1
|
van Deel ED, Snelders M, van Vliet N, Te Riet L, van den Bosch TPP, Fiedler LR, van Spreeuwel ACC, Bax NAM, Boontje N, Halabi CM, Sasaki T, Reinhardt DP, van der Velden J, Bouten CVC, von der Thüsen JH, Danser AHJ, Duncker DJ, Schneider MD, van der Pluijm I, Essers J. Induction of cardiac fibulin-4 protects against pressure overload-induced cardiac hypertrophy and heart failure. Commun Biol 2025; 8:661. [PMID: 40274989 DOI: 10.1038/s42003-025-08087-8] [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: 05/13/2024] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
The prevailing view of fibulin-4 deficient mice is that the cardiac phenotype is the result of aortic and/or valvular disease. In the present study, we have tested whether the cardiac phenotype is, at least in part, the consequence of primary cardiac effects of fibulin-4. We have found fibulin-4 expression to be activated throughout the myocardium in wildtype (fibulin-4+/+) C57Bl/6J;129 Sv mice subjected to transverse aortic constriction (TAC). In contrast, haploinsufficient fibulin-4+/R mice exposed to severe TAC do not show this increase in myocardial fibulin-4 expression, but display altered physical properties of myocardial tissue. Moreover, TAC-induced cardiac fibrosis, pulmonary congestion, and mortality are aggravated in fibulin-4+/R mice. In vitro investigations of myocardial tissue show that fibulin-4 deficiency results in cardiomyocyte hypertrophy, and a decreased beating frequency and contractile force. In conclusion, we demonstrate functions for fibulin-4 in cardiac homeostasis and show that reduced fibulin-4 expression drives myocardial disease in response to cardiac pressure overload, independent of aortic valvular pathology.
Collapse
Affiliation(s)
- E D van Deel
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Snelders
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - N van Vliet
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - L Te Riet
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - T P P van den Bosch
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - L R Fiedler
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - A C C van Spreeuwel
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N A M Bax
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - N Boontje
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), Amsterdam, the Netherlands
| | - C M Halabi
- Division of Nephrology, Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - T Sasaki
- Department of Pharmacology, Faculty of Medicine, Oita University, Oita, Japan
| | - D P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada
- Faculty of Dentistry and Oral Health Sciences, McGill University, Montreal, QC, Canada
| | - J van der Velden
- Department of Physiology, VU University Medical Center, Institute for Cardiovascular Research (ICaR-VU), Amsterdam, the Netherlands
| | - C V C Bouten
- Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - J H von der Thüsen
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - A H J Danser
- Division of Pharmacology, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - D J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M D Schneider
- British Heart Foundation Centre of Research Excellence, National Heart and Lung Institute, Imperial College London, London, UK
| | - I van der Pluijm
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - J Essers
- Department of Molecular Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.
- Department of Vascular Surgery, Cardiovascular Institute, Erasmus University Medical Center, Rotterdam, the Netherlands.
- Department of Radiotherapy, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
2
|
Gupta S, Gupta AK, Mehan S, Khan Z, Gupta GD, Narula AS. Disruptions in cellular communication: Molecular interplay between glutamate/NMDA signalling and MAPK pathways in neurological disorders. Neuroscience 2025; 569:331-353. [PMID: 39809360 DOI: 10.1016/j.neuroscience.2025.01.021] [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: 11/11/2024] [Revised: 12/30/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Neurological disorders significantly impact the central nervous system, contributing to a growing public health crisis globally. The spectrum of these disorders includes neurodevelopmental and neurodegenerative diseases. This manuscript reviews the crucial roles of cellular signalling pathways in the pathophysiology of these conditions, focusing primarily on glutaminase/glutamate/NMDA receptor signalling, alongside the mitogen-activated protein kinase (MAPK) pathways-ERK1/2, C-JNK, and P38 MAPK. Activation of these pathways is often correlated with neuronal excitotoxicity, apoptosis, and inflammation, leading to many other pathological conditions such as traumatic brain injury, stroke, and brain tumor. The interplay between glutamate overstimulation and MAPK signalling exacerbates neurodegenerative processes, underscoring the complexity of cellular communication in maintaining neuronal health. Dysfunctional signalling alters synaptic plasticity and neuronal survival, contributing to cognitive impairments in various neurological diseases. The manuscript emphasizes the potential of targeting these signalling pathways for therapeutic interventions, promoting neuroprotection and reducing neuroinflammation. Incorporating insights from precision medicine and innovative drug delivery systems could enhance treatment efficacy. Overall, understanding the intricate mechanisms of these pathways is essential for developing effective strategies to mitigate the impact of neurological disorders and improve patient outcomes. This review highlights the necessity for further exploration into these signalling cascades to facilitate advancements in therapeutic approaches, ensuring better prognoses for individuals affected by neurological conditions.
Collapse
Affiliation(s)
- Sumedha Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Abhishek Kumar Gupta
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India. https://mehanneuroscience.org
| | - Zuber Khan
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India Affiliated to IK Gujral Punjab Technical University, Jalandhar, Punjab 144603, India
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
3
|
Wang X, Liu R, Liu D. The Role of the MAPK Signaling Pathway in Cardiovascular Disease: Pathophysiological Mechanisms and Clinical Therapy. Int J Mol Sci 2025; 26:2667. [PMID: 40141309 PMCID: PMC11942496 DOI: 10.3390/ijms26062667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Cardiovascular disease (CVD) is a serious global health issue with high mortality rates worldwide. Despite the numerous advancements in the study of CVD pathogenesis in recent years, further summarization and elaboration of specific molecular pathways are required. An extensive body of research has been conducted to elucidate the association between the MAPK signaling pathway, which is present in all eukaryotic organisms, and the pathogenesis of cardiovascular disease. This review aims to provide a comprehensive summary of the research conducted on MAPK and CVD over the past five years. The primary focus is on four specific diseases: heart failure, atherosclerosis, myocardial ischemia-reperfusion injury, and cardiac hypertrophy. The review will also address the pathophysiological mechanisms of MAPK in cardiovascular diseases, with the objective of proposing novel clinical treatment strategies for CVD.
Collapse
Affiliation(s)
- Xueyang Wang
- Queen Mary College, Nanchang University, Nanchang 330001, China; (X.W.); (R.L.)
| | - Ruiqi Liu
- Queen Mary College, Nanchang University, Nanchang 330001, China; (X.W.); (R.L.)
| | - Dan Liu
- Queen Mary College, Nanchang University, Nanchang 330001, China; (X.W.); (R.L.)
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| |
Collapse
|
4
|
Avolio E, Bassani B, Campanile M, Mohammed KA, Muti P, Bruno A, Spinetti G, Madeddu P. Shared molecular, cellular, and environmental hallmarks in cardiovascular disease and cancer: Any place for drug repurposing? Pharmacol Rev 2025; 77:100033. [PMID: 40148035 DOI: 10.1016/j.pharmr.2024.100033] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 12/17/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer and cardiovascular disease (CVD) are the 2 biggest killers worldwide. Specific treatments have been developed for the 2 diseases. However, mutual therapeutic targets should be considered because of the overlap of cellular and molecular mechanisms. Cancer research has grown at a fast pace, leading to an increasing number of new mechanistic treatments. Some of these drugs could prove useful for treating CVD, which realizes the concept of cancer drug repurposing. This review provides a comprehensive outline of the shared hallmarks of cancer and CVD, primarily ischemic heart disease and heart failure. We focus on chronic inflammation, altered immune response, stromal and vascular cell activation, and underlying signaling pathways causing pathological tissue remodeling. There is an obvious scope for targeting those shared mechanisms, thereby achieving reciprocal preventive and therapeutic benefits. Major attention is devoted to illustrating the logic, advantages, challenges, and viable examples of drug repurposing and discussing the potential influence of sex, gender, age, and ethnicity in realizing this approach. Artificial intelligence will help to refine the personalized application of drug repurposing for patients with CVD. SIGNIFICANCE STATEMENT: Cancer and cardiovascular disease (CVD), the 2 biggest killers worldwide, share several underlying cellular and molecular mechanisms. So far, specific therapies have been developed to tackle the 2 diseases. However, the development of new cardiovascular drugs has been slow compared with cancer drugs. Understanding the intersection between pathological mechanisms of the 2 diseases provides the basis for repurposing cancer therapeutics for CVD treatment. This approach could allow the rapid development of new drugs for patients with CVDs.
Collapse
Affiliation(s)
- Elisa Avolio
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy
| | - Marzia Campanile
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy; Department of Biosciences, University of Milan, Milan, Italy
| | - Khaled Ak Mohammed
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom; Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paola Muti
- IRCCS MultiMedica, Milan, Italy; Department of Biomedical, Surgical and Dental Health Sciences, University of Milan, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry, and Immunology, IRCCS MultiMedica, Milan, Italy; Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy.
| | - Gaia Spinetti
- Laboratory of Cardiovascular Pathophysiology - Regenerative Medicine, IRCCS MultiMedica, Milan, Italy.
| | - Paolo Madeddu
- Bristol Heart Institute, Laboratory of Experimental Cardiovascular Medicine, Translational Health Sciences, Bristol Medical School, University of Bristol, United Kingdom.
| |
Collapse
|
5
|
Zhao Y, Sakurai T, Kamiyoshi A, Tanaka M, Ichikawa-Shindo Y, Kawate H, Matsuda Y, Zhang Y, Guo Q, Li P, Hoshiyama K, Hayashi M, Li J, Shindo T. Adrenomedullin 2/Intermedin Exerts Cardioprotective Effects by Regulating Cardiomyocyte Mitochondrial Function. Hypertension 2025; 82:e6-e21. [PMID: 39817339 DOI: 10.1161/hypertensionaha.124.23666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/03/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Adrenomedullin 2 (AM2) plays critical roles in regulating blood pressure and fluid balance. However, the specific involvement of AM2 in cardiac hypertrophy has not been comprehensively elucidated, warranting further investigation into its molecular mechanisms and therapeutic implications. METHODS Cardiac hypertrophy was induced in adult mice lacking AM2 (AM2-/-) using transverse aortic constriction surgery. Comprehensive cardiac morphology, function, histology, and transcriptome/metabolome analyses were conducted. Signal transduction underlying AM2 stimulation in the cardiomyocytes was explored. RESULTS The absence of endogenous AM2 led to the development of severe heart failure after transverse aortic constriction surgery, which was characterized by alterations in the mitochondrial morphology and function associated with glycolysis and the tricarboxylic acid cycle in the heart and cardiomyocytes of transverse aortic constriction-operated AM2-/- mice. AM2 stimulation was associated with the receptor-modifying factor RAMP2 (receptor activity-modifying protein 2), which primarily transduces signals through the MAPK (mitogen-activated protein kinase) pathway and affects the expression of genes involved in glycolysis, β-oxidation, and oxidative phosphorylation. The administration of exogenous AM2 alleviated heart failure following transverse aortic constriction. CONCLUSIONS AM2 crucially regulates mitochondrial functions associated with the glycolysis and tricarboxylic acid cycles in the cardiomyocytes, thereby exerting a protective effect on the heart under pressure overload conditions.
Collapse
Affiliation(s)
- Yunlu Zhao
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takayuki Sakurai
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| | - Akiko Kamiyoshi
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| | - Megumu Tanaka
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yuka Ichikawa-Shindo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hisaka Kawate
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yorishige Matsuda
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ophthalmology (Y.M., K.H.), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yan Zhang
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Qianqian Guo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Peixuan Li
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Ken Hoshiyama
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Ophthalmology (Y.M., K.H.), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Marina Hayashi
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Jiake Li
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takayuki Shindo
- Departments of Cardiovascular Research (Y. Zhao, T. Sakurai, A.K., M.T., Y.I.-S., H.K., Y.M., Y. Zhang, Q.G., P.L., K.H., M.H., J.L., T. Shindo), Shinshu University School of Medicine, Matsumoto, Nagano, Japan
- Department of Life Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Nagano, Japan (T. Sakurai, A.K., M.T., T. Shindo)
| |
Collapse
|
6
|
Han X, Zhu QQ, Li Z, He JK, Sun Y, Zhong QH, Tang SX, Zhang YL. 4-Hydroxychalcone attenuates AngII-induced cardiac remodeling and dysfunction via regulating PI3K/AKT pathway. Hypertens Res 2025; 48:1054-1067. [PMID: 39715792 PMCID: PMC11879844 DOI: 10.1038/s41440-024-02068-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: 09/22/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Cardiac remodeling encompasses structural alterations such as hypertrophy, fibrosis, and dilatation, alongside numerous cellular and molecular functional aberrations, constituting a pivotal process in the advancement of heart failure (HF). 4-Hydroxychalcone (4-HCH) is a class of naturally occurring compounds with variable phenolic structures, and has demonstrated the preventive efficacy in hyperaldosteronism, inflammation and renal injury. However, the role of 4-HCH in the regulation of cardiac remodeling remains uncertain. A cardiac remodeling model was established in male C57BL/6 J mice via subcutaneous Ang II (1000 or 300 ng/kg/min) for 2 weeks. Mice were treated with 4-HCH (20 or 40 mg/kg/day) or vehicle control. Systolic blood pressure (SBP) was measured using a tail-cuff method, and echocardiography assessed cardiac function. Histopathological staining evaluated cardiomyocyte hypertrophy, fibrosis, inflammation, and superoxide production. Network pharmacology analysis identified potential core targets and pathways mediating the effects of 4-HCH. Expression of inflammatory cytokines and proteins related to hypertrophy, fibrosis, inflammation, and oxidative stress was assessed by quantitative real-time PCR (qPCR) and Western blotting. Our results indicated that 4-HCH significantly ameliorated Ang II-induced hypertension, cardiomyocyte hypertrophy, fibroblast activation, fibrosis, inflammation, superoxide production, and cardiac function. Network pharmacology analysis identified the PI3K-AKT pathway as a crucial mechanism underlying the effects of 4-HCH, with experimental verification demonstrating that it inhibits cardiac remodeling by downregulating this pathway and its downstream effectors, including mTOR/ERK, TGF-β/Smad2/3, NF-κB, and NOX1 independent of its blood pressure-lowering effects. These results reveal for the first time that 4-HCH alleviates cardiac remodeling, emphasizing its potential as a therapeutic agent for HF.
Collapse
Affiliation(s)
- Xiao Han
- Institute of Cardio-Cerebrovascular Medicine, Central Hospital of Dalian University of Technology, No.826, South West Road, Shahekou District, Dalian, 116089, China
| | - Qian-Qiu Zhu
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, No. 2, Zheshan West Road, Wuhu, 241000, China
| | - Zhi Li
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, Dalian, 116011, China
| | - Jia-Kang He
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, No.193, Lianhe Road, Xigang District, Dalian, 116011, China
| | - Yan Sun
- Department of Cardiology, Longgang Central Hospital of Shenzhen, No.6802, Longgang Road, Longgang District, Shenzhen, 518000, China
| | - Qing-Hua Zhong
- Department of Cardiology, Longgang Central Hospital of Shenzhen, No.6802, Longgang Road, Longgang District, Shenzhen, 518000, China.
| | - Sheng-Xing Tang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, No. 2, Zheshan West Road, Wuhu, 241000, China.
| | - Yun-Long Zhang
- Department of Cardiology, Yijishan Hospital of Wannan Medical College, No. 2, Zheshan West Road, Wuhu, 241000, China.
| |
Collapse
|
7
|
Zhao S, Zhang Y, Zhao Y, Lu X. Cellular senescence as a key player in chronic heart failure pathogenesis: Unraveling mechanisms and therapeutic opportunities. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:8-18. [PMID: 39961550 DOI: 10.1016/j.pbiomolbio.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 02/09/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025]
Abstract
Chronic heart failure (CHF) is the final stage of heart disease and is caused by various factors. Unfortunately, CHF has a poor prognosis and a high mortality rate. Recent studies have found that aging is a significant risk factor for the development of CHF and that cellular senescence plays a vital role in its development. This article reviews different types of cellular senescence, mitochondrial dysfunction in senescent cells, autophagy in senescent cells, and senescence-associated secretory phenotype (SASP), and epigenetic regulation, to provide new perspectives on the research and treatment of CHF.
Collapse
Affiliation(s)
- Shuqing Zhao
- The First Clinical College of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Zhang
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying Zhao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Xiaohui Lu
- The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
8
|
Cairns M, Marais E, Joseph D, Essop MF. The Role of Chronic Stress in the Pathogenesis of Ischemic Heart Disease in Women. Compr Physiol 2025; 15:e70000. [PMID: 39903543 PMCID: PMC11793136 DOI: 10.1002/cph4.70000] [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: 10/16/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Psychological stress has emerged as a critical risk factor for cardiovascular disease, especially in women. While female participation in clinical research has improved, sex-specific data analysis and reporting often remain inadequate, limiting our ability to draw definitive conclusions for women. Conversely, preclinical studies consistently demonstrate adverse effects of stress on female health, yet the molecular mechanisms underlying this association remain elusive. Evidence suggests that female IHD pathogenesis is more complex than in males, involving multiple factors, including inflammation, contractile dysfunction, bioenergetic impairment, and remodeling. However, many of these mechanisms are primarily derived from male studies, and molecular investigations in female models are limited, hindering our understanding of the underlying biological pathways. This is particularly concerning given the increasing prevalence of ischemic heart disease in postmenopausal women. In order to fully elucidate the impact of stress on female cardiac health and develop targeted interventions, further preclinical research on female models is essential.
Collapse
Affiliation(s)
- Megan Cairns
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Erna Marais
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| | - Danzil Joseph
- Department of Physiological Sciences, Center for Cardio‐Metabolic Research in Africa (CARMA)Stellenbosch UniversityStellenboschSouth Africa
| | - M. Faadiel Essop
- Division of Medical PhysiologyCentre for Cardio‐Metabolic Research in Africa (CARMA)Faculty of Medicine and Health SciencesStellenbosch UniversityCape TownSouth Africa
| |
Collapse
|
9
|
Imran M, Altamimi ASA, Afzal M, Babu MA, Goyal K, Ballal S, Sharma P, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H. Targeting senescence and GATA4 in age-related cardiovascular disease: a comprehensive approach. Biogerontology 2025; 26:45. [PMID: 39831933 DOI: 10.1007/s10522-025-10189-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: 10/31/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
The growing prevalence of age-related cardiovascular diseases (CVDs) poses significant health challenges, necessitating the formulation of novel treatment approaches. GATA4, a vital transcription factor identified for modulating cardiovascular biology and cellular senescence, is recognized for its critical involvement in CVD pathogenesis. This review collected relevant studies from PubMed, Google Scholar, and Science Direct using search terms like 'GATA4,' 'cellular senescence,' 'coronary artery diseases,' 'hypertension,' 'heart failure,' 'arrhythmias,' 'congenital heart diseases,' 'cardiomyopathy,' and 'cardiovascular disease.' Additionally, studies investigating the molecular mechanisms underlying GATA4-mediated regulation of GATA4 and senescence in CVDs were analyzed to provide comprehensive insights into this critical aspect of potential treatment targeting. Dysregulation of GATA4 is involved in a variety of CVDs, as demonstrated by both experimental and clinical research, comprising CAD, hypertension, congenital heart diseases, cardiomyopathy, arrhythmias, and cardiac insufficiency. Furthermore, cellular senescence enhances the advancement of age-related CVDs. These observations suggested that therapies targeting GATA4, senescence pathways, or both as necessary may be an effective intervention in CVD progression and prognosis. Addressing age-related CVDs by targeting GATA4 and senescence is a broad mechanism approach. It implies further investigation of the molecular nature of these processes and elaboration of an effective therapeutic strategy. This review highlights the importance of GATA4 and senescence in CVD pathogenesis, emphasizing their potential as therapeutic targets for age-related CVDs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, 91911, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, UP, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Fadiyah Jadid Alanazi
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
10
|
Zhang L, Ding F, Ren Z, Cheng W, Dai H, Liang Q, Kong F, Xu W, Wang M, Zhang Y, Tao Q. Mechanisms of pathogenicity in the hypertrophic cardiomyopathy-associated TNNI3 c.235C > T variant. Int J Cardiol 2025; 419:132627. [PMID: 39426416 DOI: 10.1016/j.ijcard.2024.132627] [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: 09/16/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is typically manifested as a hereditary disorder, with 30 %-60 % of cases linked to cardiac sarcomere gene mutations. Despite numerous identified TNNI3 mutations associated with HCM, their severity, prevalence, and disease progression vary. The link between TNNI3 variants and phenotypes remains largely unexplored. This study aims to elucidate the impact of the TNNI3 c.235C > T mutation on HCM through clinical research and cell experiments and to explore its mechanism in HCM development. METHODS We screened an HCM family for pathogenic gene mutations using gene sequencing. The proband and family members were assessed through electrocardiography, echocardiography, and cardiac MRI, and a pedigree map was created for disease prediction analysis. Mutant plasmids were constructed with the TNNI3 c.235C > T mutation and transfected into the AC16 human cardiomyocyte cell line to investigate the mutation's effects. RESULTS The TNNI3 c.235C > T mutation was identified as the disease-causing variant in the family. This mutation led to the upregulation of hypertrophy-associated genes ANP, BNP, and MYH7, increased cardiomyocyte size, and activation of the ERK signaling pathway. Further investigations revealed that the TNNI3 c.235C > T mutation impaired mitochondrial function, disrupted cardiomyocyte metabolism, and increased cellular autophagy and apoptosis. CONCLUSIONS The TNNI3 c.235C > T gene mutation may be a pathogenic factor for HCM, showing heterogeneous features and clinical phenotypes. This mutation induces myocardial hypertrophy, activates the ERK signaling pathway, and exacerbates mitochondrial dysfunction, apoptosis, and autophagy in cardiomyocytes. These findings provide insights into the mechanism of HCM caused by gene mutations and may inform HCM treatment strategies.
Collapse
Affiliation(s)
- Lai Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fengzhi Ding
- Department of Physiology, Wannan Medical College, Wuhu, Anhui, 241000, China
| | - Zhongyuan Ren
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Weili Cheng
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - He Dai
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Qing Liang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Fanling Kong
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Wenjing Xu
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Minghui Wang
- College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yuqing Zhang
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
| | - Qin Tao
- Department of Cardiology, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, Jiangsu, 211100, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
11
|
Cools JMT, Goovaerts BK, Feyen E, Van den Bogaert S, Fu Y, Civati C, Van Fraeyenhove J, Tubeeckx MRL, Ott J, Nguyen L, Wülfers EM, Van Berlo B, De Vries AAF, Vandersickel N, Pijnappels DA, Audenaert D, Roderick HL, De Winter H, De Keulenaer GW, Segers VFM. Small-molecule-induced ERBB4 activation to treat heart failure. Nat Commun 2025; 16:576. [PMID: 39794341 PMCID: PMC11724075 DOI: 10.1038/s41467-024-54908-5] [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: 03/29/2024] [Accepted: 11/25/2024] [Indexed: 01/13/2025] Open
Abstract
Heart failure is a common and deadly disease requiring new treatments. The neuregulin-1/ERBB4 pathway offers cardioprotective benefits, but using recombinant neuregulin-1 as therapy has limitations due to the need for intravenous delivery and lack of receptor specificity. We hypothesize that small-molecule activation of ERBB4 could protect against heart damage and fibrosis. To test this, we conduct a screening of 10,240 compounds and identify eight structurally similar ones (EF-1 to EF-8) that induce ERBB4 dimerization, with EF-1 being the most effective. EF-1 reduces cell death and hypertrophy in cardiomyocytes and decreases collagen production in cardiac fibroblasts in an ERBB4-dependent manner. In wild-type mice, EF-1 inhibits angiotensin-II-induced fibrosis in males and females and reduces heart damage caused by doxorubicin and myocardial infarction in females, but not in Erbb4-null mice. This study shows that small-molecule ERBB4 activation is feasible and may lead to a novel class of drugs for treating heart failure.
Collapse
Grants
- This work was supported by a Geconcerteerde onderzoeksactie grant (GOA, PID36444) of the University of Antwerp; by a Senior Clinical Investigator fellowship (to V.F.S.), a PhD fellowship (to J.MT.C. and C.C.), and research grants of the Fund for Scientific Research Flanders (Application numbers 1842219N, G021019N, G021420N, 1S49323N, and 11PBU24N); VLIR/iBOF Grant 20-VLIR-iBOF-027 (to N.V., V.F.S., H.L.R., and G.W.D.K.).
Collapse
Affiliation(s)
- Julie M T Cools
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Bo K Goovaerts
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Eline Feyen
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | - Yile Fu
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Céline Civati
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | | | | | - Jasper Ott
- Laboratory of Cell Biology and Histology, University of Antwerp, Antwerp, Belgium
| | - Long Nguyen
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - Eike M Wülfers
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
- Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg - Bad Krozingen, Freiburg im Breisbau, Germany and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Benji Van Berlo
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
| | - Antoine A F De Vries
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Nele Vandersickel
- Department of Physics and Astronomy, Ghent University, Ghent, Belgium
| | - Daniël A Pijnappels
- Laboratory of Experimental Cardiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dominique Audenaert
- Screening Core, VIB, Ghent, Belgium
- Centre for Bioassay Development and Screening (C-BIOS), Ghent University, Ghent, Belgium
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Hans De Winter
- Laboratory of Medicinal Chemistry, University of Antwerp, Antwerp, Belgium
| | - Gilles W De Keulenaer
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, ZNA Hospital, Antwerp, Belgium
| | - Vincent F M Segers
- Laboratory of PhysioPharmacology, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, University Hospital Antwerp, Antwerp, Belgium.
| |
Collapse
|
12
|
Liu C, Zhang Y, Zhao J, Zhang J, Meng Z, Yang Y, Xie Y, Jiao X, Liang B, Cao J, Wang Y. Vaping/e-cigarette-induced pulmonary extracellular vesicles contribute to exacerbated cardiomyocyte impairment through the translocation of ERK5. Life Sci 2024; 358:123195. [PMID: 39481834 DOI: 10.1016/j.lfs.2024.123195] [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: 05/31/2024] [Revised: 07/30/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
AIMS The impact of e-cigarettes/vaping on cardiac function remains contradictory owing to insufficient direct evidence of interorgan communication. Extracellular vesicles (EVs) have protective or detrimental effects depending on pathological conditions, making it crucial to understand their role in lung-cardiac cell interactions mediated by vaping inhalation. METHODS AND KEY FINDINGS Pulmonary EVs were characterized from animals that underwent 12 weeks of nicotine inhalation (vaping component) (EVsNicotine) or vehicle control (EVsVehicle). EVsNicotine significantly increased in size and abundance compared with EVsVehicle. The direct effect of EVs Nicotine and EVs Vehicle on cardiomyocytes was then assessed in vitro and in vivo. EVs Nicotine led to a decrease in cardiac function as manifested by reduced cardiac contractility and impaired relaxation. EVs Nicotine induced increased levels of cleaved caspase-1 and cleaved caspase-11 in cardiomyocytes, indicating the promotion of pyroptosis. Meanwhile, EVsNicotine stimulated the secretion of fibrotic factors. Further analysis revealed that nicotine inhalation stimulated EVs Nicotine enriched with high levels of ERK5 (EVs Nicotine-ERK5). It was discovered that these EVs derived from pulmonary epithelial cells. Furthermore, inhibiting cardiac ERK5 blunted the EVs Nicotine-induced pyroptosis and fibrotic factor secretion. We further identified GATA4, a pro-pyroptosis transcription factor, as being activated through ERK5-dependent phosphorylation. SIGNIFICANCE Our research demonstrates that nicotine inhalation exacerbates cardiac injury through the activation of EVs derived from the lungs during e-cigarettes/vaping. Specifically, the EVs containing ERK5 play a crucial role in mediating the detrimental effects on cardiac function. This research provides new insights into the cardiac toxicity of vaping and highlights the role of EVs Nicotine-ERK5 in this process.
Collapse
Affiliation(s)
- Caihong Liu
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yanwei Zhang
- Department of Cardiology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jianli Zhao
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John Zhang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Zhijun Meng
- Clinical Laboratory, Shanxi Provincial People's Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yuhui Yang
- Department of Anesthesiology, Guangdong Medical University, Guangzhou 510182, Guangdong, China
| | - Yaoli Xie
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiangying Jiao
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Bin Liang
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, China; Department of Cardiology, The Second Affiliated Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jimin Cao
- Department of Physiology, Key Laboratory of Cellular Physiology, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| | - Yajing Wang
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
13
|
Zhang L, Tian L, Liang B, Wang L, Huang S, Zhou Y, Ni M, Zhang L, Li Y, Chen J, Li X. Construction of an adverse outcome pathway for the cardiac toxicity of bisphenol a by using bioinformatics analysis. Toxicology 2024; 509:153955. [PMID: 39303899 DOI: 10.1016/j.tox.2024.153955] [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: 08/01/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Bisphenol A (BPA), a common endocrine disruptor, has shown cardiovascular toxicity in several epidemiological studies, as well as in vivo and in vitro experimental studies. However, the related adverse outcome pathway (AOP) of BPA toxicity remains unraveled. This study aimed to develop an AOP for the cardiac toxicity of BPA through bioinformatics analysis. The interactions among BPA, genes, phenotypes, and cardiac toxicity were retrieved from several databases, including the Comparative Toxicogenomics Database, Computational Toxicology, DisGeNet, and MalaCards. The target genes and part of target phenotypes were obtained by Venn analysis and literature screening. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed for target genes by using the DAVID online analysis tool to obtain other target phenotypes. AOP hypotheses from BPA exposure to heart disease were established and evaluated comprehensively by a quantitative weight of evidence (QWOE) method. The target genes included ESR2, MAPK1, TGFB1, and ESR1, and the target phenotypes included heart contraction, cardiac muscle contraction, cellular Ca2+ homeostasis, cellular metabolic process, heart development, etc. Overall, the AOP of BPA cardiac toxicity was deduced to be as follows. Initially, BPA bound with ERα/β and then activated the MAPK, AKT, and IL-17 signaling pathways, leading to Ca2+ homeostasis disorder and increased inflammatory response. Subsequently, cardiac function was impaired, causing coronary heart disease, arrhythmia, cardiac dysplasia, and other heart diseases. According to the Bradford-Hill causal considerations, the score of AOP by QWOE was 69, demonstrating a moderate confidence and providing clues on cardiotoxicity-assessment procedure and further studies on BPA.
Collapse
Affiliation(s)
- Leyan Zhang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Lin Tian
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Baofang Liang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Liang Wang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Shuzhen Huang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Yongru Zhou
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Mengmei Ni
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Lishi Zhang
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Yun Li
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China
| | - Jinyao Chen
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China.
| | - Xiaomeng Li
- Department of Nutrition and Food Safety, West China School of Public Health/West China Fourth Hospital, Sichuan University, Chengdu, China; Food Safety Monitoring and Risk Assessment Key Laboratory of Sichuan Province, Chengdu, China.
| |
Collapse
|
14
|
Chang J, Zhu Y, Yang Z, Wang Z, Wang M, Chen L. Airborne polystyrene nanoplastics exposure leads to heart failure via ECM-receptor interaction and PI3K/AKT/BCL-2 pathways. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176469. [PMID: 39317253 DOI: 10.1016/j.scitotenv.2024.176469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/30/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Environmental contamination has been recognized as a significant threat to human well-being, and recent findings of microplastic presence in human cardiac tissues have raised concerns. However, research on the effects of airborne nanoplastics (NPs) on cardiac physiology remains limited. We utilized a comprehensive body exposure apparatus to simulate the impact of airborne polystyrene NPs pollution, focusing on understanding how airborne NPs affect cardiac morphology and function. Following two weeks of NPs exposure, mice exhibited a 23.89 ± 8.30 % reduction in heart mass, a 20.05 ± 2.97 % decrease in heart rate as detected, and a myocardial electrical conduction block. Echocardiography showed significant changes in cardiac contractility, with increases in cardiac ejection fraction and stroke volume of 13.00 ± 3.00 % and 43.00 ± 17.00 %, respectively. In addition, histologic assessments revealed signs of ventricular hypertrophy, ventricular myocardial hypertrophy, and myocardial necrotic fibrosis. Of particular interest, our mechanistic investigations highlighted the harmful effects of NPs on cardiac structure and function, mediated through extracellular matrix (ECM) receptor interactions and the PI3K/AKT/BCL-2 signaling pathway. The insights gained provide a foundation for understanding the risks posed by airborne NPs to human cardiac health, emphasizing the need for increased vigilance and implementation of mitigation strategies in environmental management.
Collapse
Affiliation(s)
- Jinghao Chang
- Medical School, Tianjin University, Tianjin 300072, China
| | - Yuchen Zhu
- Medical School, Tianjin University, Tianjin 300072, China
| | - Ziye Yang
- Medical School, Tianjin University, Tianjin 300072, China; School of Environmental Science and Engineering, Tianjin University, Tianjin 300072, China.
| | - Ziqi Wang
- Medical School, Tianjin University, Tianjin 300072, China
| | - Meixue Wang
- Medical School, Tianjin University, Tianjin 300072, China
| | - Liqun Chen
- Medical School, Tianjin University, Tianjin 300072, China.
| |
Collapse
|
15
|
Tian L, Huang Y, Liu Y, Liu J, Liu Y. Parecoxib inhibits tumorigenesis and angiogenesis in hepatocellular carcinoma through ERK-VEGF/MMPs signaling pathway. IUBMB Life 2024; 76:972-986. [PMID: 38873890 DOI: 10.1002/iub.2861] [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: 12/27/2023] [Accepted: 05/05/2024] [Indexed: 06/15/2024]
Abstract
Parecoxib, a well-recognized nonsteroidal anti-inflammatory drug, has been reported to possess anticancer properties in various tumor types. In this work, we aimed to investigate the potential anticancer effects of parecoxib on hepatocellular carcinoma (HCC) cells. To assess the impact of parecoxib on HCC cell proliferation, we employed Cell Counting Kit-8, colony formation, and 5-ethynyl-2'-deoxyuridine assays. Hoechst/propidium iodide (PI) double staining and flow cytometry were performed to evaluate apoptosis and cell cycle analysis. Wound healing and transwell assays were utilized to assess cell migration and invasion. Tube formation assay was employed to analyze angiogenesis. Protein levels were determined using western blotting, and mRNA expression levels were assessed using quantitative real-time polymerase chain reaction (PCR). A xenograft mouse model was used to confirm the antitumor effects of parecoxib on HCC tumors in vivo. Our data demonstrated that parecoxib effectively inhibited the proliferation of HCC cells in a dose- and time-dependent manner. In addition, parecoxib induced cell cycle arrest in the G2 phase and promoted apoptosis. Moreover, parecoxib hindered tumor migration and invasion by impeding the epithelial-mesenchymal transition process. Further investigation showed that parecoxib could significantly suppress angiogenesis through the inhibition of extracellular signal-regulated kinase (ERK)-vascular endothelial growth factor (VEGF) axis. Notably, treatment with the ERK activator phorbol myristate acetate upregulated the expression of matrix metalloproteinase (MMP)-2, MMP-9, and VEGF and reversed the function of parecoxib in HCC cells. Besides, parecoxib displayed its antitumor efficacy in vivo. Collectively, our results suggest that parecoxib ameliorates HCC progression by regulating proliferation, cell cycle, apoptosis, migration, invasion, and angiogenesis through the ERK-VEGF/MMPs signaling pathway.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/metabolism
- Liver Neoplasms/genetics
- Animals
- Isoxazoles/pharmacology
- Mice
- Cell Proliferation/drug effects
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/metabolism
- Vascular Endothelial Growth Factor A/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Apoptosis/drug effects
- Cell Movement/drug effects
- Xenograft Model Antitumor Assays
- Mice, Nude
- Signal Transduction/drug effects
- Mice, Inbred BALB C
- Gene Expression Regulation, Neoplastic/drug effects
- Carcinogenesis/drug effects
- MAP Kinase Signaling System/drug effects
- Matrix Metalloproteinase 9/metabolism
- Matrix Metalloproteinase 9/genetics
- Male
- Cell Line, Tumor
- Angiogenesis
Collapse
Affiliation(s)
- Li Tian
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - YuQi Huang
- Department of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, Hangzhou, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - JiangWei Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yan Liu
- Department of Hepatobiliary Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
16
|
Wang X, Nie X, Wang H, Ren Z. Roles of small GTPases in cardiac hypertrophy (Review). Mol Med Rep 2024; 30:208. [PMID: 39301654 PMCID: PMC11425065 DOI: 10.3892/mmr.2024.13332] [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: 04/10/2024] [Accepted: 08/20/2024] [Indexed: 09/22/2024] Open
Abstract
Cardiac hypertrophy results from the heart reacting and adapting to various pathological stimuli and its persistent development is a major contributing factor to heart failure. However, the molecular mechanisms of cardiac hypertrophy remain unclear. Small GTPases in the Ras, Rho, Rab, Arf and Ran subfamilies exhibit GTPase activity and play crucial roles in regulating various cellular responses. Previous studies have shown that Ras, Rho and Rab are closely linked to cardiac hypertrophy and that their overexpression can induce cardiac hypertrophy. Here, we review the functions of small GTPases in cardiac hypertrophy and provide additional insights and references for the prevention and treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xin Wang
- School of Mathematics and Statistics, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Xinwen Nie
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Hao Wang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Zhanhong Ren
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
17
|
Garmany R, Dasari S, Bos JM, Kim ET, Gluscevic M, Martinez KA, Tester DJ, Dos Remedios C, Maleszewski JJ, Dearani JA, Ommen SR, Geske JB, Giudicessi JR, Ackerman MJ. A multi-omics atlas of sex-specific differences in obstructive hypertrophic cardiomyopathy. J Mol Cell Cardiol 2024; 196:26-34. [PMID: 39255898 DOI: 10.1016/j.yjmcc.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is a common genetic heart disease. Women with HCM tend to have a later onset but more severe disease course. However, the underlying pathobiological mechanisms for these differences remain unknown. METHODS Myectomy samples from 97 patients (53 males/44 females) with symptomatic obstructive HCM and 23 control cardiac tissues were included in this study. RNA-sequencing was performed on all samples. Mass spectrometry-based proteomics and phosphoproteomics was performed on a representative subset of samples. RESULTS The transcriptome, proteome, and phosphoproteome was similar between sexes and did not separate on PCA plotting. Overall, there were 482 differentially expressed genes (DEGs) between control females and control males while there were only 53 DEGs between HCM females and HCM males. There were 1983 DEGs between HCM females and control females compared to 1064 DEGs between HCM males and control males. Additionally, there was increased transcriptional downregulation of hypertrophy pathways in HCM females and in HCM males. HCM females had 119 differentially expressed proteins compared to control females while HCM males only had 27 compared to control males. Finally, the phosphoproteome showed females had 341 differentially phosphorylated proteins (DPPs) compared to controls while males only had 184. Interestingly, there was hypophosphorylation and inactivation of hypertrophy pathways in females but hyperphosphorylation and activation in males. CONCLUSION There are subtle, but biologically relevant differences in the multi-omics profile of HCM. This study provides the most comprehensive atlas of sex-specific differences in the transcriptome, proteome, and phosphoproteome present at the time of surgical myectomy for obstructive HCM.
Collapse
Affiliation(s)
- Ramin Garmany
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine, and the Mayo Clinic Medical Scientist Training Program, Rochester, MN, USA; Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Department of Quantitative Health Sciences/Division of Computational Biology, Mayo Clinic, Rochester, MN, USA
| | - J Martijn Bos
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA; Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA
| | - Evelyn T Kim
- Mayo Clinic Mentorship Program, Rochester, MN, USA
| | - Martina Gluscevic
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Katherine A Martinez
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - David J Tester
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Cristobal Dos Remedios
- Mechanobiology Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, Australia
| | - Joseph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Joseph A Dearani
- Department of Cardiovascular Surgery, Mayo Clinic, Rochester, MN, USA
| | - Steve R Ommen
- Department of Cardiovascular Medicine; Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey B Geske
- Department of Cardiovascular Medicine; Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - John R Giudicessi
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA; Department of Cardiovascular Medicine; Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - Michael J Ackerman
- Department of Molecular Pharmacology & Experimental Therapeutics; Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA; Department of Pediatric and Adolescent Medicine/Division of Pediatric Cardiology, Mayo Clinic, Rochester, MN, USA; Department of Cardiovascular Medicine; Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
18
|
Cheng Y, Lin G, Xie Y, Xuan B, He S, Shang Z, Yan M, Lin J, Wei L, Peng J, Shen A. Baicalin ameliorates angiotensin II-induced cardiac hypertrophy and mitogen-activated protein kinase signaling pathway activation: A target-based network pharmacology approach. Eur J Pharmacol 2024; 981:176876. [PMID: 39127302 DOI: 10.1016/j.ejphar.2024.176876] [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: 05/08/2024] [Revised: 07/20/2024] [Accepted: 08/08/2024] [Indexed: 08/12/2024]
Abstract
Baicalin, a flavonoid glycoside from Scutellaria baicalensis Georgi., exerts anti-hypertensive effects. The present study aimed to assess the cardioprotective role of baicalin and explore its potential mechanisms. Network pharmacology analysis pointed out a total of 477 potential targets of baicalin were obtained from the PharmMapper and SwissTargetPrediction databases, while 11,280 targets were identified associating with hypertensive heart disease from GeneCards database. Based on the above 382 common targets, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed enrichment in the regulation of cardiac hypertrophy, cardiac contraction, cardiac relaxation, as well as the mitogen-activated protein kinase (MAPK) and other signaling pathways. Moreover, baicalin treatment exhibited the amelioration of increased cardiac index and pathological alterations in angiotensin II (Ang II)-infused C57BL/6 mice. Furthermore, baicalin treatment demonstrated a reduction in cell surface area and a down-regulation of hypertrophy markers (including atrial natriuretic peptide and brain natriuretic peptide) in vivo and in vitro. In addition, baicalin treatment led to a decrease in the expression of phosphorylated c-Jun N-terminal kinase (p-JNK)/JNK, phosphorylated p38 (p-p38)/p38, and phosphorylated extracellular signal-regulated kinase (p-ERK)/ERK in the cardiac tissues of Ang II-infused mice and Ang II-stimulated H9c2 cells. These findings highlight the cardioprotective effects of baicalin, as it alleviates hypertensive cardiac injury, cardiac hypertrophy, and the activation of the MAPK pathway.
Collapse
Affiliation(s)
- Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Guosheng Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Yi Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Bihan Xuan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Shuyu He
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Zucheng Shang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Mengchao Yan
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Jing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China.
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China; Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, 350122, China; Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, 350122, China.
| |
Collapse
|
19
|
Taube N, Steiner M, Ebenebe-Kasonde OV, Kabir R, Garbus-Grant H, Alam El Din SM, Illingworth E, Wang N, Lin BL, Kohr MJ. Gestational arsenite exposure alters maternal postpartum heart size and induces Ca 2+ handling dysregulation in cardiomyocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615085. [PMID: 39386735 PMCID: PMC11463392 DOI: 10.1101/2024.09.25.615085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Cardiovascular disease is the leading cause of mortality in the US. Studies suggest a role for environmental exposures in the etiology of cardiovascular disease, including exposure to arsenic through drinking water. Arsenic exposure during pregnancy has been shown to have effects on offspring, but few studies have examined impacts on maternal cardiovascular health. While our prior work documented the detrimental effect of arsenic on the maternal heart during pregnancy, our current study examines the effect of gestational arsenic exposure on the maternal heart postpartum. Timed-pregnant wild-type (C57BL/6J) mice were exposed to 0, 100 or 1000 µg/L sodium arsenite (NaAsO2) via drinking water from embryonic day 2.5 (E2.5) until parturition. Postpartum heart structure and function was assessed via transthoracic echocardiography and gravimetric measurement. Hypertrophic markers were probed via qRT-PCR and western blot. Isolated cardiomyocyte Ca 2+ -handling and contraction were also assessed, and expression of proteins associated with Ca 2+ handling and contraction. Interestingly, we found that exposure to either 100 or 1000 µg/L sodium arsenite increased postpartum heart size at P12 vs. non-exposed postpartum controls. At the cellular level, we found altered cardiomyocyte Ca 2+ -handling and contraction. We also found altered expression of key contractile proteins, including α-Actin and cardiac myosin binding protein C (cMyBP-c). Together, these findings suggest that gestational arsenic exposure impacts the postpartum maternal heart, possibly inducing long-term cardiovascular changes. Furthermore, these findings highlight the importance of reducing arsenic exposure during pregnancy, and the need for more research on the impact of arsenic and other environmental exposures on maternal heart health and adverse pregnancy events. New & Noteworthy Gestational exposure to sodium arsenite at environmentally relevant doses (100 and 1000 µg/L) increases postpartum heart size, and induces dysregulated Ca 2+ homeostasis and impaired shortening in isolated cardiomyocytes. This is the first study to demonstrate that gestational arsenic exposure impacts postpartum heart structure and function beyond the exposure period.
Collapse
|
20
|
Ibrahim Z, Khan NA, Siddiqui R, Qaisar R, Marzook H, Soares NC, Elmoselhi AB. Gut matters in microgravity: potential link of gut microbiota and its metabolites to cardiovascular and musculoskeletal well-being. Nutr Metab (Lond) 2024; 21:66. [PMID: 39123239 PMCID: PMC11316329 DOI: 10.1186/s12986-024-00836-6] [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: 02/08/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
The gut microbiota and its secreted metabolites play a significant role in cardiovascular and musculoskeletal health and diseases. The dysregulation of the intestinal microbiota poses a significant threat to cardiovascular and skeletal muscle well-being. Nonetheless, the precise molecular mechanisms underlying these changes remain unclear. Furthermore, microgravity presents several challenges to cardiovascular and musculoskeletal health compromising muscle strength, endothelial dysfunction, and metabolic changes. The purpose of this review is to critically examine the role of gut microbiota metabolites on cardiovascular and skeletal muscle functions and dysfunctions. It also explores the molecular mechanisms that drive microgravity-induced deconditioning in both cardiovascular and skeletal muscle. Key findings in this review highlight that several alterations in gut microbiota and secreted metabolites in microgravity mirror characteristics seen in cardiovascular and skeletal muscle diseases. Those alterations include increased levels of Firmicutes/Bacteroidetes (F/B) ratio, elevated lipopolysaccharide levels (LPS), increased in para-cresol (p-cresol) and secondary metabolites, along with reduction in bile acids and Akkermansia muciniphila bacteria. Highlighting the potential, modulating gut microbiota in microgravity conditions could play a significant role in mitigating cardiovascular and skeletal muscle diseases not only during space flight but also in prolonged bed rest scenarios here on Earth.
Collapse
Affiliation(s)
- Zeinab Ibrahim
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Naveed A Khan
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Ruqaiyyah Siddiqui
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS,, UK
- Microbiota Research Center, Istinye University, Istanbul, 34010, Turkey
| | - Rizwan Qaisar
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hezlin Marzook
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE
| | - Nelson C Soares
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid university of Medicine and Health Sciences, Dubai, 0000, United Arab Emirates
- Laboratory of Proteomics, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA), Av Padre Cruz, Lisbon, 1649-016, Portugal
| | - Adel B Elmoselhi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, 27272, UAE.
- Basic Medical Sciences Department, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
21
|
Adasheva DA, Serebryanaya DV. IGF Signaling in the Heart in Health and Disease. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1402-1428. [PMID: 39245453 DOI: 10.1134/s0006297924080042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/22/2024] [Indexed: 09/10/2024]
Abstract
One of the most vital processes of the body is the cardiovascular system's proper operation. Physiological processes in the heart are regulated by the balance of cardioprotective and pathological mechanisms. The insulin-like growth factor system (IGF system, IGF signaling pathway) plays a pivotal role in regulating growth and development of various cells and tissues. In myocardium, the IGF system provides cardioprotective effects as well as participates in pathological processes. This review summarizes recent data on the role of IGF signaling in cardioprotection and pathogenesis of various cardiovascular diseases, as well as analyzes severity of these effects in various scenarios.
Collapse
Affiliation(s)
- Daria A Adasheva
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Daria V Serebryanaya
- Department of Biochemistry, Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.
- Pirogov Russian National Research Medical University, Moscow, 117997, Russia
| |
Collapse
|
22
|
Zhao R, Hu J, Wen H, Zhao J, Wang Y, Niu X, Zhang M, Wang T, Li Y. Inhibition of N-acetylglucosaminyltransferase V alleviates diabetic cardiomyopathy in mice by attenuating cardiac hypertrophy and fibrosis. Nutr Metab (Lond) 2024; 21:53. [PMID: 39080739 PMCID: PMC11290217 DOI: 10.1186/s12986-024-00797-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/18/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND The pathogenesis of diabetic cardiomyopathy is closely linked to abnormal glycosylation modifications. N-acetylglucosaminyltransferase V (GnT-V), which catalyzes the production of N-linked -1-6 branching of oligosaccharides, is involved in several pathophysiological mechanisms of many disorders, including cardiac hypertrophy and heart failure. However, the mechanism by which GnT-V regulates cardiac hypertrophy in diabetic cardiomyopathy is currently poorly understood. In this study, we investigated the role of GnT-V on myocardial hypertrophy in diabetic cardiomyopathy and elucidated the underlying mechanisms. MATERIAL AND METHODS Streptozotocin (STZ) was intraperitoneally injected into mice to induce diabetic cardiomyopathy. An adeno-associated virus (AAV) carrying negative control small hairpin RNA (shNC) or GnT-V-specifc small hairpin RNA (shGnT-V) was used to manipulate GnT-V expression. In our study, forty male C57BL/6J mice were randomly divided into four groups (10 mice per group): control mice with AAV-shNC, diabetic cardiomyopathy mice with AAV-shNC, control mice with AAV-shGnT-V, and diabetic cardiomyopathy mice with AAV-shGnT-V. In addition, H9C2 cells and primary neonatal cardiac fibroblasts treated with high glucose were used as a cell model of diabetes. Analysis of cardiac hypertrophy and fibrosis, as well as functional studies, were used to investigate the underlying molecular pathways. RESULTS AAV-mediated GnT-V silencing dramatically improved cardiac function and alleviated myocardial hypertrophy and fibrosis in diabetic mice. In vitro experiments demonstrated that GnT-V was elevated in cardiomyocytes and induced cardiomyocyte hypertrophy in response to high glucose stimulation. GnT-V knockdown significantly reduced the expression of the integrinβ1 signaling pathway, as evidenced by decreased downstream ERK1/2 activity, which inhibited cardiomyocyte hypertrophy accompanied by reduced ANP, BNP, and β-MHC expression. Furthermore, knocking down GnT-V expression lowered the TGF-β1-Smads signaling pathway, which reduced the expression of α-SMA, collagen I, and collagen III. CONCLUSIONS Overall, our research indicated that GnT-V may be a useful therapeutic target to treat diabetic cardiomyopathy, primarily in the inhibition of myocardial hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Ran Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Jianqiang Hu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - He Wen
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Jieqiong Zhao
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Ying Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Xiaona Niu
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China
| | - Mingming Zhang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| | - Tingting Wang
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| | - Yan Li
- Department of Cardiology, Tangdu Hospital, Air Force Medical University, Xinsi Road No.569, Xi'an, 710038, People's Republic of China.
| |
Collapse
|
23
|
Wan S, Wang S, Yang X, Cui Y, Guan H, Xiao W, Liu F. Regulation of H9C2 cell hypertrophy by 14-3-3η via inhibiting glycolysis. PLoS One 2024; 19:e0307696. [PMID: 39038022 PMCID: PMC11262655 DOI: 10.1371/journal.pone.0307696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
It has been reported that Ywhah (14-3-3η) reduces glycolysis. However, it remains unclear about the downstream mechanism by which glycolysis is regulated by 14-3-3η in cardiac hypertrophy. As an important regulator, Yes-associated protein (YAP) interacts with 14-3-3η to participate in the initiation and progression of various diseases in vivo. In this study, the model of H9C2 cardiomyocyte hypertrophy was established by triiodothyronine (T3) or rotenone stimulation to probe into the action mechanism of 14-3-3η. Interestingly, the overexpression of 14-3-3η attenuated T3 or rotenone induced cardiomyocyte hypertrophy and decreased glycolysis in H9C2 cardiomyocytes, whereas the knockdown of 14-3-3η had an opposite effect. Mechanistically, 14-3-3η can reduce the expression level of YAP and bind to it to reduce its nuclear translocation. In addition, changing YAP may affect the expression of lactate dehydrogenase A (LDHA), a glycolysis-related protein. Meanwhile, LDHA is also a possible target for 14-3-3η to mediate glycolysis based on changes in pyruvate, a substrate of LDHA. Collectively, 14-3-3η can suppress cardiomyocyte hypertrophy via decreasing the nucleus translocation of YAP and glycolysis, which indicates that 14-3-3η could be a promising target for inhibiting cardiac hypertrophy.
Collapse
Affiliation(s)
- Sha Wan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Songhao Wang
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Xianfei Yang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
| | - Yalan Cui
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
- Clinical Pathology Department, The Second People’s Hospital of Yichang, Yichang, China
| | - Heng Guan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Wenping Xiao
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Fang Liu
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, China
- Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, China
| |
Collapse
|
24
|
Wu T, Gao R, Wang X, Guo D, Xie Y, Dong B, Hao X, Zhu C. Pancreatobiliary reflux increases macrophage-secreted IL-8 and activates the PI3K/NFκB pathway to promote cholangiocarcinoma progression. Transl Oncol 2024; 45:101967. [PMID: 38653100 PMCID: PMC11059331 DOI: 10.1016/j.tranon.2024.101967] [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: 01/22/2024] [Revised: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Persistent pancreaticobiliary reflux (PBR) is associated with a high risk of biliary malignancy. This study aimed to evaluate the proportion of PBR in biliary tract diseases and mechanisms by which PBR promoted cholangiocarcinoma progression. METHODS Overall 227 consecutive patients with primary biliary tract disease participated in this study. The amylase levels in the collected bile were analyzed. The mechanisms underlying the effect of high-amylase bile on bile duct epithelial and cholangiocarcinoma cells progression were analyzed. The source of interleukin-8 (IL-8) and its effects on the biological functions of cholangiocarcinoma cells were investigated. RESULTS The bile amylase levels in 148 of 227 patients were higher than the upper serum amylase limit of 135 IU/L. PBR was significantly correlated with sex, pyrexia, and serum gamma-glutamyl transferase (GGT) levels in the patient cohort. High-amylase bile-induced DNA damage and genetic differences in the transcript levels of the gallbladder mucosa and facilitated the proliferation and migration of bile duct cancer cells (HUCCT1 and QBC939 cells). The concentration of many cytokines increased in high-amylase bile. IL-8 is secreted primarily by macrophages via the mitogen-activated protein kinase pathway and partially by bile duct epithelial cells. IL-8 promotes the progression of HUCCT1 and QBC939 cells by regulating the expression of epithelial-mesenchymal transition-associated proteins and activating the phosphatidylinositol 3-kinase/nuclear factor kappa-B pathway. CONCLUSIONS PBR is one of the primary causes of biliary disease. IL-8 secreted by macrophages or bile duct epithelial cells stimulated by high-amylase bile promotes cholangiocarcinoma progression.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Ruiqian Gao
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Xiaowei Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Dong Guo
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Yuwei Xie
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China
| | - Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266003, China
| | - Xiwei Hao
- Department of Pediatric Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China.
| | - Chengzhan Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao 266003, China; Shandong Key Laboratory of Digital Medicine and Computer Assisted Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
25
|
Mohammed KAK, Madeddu P, Avolio E. MEK inhibitors: a promising targeted therapy for cardiovascular disease. Front Cardiovasc Med 2024; 11:1404253. [PMID: 39011492 PMCID: PMC11247000 DOI: 10.3389/fcvm.2024.1404253] [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] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Cardiovascular disease (CVD) represents the leading cause of mortality and disability all over the world. Identifying new targeted therapeutic approaches has become a priority of biomedical research to improve patient outcomes and quality of life. The RAS-RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase) pathway is gaining growing interest as a potential signaling cascade implicated in the pathogenesis of CVD. This pathway is pivotal in regulating cellular processes like proliferation, growth, migration, differentiation, and survival, which are vital in maintaining cardiovascular homeostasis. In addition, ERK signaling is involved in controlling angiogenesis, vascular tone, myocardial contractility, and oxidative stress. Dysregulation of this signaling cascade has been linked to cell dysfunction and vascular and cardiac pathological remodeling, which contribute to the onset and progression of CVD. Recent and ongoing research has provided insights into potential therapeutic interventions targeting the RAS-RAF-MEK-ERK pathway to improve cardiovascular pathologies. Preclinical studies have demonstrated the efficacy of targeted therapy with MEK inhibitors (MEKI) in attenuating ERK activation and mitigating CVD progression in animal models. In this article, we first describe how ERK signaling contributes to preserving cardiovascular health. We then summarize current knowledge of the roles played by ERK in the development and progression of cardiac and vascular disorders, including atherosclerosis, myocardial infarction, cardiac hypertrophy, heart failure, and aortic aneurysm. We finally report novel therapeutic strategies for these CVDs encompassing MEKI and discuss advantages, challenges, and future developments for MEKI therapeutics.
Collapse
Affiliation(s)
- Khaled A K Mohammed
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Paolo Madeddu
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Elisa Avolio
- Bristol Heart Institute, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
26
|
Fuentes-Mateos R, García-Navas R, Fernández-Infante C, Hernández-Cano L, Calzada-Nieto N, Juan AOS, Guerrero C, Santos E, Fernández-Medarde A. Combined HRAS and NRAS ablation induces a RASopathy phenotype in mice. Cell Commun Signal 2024; 22:332. [PMID: 38886790 PMCID: PMC11184836 DOI: 10.1186/s12964-024-01717-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND HRASKO/NRASKO double knockout mice exhibit exceedingly high rates of perinatal lethality due to respiratory failure caused by a significant lung maturation delay. The few animals that reach adulthood have a normal lifespan, but present areas of atelectasis mixed with patches of emphysema and normal tissue in the lung. METHODS Eight double knockout and eight control mice were analyzed using micro-X-ray computerized tomography and a Small Animal Physiological Monitoring system. Tissues and samples from these mice were analyzed using standard histological and Molecular Biology methods and the significance of the results analyzed using a Student´s T-test. RESULTS The very few double knockout mice surviving up to adulthood display clear craniofacial abnormalities reminiscent of those seen in RASopathy mouse models, as well as thrombocytopenia, bleeding anomalies, and reduced platelet activation induced by thrombin. These surviving mice also present heart and spleen hyperplasia, and elevated numbers of myeloid-derived suppressor cells in the spleen. Mechanistically, we observed that these phenotypic alterations are accompanied by increased KRAS-GTP levels in heart, platelets and primary mouse embryonic fibroblasts from these animals. CONCLUSIONS Our data uncovers a new, previously unidentified mechanism capable of triggering a RASopathy phenotype in mice as a result of the combined removal of HRAS and NRAS.
Collapse
Affiliation(s)
- Rocío Fuentes-Mateos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
- Present address: Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, University of Groningen, Groningen, Netherlands
| | - Rósula García-Navas
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Cristina Fernández-Infante
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Luis Hernández-Cano
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Present address: Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, Groningen, Netherlands
| | - Nuria Calzada-Nieto
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Andrea Olarte-San Juan
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain
| | - Carmen Guerrero
- Instituto de Biología Molecular y Celular del Cáncer (IMBCC), USAL-CSIC. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Eugenio Santos
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain.
| | - Alberto Fernández-Medarde
- Centro de Investigación del Cáncer-Instituto de Biología Molecular y Celular del Cáncer (CSIC-Universidad de Salamanca) and CIBERONC, Campus Unamuno, University of Salamanca, 37007, Salamanca, Spain.
| |
Collapse
|
27
|
Huang S, Zhang Y, Shu H, Liu W, Zhou X, Zhou X. Advances of the MAPK pathway in the treatment of spinal cord injury. CNS Neurosci Ther 2024; 30:e14807. [PMID: 38887853 PMCID: PMC11183187 DOI: 10.1111/cns.14807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/23/2024] [Accepted: 05/27/2024] [Indexed: 06/20/2024] Open
Abstract
Spinal cord injury (SCI) represents a complex pathology within the central nervous system (CNS), leading to severe sensory and motor impairments. It activates various signaling pathways, notably the mitogen-activated protein kinase (MAPK) pathway. Present treatment approaches primarily focus on symptomatic relief, lacking efficacy in addressing the underlying pathophysiological mechanisms. Emerging research underscores the significance of the MAPK pathway in neuronal differentiation, growth, survival, axonal regeneration, and inflammatory responses post-SCI. Modulating this pathway post-injury has shown promise in attenuating inflammation, minimizing apoptosis, alleviating neuropathic pain, and fostering neural regeneration. Given its pivotal role, the MAPK pathway emerges as a potential therapeutic target in SCI management. This review synthesizes current knowledge on SCI pathology, delineates the MAPK pathway's characteristics, and explores its dual roles in SCI pathology and therapeutic interventions. Furthermore, it addresses the existing challenges in MAPK research in the context of SCI, proposing solutions to overcome these hurdles. Our aim is to offer a comprehensive reference for future research on the MAPK pathway and SCI, laying the groundwork for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Shixue Huang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Yinuo Zhang
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Haoming Shu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Wei Liu
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xin Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Xuhui Zhou
- Department of Orthopedics, Changzheng HospitalSecond Affiliated Hospital of Naval Medical UniversityShanghaiChina
- Translational Research Centre of Orthopedics, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
28
|
Cooper STE, Lokman AB, Riley PR. Role of the Lymphatics in Cardiac Disease. Arterioscler Thromb Vasc Biol 2024; 44:1181-1190. [PMID: 38634279 DOI: 10.1161/atvbaha.124.319854] [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: 04/19/2024]
Abstract
Cardiovascular diseases remain the largest cause of death worldwide with recent evidence increasingly attributing the development and progression of these diseases to an exacerbated inflammatory response. As a result, significant research is now focused on modifying the immune environment to prevent the disease progression. This in turn has highlighted the lymphatic system in the pathophysiology of cardiovascular diseases owing, in part, to its established function in immune cell surveillance and trafficking. In this review, we highlight the role of the cardiac lymphatic system and its potential as an immunomodulatory therapeutic target in selected cardiovascular diseases.
Collapse
Affiliation(s)
- Susanna T E Cooper
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Adam B Lokman
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| | - Paul R Riley
- Institute of Developmental and Regenerative Medicine, Department of Physiology, Anatomy and Genetics, University of Oxford, United Kingdom
| |
Collapse
|
29
|
Bisht K, Verma VK, Abdullah Z, Prajapati V, Rajiv N, Bhatia J, Ray R, Nag TC, Arya DS. Arglabin: A mediator of inflammasome modulated and independent myocardial injury (PARA-AMI study). Eur J Pharmacol 2024; 970:176465. [PMID: 38479722 DOI: 10.1016/j.ejphar.2024.176465] [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: 10/22/2023] [Revised: 02/01/2024] [Accepted: 02/28/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Arglabin is a plant alkaloid (sesquiterpene lactone) that is used as an anticancer drug. It has potential anti-diabetic and anti-atherogenic effects. PURPOSE Arglabin has drawn particular attention because of its therapeutic effects as an anti-inflammatory agent in multiple diseases. Since arglabin inhibits Epidermal Growth Factor Receptor (EGFR) tyrosine kinase, concerns for cardiotoxic effects are valid. The present study was designed to investigate the protective effects of arglabin on the myocardium. STUDY DESIGN This study was designed to evaluate the effect of arglabin on the myocardium in an experimental model of myocardial necrosis in rats. Different doses of arglabin (2.5, 5, and 10 μg/kg) were investigated as pre-treatment for 21 days in the isoproterenol (ISO) model of myocardial necrosis groups and per se groups. METHODS On the 22nd day, hemodynamic, histopathological, electron microscopy, oxidative stress markers, inflammatory mediators, apoptotic markers, inflammasome mediators, and Western blot analysis were performed to evaluate the effects of arglabin. RESULTS Arglabin pre-treatment showed improvement in hemodynamic parameters and histopathological findings at low doses in isoproterenol-induced myocardial necrosis model of rats. Arglabin administration altered myocardial structure and modulated myocardial function via activation of NFκB/MAPK pathway that led to myocardial injury with an increase in dose. CONCLUSION Arglabin imparted partial cardio-protection via an inflammasome-dependent pathway and mediated injury through the inflammasome-independent pathway.
Collapse
Affiliation(s)
- Khushboo Bisht
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Vipin Kumar Verma
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Zia Abdullah
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Prajapati
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Narang Rajiv
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Jagriti Bhatia
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Ruma Ray
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Dharamvir Singh Arya
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
30
|
Julovi SM, Trinh K, Robertson H, Xu C, Minhas N, Viswanathan S, Patrick E, Horowitz JD, Meijles DN, Rogers NM. Thrombospondin-1 Drives Cardiac Remodeling in Chronic Kidney Disease. JACC Basic Transl Sci 2024; 9:607-627. [PMID: 38984053 PMCID: PMC11228122 DOI: 10.1016/j.jacbts.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 07/11/2024]
Abstract
Patients with chronic kidney disease (CKD) face a high risk of cardiovascular disease. Previous studies reported that endogenous thrombospondin 1 (TSP1) involves right ventricular remodeling and dysfunction. Here we show that a murine model of CKD increased myocardial TSP1 expression and produced left ventricular hypertrophy, fibrosis, and dysfunction. TSP1 knockout mice were protected from these features. In vitro, indoxyl sulfate is driving deleterious changes in cardiomyocyte through the TSP1. In patients with CKD, TSP1 and aryl hydrocarbon receptor were both differentially expressed in the myocardium. Our findings summon large clinical studies to confirm the translational role of TSP1 in patients with CKD.
Collapse
Affiliation(s)
- Sohel M Julovi
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
| | - Katie Trinh
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Harry Robertson
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
| | - Cuicui Xu
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Nikita Minhas
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Seethalakshmi Viswanathan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Tissue Pathology and Diagnostic Oncology, Institute of Clinical Pathology and Medical Research, Westmead Hospital, Westmead, New South Wales, Australia
| | - Ellis Patrick
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Sydney Precision Data Science Centre, University of Sydney, New South Wales, Australia
- School of Mathematics, University of Sydney, New South Wales, Australia
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong Special Administrative Region, China
| | - John D Horowitz
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
- Cardiovascular Pathophysiology and Therapeutics Research Group, Basil Hetzel Institute, Woodville, South Australia, Australia
- Department of Medicine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Daniel N Meijles
- Molecular and Clinical Sciences Research Institute, St George's University of London, London, United Kingdom
| | - Natasha M Rogers
- Kidney Injury Group, Centre for Transplant and Renal Research, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Renal and Transplantation Unit, Westmead Hospital, New South Wales, Australia
| |
Collapse
|
31
|
Pontes CNR, Bessa ADSMD, Macedo LM, Ferreira-Junior MD, Cavalcante KVN, Campos HM, Cruz-Leite VRM, Neves ÂR, Gomes RM, Ghedini PC, Biancardi MF, Mendes EP, Borges CL, Pedrino GR, Castro CH. Angiotensin-(1-7) Treatment Early in Life Prevents Cardiac Hypertrophy in Adult Hypertensive Rats. J Cardiovasc Pharmacol 2024; 83:457-465. [PMID: 39536129 DOI: 10.1097/fjc.0000000000001530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/30/2023] [Indexed: 03/20/2024]
Abstract
ABSTRACT Angiotensin (Ang)-(1-7) is a cardioprotective peptide of the renin-angiotensin system. Prepuberty has been considered as a later susceptible window of development, and stressful factors in this life phase can induce chronic diseases in adulthood. We aimed to investigate whether the treatment with Ang-(1-7) during the prepuberty could attenuate the development of hypertension and cardiac injury in adult spontaneously hypertensive rats (SHRs). SHRs were treated with Ang-(1-7) (24 μg/kg/h) from age 4 to 7 weeks. Systolic blood pressure was measured by tail-cuff plethysmography up to 17th week. Thereafter, echocardiography was performed, and the rats were euthanized for the collection of tissues and blood. Ang-(1-7) did not change the systolic blood pressure but reduced the septal and posterior wall thickness, and cardiomyocyte hypertrophy and fibrosis in SHR. In addition, Ang-(1-7) reduced the gene expression of atrial natriuretic peptide and brain natriuretic peptide, increased the metalloproteinase 9 expression, and reduced the extracellular signal-regulated kinases 1/2 phosphorylation. Ang-(1-7) also prevented the reduction of Mas receptor but did not change the protein expression of angiotensin-converting enzyme, angiotensin-converting enzyme 2, AT1, and AT2. The treatment with Ang-(1-7) decreased the malondialdehyde (MDA) levels and increased superoxide dismutase-1 and catalase activities and protein expression of catalase. Our findings demonstrate that the treatment of SHR with Ang-(1-7) for 3 weeks early in life promotes beneficial effects in the heart later in life, even without altering blood pressure, through mechanisms involving the reduction of oxidative stress and ERK1/2 phosphorylation. In addition, this study supports the prepuberty as an important programming window.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ângela Ribeiro Neves
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
| | | | | | | | - Clayton Luiz Borges
- Department of Biochemistry and Molecular Biology, Federal University of Goiás, Goiânia, Brazil
| | | | - Carlos Henrique Castro
- Department of Physiological Sciences, Federal University of Goiás, Goiânia, Brazil
- National Institute of Science and Technology in Nanobiopharmaceutics
| |
Collapse
|
32
|
Sahu R, Rawal RK. Modulation of the c-JNK/p38-MAPK signaling pathway: Investigating the therapeutic potential of natural products in hypertension. PHYTOMEDICINE PLUS 2024; 4:100564. [DOI: 10.1016/j.phyplu.2024.100564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
33
|
Todorova VK, Bauer MA, Azhar G, Wei JY. RNA sequencing of formalin fixed paraffin-embedded heart tissue provides transcriptomic information about chemotherapy-induced cardiotoxicity. Pathol Res Pract 2024; 257:155309. [PMID: 38678848 DOI: 10.1016/j.prp.2024.155309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 04/11/2024] [Indexed: 05/01/2024]
Abstract
Gene expression of formalin-fixed paraffin-embedded (FFPE) tissue may serve for molecular studies on cardiovascular diseases. Chemotherapeutics, such as doxorubicin (DOX) may cause heart injury, but the mechanisms of these side effects of DOX are not well understood. This study aimed to investigate whether DOX-induced gene expression in archival FFPE heart tissue in experimental rats would correlate with the gene expression in fresh-frozen heart tissue by applying RNA sequencing technology. The results showed RNA from FFPE samples was degraded, resulting in a lower number of uniquely mapped reads. However, DOX-induced differentially expressed genes in FFPE were related to molecular mechanisms of DOX-induced cardiotoxicity, such as inflammation, calcium binding, endothelial dysfunction, senescence, and cardiac hypertrophy signaling. Our data suggest that, despite the limitations, RNA sequencing of archival FFPE heart tissue supports utilizing FFPE tissues from retrospective studies on cardiovascular disorders, including DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Valentina K Todorova
- Division of Hematology/Oncology, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| | - Michael A Bauer
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Gohar Azhar
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jeanne Y Wei
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
34
|
Lee J, Rogers HM, Springer DA, Noguchi CT. Neuronal nitric oxide synthase required for erythropoietin modulation of heart function in mice. Front Physiol 2024; 15:1338476. [PMID: 38628440 PMCID: PMC11019009 DOI: 10.3389/fphys.2024.1338476] [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: 11/14/2023] [Accepted: 03/04/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: Erythropoietin (EPO) acts primarily in regulating red blood cell production mediated by high EPO receptor (EPOR) expression in erythroid progenitor cells. EPO activity in non-erythroid tissue is evident in mice with EPOR restricted to erythroid tissues (ΔEPORE) that become obese, glucose-intolerant, and insulin-resistant. In animal models, nitric oxide synthase (NOS) contributes to EPO activities including erythropoiesis, neuroprotection, and cardioprotection against ischemia-reperfusion injury. However, we found that extended EPO treatment to increase hematocrit compromised heart function, while the loss of neuronal NOS (nNOS) was protective against the deleterious activity of EPO to promote heart failure. Methods: Wild-type (WT) mice, ΔEPORE mice, and nNOS-knockout mice (nNOS-/-) were placed on a high-fat diet to match the ΔEPORE obese phenotype and were treated with EPO for 3 weeks. Hematocrit and metabolic response to EPO treatment were monitored. Cardiac function was assessed by echocardiography and ultrasonography. Results: ΔEPORE mice showed a decrease in the left ventricular outflow tract (LVOT) peak velocity, ejection fraction, and fractional shortening, showing that endogenous non-erythroid EPO response is protective for heart function. EPO treatment increased hematocrit in all mice and decreased fat mass in male WT, demonstrating that EPO regulation of fat mass requires non-erythroid EPOR. EPO treatment also compromised heart function in WT mice, and decreased the pulmonary artery peak velocity (PA peak velocity), LVOT peak velocity, ejection fraction, and fractional shortening, but it had minimal effect in further reducing the heart function in ΔEPORE mice, indicating that the adverse effect of EPO on heart function is not related to EPO-stimulated erythropoiesis. ΔEPORE mice had increased expression of heart failure-associated genes, hypertrophic cardiomyopathy-related genes, and sarcomeric genes that were also elevated with EPO treatment in WT mice. Male and female nNOS-/- mice were protected against diet-induced obesity. EPO treatment in nNOS-/- mice increased the hematocrit that tended to be lower than WT mice and decreased the PA peak velocity but did not affect the LVOT peak velocity, ejection fraction, and fractional shortening, suggesting that nNOS is required for the adverse effect of EPO treatment on heart function in WT mice. EPO treatment did not change expression of heart failure-associated gene expression in nNOS-/- mice. Discussion: Endogenous EPO has a protective effect on heart function. With EPO administration, in contrast to the protective effect to the cardiac injury of acute EPO treatment, extended EPO treatment to increase hematocrit in WT mice adversely affected the heart function with a corresponding increase in expression of heart failure-associated genes. This EPO activity was independent of EPO-stimulated erythropoiesis and required EPOR in non-erythroid tissue and nNOS activity, while nNOS-/- mice were protected from the EPO-associated adverse effect on heart function. These data provide evidence that nNOS contributes to the negative impact on the heart function of high-dose EPO treatment for anemia.
Collapse
Affiliation(s)
- Jeeyoung Lee
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Heather M. Rogers
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Danielle A. Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Constance T. Noguchi
- Molecular Medicine Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
35
|
Zhang M, Jin Y, Guo X, Shan W, Zhang J, Yuan A, Shi Y. Resveratrol protects mesangial cells under high glucose by regulating the miR-1231/IGF1/ERK pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:2326-2339. [PMID: 38156429 DOI: 10.1002/tox.24103] [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: 10/03/2023] [Revised: 11/13/2023] [Accepted: 12/10/2023] [Indexed: 12/30/2023]
Abstract
Diabetic nephropathy (DN) is one of the complications of diabetes mellitus and the main cause of end-stage renal disease (ESRD), which is a serious threat to human health. In DN, mesangial cells (MCs) are a critical target cell that perform a variety of key functions, and abnormal proliferation of MCs is a common and prominent pathological change in DN. In recent years, the investigation of Chinese medicine interventions for DN has increased significantly in recent years due to the many potential adverse effects and controversies associated with the treatment of DN with Western medicines. In this study, we evaluated the protective effect of resveratrol (RES), an active ingredient known as a natural antioxidant, on HMCs under high glucose and explored its possible mechanism of action. We found that RES inhibited the proliferation of human mesangial cell (HMC) under high glucose and blocked cell cycle progression. In the high glucose environment, RES upregulated miR-1231, reduced IGF1 expression, inhibited the activity of the extracellular signal-regulated kinase (ERK) signaling pathway and reduced levels of the inflammatory factors TNF-α and IL-6. In addition, we found that miR-1231 mimics were synergistically inhibited with RES, whereas miR-1231 inhibitor attenuated the protective effect of RES on HMCs. Thus, our results suggest that the protective effect of RES on HMCs under high glucose is achieved, at least in part, through modulation of the miR-1231/IGF1/ERK pathway. The discovery of this potential mechanism may provide a new molecular therapeutic target for the prevention and treatment of DN, and may also bring new ideas for the clinical research in DN.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| | - Yingli Jin
- Department of Pharmacology, School of Basic Medical Science, Jilin University, Changchun, Jilin Province, China
| | - Xuerui Guo
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| | - Wanxin Shan
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| | - Jinlong Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| | - Aoxue Yuan
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| | - Yan Shi
- Department of Experimental Pharmacology and Toxicology, School of Pharmacy, Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
36
|
Sun H, Du B, Fu H, Yue Z, Wang X, Yu S, Zhang Z. Canagliflozin combined with aerobic exercise protects against chronic heart failure in rats. iScience 2024; 27:109014. [PMID: 38439968 PMCID: PMC10910240 DOI: 10.1016/j.isci.2024.109014] [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: 07/05/2023] [Revised: 12/01/2023] [Accepted: 01/22/2024] [Indexed: 03/06/2024] Open
Abstract
To determine the efficacy and potential protective mechanism of canagliflozin combined with aerobic exercise in treating chronic heart failure (CHF). Isoproterenol was injected into rats to create CHF models. The rats were then subsequently divided into saline, canagliflozin (3 mg/kg/d), aerobic exercise training, and canagliflozin combined with aerobic exercise training. Compared to the CHF group, the canagliflozin combined with the aerobic exercise group had superior ventricular remodeling and cardiac function. In rats treated with canagliflozin combined with aerobic exercise, the expression of cytochrome P450 (CYP) 4A3, CYP4A8, COL1A1, COL3A1, and FN1 was reduced, while the expression of CYP26B1, ALDH1A2, and CYP1A1 increased significantly. Additionally, canagliflozin combined with aerobic exercise decreased the phosphorylation of AKT and ERK1/2. Canagliflozin combined with aerobic exercise has a positive effect on the development of CHF via the regulation of retinol metabolism and the AKT/ERK signaling pathway.
Collapse
Affiliation(s)
- Helin Sun
- Department of Endocrinology and Metabology, Shenzhen Research Institute of Shandong University, Shenzhen, China, Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bingyu Du
- Department of Endocrinology and Metabology, Shenzhen Research Institute of Shandong University, Shenzhen, China, Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hui Fu
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhaodi Yue
- Teaching and Research Section of Internal Medicine, College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of rehabilitation medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xueyin Wang
- Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Shaohong Yu
- Teaching and Research Section of Internal Medicine, College of Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Department of rehabilitation medicine, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhongwen Zhang
- Department of Endocrinology and Metabology, Shenzhen Research Institute of Shandong University, Shenzhen, China, Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Endocrinology and Metabology, The Third Affiliated Hospital of Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
37
|
Moreira-Costa L, Tavares-Silva M, Almeida-Coelho J, Gonçalves A, Trindade F, Vasques-Nóvoa F, Sousa-Mendes C, Leite S, Vitorino R, Falcão-Pires I, Leite-Moreira AF, Lourenço AP. Acute and chronic effects of levosimendan in the ZSF1 obese rat model of heart failure with preserved ejection fraction. Eur J Pharmacol 2024; 966:176336. [PMID: 38272343 DOI: 10.1016/j.ejphar.2024.176336] [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: 09/22/2023] [Revised: 01/02/2024] [Accepted: 01/16/2024] [Indexed: 01/27/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a syndrome characterized by impaired cardiovascular reserve in which therapeutic options are scarce. Our aim was to evaluate the inodilator levosimendan in the ZSF1 obese rat model of HFpEF. Twenty-week-old male Wistar-Kyoto (WKY), ZSF1 lean (ZSF1 Ln) and ZSF1 obese rats chronically treated for 6-weeks with either levosimendan (1 mg/kg/day, ZSF1 Ob + Levo) or vehicle (ZSF1 Ob + Veh) underwent peak-effort testing, pressure-volume (PV) haemodynamic evaluation and echocardiography (n = 7 each). Samples were collected for histology and western blotting. In obese rats, skinned and intact left ventricular (LV) cardiomyocytes underwent in vitro functional evaluation. Seven additional ZSF1 obese rats underwent PV evaluation to assess acute levosimendan effects (10 μg/kg + 0.1 μg/kg/min). ZSF1 Ob + Veh presented all hallmarks of HFpEF, namely effort intolerance, elevated end-diastolic pressures and reduced diastolic compliance as well as increased LV mass and left atrial area, cardiomyocyte hypertrophy and increased interstitial fibrosis. Levosimendan decreased systemic arterial pressures, raised cardiac index, and enhanced LV relaxation and diastolic compliance in both acute and chronic experiments. ZSF1 Ob + Levo showed pronounced attenuation of hypertrophy and interstitial fibrosis alongside increased effort tolerance (endured workload raised 38 %) and maximum O2 consumption. Skinned cardiomyocytes from ZSF 1 Ob + Levo showed a downward shift in sarcomere length-passive tension relationship and intact cardiomyocytes showed decreased diastolic Ca2+ levels and enhanced Ca2+ sensitivity. On molecular grounds, levosimendan enhanced phosphorylation of phospholamban and mammalian target of rapamycin. The observed effects encourage future clinical trials with levosimendan in a broad population of HFpEF patients.
Collapse
Affiliation(s)
- Liliana Moreira-Costa
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal.
| | - Marta Tavares-Silva
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Cardiology, Centro Hospitalar Universitário São João, Porto, Portugal
| | - João Almeida-Coelho
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Alexandre Gonçalves
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Fábio Trindade
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Francisco Vasques-Nóvoa
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Medicine, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Cláudia Sousa-Mendes
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Sara Leite
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Rui Vitorino
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Medical Sciences, Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Inês Falcão-Pires
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Adelino F Leite-Moreira
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Cardiothoracic Surgery, Centro Hospitalar Universitário São João, Porto, Portugal
| | - André P Lourenço
- Cardiovascular R&D Centre - UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Porto, Portugal; Department of Anaesthesiology, Centro Hospitalar Universitário São João, Porto, Portugal
| |
Collapse
|
38
|
Jolfayi AG, Kohansal E, Ghasemi S, Naderi N, Hesami M, MozafaryBazargany M, Moghadam MH, Fazelifar AF, Maleki M, Kalayinia S. Exploring TTN variants as genetic insights into cardiomyopathy pathogenesis and potential emerging clues to molecular mechanisms in cardiomyopathies. Sci Rep 2024; 14:5313. [PMID: 38438525 PMCID: PMC10912352 DOI: 10.1038/s41598-024-56154-7] [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: 11/22/2023] [Accepted: 03/01/2024] [Indexed: 03/06/2024] Open
Abstract
The giant protein titin (TTN) is a sarcomeric protein that forms the myofibrillar backbone for the components of the contractile machinery which plays a crucial role in muscle disorders and cardiomyopathies. Diagnosing TTN pathogenic variants has important implications for patient management and genetic counseling. Genetic testing for TTN variants can help identify individuals at risk for developing cardiomyopathies, allowing for early intervention and personalized treatment strategies. Furthermore, identifying TTN variants can inform prognosis and guide therapeutic decisions. Deciphering the intricate genotype-phenotype correlations between TTN variants and their pathologic traits in cardiomyopathies is imperative for gene-based diagnosis, risk assessment, and personalized clinical management. With the increasing use of next-generation sequencing (NGS), a high number of variants in the TTN gene have been detected in patients with cardiomyopathies. However, not all TTN variants detected in cardiomyopathy cohorts can be assumed to be disease-causing. The interpretation of TTN variants remains challenging due to high background population variation. This narrative review aimed to comprehensively summarize current evidence on TTN variants identified in published cardiomyopathy studies and determine which specific variants are likely pathogenic contributors to cardiomyopathy development.
Collapse
Affiliation(s)
- Amir Ghaffari Jolfayi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Kohansal
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Serwa Ghasemi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Naderi
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hesami
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Maryam Hosseini Moghadam
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Farjam Fazelifar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Maleki
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Kalayinia
- Cardiogenetic Research Center, Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
Garmany R, Dasari S, Bos JM, Kim ET, Martinez KA, Tester DJ, Dos Remedios C, Maleszewski JJ, Dearani JA, Ommen SR, Geske JB, Giudicessi JR, Ackerman MJ. A Multi-Omics Atlas of Sex-Specific Differences in Obstructive Hypertrophic Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.22.581621. [PMID: 38464071 PMCID: PMC10925216 DOI: 10.1101/2024.02.22.581621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Hypertrophic cardiomyopathy (HCM) is a common genetic heart disease. Women with HCM tend to have a later onset but more severe disease course. However, the underlying pathobiological mechanisms for these differences remain unknown. Methods Myectomy samples from 97 patients (53 males/44 females) with symptomatic obstructive HCM and 23 control cardiac tissues were included in this study. RNA-sequencing was performed on all samples. Mass spectrometry-based proteomics and phosphoproteomics was performed on a representative subset of samples. Results The transcriptome, proteome, and phosphoproteome was similar between sexes and did not separate on PCA plotting. Overall, there were 482 differentially expressed genes (DEGs) between control females and control males while there were only 53 DEGs between HCM females and HCM males. There were 1963 DEGs between HCM females and control females compared to 1064 DEGs between HCM males and control males. Additionally, there was increased transcriptional downregulation of hypertrophy pathways in HCM females and in HCM males. HCM females had 119 differentially expressed proteins compared to control females while HCM males only had 27 compared to control males. Finally, the phosphoproteome showed females had 341 differentially phosphorylated proteins (DPPs) compared to controls while males only had 184. Interestingly, there was hypophosphorylation and inactivation of hypertrophy pathways in females but hyperphosphorylation and activation in males. Conclusion There are subtle, but biologically relevant differences in the multi-omics profile of HCM. This study provides the most comprehensive atlas of sex-specific differences in the transcriptome, proteome, and phosphoproteome present at the time of surgical myectomy for obstructive HCM.
Collapse
|
40
|
Peng Y, Qu R, Xu S, Bi H, Guo D. Regulatory mechanism and therapeutic potentials of naringin against inflammatory disorders. Heliyon 2024; 10:e24619. [PMID: 38317884 PMCID: PMC10839891 DOI: 10.1016/j.heliyon.2024.e24619] [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: 08/04/2023] [Revised: 12/02/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024] Open
Abstract
Naringin is a natural flavonoid with therapeutic properties found in citrus fruits and an active natural product from herbal plants. Naringin has become a focus of attention in recent years because of its ability to actively participate in the body's immune response and maintain the integrity of the immune barrier. This review aims to elucidate the mechanism of action and therapeutic efficacy of naringin in various inflammatory diseases and to provide a valuable reference for further research in this field. The review provided the chemical structure, bioavailability, pharmacological properties, and pharmacokinetics of naringin and found that naringin has good therapeutic potential for inflammatory diseases, exerting anti-inflammatory, anti-apoptotic, anti-oxidative stress, anti-ulcerative and detoxifying effects in the disease. Moreover, we found that the great advantage of naringin treatment is that it is safe and can even alleviate the toxic side effects associated with some of the other drugs, which may become a highlight of naringin research. Naringin, an active natural product, plays a significant role in systemic diseases' anti-inflammatory and antioxidant regulation through various signaling pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Yuan Peng
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Ruyi Qu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Shuqin Xu
- Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, 250002, China
| |
Collapse
|
41
|
Xu FF, Xie XF, Hu HY, Tong RS, Peng C. Shenfu injection: a review of pharmacological effects on cardiovascular diseases. Front Pharmacol 2024; 15:1279584. [PMID: 38420190 PMCID: PMC10899515 DOI: 10.3389/fphar.2024.1279584] [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: 08/18/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
Shenfu injection (SFI), composed of ginseng and aconite, is a Chinese patent developed from the classic traditional prescription Shenfu Decoction created more than 700 years ago. SFI has been widely used in China for over 30 years for treating cardiovascular diseases. The main components in it include ginsenosides and aconitum alkaloids. In recent years, the role of SFI in the treatment of cardiovascular diseases has attracted much attention. The pharmacological effects and therapeutic applications of SFI in cardiovascular diseases are summarized here, highlighting pharmacological features and potential mechanisms developments, confirming that SFI can play a role in multiple ways and is a promising drug for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Fei-Fei Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao-Fang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai-Yan Hu
- Sichuan Nursing Vocational College, Chengdu, China
| | - Rong-Sheng Tong
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
42
|
Tsutsumi K, Tippayamontri T, Hayashi M, Matsuda N, Goto Y. The dynamic relationship between inorganic polyphosphate and adenosine triphosphate in human non-small cell lung cancer H1299 cells. FEBS Open Bio 2024; 14:344-354. [PMID: 38105501 PMCID: PMC10839297 DOI: 10.1002/2211-5463.13753] [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: 10/10/2023] [Revised: 11/28/2023] [Accepted: 12/15/2023] [Indexed: 12/19/2023] Open
Abstract
Inorganic polyphosphate (polyP) plays a vital role in cellular energy metabolism and signaling, owing to its structure and high-energy phosphate bonds. Intracellular ATP functions both as a cellular energy source and a key factor in cell death, and ATP dynamics in tumor cells are crucial for advancing cancer therapy. In this study, we explored the interplay between polyP and ATP in cellular energy metabolism. Treatment with polyP did not affect cell proliferation of human non-small cell lung cancer H1299 and human glioblastoma T98G cell lines as compared to their respective control cells until 72 h post-treatment. The mitochondrial membrane potential (MMP) in polyP-treated cells was low, contrasting with the time-dependent increase observed in control cells. While the ATP content increased over time in untreated and Na-phosphate-treated control cells, it remained unchanged in polyP-treated cells. Furthermore, the addition of cyclosporine A, a mitochondrial permeability transition pore (mPTP) inhibitor, failed to restore ATP levels in polyP-treated cells. We performed lactate assays and western blot analysis to evaluate the effect of polyP on glucose metabolism and found no significant differences in lactate secretion or glucose-6-phosphate dehydrogenase (G6PD) activity between polyP-treated and control cells. Additional pyruvate restored MMP but had no effect on the cellular ATP content in polyP-treated cells. We observed no correlation between the Warburg effect and glucose metabolism during ATP depletion in polyP-treated cells. Further investigation is warranted to explore the roles of polyP and ATP in cancer cell energy metabolism, which might offer potential avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Kaori Tsutsumi
- Department of Biomedical Science and EngineeringFaculty of Health SciencesHokkaido UniversitySapporoJapan
| | - Thititip Tippayamontri
- Department of Radiological Technology and Medical PhysicsFaculty of Allied Health SciencesChulalongkorn UniversityBangkokThailand
| | - Mari Hayashi
- Department of Health Sciences, School of MedicineHokkaido UniversitySapporoJapan
| | - Nobuto Matsuda
- Department of Health Sciences, School of MedicineHokkaido UniversitySapporoJapan
| | - Yusaku Goto
- Department of Health Sciences, School of MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
43
|
Gao W, Guo N, Yan H, Zhao S, Sun Y, Chen Z. Mycn ameliorates cardiac hypertrophy-induced heart failure in mice by mediating the USP2/JUP/Akt/β-catenin cascade. BMC Cardiovasc Disord 2024; 24:82. [PMID: 38297207 PMCID: PMC10829249 DOI: 10.1186/s12872-024-03748-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Pathological cardiac hypertrophy is associated with cardiac dysfunction and is a key risk factor for heart failure and even sudden death. This study investigates the function of Mycn in cardiac hypertrophy and explores the interacting molecules. METHODS A mouse model of cardiac hypertrophy was induced by isoproterenol (ISO). The cardiac dysfunction was assessed by the heart weight-to-body weight ratio (HW/BW), echocardiography assessment, pathological staining, biomarker detection, and cell apoptosis. Transcriptome alteration in cardiac hypertrophy was analyzed by bioinformatics analysis. Gain- or loss-of-function studies of MYCN proto-oncogene (Mycn), ubiquitin specific peptidase 2 (USP2), and junction plakoglobin (JUP) were performed. The biological functions of Mycn were further examined in ISO-treated cardiomyocytes. The molecular interactions were verified by luciferase assay or immunoprecipitation assays. RESULTS Mycn was poorly expressed in ISO-treated mice, and its upregulation reduced HW/BW, cell surface area, oxidative stress, and inflammation while improving cardiac function of mice. It also reduced apoptosis of cardiomyocytes in mice and those in vitro induced by ISO. Mycn bound to the USP2 promoter to activate its transcription. USP2 overexpression exerted similar myocardial protective functions. It stabilized JUP protein by deubiquitination modification, which blocked the Akt/β-catenin pathway. Knockdown of JUP restored phosphorylation of Akt and β-catenin protein level, which negated the protective effects of USP2. CONCLUSION This study demonstrates that Mycn activates USP2 transcription, which mediates ubiquitination and protein stabilization of JUP, thus inactivating the Akt/β-catenin axis and alleviating cardiac hypertrophy-induced heart failure.
Collapse
Affiliation(s)
- Weinian Gao
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
- Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Na Guo
- Department of Geriatry II, TCM Hospital of Shijiazhuang city, Shijiazhuang, Hebei, 050000, P.R. China
| | - Hongjiang Yan
- Department of Thoracic surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Shuguang Zhao
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Yongquan Sun
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China
| | - Ziying Chen
- Department of Cardiac Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, P.R. China.
| |
Collapse
|
44
|
Zheng L, Shi W, Liu B, Duan B, Sorgen P. Evaluation of Tyrosine Kinase Inhibitors Loaded Injectable Hydrogels for Improving Connexin43 Gap Junction Intercellular Communication. ACS APPLIED MATERIALS & INTERFACES 2024; 16:1985-1998. [PMID: 38175743 PMCID: PMC11061860 DOI: 10.1021/acsami.3c10923] [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] [Indexed: 01/06/2024]
Abstract
Myocardial infarction (MI) is one of the leading causes of death in the developed world, and the loss of cardiomyocytes plays a critical role in the pathogenesis of heart failure. Implicated in this process is a decrease in gap junction intercellular communication due to remodeling of Connexin43 (Cx43). We previously identified that intraperitoneal injection of the Pyk2 inhibitor PF4618433 reduced infarct size, maintained Cx43 at the intercalated disc in left ventricle hypertrophic myocytes, and improved cardiac function in an MI animal model of heart failure. With the emergence of injectable hydrogels as a therapeutic toward the regeneration of cardiac tissue after MI, here, we provide proof of concept that the release of tyrosine kinase inhibitors from hydrogels could have beneficial effects on cardiomyocytes. We developed an injectable hydrogel consisting of thiolated hyaluronic acid and P123-maleimide micelles that can incorporate PF4618433 as well as the Src inhibitor Saracatinib and achieved sustained release (of note, Src activates Pyk2). Using neonatal rat ventricular myocytes in the presence of a phorbol ester, endothelin-1, or phenylephrine to stimulate cardiac hypertrophy, the release of PF4618433 from the hydrogel had the same ability to decrease Cx43 tyrosine phosphorylation and maintain Cx43 localization at the plasma membrane as when directly added to the growth media. Additional beneficial effects included decreases in apoptosis, the hypertrophic marker atrial natriuretic peptide (ANP), and serine kinases upregulated in hypertrophy. Finally, the presence of both PF4618433 and Saracatinib further decreased the level of ANP and apoptosis than each inhibitor alone, suggesting that a combinatorial approach may be most beneficial. These findings provide the groundwork to test if tyrosine kinase inhibitor release from hydrogels will have a beneficial effect in an animal model of MI-induced heart failure.
Collapse
Affiliation(s)
- Li Zheng
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bo Liu
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Paul Sorgen
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
45
|
Luo G, Chen L, Chen M, Mao L, Zeng Q, Zou Y, Xue J, Liu P, Wu Q, Yang S, Liu M. Hirudin inhibit the formation of NLRP3 inflammasome in cardiomyocytes via suppressing oxidative stress and activating mitophagy. Heliyon 2024; 10:e23077. [PMID: 38163129 PMCID: PMC10754874 DOI: 10.1016/j.heliyon.2023.e23077] [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: 05/23/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Context Cardiomyocyte hypertrophy due to hemodynamic overload eventually leads to heart failure. Hirudin has been widely used in the treatment of cardiovascular diseases and NLRP3 inflammasome was proven to induce cardiomyocyte pyroptosis. However, the mechanism by which it inhibits cardiomyocyte hypertrophy remains unclear. Objective To explore the mechanism of hirudin inhibiting cardiomyocyte hypertrophy based on NLRP3 inflammasome activation and mitophagy. Materials & methods 1 μM AngII was used for cardiac hypertrophy modeling in H9C2 cells, and cell viability was quantified by CCK-8 assay to screen the appropriate action concentrations of hirudin. After that, we cultured AngII induced-H9C2 cells for 24 h with 0, 0.3, 0.6, and 1.2 mM hirudin, respectively. Next, we marked H9C2 cells with phalloidine and observed them using fluorescence microscope. IL-1β, IL-18, IL-6, TNF-α, ANP, BNP, β-MHC, and mtDNA were analyzed by qRT-PCR; ROS were quantified by Flow cytometry; SOD, MDA, and GSH-Px were detected by ELISA; and proteins including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18, PINK-1, Parkin, beclin-1, LC3-Ⅰ, LC3-Ⅱ, p62, were quantified by western blotting. Results It was discovered that hirudin reduced the superficial area of AngII-induced H9C2 cells and inhibited the AngII-induced up-regulation of ANP, BNP, and β-MHC. Besides, hirudin down-regulated the expressions of NLRP3 inflammasome-related cytokines, containing IL-1β, IL-18, IL-6, TNF-α. It also down-regulated the expression of mtDNA and ROS, decreased the expression levels of NLRP3 inflammasome activation related proteins, including NLRP3, ASC, caspase-1, pro-caspase-1, IL-1β, IL-18; and increased the expressions of PINK-1, Parkin, beclin-1, LC3-Ⅱ/LC3-Ⅰ, p62 in AngII-induced H9C2 cells. Discussion Hirudin promoted the process of mitophagy, inhibited the development of inflammation and oxidative stress, and inhibited the activation of the NLRP3 inflammasome and the PINK-1/Parkin pathway. Conclusion Hirudin has the activity to suppress cardiac hypertrophy may benefit from the inhibition of NLRP3 inflammasome and activating of PINK-1/Parkin related-mitophagy.
Collapse
Affiliation(s)
- Gang Luo
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Li Chen
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Mingtai Chen
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linshen Mao
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qihu Zeng
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Yuan Zou
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinyi Xue
- School of Integrated Traditional and Western Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Ping Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Qibiao Wu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Sijin Yang
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| | - Mengnan Liu
- National Traditional Chinese Medicine Clinical Research Base and Department of Cardiovascular Medicine, the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, PR China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Tapai, Macau
| |
Collapse
|
46
|
Wei M, Lu Z, Zhang H, Fan X, Zhang X, Jiang B, Li J, Xue M. Aspirin and Celecoxib Regulate Notch1/Hes1 Pathway to Prevent Pressure Overload-Induced Myocardial Hypertrophy. Int Heart J 2024; 65:475-486. [PMID: 38825493 DOI: 10.1536/ihj.23-614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
This study aimed to investigate the molecular mechanisms underlying the protective effects of cyclooxygenase (cox) inhibitors against myocardial hypertrophy.Rat H9c2 cardiomyocytes were induced by mechanical stretching. SD rats underwent transverse aortic constriction to induce pressure overload myocardial hypertrophy. Rats were subjected to echocardiography and tail arterial pressure in 12W. qPCR and western blot were used to detect the expression of Notch-related signaling. The inflammatory factors were tested by ELISA in serum, heart tissue, and cell culture supernatant.Compared with control, levels of pro-inflammatory cytokines IL-6, TNF-α, and IL-1β were increased and anti-inflammatory cytokine IL-10 was reduced in myocardial tissues and serum of rat models. Levels of Notch1 and Hes1 were reduced in myocardial tissues. However, cox inhibitor treatment (aspirin and celecoxib), the improvement of exacerbated myocardial hypertrophy, fibrosis, dysfunction, and inflammation was parallel to the activation of Notch1/Hes1 pathway. Moreover, in vitro experiments showed that, in cardiomyocyte H9c2 cells, application of ~20% mechanical stretching activated inflammatory mediators (IL-6, TNF-α, and IL-1β) and hypertrophic markers (ANP and BNP). Moreover, expression levels of Notch1 and Hes1 were decreased. These changes were effectively alleviated by aspirin and celecoxib.Cox inhibitors may protect heart from hypertrophy and inflammation possibly via the Notch1/Hes1 signaling pathway.
Collapse
Affiliation(s)
- Minghui Wei
- School of Basic Medicine, Inner Mongolia Medical University
| | - Ziyu Lu
- School of Basic Medicine, Inner Mongolia Medical University
| | - Haifeng Zhang
- Office of Academic Affairs, Inner Mongolia Medical University
| | - Xiaomei Fan
- Department of Physiology, Inner Mongolia Medical University
| | - Xin Zhang
- Department of Physiology, Inner Mongolia Medical University
| | - Bihui Jiang
- School of Basic Medicine, Inner Mongolia Medical University
| | - Jianying Li
- School of Basic Medicine, Inner Mongolia Medical University
| | - Mingming Xue
- Office of Academic Affairs, Inner Mongolia Medical University
| |
Collapse
|
47
|
Gupta P, Ekbbal R. Liraglutide Improves Diabetic Cardiomyopathy by Downregulation of Cardiac Inflammatory and Apoptosis Markers. Curr Drug Res Rev 2024; 16:289-299. [PMID: 37966282 DOI: 10.2174/0125899775243787231103075804] [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: 01/31/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023]
Abstract
BACKGROUND Diabetic cardiomyopathy is one of the leading causes of mortality for people with diabetes worldwide. The majority of the formalistic alterations in the heart associated with diabetic cardiomyopathy have been found to be primarily caused by the ongoing oxidative stress brought on by hyperglycemia, which leads to the dysfunctional reactions of apoptosis and inflammation. Liraglutide, a long-acting counterpart of glucagon-like peptide-1, has been demonstrated to have a number of therapeutic applications in medicine and other biological processes. METHODS The PubMed database was searched using the terms liraglutide, DCM, and all associated inflammatory markers. RESULTS There has been a lot of research on liraglutide's potential to protect the heart from cardiomyopathy brought on by diabetes. Liraglutide's therapeutic actions as an antioxidant, antihyperglycemic, anti-apoptotic, and anti-inflammatory medicine may help to lessen diabetic cardiomyopathy. CONCLUSION The most recent studies on the effects of liraglutide therapy on DCM are presented in this review, along with an explanation of the underlying mechanisms.
Collapse
Affiliation(s)
- Polly Gupta
- Department of Pharmaceutical Sciences, IIMT College of Medical Sciences (Pharmacy), IIMT University, Meerut, UP, India
| | - Rustam Ekbbal
- Department of Pharmacology, IIMT College of Medical Sciences (Pharmacy), IIMT University, Meerut, UP, India
| |
Collapse
|
48
|
Avagimyan A, Fogacci F, Pogosova N, Kakrurskiy L, Kogan E, Urazova O, Kobalava Z, Mikhaleva L, Vandysheva R, Zarina G, Trofimenko A, Navasardyan G, Mkrtchyan L, Galli M, Jndoyan Z, Aznauryan A, Saahakyan K, Agati L, Shafie D, Cicero A, Salvo GD, Sarrafzadegan N. Diabetic Cardiomyopathy: 2023 Update by the International Multidisciplinary Board of Experts. Curr Probl Cardiol 2024; 49:102052. [PMID: 37640176 DOI: 10.1016/j.cpcardiol.2023.102052] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 08/31/2023]
Abstract
Diabetes mellitus (DM) is considered by many the pandemic of the 21st century and is associated with multiple organ damages. Among these, cardiovascular complications are responsible for an incredible burden of mortality and morbidity in Western Countries. The study of the pathological mechanisms responsible for the cardiovascular complications in DM patients is key for the development of new therapeutic strategies. The metabolic disorders caused by hyperglycemia, insulin resistance, and dyslipidemia, results in a cascade of pathomorphological changes favoring the atherosclerotic process and leading to myocardial remodeling. Parallel to this, oxidative stress, calcium overload, mitochondrial dysfunction, activation of protein kinase C signaling pathways, myocardial lipomatosis, and low-grade inflammation of the myocardium - are the main pathways responsible for the diabetic cardiomyopathy development. This review aims to appraise and discuss the pathogenetic mechanisms behind the diabetic cardiomyopathy development.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University, Yerevan, Armenia.
| | - Federica Fogacci
- Atherosclerosis and Metabolic Disorders Unit, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Nana Pogosova
- Research and Preventive Cardiology, National Medical Research Centre of Cardiology, Moscow, Russia
| | - Lev Kakrurskiy
- A.P. Avtsyn Research Institute of Human Morphology FSBI "Petrovskiy NRCS" Moscow, Russia
| | - Eugenia Kogan
- Pathology Department, Immunohistochemistry Reference Centre of Institute of Clinical Morphology and Digital Pathology, I. M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Olga Urazova
- Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Zhanna Kobalava
- Internal Disease and Cardiology Department, Peoples Friendship University of Russia, Moscow, Russia
| | - Liudmila Mikhaleva
- A.P. Avtsyn Research Institute of Human Morphology FSBI "Petrovskiy NRCS" Moscow, Russia
| | - Rositsa Vandysheva
- A.P. Avtsyn Research Institute of Human Morphology FSBI "Petrovskiy NRCS" Moscow, Russia
| | - Gioeva Zarina
- A.P. Avtsyn Research Institute of Human Morphology FSBI "Petrovskiy NRCS" Moscow, Russia
| | - Artem Trofimenko
- Pathophysiology Department, Kuban State Medical University, Krasnodar, Russia
| | | | - Lusine Mkrtchyan
- Cardiology Department, Yerevan State Medical University, Yerevan, Armenia
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Zinaida Jndoyan
- Internal Diseases Propaedeutic Department, Yerevan State Medical University, Yerevan, Armenia
| | - Anait Aznauryan
- Histology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Karmen Saahakyan
- Cardiology Department, Azienda Umberto I, Sapienza University, Rome, Italy
| | - Luciano Agati
- Cardiology Department, Azienda Umberto I, Sapienza University, Rome, Italy
| | - Davood Shafie
- Heart Failure Research Center, Isfahan Cardiovascular Research Institute, Isfahan, Iran
| | - Arrigo Cicero
- Atherosclerosis and Metabolic Disorders Unit, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | | | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
49
|
Li Y, Bertozzi A, Mann MRW, Kühn B. Interdependent changes of nuclear lamins, nuclear pore complexes, and ploidy regulate cellular regeneration and stress response in the heart. Nucleus 2023; 14:2246310. [PMID: 37606283 PMCID: PMC10446781 DOI: 10.1080/19491034.2023.2246310] [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: 12/20/2022] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/23/2023] Open
Abstract
In adult mammals, many heart muscle cells (cardiomyocytes) are polyploid, do not proliferate (post-mitotic), and, consequently, cannot contribute to heart regeneration. In contrast, fetal and neonatal heart muscle cells are diploid, proliferate, and contribute to heart regeneration. We have identified interdependent changes of the nuclear lamina, nuclear pore complexes, and DNA-content (ploidy) in heart muscle cell maturation. These results offer new perspectives on how cells alter their nuclear transport and, with that, their gene regulation in response to extracellular signals. We present how changes of the nuclear lamina alter nuclear pore complexes in heart muscle cells. The consequences of these changes for cellular regeneration and stress response in the heart are discussed.
Collapse
Affiliation(s)
- Yao Li
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alberto Bertozzi
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mellissa RW Mann
- Department of Obstetrics, Gynaecology and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Magee-Womens Research Institute, Pittsburgh, PA, USA
| | - Bernhard Kühn
- Division of Pediatric Cardiology, Pediatric Institute for Heart Regeneration and Therapeutics (I-HRT), UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
50
|
Wang X, Yu Q, Liao X, Fan M, Liu X, Liu Q, Wang M, Wu X, Huang CK, Tan R, Yuan J. Mitochondrial Dysfunction in Arrhythmia and Cardiac Hypertrophy. Rev Cardiovasc Med 2023; 24:364. [PMID: 39077079 PMCID: PMC11272842 DOI: 10.31083/j.rcm2412364] [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: 05/27/2023] [Revised: 08/18/2023] [Accepted: 09/04/2023] [Indexed: 07/31/2024] Open
Abstract
Arrhythmia and cardiac hypertrophy are two very common cardiovascular diseases that can lead to heart failure and even sudden death, thus presenting a serious threat to human life and health. According to global statistics, nearly one million people per year die from arrhythmia, cardiac hypertrophy and other associated cardiovascular diseases. Hence, there is an urgent need to find new treatment targets and to develop new intervention measures. Recently, mitochondrial dysfunction has been examined in relation to heart disease with a view to lowering the incidence of arrhythmia and cardiac hypertrophy. The heart is the body's largest energy consuming organ, turning over about 20 kg of adenosine triphosphate (ATP) per day in the mitochondria. Mitochondrial oxidative phosphorylation (OXPHOS) produces up to 90% of the ATP needed by cardiac muscle cells for contraction and relaxation. Dysfunction of heart mitochondria can therefore induce arrhythmia, cardiac hypertrophy and other cardiovascular diseases. Mitochondrial DNA (mtDNA) mutations cause disorders in OXPHOS and defects in the synthesis of muscle contraction proteins. These lead to insufficient production of secondary ATP, increased metabolic requirements for ATP by the myocardium, and the accumulation of reactive oxygen species (ROS). The resulting damage to myocardial cells eventually induces arrhythmia and cardiac hypertrophy. Mitochondrial damage decreases the efficiency of energy production, which further increases the production of ROS. The accumulation of ROS causes mitochondrial damage and eventually leads to a vicious cycle of mitochondrial damage and low efficiency of mitochondrial energy production. In this review, the mechanism underlying the development of arrhythmia and cardiac hypertrophy is described in relation to mitochondrial energy supply, oxidative stress, mtDNA mutation and Mitochondrial dynamics. Targeted therapy for arrhythmia and cardiac hypertrophy induced by mitochondrial dysfunction is also discussed in terms of its potential clinical value. These strategies should improve our understanding of mitochondrial biology and the pathogenesis of arrhythmia and cardiac hypertrophy. They may also identify novel strategies for targeting mitochondria in the treatment of these diseases.
Collapse
Affiliation(s)
- Xiaomei Wang
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
| | - Qianxue Yu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Xuemei Liao
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Mengying Fan
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Xibin Liu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Qian Liu
- College of Basic Medical, Jining Medical University, 272067 Jining,
Shandong, China
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
| | - Manru Wang
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Xinyu Wu
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- College of Second Clinical Medicine, Jining Medical University, 272067
Jining, Shandong, China
| | - Chun-Kai Huang
- Department of Cardiology, Ruijin Hospital, Shanghai Jiao Tong University,
School of Medicine, 200025 Shanghai, China
| | - Rubin Tan
- College of Basic Medical, Xuzhou Medical University, 221004 Xuzhou,
Jiangsu, China
| | - Jinxiang Yuan
- Collaborative Innovation Center for Birth Defect Research and
Transformation of Shandong Province, Jining Medical University, 272067 Jining,
Shandong, China
- Lin He's Academician Workstation of New Medicine and Clinical Translation,
Jining Medical University, 272067 Jining, Shandong, China
| |
Collapse
|