1
|
Fan S, Zhao M, Wang K, Deng Y, Yu X, Ma K, Zhang Y, Xiao H. Exercise training attenuates cardiac dysfunction induced by excessive sympathetic activation through an AMPK-KLF4-FMO2 axis. J Mol Cell Cardiol 2024; 197:136-149. [PMID: 39491669 DOI: 10.1016/j.yjmcc.2024.10.015] [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: 05/16/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Cardiovascular diseases (CVDs) are a leading cause of mortality worldwide and are associated with an overactivated sympathetic system. Although exercise training has shown promise in mitigating sympathetic stress-induced cardiac remodeling, the precise mechanisms remain elusive. Here, we demonstrate that exercise significantly upregulates cardiac flavin-containing monooxygenase 2 (FMO2) expression. Notably, we find that exercise training effectively counteracts sympathetic overactivation-induced cardiac dysfunction and fibrosis by enhancing FMO2 expression via adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activation. Functional investigations employing FMO2 knockdown with adeno-associated virus 9 (AAV9) underscore the necessity for FMO2 expression to protect the heart during exercise in vivo. Furthermore, we identify the krüppel-like factor 4 (KLF4) as a transcriptional mediator of FMO2 that is crucial for the mechanism through which AMPK activation protects against sympathetic overactivation-induced cardiac dysfunction and fibrosis. Taken together, our study reveals a cardioprotective mechanism for exercise training through an AMPK-KLF4-FMO2 signaling pathway that underscores how exercise alleviates cardiac dysfunction induced by excessive sympathetic activation.
Collapse
Affiliation(s)
- Shiyu Fan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
| | - Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China
| | - Kang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China
| | - Yawen Deng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China
| | - Xiaoyue Yu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China
| | - Ketao Ma
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China.
| | - Han Xiao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Ministry of Education, Shihezi University School of Medicine, Shihezi 832003, China; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Institute of Advanced Clinical Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing 100191, China.
| |
Collapse
|
2
|
Yang Y, Jiang S, Mu Y, Liu C, Han Y, Jiang J, Wang Y. Berberine prevents against myocardial injury induced by acute β-adrenergic overactivation in rats. J Appl Toxicol 2024; 44:1700-1713. [PMID: 38981847 DOI: 10.1002/jat.4659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/23/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024]
Abstract
The overactivation of β-adrenergic receptors (β-ARs) can result in acute myocardial ischemic injury, culminating in myocardial necrosis. Berberine (BBR) has exhibited promising potential for prevention and treatment in various heart diseases. However, its specific role in mitigating myocardial injury induced by acute β-AR overactivation remains unexplored. This study aimed to investigate the effects and underlying mechanisms of BBR pretreatment in a rat model of acute β-AR overactivation induced by a single dose of the nonselective β-adrenergic agonist isoprenaline (ISO). Rats were pretreated with saline or BBR (100 mg/kg/day) via gavage for 14 consecutive days, followed by a subcutaneous injection of ISO or saline on the 14th day. The findings indicated that BBR pretreatment significantly attenuated myocardial injury in ISO-stimulated rats, as evidenced by reduced pathological inflammatory infiltration, necrosis, and serum markers of myocardial damage. Additionally, BBR decreased oxidative stress and inflammation in the system and heart. Furthermore, BBR pretreatment enhanced myocardial ATP levels, improved mitochondrial dysfunction through increased Drp1 phosphorylation, and augmented myocardial autophagy. In a CoCl2-induced H9c2 cell hypoxic injury model, BBR pretreatment mitigated cellular injury, apoptosis, and oxidative stress while upregulating Drp1 and autophagy-associated proteins. Mechanistically, BBR pretreatment activated AKT, AMPK, and LKB1 both in vivo and in vitro, implicating the involvement of the AKT and LKB1/AMPK signaling pathways in its cardioprotective effects. Our study demonstrated the protective effects of BBR against myocardial injury induced by acute β-AR overactivation in rats, highlighting the potential of BBR as a preventive agent for myocardial injury associated with β-adrenergic overactivation.
Collapse
Affiliation(s)
- Yalin Yang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuang Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Mu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chilu Liu
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuhong Wang
- State Key Laboratory of Bioactive Substances and Function of Natural Medicine, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Zhao M, Cao N, Gu H, Xu J, Xu W, Zhang D, Wei TYW, Wang K, Guo R, Cui H, Wang X, Guo X, Li Z, He K, Li Z, Zhang Y, Shyy JYJ, Dong E, Xiao H. AMPK Attenuation of β-Adrenergic Receptor-Induced Cardiac Injury via Phosphorylation of β-Arrestin-1-ser330. Circ Res 2024; 135:651-667. [PMID: 39082138 DOI: 10.1161/circresaha.124.324762] [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: 05/21/2024] [Revised: 07/16/2024] [Accepted: 07/22/2024] [Indexed: 09/01/2024]
Abstract
BACKGROUND β-adrenergic receptor (β-AR) overactivation is a major pathological cue associated with cardiac injury and diseases. AMPK (AMP-activated protein kinase), a conserved energy sensor, regulates energy metabolism and is cardioprotective. However, whether AMPK exerts cardioprotective effects via regulating the signaling pathway downstream of β-AR remains unclear. METHODS Using immunoprecipitation, mass spectrometry, site-specific mutation, in vitro kinase assay, and in vivo animal studies, we determined whether AMPK phosphorylates β-arrestin-1 at serine (Ser) 330. Wild-type mice and mice with site-specific mutagenesis (S330A knock-in [KI]/S330D KI) were subcutaneously injected with the β-AR agonist isoproterenol (5 mg/kg) to evaluate the causality between β-adrenergic insult and β-arrestin-1 Ser330 phosphorylation. Cardiac transcriptomics was used to identify changes in gene expression from β-arrestin-1-S330A/S330D mutation and β-adrenergic insult. RESULTS Metformin could decrease cAMP/PKA (protein kinase A) signaling induced by isoproterenol. AMPK bound to β-arrestin-1 and phosphorylated Ser330 with the highest phosphorylated mass spectrometry score. AMPK activation promoted β-arrestin-1 Ser330 phosphorylation in vitro and in vivo. Neonatal mouse cardiomyocytes overexpressing β-arrestin-1-S330D (active form) inhibited the β-AR/cAMP/PKA axis by increasing PDE (phosphodiesterase) 4 expression and activity. Cardiac transcriptomics revealed that the differentially expressed genes between isoproterenol-treated S330A KI and S330D KI mice were mainly involved in immune processes and inflammatory response. β-arrestin-1 Ser330 phosphorylation inhibited isoproterenol-induced reactive oxygen species production and NLRP3 (NOD-like receptor protein 3) inflammasome activation in neonatal mouse cardiomyocytes. In S330D KI mice, the β-AR-activated cAMP/PKA pathways were attenuated, leading to repressed inflammasome activation, reduced expression of proinflammatory cytokines, and mitigated macrophage infiltration. Compared with S330A KI mice, S330D KI mice showed diminished cardiac fibrosis and improved cardiac function upon isoproterenol exposure. However, the cardiac protection exerted by AMPK was abolished in S330A KI mice. CONCLUSIONS AMPK phosphorylation of β-arrestin-1 Ser330 potentiated PDE4 expression and activity, thereby inhibiting β-AR/cAMP/PKA activation. Subsequently, β-arrestin-1 Ser330 phosphorylation blocks β-AR-induced cardiac inflammasome activation and remodeling.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ning Cao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital (N.C.), Capital Medical University, Beijing, China
| | - Huijun Gu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Jiachao Xu
- Laboratory for Clinical Medicine (N.C.), Capital Medical University, Beijing, China
| | - Wenli Xu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Di Zhang
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Tong-You Wade Wei
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Kang Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Ruiping Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Hongtu Cui
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Xiaofeng Wang
- Laboratory of Inflammation and Vaccines, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (X.W.)
| | - Xin Guo
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Zhiyuan Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies (D.Z., Zhiyuan Li), Peking University, Beijing, China
| | - Kangmin He
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China (J.X., K.H.)
- University of Chinese Academy of Sciences, Beijing, China (K.H.)
| | - Zijian Li
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla (T.-Y.W.W., J.Y.-J.S.)
| | - Erdan Dong
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- Institute of Cardiovascular Sciences (E.D.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- State Key Laboratory of Vascular Homeostasis and Remodeling, Institute of Advanced Clinical Medicine (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.), Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Beijing Key Laboratory of Cardiovascular Receptors Research, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Haihe Laboratory of Cell Ecosystem, Beijing, China (M.Z., N.C., H.G., W.X., K.W., R.G., H.C., X.G., Zijian Li, Y.Z., E.D., H.X.)
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China (W.X., E.D., H.X.)
| |
Collapse
|
4
|
Tallapalli PS, Reddy YD, Yaraguppi DA, Matangi SP, Challa RR, Vallamkonda B, Ahmad SF, Al-Mazroua HA, Rudrapal M, Dintakurthi Sree Naga Bala Krishna P, Pasala PK. In Silico and In Vivo Studies of β-Sitosterol Nanoparticles as a Potential Therapy for Isoprenaline-Induced Cognitive Impairment in Myocardial Infarction, Targeting Myeloperoxidase. Pharmaceuticals (Basel) 2024; 17:1093. [PMID: 39204198 PMCID: PMC11359034 DOI: 10.3390/ph17081093] [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: 07/22/2024] [Revised: 08/03/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
OBJECTIVE This study aimed to compare the effects of β-sitosterol nanoparticles (BETNs) and β-sitosterol (BET) on cognitive impairment, oxidative stress, and inflammation in a myocardial infarction (MI) rat model using in silico and in vivo methods. METHODS β-Sitosterol (BET) and myeloperoxidase (MPO) ligand-receptor binding affinities were evaluated using Autodock Vina for docking and Gromacs for dynamics simulations. BET nanoparticles, prepared via solvent evaporation, had their size confirmed by a nanoparticle analyzer. ISO-induced cognitive impairment in rats was assessed through Morris water maze and Cook's pole climbing tests. Oxidative stress, inflammation, and cardiac injury were evaluated by measuring GSH, SOD, MDA, MPO, CkMB, LDH, lipid profiles, and ECGs. Histopathology of the CA1 hippocampus and myocardial tissue was performed using H&E staining. RESULTS In silico analyses revealed strong binding affinities between BET and MPO, suggesting BET's potential anti-inflammatory effect. BETN (119.6 ± 42.6 nm; PDI: 0.809) significantly improved MI-induced cognitive dysfunction in rats (p < 0.001 ***), increased hippocampal GSH (p < 0.01 **) and SOD (p < 0.01 **) levels, and decreased hippocampal MDA (p < 0.05 *) and MPO levels (p < 0.01 **). BETNs also elevated cardiac GSH (p < 0.01 **) and SOD (p < 0.01 **) levels and reduced cardiac MPO (p < 0.01 **), CkMB (p < 0.001 **) and LDH (p < 0.001 **) levels. It restored lipid profiles, normalized ECG patterns, and improved histology in the hippocampal CA1 region and myocardium. CONCLUSIONS Compared with BET treatment, BETNs were more effective in improving cognitive impairment, oxidative damage, and inflammation in MI rats, suggesting its potential in treating cognitive dysfunction and associated pathological changes in MI.
Collapse
Affiliation(s)
- Partha Saradhi Tallapalli
- Department of Pharmacology, Santhiram College of Pharmacy, JNTUA, Nandyal 518112, Andhra Pradesh, India; (P.S.T.); (Y.D.R.)
| | - Yennam Dastagiri Reddy
- Department of Pharmacology, Santhiram College of Pharmacy, JNTUA, Nandyal 518112, Andhra Pradesh, India; (P.S.T.); (Y.D.R.)
| | - Deepak A. Yaraguppi
- Department of Biotechnology, KLE Technological University, Hubli 580020, Karnataka, India;
| | - Surya Prabha Matangi
- Department of Pharmaceutics, School of Biotechnology and Pharmaceutical Sciences, Vignan’s Foundation for Science, Technology & Research, Guntur 522201, Andhra Pradesh, India; (S.P.M.); (M.R.)
| | - Ranadheer Reddy Challa
- Department of Formulation and Development, Quotient Sciences, 3080 McCann Farm Dr, Garnet Valley, PA 19060, USA;
| | - Bhaskar Vallamkonda
- Department of Pharmaceutical Analysis, Odin Pharmaceutical LLC, Somerset, NJ 08873, USA;
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (H.A.A.-M.)
| | - Haneen A. Al-Mazroua
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia (H.A.A.-M.)
| | - Mithun Rudrapal
- Department of Pharmaceutics, School of Biotechnology and Pharmaceutical Sciences, Vignan’s Foundation for Science, Technology & Research, Guntur 522201, Andhra Pradesh, India; (S.P.M.); (M.R.)
| | | | - Praveen Kumar Pasala
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, JNTUA, Anantapuramu 515721, Andhra Pradesh, India
| |
Collapse
|
5
|
Wang L, Wan W, Zhang S, Keswani T, Li G, Xiao J. RNA-mediated epigenetic regulation in exercised heart: Mechanisms and opportunities for intervention. Mol Aspects Med 2024; 97:101274. [PMID: 38653129 DOI: 10.1016/j.mam.2024.101274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Physical exercise has been widely acknowledged as a beneficial lifestyle alteration and a potent non-pharmacological treatment for heart disease. Extensive investigations have revealed the beneficial effects of exercise on the heart and the underlying mechanisms involved. Exercise is considered one of the key factors that can lead to epigenetic alterations. The increasing number of identified molecules in the exercised heart has led to many studies in recent years that have explored the cellular function of ncRNAs and RNA modifications in the heart. Investigating the regulatory role of RNA-mediated epigenetic regulation in exercised hearts will contribute to the development of therapeutic strategies for the management of heart diseases. This review aims to summarize the positive impact of exercise on cardiac health. We will first provide an overview of the mechanisms through which exercise offers protection to the heart. Subsequently, we will delve into the current understanding of ncRNAs, specifically miRNAs, lncRNAs, and circRNAs, as well as RNA modification, focusing on RNA m6A and RNA A-to-I editing, and how they contribute to exercise-induced benefits for the heart. Lastly, we will explore the emerging therapeutic strategies that utilize exercise-mediated RNA epigenetic regulation in the treatment of heart diseases, while also addressing the challenges faced in this field.
Collapse
Affiliation(s)
- Lijun Wang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Wensi Wan
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Shuang Zhang
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Tarun Keswani
- Center for Immunological and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02129, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, 200444, China; Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
6
|
Peng Y, Qin D, Wang Y, Gao W, Xu X. Pharmacological inhibition of ICOS attenuates the protective effect of exercise on cardiac fibrosis induced by isoproterenol. Eur J Pharmacol 2024; 965:176327. [PMID: 38224847 DOI: 10.1016/j.ejphar.2024.176327] [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: 08/27/2023] [Revised: 12/14/2023] [Accepted: 01/09/2024] [Indexed: 01/17/2024]
Abstract
AIMS To investigate the cardioprotective mechanism of exercise or exercise combined with inducible costimulatory molecules (ICOS) monoclonal antibody (mAb) therapy against isoproterenol (ISO)-induced cardiac remodeling. MAIN METHODS Totally 24 male C57BL/6J mice were randomly divided into four groups: the control group (normal saline treatment), ISO group (subcutaneous injection of isoproterenol, 10 mg/kg/day, once daily for 5 consecutive days), the exercise with subcutaneous ISO injection group (EPI), and the exercise with injected with ISO and ICOS mAb group (EPII). The mice in EPI and EPII group were trained on a small animal treadmill for 4 weeks (13 m/min, 0% grade, 60min/day). KEY FINDINGS Exercise significantly attenuated CD45+, Mac-2 inflammatory cell infiltration, cardiac fibrosis and inhibited the RIPK1/RIPK3/MLKL/CaMKII and cardiomyocyte pyroptosis pathways to counter ISO-induced severe cardiac injury. The administration of the ICOS mAb may inhibit the cardioprotection of exercise against ISO-induced heart damage. Compared to those in EPI, our data showed that the increasing levels of myocardial fibrosis, the leukocyte infiltration of cardiac tissue and proteins expression of cardiac myocyte necrosis and pyroptosis signaling pathways in the EPII group. SIGNIFICANCE Our results demonstrated that exercise decreased leukocyte infiltration in heart, inhibited the cardiomyocyte pyroptosis and necroptosis signaling pathways, and attenuated inflammatory responses to alleviate ISO-induced cardiac fibrosis. However, the antifibrotic effects of combined treatment with exercise and ICOS mAb intervention did not exhibit synergistic enhancement.
Collapse
Affiliation(s)
- Yong Peng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China; Jiangsu Collaborative Innovation Center for Sports and Health Project, Nanjing Sport Institute, Nanjing, Jiangsu, China; Key Laboratory of Exercise Training and Rehabilitation for Jiangsu Province, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Di Qin
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Yudi Wang
- School of Physical Education and Nursing, Chengdu College of Arts and Sciences, Chengdu, Sichuan, China
| | - Wenyue Gao
- School of Sport and Health, Nanjing Sport Institute, Nanjing, Jiangsu, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China.
| |
Collapse
|
7
|
Dang W, Cao N, Zhang Y, Li W, Li H. Association among β2-adrenergic receptor autoantibodies and proximal left anterior descending artery lesions in patients with initial ST-segment elevation myocardial infarction. Clin Cardiol 2023; 46:1371-1379. [PMID: 37587904 PMCID: PMC10642316 DOI: 10.1002/clc.24129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND β2 -adrenergic receptor autoantibody (β2 -AA) are widely present in patients with many different types of cardiovascular diseases. Proximal left anterior descending (LAD) artery lesions are associated with adverse prognostic events in patients with ST-segment elevation myocardial infarction (STEMI). HYPOTHESIS β2 -AA is associated with the presence of proximal LAD lesions in patients with STEMI. METHODS A cohort of 153 patients with STEMI who underwent primary percutaneous coronary intervention (PPCI) was enrolled in the study. Baseline characteristics were compared between the proximal LAD group (n = 62) and the nonproximal LAD group (n = 91). Admission serum of patients was collected to detect the level of β2 -AA. Data for echocardiogram within 24 hours after PPCI and at the 6-month follow-up were recorded. RESULTS The optical density values and positive rates of β2 -AA in the proximal LAD group were higher than those in the nonproximal LAD group (p < 0.05). β2 -AA positively correlated with high sensitivity C-reactive protein and peak N-terminal pro-B type natriuretic peptide levels in the proximal LAD group, but those were not relevant in the nonproximal LAD group. Multivariate logistic regression analysis revealed that high β2 -AA levels was independently associated with the presence of proximal LAD lesions in patients with STEMI. Furthermore, a receiver operating characteristic curve was used to show the efficiency of β2 -AA levels to detect proximal LAD lesions, and the AUC of the β2-AA OD value was 0.658 (95% confidence interval 0.568-0.749; p = 0.001). CONCLUSIONS The STEMI patients with high β2 -AA levels had a greater possibility having proximal LAD lesions.
Collapse
Affiliation(s)
- Wenxi Dang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
| | - Ning Cao
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| | - Yue Zhang
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Weiping Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular DiseaseBeijingChina
- Laboratory of Clinical MedicineCapital Medical UniversityBeijingChina
| |
Collapse
|
8
|
Yusifov A, Borders MO, DeHoff MA, Polson SM, Schmitt EE, Bruns DR. Juvenile physical activity protects against isoproterenol-induced cardiac dysfunction later in life. J Appl Physiol (1985) 2023; 135:572-583. [PMID: 37439235 PMCID: PMC10538985 DOI: 10.1152/japplphysiol.00010.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023] Open
Abstract
Cardiovascular disease is an enormous public health problem, particularly in older populations. Exercise is the most potent cardioprotective intervention identified to date, with exercise in the juvenile period potentially imparting greater protection, given the plasticity of the developing heart. To test the hypothesis that voluntary wheel running early in life would be cardioprotective later in life when risk for disease is high, we provided male and female juvenile (3 wk old) mice access to a running wheel for 2 wk. Mice then returned to a home cage to age to adulthood (4-6 mo) before exposure to isoproterenol (ISO) to induce cardiac stress. Cardiac function and remodeling were compared with sedentary control mice, sedentary mice exposed to ISO, and mice that exercised in adulthood immediately before ISO. Early in life activity protected against ISO-induced stress as evidenced by attenuated cardiac mass, myocyte size, and fibrosis compared with sedentary mice exposed to ISO. ISO-induced changes in cardiac function were ameliorated in male mice that engaged in wheel running, with ejection fraction and fractional shortening reversed to control values. Adrenergic receptor expression was downregulated in juvenile male runners. This suppression persisted in adulthood following ISO, providing a putative mechanism by which exercise in the young male heart provides resilience to cardiac stress later in life. Together, we show that activity early in life induces persistent cardiac changes that attenuate ISO-induced stress in adulthood. Identification of the mechanisms by which early in life exercise is protective will yield valuable insights into how exercise is medicine across the life course.NEW & NOTEWORTHY Voluntary wheel running activity early in life induces persistent changes in the heart that attenuate isoproterenol-induced hypertrophy and fibrosis in adulthood. Though the mechanisms of this protection remain incompletely understood, activity-induced downregulation of adrenergic receptor expression early in life may contribute to later protection against adrenergic stress. Together these data suggest that efforts to maintain an active lifestyle early in life may have long-lasting cardioprotective benefits.
Collapse
Affiliation(s)
- Aykhan Yusifov
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Megan O Borders
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - MacKenzie A DeHoff
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Sydney M Polson
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
| | - Emily E Schmitt
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
- WWAMI Medical Education Program, University of Wyoming, Laramie, Wyoming, United States
| | - Danielle R Bruns
- Division of Kinesiology and Health, University of Wyoming, Laramie, Wyoming, United States
- WWAMI Medical Education Program, University of Wyoming, Laramie, Wyoming, United States
| |
Collapse
|
9
|
Li W, Zhu S, Liu J, Liu Z, Zhou H, Zhang Q, Yang Y, Chen L, Guo X, Zhang T, Meng L, Chai D, Tang G, Li X, Yang C. Zanubrutinib Ameliorates Cardiac Fibrosis and Inflammation Induced by Chronic Sympathetic Activation. Molecules 2023; 28:6035. [PMID: 37630287 PMCID: PMC10458081 DOI: 10.3390/molecules28166035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/19/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
(1) Background: Heart failure (HF) is the final stage of multiple cardiac diseases, which have now become a severe public health problem worldwide. β-Adrenergic receptor (β-AR) overactivation is a major pathological factor associated with multiple cardiac diseases and mediates cardiac fibrosis and inflammation. Previous research has demonstrated that Bruton's tyrosine kinase (BTK) mediated cardiac fibrosis by TGF-β related signal pathways, indicating that BTK was a potential drug target for cardiac fibrosis. Zanubrutinib, a second-generation BTK inhibitor, has shown anti-fibrosis effects in previous research. However, it is unclear whether Zanubrutinib can alleviate cardiac fibrosis induced by β-AR overactivation; (2) Methods: In vivo: Male C57BL/6J mice were treated with or without the β-AR agonist isoproterenol (ISO) to establish a cardiac fibrosis animal model; (3) Results: In vivo: Results showed that the BTK inhibitor Zanubrutinib (ZB) had a great effect on cardiac fibrosis and inflammation induced by β-AR. In vitro: Results showed that ZB alleviated β-AR-induced cardiac fibroblast activation and macrophage pro-inflammatory cytokine production. Further mechanism studies demonstrated that ZB inhibited β-AR-induced cardiac fibrosis and inflammation by the BTK, STAT3, NF-κB, and PI3K/Akt signal pathways both in vivo and in vitro; (4) Conclusions: our research provides evidence that ZB ameliorates β-AR-induced cardiac fibrosis and inflammation.
Collapse
Affiliation(s)
- Wenqi Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Shuwen Zhu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Jing Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Zhigang Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
- Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| | - Qianyi Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Yue Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Li Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Xiaowei Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Tiantian Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Lingxin Meng
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Dan Chai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Guodong Tang
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, China; (W.L.); (S.Z.); (J.L.); (Z.L.); (H.Z.); (Q.Z.); (Y.Y.); (L.C.); (X.G.); (T.Z.); (L.M.); (D.C.)
- Tianjin International Joint Academy of Biomedicine, Tianjin 300457, China
| |
Collapse
|
10
|
Xu R, Ding Z, Li H, Shi J, Cheng L, Xu H, Wu J, Zou Y. Identification of early cardiac dysfunction and heterogeneity after pressure and volume overload in mice by high-frequency echocardiographic strain imaging. Front Cardiovasc Med 2023; 9:1071249. [PMID: 36712248 PMCID: PMC9880208 DOI: 10.3389/fcvm.2022.1071249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023] Open
Abstract
Object Aortic stenosis and regurgitation are clinically important conditions characterized with different hypertrophic types induced by pressure or volume overload, respectively, but with comparable cardiac function in compensated stage. Speckle-tracking based strain imaging has been applied to assess subtle alterations in cardiac abnormality, but its application in differentiating these two types of ventricular hypertrophy is still sparse. Here, we performed strain imaging analysis of cardiac remodeling in these two loading conditions. Methods C57BL/6J mice were subjected to transverse aortic constriction (TAC)-induced pressure overload or aortic regurgitation (AR)-induced volume overload. Conventional echocardiography and strain imaging were comprehensively assessed to detect stimulus-specific alterations in TAC and AR hearts. Results Conventional echocardiography did not detect significant changes in left ventricular systolic (ejection fraction and fractional shortening) and diastolic (E/E') function in either TAC or AR mice. On the contrary, global strain analysis revealed global longitudinal strain and strain rate were remarkably impaired in TAC while preserved in AR mice, although global radial, and circumferential strain and strain rate were significantly reduced in both models. Regional strain analysis in the long axis demonstrated that longitudinal strain and strain rate in all or most segments were decreased in TAC but maintained or slightly dented in AR mice, while radial strain and strain rate indicated overt decline in both models. Moreover, decreased radial and circumferential strain and strain rate were observed in most segments of TAC and AR mice in the short axis. Conclusion Strain imaging is superior to conventional echocardiography to detect subtle changes in myocardial deformation, with longitudinal strain and strain rate indicating distinct functional changes in pressure versus volume overload myocardial hypertrophy, making it potentially an advanced approach for early detection and differential diagnosis of cardiac dysfunction.
Collapse
Affiliation(s)
- Ran Xu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Zhiwen Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Hao Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing Shi
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Leilei Cheng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Department of Echocardiography, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Huixiong Xu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,*Correspondence: Jian Wu,
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China,Yunzeng Zou,
| |
Collapse
|
11
|
Exercise and Cardiac Fibrosis. CURRENT OPINION IN PHYSIOLOGY 2023. [DOI: 10.1016/j.cophys.2022.100630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
12
|
Tóth K, Oroszi T, Nyakas C, van der Zee EA, Schoemaker RG. Whole-body vibration as a passive alternative to exercise after myocardial damage in middle-aged female rats: Effects on the heart, the brain, and behavior. Front Aging Neurosci 2023; 15:1034474. [PMID: 36960421 PMCID: PMC10028093 DOI: 10.3389/fnagi.2023.1034474] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/02/2023] [Indexed: 03/09/2023] Open
Abstract
Background Females with cardiovascular disease seem more vulnerable to develop concomitant mental problems, such as depression and cognitive decline. Although exercise is shown beneficial in cardiovascular disease as well as in mental functions, these patients may be incapable or unmotivated to perform exercise. Whole body vibration (WBV) could provide a passive alternative to exercise. Aim of the present study was to compare WBV to exercise after isoproterenol (ISO)-induced myocardial damage in female rats, regarding effects on heart, brain and behavior. Methods One week after ISO (70 mg/kg s.c., on 2 consecutive days) or saline injections, 12 months old female rats were assigned to WBV (10 minutes daily), treadmill running (30 minutes daily) or pseudo intervention for 5 weeks. During the last 10 days, behavioral tests were performed regarding depressive-like behavior, cognitive function, and motor performance. Rats were sacrificed, brains and hearts were dissected for (immuno)histochemistry. Results Significant ISO-induced cardiac collagen deposition (0.67 ± 0.10 vs 0.18 ± 0.03%) was absent after running (0.45 ± 0.26 vs 0.46 ± 0.08%), but not after WBV (0.83 ± 0.12 vs 0.41 ± 0.05%). However, WBV as well as running significantly reduced hippocampal (CA3) collagen content in ISO-treated rats. Significant regional differences in hippocampal microglia activity and brain derived neurotrophic factor (BDNF) expression were observed. Significant ISO-induced CA1 microglia activation was reduced after WBV as well as running, while opposite effects were observed in the CA3; significant reduction after ISO that was restored by WBV and running. Both WBV and running reversed the ISO-induced increased BDNF expression in the CA1, Dentate gyrus and Hilus, but not in the CA3 area. Whereas running had no significant effect on behavior in the ISO-treated rats, WBV may be associated with short-term spatial memory in the novel location recognition test. Conclusion Although the female rats did not show the anticipated depressive-like behavior or cognitive decline after ISO, our data indicated regional effects on neuroinflammation and BDNF expression in the hippocampus, that were merely normalized by both WBV and exercise. Therefore, apart from the potential concern about the lack of cardiac collagen reduction, WBV may provide a relevant alternative for physical exercise.
Collapse
Affiliation(s)
- Kata Tóth
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- Research Center for Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
| | - Tamás Oroszi
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- Research Center for Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
| | - Csaba Nyakas
- Research Center for Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
- Behavioral Physiology Research Laboratory, Health Science Faculty, Semmelweis University, Budapest, Hungary
| | - Eddy A. van der Zee
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
| | - Regien G. Schoemaker
- Department of Neurobiology, Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Groningen, Netherlands
- Department of Cardiology, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Regien G. Schoemaker
| |
Collapse
|
13
|
Feng N, Yu H, Wang Y, Zhang Y, Xiao H, Gao W. Exercise training attenuates angiotensin II-induced cardiac fibrosis by reducing POU2F1 expression. JOURNAL OF SPORT AND HEALTH SCIENCE 2022:S2095-2546(22)00104-1. [PMID: 36374849 PMCID: PMC10362488 DOI: 10.1016/j.jshs.2022.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 09/09/2022] [Accepted: 09/24/2022] [Indexed: 06/16/2023]
Abstract
PURPOSE Exercise training protects against heart failure. However, the mechanism underlying the protective effect of exercise training on angiotensin II (Ang II)-induced cardiac fibrosis remains unclear. METHODS An exercise model involving C57BL/6N mice and 6 weeks of treadmill training was used. Ang II (1.44 mg/kg/day) was administered to induce cardiac fibrosis. RNA sequencing and bioinformatic analysis were used to identify the key factors mediating the effects of exercise training on cardiac fibrosis. Primary adult mouse cardiac fibroblasts (CFs) were used in vitro. Adeno-associated virus serotype 9 was used to overexpress POU domain, class 2, transcription factor 1 (POU2F1) in vivo. RESULTS Exercise training attenuated Ang II-induced cardiac fibrosis and reversed 39 gene expression changes. The transcription factor regulating the largest number of these genes was POU2F1. Compared to controls, POU2F1 was shown to be significantly upregulated by Ang II, which is itself reduced by exercise training. In vivo, POU2F1 overexpression nullified the benefits of exercise training on cardiac fibrosis. In CFs, POU2F1 promoted cardiac fibrosis. CCAAT enhancer-binding protein β (C/EBPβ) was predicted to be the transcription factor of POU2F1 and verified using a dual-luciferase reporter assay. In vivo, exercise training activated AMP-activated protein kinase (AMPK) and alleviated the increase in C/EBPβ induced by Ang II. In CFs, AMPK agonist inhibited the increase in C/EBPβ and POU2F1 induced by Ang II, whereas AMPK inhibitor reversed this effect. CONCLUSION Exercise training attenuates Ang II-induced cardiac fibrosis by reducing POU2F1. Exercise training inhibits POU2F1 by activating AMPK, which is followed by the downregulation of C/EBPβ, the transcription factor of POU2F1.
Collapse
Affiliation(s)
- Na Feng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Haiyi Yu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Yueshen Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Youyi Zhang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, 100191, China.
| | - Wei Gao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, National Health Commission Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
14
|
Tóth K, Oroszi T, van der Zee EA, Nyakas C, Schoemaker RG. Sex dimorphism in isoproterenol-induced cardiac damage associated neuroinflammation and behavior in old rats. Front Aging Neurosci 2022; 14:854811. [PMID: 35936761 PMCID: PMC9354817 DOI: 10.3389/fnagi.2022.854811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Acute cardiac damage can be induced by isoproterenol injections in animals. The associated inflammatory response could be reflected in the brain as neuroinflammation, with potential consequences for brain function and behavior. Although cardiac responses are reported age and sex-related, for neuroinflammation and brain function this is virtually unknown. Therefore, cardiac damage and its consequences for neuroinflammation, brain function and behavior were compared in aged male and female rats. Wistar rats of 24 months of age were treated with isoproterenol (ISO, twice s.c.) or saline. Four weeks after injections, exploratory behavior and short-term memory were tested. Then, rats were sacrificed. Hearts were collected to measure cardiac damage. Brain tissue was collected to obtain measures of neuroinflammation and brain function. In male-, but not in female rats, ISO induced significant cardiac damage. Accordingly, mortality was higher in males than in females. Baseline hippocampal microglia activity was lower in females, while ISO induced neuroinflammation in both sexes, Hippocampal brain-derived neurotrophic factor expression appeared lower in females, without effects of ISO. In the open field test, ISO-treated males, but not females, displayed anxiety-like behavior. No effects of ISO were observed on short-term memory in either sex. In conclusion, sex dimorphism in effects of ISO was observed for cardiac damage and open field behavior. However, these effects could not be related to differences in hippocampal neuroinflammation or neuronal function.
Collapse
Affiliation(s)
- Kata Tóth
- Department of Neurobiology, Faculty of Science and Engineering, GELIFES, University of Groningen, Groningen, Netherlands
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Tamás Oroszi
- Department of Neurobiology, Faculty of Science and Engineering, GELIFES, University of Groningen, Groningen, Netherlands
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Eddy A. van der Zee
- Department of Neurobiology, Faculty of Science and Engineering, GELIFES, University of Groningen, Groningen, Netherlands
| | - Csaba Nyakas
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
- Behavioral Physiology Research Laboratory, Health Science Faculty, Semmelweis University, Budapest, Hungary
| | - Regien G. Schoemaker
- Department of Neurobiology, Faculty of Science and Engineering, GELIFES, University of Groningen, Groningen, Netherlands
- Department of Mobility, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Regien G. Schoemaker,
| |
Collapse
|
15
|
Huston P. A Sedentary and Unhealthy Lifestyle Fuels Chronic Disease Progression by Changing Interstitial Cell Behaviour: A Network Analysis. Front Physiol 2022; 13:904107. [PMID: 35874511 PMCID: PMC9304814 DOI: 10.3389/fphys.2022.904107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Managing chronic diseases, such as heart disease, stroke, diabetes, chronic lung disease and Alzheimer’s disease, account for a large proportion of health care spending, yet they remain in the top causes of premature mortality and are preventable. It is currently accepted that an unhealthy lifestyle fosters a state of chronic low-grade inflammation that is linked to chronic disease progression. Although this is known to be related to inflammatory cytokines, how an unhealthy lifestyle causes cytokine release and how that in turn leads to chronic disease progression are not well known. This article presents a theory that an unhealthy lifestyle fosters chronic disease by changing interstitial cell behavior and is supported by a six-level hierarchical network analysis. The top three networks include the macroenvironment, social and cultural factors, and lifestyle itself. The fourth network includes the immune, autonomic and neuroendocrine systems and how they interact with lifestyle factors and with each other. The fifth network identifies the effects these systems have on the microenvironment and two types of interstitial cells: macrophages and fibroblasts. Depending on their behaviour, these cells can either help maintain and restore normal function or foster chronic disease progression. When macrophages and fibroblasts dysregulate, it leads to chronic low-grade inflammation, fibrosis, and eventually damage to parenchymal (organ-specific) cells. The sixth network considers how macrophages change phenotype. Thus, a pathway is identified through this hierarchical network to reveal how external factors and lifestyle affect interstitial cell behaviour. This theory can be tested and it needs to be tested because, if correct, it has profound implications. Not only does this theory explain how chronic low-grade inflammation causes chronic disease progression, it also provides insight into salutogenesis, or the process by which health is maintained and restored. Understanding low-grade inflammation as a stalled healing process offers a new strategy for chronic disease management. Rather than treating each chronic disease separately by a focus on parenchymal pathology, a salutogenic strategy of optimizing interstitial health could prevent and mitigate multiple chronic diseases simultaneously.
Collapse
Affiliation(s)
- Patricia Huston
- Department of Family Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Institut du Savoir Montfort (Research), University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Patricia Huston, , orcid.org/0000-0002-2927-1176
| |
Collapse
|
16
|
The effects of exercise training on heart, brain and behavior, in the isoproterenol-induced cardiac infarct model in middle-aged female rats. Sci Rep 2022; 12:10095. [PMID: 35710575 PMCID: PMC9203707 DOI: 10.1038/s41598-022-14168-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022] Open
Abstract
Women with cardiovascular disease may be more susceptible to concomitant mental health problems, such as depression and cognitive decline. Exercise training has beneficial effects on the cardiovascular system as well as on mental functions. Aim of the present study was to study the effects of exercise training on heart, brain and behavior in the isoproterenol (ISO) model in middle-aged female rats. Twelve months old female Wistar rats were submitted to ISO injections (70 mg/kg s.c., on two consecutive days) or received saline. One week later, rats were assigned to either exercise training (treadmill running) or control handling for five weeks. During the last 7 days, tests were performed regarding depressive-like behavior and cognitive function. Then, rats were sacrificed and heart and brains were dissected for (immuno)histochemistry. ISO-induced cardiac effects were eminent from cardiac fibrosis and declined cardiac function. Exercise training reversed cardiac damage and partly restored ISO-induced cardiac dysfunction. However, ISO treatment could not be associated with neuroinflammation, nor impaired hippocampal neurogenesis or neuronal function. Accordingly, no cognitive impairment or depressive-like behavior were observed. Actually, hippocampal microglia hyper-ramification was observed after ISO. Exercise left neuroinflammation and behavior merely unaltered, and even reduced neuronal function. Our data indicated that the cardiac damage after ISO in middle-aged female rats, and the subsequent beneficial effects of five weeks exercise training on the heart, were not reflected in changes in the brain nor in altered behavior.
Collapse
|
17
|
Glibenclamide alleviates β adrenergic receptor activation-induced cardiac inflammation. Acta Pharmacol Sin 2022; 43:1243-1250. [PMID: 34349235 PMCID: PMC9061800 DOI: 10.1038/s41401-021-00734-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/29/2021] [Indexed: 11/08/2022] Open
Abstract
β-Adrenergic receptor (β-AR) overactivation is a major pathological factor associated with cardiac diseases and mediates cardiac inflammatory injury. Glibenclamide has shown anti-inflammatory effects in previous research. However, it is unclear whether and how glibenclamide can alleviate cardiac inflammatory injury induced by β-AR overactivation. In the present study, male C57BL/6J mice were treated with or without the β-AR agonist isoprenaline (ISO) with or without glibenclamide pretreatment. The results indicated that glibenclamide alleviated ISO-induced macrophage infiltration in the heart, as determined by Mac-3 staining. Consistent with this finding, glibenclamide also inhibited ISO-induced chemokines and proinflammatory cytokines expression in the heart. Moreover, glibenclamide inhibited ISO-induced cardiac fibrosis and dysfunction in mice. To reveal the protective mechanism of glibenclamide, the NLRP3 inflammasome was further analysed. ISO activated the NLRP3 inflammasome in both cardiomyocytes and mouse hearts, but this effect was alleviated by glibenclamide pretreatment. Furthermore, in cardiomyocytes, ISO increased the efflux of potassium and the generation of ROS, which are recognized as activators of the NLRP3 inflammasome. The ISO-induced increases in these processes were inhibited by glibenclamide pretreatment. Moreover, glibenclamide inhibited the cAMP/PKA signalling pathway, which is downstream of β-AR, by increasing phosphodiesterase activity in mouse hearts and cardiomyocytes. In conclusion, glibenclamide alleviates β-AR overactivation-induced cardiac inflammation by inhibiting the NLRP3 inflammasome. The underlying mechanism involves glibenclamide-mediated suppression of potassium efflux and ROS generation by inhibiting the cAMP/PKA pathway.
Collapse
|
18
|
Shi X, Chen X, Qiu X, Luo W, Luo X, Liu H, Geng Q, Ma H, Xue L, Guo L. Effect of High-Intensity Interval Training, Moderate Continuous Training, or Guideline-Based Physical Activity on Peak Oxygen Uptake and Myocardial Fibrosis in Patients With Myocardial Infarction: Protocol for a Randomized Controlled Trial. Front Cardiovasc Med 2022; 9:860071. [PMID: 35479268 PMCID: PMC9035926 DOI: 10.3389/fcvm.2022.860071] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/04/2022] [Indexed: 11/30/2022] Open
Abstract
Introduction High-intensity interval training (HIIT) is an emerging method of cardiac rehabilitation, which is more and more popular in recent years. Research into the effect of HIIT on peak oxygen uptake (VO2 peak) and myocardial fibrosis among patients with myocardial infarction (MI) is lacking. Here, we describe the rationale along with the protocol for a clinical trial to test the following hypotheses: (1) compared with the control group, VO2 peak will be increased in both the moderate-intensity continuous training (MICT) and HIIT groups and (2) compared with the control group, myocardial fibrosis due to MI will be improved by HIIT and MICT. Methods and Analysis This is a single-center, randomized controlled clinical trial. In total, 180 patients with MI are to be recruited for this study. VO2 peak will be tested by cardiopulmonary exercise testing (CPET) and myocardial fibrosis will be evaluated by cardiac MR. A variety of blood and psychometric tests and also the peripheral arterial tonometry, reactive hyperemia index for microvascular endothelial function, and microvascular blockage or digital vasomotor response are included. Ethics and Dissemination The ethics committee of the Guangdong Provincial People's Hospital has authorized this mechanistic clinical research. Peer-reviewed articles and conference presentations will be used to disseminate the findings. Trial Registration Number NCT04863677.
Collapse
Affiliation(s)
- Xiaohe Shi
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xianyuan Chen
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinfan Qiu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Wei Luo
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinyi Luo
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hui Liu
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qingshan Geng
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Huan Ma
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- *Correspondence: Huan Ma
| | - Ling Xue
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Ling Xue
| | - Lan Guo
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Lan Guo
| |
Collapse
|
19
|
Tóth K, Oroszi T, van der Zee EA, Nyakas C, Schoemaker RG. Effects of exercise training on behavior and brain function after high dose isoproterenol-induced cardiac damage. Sci Rep 2021; 11:23576. [PMID: 34880374 PMCID: PMC8654950 DOI: 10.1038/s41598-021-03107-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/26/2021] [Indexed: 02/02/2023] Open
Abstract
Acute sympathetic stress can result in cardiac fibrosis, but may also lead to mental dysfunction. Exercise training after isoproterenol (ISO)-induced acute sympathetic stress was investigated regarding cardiac damage, neuroinflammation, brain function and behavior. Male Wistar rats (12 months) received ISO or saline. One week later, treadmill running or control handling (sedentary) started. After 4 weeks, cognitive- and exploratory behavior were evaluated, and heart and brain tissues were analyzed regarding cardiac damage, hippocampal neuroinflammation and neuronal function. ISO did not affect cognitive performance nor hippocampal function. However, ISO reduced anxiety, coinciding with locally reduced microglia (processes) size in the hippocampus. Exercise in ISO rats reversed anxiety, did not affect microglia morphology, but increased brain function. Thus, exercise after ISO did not affect cardiac damage, cognition or hippocampal neuroinflammation, but normalized anxiety. Increased localized BDNF expression may indicate improved brain function.
Collapse
Affiliation(s)
- Kata Tóth
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.,Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Tamás Oroszi
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.,Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary
| | - Eddy A van der Zee
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Csaba Nyakas
- Research Center for Molecular Exercise Science, University of Physical Education, Budapest, Hungary.,Behavioral Physiology Research Laboratory, Health Science Faculty, Semmelweis University, Budapest, Hungary
| | - Regien G Schoemaker
- Department of Neurobiology, GELIFES, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands. .,University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
20
|
Domínguez F, Lalaguna L, López-Olañeta M, Villalba-Orero M, Padrón-Barthe L, Román M, Bello-Arroyo E, Briceño A, Gonzalez-Lopez E, Segovia-Cubero J, García-Pavía P, Lara-Pezzi E. Early Preventive Treatment With Enalapril Improves Cardiac Function and Delays Mortality in Mice With Arrhythmogenic Right Ventricular Cardiomyopathy Type 5. Circ Heart Fail 2021; 14:e007616. [PMID: 34412508 DOI: 10.1161/circheartfailure.120.007616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Arrhythmogenic right ventricular cardiomyopathy type 5 (ARVC5) is an inherited cardiac disease with complete penetrance and an aggressive clinical course caused by mutations in TMEM43 (transmembrane protein 43). There is no cure for ARVC5 and palliative treatment is started once the phenotype is present. A transgenic mouse model of ARVC5 expressing human TMEM43-S358L (TMEM43mut) recapitulates the human disease, enabling the exploration of preventive treatments. The aim of this study is to determine whether preventive treatment with heart failure drugs (β-blockers, ACE [angiotensin-converting enzyme] inhibitors, mineralocorticoid-receptor antagonists) improves the disease course of ARVC5 in TMEM43mut mice. METHODS TMEM43mut male/female mice were treated with metoprolol (β-blockers), enalapril (ACE inhibitor), spironolactone (mineralocorticoid-receptor antagonist), ACE inhibitor + mineralocorticoid-receptor antagonist, ACE inhibitor + mineralocorticoid-receptor antagonist + β-blockers or left untreated. Drugs were initiated at 3 weeks of age, before ARVC5 phenotype, and serial ECG and echocardiograms were performed. RESULTS TMEM43mut mice treated with enalapril showed a significantly increased median survival compared with untreated mice (26 versus 21 weeks; P=0.003). Enalapril-treated mice also exhibited increased left ventricular ejection fraction at 4 months compared with controls (37.0% versus 24.9%; P=0.004), shorter QRS duration and reduced left ventricle fibrosis. Combined regimens including enalapril also showed positive effects. Metoprolol decreased QRS voltage prematurely and resulted in a nonsignificant decrease in left ventricular ejection fraction compared with untreated TMEM43mut mice. CONCLUSIONS Preventive enalapril-based regimens reduced fibrosis, improved ECG, echocardiographic parameters and survival of ARVC5 mice. Early metoprolol did not show positive effects and caused premature ECG abnormalities. Our findings pave the way to consider prophylactic enalapril in asymptomatic ARVC5 genetic carriers.
Collapse
Affiliation(s)
- Fernando Domínguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.).,Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (F.D., A.B., E.G.-L., J.S.-C., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (F.D., E.G.-L., J.S.-C., P.G.-P., E.L.-P.)
| | - Laura Lalaguna
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - Marina López-Olañeta
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - María Villalba-Orero
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - Laura Padrón-Barthe
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - Marta Román
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - Elísabet Bello-Arroyo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.)
| | - Ana Briceño
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (F.D., A.B., E.G.-L., J.S.-C., P.G.-P.)
| | - Esther Gonzalez-Lopez
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (F.D., A.B., E.G.-L., J.S.-C., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (F.D., E.G.-L., J.S.-C., P.G.-P., E.L.-P.)
| | - Javier Segovia-Cubero
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (F.D., A.B., E.G.-L., J.S.-C., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (F.D., E.G.-L., J.S.-C., P.G.-P., E.L.-P.)
| | - Pablo García-Pavía
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain (F.D., A.B., E.G.-L., J.S.-C., P.G.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (F.D., E.G.-L., J.S.-C., P.G.-P., E.L.-P.).,Francisco de Vitoria University, Madrid, Spain (P.G.-P.)
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (F.D., L.L., M.L.-O., M.V.-O., L.P.-B., M.R., E.B.-A., E.L.-P.).,CIBER Cardiovascular Diseases (CIBERCV), Madrid, Spain (F.D., E.G.-L., J.S.-C., P.G.-P., E.L.-P.)
| |
Collapse
|
21
|
Zhang W, Zhu B, Ding S, Wang X, Wu J, Zhu X, Zou Y, Ge J, Tong M, Yang X. Disruption of STAT6 Signal Promotes Cardiac Fibrosis Through the Mobilization and Transformation of CD11b + Immature Myeloid Cells. Front Physiol 2020; 11:579712. [PMID: 33192584 PMCID: PMC7642801 DOI: 10.3389/fphys.2020.579712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 12/23/2022] Open
Abstract
Cardiac fibrosis is an important pathological basis of various cardiovascular diseases. The roles of STAT6 signal in allergy, immune regulation, tumorigenesis, and renal fibrosis have been documented. However, the function and mechanism of STAT6 signal in sympathetic overactivation-induced cardiac fibrosis have not been fully elucidated. This study explores the novel role of STAT6 signal in isoproterenol (ISO)-induced cardiac fibrosis through the regulation of inflammatory response and the differentiation of macrophages from immature myeloid cells. The expression levels of STAT6, β1-adrenergic receptor (β1-AR), and inflammatory factors [interleukin α (IL-1α), IL-6, IL-18, and transforming growth factor β (TGF-β)] in CD11b+ myeloid cells were analyzed with a microarray study. The levels of IL-6 and TGF-β1 in the CD11b+ myeloid cells-derived macrophages were detected with reverse transcriptase-polymerase chain reaction (RT-PCR). STAT6-knockout (KO) and WT mice were used to establish a murine cardiac fibrosis model by ISO injection. Cardiac fibroblasts were isolated from the hearts of newborn STAT6-KO and WT mice, and STAT6 expression was measured by Western blotting and RT-PCR after ISO stimulation, while α-smooth muscle actin (α-SMA) expression was detected by immunofluorescence and immunohistochemistry staining. Cardiac function and pathological characteristics were examined by echocardiography and immunohistochemistry staining, respectively. Immunohistochemistry staining with anti-CD11b was performed to detect the infiltration of CD11b+ myeloid cells in heart tissue. Flow cytometry analysis was used to measure the percentages of CD11b+ myeloid cells and CD11b+Ly6C+ macrophages in the peripheral blood. The results showed that STAT6 was highly expressed in CD11b+ myeloid cells located in injured hearts, and STAT6 expression in cardiac fibroblasts was down-regulated after ISO treatment. STAT6 deficiency further aggravated ISO-induced increased expression of α-SMA in cardiac fibroblasts, myocardial fibrosis, and cardiac dysfunction. The activation of ISO/β1-AR signal aggravated cardiac inflammatory infiltration, promoted CD11b+ myeloid cell mobilization, and enhanced CD11b+Ly6C+/low macrophage differentiation, which was further exacerbated by STAT6 deficiency. Furthermore, β1-AR mRNA expression significantly increased in splenic CD11b+ myeloid cells compared to their bone marrow-derived controls, and STAT6 deficiency promoted β1-AR expression in an MI-induced sensitive cardiac fibrosis mouse model. The spleen-derived CD11b+ myeloid cells of STAT6-KO mice produced more IL-1α, IL-18, and TGF-β than their WT counterparts. Taken together, these results suggest that STAT6 signal plays a critical role in ISO-induced β1-AR overactivation and systemic inflammatory cascades, contributing to cardiac fibrogenesis. STAT6 should be a promising cardioprotective target against myocardial fibrosis and heart failure after β1-AR overactivation-induced myocardial injury.
Collapse
Affiliation(s)
- Weiwei Zhang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Baoling Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Suling Ding
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiangfei Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaowei Zhu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Minghong Tong
- Division of Clinical Laboratory, TongRen Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiangdong Yang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.,Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Xu M, Jiang H, Xiao J. Exercise Protects Sympathetic Stress-Induced Myocardial Fibrosis by Regulating Cytokines. J Cardiovasc Transl Res 2019; 13:570-571. [PMID: 31745712 DOI: 10.1007/s12265-019-09933-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Minjun Xu
- School of Medicine (in preparation), Shanghai University, Shanghai, 200444, China
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China
| | - Huimin Jiang
- Clinical Laboratory Center, Beijing Hospital of Traditional Chinese Medicine, Beijing, 100010, China
| | - Junjie Xiao
- School of Medicine (in preparation), Shanghai University, Shanghai, 200444, China.
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|