1
|
Zhang M, Sha Y, Wang J, Qi H, Shi P, Liu Y, Jiang M, Ba L, Liu Y, Cao Y, Zhang Q, Sun H. Inhibition of ULK1 attenuates ferroptosis-mediated cardiac hypertrophy via HMGA2/METTL14/SLC7A11 axis in mice. Eur J Pharmacol 2025; 995:177416. [PMID: 39993699 DOI: 10.1016/j.ejphar.2025.177416] [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: 06/26/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
UNC-51-like kinase 1 (ULK1), a primary serine/threonine kinase, is implicated in diverse pathophysiological processes. Previous findings have linked ULK1-dependent autophagy to cardiac hypertrophy. Our study further explored the functional role and molecular mechanisms of ULK1 in non-autophagic signaling pathways. Notably, ULK1 expression was significantly elevated in both transverse aortic constriction (TAC)-induced hypertrophic mouse hearts and Angiotensin II (Ang II)-treated cardiomyocytes, suggesting an increased sensitivity to hypertrophic stimuli potentially mediated by ULK1-induced ferroptosis in hypertrophic cardiomyocytes. Treatment with the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively reduced ULK1-induced cardiomyocyte hypertrophy and ferroptosis. Proteomic analysis identified the upregulation of transcription factor high mobility group A2 (HMGA2) as a key mechanism in this ferroptotic process. Elevated HMGA2 levels exacerbated ferroptosis, evidenced by increased cell death, lipid peroxidation, ROS production, and reduced GPX4 expression. Furthermore, HMGA2 was shown to promote cardiomyocyte ferroptosis via binding to methyltransferase-like 14 (METTL14), which in turn enhanced ferroptosis in cardiomyocytes through solute carrier family 7 member 11 (SLC7A11) m6A modification. In vivo, a delivery system using neutrophil membrane (NM)-coated mesoporous silica nanoparticles (MSN) was developed to inhibit cardiac hypertrophy, underscoring the therapeutic potential of targeting ULK1. Overall, this study demonstrates that ULK1 promotes cardiac hypertrophy through HMGA2/METTL14/SLC7A11 axis-mediated cardiomyocyte ferroptosis, suggesting a novel therapeutic approach for cardiac hypertrophy.
Collapse
Affiliation(s)
- Meitian Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Jiaxin Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Man Jiang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuhang Liu
- Department of Physiology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| |
Collapse
|
2
|
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
|
3
|
Davis E, Shokri MR, Rowland MB, York T, Cohen CT, Johnson AG, Moore PE, Schweitzer S, Sin J, Bui C, Correll RN. ATF6β is not essential for the development of physiological cardiac hypertrophy. PLoS One 2025; 20:e0320178. [PMID: 40193383 PMCID: PMC11975135 DOI: 10.1371/journal.pone.0320178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/15/2025] [Indexed: 04/09/2025] Open
Abstract
Physiological cardiac hypertrophy is a compensatory remodeling of the heart in response to stimuli such as exercise training or pregnancy that is reversible and well-tolerated. We previously described how the activating transcription factor 6 (ATF6) proteins, ATF6α and ATF6β, were required for pathological hypertrophy in response to hemodynamic stress. Here, we examine the functional roles of both ATF6 proteins in the context of exercise-induced physiological hypertrophy. After 20 days of swim training, we found differential roles: whole body gene-deleted mice lacking ATF6α had an attenuated hypertrophic response compared to wild-type mice but those lacking ATF6β did not. Additionally, mice lacking ATF6α displayed ventricular dilation and reduced fractional shortening after swimming. While we observed no differences in the expression of downstream UPR signaling between the exercise groups, mice lacking ATF6α showed enhanced phosphorylation of extracellular signal-regulated kinase 1/2 (ERK1/2). Thus, in response to swim training, loss of ATF6β did not hinder the development of physiological hypertrophy, but loss of ATF6α resulted in significantly reduced cardiac fractional shortening.
Collapse
Affiliation(s)
- Emery Davis
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Mohammad-Reza Shokri
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Mary B. Rowland
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Thomas York
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Caroline T. Cohen
- Department of Psychology, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Anna Grace Johnson
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Patrick E. Moore
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Saige Schweitzer
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Jon Sin
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, United States of America
| | - Chuong Bui
- Alabama Life Research Institute, University of Alabama, Tuscaloosa, Alabama, United States of America
| | - Robert N. Correll
- Department of Biological Sciences, University of Alabama, Tuscaloosa, Alabama, United States of America
- Center for Convergent Bioscience and Medicine, University of Alabama, Tuscaloosa, Alabama, United States of America
| |
Collapse
|
4
|
Ma Z, Cen Y, Xun W, Mou C, Yu J, Hu Y, Liu C, Sun J, Bi R, Qiu Y, Ding M, Jin L. Exercise enhances cardiomyocyte mitochondrial homeostasis to alleviate left ventricular dysfunction in pressure overload induced remodelling. Sci Rep 2025; 15:11698. [PMID: 40188200 PMCID: PMC11972341 DOI: 10.1038/s41598-025-95637-z] [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: 05/23/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
This study aims to explore how exercise enhances mitochondrial regulation and mitigates pathological cardiac hypertrophy. Rat groups were assigned as the control group (CN, n = 8), sham group (sham, n = 8), model group (SC, n = 16) and exercise group (SE, n = 20). A bioinformatics analysis was conducted to uncover the underlying mechanisms.H9C2 cells were divided into: the Ang II 0 h group (CON), Ang II 48 h group (Ang II), Ang II 48 h + sh-control group (sh-GFP + Ang II), Ang II 48 h + sh-ndufb10 group (sh-ndufb10 + Ang II), Ang II 48 h + overexpressedndufb10 control group (Ad-GFP + Ang II) and Ang II 48 h + over-expressedndufb10group (Ad-ndufb10 + Ang II). Mitochondrial function was measured. mRNA and protein expression were assessed by qPCR or western blot analysis respectively. In the SC group, a significant increase was observed in cardiomyocyte diameter, cardiac function, autophagy, and apoptosis. After 8 weeks of swimming exercise, there was a substantial reduction in cardiomyocyte diameter, an improvement in cardiac function, a mitigation of mitochondrial fission and autophagy. Ndufb10 was markedly enriched in oxidative phosphorylation and downregulated in the SC group, while it was upregulated in the SE group. In the sh-ndufb10 group, mitochondrial fusion was suppressed; fission and autophagy were further facilitated; mitochondrial membrane potential, mPTP, and ROS levels increased; and TUNEL positive nuclei and apoptosis-related proteins showed significant upregulation. Overexpression of ndufb10 reversed pathological hypertrophy, mitochondrial autophagy, mitochondrial dysfunction, and cardiomyocyte apoptosis in vitro. Swimming exercise improves mitochondrial abnormalities and reduces cardiomyocyte hypertrophy through regulation of the ndufb10 in left ventricular hypertrophy.
Collapse
Affiliation(s)
- Zhichao Ma
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China.
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Equine Science Research and Horse Doping Control Laboratory, Wuhan Business University, Wuhan, 430056, China.
| | - Yanling Cen
- School of Physical Education, Wuhan Business University, Wuhan, 430056, China
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Weiwei Xun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Caiying Mou
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Junwen Yu
- Aquinas International Academy, Ontario, CA, 90623, USA
| | - Yarui Hu
- Chiko Sports Institute, Sichuan University of Science and Technology, Meishan, 620000, China
| | - Chen Liu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Jun Sun
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Rui Bi
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Yanli Qiu
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Mingchao Ding
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China
| | - Li Jin
- College of Health Science, Wuhan Sports University, Wuhan, 430079, China.
- Hubei Exercise Training and Monitoring Key Laboratory, Wuhan Sports University, Wuhan, 430079, China.
| |
Collapse
|
5
|
Alammari AH, Isse FA, O'Croinin C, Davies NM, El-Kadi AOS. Effect of Cannabistilbene I in Attenuating Angiotensin II-Induced Cardiac Hypertrophy: Insights into Cytochrome P450s and Arachidonic Acid Metabolites Modulation. Cannabis Cannabinoid Res 2025; 10:277-288. [PMID: 39324890 DOI: 10.1089/can.2024.0148] [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: 09/27/2024] Open
Abstract
Introduction: This research investigated the impact of Cannabistilbene I on Angiotensin II (Ang II)-induced cardiac hypertrophy and its potential role in cytochrome P450 (CYP) enzymes and arachidonic acid (AA) metabolic pathways. Cardiac hypertrophy, a response to increased stress on the heart, can lead to severe cardiovascular diseases if not managed effectively. CYP enzymes and AA metabolites play critical roles in cardiac function and hypertrophy, making them important targets for therapeutic intervention. Methods: Adult human ventricular cardiomyocyte cell line (AC16) was cultured and treated with Cannabistilbene I in the presence and absence of Ang II. The effects on mRNA expression related to cardiac hypertrophic markers and CYP were analyzed using real-time polymerase chain reaction, while CYP protein levels were measured by Western blot analysis. AA metabolites were quantified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Results: Results showed that Ang II triggered hypertrophy, as evidenced by the increase in hypertrophic marker expression, and enlarged the cell surface area, effects that were alleviated by Cannabistilbene I. Gene expression analysis indicated that Cannabistilbene I upregulated CYP1A1, leading to increased enzymatic activity, as evidenced by 7-ethoxyresorufin-O-deethylase assay. Furthermore, LC-MS/MS analysis of AA metabolites revealed that Ang II elevated midchain (R/S)-hydroxyeicosatetraenoic acid (HETE) concentrations, which were reduced by Cannabistilbene I. Notably, Cannabistilbene I selectively increased 19(S)-HETE concentration and reversed the Ang II-induced decline in 19(S)-HETE, suggesting a unique protective role. Conclusion: This study provides new insights into the potential of Cannabistilbene I in modulating AA metabolites and reducing Ang II-induced cardiac hypertrophy, revealing a new candidate as a therapeutic agent for cardiac hypertrophy.
Collapse
Affiliation(s)
- Ahmad H Alammari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Fadumo Ahmed Isse
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Conor O'Croinin
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ayman O S El-Kadi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Abolfazli S, Karav S, Johnston TP, Sahebkar A. Regulatory effects of resveratrol on nitric oxide signaling in cardiovascular diseases. Pharmacol Rep 2025; 77:355-374. [PMID: 39832074 DOI: 10.1007/s43440-025-00694-w] [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: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular illnesses are multifactorial disorders and represent the primary reasons for death worldwide, according to the World Health Organization. As a signaling molecule, nitric oxide (NO) is extremely permeable across cellular membranes owing to its unique molecular features, like its small molecular size, lipophilicity, and free radical properties. Some of the biological effects of NO are vasodilation, inhibition in the growth of vascular smooth muscle cells, and functional regulation of cardiac cells. Several therapeutic approaches have been tested to increase the production of NO or some downstream NO signaling pathways. The health benefits of red wine are typically attributed to the polyphenolic phytoalexin, resveratrol (3,5,4'-trihydroxy-trans-stilbene), which is found in several plant species. Resveratrol has beneficial cardiovascular properties, some of which are mediated through endothelial nitric oxide synthase production (eNOS). Resveratrol promotes NO generation from eNOS through various methods, including upregulation of eNOS expression, activation in the enzymatic activity of eNOS, and reversal of eNOS uncoupling. Additionally, by reducing of oxidative stress, resveratrol inhibits the formation of superoxide and inactivation NO, increasing NO bioavailability. This review discusses the scientific literature on resveratrol's beneficial impact on NO signaling and how this effect improves the function of vascular endothelium.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Zhang S, Xie X, Zhao J, Jiang Y, Huang C, Li Q, Xia B, Yin L, Yuan X, You Q. Andrographolide and its Derivatives in Cardiovascular Disease: A Comprehensive Review. PLANTA MEDICA 2025; 91:259-270. [PMID: 40054492 DOI: 10.1055/a-2542-0756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Cardiovascular disease is one of the main causes of mortality worldwide. Andrographolide represents an important category of natural phytochemicals that has significant therapeutic potential in various conditions such as acute lung injury, heart disease, and viral infections due to its anti-oxidative, anti-inflammatory, and anti-apoptotic properties. This compound plays a protective role in human pathophysiology. This review provides a comprehensive overview of the effects of andrographolide on cardiovascular disease and examines its essential roles and mechanisms in cardiovascular disease and other vascular dysfunctions. The data collected in this review serve as a comprehensive reference for the role of andrographolide in cardiovascular disease and provide valuable insights for further research and the development of andrographolide as a novel therapeutic approach for cardiovascular disease.
Collapse
Affiliation(s)
- Shenjie Zhang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Xiaokai Xie
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Juan Zhao
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Yilong Jiang
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Qi Li
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Boyu Xia
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Le Yin
- Department of Cardiology, Tongzhou People's Hospital, Nantong, China
| | - Xiaomei Yuan
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qingsheng You
- Department of Cardiothoracic Surgery, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
8
|
Di Mattía RA, Gallo D, Ciarrocchi S, Gonano LA, Blanco PG, Valverde CA, Portiansky EL, Sommese LM, Toischer K, Bleckwedel F, Zelarayán LC, Aiello EA, Orlowski A. Cardiac hypertrophy induced by overexpression of IP3-released inositol 1, 4, 5-trisphosphate receptor-binding protein (IRBIT). J Mol Cell Cardiol 2025; 201:1-15. [PMID: 39929439 DOI: 10.1016/j.yjmcc.2025.02.001] [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: 03/06/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION IRBIT, also known as Ahcyl1, is an IP3 receptor (IP3R)-binding protein released with IP3 and was first described as a competitive inhibitor of the mentioned receptor. Studies have shown that overexpression of IP3Rs is associated with cardiac hypertrophy in both human and animal models. Given that IP3Rs play a role in cardiac hypertrophy, IRBIT may also be involved in this condition. AIM Although IRBIT heart expression has been reported, its function in cardiac tissues remains unclear. Thus, we aimed to study the cardiac outcomes of up-and downregulation of IRBIT to establish its pathophysiological role. METHODS AND RESULTS We found that IRBIT is expressed in mouse ventricles and atria, fibroblasts and cardiomyocytes isolated from neonatal mice, and in the myoblast cell line H9c2. Mice with transverse aortic constriction showed a significant increase in both the mRNA and protein expression of IRBIT. Furthermore, we described the differential expression of IRBIT in human myocardial samples of dilated and ischemic cardiomyopathy. IRBIT cardiac overexpression in mice using an adenoassociated virus (AAV9) at two different time points (neonatal mice, day 4 and adult mice, 3 months) resulted in the development of cardiac hypertrophy with impaired systolic function by four months of age. A decrease in the mRNA levels of the IP3 receptor was also observed in both models. Isolated myocytes from the IRBIT-overexpressing neonatal model showed a significantly decreased Ca2+ transient amplitude and slower rise of the global Ca2+ transient, without changes in sarcoplasmic reticulum (SR) Ca2+ content or spontaneous Ca2+ wave frequency. However, the velocity of Ca2+ wave propagation was reduced. Moreover, we found that the dyssynchrony index (DI) is significantly increased under IRBIT overexpression. Nuclear Ca2+ dynamics were assessed, showing no significant changes, but IRBIT overexpression reduced the number of nuclear envelope invaginations. In addition, reducing IRBIT expression using AAV9-shRNA did not result in any changes in the heart morphometric parameters. CONCLUSION Our study describes for the first time that IRBIT plays a critical role in the pathophysiology of the heart. Our findings demonstrate that IRBIT overexpression disrupts Ca2+ signaling, contributing to hypertrophic remodeling and impaired cardiac function. The altered wave propagation, the increase in DI and the decrease of the rate of the Ca2+ transient suggests that IRBIT influences Ca2+ - induced Ca2+ release. This study provides the first evidence linking IRBIT to pathological cardiac remodeling and Ca2+ handling dysregulation. Although significant progress has been made, further research is required to better understand the cardiovascular function of IRBIT and its mechanisms.
Collapse
MESH Headings
- Animals
- Cardiomegaly/metabolism
- Cardiomegaly/genetics
- Cardiomegaly/pathology
- Humans
- Mice
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Inositol 1,4,5-Trisphosphate Receptors/genetics
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Male
- Disease Models, Animal
- Calcium Signaling
- Cell Line
- Calcium/metabolism
- Mice, Inbred C57BL
- Inositol 1,4,5-Trisphosphate/metabolism
Collapse
Affiliation(s)
- R A Di Mattía
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - D Gallo
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - S Ciarrocchi
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - L A Gonano
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - P G Blanco
- Centro de Fisiología Reproductiva & Métodos Complementarios de Diagnóstico (CEFIRE & MECODIAG), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - C A Valverde
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - E L Portiansky
- Laboratorio de Análisis de Imágenes, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - L M Sommese
- Departamento de Ciencia y Tecnología, CONICET, Universidad Nacional de Quilmes, Bernal, Argentina
| | - K Toischer
- German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany; Clinic for Cardiology and Pneumology, University Medical Center, Göttingen, Germany
| | - F Bleckwedel
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany
| | - L C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany; Justus Liebig University, Medical Clinic I, Department of Cardiology and Angiology, Klinikstraße 33, 35392 Giessen, Germany
| | - E A Aiello
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - A Orlowski
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina.
| |
Collapse
|
9
|
Medina-Terol GJ, Chimal L, Huerta de la Cruz S, Ávila G, Aranda A, Cruz-Robles D, Centurión D, Altamirano J, Rojo R, Gómez-Viquez NL. H 2S treatment reverts cardiac hypertrophy and increases SERCA2a activity but does not fully restore cardiac Ca 2+ handling in hypertensive rats. Cell Calcium 2025; 128:103015. [PMID: 40184980 DOI: 10.1016/j.ceca.2025.103015] [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: 12/18/2024] [Revised: 03/15/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
Hydrogen sulfide (H2S) has been proposed to play a cardioprotective role, particularly due to its ability to revert left ventricular hypertrophy (LVH) and mitigate cardiac dysfunction in various cardiomyopathies, including hypertensive heart disease. However, the extent to which cardioprotection by H2S involves improvement in Ca2+ handling remains unclear. Although H2S has been reported to influence the function of key Ca2+ handling proteins, most studies have focused on acute administration of H2S donors in isolated cardiomyocytes, rather than in a therapeutic context. In this study, we used a rat model of hypertension induced by abdominal aortic coarctation (AAC) to evaluate the therapeutic potential of NaHS, an H2S donor, on LVH and Ca2+ handling. After 8 weeks of AAC, hypertensive rats developed moderate LVH, which was accompanied by a reduction in both the amplitude and the rate of rise of systolic Ca2+ transients, as well as a decrease in sarcoplasmic reticulum (SR) Ca2+ load. Despite the reduced SR Ca2+ load, the frequency of diastolic Ca2+ sparks remained high, while the incidence and propagation rate of spontaneous Ca2+ waves significantly increased, suggesting enhanced diastolic SR Ca2+ leak, most likely due to hypersensitivity of ryanodine receptors (RyR2) to Ca2+. On the other hand, NaHS administration during the final 4 weeks of AAC reverted both LVH and hypertension, and increased SR Ca2+ reuptake mediated by the SR Ca2+ ATPase (SERCA2a). However, NaHS treatment failed to restore the amplitude and rate of rise of systolic Ca2+ transients or SR Ca2+ load. Furthermore, SR Ca2+ leak might have worsened, since spontaneous Ca2+ waves increased. In conclusion, NaHS treatment does not appear to normalize all Ca2+ handling properties during hypertensive LVH. On the contrary, NaHS may exert an arrhythmogenic effect, likely due to enhanced SERCA2a activity under conditions of unresolved RyR2 Ca2+ hypersensitivity.
Collapse
Affiliation(s)
- Grecia J Medina-Terol
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Luis Chimal
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Saúl Huerta de la Cruz
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Guillermo Ávila
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Alberto Aranda
- Instituto Nacional de Cardiología Ignacio Chávez, Departamento de Anatomía Patológica, Ciudad de México, Mexico
| | - David Cruz-Robles
- Instituto Nacional de Cardiología Ignacio Chávez, Departamento de Biología Molecular, Ciudad de México, Mexico
| | - David Centurión
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Julio Altamirano
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Chihuahua, Mexico
| | - Rocio Rojo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Chihuahua, Mexico
| | - Norma Leticia Gómez-Viquez
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados-Instituto Politécnico Nacional, Ciudad de México, Mexico.
| |
Collapse
|
10
|
Bevere M, Morabito C, Verucci D, Di Sinno N, Mariggiò MA, Guarnieri S. Growth-Associated Protein-43 Loss Promotes Ca 2+ and ROS Imbalance in Cardiomyocytes. Antioxidants (Basel) 2025; 14:361. [PMID: 40227418 PMCID: PMC11939155 DOI: 10.3390/antiox14030361] [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: 01/22/2025] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Growth-Associated Protein-43 (GAP-43) is a calmodulin-binding protein, originally found in neurons, that in skeletal muscle regulates the handling of intracellular Ca2+ dynamics. According to its role in Ca2+ regulation, myotubes from GAP-43 knockout (GAP-43-/-) mice display alterations in spontaneous Ca2+ oscillations and increased Ca2+ release. The emerging hypothesis is that GAP-43 regulates CaM interactions with RyR and DHPR Ca2+ channels. The loss of GAP-43 promotes cardiac hypertrophy in newborn GAP-43-/- mice, extending the physiological role of GAP-43 in cardiac muscle. We investigated the role of GAP-43 in cardiomyocytes derived from the hearts of GAP-43-/- mice, evaluating intracellular Ca2+ variations and the correlation with the levels of reactive oxygen species (ROS), considering their importance in cardiovascular physiology. In GAP-43-/- cardiomyocytes, we found the increased expression of markers of cardiac hypertrophy, Ca2+ alterations, and high mitochondria ROS levels (O2•-) together with increased oxidized functional proteins. Treatment with a CaM inhibitor (W7) restored Ca2+ and ROS alterations, possibly due to high mitochondrial Ca2+ entry by a mitochondrial Ca2+ uniporter. Indeed, Ru360 was able to abolish O2•- mitochondrial production. Our results suggest that GAP-43 has a key role in the regulation of Ca2+ and ROS homeostasis, alterations to which could trigger heart disease.
Collapse
Affiliation(s)
- Michele Bevere
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
- ARC-Net Applied Research on Cancer Centre, University and Hospital Trust of Verona, 37134 Verona, Italy
| | - Caterina Morabito
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
| | - Delia Verucci
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
| | - Noemi Di Sinno
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
| | - Maria A. Mariggiò
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
| | - Simone Guarnieri
- Department of Neuroscience, Imaging and Clinical Sciences and Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (M.B.); (C.M.); (D.V.); (N.D.S.); (M.A.M.)
| |
Collapse
|
11
|
Liu RP, McMullen JR. Emerging Role of Targeting Deubiquitinating Enzymes to Inhibit Pathological Cardiac Hypertrophy. J Am Heart Assoc 2025; 14:e039732. [PMID: 40040615 DOI: 10.1161/jaha.124.039732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/14/2025] [Indexed: 03/06/2025]
|
12
|
Ding J, Yang A, Zhou L, Zhang F, Zhou H, Zhang Y, Wang Y, Liu Y, Liang D, Liu Y, Wu Y. PLK1 Downregulation Attenuates ET-1-Induced Cardiomyocyte Hypertrophy by Suppressing the ERK1/2 Pathway. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10604-3. [PMID: 40095199 DOI: 10.1007/s12265-025-10604-3] [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: 01/14/2025] [Accepted: 03/05/2025] [Indexed: 03/19/2025]
Abstract
Cardiomyocyte hypertrophy is a key remodeling response to cardiac stress and an independent risk factor for heart failure. However, the molecular mechanism of cardiomyocyte hypertrophy is not yet fully understood. We here found Polo-like kinase 1 (PLK1) was crucial in regulating endothelin-1 (ET-1)-induced cardiomyocyte hypertrophy. Notably, PLK1 expression was significantly elevated in ET-1-induced hypertrophic cardiomyocytes and pressure overload-induced hypertrophic cardiac tissue. Knocking down Plk1 reduced the cell size of hypertrophic cardiomyocytes and suppressed the expression of hypertrophic markers, including ANP, BNP and β-MHC. The PLK1 inhibitor BI2536 had similar effects on hypertrophic cardiomyocytes. Mechanistically, the ERK1/2 pathway was identified as the key downstream pathway mediating the effects of PLK1 on ET-1-induced cardiomyocyte hypertrophy. Finally, the deficiency of PLK1 attenuated the hypertrophy of hiPSC-CMs. In summary, our study revealed that PLK1 regulates ET-1-induced cardiomyocyte hypertrophy through the ERK1/2 pathway, providing insights into the pathogenesis and potential therapies for pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Jie Ding
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Anqi Yang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liping Zhou
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Fulei Zhang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Huixing Zhou
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuemei Zhang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yan Wang
- Jinzhou Medical University, Jinzhou, 121000, China
| | - Yi Liu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Dandan Liang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Yuanyuan Liu
- Jinzhou Medical University, Jinzhou, 121000, China.
| | - Yahan Wu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
13
|
Sheng S, Liu G, Lv P, Liu J, Lv L, Yuan M, Luo D, Xiong J, Dong P, Zhang J, Xie B, Dong Z, Shi Y. Therapeutic inhibition of PHF21B attenuates pathological cardiac hypertrophy by inhibiting the BMP4/GSK3β/β-catenin axis. Eur J Pharmacol 2025; 991:177346. [PMID: 39900327 DOI: 10.1016/j.ejphar.2025.177346] [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: 12/26/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
BACKGROUND Pathological cardiac hypertrophy is a hallmark of various cardiovascular diseases, unfortunately, effective targeted therapies are still lacking. This study aims to verify the role of plant-homeodomain finger protein21b (PHF21B) in pathological cardiac hypertrophy. METHODS Angiotensin-II (Ang II) induced cardiomyocyte hypertrophy in vitro, and short hairpin (sh) RNA-mediated PHF21B silencing was used to assess its role in hypertrophic growth. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy in mice. To assess the effect of PHF21B on pathological cardiac hypertrophy in vivo, the myocardium was transduced with adeno-associated virus 9 (AAV9) encoding a PHF21B-targeting shRNA for gene ablation. Chromatin immunoprecipitation-polymerase chain reaction (PCR), western blotting, and quantitative reverse transcription-PCR were performed to elucidate the mechanisms through which PHF21B regulates pathological cardiac hypertrophy. RESULTS This investigation revealed that PHF21B levels were elevated in patients with pathological cardiac hypertrophy. PHF21B inhibition alleviated pressure overload-induced cardiac dysfunction and hypertrophy in vivo, and Ang-II-induced cardiomyocyte hypertrophy in vitro. Genome-wide transcriptome analysis and biological experiments revealed that PHF21B silencing inhibited the Wnt signalling pathway, include the protein expression of β-catenin, and the phosphorylation of glycogen synthase kinase (GSK)-3β. Mechanistically, PHF21B influenced the translation of bone morphogenetic protein (BMP)-4 and facilitated the activation of the GSK3β/β-catenin pathway. The anti-hypertrophic effects of PHF21B knockdown were blocked by BMP4 supplementation. CONCLUSIONS Collectively, our results demonstrated that PHF21B is contributes to pathological cardiac hypertrophy by regulating BMP4 expression and the GSK3β/β-catenin pathway. The inhibition of PHF21B is a potential new therapeutic strategy to mitigate pathological cardiiac hypertrophy.
Collapse
Affiliation(s)
- Siqi Sheng
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China; The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Guannan Liu
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Pengcheng Lv
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Jialiang Liu
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Lin Lv
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China; Experimental Animal Center, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Meng Yuan
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Dankun Luo
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Jie Xiong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Pengwei Dong
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Jingyue Zhang
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China
| | - Baodong Xie
- Cardiovascular Surgery, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China.
| | - Zengxiang Dong
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China; NHC Key Laboratory of Cell Transplantation, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China.
| | - Yuanqi Shi
- The Key Laboratory of Cardiovascular Disease Acousto-Optic Electromagnetic Diagnosis and Treatment in Heilongjiang Province, The First Affiliated Hospital of Harbin Medical University, Youzheng Street, Nangang District, Harbin, 150001, China.
| |
Collapse
|
14
|
Eggertsen TG, Travers JG, Hardy EJ, Wolf MJ, McKinsey TA, Saucerman JJ. Logic-based machine learning predicts how escitalopram attenuates cardiomyocyte hypertrophy. Proc Natl Acad Sci U S A 2025; 122:e2420499122. [PMID: 40035765 PMCID: PMC11912418 DOI: 10.1073/pnas.2420499122] [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: 10/06/2024] [Accepted: 01/14/2025] [Indexed: 03/06/2025] Open
Abstract
Cardiomyocyte hypertrophy is a key clinical predictor of heart failure. High-throughput and AI-driven screens have the potential to identify drugs and downstream pathways that modulate cardiomyocyte hypertrophy. Here, we developed LogiRx, a logic-based mechanistic machine learning method that predicts drug-induced pathways. We applied LogiRx to discover how drugs discovered in a previous compound screen attenuate cardiomyocyte hypertrophy. We experimentally validated LogiRx predictions in neonatal cardiomyocytes, adult mice, and two patient databases. Using LogiRx, we predicted antihypertrophic pathways for seven drugs currently used to treat noncardiac disease. We experimentally validated that escitalopram (Lexapro) and mifepristone inhibit hypertrophy of cultured cardiomyocytes in two contexts. The LogiRx model predicted that escitalopram prevents hypertrophy through an "off-target" serotonin receptor/PI3Kγ pathway, mechanistically validated using additional investigational drugs. Further, escitalopram reduced cardiomyocyte hypertrophy in a mouse model of hypertrophy and fibrosis. Finally, mining of both FDA and University of Virginia databases showed that patients with depression on escitalopram have a lower incidence of cardiac hypertrophy than those prescribed other serotonin reuptake inhibitors that do not target the serotonin receptor. Mechanistic machine learning by LogiRx discovers drug pathways that perturb cell states, which may enable repurposing of escitalopram and other drugs to limit cardiac remodeling through off-target pathways.
Collapse
Affiliation(s)
- Taylor G. Eggertsen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA22908
| | - Joshua G. Travers
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045-2507
| | - Elizabeth J. Hardy
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045-2507
| | - Matthew J. Wolf
- Division of Cardiovascular Medicine, University of Virginia, Charlottesville, VA22908
| | - Timothy A. McKinsey
- Division of Cardiology and Consortium for Fibrosis Research and Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO80045-2507
| | - Jeffrey J. Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA22908
| |
Collapse
|
15
|
Yang X, Liu H, Wu X. High-altitude pulmonary hypertension: a comprehensive review of mechanisms and management. Clin Exp Med 2025; 25:79. [PMID: 40063280 PMCID: PMC11893705 DOI: 10.1007/s10238-025-01577-3] [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: 07/23/2024] [Accepted: 01/26/2025] [Indexed: 03/14/2025]
Abstract
High-altitude pulmonary hypertension (HAPH) is characterized by an increase in pulmonary artery pressure due to prolonged exposure to hypoxic environment at high altitudes. The development of HAPH involves various factors such as pressure changes, inflammation, oxidative stress, gene regulation, and signal transduction. The pathophysiological mechanisms of this condition operate at molecular, cellular, and genetic levels. Diagnosis of HAPH often relies on echocardiography, cardiac catheterization, and other methods to assess pulmonary artery pressure and its impact on cardiac function. Treatment options for HAPH encompass both nondrug and drug therapies. While advancements have been made in understanding the pathological mechanisms through research on animal models and clinical trials, there are still limitations to be addressed. Future research should focus on exploring molecular targets, personalized medicine, long-term management strategies, and interdisciplinary approaches. By leveraging advanced technologies like systems biology, omics technology, big data, and artificial intelligence, a comprehensive analysis of HAPH pathogenesis can lead to the identification of new treatment targets and strategies, ultimately enhancing patient quality of life and prognosis. Furthermore, research on health monitoring and preventive measures for populations living at high altitudes should be intensified to reduce the incidence and mortality of HAPH.
Collapse
Affiliation(s)
- Xitong Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Hong Liu
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China
| | - Xinhua Wu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Medical School, Kunming University of Science and Technology, Kunming, Yunnan, China.
- The First Affiliated Hospital of Dali University, Dali, Yunnan, China.
| |
Collapse
|
16
|
Ge G, Zhao W, Zhong Z, Huang Y, Hua Y, Chen K, Yu Y, Wu T, Lu Y, Yadav N, Zhang F. Acacetin ameliorates pressure overload-induced cardiac remodeling by targeting USP10 and inhibiting maladaptive cardiomyocyte autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156588. [PMID: 40118748 DOI: 10.1016/j.phymed.2025.156588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/11/2025] [Accepted: 02/26/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Numerous drugs have been developed to meet the critical demand for treatments inhibiting cardiac remodeling following cardiovascular disease. Acacetin is a flavonoid with potential therapeutic effects against various cardiovascular diseases. PURPOSE This study investigated the effect of acacetin on pressure overload-induced cardiac remodeling and its underlying molecular regulatory mechanisms. METHODS We simulated pressure overload-induced cardiac remodeling in male C57BL/6 mice by constricting the thoracic aortic arch and assessed the effect of acacetin on cardiac remodeling. RESULTS Acacetin significantly ameliorated cardiac remodeling by downregulating ubiquitin-specific peptidase 10 (USP10) protein expression and reducing autophagy levels in cardiomyocytes. These findings confirm that acacetin improves cardiac remodeling by suppressing cardiomyocyte autophagy and highlight the crucial role of USP10 in the Beclin 1 ubiquitination degradation-mediated inhibition of the cardiomyocyte autophagy signaling pathway. CONCLUSION These results suggest that acacetin is a promising candidate drug for the treatment of cardiac remodeling induced by pressure overload.
Collapse
Affiliation(s)
- Gaoyuan Ge
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China; Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Hanjiang Middle Road 368, Yangzhou 225000, Jiangsu, PR China
| | - Wei Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Zhuen Zhong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Youfu Huang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yan Hua
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Kaiyan Chen
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yue Yu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Tianyu Wu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Yao Lu
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Nishant Yadav
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China
| | - Fengxiang Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, Jiangsu, PR China.
| |
Collapse
|
17
|
Luo D, Wang J, Zheng S, Li W, Yu B, Peng H, Gui F, Mao B, Chen Z. Crocin ameliorates hypertension-induced cardiac hypertrophy and apoptosis by activating AMPKα signalling. CLIN INVEST MED 2025; 48:11-23. [PMID: 40131211 DOI: 10.3138/cim-2024-0118] [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: 03/26/2025]
Abstract
PURPOSE Cardiac hypertrophy is a critical contributor to heart failure. Therapies that effectively manage cardiac hypertrophy are still inadequate. Crocin is a natural component of saffron, and its beneficial properties have been previously documented. This study aimed to investigate the role of crocin in cardiac hypertrophy and apoptosis and its related mechanisms. METHODS Sprague-Dawley rats were infused with angiotensin II (Ang II; 520 ng/kg/min) or normal saline and then intraperitoneally injected with crocin (40 mg/kg) or dimethyl sulfoxide for 4 weeks. Systolic and diastolic blood pressure were recorded. Cardiac hypertrophy was evaluated by echocardiography, heart weight, hematoxylin-eosin staining, TUNEL assay, and gene expression. For in vitro studies, H9C2 cells were treated with Ang II (1 μM) for 48 hours to induce cardiac hypertrophy-like conditions. An immunofluorescence assay was used for [Formula: see text]-actinin staining. reverse transcription quantitative real-time polymerase chain reaction was performed to measure the expression of hypertrophic markers, and western blotting was used to detect apoptosis and underlying mechanisms. RESULTS Our findings revealed that crocin attenuated diastolic dysfunction, cardiac hypertrophy, and apoptosis caused by Ang II in vivo. Additionally, crocin prevented Ang II-stimulated cardiomyocyte enlargement and apoptosis in vitro. Mechanistically, crocin induced AMP-activated protein kinase (AMPK)[Formula: see text] activation and mTOR/p70S6K inhibition in cellular and animal models of cardiac hypertrophy. Moreover, AMPK inhibition abolished the anti-hypertrophic effect of crocin in vitro, while mTOR inhibition enhanced the protective effect of crocin against Ang II-induced cardiomyocyte hypertrophy. CONCLUSION This study demonstrates that crocin can ameliorate Ang II-stimulated cardiac hypertrophy in vivo and in vitro by regulating AMPK[Formula: see text]/mTOR/ p70S6K signalling.
Collapse
Affiliation(s)
- Dan Luo
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jueyan Wang
- School of Medicine, Jianghan University, Wuhan, Hubei, China
| | - Shijiao Zheng
- Department of Nephrology, The 7th Hospital of Wuhan, Wuhan, Hubei, China
| | - Wei Li
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bo Yu
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huan Peng
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Feng Gui
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bing Mao
- Department of Pediatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhen Chen
- Department of Emergency Medicine, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
18
|
Mancuso G, Marsan M, Neroni P, Soddu C, Lai F, Serventi L, Cau M, Coiana A, Incani F, Murru S, Savasta S. Clinical and Genetic Heterogeneity of HCM: The Possible Role of a Deletion Involving MYH6 and MYH7. Genes (Basel) 2025; 16:212. [PMID: 40004541 PMCID: PMC11855101 DOI: 10.3390/genes16020212] [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: 01/20/2025] [Revised: 01/29/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Pediatric hypertrophic cardiomyopathy (HCM) is the most common genetic myocardial disorder in children and a leading cause of sudden cardiac death (SCD) among the young. Its phenotypic variability, driven by incomplete penetrance and variable expressivity, presents significant challenges in diagnosis and clinical management. METHODS In this study, we report a unique case of a 16-month-old female diagnosed with HCM caused by a rare genetic deletion. Molecular analysis was performed using a multigene panel and chromosomal microarray analysis (CMA). RESULTS Molecular tests identified a 30 kb deletion encompassing the MYH6 and MYH7 genes. These genes are critical components of sarcomeric architecture, with known associations to HCM and other cardiomyopathies. CONCLUSIONS This case underscores the clinical and genetic heterogeneity of HCM, highlighting the importance of considering genomic deletions involving key sarcomeric genes in the diagnostic evaluation.
Collapse
Affiliation(s)
- Giancarlo Mancuso
- Medical Genetics Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (G.M.)
| | - Marina Marsan
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital “A. Cao”, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
| | - Paola Neroni
- Neonatal Intensive Care Unit, Department of Surgical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Consolata Soddu
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
| | - Francesco Lai
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
- Unit of Oncology and Molecular Pathology, Department of Biomedical Sciences, University of Cagliari, 09124 Cagliari, Italy
| | - Laura Serventi
- Medical Genetics Unit, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy; (G.M.)
| | - Milena Cau
- Genetic and Genomic Laboratory, Pediatric Children Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Alessandra Coiana
- Genetic and Genomic Laboratory, Pediatric Children Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
- Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
| | - Federica Incani
- Genetic and Genomic Laboratory, Pediatric Children Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
| | - Stefania Murru
- Genetic and Genomic Laboratory, Pediatric Children Hospital “A. Cao”, ASL 8 Cagliari, 09121 Cagliari, Italy
| | - Salvatore Savasta
- Pediatric and Rare Diseases Clinic, Microcitemico Hospital “A. Cao”, Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy;
| |
Collapse
|
19
|
Wang J, Huang Y, Wang Z, Liu J, Liu Z, Yang J, He Z. The mTOR Signaling Pathway: Key Regulator and Therapeutic Target for Heart Disease. Biomedicines 2025; 13:397. [PMID: 40002810 PMCID: PMC11853667 DOI: 10.3390/biomedicines13020397] [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: 12/15/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Heart disease, including myocardial infarction, heart failure, cardiac hypertrophy, and cardiomyopathy, remains a leading cause of mortality worldwide. The mammalian target of rapamycin (mTOR) is a centrally regulated kinase that governs key cellular processes, including growth, proliferation, metabolism, and survival. Notably, mTOR plays a pivotal role in cardiovascular health and disease, particularly in the onset and progression of cardiac conditions. In this review, we discuss mTOR's structure and function as well as the regulatory mechanisms of its associated signaling pathways. We focus on the molecular mechanisms by which mTOR signaling regulates cardiac diseases and the potential of mTOR inhibitors and related regulatory drugs in preventing these conditions. We conclude that the mTOR signaling pathway is a promising therapeutic target for heart disease.
Collapse
Affiliation(s)
- Jieyu Wang
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China; (J.W.); (Y.H.); (Z.W.); (J.L.)
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Yuxuan Huang
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China; (J.W.); (Y.H.); (Z.W.); (J.L.)
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Zhaoxia Wang
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China; (J.W.); (Y.H.); (Z.W.); (J.L.)
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Jing Liu
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China; (J.W.); (Y.H.); (Z.W.); (J.L.)
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| | - Zhijian Liu
- Department of Anesthesiology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya, School of Medicine, Central South University, Changsha 410013, China;
| | - Jinfeng Yang
- Department of Anesthesiology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya, School of Medicine, Central South University, Changsha 410013, China;
| | - Zuping He
- Department of Basic Medicine, School of Medicine, Hunan Normal University, Changsha 410013, China; (J.W.); (Y.H.); (Z.W.); (J.L.)
- The Key Laboratory of Model Animal and Stem Cell Biology in Hunan Province, Hunan Normal University, Changsha 410013, China
| |
Collapse
|
20
|
Hu M, Huang SY, Gao YP, Hu YX, Wang SS, Teng T, Zeng XF, Tang QZ. KLF12 Aggravates Angiotensin II-Induced Cardiac Remodeling in Male Mice by Transcriptionally Inhibiting SMAD7. J Am Heart Assoc 2025; 14:e037455. [PMID: 39895521 DOI: 10.1161/jaha.124.037455] [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: 07/01/2024] [Accepted: 10/30/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Adverse left ventricular remodeling and subsequent heart failure remain a major cause of patient morbidity and mortality worldwide. The KLF family of transcription factors plays crucial roles in heart injury. KLF12 (Krüppel-like factor 12) is a transcription factor that regulates multiple disease processes, although the specific role of KLF12 in cardiac remodeling remains unclear. METHODS AND RESULTS In our study, we observed a significant upregulation of KLF12 expression in remodeling hearts. The increased expression of KLF12 primarily originated from cardiac fibroblasts during the fibrotic response induced by angiotensin II. To investigate the effects of KLF12, we performed RNA-seq and found that KLF12 overexpression significantly upregulated the cardiac remodeling associated pathway. Hence, we generated adult mice with cardiac fibroblast-specific overexpression of KLF12 using lentivirus or miRNA (miR-1/133TS) technology. Compared with control mice, KLF12-miR1/133TS transfected mice exhibited exacerbated cardiac remodeling and function. Mechanistically, we discovered that KLF12 directly binds to the promoter of Smad7, leading to the activation of the TGF-β (transforming growth factor beta)-Smad3 pathway. CONCLUSIONS In conclusion, KLF12 promoted the development of angiotensin II-induced cardiac remodeling in male mice. Targeting KLF12 may be a promising therapeutic approach to treat cardiac remodeling.
Collapse
Affiliation(s)
- Min Hu
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Shi-Yu Huang
- Department of Urology Renmin Hospital of Wuhan University Wuhan Hubei China
| | - Yi-Peng Gao
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Yu-Xin Hu
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Sha-Sha Wang
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Teng Teng
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Xiao-Feng Zeng
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| | - Qi-Zhu Tang
- Department of Cardiology Renmin Hospital of Wuhan University Wuhan China
- Hubei Key Laboratory of Metabolic and Chronic Diseases Wuhan China
| |
Collapse
|
21
|
Huang S, Zhou Y, Zhang Y, Liu N, Liu J, Liu L, Fan C. Advances in MicroRNA Therapy for Heart Failure: Clinical Trials, Preclinical Studies, and Controversies. Cardiovasc Drugs Ther 2025; 39:221-232. [PMID: 37505309 DOI: 10.1007/s10557-023-07492-7] [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] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Heart failure (HF) is a rapidly growing public health issue with more than 37.7 million patients worldwide and an annual healthcare cost of $108 billion. However, HF-related drugs have not changed significantly for decades, and it is essential to find biological drugs to provide better treatment for HF patients. MicroRNAs (miRNAs) are non-coding RNAs (ncRNAs) with a length of approximately 21 nucleotides and play an important role in the onset and progression of cardiovascular diseases. Increasing studies have shown that miRNAs are widely involved in the pathophysiology of HF, and the regulation of miRNAs has promising therapeutic effects. Among them, there is great interest in miRNA-132, since the encouraging success of anti-miRNA-132 therapy in a phase 1b clinical trial in 2020. However, it is worth noting that the multi-target effect of miRNA may produce side effects such as thrombocytopenia, revascularization dysfunction, severe immune response, and even death. Advances in drug delivery modalities, delivery vehicles, chemical modifications, and plant-derived miRNAs are expected to address safety concerns and further improve miRNA therapy. Here, we reviewed the preclinical studies and clinical trials of HF-related miRNAs (especially miRNA-132) in the past 5 years and summarized the controversies of miRNA therapy.
Collapse
Affiliation(s)
- Shengyuan Huang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yong Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yiru Zhang
- Department of Laboratory Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ningyuan Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Liming Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
22
|
Pan L, Huang C, Jin X, Wu J, Jin K, Lin J, Wang Y, Li J, Yin C, Wang X, Zhang L, Zhang G, Dong H, Guo J, Komuro I, Dai Y, Zou Y, Gong H. Cardiac secreted HSP90α exacerbates pressure overload myocardial hypertrophy and heart failure. Redox Biol 2025; 79:103466. [PMID: 39721497 PMCID: PMC11732234 DOI: 10.1016/j.redox.2024.103466] [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: 11/04/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Sustained myocardial hypertrophy or left ventricular hypertrophy (LVH) triggered by pressure overload is strongly linked to adverse cardiovascular outcomes. Here, we investigated the clinical relationship between serum HSP90α (an isoform of HSP90) levels and LVH in patients with hypertension or aortic stenosis (AS) and explored underlying mechanisms in pressure overload mouse model. We built a pressure overload mouse model via transverse aortic constriction (TAC). Compared to controls, elevated serum HSP90α levels were observed in patients with hypertension or AS, and the levels positively correlated with LVH. Similarly, HSP90α levels increased in heart tissues from patients with obstructive hypertrophic cardiomyopathy (HCM), and in mice post-TAC. TAC induced the enhanced cardiac expression and secretion of HSP90α from cardiomyocytes and cardiac fibroblasts. Knockdown of HSP90α or blockade of extracellular HSP90α (eHSP90α) attenuated cardiac hypertrophy and dysfunction by inhibition of β-catenin/TCF7 signaling under pressure overload. Further analysis revealed that eHSP90α interacted with EC1-EC2 region of N-cadherin to activate β-catenin, enhancing the transcription of hypertrophic genes by TCF7, resulting in cardiac hypertrophy and dysfunction under pressure overload. These insights suggest the therapeutic potential of targeting HSP90α-initiated signaling pathway against cardiac hypertrophy and heart failure under pressure overload.
Collapse
Affiliation(s)
- Le Pan
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenxing Huang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xuejuan Jin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China
| | - Kejia Jin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jingyi Lin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianxuan Li
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Lei Zhang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Guoping Zhang
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hangming Dong
- Department of Respiratory and Critical Care Medicine, Chronic Airways Diseases Laboratory, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, 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, 571199, China
| | - Issei Komuro
- Department of Frontier Cardiovascular Science, Graduate School of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
| | - Yuxiang Dai
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China; NHC Key Laboratory of Ischemic Heart Diseases, and Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China.
| |
Collapse
|
23
|
Schopohl B, Kohlhaas M, Nickel AG, Schiuma AF, Maas SL, van der Vorst EPC, Shia YX, Maack C, Steffens S, Puhl SL. Gpr55 deficiency crucially alters cardiomyocyte homeostasis and counteracts angiotensin II induced maladaption in female mice. Br J Pharmacol 2025; 182:670-691. [PMID: 39428581 DOI: 10.1111/bph.17350] [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: 01/18/2024] [Revised: 06/24/2024] [Accepted: 07/20/2024] [Indexed: 10/22/2024] Open
Abstract
BACKGROUND AND PURPOSE Cannabis stimulates several G-protein-coupled-receptors and causes bradycardia and hypotension upon sustained consumption. Moreover, in vitro studies suggest an interference of cannabinoid-signalling with cardiomyocyte contractility and hypertrophy. We aimed at revealing a functional contribution of the cannabinoid-sensitive receptor GPR55 to cardiomyocyte homeostasis and neurohumorally induced hypertrophy in vivo. EXPERIMENTAL APPROACH Gpr55-/- and wild-type (WT) mice were characterized after 28-day angiotensin II (AngII; 1·μg·kg-1 min-1) or vehicle infusion. In isolated adult Gpr55-/- and WT cardiomyocytes, mitochondrial function was assessed under naïve conditions, while cytosolic Ca2+ handling was additionally determined following application of the selective GPR55 antagonist CID16020046. KEY RESULTS Gpr55 deficiency did not affect angiotensin II (AngII) mediated hypertrophic growth, yet, especially in females, it alleviated maladaptive pro-hypertrophic and -inflammatory gene expression and improved inotropy and adrenergic responsiveness compared to WT. In-depth analyses implied increased cytosolic Ca2+ concentrations and transient amplitudes, and accelerated sarcomere contraction kinetics in Gpr55-/- myocytes, which could be mimicked by GPR55 blockade with CID16020046 in female WT cells. Moreover, Gpr55 deficiency up-regulated factors involved in glucose and fatty acid transport independent of the AngII challenge, accelerated basal mitochondrial respiration and reduced basal protein kinase (PK) A, G and C activity and phospholemman (PLM) phosphorylation. CONCLUSIONS AND IMPLICATIONS Our study suggests GPR55 as crucial regulator of cardiomyocyte hypertrophy and homeostasis presumably by regulating PKC/PKA-PLM and PKG signalling, and identifies the receptor as potential target to counteract maladaptation, adrenergic desensitization and metabolic shifts as unfavourable features of the hypertrophied heart in females.
Collapse
Affiliation(s)
- Brigitte Schopohl
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Michael Kohlhaas
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Alexander G Nickel
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | | | - Sanne L Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany
| | - Yi Xuan Shia
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
- Medical Clinic I, University Clinic Würzburg, Würzburg, Germany
| | - Sabine Steffens
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Sarah-Lena Puhl
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| |
Collapse
|
24
|
Zhao M, He X, Min X, Yang H, Wu W, Zhong J, Xu H, Chen J. Recent Clinical Updates of Hypertrophic Cardiomyopathy and Future Therapeutic Strategies. Rev Cardiovasc Med 2025; 26:25132. [PMID: 40026515 PMCID: PMC11868910 DOI: 10.31083/rcm25132] [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: 06/05/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 03/05/2025] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent inherited cardiomyopathy transmitted in an autosomal dominant manner to offspring. It is characterized by unexplained asymmetrical hypertrophy primarily affecting the left ventricle and interventricular septum while potentially causing obstruction within the left ventricular outflow tract (LVOT). The clinical manifestations of HCM are diverse, ranging from asymptomatic to severe heart failure (HF) and sudden cardiac death. Most patients present with obvious symptoms of left ventricular outflow tract obstruction (LVOTO). The diagnosis of HCM mainly depends on echocardiography and other imaging examinations. In recent years, myosin inhibitors have undergone clinical trials and gene therapy, which is expected to become a new treatment for HCM, has been studied. This article summarizes recent clinical updates on the epidemiology, pathogenesis, diagnostic methods, treatment principles, and complication prevention and treatment of HCM, to provide new ideas for follow-up research.
Collapse
Affiliation(s)
- Mengya Zhao
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Xianzhen He
- Children’s Medical Center, Renmin Hospital, Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Xinwen Min
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Handong Yang
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Wenwen Wu
- School of Public Health, Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Hao Xu
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei University of Medicine, 442000 Shiyan, Hubei, China
| | - Jun Chen
- Sinopharm Dongfeng General Hospital (Hubei Clinical Research Center of Hypertension), Hubei University of Medicine, 442000 Shiyan, Hubei, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, 442000 Shiyan, Hubei, China
| |
Collapse
|
25
|
Shen Y, Gleghorn JP. Class III Phosphatidylinositol-3 Kinase/Vacuolar Protein Sorting 34 in Cardiovascular Health and Disease. J Cardiovasc Transl Res 2025:10.1007/s12265-024-10581-z. [PMID: 39821606 DOI: 10.1007/s12265-024-10581-z] [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: 06/24/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Phosphatidylinositol-3 kinases (PI3Ks) play a critical role in maintaining cardiovascular health and the development of cardiovascular diseases (CVDs). Specifically, vacuolar Protein Sorting 34 (VPS34) or PIK3C3, the only member of Class III PI3K, plays an important role in CVD progression. The main function of VPS34 is inducing the production of phosphatidylinositol 3-phosphate, which, together with other essential structural and regulatory proteins in forming VPS34 complexes, further regulates the mammalian target of rapamycin activation, autophagy, and endocytosis. VPS34 is found to have crucial functions in the cardiovascular system, including dictating the proliferation and survival of vascular smooth muscle cells and cardiomyocytes and the formation of thrombosis. This review aims to summarize our current knowledge and recent advances in understanding the function and regulation of VPS34 in cardiovascular health and disease. We also discuss the current development of VPS34 inhibitors and their potential to treat CVDs.
Collapse
Affiliation(s)
- Yuanjun Shen
- Departments of Biomedical Engineering, University of Delaware, Newark, DE, USA.
- School of Pharmacy and Pharmceutical Sciences, Binghamton University, Johnson City, NY, USA.
| | - Jason P Gleghorn
- Departments of Biomedical Engineering, University of Delaware, Newark, DE, USA
- Biological Sciences, University of Delaware, Newark, DE, USA
| |
Collapse
|
26
|
Tian RZ, Zhuang DL, Vong CT, He X, Ouyang Q, Liang JH, Guo YP, Wang YH, Zhao S, Yuan H, Ide Nasser M, Li G, Zhu P. Role of Autophagy in Myocardial Remodeling After Myocardial Infarction. J Cardiovasc Pharmacol 2025; 85:1-11. [PMID: 39454200 DOI: 10.1097/fjc.0000000000001646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/08/2024] [Indexed: 10/27/2024]
Abstract
ABSTRACT Autophagy is the process of reusing the body's senescent and damaged cell components, which can be regarded as the cellular circulatory system. There are 3 distinct forms of autophagy: macroautophagy, microautophagy, and chaperone-mediated autophagy. In the heart, autophagy is regulated mainly through mitophagy because of the metabolic changes of cardiomyocytes caused by ischemia and hypoxia. Myocardial remodeling is characterized by gradual heart enlargement, cardiac dysfunction, and extraordinary molecular changes. Cardiac remodeling after myocardial infarction is almost inevitable, which is the leading cause of heart failure. Autophagy has a protective effect on myocardial remodeling improvement. Autophagy can minimize cardiac remodeling by preventing misfolded protein accumulation and oxidative stress. This review summarizes the nestest molecular mechanisms of autophagy and myocardial remodeling, the protective effects, and the new target of autophagy medicine in cardiac remodeling. The future development and challenges of autophagy in heart disease are also summarized.
Collapse
Affiliation(s)
- Run-Ze Tian
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Dong-Lin Zhuang
- Department of Structural Heart Disease, National Center for Cardiovascular Disease, China & Fuwai Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chi Teng Vong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
- Macau Centre for Research and Development in Chinese Medicine, University of Macau, Macau, China
| | - Xuyu He
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Ouyang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Jing-Hua Liang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yan-Ping Guo
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yu-Hong Wang
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Shuang Zhao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China; and
| | - Haiyun Yuan
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Moussa Ide Nasser
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Ping Zhu
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Wu XW, Huang YX, Li CJ, Li YF, Wang BB, Zeb MA, Xiao WL, Zheng CB, Li XL. Podocarpane and cleistanthane diterpenoids from Strophioblachia glandulosa: structural elucidation, anti-hypertrophy activity and network pharmacology. Bioorg Chem 2025; 154:108026. [PMID: 39644615 DOI: 10.1016/j.bioorg.2024.108026] [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: 09/29/2024] [Revised: 11/21/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
In the present investigation, fourteen unprecedented podocarpane diterpenoids strophiolosas A-J, L-N and P (1-10, 12-14, 16), two new cleistanthane derivatives strophiolosas Q-R (17-18), two new dibenzopyroan-ones and one new tetralone strophiolosas S-U (19-21), were isolated from the whole plant of Strophioblachia glandulosa. The structures were elucidated via various spectroscopic analysis, quantum chemistry calculations, and X-ray diffraction. Bioactivity test indicated that compounds 5 and 17 possessed promising anti-cardiac hypertrophy effect in vitro (IC50 values of 16.50 and 9.67 μM). Additionally, through network pharmacology prediction, PARP1 may be a potential target of compound 17, mediating its anti-hypertrophic effects through multiple pathways.
Collapse
Affiliation(s)
- Xue-Wen Wu
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China
| | - Yong-Xiang Huang
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Chao-Jun Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China
| | - Yan-Fang Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China
| | - Bin-Bao Wang
- Faculty of Basic Medical Science, Kunming Medical University, Kunming 650500, People's Republic of China
| | - Muhammad Aurang Zeb
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China; Southwest United Graduate School, Kunming 650592, People's Republic of China.
| | - Chang-Bo Zheng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming 650500, People's Republic of China.
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, People's Republic of China.
| |
Collapse
|
28
|
Palazzolo S, Natale S, Capparucci F, Piro MG, Cuzzocrea S, Peritore AF, Crupi R, Britti D. Freshwater pollution: cardiotoxicity effect of perfluorooctane sulfonic acid and neonicotinoid imidacloprid mixture. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2025; 44:136-145. [PMID: 39887270 DOI: 10.1093/etojnl/vgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 02/01/2025]
Abstract
Perfluorooctanesulfonate (PFOS) is a widely used chemical that accumulates in living things and the environment, especially the aquatic, over time. It is also known as a "forever chemical". Furthermore, different anthropogenic substances are rarely found individually in the environment. Some of these substances are very toxic to aquatic species, such as imidacloprid (IMI), an insecticide belonging to the neonicotinoid family. The main objectives of this study were to investigate the effect of coexposure of these two contaminants at individual nontoxic concentration. In this study, we first analyzed different nominal concentrations of PFOS (from 0.1 to 10 μM) and IMI (from 75 to 1,000 μM) to highlight the morphological effects at 96 hr postfertilization and subsequently assessed the toxicity of mixture coexposure at both lethal and sublethal levels. Coexposure of PFOS and IMI at two individually nontoxic concentrations resulted in increased toxicity in terms of morphological alterations, accompanied by increased cell death in the pericardium. Molecular investigations confirmed the increased cardiotoxicity accompanied by cell death, showing overexpression of apoptosis-associated genes (caspase 3, bax, and bcl-2.) and a dysregulation of oxidative stress-related genes (cat, sod1, and gstp2). These results suggest that IMI could potentiate PFOS cardiotoxicity on zebrafish embryo development by alteration of antioxidative balance and induced apoptosis.
Collapse
Affiliation(s)
- Simone Palazzolo
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Sabrina Natale
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Fabiano Capparucci
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Maria Giovanna Piro
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical, and Environmental Science, University of Messina, Messina, Italy
- Link Campus University, Rome, Italy
| | | | - Rosalia Crupi
- Department of Veterinary Science, University of Messina, Messina, Italy
| | - Domenico Britti
- Department of Health Sciences, "Magna Græcia University" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
29
|
Alhasan KA, King MA, Pattar BSB, Lewis IA, Lopaschuk GD, Greenway SC. Anaplerotic filling in heart failure: a review of mechanism and potential therapeutics. Cardiovasc Res 2024; 120:2166-2178. [PMID: 39570879 PMCID: PMC11687400 DOI: 10.1093/cvr/cvae248] [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: 06/18/2024] [Revised: 09/08/2024] [Accepted: 11/12/2024] [Indexed: 01/01/2025] Open
Abstract
Heart failure (HF) is a complex syndrome and a leading cause of mortality worldwide. While current medical treatment is based on known pathophysiology and is effective for many patients, the underlying cellular mechanisms are poorly understood. Energy deficiency is a characteristic of HF, marked by complex alterations in metabolism. Within the tricarboxylic acid cycle, anaplerosis emerges as an essential metabolic process responsible for replenishing lost intermediates, thereby playing a crucial role in sustaining energy metabolism and consequently cardiac function. Alterations in cardiac anaplerosis are commonly observed in HF, demonstrating potential for therapeutic intervention. This review discusses recent advances in understanding the anaplerotic adaptations that occur in HF. We also explore therapeutics that can directly modulate anaplerosis or are likely to confer cardioprotective effects through anaplerosis, which could potentially be implemented to rescue the failing heart.
Collapse
Affiliation(s)
- Karm A Alhasan
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Pediatrics and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 1N4
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Melissa A King
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada T2N 1N4
| | - Badal S B Pattar
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| | - Ian A Lewis
- Alberta Centre for Advanced Diagnostics, Department of Biological Sciences, University of Calgary, Calgary, AB, Canada T2N 1N4
| | - Gary D Lopaschuk
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | - Steven C Greenway
- Department of Cardiac Sciences and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
- Department of Pediatrics and Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 1N4
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada T2N 4N1
| |
Collapse
|
30
|
Qin Y, Shirakawa J, Xu C, Chen R, Yang X, Ng C, Nakano S, Elguindy M, Deng Z, Prasanth KV, Eissmann MF, Nakagawa S, Ricci WM, Zhao B. Long non-coding RNA Malat1 fine-tunes bone homeostasis and repair by orchestrating cellular crosstalk and β-catenin-OPG/Jagged1 pathway. eLife 2024; 13:RP98900. [PMID: 39714456 DOI: 10.7554/elife.98900] [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] [Indexed: 12/24/2024] Open
Abstract
The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts, and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to β-catenin and functioning through the β-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.
Collapse
Affiliation(s)
- Yongli Qin
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
| | - Jumpei Shirakawa
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Cheng Xu
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Ruge Chen
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Xu Yang
- Research Institute, Hospital for Special Surgery, New York, United States
- Department of Orthopaedic Surgery, Weill Cornell Medicine, New York, United States
| | - Courtney Ng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Shinichi Nakano
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Mahmoud Elguindy
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Zhonghao Deng
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
| | - Kannanganattu V Prasanth
- Department of Cell and Developmental Biology, Cancer center at Illinois, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Moritz F Eissmann
- Institute for Tumor Biology and Experimental Therapy, Frankfurt, Germany
| | - Shinichi Nakagawa
- RNA Biology Laboratory, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - William M Ricci
- Orthopaedic Trauma Service, Hospital for Special Surgery & NewYork-Presbyterian Hospital, NewYork, United States
| | - Baohong Zhao
- Arthritis and Tissue Degeneration Program and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, United States
- Department of Medicine, Weill Cornell Medical College, New York, United States
- Graduate Program in Cell and Development Biology, Weill Cornell Graduate School of Medical Sciences, New York, United States
| |
Collapse
|
31
|
Zhi Y, Duan Y, Zhang Y, Hu H, Hu F, Wang P, Liu B, Wang C, Liu D, Gu G. miR-421-mediated suppression of FGF13 as a novel mechanism ameliorates cardiac hypertrophy by inhibiting endoplasmic reticulum stress. Eur J Pharmacol 2024; 985:177085. [PMID: 39486770 DOI: 10.1016/j.ejphar.2024.177085] [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: 05/17/2024] [Revised: 09/23/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Pathological cardiac hypertrophy is an independent risk factor for heart failure. Currently, clinical treatments offer limited effectiveness, and both mortality and morbidity from cardiac hypertrophy and heart failure continue to be significant. Therefore, it is extremely urgent to find new intervention targets to prevent and alleviate pathological cardiac hypertrophy. In this study, we explored FGF13 expression and its upstream regulators in hypertrophic hearts. Firstly, we observed an increase in FGF13 expression levels in human hypertrophic myocardium tissues, as well as in mouse models of TAC-induced hypertrophy and in neonatal rat cardiomyocyte (NRCM) models induced by isoproterenol (ISO). Moreover, these elevated levels of FGF13 were shown to positively correlate with hypertrophic markers, including ANP and BNP. By using both gain-of-function and loss-of-function approaches in an in vitro hypertrophy model, we demonstrated that FGF13 knockdown could inhibit endoplasmic reticulum stress (ERS), thereby ameliorating cardiomyocyte hypertrophy. Meanwhile, we investigated the upstream regulators of FGF13 in hypertrophic hearts, and a dual-luciferase reporter assay confirmed that FGF13 is a direct target of miR-421. Overexpression of miR-421 decreased the protein level of FGF13 and ameliorated ISO-induced cardiomyocyte hypertrophy via modulating ER stress. In contrast, overexpression of FGF13 attenuated the ameliorative effect of miR-421 on ISO-induced cardiomyocyte hypertrophy. Taken together, the present results suggested that miR-421 ameliorated ISO-induced cardiomyocyte hypertrophy by negatively regulating FGF13 expression. This finding may offer a novel approach for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Yaxin Zhi
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yanru Duan
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Ying Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Haijuan Hu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Fengli Hu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Pengfei Wang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Bin Liu
- Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chuan Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, 050000, China.
| | - Demin Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Guoqiang Gu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
32
|
Polecka A, Nawrocki J, Pulido MA, Olszewska E. Mandibular Advancement Devices in Obstructive Sleep Apnea and Its Effects on the Cardiovascular System: A Comprehensive Literature Review. J Clin Med 2024; 13:6757. [PMID: 39597901 PMCID: PMC11594591 DOI: 10.3390/jcm13226757] [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: 10/06/2024] [Revised: 11/03/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Obstructive sleep apnea syndrome (OSA) is a chronic inflammatory disease characterized by endothelial dysfunction and cardiovascular complications. Continuous positive airway pressure (CPAP) is the standard treatment, hence poor adherence has prompted interest in mandibular advancement devices (MAD) as an alternative. This comprehensive review aimed to explore the effects of MAD therapy on oxidative stress, inflammation, endothelial function, and its impact on the cardiovascular risk in OSA patients. RESULTS MAD therapy significantly reduces the apnea-hypopnea index (AHI), improves serum nitric oxide (NOx) concentrations, reduces oxidative stress markers, and enhances endothelial function. Animal studies indicated that MAD reduces myocardial fibrosis and attenuates inflammatory markers. While both CPAP and MADs improve endothelial function and heart rate variability, CPAP is more effective in reducing OSA severity. Nevertheless, MAD has higher compliance, contributing to its positive impact on cardiovascular function. Moreover, CPAP and MADs have similar effectiveness in reducing cardiovascular risk. CONCLUSIONS MAD therapy is an effective alternative to CPAP, particularly for patients with mild to moderate OSA as well as those intolerant to CPAP. It offers significant improvements in endothelial function and oxidative stress. Further studies are needed to assess MAD therapy in comprehensive OSA management.
Collapse
Affiliation(s)
- Agnieszka Polecka
- Department of Cardiology and Internal Medicine with Cardiac Intensive Care Unit, Doctoral School of the Medical University of Bialystok, Medical University of Bialystok, 15-089 Bialystok, Poland
| | - Jakub Nawrocki
- Clinic of Orthodontics, Wroclaw University Dental Center, Krakowska 26, 50-425 Wroclaw, Poland;
| | | | - Ewa Olszewska
- Sleep Apnea Surgery Center, Department of Otolaryngology, Medical University of Bialystok, 15-089 Bialystok, Poland
| |
Collapse
|
33
|
Maruyama N, Ogata T, Kasahara T, Hamaoka T, Higuchi Y, Tsuji Y, Tomita S, Sakamoto A, Nakanishi N, Matoba S. Loss of Cavin-2 destabilizes phosphatase and tensin homologue and enhances Akt signalling pathway in cardiomyocytes. Cardiovasc Res 2024; 120:1562-1576. [PMID: 38861679 DOI: 10.1093/cvr/cvae130] [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: 09/10/2023] [Revised: 03/19/2024] [Accepted: 05/03/2024] [Indexed: 06/13/2024] Open
Abstract
AIMS Specific cavins and caveolins, known as caveola-related proteins, have been implicated in cardiac hypertrophy and myocardial injury. Cavin-2 forms complexes with other caveola-related proteins, but the role of Cavin-2 in cardiomyocytes (CMs) is poorly understood. Here, we investigated an unknown function of Cavin-2 in CMs. METHODS AND RESULTS Under cardiac stress-free conditions, systemic Cavin-2 knockout (KO) induced mild and significant CM hypertrophy. Cavin-2 KO suppressed phosphatase and tensin homologue (PTEN) associated with Akt signalling, whereas there was no difference in Akt activity between the hearts of the wild-type and the Cavin-2 KO mice under cardiac stress-free conditions. However, after swim training, CM hypertrophy was more facilitated with enhanced phosphoinositide 3-kinase (PI3K)-Akt activity in the hearts of Cavin-2 KO mice. Cavin-2 knockdown neonatal rat CMs (NRCMs) using adenovirus expressing Cavin-2 short hairpin RNA were hypertrophied and resistant to hypoxia and H2O2-induced apoptosis. Cavin-2 knockdown increased Akt phosphorylation in NRCMs, and an Akt inhibitor inhibited Cavin-2 knockdown-induced anti-apoptotic responses in a dose-dependent manner. Cavin-2 knockdown increased phosphatidylinositol-3,4,5-triphosphate production and attenuated PTEN at the membrane fraction of NRCMs. Immunostaining and immunoprecipitation showed that Cavin-2 was associated with PTEN at the plasma membrane of NRCMs. A protein stability assay showed that Cavin-2 knockdown promoted PTEN destabilization in NRCMs. In an Angiotensin II (2-week continuous infusion)-induced pathological cardiac hypertrophy model, CM hypertrophy and CM apoptosis were suppressed in CM-specific Cavin-2 conditional KO (Cavin-2 cKO) mice. Because Cavin-2 cKO mouse hearts showed increased Akt activity but not decreased extracellular signal-regulated kinase activity, suppression of pathological hypertrophy by Cavin-2 loss may be due to increased survival of healthy CMs. CONCLUSION Cavin-2 plays a negative regulator in the PI3K-Akt signalling in CMs through interaction with PTEN. Loss of Cavin-2 enhances Akt activity by promoting PTEN destabilization, which promotes physiological CM hypertrophy and may enhance Akt-mediated cardioprotective effects against pathological CM hypertrophy.
Collapse
Affiliation(s)
- Naoki Maruyama
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Pathology and Cell Regulation, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Takeru Kasahara
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tetsuro Hamaoka
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Akira Sakamoto
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Naohiko Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
34
|
Yin W, Chen Y, Wang W, Guo M, Tong L, Zhang M, Wang Z, Yuan H. Macrophage-mediated heart repair and remodeling: A promising therapeutic target for post-myocardial infarction heart failure. J Cell Physiol 2024; 239:e31372. [PMID: 39014935 DOI: 10.1002/jcp.31372] [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: 03/04/2024] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 07/18/2024]
Abstract
Heart failure (HF) remains prevalent in patients who survived myocardial infarction (MI). Despite the accessibility of the primary percutaneous coronary intervention and medications that alleviate ventricular remodeling with functional improvement, there is an urgent need for clinicians and basic scientists to further reveal the mechanisms behind post-MI HF as well as investigate earlier and more efficient treatment after MI. Growing numbers of studies have highlighted the crucial role of macrophages in cardiac repair and remodeling following MI, and timely intervention targeting the immune response via macrophages may represent a promising therapeutic avenue. Recently, technology such as single-cell sequencing has provided us with an updated and in-depth understanding of the role of macrophages in MI. Meanwhile, the development of biomaterials has made it possible for macrophage-targeted therapy. Thus, an overall and thorough understanding of the role of macrophages in post-MI HF and the current development status of macrophage-based therapy will assist in the further study and development of macrophage-targeted treatment for post-infarction cardiac remodeling. This review synthesizes the spatiotemporal dynamics, function, mechanism and signaling of macrophages in the process of HF after MI, as well as discusses the emerging bio-materials and possible therapeutic agents targeting macrophages for post-MI HF.
Collapse
Affiliation(s)
- Wenchao Yin
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yong Chen
- Department of Emergency, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Wenjun Wang
- Department of Intensive Care Unit, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Mengqi Guo
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Lingjun Tong
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingxiang Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Department of Cardiology, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zhaoyang Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
35
|
Eteng OE, Ugwor EI, James AS, Moses CA, Ogbonna CU, Iwara IA, Akamo AJ, Akintunde JK, Blessing OA, Tola YM, Eru EM, Igiri AO. Vanillic acid ameliorates diethyl phthalate and bisphenol S-induced oxidative stress and neuroinflammation in the hippocampus of experimental rats. J Biochem Mol Toxicol 2024; 38:e70017. [PMID: 39415675 DOI: 10.1002/jbt.70017] [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: 12/11/2023] [Revised: 09/18/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
Long-term adverse effects on human health are caused by exogenous compounds that alter the functions of biological systems, especially neuroendocrine disruptors like diethyl phthalate (DEP) and bisphenol S (BPS). Although vanillic acid (VA) has pertinent neuropharmacological characteristics, its effect against DEP + BPS-induced neurotoxicity has not been explored. This study proposed that VA may offer protection against the neurotoxicity caused by DEP + BPS. Thirty male Wistar rats were randomly distributed across five groups: a control group receiving DMSO, a group exposed to a mixture of BPS and DEP, two BPS + DEP-exposed groups treated with VA at doses of 25 mg/kg or 50 mg/kg, and a nonexposed group treated with 50 mg VA/kg. After 21 days, the hippocampal tissues were processed for biochemical analyses. Our results indicate that exposure to DEP + BPS upregulated neurosignaling mediators (NTPDase, ADA, MAO-A, and Ca2+), inhibited others (AChE and Ca2+/Mg2+-ATPase), decreased hippocampus antioxidants (GSH, GPx, CAT, and SOD), and elevated markers of oxidative stress/damage (NO, H2O2, MDA, and AOPP). AR, BAX, TNF-α, BAK1, and IL-1β expressions were upregulated, while IL-10 and BDNF expressions were downregulated. NF-κB and caspase-3/9 pathways were also upregulated. Co-treatment with vanillic acid remarkably precluded these neurotoxic outcomes by improving neurosignaling, augmenting antioxidant status, abrogating oxidative damage, inflammation (TNF-α, IL-1β), and apoptosis (BAX, BAK1, caspase-3/9). Vanillic acid also restored IL-10 and BDNF levels, thereby exhibiting neuroprotective effects, corroborated by histological examinations. We posit vanillic acid as a safe and effective therapeutic agent against neurotoxicity occasioned by exposure to neuroendocrine disruptors.
Collapse
Affiliation(s)
- Ofem Effiom Eteng
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | | | - Adewale Segun James
- Department of Chemical Sciences, Faculty of Science, Augustine University, Ilara-Epe P.M.B1100, Lagos State, Nigeria
| | - Ceaser Antiya Moses
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Chukwuka Uzoamaka Ogbonna
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
- Department of Biochemistry, Pollution Control and Biotechnology Unit, University of Nigeria, Nsukka, Enugu State, Nigeria
| | - Iwara Arikpo Iwara
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Adio Jamiu Akamo
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Jacob Kehinde Akintunde
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | | | - Yinus Mujeeb Tola
- Department of Biochemistry, Federal University of Agriculture, Abeokuta, Ogun State, Nigeria
| | - Eru Mba Eru
- Department of Biochemistry, University of Calabar, Cross River State
| | | |
Collapse
|
36
|
Zhou H, Liu P, Guo X, Fang W, Wu C, Zhang M, Ji Z. Fibroblast-derived miR-425-5p alleviates cardiac remodelling in heart failure via inhibiting the TGF-β1/Smad signalling. J Cell Mol Med 2024; 28:e70199. [PMID: 39527465 PMCID: PMC11552651 DOI: 10.1111/jcmm.70199] [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: 06/28/2024] [Revised: 10/15/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
The pathological activation of cardiac fibroblasts (CFs) plays a crucial role in the development of pressure overload-induced cardiac remodelling and subsequent heart failure (HF). Growing evidence demonstrates that multiple microRNAs (miRNAs) are abnormally expressed in the pathophysiologic process of cardiovascular diseases, with miR-425 recently reported to be potentially involved in HF. In this study, we aimed to investigate the effects of fibroblast-derived miR-425-5p in pressure overload-induced HF and explore the underlying mechanisms. C57BL/6 mice were injected with a recombinant adeno-associated virus specifically designed to overexpress miR-425-5p in CFs, followed by transverse aortic constriction (TAC) surgery. Neonatal mouse CFs (NMCFs) were transfected with miR-425-5p mimics and subsequently stimulated with angiotensin II (Ang II). We found that miR-425-5p levels were significantly downregulated in HF mice and Ang II-treated NMCFs. Notably, fibroblast-specific overexpression of miR-425-5p markedly inhibited the proliferation and differentiation of CFs, thereby alleviating myocardial fibrosis, cardiac hypertrophy and systolic dysfunction. Mechanistically, the cardioprotective actions of miR-425-5p may be achieved by targeting the TGF-β1/Smad signalling. Interestingly, miR-425-5p mimics-treated CFs could also indirectly affect cardiomyocyte hypertrophy in this course. Together, our findings suggest that fibroblast-derived miR-425-5p mitigates TAC-induced HF, highlighting miR-425-5p as a potential diagnostic and therapeutic target for treating HF patients.
Collapse
Affiliation(s)
- Haijia Zhou
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Pengyun Liu
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Xuelin Guo
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Wei Fang
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Chan Wu
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Mingming Zhang
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| | - Zhaole Ji
- Department of CardiologyTangdu Hospital, Second Affiliated Hospital of Air Force Medical UniversityXi'anChina
| |
Collapse
|
37
|
Wang C, Lai Z, Tan H, Zhang H, Tan L, Luo Q, Li S, Xiong Z, Yang G, Xiong Z. Impaired cardiomyocytes accelerate cardiac hypertrophy and fibrosis by delivering exosomes containing Shh/N-Shh/Gli1 in angiotensin II infused mice. Heliyon 2024; 10:e39332. [PMID: 39640644 PMCID: PMC11620221 DOI: 10.1016/j.heliyon.2024.e39332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/11/2024] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Backgrounds Heart failure (HF) is characterized by progressive cardiac hypertrophy and fibrosis, yet the underlying pathological mechanisms remain unclear. Exosomes are pivotal in cellular communication and are key signaling carriers in HFs. This study investigated the roles of exosomes in HF. Methods Eight-week-old male mice were divided into three groups: a control group, an Ang II group receiving angiotensin II (Ang II) infusion for 4 weeks, and an Ang II + DMA group receiving Ang II and dimethyl amiloride (DMA) infusion. This study examined the associations between cardiac injury, exosomes, and their substrate Shh. Furthermore, we conducted cellular experiments to assess the effects of Ang II-induced injury in primary cardiomyocytes on other cardiomyocytes and fibroblasts, and to test the therapeutic effects of the exosome inhibitor DMA and the Shh signaling inhibitor cyclopamine (CPN). Results Ang II-induced cardiac hypertrophy and fibrosis, which were accompanied by exosome secretion and Shh upregulation in vivo. DMA relieved these cardiac lesions. Furthermore, cellular experiments revealed that Ang II-induced cardiomyocytes hypertrophy and activated cardiac fibroblasts by promoting the release of exosomes containing Shh/N-Shh/Gli1. Both DMA and CPN nullified fibroblast activation and proliferation. Conclusions Ang II-induced cardiomyocyte injury leads to cardiac hypertrophy and fibrosis through the release of exosomes carrying Shh signaling. The suppression of exosome secretion or the Shh pathway could offer new strategies for treating HF.
Collapse
Affiliation(s)
- Cong Wang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Zhiwei Lai
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Huishi Tan
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hua Zhang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Lishan Tan
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Qingyun Luo
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Sanmu Li
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Zibo Xiong
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, Guangdong, China
| | - Guang Yang
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, Guangdong, China
- Institute of Nephrology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Zuying Xiong
- Division of Renal Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
- Shenzhen Clinical Research Center for Urology and Nephrology, Shenzhen, Guangdong, China
- Institute of Nephrology, Shenzhen Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| |
Collapse
|
38
|
Appunni S, Rubens M, Ramamoorthy V, Saxena A, McGranaghan P, Khosla A, Doke M, Chaparro S, Jimenez J. Molecular remodeling in comorbidities associated with heart failure: a current update. Mol Biol Rep 2024; 51:1092. [PMID: 39460797 PMCID: PMC11512903 DOI: 10.1007/s11033-024-10024-7] [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: 05/19/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Recent advances in genomics and proteomics have helped in understanding the molecular mechanisms and pathways of comorbidities and heart failure. In this narrative review, we reviewed molecular alterations in common comorbidities associated with heart failure such as obesity, diabetes mellitus, systemic hypertension, pulmonary hypertension, coronary artery disease, hypercholesteremia and lipoprotein abnormalities, chronic kidney disease, and atrial fibrillation. We searched the electronic databases, PubMed, Ovid, EMBASE, Google Scholar, CINAHL, and PhysioNet for articles without time restriction. Although the association between comorbidities and heart failure is already well established, recent studies have explored the molecular pathways in much detail. These molecular pathways demonstrate how novels drugs for heart failure works with respect to the pathways associated with comorbidities. Understanding the altered molecular milieu in heart failure and associated comorbidities could help to develop newer medications and targeted therapies that incorporate these molecular alterations as well as key molecular variations across individuals to improve therapeutic outcomes. The molecular alterations described in this study could be targeted for novel and personalized therapeutic approaches in the future. This knowledge is also critical for developing precision medicine strategies to improve the outcomes for patients living with these conditions.
Collapse
Affiliation(s)
| | - Muni Rubens
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL, USA
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Universidad Espíritu Santo, Samborondón, Ecuador
| | | | - Anshul Saxena
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Center for Advanced Analytics, Baptist Health South Florida, Miami, FL, USA
| | - Peter McGranaghan
- Semmelweis University, Budapest, Hungary.
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Augustenburger Platz 1, 10117, Berlin, Germany.
| | - Atulya Khosla
- William Beaumont University Hospital, Royal Oak, MI, USA
| | | | - Sandra Chaparro
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA
- Miami Cardiac & Vascular Institute, Baptist Health South Florida, Miami, FL, USA
| | - Javier Jimenez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL, USA.
- Miami Cardiac & Vascular Institute, Baptist Health South Florida, Miami, FL, USA.
- Advance Heart Failure and Pulmonary Hypertension, South Miami Hospital, Baptist Health South, Miami, FL, USA.
| |
Collapse
|
39
|
Zakaria ZZ, Suleiman M, Benslimane FM, Al-Badr M, Sivaraman S, Korashy HM, Ahmad F, Uddin S, Mraiche F, Yalcin HC. Imatinib‑ and ponatinib‑mediated cardiotoxicity in zebrafish embryos and H9c2 cardiomyoblasts. Mol Med Rep 2024; 30:187. [PMID: 39219269 PMCID: PMC11350628 DOI: 10.3892/mmr.2024.13311] [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: 08/04/2023] [Accepted: 04/26/2024] [Indexed: 09/04/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) offer targeted therapy for cancers but can cause severe cardiotoxicities. Determining their dose‑dependent impact on cardiac function is required to optimize therapy and minimize adverse effects. The dose‑dependent cardiotoxic effects of two TKIs, imatinib and ponatinib, were assessed in vitro using H9c2 cardiomyoblasts and in vivo using zebrafish embryos. In vitro, H9c2 cardiomyocyte viability, apoptosis, size, and surface area were evaluated to assess the impact on cellular health. In vivo, zebrafish embryos were analyzed for heart rate, blood flow velocity, and morphological malformations to determine functional and structural changes. Additionally, reverse transcription‑quantitative PCR (RT‑qPCR) was employed to measure the gene expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP), established markers of cardiac injury. This comprehensive approach, utilizing both in vitro and in vivo models alongside functional and molecular analyses, provides a robust assessment of the potential cardiotoxic effects. TKI exposure decreased viability and surface area in H9c2 cells in a dose‑dependent manner. Similarly, zebrafish embryos exposed to TKIs exhibited dose‑dependent heart malformation. Both TKIs upregulated ANP and BNP expression, indicating heart injury. The present study demonstrated dose‑dependent cardiotoxic effects of imatinib and ponatinib in H9c2 cells and zebrafish models. These findings emphasize the importance of tailoring TKI dosage to minimize cardiac risks while maintaining therapeutic efficacy. Future research should explore the underlying mechanisms and potential mitigation strategies of TKI‑induced cardiotoxicities.
Collapse
Affiliation(s)
- Zain Z. Zakaria
- Vice President of Health and Medical Sciences Office, QU Health, Qatar University, Doha 2713, Qatar
| | - Muna Suleiman
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | | | - Mashael Al-Badr
- Department of Biology, College of Art and Science, Qatar University, Doha 2713, Qatar
- National Reference Laboratory, Ministry of Public Health, Doha 7744, Qatar
| | - Siveen Sivaraman
- Translational Research Institute, Hamad Medical Corporation, Doha 3050, Qatar
| | - Hesham M. Korashy
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | - Fareed Ahmad
- Translational Research Institute and Dermatology Institute, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Hamad Medical Corporation, Doha 3050, Qatar
- Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Fatima Mraiche
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar
| | | |
Collapse
|
40
|
Li Y, Li Z, Ren Y, Lei Y, Yang S, Shi Y, Peng H, Yang W, Guo T, Yu Y, Xiong Y. Mitochondrial-derived peptides in cardiovascular disease: Novel insights and therapeutic opportunities. J Adv Res 2024; 64:99-115. [PMID: 38008175 PMCID: PMC11464474 DOI: 10.1016/j.jare.2023.11.018] [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: 09/20/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 11/28/2023] Open
Abstract
BACKGROUND Mitochondria-derived peptides (MDPs) represent a recently discovered family of peptides encoded by short open reading frames (ORFs) found within mitochondrial genes. This group includes notable members including humanin (HN), mitochondrial ORF of the 12S rDNA type-c (MOTS-c), and small humanin-like peptides 1-6 (SHLP1-6). MDPs assume pivotal roles in the regulation of diverse cellular processes, encompassing apoptosis, inflammation, and oxidative stress, which are all essential for sustaining cellular viability and normal physiological functions. Their emerging significance extends beyond this, prompting a deeper exploration into their multifaceted roles and potential applications. AIM OF REVIEW This review aims to comprehensively explore the biogenesis, various types, and diverse functions of MDPs. It seeks to elucidate the central roles and underlying mechanisms by which MDPs participate in the onset and development of cardiovascular diseases (CVDs), bridging the connections between cell apoptosis, inflammation, and oxidative stress. Furthermore, the review highlights recent advancements in clinical research related to the utilization of MDPs in CVD diagnosis and treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW MDPs levels are diminished with aging and in the presence of CVDs, rendering them potential new indicators for the diagnosis of CVDs. Also, MDPs may represent a novel and promising strategy for CVD therapy. In this review, we delve into the biogenesis, various types, and diverse functions of MDPs. We aim to shed light on the pivotal roles and the underlying mechanisms through which MDPs contribute to the onset and advancement of CVDs connecting cell apoptosis, inflammation, and oxidative stress. We also provide insights into the current advancements in clinical research related to the utilization of MDPs in the treatment of CVDs. This review may provide valuable information with MDPs for CVD diagnosis and treatment.
Collapse
Affiliation(s)
- Yang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Zhuozhuo Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Ying Lei
- School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Silong Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuqi Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Han Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Weijie Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Tiantian Guo
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; School of Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, College of Life Sciences, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
41
|
Shao Y, Xu J, Chen W, Hao M, Liu X, Zhang R, Wang Y, Dong Y. miR-135b: An emerging player in cardio-cerebrovascular diseases. J Pharm Anal 2024; 14:100997. [PMID: 39211791 PMCID: PMC11350494 DOI: 10.1016/j.jpha.2024.100997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 09/04/2024] Open
Abstract
miR-135 is a highly conserved miRNA in mammals and includes miR-135a and miR-135b. Recent studies have shown that miR-135b is a key regulatory factor in cardio-cerebrovascular diseases. It is involved in regulating the pathological process of myocardial infarction, myocardial ischemia/reperfusion injury, cardiac hypertrophy, atrial fibrillation, diabetic cardiomyopathy, atherosclerosis, pulmonary hypertension, cerebral ischemia/reperfusion injury, Parkinson's disease, and Alzheimer's disease. Obviously, miR-135b is an emerging player in cardio-cerebrovascular diseases and is expected to be an important target for the treatment of cardio-cerebrovascular diseases. However, the crucial role of miR-135b in cardio-cerebrovascular diseases and its underlying mechanism of action has not been reviewed. Therefore, in this review, we aimed to comprehensively summarize the role of miR-135b and the signaling pathway mediated by miR-135b in cardio-cerebrovascular diseases. Drugs targeting miR-135b for the treatment of diseases and related patents, highlighting the importance of this target and its utility as a therapeutic target for cardio-cerebrovascular diseases, have been discussed.
Collapse
Affiliation(s)
- Yingchun Shao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Wujun Chen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Xinlin Liu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Renshuai Zhang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Yanhong Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
| | - Yinying Dong
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, China
- Department of Radiation Oncology, The Affiliated Hospital of Qingdao University, Qingdao, 266071, China
| |
Collapse
|
42
|
Wu S, Ding D, Wang D. Regulated Cell Death Pathways in Pathological Cardiac Hypertrophy. Rev Cardiovasc Med 2024; 25:366. [PMID: 39484135 PMCID: PMC11522757 DOI: 10.31083/j.rcm2510366] [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: 03/20/2024] [Revised: 05/19/2024] [Accepted: 06/03/2024] [Indexed: 11/03/2024] Open
Abstract
Cardiac hypertrophy is characterized by an increased volume of individual cardiomyocytes rather than an increase in their number. Myocardial hypertrophy due to pathological stimuli encountered by the heart, which reduces pressure on the ventricular walls to maintain cardiac function, is known as pathological hypertrophy. This eventually progresses to heart failure. Certain varieties of regulated cell death (RCD) pathways, including apoptosis, pyroptosis, ferroptosis, necroptosis, and autophagy, are crucial in the development of pathological cardiac hypertrophy. This review summarizes the molecular mechanisms and signaling pathways underlying these RCD pathways, focusing on their mechanism of action findings for pathological cardiac hypertrophy. It intends to provide new ideas for developing therapeutic approaches targeted at the cellular level to prevent or reverse pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Shengnan Wu
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| | - Ding Ding
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| | - Deguo Wang
- Department of Geriatrics, The First Affiliated Hospital of Wannan Medical College, 241001 Wuhu, Anhui, China
| |
Collapse
|
43
|
Zhang D, Wen Q, Zhang R, Kou K, Lin M, Zhang S, Yang J, Shi H, Yang Y, Tan X, Yin S, Ou X. From Cell to Gene: Deciphering the Mechanism of Heart Failure With Single-Cell Sequencing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308900. [PMID: 39159065 PMCID: PMC11497092 DOI: 10.1002/advs.202308900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 07/24/2024] [Indexed: 08/21/2024]
Abstract
Heart failure (HF) is a prevalent cardiovascular disease with significant morbidity and mortality rates worldwide. Due to the intricate structure of the heart, diverse cell types, and the complex pathogenesis of HF, further in-depth investigation into the underlying mechanisms is required. The elucidation of the heterogeneity of cardiomyocytes and the intercellular communication network is particularly important. Traditional high-throughput sequencing methods provide an average measure of gene expression, failing to capture the "heterogeneity" between cells and impacting the accuracy of gene function knowledge. In contrast, single-cell sequencing techniques allow for the amplification of the entire genome or transcriptome at the individual cell level, facilitating the examination of gene structure and expression with unparalleled precision. This approach offers valuable insights into disease mechanisms, enabling the identification of changes in cellular components and gene expressions during hypertrophy associated with HF. Moreover, it reveals distinct cell populations and their unique roles in the HF microenvironment, providing a comprehensive understanding of the cellular landscape that underpins HF pathogenesis. This review focuses on the insights provided by single-cell sequencing techniques into the mechanisms underlying HF and discusses the challenges encountered in current cardiovascular research.
Collapse
Affiliation(s)
- Dan Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- Department of Rehabilitation MedicineSouthwest Medical UniversityLuzhouSichuan646000China
| | - Qiang Wen
- Department of CardiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang RdWuhanHubei430022China
| | - Rui Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Kun Kou
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Miao Lin
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Shiyu Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Jun Yang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Hangchuan Shi
- Department of Clinical & Translational ResearchUniversity of Rochester Medical Center265 Crittenden BlvdRochesterNY14642USA
- Department of Pathology and Laboratory MedicineUniversity of Rochester Medical Center601 Elmwood AveRochesterNY14642USA
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
| | - Xiaoqiu Tan
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- Department of PhysiologySchool of Basic Medical SciencesSouthwest Medical UniversityLuzhouSichuan646000China
| | - Shigang Yin
- Luzhou Key Laboratory of Nervous system disease and Brain FunctionSouthwest Medical UniversityLuzhouSichuan646000China
| | - Xianhong Ou
- Key Laboratory of Medical Electrophysiology of Ministry of EducationInstitute of Cardiovascular MedicineDepartment of Cardiology of the Affiliated HospitalSouthwest Medical UniversityLuzhouSichuan646000China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal ResourcesGuangxi Normal UniversityGuilinGuangxi541004China
| |
Collapse
|
44
|
Du P, Xu L, Wang Y, Jiao T, Cheng J, Zhang C, Tapu MSR, Dai J, Li J. Astragaloside IV ameliorates pressure overload-induced heart failure by enhancing angiogenesis through HSF1/VEGF pathway. Heliyon 2024; 10:e37019. [PMID: 39296120 PMCID: PMC11408759 DOI: 10.1016/j.heliyon.2024.e37019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Astragaloside IV(AS-IV), the main active ingredient of Astragalus, has been used as a treatment for heart failure with favorable effects, but its molecular mechanism has not been fully elucidated. Network pharmacological analysis and molecular docking revealed that Heat shock transcription factor 1 (HSF1) is a potential target of AS-IV. We designed cellular and animal experiments to investigate the role and intrinsic molecular mechanisms of AS-IV in ameliorating pressure overload-induced heart failure. In cellular experiments, Myocardial microvascular endothelial cells (MMVECs) were cultured in isolation and stimulated by adding high and low concentrations of AS-IV, and a cell model with down-regulation of HSF1 expression was constructed by using siRNA technology. Changes in the expression of key molecules of HSF1/VEGF signaling pathway and differences in tube-forming ability were detected in different groups of cells using PCR, WB and tube-forming assay. In animal experiments, TAC technology was applied to establish a pressure overload-induced heart failure model in C57 mice, postoperative mice were ingested AS-IV by gavage, and adenoviral transfection technology was applied to construct a mouse model with down-regulation of HSF1 expression.Small animal ultrasound for cardiac function assessment, MASSON staining, CD31 immunohistochemistry, and Western blotting (WB) were performed on the mice. The results showed that AS-IV could promote the expression of key molecules of HSF1/VEGF signaling pathway, enhance the tube-forming ability of MMVECs, increase the density of myocardial capillaries, reduce myocardial fibrosis, and improve the cardiac function of mice with TAC.AS-IV could modulate the HSF1/VEGF signaling pathway to promote the angiogenesis and improve the pressure overload-induced heart failure.
Collapse
Affiliation(s)
- Peizhao Du
- Department of Cardiology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, China
| | - Linghao Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yuanqi Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Tiantian Jiao
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jing Cheng
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chunsheng Zhang
- Department of Cardiology, East Hospital of Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Md Sakibur Rahman Tapu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jian Dai
- Department of Cardiology, Baoshan District Hospital of Integrated Traditional Chinese and Western Medicine of Shanghai, Shanghai University of Traditional Chinese Medicine, Shanghai, 201999, China
| | - Jiming Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| |
Collapse
|
45
|
Sigle M, Rohlfing AK, Cruz Santos M, Kopp T, Krutzke K, Gidlund V, Kollotzek F, Marzi J, von Ungern-Sternberg S, Poso A, Heikenwälder M, Schenke-Layland K, Seizer P, Möllmann J, Marx N, Feil R, Feil S, Lukowski R, Borst O, Schäffer TE, Müller KAL, Gawaz MP, Heinzmann D. Targeting Cyclophilin A in the Cardiac Microenvironment Preserves Heart Function and Structure in Failing Hearts. Circ Res 2024; 135:758-773. [PMID: 39140165 DOI: 10.1161/circresaha.124.324812] [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: 04/27/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Cardiac hypertrophy is characterized by remodeling of the myocardium, which involves alterations in the ECM (extracellular matrix) and cardiomyocyte structure. These alterations critically contribute to impaired contractility and relaxation, ultimately leading to heart failure. Emerging evidence implicates that extracellular signaling molecules are critically involved in the pathogenesis of cardiac hypertrophy and remodeling. The immunophilin CyPA (cyclophilin A) has been identified as a potential culprit. In this study, we aimed to unravel the interplay between eCyPA (extracellular CyPA) and myocardial dysfunction and evaluate the therapeutic potential of inhibiting its extracellular accumulation to improve heart function. METHODS Employing a multidisciplinary approach encompassing in silico, in vitro, in vivo, and ex vivo experiments we studied a mouse model of cardiac hypertrophy and human heart specimen to decipher the interaction of CyPA and the cardiac microenvironment in highly relevant pre-/clinical settings. Myocardial expression of CyPA (immunohistology) and the inflammatory transcriptome (NanoString) was analyzed in human cardiac tissue derived from patients with nonischemic, noninflammatory congestive heart failure (n=187). These analyses were paralleled by a mouse model of Ang (angiotensin) II-induced heart failure, which was assessed by functional (echocardiography), structural (immunohistology, atomic force microscopy), and biomolecular (Raman spectroscopy) analyses. The effect of inhibiting eCyPA in the cardiac microenvironment was evaluated using a newly developed neutralizing anti-eCyPA monoclonal antibody. RESULTS We observed a significant accumulation of eCyPA in both human and murine-failing hearts. Importantly, higher eCyPA expression was associated with poor clinical outcomes in patients (P=0.043) and contractile dysfunction in mice (Pearson correlation coefficient, -0.73). Further, myocardial expression of eCyPA was critically associated with an increase in myocardial hypertrophy, inflammation, fibrosis, stiffness, and cardiac dysfunction in vivo. Antibody-based inhibition of eCyPA prevented (Ang II)-induced myocardial remodeling and dysfunction in mice. CONCLUSIONS Our study provides strong evidence of the pathogenic role of eCyPA in remodeling, myocardial stiffening, and dysfunction in heart failure. The findings suggest that antibody-based inhibition of eCyPA may offer a novel therapeutic strategy for nonischemic heart failure. Further research is needed to evaluate the translational potential of these interventions in human patients with cardiac hypertrophy.
Collapse
Affiliation(s)
- Manuel Sigle
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
| | - Anne-Katrin Rohlfing
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
| | - Melanie Cruz Santos
- Institute of Pharmacy, Pharmacology, Toxicology and Clinical Pharmacy (M.C.S., R.L.), University of Tübingen, Germany
| | - Timo Kopp
- Interfaculty Institute of Biochemistry (IFIB) (T.K., R.F., S.F.), University of Tübingen, Germany
| | - Konstantin Krutzke
- Institute for Applied Physics (K.K., V.G., T.E.S.), University of Tübingen, Germany
| | - Vincent Gidlund
- Interfaculty Institute of Biochemistry (IFIB) (T.K., R.F., S.F.), University of Tübingen, Germany
- Institute for Applied Physics (K.K., V.G., T.E.S.), University of Tübingen, Germany
| | - Ferdinand Kollotzek
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
- DFG Heisenberg Group Cardiovascular Thrombo-Inflammation and Translational Thrombocardiology (F.K., O.B.), University of Tübingen, Germany
| | - Julia Marzi
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine (J. Marzi, K.S.-L.), Eberhard Karls University Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," (J. Marzi, A.P., K.S.-L.), University of Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen Reutlingen, Germany (J. Marzi, K.S.-L.)
| | - Saskia von Ungern-Sternberg
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
- Now with Center for Thrombosis and Hemostasis (CTH), University Medical Center of the Johannes Gutenberg-University Mainz, Germany (S.U.-S.)
| | - Antti Poso
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," (J. Marzi, A.P., K.S.-L.), University of Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland Kuopio (A.P.)
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard-Karls-Universität Tübingen, Germany (A.P.)
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany (A.P.)
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Centre Heidelberg (DKFZ), Germany (M.H.)
- University Tübingen, Faculty of Medicine, Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome (M.H.)
| | - Katja Schenke-Layland
- Institute of Biomedical Engineering, Department for Medical Technologies and Regenerative Medicine (J. Marzi, K.S.-L.), Eberhard Karls University Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," (J. Marzi, A.P., K.S.-L.), University of Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen Reutlingen, Germany (J. Marzi, K.S.-L.)
| | - Peter Seizer
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
- Now with Aalen, Germany (P.S.)
| | - Julia Möllmann
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Germany (J. Möllmann, N.M.)
| | - Nikolaus Marx
- Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, Germany (J. Möllmann, N.M.)
| | - Robert Feil
- Interfaculty Institute of Biochemistry (IFIB) (T.K., R.F., S.F.), University of Tübingen, Germany
| | - Susanne Feil
- Interfaculty Institute of Biochemistry (IFIB) (T.K., R.F., S.F.), University of Tübingen, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Pharmacology, Toxicology and Clinical Pharmacy (M.C.S., R.L.), University of Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
- DFG Heisenberg Group Cardiovascular Thrombo-Inflammation and Translational Thrombocardiology (F.K., O.B.), University of Tübingen, Germany
| | - Tilman E Schäffer
- Institute for Applied Physics (K.K., V.G., T.E.S.), University of Tübingen, Germany
| | - Karin Anne Lydia Müller
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
| | - Meinrad P Gawaz
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
| | - David Heinzmann
- Department of Cardiology and Angiology (M.S., A.-K.R., F.K., S.U.-S., P.S., O.B., K.A.L.M., M.P.G., D.H.), Eberhard Karls University Tübingen, Germany
| |
Collapse
|
46
|
Zhao J, Huang H. Extracellular Vesicle-Derived Non-Coding RNAs: Key Mediators in Remodelling Heart Failure. Curr Issues Mol Biol 2024; 46:9430-9448. [PMID: 39329911 PMCID: PMC11430706 DOI: 10.3390/cimb46090559] [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: 07/27/2024] [Revised: 08/18/2024] [Accepted: 08/23/2024] [Indexed: 09/28/2024] Open
Abstract
Heart failure (HF), a syndrome of persistent development of cardiac insufficiency due to various heart diseases, is a serious and lethal disease for which specific curative therapies are lacking and poses a severe burden on all aspects of global public health. Extracellular vesicles (EVs) are essential mediators of intercellular and interorgan communication, and are enclosed nanoscale vesicles carrying biomolecules such as RNA, DNA, and proteins. Recent studies have showed, among other things, that non-coding RNAs (ncRNAs), especially microRNAs (miRNAs), long ncRNAs (lncRNA), and circular RNAs (circRNAs) can be selectively sorted into EVs and modulate the pathophysiological processes of HF in recipient cells, acting on both healthy and diseased hearts, which makes them promising targets for the diagnosis and therapy of HF. This review aims to explore the mechanism of action of EV-ncRNAs in heart failure, with emphasis on the potential use of differentially expressed miRNAs and circRNAs as biomarkers of cardiovascular disease, and recent research advances in the diagnosis and treatment of heart failure. Finally, we focus on summarising the latest advances and challenges in engineering EVs for HF, providing novel concepts for the diagnosis and treatment of heart failure.
Collapse
Affiliation(s)
- Jiayi Zhao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China;
- Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Huang Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China;
- Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
47
|
Yan N, Wang X, Xu Z, Zhong L, Yang J. Apigenin Attenuates Transverse Aortic Constriction-Induced Myocardial Hypertrophy: The Key Role of miR-185-5p/SREBP2-Mediated Autophagy. Drug Des Devel Ther 2024; 18:3841-3851. [PMID: 39219698 PMCID: PMC11365498 DOI: 10.2147/dddt.s464004] [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: 04/11/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Apigenin is a natural flavonoid compound with promising potential for the attenuation of myocardial hypertrophy (MH). The compound can also modulate the expression of miR-185-5p that both promote MH and suppress autophagy. The current attempts to explain the anti-MH effect of apigenin by focusing on changes in miR-185-5p-mediated autophagy. Methods Hypertrophic symptoms were induced in rats using transverse aortic constriction (TAC) method and in cardiomyocytes using Ang II and then handled with apigenin. Changes in myocardial function and structure and cell viability and surface area were measured. The role of miR-185-5p in the anti-MH function of apigenin was explored by detecting changes in autophagic processes and miR-185-5p/SREBP2 axis. Results TAC surgery induced weight increase, structure destruction, and collagen deposition in hearts of model rats. Ang II suppresses cardiomyocyte viability and increased cell surface area. All these impairments were attenuated by apigenin and were associated with the restored level of autophagy. At the molecular level, the expression of miR-185-5p was up-regulated by TAC, while the expression of SREBP2 was down-regulated, which was reserved by apigenin both in vivo and in vitro. The induction of miR-185-5p in cardiomyocytes could counteracted the protective effects of apigenin. Discussion Collectively, the findings outlined in the current study highlighted that apigenin showed anti-MH effects. The effects were related to the inhibition of miR-185-5p and activation of SREBP, which contributed to the increased autophagy.
Collapse
Affiliation(s)
- Na Yan
- Department of Vasculocardiology, Ganzhou People’s Hospital, Ganzhou, People’s Republic of China
| | - Xianggui Wang
- Department of Vasculocardiology, Ganzhou People’s Hospital, Ganzhou, People’s Republic of China
| | - Zufang Xu
- Department of Vasculocardiology, Ganzhou People’s Hospital, Ganzhou, People’s Republic of China
| | - Linling Zhong
- Department of Vasculocardiology, Ganzhou People’s Hospital, Ganzhou, People’s Republic of China
| | - Jiangyong Yang
- Department of Cardiology, Ganzhou Hospital of Guangdong Provincial People’s Hospital, Ganzhou Municipal Hospital, Ganzhou, People’s Republic of China
| |
Collapse
|
48
|
Golatkar V, Bhatt LK. Artesunate attenuates isoprenaline induced cardiac hypertrophy in rats via SIRT1 inhibiting NF-κB activation. Eur J Pharmacol 2024; 977:176709. [PMID: 38843948 DOI: 10.1016/j.ejphar.2024.176709] [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: 02/12/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Cardiac Hypertrophy is an adaptive response of the body to physiological and pathological stimuli, which increases cardiomyocyte size, thickening of cardiac muscles and progresses to heart failure. Downregulation of SIRT1 in cardiomyocytes has been linked with the pathogenesis of cardiac hypertrophy. The present study aimed to investigate the effect of Artesunate against isoprenaline induced cardiac hypertrophy in rats via SIRT1 inhibiting NF-κB activation. Experimental cardiac hypertrophy was induced in rats by subcutaneous administration of isoprenaline (5 mg/kg) for 14 days. Artesunate was administered simultaneously for 14 days at a dose of 25 mg/kg and 50 mg/kg. Artesunate administration showed significant dose dependent attenuation in mean arterial pressure, electrocardiogram, hypertrophy index and left ventricular wall thickness compared to the disease control group. It also alleviated cardiac injury biomarkers and oxidative stress. Histological observation showed amelioration of tissue injury in the artesunate treated groups compared to the disease control group. Further, artesunate treatment increased SIRT1 expression and decreased NF-kB expression in the heart. The results of the study show the cardioprotective effect of artesunate via SIRT1 inhibiting NF-κB activation in cardiomyocytes.
Collapse
Affiliation(s)
- Vaishnavi Golatkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
49
|
Cao H, Zhao L, Yuan Y, Liao C, Zeng W, Li A, Huang Q, Zhao Y, Fan Y, Jiang L, Song D, Li S, Zhang B. Lipoamide Attenuates Hypertensive Myocardial Hypertrophy Through PI3K/Akt-Mediated Nrf2 Signaling Pathway. J Cardiovasc Transl Res 2024; 17:910-922. [PMID: 38334841 PMCID: PMC11371882 DOI: 10.1007/s12265-024-10488-9] [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: 09/21/2023] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
The process of myocardial hypertrophy in hypertension can lead to excessive activation of oxidative stress. Lipoamide (ALM) has significant antioxidant and anti-inflammatory effects. This study aimed to investigate the effects of ALM on hypertension-induced cardiac hypertrophy, as well as explore its underlying mechanisms. We evaluated the effects of ALM on spontaneously hypertensive rats and rat cardiomyocytes treated with Ang II. We found that ALM was not effective in lowering blood pressure in SHR, but it attenuated hypertension-mediated cardiac fibrosis, oxidative stress, inflammation, and hypertrophy in rats. After that, in cultured H9C2 cells stimulated with Ang II, ALM increased the expression of antioxidant proteins that were decreased in the Ang II group. ALM also alleviated cell hypertrophy and the accumulation of ROS, while LY294002 partially abrogated these effects. Collectively, these results demonstrate that ALM could alleviate oxidative stress in cardiac hypertrophy, potentially through the activation of the PI3K/Akt-mediated Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Hongjuan Cao
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Lina Zhao
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yao Yuan
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Chunyan Liao
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Weidan Zeng
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Aiyue Li
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Quanfeng Huang
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yueyao Zhao
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yubing Fan
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Liu Jiang
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Dandan Song
- Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Sha Li
- Guizhou Medical University, Guiyang, Guizhou Province, China
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Bei Zhang
- Guizhou Medical University, Guiyang, Guizhou Province, China.
- Department of Ultrasound Center, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China.
| |
Collapse
|
50
|
Yu S, Sun Z, Ju T, Liu Y, Mei Z, Wang C, Qu Z, Li N, Wu F, Liu K, Lu M, Huang M, Pang X, Jia Y, Li Y, Zhang Y, Dou S, Jiang J, Dong X, Huang C, Li W, zhang Y, Yuan Y, Yang B, Du W. The m7G Methyltransferase Mettl1 Drives Cardiac Hypertrophy by Regulating SRSF9-Mediated Splicing of NFATc4. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308769. [PMID: 38810124 PMCID: PMC11304317 DOI: 10.1002/advs.202308769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/11/2024] [Indexed: 05/31/2024]
Abstract
Cardiac hypertrophy is a key factor driving heart failure (HF), yet its pathogenesis remains incompletely elucidated. Mettl1-catalyzed RNA N7-methylguanosine (m7G) modification has been implicated in ischemic cardiac injury and fibrosis. This study aims to elucidate the role of Mettl1 and the mechanism underlying non-ischemic cardiac hypertrophy and HF. It is found that Mettl1 is upregulated in human failing hearts and hypertrophic murine hearts following transverse aortic constriction (TAC) and Angiotensin II (Ang II) infusion. YY1 acts as a transcriptional factor for Mettl1 during cardiac hypertrophy. Mettl1 knockout alleviates cardiac hypertrophy and dysfunction upon pressure overload from TAC or Ang II stimulation. Conversely, cardiac-specific overexpression of Mettl1 results in cardiac remodeling. Mechanically, Mettl1 increases SRSF9 expression by inducing m7G modification of SRSF9 mRNA, facilitating alternative splicing and stabilization of NFATc4, thereby promoting cardiac hypertrophy. Moreover, the knockdown of SRSF9 protects against TAC- or Mettl1-induced cardiac hypertrophic phenotypes in vivo and in vitro. The study identifies Mettl1 as a crucial regulator of cardiac hypertrophy, providing a novel therapeutic target for HF.
Collapse
Affiliation(s)
- Shuting Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - ZhiYong Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Tiantian Ju
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Yingqi Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Zhongting Mei
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Changhao Wang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Zhezhe Qu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Na Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Fan Wu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - KuiWu Liu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Meixi Lu
- Traditional Chinese Medicine SchoolBeijing University of Chinese MedicineBeijing100013China
| | - Min Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Xiaochen Pang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Yingqiong Jia
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Ying Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Yaozhi Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Shunkang Dou
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Jianhao Jiang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Xianhui Dong
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Chuanhao Huang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Wanhong Li
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Yi zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
| | - Ye Yuan
- Department of Pharmacy (The University Key Laboratory of Drug ResearchHeilongjiang Province)The Second Affiliated Hospital of Harbin Medical UniversityHarbin150086China
| | - Baofeng Yang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
- Northern Translational Medicine Research and Cooperation CenterHeilongjiang Academy of Medical SciencesHarbin Medical UniversityHarbin150081China
- Research Unit of Noninfectious Chronic Diseases in Frigid ZoneChinese Academy of Medical Sciences2019RU070Harbin150081China
| | - Weijie Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD)Department of Pharmacology (The State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of ChinaKey Laboratory of Cardiovascular ResearchMinistry of Education)College of PharmacyHarbin Medical UniversityHarbin150081China
- Northern Translational Medicine Research and Cooperation CenterHeilongjiang Academy of Medical SciencesHarbin Medical UniversityHarbin150081China
- Research Unit of Noninfectious Chronic Diseases in Frigid ZoneChinese Academy of Medical Sciences2019RU070Harbin150081China
| |
Collapse
|