCLUSION SIRT3 activates DHRS2-PKCδ to stop aging in endothelial cells by inhibiting uncoupled eNOS translocating to mitochondria. BZBS rescued vascular aging and endothelial dysfunction via the SIRT3-DHRS2 axis. Revealing a protective mechanism by which SIRT3 inhibits endothelial senescence, this study provides evidence for BZBS in delaying vascular aging.
Collapse
Affiliation(s)
- Yuan Zhao
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Chunmei Zhang
- Department of Cardiology, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Chen Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Xuehui Zheng
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Yan Qi
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Binghui Kong
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China
| | - Yunlong Hou
- State Key Laboratory for Innovation and Transformation of Luobing Theory, Shijiazhuang 050035, China; Hebei Provincial Key Laboratory of Luobing, Shijiazhuang 050035, China
| | - Yun Ti
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China.
| | - Peili Bu
- State Key Laboratory for Innovation and Transformation of Luobing Theory, China; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province, China; Department of Cardiology, Qilu Hospital of Shandong University, 107 Wenhuaxi Road, Jinan 250012, China.
| |
Collapse
|
10
|
Fan Y, Zheng Y, Zhang Y, Xu G, Liu C, Hu J, Ji Q, Zhang S, Fang S, Lei J, Li LZ, Wang X, Xu X, Wang C, Wang S, Ma S, Song M, Jiang W, Zhu J, Feng Y, Wang J, Yang Y, Zhu G, Tian XL, Zhang H, Song W, Yang J, Yao Y, Liu GH, Qu J, Zhang W. ARID5A orchestrates cardiac aging and inflammation through MAVS mRNA stabilization. NATURE CARDIOVASCULAR RESEARCH 2025; 4:602-623. [PMID: 40301689 DOI: 10.1038/s44161-025-00635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/10/2025] [Indexed: 05/01/2025]
Abstract
Elucidating the regulatory mechanisms of human cardiac aging remains a great challenge. Here, using human heart tissues from 74 individuals ranging from young (≤35 years) to old (≥65 years), we provide an overview of the histological, cellular and molecular alterations underpinning the aging of human hearts. We decoded aging-related gene expression changes at single-cell resolution and identified increased inflammation as the key event, driven by upregulation of ARID5A, an RNA-binding protein. ARID5A epi-transcriptionally regulated Mitochondrial Antiviral Signaling Protein (MAVS) mRNA stability, leading to NF-κB and TBK1 activation, amplifying aging and inflammation phenotypes. The application of gene therapy using lentiviral vectors encoding shRNA targeting ARID5A into the myocardium not only mitigated the inflammatory and aging phenotypes but also bolstered cardiac function in aged mice. Altogether, our study provides a valuable resource and advances our understanding of cardiac aging mechanisms by deciphering the ARID5A-MAVS axis in post-transcriptional regulation.
Collapse
Affiliation(s)
- Yanling Fan
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yandong Zheng
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiyuan Zhang
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Chun Liu
- Department of Physiology and Medicine, Cardiovascular Center, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jianli Hu
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qianzhao Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuaiqi Fang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lan-Zhu Li
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xing Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xi Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Cui Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuai Ma
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Moshi Song
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junming Zhu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yijia Feng
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiangang Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ying Yang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guodong Zhu
- Institute of Gerontology, Guangzhou Geriatric Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weihong Song
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Yao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jing Qu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Weiqi Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
11
|
Liu F, Cai H. Diabetes and calcific aortic valve disease: implications of glucose-lowering medication as potential therapy. Front Pharmacol 2025; 16:1583267. [PMID: 40356984 PMCID: PMC12066769 DOI: 10.3389/fphar.2025.1583267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Calcific aortic valve disease (CAVD) is a progressive disease, of which the 2-year mortality is >50% for symptomatic disease. However, there are currently no pharmacotherapies to prevent the progression of CAVD unless transcatheter or surgical aortic valve replacement is performed. The prevalence of diabetes among CAVD has increased rapidly in recent decades, especially among those undergoing aortic valve replacement. Diabetes and its comorbidities, such as hypertension, hyperlipidemia, chronic kidney disease and ageing, participated jointly in the initiation and progression of CAVD, which increased the management complexity in patients with CAVD. Except from hyperglycemia, the molecular links between diabetes and CAVD included inflammation, oxidative stress and endothelial dysfunction. Traditional cardiovascular drugs like lipid-lowering agents and renin-angiotensin system blocking drugs have proven to be unsuccessful in retarding the progression of CAVD in clinical trials. In recent years, almost all kinds of glucose-lowering medications have been specifically assessed for decelerating the development of CAVD. Based on the efficacy for atherosclerotic cardiovascular disease and CAVD, this review summarized current knowledge about glucose-lowering medications as promising treatment options with the potential to retard CAVD.
Collapse
Affiliation(s)
| | - Haipeng Cai
- Department of Cardiology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| |
Collapse
|
12
|
Cai J, Yu R, Zhang N, Zhang H, Zhang Y, Xiang Y, Xu H, Xiao X, Zhao X. Association Between Cardiovascular Biological Age and Cardiovascular Disease - A Prospective Cohort Study. Circ J 2025; 89:620-628. [PMID: 40074355 DOI: 10.1253/circj.cj-24-0824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
BACKGROUND Biological age serves as a common starting point for various age-related diseases and can be associated with a wide range of cardiovascular outcomes. However, associations between cardiovascular biological age (CBA) and various types of cardiovascular disease (CVD) remain unclear. METHODS AND RESULTS Analyzing 262,343 UK Biobank participants, we constructed CBA based on composite biomarkers using the Klemera-Doubal method (denoted as KDM-CBA). We measured KDM-CBA acceleration as the difference between KDM-CBA and chronological age. We then examined the associations between KDM-CBA and 17 CVD types using Cox proportional hazard models. We used restricted cubic spline models to assess potential nonlinear associations of KDM-CBA and KDM-CBA acceleration with different types of CVDs. We observed that KDM-CBA (per 1SD increase) was associated with various CVD types, but with different extent (hypertension: hazard ratio (HR)=2.115, 95% confidence interval (CI): 2.083-2.148; coronary atherosclerosis: HR=1.711, 95% CI: 1.545-1.896). We observed similar results for KDM-CBA acceleration and KDM-CBA. KDM-CBA and KDM-CBA acceleration showed J-type nonlinear associations with nearly all CVD types (cutoff values of ≈55 and -1.7 years for KDM-CBA and KDM-CBA acceleration, respectively). CONCLUSIONS Our study showed that CBA is associated with increased incidence of CVD, which further validates aging as a common starting point for different CVD types as well as highlighting CBA's role as an early CVD indicator, providing valuable insights for CVD interventions.
Collapse
Affiliation(s)
- Jiajie Cai
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Rui Yu
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Ning Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Hongmei Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Yuan Zhang
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Yi Xiang
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Hao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University
| | - Xiong Xiao
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| | - Xing Zhao
- West China School of Public Health and West China Fourth Hospital, Sichuan University
| |
Collapse
|
13
|
Jin M, Li C, Wu Z, Tang Z, Xie J, Wei G, Yang Z, Huang S, Chen Y, Li X, Chen Y, Liao W, Liao Y, Chen G, Zheng H, Bin J. Inhibiting the Histone Demethylase Kdm4a Restrains Cardiac Fibrosis After Myocardial Infarction by Promoting Autophagy in Premature Senescent Fibroblasts. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414830. [PMID: 40231733 DOI: 10.1002/advs.202414830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 03/26/2025] [Indexed: 04/16/2025]
Abstract
Premature senescent fibroblasts (PSFs) play an important role in regulating the fibrotic process after myocardial infarction (MI), but their effect on cardiac fibrosis remains unknown. Here, the investigation is aimed to determine whether PSFs contribute to cardiac fibrosis and the underlying mechanisms involved. It is observed that premature senescence of fibroblasts is strongly activated in the injured myocardium at 7 days after MI and identified that Kdm4a is located in PSFs by the analysis of scRNA-seq data and immunostaining staining. Moreover, fibroblast specific gain- and loss-of-function assays showed that Kdm4a promoted the premature senescence of fibroblasts and cardiac interstitial fibrosis, contributing to cardiac remodeling in the advanced stage after MI, without influencing early cardiac rupture. ChIP-seq and ChIP-PCR revealed that Kdm4a deficiency promoted autophagy in PSFs by reducing Trim44 expression through increased levels of the H3K9me3 modification in the Trim44 promoter region. Furthermore, a coculture system revealed that Kdm4a overexpression increased the accumulation of PSFs and the secretion of senescence-associated secretory phenotype (SASP) factors, subsequently inducing cardiac fibrosis, which could be reversed by Trim44 interference. Kdm4a induces the premature senescence of fibroblasts through Trim44-mediated autophagy and then facilitates interstitial fibrosis after MI, ultimately resulting in cardiac remodeling, but not affecting ventricular rupture.
Collapse
Affiliation(s)
- Ming Jin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Chuling Li
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| | - Zhaoyi Wu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Zhenquan Tang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Jingfang Xie
- Guangdong Provincial Geriatrics Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Guoquan Wei
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Zhiwen Yang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Senlin Huang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Yijin Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Xinzhong Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Yanmei Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou, 510515, China
- Cardiovascular Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, 528200, China
| |
Collapse
|
14
|
Liang N, Liu S, Wang Y, Ying L, Zhang K, Li H, Xiao L, Hu Y, Luo G. Nicotinamide Mononucleotide (NMN) Improves the Senescence of Mouse Vascular Smooth Muscle Cells Induced by Ang II Through Activating p-AMPK/KLF4 Pathway. Pharmaceuticals (Basel) 2025; 18:553. [PMID: 40283988 PMCID: PMC12030317 DOI: 10.3390/ph18040553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Vascular smooth muscle cells (VSMCs) senescence exacerbates vascular diseases like atherosclerosis and hypertension. Angiotensin II (Ang II) is a strong inducer of VSMCs senescence, causing vascular damage, though its exact mechanism is unclear. Nicotinamide mononucleotide (NMN), a NAD+ precursor, has gained attention for its anti-senescence potential, yet its role in inhibiting VSMCs senescence is not fully understood. Methods: This study assessed senescence markers, including β-galactosidase activity (SA-β-gal) and the senescence-associated secretory phenotype (SASP), in mouse VSMCs treated with Ang II alone or with NMN and relevant activators/inhibitors. Results: Compared to controls, SA-β-gal levels and SASP secretion significantly increased in Ang II-exposed cells. In contrast, NMN reduced the expression of both markers. NMN also reversed Ang II-induced VSMCs senescence by downregulating KLF4 and p16 through AMPK activation, which Ang II inhibited, while decreasing mRNA levels of key SASP components. The effects of the AMPK activator AICAR were similar to those of NMN, whereas the AMPK inhibitor Compound C negated NMN's effects. Conclusions: In summary, NMN mitigates Ang II-induced mouse VSMCs senescence via the AMPK/KLF4/p16 pathway. This study underscores the anti-senescence effects of NMN on mouse VSMCs, supporting further exploration of its potential as a food supplement for preventing and treating vascular senescence.
Collapse
Affiliation(s)
- Na Liang
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Si Liu
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Yan Wang
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Linyao Ying
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Keyi Zhang
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Hao Li
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Lin Xiao
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| | - Yuming Hu
- Hunan Provincial Center for Disease Control and Prevention, Changsha 410078, China
- Hunan Academy of Preventive Medicine, Changsha 410078, China
| | - Gang Luo
- Xiangya School of Public Health, Central South University, Changsha 410078, China; (N.L.); (S.L.); (Y.W.); (L.Y.); (K.Z.); (H.L.); (L.X.)
| |
Collapse
|
15
|
Yang Z, Potenza DM, Ming XF. Is Senolytic Therapy in Cardiovascular Diseases Ready for Translation to Clinics? Biomolecules 2025; 15:545. [PMID: 40305307 PMCID: PMC12024785 DOI: 10.3390/biom15040545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Aging is a predominant risk factor for cardiovascular diseases. There is evidence demonstrating that senescent cells not only play a significant role in organism aging but also contribute to the pathogenesis of cardiovascular diseases in younger ages. Encouraged by recent findings that the elimination of senescent cells by pharmacogenetic tools could slow down and even reverse organism aging in animal models, senolytic drugs have been developed, and the translation of results from basic research to clinical settings has been initiated. Because numerous studies in the literature show beneficial therapeutic effects of targeting senescent cells in cardiomyopathies associated with aging and ischemia/reperfusion and in atherosclerotic vascular disease, senolytic drugs are considered the next generation of therapies for cardiovascular disorders. However, recent studies have reported controversial results or detrimental effects caused by senolytic therapeutic approaches, including worsening of cardiac dysfunction, instability of atherosclerotic plaques, and even an increase in mortality in animal models, which challenges the translation of senolytic therapy into the clinical practice. This brief review article will focus on (1) analyzing and discussing the beneficial and detrimental effects of senolytic therapeutic approaches in cardiovascular diseases and cardiovascular aging and (2) future research directions and questions that are essential to understand the controversies and to translate preclinical results of senolytic therapies into clinical practice.
Collapse
Affiliation(s)
- Zhihong Yang
- Laboratory of Cardiovascular and Aging Research, Department of Endocrinology, Metabolism, and Cardiovascular System, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland; (D.M.P.); (X.-F.M.)
| | | | | |
Collapse
|
16
|
Boichenko V, Noakes VM, Reilly-O’Donnell B, Luciani GB, Emanueli C, Martelli F, Gorelik J. Circulating Non-Coding RNAs as Indicators of Fibrosis and Heart Failure Severity. Cells 2025; 14:553. [PMID: 40214506 PMCID: PMC11989213 DOI: 10.3390/cells14070553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality worldwide, representing a complex clinical syndrome in which the heart's ability to pump blood efficiently is impaired. HF can be subclassified into heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF), each with distinct pathophysiological mechanisms and varying levels of severity. The progression of HF is significantly driven by cardiac fibrosis, a pathological process in which the extracellular matrix undergoes abnormal and uncontrolled remodelling. Cardiac fibrosis is characterized by excessive matrix protein deposition and the activation of myofibroblasts, increasing the stiffness of the heart, thus disrupting its normal structure and function and promoting lethal arrythmia. MicroRNAs, long non-coding RNAs, and circular RNAs, collectively known as non-coding RNAs (ncRNAs), have recently gained significant attention due to a growing body of evidence suggesting their involvement in cardiac remodelling such as fibrosis. ncRNAs can be found in the peripheral blood, indicating their potential as biomarkers for assessing HF severity. In this review, we critically examine recent advancements and findings related to the use of ncRNAs as biomarkers of HF and discuss their implication in fibrosis development.
Collapse
Affiliation(s)
- Veronika Boichenko
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular and Surgical Sciences, The University of Verona, Policlinico G. B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milano, Italy
| | - Victoria Maria Noakes
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Benedict Reilly-O’Donnell
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Giovanni Battista Luciani
- Department of Surgery, Dentistry, Pediatrics and Gynecology, Cardiovascular and Surgical Sciences, The University of Verona, Policlinico G. B. Rossi, P.le. La Scuro 10, 37134 Verona, Italy
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097 Milano, Italy
| | - Julia Gorelik
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
17
|
Li Q, Liu Q, Lin Z, Lin W, Lin Z, Huang F, Zhu P. Comparison Between the Effect of Mid-Late-Life High-Intensity Interval Training and Continuous Moderate-Intensity Training in Old Mouse Hearts. J Gerontol A Biol Sci Med Sci 2025; 80:glaf025. [PMID: 39928548 PMCID: PMC11973967 DOI: 10.1093/gerona/glaf025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Indexed: 02/12/2025] Open
Abstract
The impact of mid-late-life exercise on the aging heart remains unclear, particularly the effects of high-intensity interval training (HIIT) and continuous moderate-intensity training (CMIT). This study was the first to examine cardiac function, tissue characteristics, electrical remodeling, mitochondrial morphology, and homeostasis in old mice subjected to CMIT or HIIT, compared to untrained controls. Our results showed that 8-week HIIT significantly improved the survival rate of old mice. HIIT presented advantages on cardiac function, deposition of collagen fibers, neovascularization, aging biomarkers, and mitochondrial homeostasis. Only CMIT alleviated age-related cardiac hypertrophy. However, CMIT potentially exacerbated adverse cardiac electrical remodeling. Those findings suggested HIIT as a particularly appealing option for clinical application for aging populations.
Collapse
Affiliation(s)
- Qiaowei Li
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Qin Liu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhong Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenwen Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Zhonghua Lin
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Feng Huang
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Pengli Zhu
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, China
- Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| |
Collapse
|
18
|
Shen E, Wu Y, Ye W, Li S, Zhu J, Jiang M, Hu Z, Cao G, Yi X, Li F, Tang Z, Li X, Lee KY, Jin L, Wang X, Cong W. The FGF13-Caveolin-1 Axis: A Key Player in the Pathogenesis of Doxorubicin- and D-Galactose-Induced Premature Cardiac Aging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501055. [PMID: 40184605 DOI: 10.1002/advs.202501055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/17/2025] [Indexed: 04/06/2025]
Abstract
Delaying senescence of cardiomyocytes has garnered widespread attention as a potential target for preventing cardiovascular diseases (CVDs). FGF13 (Fibroblast growth factor 13) has been implicated in various pathophysiological processes. However, its role in premature myocardial aging and cardiomyocyte senescence remains unknown. Adeno-associated virus 9 (AAV9) vectors expressing FGF13 and cardiac-specific Fgf13 knockout (Fgf13KO) mice are utilized to reveal that FGF13 overexpression and deficiency exacerbated and alleviated Doxorubicin/D-galactose-induced myocardial aging characteristics and functional impairment, respectively. Transcriptomics are employed to identify an association between FGF13 and Caveolin-1 (Cav1). Mechanistic studies indicated that FGF13 regulated the Cav1 promoter activity and expression through the p38/MAPK pathway and nuclear translocation of p65, as well as the binding level of PTRF to Cav1 to mediate cardiomyocyte senescence. Furthermore, Cav1 overexpression in murine hearts reversed the alleviatory effects of FGF13 deficiency on the Doxorubicin/D-galactose-induced myocardial aging phenotype and dysfunction. This study has demonstrated that FGF13 regulated the Cav1-p53-p21 axis to augment cardiomyocyte senescence and thereby exacerbated cardiac premature aging and suggests that FGF13 knockdown may be a promising approach to combat CVDs in response to aging and chemotoxicity.
Collapse
Affiliation(s)
- Enzhao Shen
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yuecheng Wu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Weijian Ye
- Department of Pharmacy, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, P. R China
| | - Sihang Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Junjie Zhu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Meifan Jiang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Zhicheng Hu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Gaoyong Cao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Xiaojing Yi
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Fan Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Zhouhao Tang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Xiaokun Li
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| | - Kwang Youl Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Litai Jin
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Ningbo Key Laboratory of Skin Science, Ningbo College of Health Sciences, Ningbo, 315000, P. R. China
| | - Xu Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
| | - Weitao Cong
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, P. R. China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, P. R. China
| |
Collapse
|
19
|
Wang XJ, Huang L, Hou M, Guo J. Senescence-related Genes as Prognostic Markers for STEMI Patients: LASSO Regression-Based Bioinformatics and External Validation. J Cardiovasc Transl Res 2025; 18:354-365. [PMID: 39786668 DOI: 10.1007/s12265-024-10583-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 10/14/2024] [Indexed: 01/12/2025]
Abstract
The prognostic value of differentially expressed senescence-related genes(DESRGs) in ST-segment elevation myocardial infarction(STEMI) patients is unclear. We used GEO2R to identify DESRGs from GSE60993 and performed functional enrichment analysis. We built an optimal prognostic model with LASSO penalized Cox regression via GSE49925. We evaluated the model with survival analysis, ROC curve, decision curve analysis, nomogram, and external validation with plasma samples. We created a prognostic signature with three dysregulated DESRGs (CDC25B, FKBP5, and ECHDC3) and two clinical variables (serum creatinine, Gensini score). The signature stratified patients into low- and high-risk groups and showed strong predictive performance within two years. The external validation confirmed the survival difference between the groups. We identified three DESRGs that were differentially expressed and prognostic in STEMI patients. The model incorporating three DESRGs showed promising prediction and utility for stratifying patients and estimating survival risk in STEMI patients.
Collapse
Affiliation(s)
- Xing-Jie Wang
- Clinical Laboratory of Tianjin Chest Hospital, 261 Taierzhuang South Road, Tianjin, 300222, Jinnan District, China.
| | - Lei Huang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Tianjin, 300170, Hedong District, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin University Central Hospital, Tianjin, China
| | - Min Hou
- Clinical Laboratory of Tianjin Chest Hospital, 261 Taierzhuang South Road, Tianjin, 300222, Jinnan District, China
| | - Jie Guo
- Clinical Laboratory of Tianjin Chest Hospital, 261 Taierzhuang South Road, Tianjin, 300222, Jinnan District, China
| |
Collapse
|
20
|
Fang Z, Raza U, Song J, Lu J, Yao S, Liu X, Zhang W, Li S. Systemic aging fuels heart failure: Molecular mechanisms and therapeutic avenues. ESC Heart Fail 2025; 12:1059-1080. [PMID: 39034866 PMCID: PMC11911610 DOI: 10.1002/ehf2.14947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Systemic aging influences various physiological processes and contributes to structural and functional decline in cardiac tissue. These alterations include an increased incidence of left ventricular hypertrophy, a decline in left ventricular diastolic function, left atrial dilation, atrial fibrillation, myocardial fibrosis and cardiac amyloidosis, elevating susceptibility to chronic heart failure (HF) in the elderly. Age-related cardiac dysfunction stems from prolonged exposure to genomic, epigenetic, oxidative, autophagic, inflammatory and regenerative stresses, along with the accumulation of senescent cells. Concurrently, age-related structural and functional changes in the vascular system, attributed to endothelial dysfunction, arterial stiffness, impaired angiogenesis, oxidative stress and inflammation, impose additional strain on the heart. Dysregulated mechanosignalling and impaired nitric oxide signalling play critical roles in the age-related vascular dysfunction associated with HF. Metabolic aging drives intricate shifts in glucose and lipid metabolism, leading to insulin resistance, mitochondrial dysfunction and lipid accumulation within cardiomyocytes. These alterations contribute to cardiac hypertrophy, fibrosis and impaired contractility, ultimately propelling HF. Systemic low-grade chronic inflammation, in conjunction with the senescence-associated secretory phenotype, aggravates cardiac dysfunction with age by promoting immune cell infiltration into the myocardium, fostering HF. This is further exacerbated by age-related comorbidities like coronary artery disease (CAD), atherosclerosis, hypertension, obesity, diabetes and chronic kidney disease (CKD). CAD and atherosclerosis induce myocardial ischaemia and adverse remodelling, while hypertension contributes to cardiac hypertrophy and fibrosis. Obesity-associated insulin resistance, inflammation and dyslipidaemia create a profibrotic cardiac environment, whereas diabetes-related metabolic disturbances further impair cardiac function. CKD-related fluid overload, electrolyte imbalances and uraemic toxins exacerbate HF through systemic inflammation and neurohormonal renin-angiotensin-aldosterone system (RAAS) activation. Recognizing aging as a modifiable process has opened avenues to target systemic aging in HF through both lifestyle interventions and therapeutics. Exercise, known for its antioxidant effects, can partly reverse pathological cardiac remodelling in the elderly by countering processes linked to age-related chronic HF, such as mitochondrial dysfunction, inflammation, senescence and declining cardiomyocyte regeneration. Dietary interventions such as plant-based and ketogenic diets, caloric restriction and macronutrient supplementation are instrumental in maintaining energy balance, reducing adiposity and addressing micronutrient and macronutrient imbalances associated with age-related HF. Therapeutic advancements targeting systemic aging in HF are underway. Key approaches include senomorphics and senolytics to limit senescence, antioxidants targeting mitochondrial stress, anti-inflammatory drugs like interleukin (IL)-1β inhibitors, metabolic rejuvenators such as nicotinamide riboside, resveratrol and sirtuin (SIRT) activators and autophagy enhancers like metformin and sodium-glucose cotransporter 2 (SGLT2) inhibitors, all of which offer potential for preserving cardiac function and alleviating the age-related HF burden.
Collapse
Affiliation(s)
- Zhuyubing Fang
- Cardiovascular Department of Internal MedicineKaramay Hospital of People's Hospital of Xinjiang Uygur Autonomous RegionKaramayXinjiang Uygur Autonomous RegionChina
| | - Umar Raza
- School of Basic Medical SciencesShenzhen UniversityShenzhenGuangdong ProvinceChina
| | - Jia Song
- Department of Medicine (Cardiovascular Research)Baylor College of MedicineHoustonTexasUSA
| | - Junyan Lu
- Department of CardiologyZengcheng Branch of Nanfang Hospital, Southern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Shun Yao
- Department of NeurosurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| | - Xiaohong Liu
- Cardiovascular Department of Internal MedicineKaramay Hospital of People's Hospital of Xinjiang Uygur Autonomous RegionKaramayXinjiang Uygur Autonomous RegionChina
| | - Wei Zhang
- Outpatient Clinic of SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| | - Shujuan Li
- Department of Pediatric CardiologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouGuangdong ProvinceChina
| |
Collapse
|
21
|
Jiang T, Zeng Q, Wang J. Unlocking the secrets of Cardiac development and function: the critical role of FHL2. Mol Cell Biochem 2025; 480:2143-2157. [PMID: 39466483 DOI: 10.1007/s11010-024-05142-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 10/12/2024] [Indexed: 10/30/2024]
Abstract
FHL2 (Four-and-a-half LIM domain protein 2) is a crucial factor involved in cardiac morphogenesis, the process by which the heart develops its complex structure. It is expressed in various tissues during embryonic development, including the developing heart, and has been shown to play important roles in cell proliferation, differentiation, and migration. FHL2 interacts with multiple proteins to regulate cardiac development as a coactivator or a corepressor. It is involved in cardiac specification and determination of cell fate, cardiomyocyte growth, cardiac remodeling, myofibrillogenesis, and the regulation of HERG channels. Targeting FHL2 has therapeutic implications as it could improve cardiac function, control arrhythmias, alleviate heart failure, and maintain cardiac integrity in various pathological conditions. The identification of FHL2 as a signature gene in atrial fibrillation suggests its potential as a diagnostic marker and therapeutic target for this common arrhythmia.
Collapse
Affiliation(s)
- Tingting Jiang
- Department of Clinical Laboratory, Hengyang Medical School, the Affiliated Nanhua Hospital, University of South China, Hengyang, 421000, China
| | - Qun Zeng
- Department of Biochemistry and Molecular Biology, Hengyang Medical School, University of South China, Hengyang, 421000, China
| | - Jing Wang
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, 410219, China.
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research On Functional Nucleic Acid, Changsha Medical University, Changsha, 410219, China.
- The First Clinical College, Changsha Medical University, Changsha, 410219, China.
| |
Collapse
|
22
|
Chen Y, Jiang F, Zeng Y, Zhang M. The role of retinal pigment epithelial senescence and the potential of senotherapeutics in age-related macular degeneration. Surv Ophthalmol 2025:S0039-6257(25)00053-0. [PMID: 40089029 DOI: 10.1016/j.survophthal.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of visual impairment in the aging population. Evidence showing the presence of cellular senescence in retinal pigment epithelium (RPE) of patients with AMD is growing. Senescent RPE play a pivotal role in its pathogenesis. The senescent RPE suffers from structural and functional alterations and disruption of the surrounding microenvironment due to the development of the senescence-associated secretory phenotype, which contributes to metabolic dysfunctions and inflammatory responses in the retina. Senotherapeutics, including senolytics, senomorphics and others, are novel treatments targeting senescent cells and are promising treatments for AMD. As senotherapeutic targets are being developed, it is promising that the burden of AMD could be decreased.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Feipeng Jiang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Yue Zeng
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| | - Meixia Zhang
- Department of Ophthalmology and Laboratory of Macular Disease, West China Hospital, Sichuan University, China.
| |
Collapse
|
23
|
Wu L, Zhu X, Pan S, Chen Y, Luo C, Zhao Y, Xing J, Shi K, Zhang S, Li J, Chai J, Ling X, Qiu J, Wang Y, Shen Z, Jie W, Guo J. Diabetes Advances Cardiomyocyte Senescence Through Interfering Rnd3 Expression and Function. Aging Cell 2025:e70031. [PMID: 40025898 DOI: 10.1111/acel.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 03/04/2025] Open
Abstract
Rnd3 is a small Rho-GTPase that has been implicated in various cardiovascular diseases. Yet, its role in diabetes-induced cardiomyocyte senescence remains unknown. Here we tested the role of Rnd3 in cardiomyocyte senescence and diabetic cardiomyopathy (DCM). The expression of Rnd3 was found to be reduced in peripheral blood mononuclear cells from diabetic patients and correlated negatively with age but positively with cardiac function. In 96-week-old Sprague Dawley (SD) rats, cardiac function was impaired, accompanied by an increased number of SA-β-gal-positive cells and elevated levels of the senescence-associated secretory phenotype (SASP) related factors, compared to those of 12-week-old rats. Diabetes and high glucose (HG, 35 mmol/L D-glucose) suppressed Rnd3 expression in cardiomyocytes and induced cardiomyocyte senescence. The deficiency of Rnd3 exacerbated cardiomyocyte senescence in vitro and in vivo. MicroRNA sequencing in AC16 cells identified a conserved miR-103a-3p (present in humans and rats) as a key HG-upregulated microRNA that bound to the Rnd3 3'-UTR. In cultured cardiomyocytes, miR-103a-3p inhibitors antagonized HG-induced cardiomyocyte senescence dependent on Rnd3 expression. Treatment with AAV9 vectors carrying miR-103a-3p sponges and Rnd3-overexpressing plasmids alleviated cardiomyocyte senescence and restored cardiac function in diabetic SD rats. HG stimulation increased STAT3 (Tyr705) phosphorylation and promoted its nuclear translocation in H9C2 cells, an effect exacerbated by Rnd3 knockout. Mechanistically, Rnd3 interacted with p-STAT3 in the cytoplasm, facilitating proteasome-mediated ubiquitination and p-STAT3 degradation. The STAT3 inhibitor S3I-201 blocked HG-induced STAT3 activation and mitigated cardiomyocyte senescence. These findings suggest that diabetes induces cardiomyocyte senescence via the miR-103a-3p/Rnd3/STAT3 signaling pathway, highlighting a potential therapeutic target for DCM.
Collapse
Affiliation(s)
- Linxu Wu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
- Public Research Center of Hainan Medical University, Haikou, China
| | - Xinglin Zhu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Shanshan Pan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Yan Chen
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Cai Luo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Yangyang Zhao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Jingci Xing
- Department of Pathology and Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang, China
| | - Kaijia Shi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Shuya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Jiaqi Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Jinxuan Chai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Xuebin Ling
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Jianmin Qiu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
| | - Yan Wang
- Public Research Center of Hainan Medical University, Haikou, China
| | - Zhihua Shen
- Department of Pathology and Pathophysiology, School of Basic Medicine Sciences, Guangdong Medical University, Zhanjiang, China
| | - Wei Jie
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Junli Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory for Tropical Cardiovascular Diseases Research, School of Public Health, Hainan Medical University, Haikou, China
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
24
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
25
|
Sato M, Kadomatsu T, Morinaga J, Kinoshita Y, Torigoe D, Horiguchi H, Ohtsuki S, Yamamura S, Kusaba R, Yamaguchi T, Yoshioka G, Araki K, Wakayama T, Miyata K, Node K, Oike Y. HINT1 suppression protects against age-related cardiac dysfunction by enhancing mitochondrial biogenesis. Mol Metab 2025; 93:102107. [PMID: 39909188 PMCID: PMC11850129 DOI: 10.1016/j.molmet.2025.102107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/07/2025] Open
Abstract
OBJECTIVE Cardiac function declines with age, impairing exercise tolerance and negatively impacting healthy aging. However, mechanisms driving age-related declines in cardiac function are not fully understood. METHODS We examined mechanisms underlying age-related cardiac dysfunction using 3- and 24-month-old wild-type mice fed ad libitum or 24-month-old wild-type mice subjected to 70% calorie restriction (CR) starting at 2-month-old. In addition, cardiac aging phenotypes and mitochondrial biogenesis were also analyzed in 25-month-old cardiac-specific Hint1 knockout mice, 24-month-old CAG-Caren Tg mice, and 24-month-old wild-type mice injected with AAV6-Caren. RESULTS We observed inactivation of mitochondrial biogenesis in hearts of aged mice. We also showed that activity of the BAF chromatin remodeling complex is repressed by HINT1, whose expression in heart increases with age, leading to decreased transcription of Tfam, which promotes mitochondrial biogenesis. Interestingly, CR not only suppressed age-related declines in cardiac function and mitochondrial biogenesis but blocked concomitant increases in cardiac HINT1 protein levels and maintained Tfam transcription. Furthermore, expression of the lncRNA Caren, which inhibits Hint1 mRNA translation, decreased with age in heart, and CR suppressed this effect. Finally, decreased HINT1 expression due to Caren overexpression antagonized age-related declines in mitochondrial biogenesis, ameliorating age-related cardiac dysfunction, exercise intolerance, and exercise-induced cardiac damage and subsequent death of mice. CONCLUSION Our findings suggest that mitochondrial biogenesis in cardiomyocytes decreases with age and could underlie cardiac dysfunction, and that the Caren-HINT1-mitochondrial biogenesis axis may constitute a mechanism linking CR to resistance to cardiac aging. We also show that ameliorating declines in mitochondrial biogenesis in cardiomyocytes could counteract age-related declines in cardiac function, and that this strategy may improve exercise tolerance and extend so-called "healthy life span".
Collapse
Affiliation(s)
- Michio Sato
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Division of Kumamoto Mouse Clinic (KMC), Institute of Resource Developmental and Analysis (IRDA), Kumamoto University, Kumamoto, Japan; Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
| | - Tsuyoshi Kadomatsu
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Jun Morinaga
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Yuya Kinoshita
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Daisuke Torigoe
- Division of Experimental Genetics, IRDA, Kumamoto University, Kumamoto, Japan
| | - Haruki Horiguchi
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Department of Aging and Geriatric Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Graduate School of Pharmacological Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuji Yamamura
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryoko Kusaba
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Takanori Yamaguchi
- Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
| | - Goro Yoshioka
- Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
| | - Kimi Araki
- Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Division of Developmental Genetics, IRDA, Kumamoto University, Kumamoto, Japan
| | - Tomohiko Wakayama
- Department of Histology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Keishi Miyata
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan
| | - Koichi Node
- Department of Cardiovascular Medicine, School of Medicine, Saga University, Saga, Japan
| | - Yuichi Oike
- Department of Molecular Genetics, Kumamoto University, Kumamoto, Japan; Center for Metabolic Regulation of Healthy Aging (CMHA), Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan; Department of Aging and Geriatric Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
26
|
Chen Y, Li J, Liu X, Geng Z, Xu K, Su J. Advances in biomarkers and diagnostic significance of organ aging. FUNDAMENTAL RESEARCH 2025; 5:683-696. [PMID: 40242549 PMCID: PMC11997494 DOI: 10.1016/j.fmre.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 04/18/2025] Open
Abstract
A complete understanding of aging is a critical first step in treating age-related diseases and postponing aging dysfunction in the context of an aging global population. Aging in organisms is driven by related molecular alterations that gradually occur in many organs. There has previously been a wealth of knowledge of how cells behave as they age, but when aging is investigated as a disease, the discovery and selection of aging biomarkers and how to diagnose the aging of the organism are crucial. Here, we provide a summary of the state of the field and suggest future potential routes for research on organ senescence markers. We reviewed research on biomarkers of risk of aging from the perspective of organ aging and summarized the biomarkers currently used on three scales. We emphasize that the combination of traditional markers with emerging multifaceted biomarkers may be a better way to diagnose age-related diseases.
Collapse
Affiliation(s)
- Yulin Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Jiadong Li
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
- School of Medicine, Shanghai University, Shanghai 200444, China
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Xinru Liu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Zhen Geng
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China
- Organoid Research Center, Shanghai University, Shanghai 200444, China
| |
Collapse
|
27
|
Li H, Yang Y, Li B, Yang J, Liu P, Gao Y, Zhang M, Ning G. Comprehensive Analysis Reveals the Potential Diagnostic Value of Biomarkers Associated With Aging and Circadian Rhythm in Knee Osteoarthritis. Orthop Surg 2025; 17:922-938. [PMID: 39846237 PMCID: PMC11872380 DOI: 10.1111/os.14370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/24/2025] Open
Abstract
OBJECTIVE Knee osteoarthritis (KOA) is characterized by structural changes. Aging is a major risk factor for KOA. Therefore, the objective of this study was to examine the role of genes related to aging and circadian rhythms in KOA. METHODS This study identified differentially expressed genes (DEGs) by comparing gene expression levels between normal and KOA samples from the GEO database. Subsequently, we intersected the DEGs with aging-related circadian rhythm genes to obtain a set of aging-associated circadian rhythm genes differentially expressed in KOA. Next, we conducted Mendelian randomization (MR) analysis, using the differentially expressed aging-related circadian rhythm genes in KOA as the exposure factors, their SNPs as instrumental variables, and KOA as the outcome event, to explore the causal relationship between these genes and KOA. We then performed Gene Set Enrichment Analysis (GSEA) to investigate the pathways associated with the selected biomarkers, conducted immune infiltration analysis, built a competing endogenous RNA (ceRNA) network, and performed molecular docking studies. Additionally, the findings and functional roles of the biomarkers were further validated through experiments on human cartilage tissue and cell models. RESULTS A total of 75 differentially expressed aging-circadian rhythm related genes between the normal group and the KOA group were identified by difference analysis, primarily enriched in the circadian rhythm pathway. Two biomarkers (PFKFB4 and DDIT4) were screened by MR analysis. Then, immune infiltration analysis showed significant differences in three types of immune cells (resting dendritic cells, resting mast cells, and M2 macrophages), between the normal and KOA groups. Drug prediction and molecular docking results indicated stable binding of PFKFB4 to estradiol and bisphenol_A, while DDIT4 binds stably to nortriptyline and trimipramine. Finally, cell lines with stable expression of the biomarkers were established by lentiviral infection and resistance screening, Gene expression was significantly elevated in overexpressing cells of PFKFB4 and DDIT4 and reversed the proliferation and migration ability of cells after IL-1β treatment. CONCLUSIONS Two diagnostic and therapeutic biomarkers associated with aging-circadian rhythm in KOA were identified. Functional analysis, molecular mechanism exploration, and experimental validation further elucidated their roles in KOA, offering novel perspectives for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Hao Li
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| | - Yuze Yang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Bo Li
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| | - Jiaju Yang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Pengyu Liu
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Yuanpeng Gao
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Min Zhang
- Department of OrthopedicsThe Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury RepairTaiyuanChina
| | - Guangzhi Ning
- Department of OrthopedicsTianjin Medical University General Hospital, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal CordTianjinChina
| |
Collapse
|
28
|
Sun Y, Xiao L, Chen L, Wang X. Doxorubicin-Induced Cardiac Remodeling: Mechanisms and Mitigation Strategies. Cardiovasc Drugs Ther 2025:10.1007/s10557-025-07673-6. [PMID: 40009315 DOI: 10.1007/s10557-025-07673-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/16/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND The therapeutic prowess of doxorubicin in oncology is marred by its cardiotoxic consequences, manifesting as cardiac remodeling. Pathophysiological alterations triggered by doxorubicin include inflammatory cascades, fibrotic tissue deposition, vascular and valvular changes, and finally cardiomyopathy. These multifarious consequences collectively orchestrate the deterioration of cardiac architecture and function. METHOD By charting the molecular underpinnings and remedial prospects, this review aspires to contribute a novel perspective using latest publications to the ongoing quest for cardioprotection in cancer therapy. RESULTS AND DISCUSSION Experimental analyses demonstrate the pivotal roles of oxidative stress and subsequent necrosis and apoptosis of cardiomyocytes, muscle cells, endothelial cells, and small muscle cells in different parts of the heart. In addition, severe and unusual infiltration of macrophages, mast cells, and neutrophils can amplify oxidative damage and subsequent impacts such as chronic inflammatory responses, vascular and valvular remodeling, and fibrosis. These modifications can render cardiomyopathy, ischemia, heart attack, and other disorders. In an endeavor to counteract these ramifications, a spectrum of emerging adjuvants and strategies are poised to fortify the heart against doxorubicin's deleterious effects. CONCLUSION The compendium of mitigation tactics such as innovative pharmacological agents hold the potential to attenuate the cardiotoxic burden.
Collapse
Affiliation(s)
- Yanna Sun
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Lili Xiao
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Linlin Chen
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China
| | - Xiaofang Wang
- Department of Cardiology, The First Affiliated of Zhengzhou University, Zhengzhou City Henan Province, 450052, China.
| |
Collapse
|
29
|
Liberale L, Tual-Chalot S, Sedej S, Ministrini S, Georgiopoulos G, Grunewald M, Bäck M, Bochaton-Piallat ML, Boon RA, Ramos GC, de Winther MPJ, Drosatos K, Evans PC, Ferguson JF, Forslund-Startceva SK, Goettsch C, Giacca M, Haendeler J, Kallikourdis M, Ketelhuth DFJ, Koenen RR, Lacolley P, Lutgens E, Maffia P, Miwa S, Monaco C, Montecucco F, Norata GD, Osto E, Richardson GD, Riksen NP, Soehnlein O, Spyridopoulos I, Van Linthout S, Vilahur G, Wentzel JJ, Andrés V, Badimon L, Benetos A, Binder CJ, Brandes RP, Crea F, Furman D, Gorbunova V, Guzik TJ, Hill JA, Lüscher TF, Mittelbrunn M, Nencioni A, Netea MG, Passos JF, Stamatelopoulos KS, Tavernarakis N, Ungvari Z, Wu JC, Kirkland JL, Camici GG, Dimmeler S, Kroemer G, Abdellatif M, Stellos K. Roadmap for alleviating the manifestations of ageing in the cardiovascular system. Nat Rev Cardiol 2025:10.1038/s41569-025-01130-5. [PMID: 39972009 DOI: 10.1038/s41569-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2025] [Indexed: 02/21/2025]
Abstract
Ageing of the cardiovascular system is associated with frailty and various life-threatening diseases. As global populations grow older, age-related conditions increasingly determine healthspan and lifespan. The circulatory system not only supplies nutrients and oxygen to all tissues of the human body and removes by-products but also builds the largest interorgan communication network, thereby serving as a gatekeeper for healthy ageing. Therefore, elucidating organ-specific and cell-specific ageing mechanisms that compromise circulatory system functions could have the potential to prevent or ameliorate age-related cardiovascular diseases. In support of this concept, emerging evidence suggests that targeting the circulatory system might restore organ function. In this Roadmap, we delve into the organ-specific and cell-specific mechanisms that underlie ageing-related changes in the cardiovascular system. We raise unanswered questions regarding the optimal design of clinical trials, in which markers of biological ageing in humans could be assessed. We provide guidance for the development of gerotherapeutics, which will rely on the technological progress of the diagnostic toolbox to measure residual risk in elderly individuals. A major challenge in the quest to discover interventions that delay age-related conditions in humans is to identify molecular switches that can delay the onset of ageing changes. To overcome this roadblock, future clinical trials need to provide evidence that gerotherapeutics directly affect one or several hallmarks of ageing in such a manner as to delay, prevent, alleviate or treat age-associated dysfunction and diseases.
Collapse
Affiliation(s)
- Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Stefano Ministrini
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | | | - Myriam Grunewald
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Magnus Bäck
- Translational Cardiology, Centre for Molecular Medicine, Department of Medicine Solna, and Department of Cardiology, Heart and Vascular Centre, Karolinska Institutet, Stockholm, Sweden
- Inserm, DCAC, Université de Lorraine, Nancy, France
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC location VUmc, Amsterdam, Netherlands
| | - Gustavo Campos Ramos
- Department of Internal Medicine I/Comprehensive Heart Failure Centre, University Hospital Würzburg, Würzburg, Germany
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis and Ischaemic Syndromes; Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location AMC, Amsterdam, Netherlands
| | - Konstantinos Drosatos
- Metabolic Biology Laboratory, Cardiovascular Center, Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Paul C Evans
- William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Jane F Ferguson
- Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Mauro Giacca
- British Heart foundation Centre of Reseach Excellence, King's College London, London, UK
| | - Judith Haendeler
- Cardiovascular Degeneration, Medical Faculty, University Hospital and Heinrich-Heine University, Düsseldorf, Germany
| | - Marinos Kallikourdis
- Adaptive Immunity Lab, IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Daniel F J Ketelhuth
- Cardiovascular and Renal Research Unit, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rory R Koenen
- CARIM-School for Cardiovascular Diseases, Department of Biochemistry, Maastricht University, Maastricht, Netherlands
| | | | - Esther Lutgens
- Department of Cardiovascular Medicine & Immunology, Mayo Clinic, Rochester, MN, USA
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Satomi Miwa
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Claudia Monaco
- Kennedy Institute, NDORMS, University of Oxford, Oxford, UK
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Osto
- Division of Physiology and Pathophysiology, Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Medical University of Graz, Graz, Austria
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Oliver Soehnlein
- Institute of Experimental Pathology, University of Münster, Münster, Germany
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Sophie Van Linthout
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité - Universitätmedizin Berlin, Berlin, Germany
| | - Gemma Vilahur
- Research Institute, Hospital de la Santa Creu y Sant Pau l, IIB-Sant Pau, Barcelona, Spain
| | - Jolanda J Wentzel
- Cardiology, Biomedical Engineering, Erasmus MC, Rotterdam, Netherlands
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBERCV, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Health and Innovation Research Foundation (FICSI) and Cardiovascular Health and Network Medicine Department, University of Vic (UVIC-UCC), Barcelona, Spain
| | - Athanase Benetos
- Department of Geriatrics, University Hospital of Nancy and Inserm DCAC, Université de Lorraine, Nancy, France
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University, Frankfurt am Main, Germany
| | - Filippo Crea
- Centre of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Roma, Italy
| | - David Furman
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Vera Gorbunova
- Departments of Biology and Medicine, University of Rochester, Rochester, NY, USA
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Joseph A Hill
- University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and National Heart and Lung Institute, Imperial College, London, UK
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas (CSIC), Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Alessio Nencioni
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, Genoa, Italy
- Dipartimento di Medicina Interna e Specialità Mediche-DIMI, Università degli Studi di Genova, Genova, Italy
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, USA
| | - Kimon S Stamatelopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Nektarios Tavernarakis
- Medical School, University of Crete, and Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Zoltan Ungvari
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James L Kirkland
- Center for Advanced Gerotherapeutics, Division of Endocrinology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université Paris Cité, Sorbonne Université, Inserm, Institut Universitaire de France, Paris, France
| | | | - Konstantinos Stellos
- Department of Cardiovascular Research, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
30
|
Li F, Zhu J, Zhou J, Zeng G, Zhou Y, Lin Q, Zhang Z, Tan S, Liu Q. Analysis risk factors of long-term adverse outcomes and a prediction nomogram for coronary artery disease patients underwent fractional flow reserve. Int J Med Sci 2025; 22:1292-1300. [PMID: 40084257 PMCID: PMC11898858 DOI: 10.7150/ijms.106807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 03/16/2025] Open
Abstract
Background: The role of fractional flow reserve (FFR) in intermediate lesions has been widely used and recommended by guidelines. However, the long-term outcomes in patient with an intermediate stenosis received FFR have not yet been investigated comprehensively. Methods: We retrospectively included 558 patients underwent both coronary artery angiography (CAG) and FFR. Multivariate logistic regression analysis was employed to identify the independent predictors of major adverse cardiovascular and cerebrovascular events (MACCEs). Additionally, we constructed a prediction nomogram and tested its performance by multiple methods. Results: During a median follow-up of 6.2 years, 87 (15.59%) adverse events were documented. Multivariate logistic regression results revealed that age (OR 1.13, p<0.01), diabetes mellitus (OR, 5.87, p<0.01), hyperuricemia (OR, 2.91, p<0.01) were independently associated with MACCEs. The nomogram consists of age, smoking, hypertension, diabetes mellitus (DM), hyperuricemia, and FFR≤0.8 six factors. The AUC of 3-year, 5-year, 7-year receiver operating characteristic (ROC) curves of training set were 0.697, 0.823, 0.854, and of validation set were 0.845, 0.924, 0.856. The calibration curves and decision curve analysis (DCA) illustrated the ability of the nomogram to predict long-term adverse outcomes and its net benefits in clinical practice. Conclusions: Age, DM, and hyperuricemia were independently associated with long-term adverse outcomes, and the constructed nomogram may be used as a visible tool to predict long-term adverse outcomes for patients underwent FFR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Qiming Liu
- Department of Cardiology, The Second Xiangya Hospital of Central South University, Changsha 410000, China
| |
Collapse
|
31
|
Peng T, Xiang J, Tian Y, Tang X, Wang L, Gao L, Luo OJ, Huang L, Chen G. Lycium barbarum glycopeptide ameliorates aging phenotypes and enhances cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. Exp Gerontol 2025; 200:112686. [PMID: 39827719 DOI: 10.1016/j.exger.2025.112686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Aging is a complex biological process that disrupts tissue structure and impairs physiological function, which contributes to the development of age-related diseases such as cardiovascular disorders. However, effective treatment strategies are lacking. OBJECTIVE To investigate the geroprotective effects of Lycium barbarum glycopeptide (LbGp) and its potential mechanisms in a D-galactose-induced accelerated aging mouse model. METHODS Mice were subcutaneously injected with D-galactose (500 mg/kg/day) for 12 weeks to induce aging, while LbGp was orally administered (100 mg/kg/day) throughout the study. The geroprotective effects of LbGp were assessed by behavioral tests, cardiac echocardiography, pathohistological and transcriptomic analyses. Transmission electron microscopy was used to observe the ultrastructure of mitochondria. Mitochondrial stress assays and JC-1 fluorescent probe were conducted to evaluate mitochondrial function. Flow cytometer and western blot were performed to assess mitophagy flux. RESULTS LbGp treatment improved the aging phenotypes of D-galactose-induced mice, with a pronounced enhancement in cardiac function compared to neurocognitive and skeletal muscle functions. Transcriptome analysis indicated that LbGp ameliorated energy metabolism in the heart. Mitochondrial assays revealed LbGp improved mitochondrial function and preserved structural integrity of the mitochondrial inner membrane. LbGp attenuated mitochondrial fission and restored impaired PINK1/Parkin-mediated mitophagy pathway caused by D-galactose in cardiomyocytes. CONCLUSION LbGp can ameliorate aging phenotypes and enhance cardiac metabolism by activating the PINK1/Parkin-mediated mitophagy pathway in D-galactose-induced mice. These findings underscore its potential as a therapeutic agent for aging and aging-related cardiovascular diseases.
Collapse
Affiliation(s)
- Tianchan Peng
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Xiang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yun Tian
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaogen Tang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lina Wang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Oscar Junhong Luo
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China
| | - Li'an Huang
- Department of Neurology, Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou 510632, China; Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou 510632, China; Zhuhai Institute of Jinan University, Zhuhai 519070, China.
| |
Collapse
|
32
|
Fang Z, Wang L, Wang Y, Ma Y, Fang Y, Zhang W, Cao R, Zhang Y, Li H, Chen S, Tian L, Shen X, Cao F. Protective effects and bioinformatic analysis of narciclasine on vascular aging via cross-talk between inflammation and metabolism through inhibiting skeletal muscle-specific ceramide synthase 1. Mech Ageing Dev 2025; 223:112021. [PMID: 39706373 DOI: 10.1016/j.mad.2024.112021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVE The senescence of smooth muscle is one of the independent risk factors in atherosclerosis progression in which the vascular inflammation plays an important role on vascular dysfunction. This study is designed to explore the novel vascular aging biomarkers and screen the potential molecular interventional targets through bioinformatic analysis. RESULTS Transcriptional analysis was conducted based on the GSE16487 open access database, which included 15 human vascular tissue samples from two groups: young group (≤ 60 years old, n = 8) and aged group (≥ 75 years old, n = 7). There were 275 differential expression genes (119 upregulated and 156 downregulated genes) with minimum 1.5-fold change between two groups. 9 genes were mainly participated in inflammation-related signaling pathways, in which narciclasine was validated as the most effective candidate for modulation the ceramide synthesis. In vitro and animal study demonstrated that narciclasine reversed vascular aging by inhibiting skeletal muscle-specific ceramide synthase 1 (CerS1), reducing the ceramide level derived from CerS1, and improving fat deposition and circulating glycolipid metabolism. CONCLUSION Narciclasine attenuates vascular aging and modulates the cross-talk between inflammation and metabolism via inhibiting skeletal muscle-specific ceramide synthase 1.
Collapse
Affiliation(s)
- Zhiyi Fang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Linghuan Wang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yabin Wang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yan Ma
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yan Fang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Weiwei Zhang
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Ruihua Cao
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Yingjie Zhang
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Hui Li
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Sijia Chen
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Lei Tian
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Xiaoying Shen
- Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China
| | - Feng Cao
- School of Medicine, Nankai University, 94 Weijin Road, Tianjin 30071, China; Institude of Chinese PLA Geriatric Medicine, The Second Medical Centre, Chinese PLA General Hospital & National Clinical Research Center for Geriatric Diseases, Beijing 100853, China.
| |
Collapse
|
33
|
Chung S, Gouveia Z, Shrestha S, Coles JG, Maynes JT, Santerre JP. Nanoparticles for the Delivery of Pro-regenerative Cardiac Progenitor Secretory Proteins Targeting Cellular Senescence and Vasculogenesis. ACS APPLIED BIO MATERIALS 2025; 8:386-398. [PMID: 39763341 DOI: 10.1021/acsabm.4c01361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
Contemporary therapies following heart failure center on regenerative approaches to account for the loss of cardiomyocytes and limited regenerative capacity of the adult heart. While the delivery of cardiac progenitor cells has been shown to improve cardiac function and repair following injury, recent evidence has suggested that their paracrine effects (or secretome) provides a significant contribution towards modulating regeneration, rather than the progenitor cells intrinsically. The direct delivery of secretory biomolecules, however, remains a challenge due to their lack of stability and tissue retention, limiting their prolonged therapeutic efficacy. We hypothesized that polyurethane-based nanoparticles with heteropolar-hydrophobic-ionic chemistry (DPHI-NPs) could enable the delivery of a subset of pro-regenerative cardiac progenitor cell proteins [bone morphogenetic protein-4 (BMP-4) and angiotensin 1-7 (Ang1-7)] to promote biological pathways conducive to repair processes such as antisenescence (through the quantification of β-galactosidase and interleukin-6) and vasculogenesis (through the formation of endothelial tubes), demonstrated in vitro with human cardiac fibroblasts (hCFs) and human microvascular endothelial cells (hMECs), respectively. DPHI-NPs with a diameter of 190 ± 2 nm (polydispersity index < 0.2) and a zeta potential of -40 ± 1 mV were generated using an emulsion inversion technique and loaded with both therapeutic proteins (BMP-4 and Ang1-7) by optimizing surface charge, loading solution concentration, coating duration, and coating efficiency. Senescence-induced hCFs treated with functionalized DPHI-NPs were found to exhibit a significant reduction in expressed β-galactosidase and IL-6 (p < 0.05). Additionally, hMECs treated with NPBMP-4 were found to display enhanced vasculogenesis compared to control culture conditions alone (p < 0.05). The development of a DPHI-NP vector for the delivery of pro-regenerative secretome biomolecules may present an effective translatable strategy to improve their therapeutic efficacy with respect to cell function.
Collapse
Affiliation(s)
- Shirley Chung
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3E2, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, Ontario M5G 1M1, Canada
| | - Zach Gouveia
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, Ontario M5G 1M1, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, Ontario M5G 1G6, Canada
| | - Suja Shrestha
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, Ontario M5G 1M1, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, Ontario M5G 1G6, Canada
| | - John G Coles
- Division of Cardiovascular Surgery, The Hospital for Sick Children, 170 Elizabeth Street, Toronto, Ontario M5G 1E8, Canada
| | - Jason T Maynes
- Program in Molecular Medicine, SickKids Research Institute, 686 Bay Street, Toronto, Ontario M5G 0A4, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, 170 Elizabeth Street, Toronto, Ontario M5G 1E8, Canada
- Department of Anesthesiology and Pain Medicine, University of Toronto, 170 Elizabeth Street, Toronto, Ontario M5G 1E8, Canada
| | - J Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3E2, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Ave, Toronto, Ontario M5G 1M1, Canada
- Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, Ontario M5G 1G6, Canada
| |
Collapse
|
34
|
Morgado LAL, Rodrigues LMZ, Silva DCF, da Silva BD, Irigoyen MCC, Takano APC. NF-κB-Specific Suppression in Cardiomyocytes Unveils Aging-Associated Responses in Cardiac Tissue. Biomedicines 2025; 13:224. [PMID: 39857807 PMCID: PMC11762954 DOI: 10.3390/biomedicines13010224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Aging is associated with structural and functional changes in the heart, including hypertrophy, fibrosis, and impaired contractility. Cellular mechanisms such as senescence, telomere shortening, and DNA damage contribute to these processes. Nuclear factor kappa B (NF-κB) has been implicated in mediating cellular responses in aging tissues, and increased NF-κB expression has been observed in the hearts of aging rodents. Therefore, NF-κB is suspected to play an important regulatory role in the cellular and molecular processes occurring in the heart during aging. This study investigates the in vivo role of NF-κB in aging-related cardiac alterations, focusing on senescence and associated cellular events. Methods: Young and old wild-type (WT) and transgenic male mice with cardiomyocyte-specific NF-κB suppression (3M) were used to assess cardiac function, morphology, senescence markers, lipofuscin deposition, DNA damage, and apoptosis. Results: Kaplan-Meier analysis revealed reduced survival in 3M mice compared to WT. Echocardiography showed evidence of eccentric hypertrophy, and both diastolic and systolic dysfunction in 3M mice. Both aged WT and 3M mice exhibited cardiac hypertrophy, with more pronounced hypertrophic changes in cardiomyocytes from 3M mice. Additionally, cardiac fibrosis, senescence-associated β-galactosidase activity, p21 protein expression, and DNA damage (marked by phosphorylated H2A.X) were elevated in aged WT and both young and aged 3M mice. Conclusions: The suppression of NF-κB in cardiomyocytes leads to pronounced cardiac remodeling, dysfunction, and cellular damage associated with the aging process. These findings suggest that NF-κB plays a critical regulatory role in cardiac aging, influencing both cellular senescence and molecular damage pathways. This has important implications for the development of therapeutic strategies aimed at mitigating age-related cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Bruno Durante da Silva
- Unidade de Hipertensao, Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-000, Brazil
| | - Maria Claudia Costa Irigoyen
- Unidade de Hipertensao, Instituto do Coracao, Hospital das Clinicas, Faculdade de Medicina, Universidade de Sao Paulo (InCor-HCFMUSP), Sao Paulo 05403-000, Brazil
| | - Ana Paula Cremasco Takano
- Department of Anatomy, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| |
Collapse
|
35
|
Li H, Wei D, Cao H, Han Y, Li L, Liu Y, Qi J, Wu X, Zhang Z. Bioinformatics-Based Exploration of the Ability of Ginkgetin to Alleviate the Senescence of Cardiomyocytes After Myocardial Infarction and Its Cardioprotective Effects. J Inflamm Res 2025; 18:301-323. [PMID: 39802510 PMCID: PMC11724673 DOI: 10.2147/jir.s491535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 12/21/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose Myocardial infarction (MI) is a prevalent cardiovascular disorder affecting individuals worldwide. There is a need to identify more effective therapeutic agents to minimize cardiomyocyte damage and enhance cardioprotection. Ginkgo biloba extract is extensively used to treat neurological disorders and peripheral vascular diseases. The aim of this study was to determine the protective effects and mechanisms of ginkgetin on postinfarction cardiomyocytes through bioinformatics and experimental validation. Methods Bioinformatics analysis was performed to predict the underlying biological mechanisms of ginkgetin in the treatment of MI. Next, we performed further validation through experiments. For in vivo studies, we used coronary ligation to construct an MI rat model. In vitro, oxygen and glucose deprivation (OGD) was performed to simulate ischemia in H9c2 cardiomyocytes. Results Bioinformatics analysis revealed that the key targets of ginkgetin for MI treatment were MMP2, MMP9, and VEGFA. Immune infiltration analysis revealed that ginkgetin might be involved in immune regulation by acting on the TCR signaling pathway. The results of the GO enrichment analysis revealed that ginkgetin might protect the heart by acting on the cell membrane to alleviate the senescent apoptosis of cardiomyocytes after MI. In vivo studies revealed that ginkgetin ameliorated myocardial pathological damage and cardiac decompensation after MI. It also alleviated the inflammatory infiltration and senescent apoptosis of cardiomyocytes after MI. Additionally, ginkgetin can downregulate the activation signals of the TCR signaling pathway by dephosphorylating CD3 and CD28. In vitro studies revealed that ginkgetin attenuated elevated OGD-induced cytotoxicity, increased cell viability, and alleviated OGD-induced senescent apoptosis, thus protecting cardiomyocytes. Conclusion Ginkgetin inhibits postinfarction myocardial fibrosis and cardiomyocyte hypertrophy, scavenges oxygen free radicals, decreases postinfarction limbic cell inflammatory infiltration, suppresses activation of the inflammatory-immune pathway, and delays postinfarction peripheral cells from undergoing senescent apoptosis, thus protecting the heart.
Collapse
Affiliation(s)
- Han Li
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Dongsheng Wei
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Huimin Cao
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Yelei Han
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Luzhen Li
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Yuting Liu
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Jiajie Qi
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Xinyue Wu
- The First School of Clinical Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
| | - Zhe Zhang
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang, 110847, People’s Republic of China
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110032, People’s Republic of China
| |
Collapse
|
36
|
Cummings SR, Lui LY, Zaira A, Mau T, Fielding RA, Atkinson EJ, Patel S, LeBrasseur N. Biomarkers of cellular senescence and major health outcomes in older adults. GeroScience 2024:10.1007/s11357-024-01474-9. [PMID: 39695064 DOI: 10.1007/s11357-024-01474-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
The geroscience hypothesis proposes that underlying biological processes, such as the accumulation of senescent cells, have deleterious effects on multiple tissues and increase the risk of many chronic conditions with aging. Senescent cells produce heterogenous biomarkers, also called senescence-associated secretory phenotype (SASP). Circulating concentrations of senescence biomarkers may reflect an underlying burden of senescent cells in various tissues. Plasma levels of these proteins have been associated with increased mortality and poorer physical function. The associations of them with the incidence of major age-related conditions including heart failure, cardiovascular disease, stroke, and dementia, have not been studied. We measured 35 senescence biomarkers in baseline plasma samples from 1678 participants aged 70-79 years old in the longitudinal Health ABC cohort study. Clinical outcomes were ascertained and validated over an average 11.5 year follow-up. In models adjusted for age, sex, and race, higher levels of most of senescence biomarkers were associated with increased risk of all-cause mortality, mobility limitation, and heart failure. Several were also associated with an increased risk of coronary heart disease, stroke, and dementia. Very few were associated with the risk of cancer. Proteins that were selected by Lasso regression for each outcome that commonly included GDF15 and IL6, significantly improved the prediction of mortality, mobility limitation, and heart failure compared with age, sex, and race alone. These results indicate that levels of senescence biomarkers predict an increased risk of several age-related clinical outcomes and may identify individuals most likely to benefit from senotherapeutics.
Collapse
Affiliation(s)
- Steven R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA.
| | - Li-Yung Lui
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Aversa Zaira
- Robert and Arlene Kogod Center on Aging, Paul F. Glenn Center for the Biology of Aging Research, and Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| | - Theresa Mau
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Roger A Fielding
- Metabolism and Basic Biology of Aging Directive, Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Medford, MA, USA
| | | | - Sheena Patel
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Nathan LeBrasseur
- Robert and Arlene Kogod Center on Aging, Paul F. Glenn Center for the Biology of Aging Research, and Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
37
|
Wen Y, Zhang X, Liu H, Ye H, Wang R, Ma C, Duo T, Wang J, Yang X, Yu M, Wang Y, Wu L, Zhao Y, Wang L. SGLT2 inhibitor downregulates ANGPTL4 to mitigate pathological aging of cardiomyocytes induced by type 2 diabetes. Cardiovasc Diabetol 2024; 23:430. [PMID: 39633372 PMCID: PMC11619200 DOI: 10.1186/s12933-024-02520-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Senescence is recognized as a principal risk factor for cardiovascular diseases, with a significant association between the senescence of cardiomyocytes and inferior cardiac function. Furthermore, type 2 diabetes exacerbates this aging process. Sodium-glucose co-transporter 2 inhibitor (SGLT2i) has well-established cardiovascular benefits and, in recent years, has been posited to possess anti-aging properties. However, there are no reported data on their improvement of cardiomyocytes function through the alleviation of aging. Consequently, our study aims to investigate the mechanism by which SGLT2i exerts anti-aging and protective effects at the cardiac level through its action on the FOXO1-ANGPTL4 pathway. METHODS To elucidate the underlying functions and mechanisms, we established both in vivo and in vitro disease models, utilizing mice with diabetic cardiomyopathy (DCM) induced by type 2 diabetes mellitus (T2DM) through high-fat diet combined with streptozotocin (STZ) administration, and AC16 human cardiomyocyte cell subjected to stimulation with high glucose (HG) and palmitic acid (PA). These models were employed to assess the changes in the senescence phenotype of cardiomyocytes and cardiac function following treatment with SGLT2i. Concurrently, we identified ANGPTL4, a key factor contributing to senescence in DCM, using RNA sequencing (RNA-seq) technology and bioinformatics methods. We further clarified ANGPTL4 role in promoting pathological aging of cardiomyocytes induced by hyperglycemia and hyperlipidemia through knockdown and overexpression of the factor, as well as analyzed the impact of SGLT2i intervention on ANGPTL4 expression. Additionally, we utilized chromatin immunoprecipitation followed by quantitative real-time PCR (ChIP-qPCR) to confirm that FOXO1 is essential for the transcriptional activation of ANGPTL4. RESULTS The therapeutic intervention with SGLT2i alleviated the senescence phenotype in cardiomyocytes of the DCM mouse model constructed by high-fat feeding combined with STZ, as well as in the AC16 model stimulated by HG and PA, while also improving cardiac function in DCM mice. We observed that the knockdown of ANGPTL4, a key senescence-promoting factor in DCM identified through RNA-seq technology and bioinformatics, mitigated the senescence of cardiomyocytes, whereas overexpression of ANGPTL4 exacerbated it. Moreover, SGLT2i improved the senescence phenotype by suppressing the overexpression of ANGPTL4. In fact, we discovered that SGLT2i exert their effects by regulating the upstream transcription factor FOXO1 of ANGPTL4. Under conditions of hyperglycemia and hyperlipidemia, compared to the control group without FOXO1, the overexpression of FOXO1 in conjunction with SGLT2i intervention significantly reduced both ANGPTL4 mRNA and protein levels. This suggests that the FOXO1-ANGPTL4 axis may be a potential target for the cardioprotective effects of SGLT2i. CONCLUSIONS Collectively, our study demonstrates that SGLT2i ameliorate the pathological aging of cardiomyocytes induced by a high glucose and high fat metabolic milieu by regulating the interaction between FOXO1 and ANGPTL4, thereby suppressing the transcriptional synthesis of the latter, and consequently restoring cardiac function.
Collapse
MESH Headings
- Sodium-Glucose Transporter 2 Inhibitors/pharmacology
- Sodium-Glucose Transporter 2 Inhibitors/therapeutic use
- Diabetic Cardiomyopathies/blood
- Diabetic Cardiomyopathies/drug therapy
- Diabetic Cardiomyopathies/etiology
- Diabetic Cardiomyopathies/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/pathology
- Streptozocin/toxicity
- Diet, High-Fat/adverse effects
- Angiopoietin-Like Protein 4/antagonists & inhibitors
- Angiopoietin-Like Protein 4/genetics
- Angiopoietin-Like Protein 4/metabolism
- Cell Line
- Humans
- Animals
- Mice
- Cellular Senescence/drug effects
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Gene Knockdown Techniques
- Mice, Inbred C57BL
- Male
- Forkhead Box Protein O1/metabolism
- Signal Transduction/drug effects
- Signal Transduction/genetics
- Down-Regulation/drug effects
- Blood Glucose/metabolism
Collapse
Affiliation(s)
- Yun Wen
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xiaofang Zhang
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, Jinan University, Guangzhou, China
| | - Han Liu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Haowen Ye
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ruxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Caixia Ma
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Tianqi Duo
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Jiaxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Xian Yang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Meixin Yu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Liangyan Wu
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yongting Zhao
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lihong Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China.
- The Academician Cooperative Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital, Jinan University, Guangzhou, China.
| |
Collapse
|
38
|
Amin A, Mohajerian A, Ghalehnoo SR, Mohamadinia M, Ahadi S, Sohbatzadeh T, Pazoki M, Hasanvand A, Faghihkhorasani F, Habibi Z. Potential Player of Platelet in the Pathogenesis of Cardiotoxicity: Molecular Insight and Future Perspective. Cardiovasc Toxicol 2024; 24:1381-1394. [PMID: 39397196 DOI: 10.1007/s12012-024-09924-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 09/18/2024] [Indexed: 10/15/2024]
Abstract
Cancer patients may encounter the onset of cardiovascular disease due to tumor advancement or chemotherapy, commonly known as "cardiotoxicity." In this respect, the conventional chemotherapy treatment protocol involves a mixture of different medications. These medications can be detrimental to cardiac tissue, consequently exposing the patient to the possibility of irreversible cardiac injury. The enhancement of oxidative stress and inflammation is an important mechanism of chemotherapeutic agents for developing cardiotoxicity. Regarding their dual pro- and anti-inflammatory functions, platelets can significantly influence the progression or suppression of cardiotoxicity. Therefore, the expression of platelet activatory markers can serve as valuable prognostic indicators for cardiotoxicity. The primary objective of this study is to examine the significance of platelets in cardiotoxicity and explore potential strategies that could effectively target malignant cells while minimizing their cytotoxic impact, such as cardiotoxicity and thrombosis.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Ahmad Mohajerian
- Department of Emergency Medicine, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sara Rashki Ghalehnoo
- Department of Cardiology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Mehdi Mohamadinia
- Department of Dental Prosthesis, School of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Shana Ahadi
- School of Medicine, Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Science, Alborz, Iran
| | - Mahboubeh Pazoki
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Hasanvand
- Department of General Surgery, Lorestan University of Medical Science, Khorramabad, Iran
| | | | - Zeinab Habibi
- Lorestan University of Medical Science, Lorestan, Iran.
| |
Collapse
|
39
|
Lin Z, Wu C, Song D, Zhu C, Wu B, Wang J, Xue Y. Sarmentosin alleviates doxorubicin-induced cardiotoxicity and ferroptosis via the p62-Keap1-Nrf2 pathway. Redox Rep 2024; 29:2392329. [PMID: 39150892 PMCID: PMC11332294 DOI: 10.1080/13510002.2024.2392329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2024] Open
Abstract
Doxorubicin (Dox) is extensively used as an antitumor agent, but its severe cardiotoxicity significantly limits its clinical use. Current treatments for Dox-induced cardiotoxicity are inadequate, necessitating alternative solutions. This study evaluated the effects of sarmentosin, a compound from Sedum sarmentosum, on Dox-induced cardiotoxicity and dysfunction. Sarmentosin was administered as a pretreatment to both mice and H9c2 cells before Dox exposure. Subsequently, markers of Dox-induced cardiotoxicity and ferroptosis in serum and cell supernatants were measured. Western blot analysis was utilized to detect levels of ferroptosis, oxidative stress, and autophagy proteins. Additionally, echocardiography, hematoxylin-eosin staining, ROS detection, and immunofluorescence techniques were employed to support our findings. Results demonstrated that sarmentosin significantly inhibited iron accumulation, lipid peroxidation, and oxidative stress, thereby reducing Dox-induced ferroptosis and cardiotoxicity in C57BL/6 mice and H9c2 cells. The mechanism involved the activation of autophagy and the Nrf2 signaling pathway. These findings suggest that sarmentosin may prevent Dox-induced cardiotoxicity by mitigating ferroptosis. The study underscores the potential of compounds like sarmentosin in treating Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhihui Lin
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Chang Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Dongyan Song
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Chenxi Zhu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Bosen Wu
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jie Wang
- Department of Endocrinology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Yangjing Xue
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children’s Hospital, Wenzhou Medical University, Wenzhou, People’s Republic of China
| |
Collapse
|
40
|
Huang TS, Wu T, Fu XL, Ren HL, He XD, Zheng DH, Tan J, Shen CH, Xiong SJ, Qian J, Zou Y, Wan JH, Ji YJ, Liu MY, Wu YD, Li XH, Li H, Zheng K, Yang XF, Wang H, Ren M, Cai WB. SREBP1 induction mediates long-term statins therapy related myocardial lipid peroxidation and lipid deposition in TIIDM mice. Redox Biol 2024; 78:103412. [PMID: 39476450 PMCID: PMC11555471 DOI: 10.1016/j.redox.2024.103412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 11/15/2024] Open
Abstract
Statins therapy is efficacious in diminishing the risk of major cardiovascular events in diabetic patients. However, our research has uncovered a correlation between the prolonged administration of statins and an elevated risk of myocardial dysfunction in patients with type II diabetes mellitus (TIIDM). Here, we report the induction of sterol regulatory element-binding protein 1 (SREBP1) activation, associated lipid peroxidation, and the consequent diabetic myocardial dysfunction after statin treatment and explored the underlying mechanisms. In db/db mice, we observed that 40 weeks atorvastatin (5 and 10 mg/kg) and rosuvastatin (20 mg/kg) administration exacerbated diabetic myocardial dysfunction by echocardiography and cardiomyocyte contractility assay, increased myocardial inflammation and fibrosis as shown by CD68, IL-1β, Masson's staining and Collagen1A1 immunohistochemistry (IHC) staining, increased respiratory exchange ratio (RER) by metabolic cage system assessment, exacerbated mitochondrial structural pathological changes by transmission electron microscopy (TEM) examination, increased deposition of lipid and glycogen by TEM, Oil-red and periodic acid-schiff stain (PAS) staining, which were corresponded with augmented levels of myocardial SREBP1 protein and lipid peroxidation marked by 4-hydroxynonenal (4-HNE) staining. Comparable myocardial fibrosis was also observed in KK-ay and low-dose streptozotocin (STZ)-induced TIIDM mice. Elevated SREBP1 levels were observed in the heart tissues from diabetic patients, which was positively correlated with their myocardial dysfunction. To elucidate the role of statin induced SREBP1 in lipid peroxidation and lipid deposition and related mechanism, we cultured neonatal mouse primary cardiomyocytes (NMPCs) and treated them with atorvastatin (10 μM, 24 h), tracing with [U-13C]-glucose and evaluating for SREBP1 expression and localization. We found that statin treatment elevated de novo lipogenesis (DNL) and the levels of SREBP1 cleavage-activating protein (SCAP), reduced the interaction of SCAP with insulin-induced gene 1 (Insig1), and enhance SCAP/SREBP1 translocation to the Golgi, which facilitate SREBP1 cleavage leading to its nuclear trans-localization and activation in NMPCs. Ultimately, SREBP1 knockdown or l-carnitine mitigated long-term statins therapy induced lipid peroxidation and myocardial fibrosis in low-dose STZ treated SREBP1+/- mice and l-carnitine treated db/db mice. In conclusion, we demonstrated that statin therapy may augment DNL by activating SREBP1, resulting in myocardial lipid peroxidation and lipid deposition.
Collapse
Affiliation(s)
- Tong-Sheng Huang
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Teng Wu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Xin-Lu Fu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Hong-Lin Ren
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Xiao-Dan He
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Ding-Hao Zheng
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China; Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jing Tan
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Cong-Hui Shen
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Shi-Jie Xiong
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Jiang Qian
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Yan Zou
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Jun-Hong Wan
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Yuan-Jun Ji
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Meng-Ying Liu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Yan-di Wu
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Xing-Hui Li
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China
| | - Hui Li
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China
| | - Kai Zheng
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, Guangdong, PR China
| | - Xiao-Feng Yang
- Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Hong Wang
- Metabolic Disease Research, Department of Cardiovascular Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
| | - Meng Ren
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, PR China.
| | - Wei-Bin Cai
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Guangzhou, 510080, Guangdong, PR China; Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, PR China.
| |
Collapse
|
41
|
Shen YH, Ding D, Lian TY, Qiu BC, Yan Y, Wang PW, Zhang WH, Jing ZC. Panorama of artery endothelial cell dysfunction in pulmonary arterial hypertension. J Mol Cell Cardiol 2024; 197:61-77. [PMID: 39437884 DOI: 10.1016/j.yjmcc.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal lung disease characterized by progressive pulmonary vascular remodeling. The initial cause of pulmonary vascular remodeling is the dysfunction of pulmonary arterial endothelial cells (PAECs), manifested by changes in the categorization of cell subtypes, endothelial programmed cell death, such as apoptosis, necroptosis, pyroptosis, ferroptosis, et al., overproliferation, senescence, metabolic reprogramming, endothelial-to-mesenchymal transition, mechanosensitivity, and regulation ability of peripheral cells. Therefore, it is essential to explore the mechanism of endothelial dysfunction in the context of PAH. This review aims to provide a comprehensive understanding of the molecular mechanisms underlying endothelial dysfunction in PAH. We highlight the developmental process of PAECs and changes in PAH and summarise the latest classification of endothelial dysfunction. Our review could offer valuable insights into potential novel EC-specific targets for preventing and treating PAH.
Collapse
Affiliation(s)
- Ying-Huizi Shen
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China
| | - Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Yan
- Heart Center and Shanghai Institute of Pediatric Congenital Heart Disease, Shanghai Children's Medical Center, National Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pei-Wen Wang
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei-Hua Zhang
- Department of Cardiovascular Medicine, The First Hospital of Jilin University, Changchun, China.
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
42
|
Zhou Z, Li M, Zhang Z, Song Z, Xu J, Zhang M, Gong M. Overview of Panax ginseng and its active ingredients protective mechanism on cardiovascular diseases. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118506. [PMID: 38964625 DOI: 10.1016/j.jep.2024.118506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/06/2024]
Abstract
ETHNIC PHARMACOLOGICAL RELEVANCE Panax ginseng is a traditional Chinese herbal medicine used to treat cardiovascular diseases (CVDs), and it is still widely used to improve the clinical symptoms of various CVDs. However, there is currently a lack of summary and analysis on the mechanism of Panax ginseng exerts its cardiovascular protective effects. This article provides a review of in vivo and in vitro pharmacological studies on Panax ginseng and its active ingredients in reducing CVDs damage. AIM OF THIS REVIEW This review summarized the latest literature on Panax ginseng and its active ingredients in CVDs research, aiming to have a comprehensive and in-depth understanding of the cardiovascular protection mechanism of Panax ginseng, and to provide new ideas for the treatment of CVDs, as well as to optimize the clinical application of Panax ginseng. METHODS Enrichment of pathways and biological terms using the traditional Chinese medicine molecular mechanism bioinformatics analysis tool (BATMAN-TCM). The literature search is based on electronic databases such as PubMed, ScienceDirect, Scopus, CNKI, with a search period of 2002-2023. The search terms include Panax ginseng, Panax ginseng ingredients, ginsenosides, ginseng polysaccharides, ginseng glycoproteins, ginseng volatile oil, CVDs, heart, and cardiac. RESULTS 132 articles were ultimately included in the review. The ingredients in Panax ginseng that manifested cardiovascular protective effects are mainly ginsenosides (especially ginsenoside Rb1). Ginsenosides protected against CVDs such as ischemic reperfusion injury, atherosclerosis and heart failure mainly through improving energy metabolism, inhibiting hyper-autophagy, antioxidant, anti-inflammatory and promoting secretion of exosomes. CONCLUSION Panax ginseng and its active ingredients have a particularly prominent effect on improving myocardial energy metabolism remodeling in protecting against CVDs. The AMPK and PPAR signaling pathways are the key targets through which Panax ginseng produces multiple mechanisms of cardiovascular protection. Extracellular vesicles and nanoparticles as carriers are potential delivery ways for optimizing the bioavailability of Panax ginseng and its active ingredients.
Collapse
Affiliation(s)
- Ziwei Zhou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Meijing Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Zekuan Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Zhimin Song
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Jingjing Xu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, 100069, China
| | - Minyu Zhang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, 100069, China.
| | - Muxin Gong
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Traditional Chinese Medicine Collateral Disease Theory Research, Beijing, 100069, China.
| |
Collapse
|
43
|
He S, Yan L, Yuan C, Li W, Wu T, Chen S, Li N, Wu M, Jiang J. The role of cardiomyocyte senescence in cardiovascular diseases: A molecular biology update. Eur J Pharmacol 2024; 983:176961. [PMID: 39209099 DOI: 10.1016/j.ejphar.2024.176961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/18/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, and advanced age is a main contributor to the prevalence of CVD. Cellular senescence is an irreversible state of cell cycle arrest that occurs in old age or after cells encounter various stresses. Senescent cells not only result in the reduction of cellular function, but also produce senescence-associated secretory phenotype (SASP) to affect surrounding cells and tissue microenvironment. There is increasing evidence that the gradual accumulation of senescent cardiomyocytes is causally involved in the decline of cardiovascular system function. To highlight the role of senescent cardiomyocytes in the pathophysiology of age-related CVD, we first introduced that senescent cardiomyoyctes can be identified by structural changes and several senescence-associated biomarkers. We subsequently provided a comprehensive summary of existing knowledge, outlining the compelling evidence on the relationship between senescent cardiomyocytes and age-related CVD phenotypes. In addition, we discussed that the significant therapeutic potential represented by the prevention of accelerated senescent cardiomyocytes, and the current status of some existing geroprotectors in the prevention and treatment of age-related CVD. Together, the review summarized the role of cardiomyocyte senescence in CVD, and explored the molecular knowledge of senescent cardiomyocytes and their potential clinical significance in developing senescent-based therapies, thereby providing important insights into their biology and potential therapeutic exploration.
Collapse
Affiliation(s)
- Shuangyi He
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Yan
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Department of Pharmacy, Wuhan Asia General Hospital, Wuhan, 430056, China
| | - Chao Yuan
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Wenxuan Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Tian Wu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Suya Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Niansheng Li
- Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, China
| | - Meiting Wu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Department of Nephrology, Institute of Nephrology, 2nd Affiliated Hospital of Hainan Medical University, Haikou, 570100, China
| | - Junlin Jiang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410078, China; Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, 410078, China.
| |
Collapse
|
44
|
Liu J, Tan G, Wang S, Tong B, Wu Y, Zhang L, Jiang B. Artesunate induces HO-1-mediated cell cycle arrest and senescence to protect against ocular fibrosis. Int Immunopharmacol 2024; 141:112882. [PMID: 39151383 DOI: 10.1016/j.intimp.2024.112882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Recent research found artesunate could inhibit ocular fibrosis; however, the underlying mechanisms are not fully known. Since the ocular fibroblast is the main effector cell in fibrosis, we hypothesized that artesunate may exert its protective effects by inhibiting the fibroblasts proliferation. TGF-β1-induced ocular fibroblasts and glaucoma filtration surgery (GFS)-treated rabbits were used as ocular fibrotic models. Firstly, we analyzed fibrosis levels by assessing the expression of fibrotic marker proteins, and used Ki67 immunofluorescence, EdU staining, flow cytometry to determine cell cycle status, and SA-β-gal staining to assess cellular senescence levels. Then to predict target genes and pathways of artesunate, we analyzed the differentially expressed genes and enriched pathways through RNA-seq. Western blot and immunohistochemistry were used to detect the pathway-related proteins. Additionally, we validated the dependence of artesunate's effects on HO-1 expression through HO-1 siRNA. Moreover, DCFDA and MitoSOX fluorescence staining were used to examine ROS level. We found artesunate significantly inhibits the expression of fibrosis-related proteins, induces cell cycle arrest and cellular senescence. Knocking down HO-1 in fibroblasts with siRNA reverses these regulatory effects of artesunate. Mechanistic studies show that artesunate significantly inhibits the activation of the Cyclin D1/CDK4-pRB pathway, induces an increase in cellular and mitochondrial ROS levels and activates the Nrf2/HO-1 pathway. In conclusion, the present study identifies that artesunate induces HO-1 expression through ROS to activate the antioxidant Nrf2/HO-1 pathway, subsequently inhibits the cell cycle regulation pathway Cyclin D1/CDK4-pRB in an HO-1-dependent way, induces cell cycle arrest and senescence, and thereby resists periorbital fibrosis.
Collapse
Affiliation(s)
- Jingyuan Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Guangshuang Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Shutong Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Boding Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Ying Wu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China.
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China.
| |
Collapse
|
45
|
Song L, Qiu Q, Ju F, Zheng C. Mechanisms of doxorubicin-induced cardiac inflammation and fibrosis; therapeutic targets and approaches. Arch Biochem Biophys 2024; 761:110140. [PMID: 39243924 DOI: 10.1016/j.abb.2024.110140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Doxorubicin plays a pivotal role in the treatment of various malignancies. Despite its efficacy, the cardiotoxicity associated with doxorubicin limits its clinical utility. The cardiotoxic nature of doxorubicin is attributed to several mechanisms, including its interference with mitochondrial function, the generation of reactive oxygen species (ROS), and the subsequent damage to cardiomyocyte DNA, proteins, and lipids. Furthermore, doxorubicin disrupts the homeostasis of cardiac-specific transcription factors and signaling pathways, exacerbating cardiac dysfunction. Oxidative stress, cell death, and other severe changes, such as mitochondrial dysfunction, activation of pro-oxidant enzymes, the renin-angiotensin system (RAS), endoplasmic reticulum (ER) stress, and infiltration of immune cells in the heart after treatment with doxorubicin, may cause inflammatory and fibrotic responses. Fibrosis and inflammation can lead to a range of disorders in the heart, resulting in potential cardiac dysfunction and disease. Various adjuvants have shown potential in preclinical studies to mitigate these challenges associated with cardiac inflammation and fibrosis. Antioxidants, plant-based products, specific inhibitors, and cardioprotective drugs may be recommended to alleviate cardiotoxicity. This review explores the complex mechanisms of doxorubicin-induced heart inflammation and fibrosis, identifies possible cellular and molecular targets, and investigates potential substances that could help reduce these harmful effects.
Collapse
Affiliation(s)
- Linghua Song
- Department of Pharmacy, Yantai Mountain Hospital, Yantai City, Shandong Province, 264001, China
| | - Qingzhuo Qiu
- Medical Imaging Department of Qingdao Women and Children's Hospital, 266000, China
| | - Fei Ju
- Department of Critical Care, Medicine East Hospital of Qingdao Municipal Hospital, 266000, China
| | - Chunyan Zheng
- Cadre Health Office of Zibo Central Hospital in Shandong Province, 255000, China.
| |
Collapse
|
46
|
Fu ZP, Ying YG, Wang RY, Wang YQ. Aged gut microbiota promotes arrhythmia susceptibility via oxidative stress. iScience 2024; 27:110888. [PMID: 39381749 PMCID: PMC11460473 DOI: 10.1016/j.isci.2024.110888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 05/24/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024] Open
Abstract
Arrhythmias and sudden cardiac death (SCD) impose a significant burden. Their prevalence rises with age and is linked to gut dysbiosis. Our study aimed to determine whether aged gut microbiota affects arrhythmogenesis. Here, we demonstrated that arrhythmia susceptibility in aged mice could be transmitted to young mice using fecal microbiota transplantation (FMT). Mechanistically, increased intestinal reactive oxygen species (ROS) in aged mice reduced ion channel protein expression and promoted arrhythmias. Gut microbiota depletion by an antibiotic cocktail reduced ROS and arrhythmia in aged mice. Interestingly, oxidative stress in heart induced by hydrogen peroxide (H2O2) increased arrhythmia. Moreover, aged gut microbiota could induce oxidative stress in young mice colon by gut microbiota metabolites transplantation. Vitexin could reduce aging and arrhythmia through OLA1-Nrf2 signaling pathway. Overall, our study demonstrated that the gut microbiota of aged mice reduced cardiac ion channel protein expression through systemic oxidative stress, thereby increased the risk of arrhythmias.
Collapse
Affiliation(s)
- Zhi-ping Fu
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yi-ge Ying
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Rui-yao Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| | - Yu-qing Wang
- Collage of Pharmacology, North China University of Science and Technology, Tangshan 063200, China
| |
Collapse
|
47
|
Tao P, Zhang HF, Zhou P, Wang YL, Tan YZ, Wang HJ. Growth differentiation factor 11 alleviates oxidative stress-induced senescence of endothelial progenitor cells via activating autophagy. Stem Cell Res Ther 2024; 15:370. [PMID: 39420391 PMCID: PMC11488219 DOI: 10.1186/s13287-024-03975-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Stem cell transplantation has been regarded as a promising therapeutic strategy for myocardial regeneration after myocardial infarction (MI). However, the survival and differentiation of the transplanted stem cells in the hostile ischaemic and inflammatory microenvironment are poor. Recent studies have focused on enhancing the survival and differentiation of the stem cells, while strategies to suppress the senescence of the transplanted stem cells is unknown. Therefore, we investigated the effect of growth differentiation factor 11 (GDF11) on attenuating oxidative stress-induced senescence in the engrafted endothelial progenitor cells (EPCs). METHODS Rat models of oxidative stress were established by hydrogen peroxide conditioning. Oxidative stress-induced senescence was assessed through senescence-associated β-galactosidase expression and lipofuscin accumulation. The effects of GDF11 treatment on senescence and autophagy of EPCs were evaluated 345, while improvement of myocardial regeneration, neovascularization and cardiac function were examined following transplantation of the self-assembling peptide (SAP) loaded EPCs and GDF11 in the rat MI models. RESULTS Following hydrogen peroxide conditioning, the level of ROS in EPCs decreased significantly upon treatment with GDF11. This resulted in reduction in the senescent cells and lipofuscin particles, as well as the damaged mitochondria and rough endoplasmic reticula. Concurrently, there was a significant increase in LC3-II expression, LC3-positive puncta and the presence of autophagic ultrastructures were increased significantly. The formulated SAP effectively adhered to EPCs and sustained the release of GDF11. Transplantation of SAP-loaded EPCs and GDF11 into the ischaemic abdominal pouch or myocardium resulted in a decreased number of the senescent EPCs. At four weeks after transplantation into the myocardium, neovascularization and myocardial regeneration were enhanced, reverse myocardial remodeling was attenuated, and cardiac function was improved effectively. CONCLUSIONS This study provides novel evidence suggesting that oxidative stress could induce senescence of the transplanted EPCs in the ischemic myocardium. GDF11 demonstrates the ability to mitigate oxidative stress-induced senescence in the transplanted EPCs within the myocardium by activating autophagy.
Collapse
Affiliation(s)
- Ping Tao
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
- Department of Laboratory Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200086, People's Republic of China
| | - Hai-Feng Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Pei Zhou
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Yong-Li Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China
| | - Yu-Zhen Tan
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
- Rehabilitation Therapy Department, School of Health Sciences, West Yunnan University of Applied Sciences, Dali, Yunnan Province, 671000, People's Republic of China.
| | - Hai-Jie Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, 138 Yixueyuan Road, Shanghai, 200032, People's Republic of China.
- Rehabilitation Therapy Department, School of Health Sciences, West Yunnan University of Applied Sciences, Dali, Yunnan Province, 671000, People's Republic of China.
| |
Collapse
|
48
|
Zhen K, Wei X, Zhi Z, Shang S, Zhang S, Xu Y, Fu X, Cheng L, Yao J, Li Y, Chen X, Liu P, Zhang H. Circulating Extracellular Vesicles from Heart Failure Patients Inhibit Human Cardiomyocyte Activities. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10571-1. [PMID: 39384702 DOI: 10.1007/s12265-024-10571-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Extracellular vesicles (EVs) have been implicated in cardiac remodeling during heart failure (HF). However, the role of circulating EVs (CEVs) in the process of HF is poorly understood. To elucidate the molecular mechanism associated with CEVs in the context of HF, the proteome of 4D label-free EVs from plasma samples was identified. Among the identified proteins, 6 exhibited upregulation while 9 demonstrated downregulation in CEVs derived from HF patients (HCEVs) compared to healthy controls (NCEVs). Our results showed that up-regulated proteins mainly participate in the primary metabolic, glycerolipid metabolic processes, oxidation-reduction process, and inflammatory amplification. In contrast, the down-regulated proteins influenced cell development, differentiation, and proliferation. Compared to NCEVs, HCEVs significantly induced inflammation and triacylglycerol (TAG) accumulation in human cardiomyocytes (HCMs) in vitro. They also compromised their regenerative capacities, triggered endoplasmic reticulum (ER) stress and increased autophagy in HCMs. Further, HCEVs induced differentiation of human cardiac fibroblasts (HCFs), amplifying pro-inflammatory, and pro-fibrotic factors, and enhancing extracellular matrix deposition. Notably, HCEVs are also associated with an increase in the HF biomarker MMP9 within HCFs and demonstrate a negative correlation with autophagic flux. In conclusion, HCEVs appear pivotal in advancing HF via pathological cardiac remodeling.
Collapse
Affiliation(s)
- Ke Zhen
- Beijing Anzhen Hospital, Capital Medical University, Beijing, 100011, China
| | - Xiaojuan Wei
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Zelun Zhi
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shiyu Shang
- The First Clinical Medical College, Hebei North University, Zhangjiakou, 075132, China
| | - Shuyan Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yilu Xu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Xiaochuan Fu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Linjia Cheng
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, 650500, China
| | - Jing Yao
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Yue Li
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Xia Chen
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China
| | - Pingsheng Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongchao Zhang
- Department of Cardiovascular Surgery, Air Force Medical Center, PLA, Beijing, 100048, China.
| |
Collapse
|
49
|
Kieronska-Rudek A, Kij A, Bar A, Kurpinska A, Mohaissen T, Grosicki M, Stojak M, Sternak M, Buczek E, Proniewski B, Kuś K, Suraj-Prazmowska J, Panek A, Pietrowska M, Zapotoczny S, Shanahan CM, Szabo C, Chlopicki S. Phylloquinone improves endothelial function, inhibits cellular senescence, and vascular inflammation. GeroScience 2024; 46:4909-4935. [PMID: 38980631 PMCID: PMC11336140 DOI: 10.1007/s11357-024-01225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/24/2024] [Indexed: 07/10/2024] Open
Abstract
Phylloquinon (PK) and menaquinones (MK) are both naturally occurring compounds belonging to vitamin K group. Present study aimed to comprehensively analyze the influence of PK in several models of vascular dysfunction to determine whether PK has vasoprotective properties, similar to those previously described for MK. Effects of PK and MK on endothelial dysfunction were studied in ApoE/LDLR-/- mice in vivo, in the isolated aorta incubated with TNF, and in vascular cells as regard inflammation and cell senescence (including replicative and stress-induced models of senescence). Moreover, the vascular conversion of exogenous vitamins to endogenous MK-4 was analyzed. PK, as well as MK, given for 8 weeks in diet (10 mg/kg) resulted in comparable improvement in endothelial function in the ApoE/LDLR-/- mice. Similarly, PK and MK prevented TNF-induced impairment of endothelium-dependent vasorelaxation in the isolated aorta. In in vitro studies in endothelial and vascular smooth muscle cells, we identified that both PK and MK displayed anti-senescence effects via decreasing DNA damage while in endothelial cells anti-inflammatory activity was ascribed to the modulation of NFκB activation. The activity of PK and MK was comparable in terms of their effect on senescence and inflammation. Presence of endogenous synthesis of MK-4 from PK in aorta and endothelial and smooth muscle cells suggests a possible involvement of MK in vascular effects of PK. In conclusion, PK and MK display comparable vasoprotective effects, which may be ascribed, at least in part, to the inhibition of cell senescence and inflammation. The vasoprotective effect of PK in the vessel wall can be related to the direct effects of PK, as well as to the action of MK formed from PK in the vascular wall.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marek Grosicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Elżbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Kuś
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Panek
- Institute of Nuclear Physics Polish Academy of Sciences, Krakow, Poland
| | - Monika Pietrowska
- Centre for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland
| | - Szczepan Zapotoczny
- Department of Physical Chemistry and Electrochemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Catherine M Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, James Black Centre, King's College London, London, UK
| | - Csaba Szabo
- Chair of Pharmacology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
50
|
Wang SY, Wang YJ, Dong MQ, Li GR. Acacetin is a Promising Drug Candidate for Cardiovascular Diseases. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1661-1692. [PMID: 39347953 DOI: 10.1142/s0192415x24500654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Phytochemical flavonoids have been proven to be effective in treating various disorders, including cardiovascular diseases. Acacetin is a natural flavone with diverse pharmacological effects, uniquely including atrial-selective anti-atrial fibrillation (AF) via the inhibition of the atrial specific potassium channel currents [Formula: see text] (ultra-rapidly delayed rectifier potassium current), [Formula: see text] (acetylcholine-activated potassium current), [Formula: see text] (calcium-activated small conductance potassium current), and [Formula: see text] (transient outward potassium current). [Formula: see text] inhibition by acacetin, notably, suppresses experimental J-wave syndromes. In addition, acacetin provides extensive cardiovascular protection against ischemia/reperfusion injury, cardiomyopathies/heart failure, autoimmune myocarditis, pulmonary artery hypertension, vascular remodeling, and atherosclerosis by restoring the downregulated intracellular signaling pathway of Sirt1/AMPK/PGC-1α followed by increasing Nrf2/HO-1/SOD thereby inhibiting oxidation, inflammation, and apoptosis. This review provides an integrated insight into the capabilities of acacetin as a drug candidate for treating cardiovascular diseases, especially atrial fibrillation and cardiomyopathies/heart failure.
Collapse
Affiliation(s)
- Shu-Ya Wang
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611137, P. R. China
| | - Ya-Jing Wang
- Department of Pharmacy, School of Pharmacy, Changzhou University Changzhou, Jiangsu 213164, P. R. China
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu 210032, P. R. China
| | - Ming-Qing Dong
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and Translation, Chengdu Fifth People's Hospital (The Second Clinical Medical College, Affiliated Fifth People's Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu 611137, P. R. China
| | - Gui-Rong Li
- Nanjing Amazigh Pharma Limited, Nanjing, Jiangsu 210032, P. R. China
| |
Collapse
|