1
|
Liu S, Su L, Li J, Zhang Y, Hu X, Wang P, Liu P, Ye J. Inhibition of miR-146b-5p alleviates isoprenaline-induced cardiac hypertrophy via regulating DFCP1. Mol Cell Endocrinol 2024; 589:112252. [PMID: 38649132 DOI: 10.1016/j.mce.2024.112252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Pathological cardiac hypertrophy often precedes heart failure due to various stimuli, yet effective clinical interventions remain limited. Recently, microRNAs (miRNAs) have been identified as critical regulators of cardiovascular development. In this study, we investigated the role of miR-146b-5p and its underlying mechanisms of action in cardiac hypertrophy. Isoprenaline (ISO) treatment induced significant hypertrophy and markedly enhanced the expression of miR-146b-5p in cultured neonatal rat cardiomyocytes and hearts of C57BL/6 mice. Transfection with the miR-146b-5p mimic led to cardiomyocyte hypertrophy accompanied by autophagy inhibition. Conversely, miR-146b-5p inhibition significantly alleviated ISO-induced autophagy depression, thereby mitigating cardiac hypertrophy both in vitro and in vivo. Our results showed that the autophagy-related mediator double FYVE domain-containing protein 1 (DFCP1) is a target of miR-146b-5p. MiR-146b-5p blocked autophagic flux in cardiomyocytes by suppressing DFCP1, thus contributing to hypertrophy. These findings revealed that miR-146b-5p is a potential regulator of autophagy associated with the onset of cardiac hypertrophy, suggesting a possible therapeutic strategy involving the inhibition of miR-146b-5p.
Collapse
Affiliation(s)
- Siling Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Linjie Su
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Jie Li
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Yuexin Zhang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Xiaopei Hu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Pengcheng Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China
| | - Peiqing Liu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China.
| | - Jiantao Ye
- School of Pharmaceutical Sciences, Sun Yat-Sen University, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, China.
| |
Collapse
|
2
|
Karmazyn M, Gan XT. Molecular and Cellular Mechanisms Underlying the Cardiac Hypertrophic and Pro-Remodelling Effects of Leptin. Int J Mol Sci 2024; 25:1137. [PMID: 38256208 PMCID: PMC10816997 DOI: 10.3390/ijms25021137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Since its initial discovery in 1994, the adipokine leptin has received extensive interest as an important satiety factor and regulator of energy expenditure. Although produced primarily by white adipocytes, leptin can be synthesized by numerous tissues including those comprising the cardiovascular system. Cardiovascular function can thus be affected by locally produced leptin via an autocrine or paracrine manner but also by circulating leptin. Leptin exerts its effects by binding to and activating specific receptors, termed ObRs or LepRs, belonging to the Class I cytokine family of receptors of which six isoforms have been identified. Although all ObRs have identical intracellular domains, they differ substantially in length in terms of their extracellular domains, which determine their ability to activate cell signalling pathways. The most important of these receptors in terms of biological effects of leptin is the so-called long form (ObRb), which possesses the complete intracellular domain linked to full cell signalling processes. The heart has been shown to express ObRb as well as to produce leptin. Leptin exerts numerous cardiac effects including the development of hypertrophy likely through a number of cell signaling processes as well as mitochondrial dynamics, thus demonstrating substantial complex underlying mechanisms. Here, we discuss mechanisms that potentially mediate leptin-induced cardiac pathological hypertrophy, which may contribute to the development of heart failure.
Collapse
|
3
|
Zhazykbayeva S, Hassoun R, Herwig M, Budde H, Kovács Á, Mannherz HG, El-Battrawy I, Tóth A, Schmidt WE, Mügge A, Hamdani N. Oxidative stress and inflammation distinctly drive molecular mechanisms of diastolic dysfunction and remodeling in female and male heart failure with preserved ejection fraction rats. Front Cardiovasc Med 2023; 10:1157398. [PMID: 37363100 PMCID: PMC10285478 DOI: 10.3389/fcvm.2023.1157398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/26/2023] [Indexed: 06/28/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a complex cardiovascular insufficiency syndrome presenting with an ejection fraction (EF) of greater than 50% along with different proinflammatory and metabolic co-morbidities. Despite previous work provided key insights into our understanding of HFpEF, effective treatments are still limited. In the current study we attempted to unravel the molecular basis of sex-dependent differences in HFpEF pathology. We analyzed left ventricular samples from 1-year-old female and male transgenic (TG) rats homozygous for the rat Ren-2 renin gene (mRen2) characterized with hypertension and diastolic dysfunction and compared it to age-matched female and male wild type rats (WT) served as control. Cardiomyocytes from female and male TG rats exhibited an elevated titin-based stiffness (Fpassive), which was corrected to control level upon treatment with reduced glutathione indicating titin oxidation. This was accompanied with high levels of oxidative stress in TG rats with more prominent effects in female group. In vitro supplementation with heat shock proteins (HSPs) reversed the elevated Fpassive indicating restoration of their cytoprotective function. Furthermore, the TG group exhibited high levels of proinflammatory cytokines with significant alterations in apoptotic and autophagy pathways in both sexes. Distinct alterations in the expression of several proteins between both sexes suggest their differential impact on disease development and necessitate distinct treatment options. Hence, our data suggested that oxidative stress and inflammation distinctly drive diastolic dysfunction and remodeling in female and male rats with HFpEF and that the sex-dependent mechanisms contribute to HF pathology.
Collapse
Affiliation(s)
- Saltanat Zhazykbayeva
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| | - Roua Hassoun
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| | - Melissa Herwig
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| | - Heidi Budde
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| | - Árpád Kovács
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| | - Hans Georg Mannherz
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany
| | - Ibrahim El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, UK RUB, Ruhr University of Bochum, Bochum, Germany
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Research Centre for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| | - Wolfgang E. Schmidt
- Department of Medicine I, St. Josef Hospital, UK RUB, Ruhr-University Bochum, Bochum, Germany
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, UK RUB, Ruhr University of Bochum, Bochum, Germany
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Bochum, Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
- Department of Cardiology, St. Josef-Hospital, UK RUB, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
4
|
Pang B, Jiang YR, Xu JY, Shao DX, Hao LY. Apelin/ELABELA-APJ system in cardiac hypertrophy: Regulatory mechanisms and therapeutic potential. Eur J Pharmacol 2023; 949:175727. [PMID: 37062502 DOI: 10.1016/j.ejphar.2023.175727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 04/18/2023]
Abstract
Heart failure is one of the most significant public health problems faced by millions of medical researchers worldwide. And pathological cardiac hypertrophy is considered one of the possible factors of increasing the risk of heart failure. Here, we introduce apelin/ELABELA-APJ system as a novel therapeutic target for cardiac hypertrophy, bringing about new directions in clinical treatment. Apelin has been proven to regulate cardiac hypertrophy through various pathways. And an increasing number of studies on ELABELA, the newly discovered endogenous ligand, suggest it can alleviate cardiac hypertrophy through mechanisms similar or different to apelin. In this review, we elaborate on the role that apelin/ELABELA-APJ system plays in cardiac hypertrophy and the intricate mechanisms that apelin/ELABELA-APJ affect cardiac hypertrophy. We also illuminate and make comparisons of the newly designed peptides and small molecules as agonists and antagonists for APJ, updating the breakthroughs in this field.
Collapse
Affiliation(s)
- Bo Pang
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Yin-Ru Jiang
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Jia-Yao Xu
- China Medical University-The Queen's University of Belfast Joint College, Queen's University Belfast, Belfast Northern Ireland, BT9 7BL, United Kingdom.
| | - Dong-Xue Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| | - Li-Ying Hao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
5
|
Zhang L, Liu W, Wu N, Wang H, Zhang Z, Liu Y, Wang X. Southern rice black-streaked dwarf virus induces incomplete autophagy for persistence in gut epithelial cells of its vector insect. PLoS Pathog 2023; 19:e1011134. [PMID: 36706154 PMCID: PMC9907856 DOI: 10.1371/journal.ppat.1011134] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 02/08/2023] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Autophagy plays an important role in virus infection of the host, because viral components and particles can be degraded by the host's autophagy and some viruses may be able to hijack and subvert autophagy for its benefit. However, details on the mechanisms that govern autophagy for immunity against viral infections or benefit viral survival remain largely unknown. Plant reoviruses such as southern rice black-streaked dwarf virus (SRBSDV), which seriously threaten crop yield, are only transmitted by vector insects. Here, we report a novel mechanism by which SRBSDV induces incomplete autophagy by blocking autophagosome-lysosome fusion, resulting in viral accumulation in gut epithelial cells of its vector, white-backed planthopper (Sogatella furcifera). SRBSDV infection leads to stimulation of the c-Jun N-terminal kinase (JNK) signaling pathway, which further activates autophagy. Mature and assembling virions were found close to the edge7 of the outer membrane of autophagosomes. Inhibition autophagy leads to the decrease of autophagosomes, which resulting in impaired maturation of virions and the decrease of virus titer, whereas activation of autophagy facilitated virus titer. Further, SRBSDV inhibited fusion of autophagosomes and lysosomes by interacting with lysosomal-associated membrane protein 1 (LAMP1) using viral P10. Thus, SRBSDV not only avoids being degrading by lysosomes, but also further hijacks these non-fusing autophagosomes for its subsistence. Our findings reveal a novel mechanism of reovirus persistence, which can explain why SRBSDV can be acquired and transmitted rapidly by its insect vector.
Collapse
Affiliation(s)
- Lu Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- Beijing Key Laboratory of Maize DNA Fingerprinting and Molecular Breeding, Maize Research Institute, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Wenwen Liu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (WL); (XW)
| | - Nan Wu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hui Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zhongkai Zhang
- Biotechnology and Germplasm Resources Institute, Yunnan Key Laboratory of Agricultural Biotechnology, Yunnan Academy of Agricultural Sciences, Kunming, China
| | - Yule Liu
- MOE Key Laboratory of Bioinformatics and Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xifeng Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
- * E-mail: (WL); (XW)
| |
Collapse
|
6
|
Wu J, Li K, Liu Y, Feng A, Liu C, Adu-Amankwaah J, Ji M, Ma Y, Hao Y, Bu H, Sun H. Daidzein ameliorates doxorubicin-induced cardiac injury by inhibiting autophagy and apoptosis in rats. Food Funct 2023; 14:934-945. [PMID: 36541083 DOI: 10.1039/d2fo03416f] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Backgrounds: Doxorubicin (Dox) is a classical antitumor antibiotic widely restricted for use due to its cardiotoxicity. Daidzein (Daid) is a soy isoflavone that enhances antioxidant enzyme systems and inhibits apoptosis to prevent cardiovascular diseases. In this study, we intended to assess whether Daid protects against Dox-induced cardiotoxicity and explored its underlying mechanisms. Methods: Male Sprague-Dawley (SD) rats were divided into five groups: control (Ctrl), 40 mg per kg per day Daidzein (Daid), 3 mg per kg per week doxorubicin (Dox), 20 mg per kg per day Daidzein + 3 mg per kg per week doxorubicin (Daid20 + Dox) and 40 mg per kg per day Daidzein + 3 mg per kg per week doxorubicin (Daid40 + Dox) groups. Cardiac function assessments, immunohistochemistry (IHC) and immunofluorescence (IF) analyses were initially performed in each group of rats. Secondly, the cell proliferative capacity analysis, AO staining, and LC3 puncta analysis were employed to evaluate the cellular response to Dox in H9c2 cells. Ultimately, the protein expressions of cleaved caspase3, LC3 II, Bcl-2, Bax, Akt, p-Akt, and cyclin D1 were examined by western blotting. Results: Pretreatment with a low dose of Daid rather than a high dose significantly enhanced cardiac function and alleviated histopathological deterioration of cardiomyocytes induced by Dox. Daid downregulated the protein levels of Bax, LC3 II, cleaved caspase3 and p-Akt, while up-regulating Bcl-2 and cyclin D1. The Akt agonist SC79 could invalidate all the protective effects of Daid both in vivo and in vitro. Conclusions: Daid reduced autophagy and apoptosis by inhibiting the PI3K/Akt pathway, thereby protecting the hearts from Dox-induced cardiac damage.
Collapse
Affiliation(s)
- Jinxia Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Kexue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Yan Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Ailu Feng
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Chunyang Liu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Miaojin Ji
- Jiangsu Province Key Laboratory of Anesthesiology and Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, School of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yanhong Ma
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Yanling Hao
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Huimin Bu
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| | - Hong Sun
- Department of Physiology, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
7
|
Jiang X, Zhang K, Gao C, Ma W, Liu M, Guo X, Bao G, Han B, Hu H, Zhao Z. Activation of FMS-like tyrosine kinase 3 protects against isoprenaline-induced cardiac hypertrophy by improving autophagy and mitochondrial dynamics. FASEB J 2022; 36:e22672. [PMID: 36440960 DOI: 10.1096/fj.202200419rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 10/28/2022] [Accepted: 11/15/2022] [Indexed: 11/30/2022]
Abstract
FMS-like receptor tyrosine kinase 3 (Flt3) expression was reported to increase in the heart in response to pathological stress, but the role of Flt3 activation and its underlying mechanisms remain poorly elucidated. This study was designed to investigate the role of Flt3 activation in sympathetic hyperactivity-induced cardiac hypertrophy and its mechanisms through autophagy and mitochondrial dynamics. In vivo, cardiac hypertrophy was established by subcutaneous injection of isoprenaline (6 mg/kg·day) in C57BL/6 mice for 7 consecutive days. The Flt3-ligand intervention was launched 2 h prior to isoprenaline each day. In vitro, experiments of cardiomyocyte hypertrophy, autophagy, and mitochondrial dynamics were performed in neonatal rat cardiomyocytes (NRCMs). Our results revealed that the expression level of Flt3 protein was significantly increased in the hypertrophic myocardium provoked by isoprenaline administration. Flt3-ligand intervention alleviated isoprenaline-induced cardiac oxidative stress, hypertrophy, fibrosis, and contractile dysfunction. Isoprenaline stimulation impaired autophagic flux in hypertrophic mouse hearts, supported by the accumulation of LC3II and P62 proteins, while Flt3-ligand restored the impairment of autophagic flux. Flt3 activation normalized the imbalance of mitochondrial fission and fusion in the hearts of mice evoked by isoprenaline as evidenced by the neutralization of elevated mitochondrial fission markers and reduced mitochondrial fusion markers. In NRCMs, Flt3-ligand treatment attenuated isoprenaline-stimulated hypertrophy, which was abolished by a Flt3-specific blocker AC220. Activating Flt3 reversed isoprenaline-induced autophagosome accumulation and impairment of autophagic flux probably by enhancing SIRT1 expression and consequently TFEB nuclear translocation. Flt3 activation improved the imbalance of mitochondrial dynamics induced by isoprenaline in NRCMs through the SIRT1/P53 pathway. Activation of Flt3 mitigated ISO-stimulated hypertrophy probably involves the restoration of autophagic flux and balance of mitochondrial dynamics. Therefore, activation of Flt3 attenuates isoprenaline-induced cardiac hypertrophy in vivo and in vitro, the potential mechanism probably attributes to SIRT1/TFEB-mediated autophagy promotion and SIRT1/P53-mediated mitochondrial dynamics balance. These findings suggest that activation of Flt3 may be a novel target for protection against cardiac remodeling and heart failure during sympathetic hyperactivity.
Collapse
Affiliation(s)
- Xixi Jiang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kaina Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chenying Gao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wenzhuo Ma
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Mengqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xinyu Guo
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Gaowa Bao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Bing Han
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Zhenghang Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
8
|
Zeng Y, Ren WQ, Wen AZ, Zhang W, Fan FY, Chen OY. Autophagy and pressure overload-induced cardiac hypertrophy. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:1101-1108. [PMID: 35043747 DOI: 10.1080/10286020.2021.2024810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
Autophagy plays an important role in the pathogenesis of cardiovascular diseases. Dysregulation of autophagy may have a huge effect on cardiac hypertrophy induced by overload pressure although reports on autophagy and cardiac hypertrophy have been contradictory. Some studies showed that autophagy activation attenuated cardiac hypertrophy. However, others suggested that inhibition of autophagy would be protective. Different research models or different pathways involved could be responsible for it. Cardiac hypertrophy may be alleviated through regulation of autophagy. This review aims to highlight the pathways and therapeutic targets identified in the prevention and treatment of cardiac hypertrophy by regulating autophagy.
Collapse
Affiliation(s)
- Yong Zeng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Wei-Qiong Ren
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ai-Zhen Wen
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Wen Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Fu-Yuan Fan
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ou-Ying Chen
- School of Nursing, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
9
|
Aboulgheit A, Karbasiafshar C, Sabra M, Zhang Z, Sodha N, Abid MR, Sellke FW. Extracellular vesicles improve diastolic function and substructure in normal and high-fat diet models of chronic myocardial ischemia. J Thorac Cardiovasc Surg 2022; 164:e371-e384. [PMID: 34756431 PMCID: PMC9005578 DOI: 10.1016/j.jtcvs.2021.07.062] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/29/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVE The burden of mortality and morbidity of cardiovascular disease is in part due to substantial fibrosis accelerated by coexisting risk factors. This study aims to evaluate the effect of extracellular vesicle therapy on diastolic function and myocardial fibrosis in the setting of chronic myocardial ischemia with and without a high-fat diet. METHODS Forty male Yorkshire swine were administered a normal or high-fat diet. At 11 weeks of age, they underwent placement of an ameroid constrictor on their left circumflex coronary artery. Both dietary groups then received either intramyocardial injection of vehicle saline as controls or extracellular vesicles as treatment into the ischemic territory (normal diet control, n = 8; high-fat diet controls, n = 11) or extracellular vesicles (normal diet extracellular vesicles, n = 9; high-fat diet extracellular vesicles, n = 12). Five weeks later, hemodynamic parameters, histology, and selected protein expression were evaluated. RESULTS Extracellular vesicles reduced end-diastolic pressure volume relationship (P = .002), perivascular collagen density (P = .031), calcium mineralization (P = .026), and cardiomyocyte diameter (P < .0001), and upregulated osteopontin (P = .0046) and mechanistic target of rapamycin (P = .021). An interaction between extracellular vesicles and diet was observed in the vimentin area (P = .044) and fraction of myofibroblast markers to total vimentin (P = .049). Significant changes across diet were found with reductions in muscle fiber area (P = .026), tumor necrosis factor α (P = .0002), NADPH oxidase 2 and 4 (P = .0036, P = .008), superoxide dismutase 1 (P = .034), and phosphorylated glycogen synthase kinase 3β (P = .020). CONCLUSIONS Extracellular vesicle therapy improved the myocardium's ability to relax and is likely due to structural improvements at the extracellular matrix and cellular levels.
Collapse
Affiliation(s)
- Ahmed Aboulgheit
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | | | - Mohamed Sabra
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI
| | - Zhiqi Zhang
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - Neel Sodha
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, RI; Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI.
| |
Collapse
|
10
|
Altered Cellular Protein Quality Control System Modulates Cardiomyocyte Function in Volume Overload-Induced Hypertrophy. Antioxidants (Basel) 2022; 11:antiox11112210. [DOI: 10.3390/antiox11112210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/10/2022] Open
Abstract
Volume-induced hypertrophy is one of the risk factors for cardiac morbidity and mortality. In addition, mechanical and metabolic dysfunction, aging, and cellular redox balance are also contributing factors to the disease progression. In this study, we used volume overload (VO), which was induced by an aortocaval fistula in 2-month-old male Wistar rats, and sham-operated animals served as control. Functional parameters were measured by transthoracic echocardiography at termination 4- or 8-months after VO. The animals showed hypertrophic remodeling that was accompanied by mechanical dysfunction and increased cardiomyocyte stiffness. These alterations were reversible upon treatment with glutathione. Cardiomyocyte dysfunction was associated with elevated oxidative stress markers with unchanged inflammatory signaling pathways. In addition, we observed altered phosphorylation status of small heat shock proteins 27 and 70 and diminished protease expression caspases 3 compared to the matched control group, indicating an impaired protein quality control system. Such alterations might be attributed to the increased oxidative stress as anticipated from the enhanced titin oxidation, ubiquitination, and the elevation in oxidative stress markers. Our study showed an early pathological response to VO, which manifests in cardiomyocyte mechanical dysfunction and dysregulated signaling pathways associated with enhanced oxidative stress and an impaired protein quality control system.
Collapse
|
11
|
Fan S, Hu Y. Role of m6A Methylation in the Occurrence and Development of Heart Failure. Front Cardiovasc Med 2022; 9:892113. [PMID: 35811741 PMCID: PMC9263194 DOI: 10.3389/fcvm.2022.892113] [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: 03/08/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
N6-methyladenosine (m6A) RNA methylation is one of the most common epigenetic modifications in RNA nucleotides. It is known that m6A methylation is involved in regulation, including gene expression, homeostasis, mRNA stability and other biological processes, affecting metabolism and a variety of biochemical regulation processes, and affecting the occurrence and development of a variety of diseases. Cardiovascular disease has high morbidity, disability rate and mortality in the world, of which heart failure is the final stage. Deeper understanding of the potential molecular mechanism of heart failure and exploring more effective treatment strategies will bring good news to the sick population. At present, m6A methylation is the latest research direction, which reveals some potential links between epigenetics and pathogenesis of heart failure. And m6A methylation will bring new directions and ideas for the prevention, diagnosis and treatment of heart failure. The purpose of this paper is to review the physiological and pathological mechanisms of m6A methylation that may be involved in cardiac remodeling in heart failure, so as to explain the possible role of m6A methylation in the occurrence and development of heart failure. And we hope to help m6A methylation obtain more in-depth research in the occurrence and development of heart failure.
Collapse
|
12
|
Deoxyelephantopin and Its Isomer Isodeoxyelephantopin: Anti-Cancer Natural Products with Multiple Modes of Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072086. [PMID: 35408483 PMCID: PMC9000713 DOI: 10.3390/molecules27072086] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/18/2022] [Accepted: 03/22/2022] [Indexed: 01/05/2023]
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. The development of cancer involves aberrations in multiple pathways, representing promising targets for anti-cancer drug discovery. Natural products are regarded as a rich source for developing anti-cancer therapies due to their unique structures and favorable pharmacology and toxicology profiles. Deoxyelephantopin and isodeoxyelephantopin, sesquiterpene lactone compounds, are major components of Elephantopus scaber and Elephantopus carolinianus, which have long been used as traditional medicines to treat multiple ailments, including liver diseases, diabetes, bronchitis, fever, diarrhea, dysentery, cancer, renal disorders, and inflammation-associated diseases. Recently, deoxyelephantopin and isodeoxyelephantopin have been extensively explored for their anti-cancer activities. This review summarizes and discusses the anti-cancer activities of deoxyelephantopin and isodeoxyelephantopin, with an emphasis on their modes of action and molecular targets. Both compounds disrupt several processes involved in cancer progression by targeting multiple signaling pathways deregulated in cancers, including cell cycle and proliferation, cell survival, autophagy, and invasion pathways. Future directions of research on these two compounds towards anti-cancer drug development are discussed.
Collapse
|
13
|
Kumari R, Ray AG, Mukherjee D, Chander V, Kar D, Kumar US, Bharadwaj P.V.P. D, Banerjee SK, Konar A, Bandyopadhyay A. Downregulation of PTEN Promotes Autophagy via Concurrent Reduction in Apoptosis in Cardiac Hypertrophy in PPAR α−/− Mice. Front Cardiovasc Med 2022; 9:798639. [PMID: 35224041 PMCID: PMC8881053 DOI: 10.3389/fcvm.2022.798639] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/14/2022] [Indexed: 01/05/2023] Open
Abstract
Cardiac hypertrophy is characterized by an increase in the size of the cardiomyocytes which is initially triggered as an adaptive response but ultimately becomes maladaptive with chronic exposure to different hypertrophic stimuli. Prolonged cardiac hypertrophy is often associated with mitochondrial dysfunctions and cardiomyocyte cell death. Peroxisome proliferator activated receptor alpha (PPAR α), which is critical for mitochondrial biogenesis and fatty acid oxidation, is down regulated in hypertrophied cardiomyocytes. Yet, the role of PPAR α in cardiomyocyte death is largely unknown. To assess the role of PPAR α in chronic hypertrophy, isoproterenol, a β-adrenergic receptor agonist was administered in PPAR α knock out (PPAR α−/−) mice for 2 weeks and hypertrophy associated changes in cardiac tissues were observed. Echocardiographic analysis ensured the development of cardiac hypertrophy and compromised hemodynamics in PPAR α−/− mice. Proteomic analysis using high resolution mass spectrometer identified about 1,200 proteins enriched in heart tissue. Proteins were classified according to biological pathway and molecular functions. We observed an unexpected down regulation of apoptotic markers, Annexin V and p53 in hypertrophied heart tissue. Further validation revealed a significant down regulation of apoptosis regulator, PTEN, along with other apoptosis markers like p53, Caspase 9 and c-PARP. The autophagy markers Atg3, Atg5, Atg7, p62, Beclin1 and LC3 A/B were up regulated in PPAR α−/− mice indicating an increase in autophagy. Similar observations were made in a high cholesterol diet fed PPAR α−/−mice. The results were further validated in vitro using NRVMs and H9C2 cell line by blocking PPAR α that resulted in enhanced autophagosome formation upon hypertrophic stimulation. The results demonstrate that in the absence of PPAR α apoptotic pathway is inhibited while autophagy is enhanced. The data suggest that PPAR α signaling might act as a molecular switch between apoptosis and autophagy thereby playing a critical role in adaptive process in cardiac hypertrophy.
Collapse
Affiliation(s)
- Ritu Kumari
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Aleepta Guha Ray
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dibyanti Mukherjee
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Vivek Chander
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Dipak Kar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Uppulapu Shravan Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Deepak Bharadwaj P.V.P.
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Sanjay K. Banerjee
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Aditya Konar
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Arun Bandyopadhyay
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- *Correspondence: Arun Bandyopadhyay ; orcid.org/0000-0002-4885-7033
| |
Collapse
|
14
|
Heidary Moghaddam R, Samimi Z, Asgary S, Mohammadi P, Hozeifi S, Hoseinzadeh-Chahkandak F, Xu S, Farzaei MH. Natural AMPK Activators in Cardiovascular Disease Prevention. Front Pharmacol 2022; 12:738420. [PMID: 35046800 PMCID: PMC8762275 DOI: 10.3389/fphar.2021.738420] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases (CVD), as a life-threatening global disease, is receiving worldwide attention. Seeking novel therapeutic strategies and agents is of utmost importance to curb CVD. AMP-activated protein kinase (AMPK) activators derived from natural products are promising agents for cardiovascular drug development owning to regulatory effects on physiological processes and diverse cardiometabolic disorders. In the past decade, different therapeutic agents from natural products and herbal medicines have been explored as good templates of AMPK activators. Hereby, we overviewed the role of AMPK signaling in the cardiovascular system, as well as evidence implicating AMPK activators as potential therapeutic tools. In the present review, efforts have been made to compile and update relevant information from both preclinical and clinical studies, which investigated the role of natural products as AMPK activators in cardiovascular therapeutics.
Collapse
Affiliation(s)
- Reza Heidary Moghaddam
- Clinical Research Development Center, Imam Ali and Taleghani Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zeinab Samimi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sedigheh Asgary
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute,.Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soroush Hozeifi
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Suowen Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Technology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Lu MC, Lee IT, Hong LZ, Ben-Arie E, Lin YH, Lin WT, Kao PY, Yang MD, Chan YC. Coffeeberry Activates the CaMKII/CREB/BDNF Pathway, Normalizes Autophagy and Apoptosis Signaling in Nonalcoholic Fatty Liver Rodent Model. Nutrients 2021; 13:nu13103652. [PMID: 34684653 PMCID: PMC8541094 DOI: 10.3390/nu13103652] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 12/02/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) shows extensive liver cell destruction with lipid accumulation, which is frequently accompanied by metabolic comorbidities and increases mortality. This study aimed to investigate the effects of coffeeberry (CB) on regulating the redox status, the CaMKII/CREB/BDNF pathway, autophagy, and apoptosis signaling by a NAFLD rodent model senescence-accelerated mice prone 8 (SAMP8). Three-month-old male SAMP8 mice were divided into a control group and three CB groups (50, 100, and 200 mg/kg BW), and fed for 12 weeks. The results show that CB reduced hepatic malondialdehyde and carbonyl protein levels. CB significantly enhanced Ca2+/calmodulin-dependent protein kinase II (CaMKII) and brain-derived neurotrophic factor (BDNF) and reduced the phospho-cAMP response element-binding protein (p-CREB)/CREB ratio. In addition, CB increased the silent information regulator T1 level, promoted Beclin 1 and microtubule-associated protein light chain 3 II expressions, and reduced phosphorylated mammalian target of rapamycin and its downstream p-p70s6k levels. CB also inhibited the expressions of apoptosis-related factors poly (ADP-ribose) polymerase-1 and the apoptosis-inducing factor. In conclusion, CB might protect the liver by reducing oxidative stress, activating the CaMKII/CREB/BDNF pathway, and improving autophagic and apoptotic expressions in a dose-dependent manner.
Collapse
Affiliation(s)
- Meng-Chun Lu
- Department of Clinical Nutrition, China Medical University Hospital, Taichung 406040, Taiwan;
- Department of Nutrition, China Medical University, Taichung 406040, Taiwan
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan; (Y.-H.L.); (W.-T.L.)
| | - I-Te Lee
- Division of Endocrinology and Metabolism, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Ling-Zong Hong
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705, Taiwan;
| | - Eyal Ben-Arie
- Graduate Institute of Acupuncture Science, Collage of Chinese Medicine, China Medical University, Taichung 406040, Taiwan;
| | - Yu-Hsuan Lin
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan; (Y.-H.L.); (W.-T.L.)
| | - Wei-Ting Lin
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan; (Y.-H.L.); (W.-T.L.)
| | - Pei-Yu Kao
- Division of Thoracic Surgery, Department of Surgery, China Medical University Hospital, Taichung 406040, Taiwan;
| | - Mei-Due Yang
- Division of General Surgery, Department of Surgery, China Medical University Hospital, Taichung 406040, Taiwan;
| | - Yin-Ching Chan
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan; (Y.-H.L.); (W.-T.L.)
- Correspondence:
| |
Collapse
|
16
|
Xu X, Su YL, Shi JY, Lu Q, Chen C. MicroRNA-17-5p Promotes Cardiac Hypertrophy by Targeting Mfn2 to Inhibit Autophagy. Cardiovasc Toxicol 2021; 21:759-771. [PMID: 34120306 DOI: 10.1007/s12012-021-09667-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 06/03/2021] [Indexed: 12/21/2022]
Abstract
Pathological cardiac hypertrophy is the leading cause of heart failure, and miRNAs have been recognized as key factors in cardiac hypertrophy. This study aimed to elucidate whether miR-17-5p affects cardiac hypertrophy by targeting the mitochondrial fusion protein mitofusin 2 (Mfn2)-mediated phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathway and regulating autophagy. miR-17-5p expression was shown to be upregulated both in vivo and in vitro. In addition, a miR-17-5p inhibitor significantly reversed AngII-induced cell hypertrophy in neonatal rat left ventricle myocytes (NRVMs). In contrast to miR-17-5p expression, Mfn2 expression was inhibited in rat hearts at 4 weeks after transverse aortic constriction (TAC) and in an Ang II-induced cell hypertrophy model. We examined miR-17-5p targeting of Mfn2 by dual luciferase reporter and Western blot assays. In addition, we also verified the relationship between Mfn2 and the PI3K/AKT/mTOR pathway. Mfn2 overexpression attenuated miR-17-5p-induced cell hypertrophy, and in rat myocardial tissue, miR-17-5p induced autophagy inhibition. In summary, the results of the present study demonstrated that miR-17-5p inhibits Mfn2 expression, activates the PI3K/AKT/mTOR pathway and suppresses autophagy to promote cardiac hypertrophy.
Collapse
Affiliation(s)
- Xuan Xu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Yi-Ling Su
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jia-Yu Shi
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Qi Lu
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Chu Chen
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
17
|
Li X, Li L, Lei W, Chua HZ, Li Z, Huang X, Wang Q, Li N, Zhang H. Traditional Chinese medicine as a therapeutic option for cardiac fibrosis: Pharmacology and mechanisms. Biomed Pharmacother 2021; 142:111979. [PMID: 34358754 DOI: 10.1016/j.biopha.2021.111979] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/05/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death worldwide and cardiac fibrosis is a common pathological process for cardiac remodeling in cardiovascular diseases. Cardiac fibrosis not only accelerates the deterioration progress of diseases but also becomes a pivotal contributor for futile treatment in clinical cardiovascular trials. Although cardiac fibrosis is common and prevalent, effective medicines to provide sufficient clinical intervention for cardiac fibrosis are still unavailable. Traditional Chinese medicine (TCM) is the natural essence experienced boiling, fry, and other processing methods, including active ingredients, extracts, and herbal formulas, which have been applied to treat human diseases for a long history. Recently, research has increasingly focused on the great potential of TCM for the prevention and treatment of cardiac fibrosis. Here, we aim to clarify the identified pro-fibrotic mechanisms and intensively summarize the application of TCM in improving cardiac fibrosis by working on these mechanisms. Through comprehensively analyzing, TCM mainly regulates the following pathways during ameliorating cardiac fibrosis: attenuation of inflammation and oxidative stress, inhibition of cardiac fibroblasts activation, reduction of extracellular matrix accumulation, modulation of the renin-angiotensin-aldosterone system, modulation of autophagy, regulation of metabolic-dependent mechanisms, and targeting microRNAs. We also discussed the deficiencies and the development direction of anti-fibrotic therapies on cardiac fibrosis. The data reviewed here demonstrates that TCM shows a robust effect on alleviating cardiac fibrosis, which provides us a rich source of new drugs or drug candidates. Besides, we also hope this review may give some enlightenment for treating cardiac fibrosis in clinical practice.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Lin Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Wei Lei
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Hui Zi Chua
- Evidence-Based Medicine Center, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Zining Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Xianglong Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin 300381, China.
| | - Qilong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Nan Li
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Han Zhang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Innovation Team of Research on Compound Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
18
|
Hautbergue T, Antigny F, Boët A, Haddad F, Masson B, Lambert M, Delaporte A, Menager JB, Savale L, Pavec JL, Fadel E, Humbert M, Junot C, Fenaille F, Colsch B, Mercier O. Right Ventricle Remodeling Metabolic Signature in Experimental Pulmonary Hypertension Models of Chronic Hypoxia and Monocrotaline Exposure. Cells 2021; 10:1559. [PMID: 34205639 PMCID: PMC8235667 DOI: 10.3390/cells10061559] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION Over time and despite optimal medical management of patients with pulmonary hypertension (PH), the right ventricle (RV) function deteriorates from an adaptive to maladaptive phenotype, leading to RV failure (RVF). Although RV function is well recognized as a prognostic factor of PH, no predictive factor of RVF episodes has been elucidated so far. We hypothesized that determining RV metabolic alterations could help to understand the mechanism link to the deterioration of RV function as well as help to identify new biomarkers of RV failure. METHODS In the current study, we aimed to characterize the metabolic reprogramming associated with the RV remodeling phenotype during experimental PH induced by chronic-hypoxia-(CH) exposure or monocrotaline-(MCT) exposure in rats. Three weeks after PH initiation, we hemodynamically characterized PH (echocardiography and RV catheterization), and then we used an untargeted metabolomics approach based on liquid chromatography coupled to high-resolution mass spectrometry to analyze RV and LV tissues in addition to plasma samples from MCT-PH and CH-PH rat models. RESULTS CH exposure induced adaptive RV phenotype as opposed to MCT exposure which induced maladaptive RV phenotype. We found that predominant alterations of arginine, pyrimidine, purine, and tryptophan metabolic pathways were detected on the heart (LV+RV) and plasma samples regardless of the PH model. Acetylspermidine, putrescine, guanidinoacetate RV biopsy levels, and cytosine, deoxycytidine, deoxyuridine, and plasmatic thymidine levels were correlated to RV function in the CH-PH model. It was less likely correlated in the MCT model. These pathways are well described to regulate cell proliferation, cell hypertrophy, and cardioprotection. These findings open novel research perspectives to find biomarkers for early detection of RV failure in PH.
Collapse
Affiliation(s)
- Thaïs Hautbergue
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Fabrice Antigny
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Angèle Boët
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Réanimation des Cardiopathies Congénitales, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - François Haddad
- Cardiovascular Medicine, Stanford Hospital, Stanford University, Stanford, CA 94305, USA;
| | - Bastien Masson
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Mélanie Lambert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
| | - Amélie Delaporte
- Service d’Anesthésie, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France;
| | - Jean-Baptiste Menager
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Laurent Savale
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Jérôme Le Pavec
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Elie Fadel
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| | - Marc Humbert
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Assistance Publique—Hôpitaux de Paris (AP-HP), Service de Pneumologie et Soins Intensifs Respiratoires, Centre de Référence de l’Hypertension Pulmonaire, Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Christophe Junot
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - François Fenaille
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Benoit Colsch
- Département Médicaments et Technologies pour la Santé (MTS), Université Paris-Saclay, CEA, INRAE, SPI, MetaboHUB, 91191 Gif-sur-Yvette, France; (T.H.); (C.J.); (F.F.); (B.C.)
| | - Olaf Mercier
- Faculté de Médecine, Université Paris-Saclay, 91191 Gif-sur-Yvette, France; (F.A.); (A.B.); (B.M.); (M.L.); (J.-B.M.); (L.S.); (J.L.P.); (E.F.); (M.H.)
- INSERM UMR_S 999 Hypertension Pulmonaire: Physiopathologie et Nouvelles Thérapies, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France
- Service de Chirurgie Thoracique, Vasculaire et Transplantation Cardio-Pulmonaire, Hôpital Marie Lannelongue, Groupe Hospitalier Paris Saint Joseph, 92350 Le Plessis-Robinson, France
| |
Collapse
|
19
|
Zullo A, Mancini FP, Schleip R, Wearing S, Klingler W. Fibrosis: Sirtuins at the checkpoints of myofibroblast differentiation and profibrotic activity. Wound Repair Regen 2021; 29:650-666. [PMID: 34077595 DOI: 10.1111/wrr.12943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 12/11/2022]
Abstract
Fibrotic diseases are still a serious concern for public health, due to their high prevalence, complex etiology and lack of successful treatments. Fibrosis consists of excessive accumulation of extracellular matrix components. As a result, the structure and function of tissues are impaired, thus potentially leading to organ failure and death in several chronic diseases. Myofibroblasts represent the principal cellular mediators of fibrosis, due to their extracellular matrix producing activity, and originate from different types of precursor cells, such as mesenchymal cells, epithelial cells and fibroblasts. Profibrotic activation of myofibroblasts can be triggered by a variety of mechanisms, including the transforming growth factor-β signalling pathway, which is a major factor driving fibrosis. Interestingly, preclinical and clinical studies showed that fibrotic degeneration can stop and even reverse by using specific antifibrotic treatments. Increasing scientific evidence is being accumulated about the role of sirtuins in modulating the molecular pathways responsible for the onset and development of fibrotic diseases. Sirtuins are NAD+ -dependent protein deacetylases that play a crucial role in several molecular pathways within the cells, many of which at the crossroad between health and disease. In this context, we will report the current knowledge supporting the role of sirtuins in the balance between healthy and diseased myofibroblast activity. In particular, we will address the signalling pathways and the molecular targets that trigger the differentiation and profibrotic activation of myofibroblasts and can be modulated by sirtuins.
Collapse
Affiliation(s)
- Alberto Zullo
- Department of Sciences and Technologies, Benevento, Italy.,CEINGE Advanced Biotechnologies s.c.a.r.l. Naples, Italy
| | | | - Robert Schleip
- Department of Sport and Health Sciences, Technical University Munich, Germany.,Fascia Research Group, Department of Neurosurgery, Ulm University, Germany.,Diploma University of Applied Sciences, Bad Sooden-Allendorf, Germany
| | - Scott Wearing
- Department of Sport and Health Sciences, Technical University Munich, Germany.,Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Werner Klingler
- Department of Sport and Health Sciences, Technical University Munich, Germany.,Fascia Research Group, Department of Neurosurgery, Ulm University, Germany.,Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia.,Department of Anaesthesiology, SRH Hospital Sigmaringen, Germany
| |
Collapse
|
20
|
Different types of cell death in vascular diseases. Mol Biol Rep 2021; 48:4687-4702. [PMID: 34013393 DOI: 10.1007/s11033-021-06402-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/08/2021] [Indexed: 10/21/2022]
Abstract
In a mature organism, tissue homeostasis is regulated by cell division and cell demise as the two major physiological procedures. There is increasing evidence that deregulation of these processes is important in the pathogenicity of main diseases, including myocardial infarction, stroke, atherosclerosis, and inflammatory diseases. Therefore, there are ongoing efforts to discover modulating factors of the cell cycle and cell demise planners aiming at shaping innovative therapeutically modalities to the therapy of such diseases. Although the life of a cell is terminated by several modes of action, a few cell deaths exist-some of which resemble apoptosis and/or necrosis, and most of them are different from one another-that contribute to a wide range of functions to either support or disrupt the homoeostasis. Even in normal physiological conditions, cell life is severe within the cardiovascular system. Cells are persistently undergoing stretch, contraction, injurious metabolic byproducts, and hemodynamic forces, and a few of cells sustain decade-long lifetimes. The duration of vascular disease causes further exposure of vascular cells to a novel range of offences, most of which induce cell death. There is growing evidence on consequences of direct damage to a cell, as well as on responses of adjacent and infiltrating cells, which also have an effect on the pathology. In this study, by focusing on different pathways of cell death in different vascular diseases, an attempt is made to open a new perspective on the therapeutic goals associated with cell death in these diseases.
Collapse
|
21
|
Liu BY, Li L, Liu GL, Ding W, Chang WG, Xu T, Ji XY, Zheng XX, Zhang J, Wang JX. Baicalein attenuates cardiac hypertrophy in mice via suppressing oxidative stress and activating autophagy in cardiomyocytes. Acta Pharmacol Sin 2021; 42:701-714. [PMID: 32796955 PMCID: PMC8115069 DOI: 10.1038/s41401-020-0496-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022] Open
Abstract
Baicalein is a natural flavonoid extracted from the root of Scutellaria baicalensis that exhibits a variety of pharmacological activities. In this study, we investigated the molecular mechanisms underlying the protective effect of baicalein against cardiac hypertrophy in vivo and in vitro. Cardiac hypertrophy was induced in mice by injection of isoproterenol (ISO, 30 mg·kg-1·d-1) for 15 days. The mice received caudal vein injection of baicalein (25 mg/kg) on 3rd, 6th, 9th, 12th, and 15th days. We showed that baicalein administration significantly attenuated ISO-induced cardiac hypertrophy and restored cardiac function. The protective effect of baicalein against cardiac hypertrophy was also observed in neonatal rat cardiomyocytes treated with ISO (10 μM). In cardiomyocytes, ISO treatment markedly increased reactive oxygen species (ROS) and inhibited autophagy, which were greatly alleviated by pretreatment with baicalein (30 μM). We found that baicalein pretreatment increased the expression of catalase and the mitophagy receptor FUN14 domain containing 1 (FUNDC1) to clear ROS and promote autophagy, thus attenuated ISO-induced cardiac hypertrophy. Furthermore, we revealed that baicalein bound to the transcription factor FOXO3a directly, promoting its transcription activity, and transactivated catalase and FUNDC1. In summary, our data provide new evidence for baicalein and FOXO3a in the regulation of ISO-induced cardiac hypertrophy. Baicalein has great potential for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Bing-Yan Liu
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Ling Li
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Gao-Li Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wei Ding
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266011, China
| | - Wen-Guang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Tao Xu
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xiao-Yu Ji
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xian-Xin Zheng
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jing Zhang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jian-Xun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China.
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China.
| |
Collapse
|
22
|
Rap1GAP Mediates Angiotensin II-Induced Cardiomyocyte Hypertrophy by Inhibiting Autophagy and Increasing Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7848027. [PMID: 33936386 PMCID: PMC8062190 DOI: 10.1155/2021/7848027] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 03/05/2021] [Accepted: 03/18/2021] [Indexed: 12/18/2022]
Abstract
Abnormal autophagy and oxidative stress contribute to angiotensin II- (Ang II-) induced cardiac hypertrophy and heart failure. We previously showed that Ang II increased Rap1GAP gene expression in cardiomyocytes associated with hypertrophy and autophagy disorders. Using real-time PCR and Western blot, we found that Rap1GAP expression was increased in the heart of Sprague Dawley (SD) rats infused by Ang II compared with saline infusion and in Ang II vs. vehicle-treated rat neonatal cardiomyocytes. Overexpression of Rap1GAP in cultured cardiomyocytes exacerbated Ang II-induced cardiomyocyte hypertrophy, reactive oxygen species (ROS) generation, and cell apoptosis and inhibited autophagy. The increased oxidative stress caused by Rap1GAP overexpression was inhibited by the treatment of autophagy agonists. Knockdown of Rap1GAP by siRNA markedly attenuated Ang II-induced cardiomyocyte hypertrophy and oxidative stress and enhanced autophagy. The AMPK/AKT/mTOR signaling pathway was inhibited by overexpression of Rap1GAP and activated by the knockdown of Rap1GAP. These results show that Rap1GAP-mediated pathway might be a new mechanism of Ang II-induced cardiomyocyte hypertrophy, which could be a potential target for the future treatment of cardiac hypertrophy and heart failure.
Collapse
|
23
|
Jiang X, Shao M, Liu X, Liu X, Zhang X, Wang Y, Yin K, Wang S, Hu Y, Jose PA, Zhou Z, Xu F, Yang Z. Reversible Treatment of Pressure Overload-Induced Left Ventricular Hypertrophy through Drd5 Nucleic Acid Delivery Mediated by Functional Polyaminoglycoside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003706. [PMID: 33717857 PMCID: PMC7927605 DOI: 10.1002/advs.202003706] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Indexed: 05/12/2023]
Abstract
Left ventricular hypertrophy and fibrosis are major risk factors for heart failure, which require timely and effective treatment. Genetic therapy has been shown to ameliorate hypertrophic cardiac damage. In this study, it is found that in mice, the dopamine D5 receptor (D5R) expression in the left ventricle (LV) progressively decreases with worsening of transverse aortic constriction-induced left ventricular hypertrophy. Then, a reversible treatment of left ventricular hypertrophy with Drd5 nucleic acids delivered by tobramycin-based hyperbranched polyaminoglycoside (SS-HPT) is studied. The heart-specific increase in D5R expression by SS-HPT/Drd5 plasmid in the early stage of left ventricular hypertrophy attenuates cardiac hypertrophy and fibrosis by preventing oxidative and endoplasmic reticulum (ER) stress and ameliorating autophagic dysregulation. By contrast, SS-HPT/Drd5 siRNA promotes the progression of left ventricular hypertrophy and accelerates the deterioration of myocardial function into heart failure. The reduction in cardiac D5R expression and dysregulated autophagy are observed in patients with hypertrophic cardiomyopathy and heart failure. The data show a cardiac-specific beneficial effect of SS-HPT/Drd5 plasmid on myocardial remodeling and dysfunction, which may provide an effective therapy of patients with left ventricular hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Meiyu Shao
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xue Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xing Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xu Zhang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Yuming Wang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Kunlun Yin
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Shuiyun Wang
- Department of Cardiovascular SurgeryState Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Pedro A Jose
- Department of Pharmacology and PhysiologyThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
- Department of MedicineDivision of Kidney Diseases & HypertensionThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Fu‐Jian Xu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Zhiwei Yang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| |
Collapse
|
24
|
He Y, Ao N, Yang J, Wang X, Jin S, Du J. The preventive effect of liraglutide on the lipotoxic liver injury via increasing autophagy. Ann Hepatol 2021; 19:44-52. [PMID: 31787541 DOI: 10.1016/j.aohep.2019.06.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND OBJECTIVES The incidence of non-alcoholic fatty liver disease (NAFLD) is increasing. Previous studies indicated that Liraglutide, glucagon-like peptide-1 analogue, could regulate glucose homeostasis as a valuable treatment for Type 2 Diabetes. However, the precise effect of Liraglutide on NAFLD model in rats and the mechanism remains unknown. In this study, we investigated the molecular mechanism by which Liraglutide ameliorates hepatic steatosis in a high-fat diet (HFD)-induced rat model of NAFLD in vivo and in vitro. MATERIALS AND METHODS NALFD rat models and hepatocyte steatosis in HepG2 cells were induced by HFD and palmitate fatty acid treatment, respectively. AMPK inhibitor, Compound C was added in HepG2 cells. Autophagy-related proteins LC3, Beclin1 and Atg7, and AMPK pathway-associated proteins were evaluated by Western blot and RT-PCR. RESULTS Liraglutide enhanced autophagy as showed by the increased expression of the autophagy markers LC3, Beclin1 and Atg7 in HFD rats and HepG2 cells treated with palmitate fatty acid. In vitro, The AMPK inhibitor exhibited an inhibitory effect on Liraglutide-induced autophagy enhancement with the deceased expression of LC3, Beclin1 and Atg7. Additionally, Liraglutide treatment elevated AMPK levels and TSC1, decreased p-mTOR expression. CONCLUSIONS Liraglutide could upregulate autophagy to decrease lipid over-accumulation via the AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Yini He
- Department of General Practice, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Ao
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jing Yang
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaochen Wang
- Department of Endocrinology, The People's Hospital of Liaoning Province, Shenyang, Liaoning, China
| | - Shi Jin
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jian Du
- Department of Endocrinology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
25
|
Miranda-Silva D, G Rodrigues P, Alves E, Rizo D, Fonseca ACRG, Lima T, Baganha F, Conceição G, Sousa C, Gonçalves A, Miranda I, Vasques-Nóvoa F, Magalhães J, Leite-Moreira A, Falcão-Pires I. Mitochondrial Reversible Changes Determine Diastolic Function Adaptations During Myocardial (Reverse) Remodeling. Circ Heart Fail 2020; 13:e006170. [PMID: 33176457 DOI: 10.1161/circheartfailure.119.006170] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Often, pressure overload-induced myocardial remodeling does not undergo complete reverse remodeling after decreasing afterload. Recently, mitochondrial abnormalities and oxidative stress have been successively implicated in the pathogenesis of several chronic pressure overload cardiac diseases. Therefore, we aim to clarify the myocardial energetic dysregulation in (reverse) remodeling, mainly focusing on the mitochondria. METHODS Thirty-five Wistar Han male rats randomly underwent sham or ascending (supravalvular) aortic banding procedure. Echocardiography revealed that banding induced concentric hypertrophy and diastolic dysfunction (early diastolic transmitral flow velocity to peak early-diastolic annular velocity ratio, E/E': sham, 13.6±2.1, banding, 18.5±4.1, P=0.014) accompanied by increased oxidative stress (dihydroethidium fluorescence: sham, 1.6×108±6.1×107, banding, 2.6×108±4.5×107, P<0.001) and augmented mitochondrial function. After 8 to 9 weeks, half of the banding animals underwent overload relief by an aortic debanding surgery (n=10). RESULTS Two weeks later, hypertrophy decreased with the decline of oxidative stress (dihydroethidium fluorescence: banding, 2.6×108±4.5×107, debanding, 1.96×108±6.8×107, P<0.001) and diastolic dysfunction improved simultaneously (E/E': banding, 18.5±4.1, debanding, 15.1±1.8, P=0.029). The reduction of energetic demands imposed by overload relief allowed the mitochondria to reduce its activity and myocardial levels of phosphocreatine, phosphocreatine/ATP, and ATP/ADP to normalize in debanding towards sham values (phosphocreatine: sham, 38.4±7.4, debanding, 35.6±8.7, P=0.71; phosphocreatine/ATP: sham, 1.22±0.23 debanding, 1.11±0.24, P=0.59; ATP/ADP: sham, 6.2±0.9, debanding, 5.6±1.6, P=0.66). Despite the decreased mitochondrial area, complex III and V expression increased in debanding compared with sham or banding. Autophagy and mitophagy-related markers increased in banding and remained higher in debanding rats. CONCLUSIONS During compensatory and maladaptive hypertrophy, mitochondria become more active. However, as the disease progresses, the myocardial energetic demands increase and the myocardium becomes energy deficient. During reverse remodeling, the concomitant attenuation of cardiac hypertrophy and oxidative stress allowed myocardial energetics, left ventricle hypertrophy, and diastolic dysfunction to recover. Autophagy and mitophagy are probably involved in the myocardial adaptation to overload and to unload. We conclude that these mitochondrial reversible changes underlie diastolic function adaptations during myocardial (reverse) remodeling.
Collapse
Affiliation(s)
- Daniela Miranda-Silva
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Patrícia G Rodrigues
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Estela Alves
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - David Rizo
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Ana Catarina R G Fonseca
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Portugal (A.C.R.G.F.)
| | - Tânia Lima
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Fabiana Baganha
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Gloria Conceição
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Cláudia Sousa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Alexandre Gonçalves
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Isabel Miranda
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Francisco Vasques-Nóvoa
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - José Magalhães
- LaMetEX, Laboratory of Metabolism and Exercise (E.A., D.R., J.M.).,CIAFEL, Research Centre in Physical Activity, Health and Leisure, Faculty of Sports, Portugal (E.A., D.R., J.M.)
| | - Adelino Leite-Moreira
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Porto, Portugal (D.M.S., P.G.R., T.L., F.B., G.C., C.S., A.G., I.M., F.V.-N., A.L.-M., I.F.-P.)
| |
Collapse
|
26
|
Nandi SS, Katsurada K, Sharma NM, Anderson DR, Mahata SK, Patel KP. MMP9 inhibition increases autophagic flux in chronic heart failure. Am J Physiol Heart Circ Physiol 2020; 319:H1414-H1437. [PMID: 33064567 DOI: 10.1152/ajpheart.00032.2020] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Increased matrix metalloprotease 9 (MMP9) after myocardial infarction (MI) exacerbates ischemia-induced chronic heart failure (CHF). Autophagy is cardioprotective during CHF; however, whether increased MMP9 suppresses autophagic activity in CHF is unknown. This study aimed to determine whether increased MMP9 suppressed autophagic flux and MMP9 inhibition increased autophagic flux in the heart of rats with post-MI CHF. Sprague-Dawley rats underwent either sham surgery or coronary artery ligation 6-8 wk before being treated with MMP9 inhibitor for 7 days, followed by cardiac autophagic flux measurement with lysosomal inhibitor bafilomycin A1. Furthermore, autophagic flux was measured in vitro by treating H9c2 cardiomyocytes with two independent pharmacological MMP9 inhibitors, salvianolic acid B (SalB) and MMP9 inhibitor-I, and CRISPR/cas9-mediated MMP9 genetic ablation. CHF rats showed cardiac infarct, significantly increased left ventricular end-diastolic pressure (LVEDP), and increased MMP9 activity and fibrosis in the peri-infarct areas of left ventricular myocardium. Measurement of the autophagic markers LC3B-II and p62 with lysosomal inhibition showed decreased autophagic flux in the peri-infarct myocardium. Treatment with SalB for 7 days in CHF rats decreased MMP9 activity and cardiac fibrosis but increased autophagic flux in the peri-infarct myocardium. As an in vitro corollary study, measurement of autophagic flux in H9c2 cardiomyocytes and fibroblasts showed that pharmacological inhibition or genetic ablation of MMP9 upregulates autophagic flux. These data are consistent with our observations that MMP9 inhibition upregulates autophagic flux in the heart of rats with CHF. In conclusion, the results in this study suggest that the beneficial outcome of MMP9 inhibition in pathological cardiac remodeling is in part mediated by improved autophagic flux.NEW & NOTEWORTHY This study elucidates that the improved cardiac extracellular matrix (ECM) remodeling and cardioprotective effect of matrix metalloprotease 9 (MMP9) inhibition in chronic heart failure (CHF) are via increased autophagic flux. Autophagy is cardioprotective; however, the mechanism of autophagy suppression in CHF is unknown. We for the first time demonstrated here that increased MMP9 suppressed cardiac autophagy and ablation of MMP9 increased cardiac autophagic flux in CHF rats. Restoring the physiological level of autophagy in the failing heart is a challenge, and our study addressed this challenge. The novelty and highlights of this report are as follows: 1) MMP9 regulates cardiomyocyte and fibroblast autophagy, 2) MMP9 inhibition protects CHF after myocardial infarction (MI) via increased cardiac autophagic flux, 3) MMP9 inhibition increased cardiac autophagy via activation of AMP-activated protein kinase (AMPK)α, Beclin-1, Atg7 pathway and suppressed mechanistic target of rapamycin (mTOR) pathway.
Collapse
Affiliation(s)
- Shyam S Nandi
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kenichi Katsurada
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Neeru M Sharma
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Daniel R Anderson
- Department of Cardiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil K Mahata
- Department of Medicine, Metabolic Physiology and Ultrastructural Biology Laboratory, University of California, San Diego, California.,Department of Veterans Affairs San Diego Healthcare System, San Diego, California
| | - Kaushik P Patel
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
27
|
Zheng D, Zhang M, Liu T, Zhou T, Shen A. Osteoprotegerin prompts cardiomyocyte hypertrophy via autophagy inhibition mediated by FAK/BECLIN1 pathway. Life Sci 2020; 264:118550. [PMID: 33035582 DOI: 10.1016/j.lfs.2020.118550] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/17/2020] [Accepted: 09/30/2020] [Indexed: 10/23/2022]
Abstract
AIM It has been reported that Osteoprotegerin (OPG) induces cardiomyocyte hypertrophy, but the mechanism remains unclear. This study was to investigate the role of Focal Adhesion Kinase (FAK) pathway in the OPG induced hypertrophy in cultured cardiomyocytes. METHODS The H9C2 line of rat cardiomyocytes were treated with OPG at different concentrations and the cellular hypertrophy was evaluated. Meanwhile, the activity of FAK and other the phosphorylation kinases were detected. Autophagy flux assay was performed in absence and presence OPG. The interaction between proteins was analyses using Co-Immunoprecipitation assay. RESULTS We found that OPG induced cardiomyocyte hypertrophic response, indicated by increased cellular size and protein content per cell. OPG increases the heart/body weight ratio in vivo. Also OPG inhibits autophagy and induces FAK phosphorylation. FAK silencing using si-RNA abrogates the effect of OPG on autophagy and cellular hypertrophy. Furthermore, Co-immunoprecipitation assay reveals that OPG inhibits autophagy through enhancing the binding of FAK and Beclin1. CONCLUSION The FAK/Beclin1 signal pathway is essential for the OPG induced autophagy inhibition and hypertrophic response in cultured H9C2 cells.
Collapse
Affiliation(s)
- Dezhong Zheng
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Tianhe District Guangzhou, 510630 Guangzhou, Guangdong, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Mingyu Zhang
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Tianhe District Guangzhou, 510630 Guangzhou, Guangdong, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Tingrong Liu
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Tianhe District Guangzhou, 510630 Guangzhou, Guangdong, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China
| | - Tao Zhou
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Tianhe District Guangzhou, 510630 Guangzhou, Guangdong, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China.
| | - Anna Shen
- Department of Cardiology, The Third Affiliated Hospital of Southern Medical University, No.183, West Zhongshan Ave, Tianhe District Guangzhou, 510630 Guangzhou, Guangdong, China; Key Laboratory for Organ Failure Research, Ministry of Education of the People's Republic of China, Guangzhou, China; The Third School of Clinical Medicine, Southern Medical University.
| |
Collapse
|
28
|
Ma W, Liang F, Zhan H, Jiang X, Gao C, Zhang X, Zhang K, Sun Q, Hu H, Zhao Z. Activated FMS-like tyrosine kinase 3 ameliorates angiotensin II-induced cardiac remodelling. Acta Physiol (Oxf) 2020; 230:e13519. [PMID: 32480429 DOI: 10.1111/apha.13519] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022]
Abstract
AIM FMS-like receptor tyrosine kinase 3 (Flt3) has been reported to be increased in cardiomyocytes responding to ischaemic stress. This study was to determine whether Flt3 activation could ameliorate pressure overload-induced heart hypertrophy and fibrosis, and to elucidate the mechanisms of action. METHODS In vivo cardiac hypertrophy and remodelling experiments were conducted by infusing angiotensin II (Ang II) chronically in male C57BL/6 mice. Flt3-specific ligand (FL) was administered intraperitoneally every two days (5 µg/mouse). In vitro experiments on hypertrophy, apoptosis and autophagy mechanism were performed in neonatal rat cardiomyocytes (NRCMs) and H9c2 cells with adenovirus vector-mediated overexpression of Flt3. RESULTS Our results demonstrated that following chronic Ang II infusion for 4 weeks, the mice exhibited heart hypertrophy, fibrosis, apoptosis and contractile dysfunction. Meanwhile, Ang II induced autophagic responses in mouse hearts, as evidenced by increased LC3 II and decreased P62 expression. These pathological alterations in Ang II-treated mice were significantly ameliorated by Flt3 activation with FL administration. In NRCMs and Flt3-overexpressed H9c2 cells, FL attenuated Ang II-induced pathological autophagy and inactivated AMPK/mTORC1/FoxO3a signalling, thereby efficiently mitigating cell hypertrophy and apoptosis. Conversely, the AMPK activator metformin or the mTORC1 inhibitor rapamycin reversed the effects of FL on the alterations of autophagy, hypertrophy and apoptosis in cardiomyocytes induced by Ang II. CONCLUSION Flt3 activation ameliorates cardiac hypertrophy, fibrosis and contractile dysfunction in the mouse model of chronic pressure overload, most likely via suppressing AMPK/mTORC1/FoxO3a-mediated autophagy. These results provide new evidence supporting Flt3 as a novel therapeutic target in maladaptive cardiac remodelling.
Collapse
Affiliation(s)
- Wenzhuo Ma
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Fanfan Liang
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Heqin Zhan
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Department of Pharmacology College of Pharmacy Xinxiang Medical University Xinxiang Henan China
| | - Xixi Jiang
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Chenying Gao
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Xin Zhang
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Kaina Zhang
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Qiang Sun
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Hao Hu
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| | - Zhenghang Zhao
- Department of Pharmacology School of Basic Medicine Sciences Xi'an Jiaotong University Health Science Center Xi’an Shaanxi China
- Key Laboratory of Environment and Genes Related to Diseases Ministry of Education Xi'an Jiaotong University Xi’an Shaanxi China
| |
Collapse
|
29
|
Zhang T, Wang H, Lu M, Zhao K, Yin J, Liu Y, Sun Y. Astragaloside IV prevents myocardial hypertrophy induced by mechanical stress by activating autophagy and reducing inflammation. Am J Transl Res 2020; 12:5332-5342. [PMID: 33042423 PMCID: PMC7540164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 08/01/2020] [Indexed: 06/11/2023]
Abstract
AIMS The aim of the present study was to investigate the effects of astragaloside IV (As-IV) on mechanical stress-induced myocardial hypertrophy, with a focus on autophagy and inflammation. METHODS A rat cardiac hypertrophy model was established by narrowing the abdominal aorta, and a cell hypertrophy model was established by mechanically stretching primary cardiomyocytes. Cardiac function index and cardiac hypertrophy were measured by echocardiography, heart weight index (HWI) and left ventriculus weight index (LVWI) in vivo. Cell size was measured by phalloidin-tetramethyl treatment in vitro, while hematoxylin and eosin (HE) staining was used to observe the arrangement and morphology of myocardial cells. The expression of ANP, BNP, LC3II, p62, NLRP3, and IL-1β in both myocardial tissue and cardiomyocytes was assessed by Western blot, while TNF-α and IL-18 levels in serum and cell supernatants were measured by ELISA. RESULTS In the aortic banding model, the cardiac function index LVEF was decreased; the hypertrophy indexes LVPWd, LVPWs, IVSd and IVSs were significantly increased; cardiomyocytes were enlarged and disordered; the expression levels of ANP, BNP, NLRP3, IL-1β and p62 were increased; and LC3II expression was decreased in both myocardial tissue and cardiomyocytes. As-IV could significantly improve cardiac function and cardiomyocyte morphology and limit hypertrophy, thereby protecting damaged hearts, while rapamycin had a similar effect as As-IV. In addition, As-IV decreased the expression of NLRP3 and IL-1β and activated autophagy, as evidenced by increased LC3II expression and decreased p62 levels. CONCLUSION As-IV prevents myocardial hypertrophy induced by mechanical stress by activating autophagy and reducing inflammation.
Collapse
Affiliation(s)
- Tong Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
| | - Hongxin Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
| | - Meili Lu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
| | - Kun Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Jiawei Yin
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
| | - Yang Liu
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
- The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou 121001, China
| | - Yang Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Drug Research of Liaoning Province, Jinzhou Medical UniversityJinzhou 121001, China
| |
Collapse
|
30
|
Pang K, Li B, Tang Z, Yang W, Hao L, Shi Z, Zhang J, Cai L, Li R, Liu Y, Lv Q, Ding J, Han C. Resveratrol inhibits hypertrophic scars formation by activating autophagy via the miR-4654/Rheb axis. Mol Med Rep 2020; 22:3440-3452. [PMID: 32945452 PMCID: PMC7453609 DOI: 10.3892/mmr.2020.11407] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Hypertrophic scars (HSs) are a type of pathological scar which are induced by surgery, burn injuries or trauma during the healing process. Due to the high recurrence rates and strong invasive properties, HSs have become a major clinical issue. Resveratrol has been identified as a potential agent to suppress scar formation; however, the underlying mechanism of action remains unclear. Therefore, the present study aimed to investigate the effect of resveratrol on HS-derived fibroblasts (HSFBs) in vitro. MTT assay was performed to evaluate cell viability following the resveratrol treatment. Western blot and RT-qPCR analysis was used to identify the expression levels and the relationship among autophagic markers, miR-4654 and resveratrol treatment. Finally, GFP-LC3 stable HSFBs cells were generated to further assess the effect of resveratrol. The results revealed that resveratrol significantly induced cell death in a dose-dependent manner and induced autophagy by downregulating the expression levels of Rheb in HSFBs. Notably, microRNA-4654 (miR-4654) was significantly decreased in the HSFBs and re-upregulated by resveratrol treatment dose-dependently. Through the bioinformatic analysis and luciferase assay, miR-4654 was identified to directly target Rheb. Transfection studies showed that miR-4654 negative correlated with Rheb expression, suggesting that the autophagic process may be altered by the miR-4654/Rheb axis under the control of resveratrol. In conclusion, the results of the present study suggested that resveratrol may promote autophagy by upregulating miR-4654, which in turn may suppress Rheb expression via directly binding to the 3′-untranslated region of Rheb. These findings provided a novel insight into the development of potential therapeutic targets for HSs.
Collapse
Affiliation(s)
- Kun Pang
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Bibo Li
- Department of Urology, Taizhou Hospital Affiliated to Nanjing University of Chinese Medicine, Taizhou, Jiangsu 225300, P.R. China
| | - Zhiming Tang
- Department of Dermatology, Xuzhou Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Xuzhou, Jiangsu 221009, P.R. China
| | - Wen Yang
- Department of Renal Disease, Shandong First Medical University, Tai'an, Shandong 271016, P.R. China
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Zhenduo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Jianjun Zhang
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Longjun Cai
- Department of Urology, Suqian People's Hospital of Nanjing Drum-Tower Hospital Group, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, Jiangsu 223800, P.R. China
| | - Rui Li
- Department of Burns and Plastic Surgery, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Ying Liu
- Department of Burns and Plastic Surgery, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Qian Lv
- Department of Burns and Plastic Surgery, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Jicun Ding
- Department of Burns and Plastic Surgery, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| | - Conghui Han
- Department of Urology, Xuzhou Central Hospital, Xuzhou Clinical College Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu 221009, P.R. China
| |
Collapse
|
31
|
Lin YB, Huang DJ, Huang HL, Chen DX, Huang JH. Sophocarpine ameliorates cardiac hypertrophy through activation of autophagic responses. Biosci Biotechnol Biochem 2020; 84:2054-2061. [PMID: 32544026 DOI: 10.1080/09168451.2020.1780111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Mounting evidences indicate that autophagy is an essential homeostatic mechanism to maintain the global cardiac structure function. Sophocarpine (SOP), a major bioactive compound derived from the natural plant Sophora flavescens. However, the role of SOP in cardiac hypertrophy remain to be fully elucidated. In the present study, we tested the hypothesis that SOP protects against Ang II-induced cardiac hypertrophy by mediating the regulation of autophagy. The results demonstrated that SOP attenuated the Ang II-induced cardiac hypertrophy, as assessed by measurements of echocardiography parameters, the ratios of heart weight/body weight and left ventricle weight/body weight, histopathological staining, cross-sectional cardiomyocyte area, and the expression levels of cardiac hypertrophic markers. The anti-hypertrophic effect of SOP was mediated by activating autophagy-related pathway, as revealed by reversal of the increased autophagy marker protein expression. These findings reveal a novel mechanism of SOP attenuating cardiac hypertrophy via activating autophagy-related signaling pathways.
Collapse
Affiliation(s)
- Yue-Bao Lin
- Department of General Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Dong-Jian Huang
- Department of Intensive Care Unit, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Huan-Liang Huang
- Department of Emergency, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - De-Xiong Chen
- Department of General Medicine, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| | - Jiong-Hua Huang
- Department of Vasculocardiology, The Third Affiliated Hospital of Guangzhou Medical University , Guangzhou, China
| |
Collapse
|
32
|
Yan W, Dong ZC, Wang JJ, Zhang YL, Wang HX, Zhang B, Li HH. Deficiency of the Immunoproteasome LMP10 Subunit Attenuates Angiotensin II-Induced Cardiac Hypertrophic Remodeling via Autophagic Degradation of gp130 and IGF1R. Front Physiol 2020; 11:625. [PMID: 32581853 PMCID: PMC7296172 DOI: 10.3389/fphys.2020.00625] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/18/2020] [Indexed: 01/26/2023] Open
Abstract
Background/Aim Hypertensive cardiac hypertrophy is the leading cause of cardiac remodeling and heart failure. We recently demonstrated that the immunoproteasome, an inducible form of the constitutive proteasome, plays a critical role in regulating cardiovascular diseases. However, the role of the immunoproteasome LMP10 (β2i) catalytic subunit in the regulation of angiotensin II (Ang II)-induced cardiac hypertrophic remodeling remains unclear. Methods Wild-type (WT) and LMP10 knockout (KO) mice were infused with Ang II 1,000 ng/kg/min for 2 weeks. Blood pressure was measured using a tail-cuff system. Cardiac function and hypertrophic remodeling were examined by echocardiography and histological staining. The expression levels of genes and proteins were examined with quantitative real-time PCR and immunoblotting analysis, respectively. Results LMP10 mRNA and protein expression was significantly increased in Ang II-stimulated hearts and primary cardiomyocytes. Moreover, Ang II infusion for 2 weeks increased systolic blood pressure, abnormal cardiac function, hypertrophy, fibrosis, and inflammation in WT mice, which were significantly reversed in KO mice. Moreover, a marked reduction in the protein levels of insulin growth factor-1 receptor (IGF1R), glycoprotein 130 (gp130), and phosphorylated AKT, mTOR, STAT3, and ERK1/2 and an increase in the LC3II/I ratio were also observed in LMP10 KO mice compared with WT mice after Ang II infusion. In vitro culture experiments confirmed that LMP10 knockdown activated autophagy and increased IGF1R and gp130 degradation, leading to the inhibition of cardiomyocyte hypertrophy. However, inhibiting autophagy with chloroquine reversed this effect. Conclusion The results of this study indicate that LMP10 KO attenuates Ang II-induced cardiac hypertrophic remodeling via the autophagy-dependent degradation of IGF1R and gp130, and suggests that LMP10 may be a novel therapeutic target for hypertrophic heart diseases.
Collapse
Affiliation(s)
- Wen Yan
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhi-Chao Dong
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jing-Jing Wang
- Department of Laboratory Animal Sciences, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yun-Long Zhang
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hong-Xia Wang
- Department of Physiology and Physiopathology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Bo Zhang
- Department of Cardiology, Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
33
|
Tang L, Yu X, Zheng Y, Zhou N. Inhibiting SLC26A4 reverses cardiac hypertrophy in H9C2 cells and in rats. PeerJ 2020; 8:e8253. [PMID: 31998553 PMCID: PMC6979409 DOI: 10.7717/peerj.8253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 11/20/2019] [Indexed: 11/20/2022] Open
Abstract
Background It has been confirmed that mutations in solute carrier family 26 member 4 (SLC26A4) contribute to pendred syndrome. However, the role of SLC26A4 in cardiac hypertrophy and the signaling pathways remain unclear. Methods Cardiomyocytes were treated by 200 µM phenylephrine (PE) to induce cardiac hypertrophy. Also, the expression of SLC26A4, GSK3, cardiac hypertrophy markers including atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) was detected through real-time quantitative polymerase chain reaction (RT-qPCR). Flow cytometry assay was used to test the apoptosis of PE-induced cardiomyocytes transfected by small interfere RNA (siRNA)-SLC26A4. Furthermore, we detected the expression of autophagy-related markers including light chain 3 (LC3) and P62. Finally, we established a rat model of abdominal aortic constriction (AAC)-induced cardiac hypertrophy in vivo. Results RT-qPCR results showed that the mRNA expression of SLC26A4 was significantly up-regulated in PE-induced cardiac hypertrophy. After inhibiting SLC26A4, the release of ANP and BNP was significantly decreased and GSK3β was elevated in vivo and in vitro. Furthermore, inhibiting SLC26A4 promoted apoptosis of cardiac hypertrophy cells. In addition, LC3 was down-regulated and P62 was enhanced after transfection of siRNA-SLC26A4. Conclusion Our findings revealed that SLC26A4 increases cardiac hypertrophy, and inhibiting SLC26A4 could decrease the release of ANP/BNP and promote the expression of GSK-3β in vitro and in vivo. Moreover, SLC26A4 silencing inhibits autophagy of cardiomyocytes and induces apoptosis of cardiomyocytes. Therefore, SLC26A4 possesses potential value to be a therapeutic target of cardiac hypertrophy, and our study provides new insights into the mechanisms of cardiac hypertrophy.
Collapse
Affiliation(s)
- Liqun Tang
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoqin Yu
- Department of Geriatrics, Zhejiang Aid Hospital, Hangzhou, Zhejiang, China
| | - Yangyang Zheng
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ning Zhou
- Department of Geriatrics, Zhejiang Province People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
34
|
Zhao Z, Liu H, Guo D. Aliskiren attenuates cardiac dysfunction by modulation of the mTOR and apoptosis pathways. ACTA ACUST UNITED AC 2020; 53:e8793. [PMID: 31994601 PMCID: PMC6984373 DOI: 10.1590/1414-431x20198793] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
Aliskiren (ALS) is well known for its antihypertensive properties. However, the potential underlying the molecular mechanism and the anti-hypertrophic effect of ALS have not yet been fully elucidated. The aim of the present study was to investigate the role of ALS in mammalian target of rapamycin (mTOR) and apoptosis signaling using in vivo and in vitro models of cardiac hypertrophy. A rat model of cardiac hypertrophy was induced by isoproterenol treatment (5 mg·kg-1·day-1) for 4 weeks, with or without ALS treatment at 20 mg·kg-1·day-1. The expression of hypertrophic, fibrotic, and apoptotic markers was determined by RT-qPCR. The protein expression of apoptotic markers mTOR and p-mTOR was assessed by western blot analysis. The proliferation of H9C2 cells was monitored using the MTS assay. Cell apoptosis was analyzed using flow cytometry. In vivo, isoproterenol-treated rats exhibited worse cardiac function, whereas ALS treatment reversed these dysfunctions, which were associated with changes in p-mTOR, Bcl-2, Bax, and cleaved caspase-3 expression, as well as the number of apoptotic cells. In vitro, H9C2 cardiomyocyte viability was significantly inhibited and cardiac hypertrophy was induced by Ang II administration, but ALS reversed Ang II-induced H9C2 cardiomyocyte hypertrophy and death. Furthermore, Ang II triggered the activation of the mTOR and apoptosis pathways in hypertrophic cardiomyocytes that were inhibited by ALS treatment. These results indicated that ALS alleviated cardiac hypertrophy through inhibition of the mTOR and apoptosis pathways in cardiomyocytes.
Collapse
Affiliation(s)
- Zhengbo Zhao
- Department of Cardiovascular Medicine, Jiulongpo District People's Hospital, Chongqing, China
| | - Han Liu
- Department of Neurology, Jiulongpo District People's Hospital, Chongqing, China
| | - Dongmei Guo
- Department of Cardiovascular Medicine, Nanchuan District People's Hospital, Chongqing, China
| |
Collapse
|
35
|
Shao J, Lin W, Lin B, Wang Q, Chen Y, Fan C. MiR-377 accelerates cardiac hypertrophy by inhibiting autophagy via targeting PPAR γ. ALL LIFE 2020. [DOI: 10.1080/26895293.2020.1808083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Affiliation(s)
- Jianzhi Shao
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Wenhui Lin
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Bin Lin
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Qizeng Wang
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Yunpeng Chen
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| | - Chenrong Fan
- Department of Cardiology, The First People’s Hospital of Wenling, Wenling, People’s Republic of China
| |
Collapse
|
36
|
Dai J, Ma J, Liao Y, Luo X, Chen G. Polydatin protects H9c2 cells from hypoxia-induced injury via up-regulating long non-coding RNA DGCR5. ACTA ACUST UNITED AC 2019; 52:e8834. [PMID: 31826181 PMCID: PMC6903803 DOI: 10.1590/1414-431x20198834] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022]
Abstract
Polydatin (PD), a monocrystalline polyphenolic drug mainly found in the roots of Polygonum cuspidatum, has various pharmacological activities. Long non-coding RNAs (lncRNA) DiGeorge syndrome critical region gene 5 (DGCR5) was found to participate in the suppression of multiple cancers. Here, we proposed to study the effect of PD on myocardial infarction (MI) by inducing DGCR5. CCK-8 assay was performed to detect the viability of H9c2 cells. Flow cytometry was utilized to test apoptosis of H9c2 cells. These results determined the optimal concentration and effect time of hypoxia as well as PD. Si-DGCR5 was transfected into cells and the expression level was determined by qRT-PCR. Western blot was utilized to evaluate the expression of apoptosis-related proteins, Bcl-2, Bax, and cleaved-caspase-3, as well as autophagy-associated proteins including Beclin-1, p62, and LC3-II/LC3-I. As a result, PD efficiently attenuated hypoxia-induced apoptosis and autophagy in H9c2 cells. The expression of DGCR5 was down-regulated by hypoxia and up-regulated by PD. Besides, knocking-down the expression of DGCR5 inhibited the protection of PD in H9c2 cells. In addition, PD up-regulated the accumulation of DGCR5, DGCR5 decreased the expression of Bcl-2 and p62, raised the expression of Bax and cleaved-caspase-3, and the proportion of LC3-II/LC3-I. PD stimulated the PI3K/AKT/mTOR and MEK/ERK signaling pathways via up-regulating the expression of DGCR5. Our data demonstrated that PD reduced cell apoptosis and autophagy induced by hypoxia in cardiomyocytes. Moreover, PD activated PI3K/AKT/mTOR and MEK/ERK signaling pathways by up-regulating the expression of DGCR5.
Collapse
Affiliation(s)
- Jinhua Dai
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Jianbo Ma
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Yufeng Liao
- Department of Clinical Laboratory, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| | - Xianhai Luo
- Department of Clinical Laboratory, Ningbo Kangning Hospital, Ningbo Mental Health Center, Ningbo, Zhejiang, China
| | - Guofang Chen
- Department of Cardiology, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, Zhejiang, China
| |
Collapse
|
37
|
Wang X, Chen J, Huang X. Rosuvastatin Attenuates Myocardial Ischemia-Reperfusion Injury via Upregulating miR-17-3p-Mediated Autophagy. Cell Reprogram 2019; 21:323-330. [PMID: 31730378 PMCID: PMC6918854 DOI: 10.1089/cell.2018.0053] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myocardial diseases usually appear ischemic. Reperfusion therapy is one of the effective methods that can improve clinical therapeutic efficacy. However, reperfusion results in myocardial injury named I/R injury. Rosuvastatin (RS) is HMG-CoA reductase inhibitor. We investigated the role of RS in the myocardial I/R injury in vitro and its active mechanism. Oxygen-glucose deprivation/reoxygenation (OGD/R) model was applied to investigate I/R in vitro. OGD/R decreased cell viability and increased levels of miR-17-3p and lactate dehydrogenase (LDH) leakage. Besides, RS decreased cleaved caspase-3 level and LDH leakage, promoted the levels of miR-17-3p and LC3II/LC3I, and increased cell viability when H9C2 cell was treated by OGD/R. miR-17-3p inhibitor reduced the H9C2 cell viability and LC3II/LC3I level, whereas miR-17-3p mimics increased H9C2 cell viability and LC3II/LC3I level. RS promoted cell viability and increased LC3II/LC3I level while it lowered LDH leakage, apoptosis rate, and the levels of cleaved caspase-3 and Cyto c. Our study suggested that RS reduced I/R injury in cardiocyte via cleaved caspase-3/Cyto c apoptosis signaling pathway and autophagy. Moreover, the autophagy happens to cardiocyte by upregulating the expression of miR-17-3p.
Collapse
Affiliation(s)
- Xiaoqin Wang
- Department of Cardiovascular Medicine, Jingmen No.1 People's Hospital, Jingmen, China
| | - Jinghan Chen
- Department of Neurology, Jingmen Recovery Hospital, Jingmen, China
| | - Xiaojiao Huang
- Department of Cardiovascular Medicine, Jingmen No.1 People's Hospital, Jingmen, China
| |
Collapse
|
38
|
Shi J, Chen C, Xu X, Lu Q. miR-29a promotes pathological cardiac hypertrophy by targeting the PTEN/AKT/mTOR signalling pathway and suppressing autophagy. Acta Physiol (Oxf) 2019; 227:e13323. [PMID: 31162869 DOI: 10.1111/apha.13323] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 05/13/2019] [Accepted: 05/31/2019] [Indexed: 12/26/2022]
Abstract
AIM Although miR-29 has emerged as a crucial non-coding RNA in the regulation of pathological cardiac hypertrophy, further exploration of its specific mechanisms is necessary to resolve controversy about its major role in this condition. This study therefore evaluated the role of miR-29a and whether it acts through the PTEN/AKT/mTOR pathway. METHODS In this study, a rat model of pressure-induced cardiac hypertrophy was established by transverse aortic constriction and verified by echocardiography, histological analysis and quantitative RT-PCR. At the cellular level, we explored the role of miR-29a in angiotensin II-stimulated hypertrophic H9c2 cardiomyoblasts by transfecting the cells with miR-29a inhibitor and mimic. The relationship between miR-29a and the signalling pathway was investigated with dual luciferase reporter assays, immunofluorescence analysis and Western blotting. We also examined whether autophagy is involved in the regulatory mechanism of miR-29a through transmission electron microscopy and detection of autophagy-associated proteins. RESULTS The results showed that miR-29a was upregulated both in rats 4 weeks after surgery and in 10-6 M angiotensin II-stimulated cells. In contrast, inhibition of miR-29a partially attenuated angiotensin II-induced hypertrophy. Additionally, bioinformatics analysis revealed that PTEN was one of the target genes of miR-29a, which was also verified by luciferase assay. The results of immunofluorescence and Western blotting indicated that overexpression of miR-29a inhibited the expression of PTEN, activated the AKT/mTOR pathway and suppressed autophagy, which ultimately led to cardiac hypertrophy. CONCLUSION In pathological cardiac hypertrophy, miR-29a was overexpressed and promoted cardiac hypertrophy by regulating the PTEN/AKT/mTOR pathway and suppressing autophagy.
Collapse
Affiliation(s)
- Jia‐yu Shi
- Department of Cardiology Affiliated Hospital of Nantong University Nantong China
| | - Chu Chen
- Department of Cardiology Affiliated Hospital of Nantong University Nantong China
| | - Xuan Xu
- Department of Cardiology Affiliated Hospital of Nantong University Nantong China
| | - Qi Lu
- Department of Cardiology Affiliated Hospital of Nantong University Nantong China
| |
Collapse
|
39
|
Guan P, Sun ZM, Wang N, Zhou J, Luo LF, Zhao YS, Ji ES. Resveratrol prevents chronic intermittent hypoxia-induced cardiac hypertrophy by targeting the PI3K/AKT/mTOR pathway. Life Sci 2019; 233:116748. [DOI: 10.1016/j.lfs.2019.116748] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/02/2019] [Accepted: 08/10/2019] [Indexed: 01/22/2023]
|
40
|
Zheng J, Tan J, Miao YY, Zhang Q. Extracellular vesicles degradation pathway based autophagy lysosome pathway. Am J Transl Res 2019; 11:1170-1183. [PMID: 30972154 PMCID: PMC6456539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 01/28/2019] [Indexed: 06/09/2023]
Abstract
As an ancient intracellular degradation pathway, the autophagy lysosome pathway exists in various cells continuously and stably and maintains cellular homeostasis by degrading damaged organelles and misfolded proteins that are prejudicial to cells. Extracellular vesicles (EVs) including microparticles and exosomes, are derived from varieties of mammalian tissue cells such as platelets, endothelial cells, cardiomyocytes. Through large quantity of active substances carried by EVs, EVs exert momentous biological functions. Recent researches have revealed the molecular mechanism of the interaction between extracellular vesicles and autophagy. In this review, we first elaborate that extracellular vesicles are identified and internalized by target cells by means of receptor-ligand. Since extracellular vesicles contain multiple functional molecules, we subsequently describe the process of intracellular autophagy pathway induced by extracellular vesicles, which activates autophagy-related pathways or delivers autophagy-associated molecules. Finally, we introduced the effects of extracellular vesicle-induced autophagy on extracellular vesicles and target cells respectively. In conclusion, this article integrates relevant theoretical knowledge of autophagy caused by extracellular vesicles and provides a new direction for the study of extracellular vesicles in the future.
Collapse
Affiliation(s)
- Jun Zheng
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics InstituteTianjin, China
| | - Jin Tan
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics InstituteTianjin, China
| | | | - Qiang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics InstituteTianjin, China
| |
Collapse
|
41
|
Abstract
Cardiotoxicity is a well-known side effect of doxorubicin (DOX), but the mechanisms leading to this phenomenon are still not completely clear. Prediction of drug-induced dysfunction onset is difficult and is still largely based on detection of cardiac troponin (cTn), a circulating marker of heart damage. In the last years, several investigations focused on the possible involvement of microRNAs (miRNAs) in DOX-induced toxicity in vitro, with contrasting results. Recently, several groups employed animal models to mimic patient’s condition, investigate the biological pathways perturbed by DOX, and identify diagnostic markers of cardiotoxicity. We reviewed the results from several studies investigating cardiac miRNAs expression in rodent models of DOX-treatment. We also discussed the data from two publications indicating the possible use of circulating miRNA as biomarkers of DOX-induced cardiotoxicity. Unfortunately, limited information was derived from these studies, as selection methods of candidate-miRNAs and heterogeneity in cardiotoxicity assessment greatly hampered the novelty and robustness of the findings. Nevertheless, at least one circulating miRNA, miR-1, showed a good potential as early biomarker of drug-mediated cardiac dysfunction onset. The use of animal models to investigate DOX-induced cardiotoxicity surely helps narrowing the gap between basic research and clinical practice. Despite this, several issues, including selection of relevant miRNAs and less-than-optimal assessment of cardiotoxicity, greatly limited the results obtained so far. Nonetheless, the association of patients-based studies with the use of preclinical models may be the key to address the many unanswered questions regarding the pathophysiology and early detection of cardiotoxicity.
Collapse
|
42
|
The potential role of tubeimosides in cancer prevention and treatment. Eur J Med Chem 2019; 162:109-121. [DOI: 10.1016/j.ejmech.2018.11.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 12/30/2022]
|
43
|
Toll-like receptor 4 contributes to a myofibroblast phenotype in cardiac fibroblasts and is associated with autophagy after myocardial infarction in a mouse model. Atherosclerosis 2018; 279:23-31. [PMID: 30399463 DOI: 10.1016/j.atherosclerosis.2018.10.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 10/11/2018] [Accepted: 10/17/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND AND AIMS Cardiac fibrosis after myocardial infarction (MI) is involved in fibroblast transforming and differentiating into myofibroblast phenoconversion, however, the underlying mechanisms are poorly understood. Toll-like receptor 4 (TLR4)-mediated pathogen-associated molecular patterns are key factors that deteriorate cardiac remodelling after MI. Moreover, autophagy has dual roles in cell survival in myocardial tissues after MI. We evaluated the relationship between TLR4 signalling and cardiac myofibroblast transformation-differentiation after MI in vivo and in vitro and analysed the role of autophagy. METHODS We reproduced a model of MI by the permanent ligation of the left anterior descending coronary artery of Tlr4-knockout (Tlr4-/-) and wild-type (WT) male mice. We evaluated scar formation, myofibroblast phenoconversion, LC3 dot formation, autophagy related proteins and α-smooth muscle actin (SMA) in cardiac tissues, 7, 14, and 28 days after myocardial infarction. Cardiac fibroblasts were cultured from Tlr4-/- or WT mice. Vimentin, α-SMA, bilayer membrane vesicle structures of autophagosomes, and autophagy related proteins were observed after treatment with lipopolysaccharide (LPS) or 3-methyladenine (3-MA) at 24 h. RESULTS After MI on 7, 14, and 28 days, Tlr4-/- mice showed that heart tissue fibrosis and expression of α-SMA, a marker of myofibroblasts, were decreased compared to WT mice. Additionally, levels of LC3II, Atg5, Atg7, and Beclin-1, which are involved in autophagy, were lower than those in WT mice. Further, p62 expression, which is negatively correlated with autophagy levels, was higher in Tlr4-/- mice. Moreover, LC3-labelled autophagosomes in cardiac tissues were reduced in these animals. In vitro, LPS, a ligand of TLR4, stimulated α-SMA expression in cardiac fibroblasts, enhanced autophagic flux, and increased autophagosome numbers. In contrast, these effects were not obvious in Tlr4-/- cardiac fibroblasts. LC3II, Atg5, Atg7, and Beclin-1 were upregulated, and p62 was downregulated in cardiac fibroblasts of WT mice stimulated with LPS. However, these effects were blocked by 3-methyladenine, an inhibitor of autophagy. CONCLUSIONS These results suggest that TLR4 signalling executes the development of a myofibroblast phenotype after MI via autophagy and could be therapeutically exploited to improve outcome after myocardial injury.
Collapse
|
44
|
Gupta I, Varshney NK, Khan S. Emergence of Members of TRAF and DUB of Ubiquitin Proteasome System in the Regulation of Hypertrophic Cardiomyopathy. Front Genet 2018; 9:336. [PMID: 30186311 PMCID: PMC6110912 DOI: 10.3389/fgene.2018.00336] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/03/2018] [Indexed: 01/10/2023] Open
Abstract
The ubiquitin proteasome system (UPS) plays an imperative role in many critical cellular processes, frequently by mediating the selective degradation of misfolded and damaged proteins and also by playing a non-degradative role especially important as in many signaling pathways. Over the last three decades, accumulated evidence indicated that UPS proteins are primal modulators of cell cycle progression, DNA replication, and repair, transcription, immune responses, and apoptosis. Comparatively, latest studies have demonstrated a substantial complexity by the UPS regulation in the heart. In addition, various UPS proteins especially ubiquitin ligases and proteasome have been identified to play a significant role in the cardiac development and dynamic physiology of cardiac pathologies such as ischemia/reperfusion injury, hypertrophy, and heart failure. However, our understanding of the contribution of UPS dysfunction in the plausible development of cardiac pathophysiology and the complete list of UPS proteins regulating these afflictions is still in infancy. The recent emergence of the roles of TNF receptor-associated factor (TRAFs) and deubiquitinating enzymes (DUBs) superfamily in hypertrophic cardiomyopathy has enhanced our knowledge. In this review, we have mainly compiled the TRAF superfamily of E3 ligases and few DUBs proteins with other well-documented E3 ligases such as MDM2, MuRF-1, Atrogin-I, and TRIM 32 that are specific to myocardial hypertrophy. In this review, we also aim to highlight their expression profile following physiological and pathological stimulation leading to the onset of hypertrophic phenotype in the heart that can serve as biomarkers and the opportunity for the development of novel therapies.
Collapse
Affiliation(s)
- Ishita Gupta
- Structural Immunology Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India.,Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Nishant K Varshney
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| | - Sameena Khan
- Drug Discovery Research Center, Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
45
|
Ding J, Cong YF, Liu B, Miao J, Wang L. Aberrant Protein Turn-Over Associated With Myofibrillar Disorganization in FHL1 Knockout Mice. Front Genet 2018; 9:273. [PMID: 30083183 PMCID: PMC6065255 DOI: 10.3389/fgene.2018.00273] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/04/2018] [Indexed: 12/12/2022] Open
Abstract
Mutations in the FHL1 gene, and FHL1 protein deletion, are associated with rare hereditary myopathies and cardiomyopathies. FHL1-null mice develop age-dependent myopathy and increased autophagic activity. However, the molecular pathway involved in contractile function and increased autophagic activity in the FHL1-null mouse has not yet been fully elucidated. In this study, FHL1 protein was knocked out in mice using Transcription Activator-like Effector Nucleases (TALENs) and the IRS1-FOXO1/mTOR signaling pathway was investigated in skeletal muscles and heart. TALEN constructs caused targeted mutations in 30% of newborn mice; these mutations caused a deletion of 1–13 base pairs which blocked synthesis of the full-length FHL1 protein. Furthermore, 2.5-month old FHL1-null male mice were not prone to global muscular fatigue when compared with WT littermates, but histological analysis and ultrastructural analysis by transmission electron microscopy confirmed the presence of myofibrillar disorganization and the accumulation of autophagosome or autolysosome-like structures in FHL1-null mice. Moreover, autophagy and mitophagy were both activated in FHL1 KO mice and the degradation of autophagic lysosomes was impeded. Enhanced autophagic activity in FHL1 KO mice was induced by FOXO1 up-regulation and protein synthesis was increased via mTOR. The cytoskeletal proteins, MYBPC2 and LDB3, were involved in the formation of pathological changes in FHL1 KO mice. Markers of early differentiation (MEF2C and MYOD1) and terminal differentiation (total MYH) were both up-regulated in tibialis anterior (TA) muscles in FHL1 KO mice. The number of type I and type II fibers increased in FHL1-null TA muscles, but the number of type| | b, and type | | d fibers were both reduced in FHL1-null TA muscles. The results obtained from the heart were consistent with those from the skeletal muscle and indicated autophagic activation by FOXO1 and an increase in protein synthesis via mTOR also occurred in the heart tissue of FHL1 knockout mice. In conclusion, aberrant protein turn-over associated with myofibrillar disorganization in FHL1 knockout mice. the up-regulation of FOXO1 was associated with enhanced autophagic activity and pathological changes in the muscle fibers of FHL1 KO mice. These results indicated that autophagy activated by FOXO1 is a promising therapeutic target for hereditary myopathies and cardiomyopathies induced by FHL1.
Collapse
Affiliation(s)
- Jingjing Ding
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, China
| | - Yan Fei Cong
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, China
| | - Bo Liu
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, China
| | - Jianing Miao
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, China
| | - Lili Wang
- Medical Research Center of Shengjing Hospital, China Medical University, Shenyang, China
| |
Collapse
|
46
|
AMPK blunts chronic heart failure by inhibiting autophagy. Biosci Rep 2018; 38:BSR20170982. [PMID: 30021848 PMCID: PMC6050195 DOI: 10.1042/bsr20170982] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 12/23/2022] Open
Abstract
AMP-activated protein kinase (AMPK), a serine/threonine protein kinase, has been shown to exert a protective effect against cardiac hypertrophy and heart failure. Our previous reports have demonstrated that AMPK can inhibit cardiac hypertrophy and block the development of heart failure by promoting autophagy. However, other investigators have demonstrated that overactive and dysregulated autophagy may also contribute to the onset and exacerbation of heart failure. Thus, a major goal of the present investigation is to explore how AMPK regulates autophagy in heart failure. First, heart failure was induced in mice by 4 weeks of pressure overload; AMPK activation was subsequently induced by injecting 5-aminoimidazole-4-carboxamide 1-β-d-ribonucleotide (AICAR) after the establishment of chronic heart failure. We showed that AMPK activation significantly attenuated the progression of heart failure and improved cardiac function, which was accompanied by decreased autophagy levels in the failing hearts. Additionally, we demonstrated that the treatment with AICAR inhibited phosphorylation of the mammalian target of rapamycin (mTOR) complex 1 (mTORC1) downstream effectors 4E-binding protein1 (4EBP1), and ribosomal protein S6 kinase (p70S6K). A major action of AICAR was significantly to activate AKT (Ser473), the downstream substrate of mTOR complex 2 (mTORC2). In conclusion, the data suggest that AMPK improved cardiac function during the development of chronic heart failure by attenuating autophagy, potentially via mTORC2 activation and the downstream effects.
Collapse
|
47
|
Deshpande S, Abdollahi M, Wang M, Lanting L, Kato M, Natarajan R. Reduced Autophagy by a microRNA-mediated Signaling Cascade in Diabetes-induced Renal Glomerular Hypertrophy. Sci Rep 2018; 8:6954. [PMID: 29725042 PMCID: PMC5934412 DOI: 10.1038/s41598-018-25295-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/18/2018] [Indexed: 12/12/2022] Open
Abstract
Autophagy plays a key role in the pathogenesis of kidney diseases, however its role in diabetic nephropathy (DN), and particularly in kidney glomerular mesangial cells (MCs) is not very clear. Transforming Growth Factor- β1 (TGF-β), a key player in the pathogenesis of DN, regulates expression of various microRNAs (miRNAs), some of which are known to regulate the expression of autophagy genes. Here we demonstrate that miR-192, induced by TGF-β signaling, plays an important role in regulating autophagy in DN. The expression of key autophagy genes was decreased in kidneys of streptozotocin-injected type-1 and type-2 (db/db) diabetic mice and this was reversed by treatment with Locked Nucleic Acid (LNA) modified miR-192 inhibitors. Changes in autophagy gene expression were also attenuated in kidneys of diabetic miR-192-KO mice. In vitro studies using mouse glomerular mesangial cells (MMCs) also showed a decrease in autophagy gene expression with TGF-β treatment. miR-192 mimic oligonucleotides also decreased the expression of certain autophagy genes. These results demonstrate that TGF-β and miR-192 decrease autophagy in MMCs under diabetic conditions and this can be reversed by inhibition or deletion of miR-192, further supporting miR-192 as a useful therapeutic target for DN.
Collapse
Affiliation(s)
- Supriya Deshpande
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Mei Wang
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA
| | - Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA.
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute and Beckman Research Institute of City of Hope, Duarte, California, 91010, USA.
| |
Collapse
|
48
|
Abstract
Ageing is the gradual decline in biological function both at the cellular and organismal level. One of the key characteristics of cellular ageing is the accumulation of damaged proteins and organelles which, in turn, can cause cellular toxicity and death. Autophagy is an evolutionarily conserved process that is responsible for the sequestration of damaged or surplus cytoplasmic components which are then delivered to the lysosome for degradation. This house-keeping mechanism is essential to maintain cellular homeostasis and survival, particularly during stress. A decline or loss of sensitivity/responsiveness of autophagy is intimately linked with an accelerated rate of ageing as well as many age-related diseases including neurodegeneration, cancer and metabolic disease where damage accumulation exceeds damage removal. This chapter summarises current knowledge regarding the relationship between autophagy and ageing and outlines some strategies that can be implemented to promote the anti-ageing effects of autophagy to improve human health and lifespan.
Collapse
Affiliation(s)
- Charlotte J Pattison
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| | | |
Collapse
|
49
|
Su F, Shi M, Zhang J, Zheng Q, Zhang D, Zhang W, Wang H, Li X. Simvastatin Protects Heart from Pressure Overload Injury by Inhibiting Excessive Autophagy. Int J Med Sci 2018; 15:1508-1516. [PMID: 30443172 PMCID: PMC6216062 DOI: 10.7150/ijms.28106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/29/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiac hypertrophy is an independent predictor of cardiovascular morbidity and mortality. To identify the mechanisms by which simvastatin inhibits cardiac hypertrophy induced by pressure overload, we determined effects of simvastatin on 14-3-3 protein expression and autophagic activity. Simvastatin was administered intragastrically to Sprague-Dawley (SD) rats before abdominal aortic banding (AAB). Neonatal rat cardiomyocytes (NRCs) were treated with simvastatin before angiotensin II (AngII) stimulation. 14-3-3, LC3, and p62 protein levels were determined by western blot. Autophagy was also measured by the double-labeled red fluorescent protein-green fluorescent protein autophagy reporter system. Simvastatin alleviated excessive autophagy, characterized by a high LC3II/LC3I ratio and low level of p62, and blunted cardiac hypertrophy while increasing 14-3-3 protein expression in rats that had undergone AAB. In addition, it increased 14-3-3 expression and inhibited excessive autophagy in NRCs exposed to AngII. Our study demonstrated that simvastatin may inhibit excessive autophagy, increase 14-3-3 expression, and finally exert beneficial effects on cardioprotection against pressure overload.
Collapse
Affiliation(s)
- Feifei Su
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Miaoqian Shi
- Department of Cardiology, PLA Army General Hospital, No.5 Nanmen Cang, Dongcheng District, Beijing, 100700, China
| | - Jian Zhang
- Department of Cardiology, Beijing Chest Hospital Heart Center, Capital Medical University, No.9. Beiguan Grand Street, Tongzhou District, Beijing, 101149, China
| | - Qiangsun Zheng
- Division of Cardiology, Second Affiliated Hospital of JiaoTong University, Xi'an, 710004, China
| | - Dongwei Zhang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Wei Zhang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, China
| |
Collapse
|
50
|
Feng Y, Zhang Y, Xiao H. AMPK and cardiac remodelling. SCIENCE CHINA-LIFE SCIENCES 2017; 61:14-23. [PMID: 29170891 DOI: 10.1007/s11427-017-9197-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/11/2017] [Indexed: 12/17/2022]
Abstract
Cardiac remodelling is generally accepted as a critical process in the progression of heart failure. Myocyte hypertrophy, inflammatory responses and cardiac fibrosis are the main pathological changes associated with cardiac remodelling. AMP-activated protein kinase (AMPK) is known as an energy sensor and a regulator of cardiac metabolism under normal and ischaemic conditions. Additionally, AMPK has been shown to play roles in cardiac remodelling extending well beyond metabolic regulation. In this review, we discuss the currently defined roles of AMPK in cardiac remodelling and summarize the effects of AMPK on cardiac hypertrophy, inflammatory responses and fibrosis and the molecular mechanisms underlying these effects. In addition, we discuss some pharmacological activators of AMPK that are promising treatments for cardiac remodelling.
Collapse
Affiliation(s)
- Yenan Feng
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, 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, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|