1
|
Islam R, Hong Z. YAP/TAZ as mechanobiological signaling pathway in cardiovascular physiological regulation and pathogenesis. MECHANOBIOLOGY IN MEDICINE 2024; 2:100085. [PMID: 39281415 PMCID: PMC11391866 DOI: 10.1016/j.mbm.2024.100085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Cardiovascular diseases (CVDs) persistently rank as a leading cause of premature death and illness worldwide. The Hippo signaling pathway, known for its highly conserved nature and integral role in regulating organ size, tissue homeostasis, and stem cell function, has been identified as a critical factor in the pathogenesis of CVDs. Recent findings underscore the significance of the Yes-associated protein (YAP) and the Transcriptional Coactivator with PDZ-binding motif (TAZ), collectively referred to as YAP/TAZ. These proteins play pivotal roles as downstream components of the Hippo pathway, in the regulation of cardiovascular development and homeostasis. YAP/TAZ can regulate various cellular processes such as cell proliferation, migration, differentiation, and apoptosis through their interactions with transcription factors, particularly those within the transcriptional enhancer associate domain (TEAD) family. The aim of this review is to provide a comprehensive overview of the current understanding of YAP/TAZ signaling in cardiovascular physiology and pathogenesis. We analyze the regulatory mechanisms of YAP/TAZ activation, explore their downstream effectors, and examine their association across numerous cardiovascular disorders, including myocardial hypertrophy, myocardial infarction, pulmonary hypertension, myocardial ischemia-reperfusion injury, atherosclerosis, angiogenesis, restenosis, and cardiac fibrosis. Furthermore, we investigate the potential therapeutic implications of targeting the YAP/TAZ pathway for the treatment of CVDs. Through this comprehensive review, our aim is to elucidate the current understanding of YAP/TAZ signaling in cardiovascular biology and underscore its potential implications for the diagnosis and therapeutic intervention of CVDs.
Collapse
Affiliation(s)
- Rakibul Islam
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| | - Zhongkui Hong
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
2
|
Wang Y, Chatterjee E, Li G, Xu J, Xiao J. Force-sensing protein expression in response to cardiovascular mechanotransduction. EBioMedicine 2024; 110:105412. [PMID: 39481337 PMCID: PMC11554632 DOI: 10.1016/j.ebiom.2024.105412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
Force-sensing biophysical cues in microenvironment, including extracellular matrix performances, stretch-mediated mechanics, shear stress and flow-induced hemodynamics, have a significant influence in regulating vascular morphogenesis and cardiac remodeling by mechanotransduction. Once cells perceive these extracellular mechanical stimuli, Piezo activation promotes calcium influx by forming integrin-adhesion-coupling receptors. This induces robust contractility of cytoskeleton structures to further transmit biomechanical alternations into nuclei by regulating Hippo-Yes associated protein (YAP) signaling pathway between cytoplasmic and nuclear translocation. Although biomechanical stimuli are widely studied in cardiovascular diseases, the expression of force-sensing proteins in response to cardiovascular mechanotransduction has not been systematically concluded. Therefore, this review will summarize the force-sensing Piezo, cytoskeleton and YAP proteins to mediate extracellular mechanics, and also give the prominent emphasis on intrinsic connection of these mechanical proteins and cardiovascular mechanotransduction. Extensive insights into cardiovascular mechanics may provide some new strategies for cardiovascular clinical therapy.
Collapse
Affiliation(s)
- Yongtao Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China
| | - Emeli Chatterjee
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Jiahong Xu
- Department of Cardiology, Shanghai Gongli Hospital, Shanghai 200135, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China; Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
3
|
Lumish HS, Sherrid MV, Janssen PML, Ferrari G, Hasegawa K, Castillero E, Adlestein E, Swistel DG, Topkara VK, Maurer MS, Reilly MP, Shimada YJ. Comprehensive Proteomic Profiling of Human Myocardium Reveals Signaling Pathways Dysregulated in Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2024; 84:1999-2011. [PMID: 39365226 DOI: 10.1016/j.jacc.2024.07.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is the most common genetic cardiac disease. Signaling pathways that link genetic sequence variants to clinically overt HCM and progression to severe forms of HCM remain unknown. OBJECTIVES The purpose of this study was to identify signaling pathways that are differentially regulated in HCM, using proteomic profiling of human myocardium, confirmed with transcriptomic profiling. METHODS In this multicenter case-control study, myocardial samples were obtained from cases with HCM and control subjects with nonfailing hearts. Proteomic profiling of 7,289 proteins from myocardial samples was performed using the SomaScan assay (SomaLogic). Pathway analysis of differentially expressed proteins was performed, using a false discovery rate <0.05. Pathway analysis of proteins whose concentrations correlated with clinical indicators of severe HCM (eg, reduced left ventricular ejection fraction, atrial fibrillation, and ventricular tachyarrhythmias) was also executed. Confirmatory analysis of differentially expressed genes was performed using myocardial transcriptomic profiling. RESULTS The study included 99 HCM cases and 15 control subjects. Pathway analysis of differentially expressed proteins revealed dysregulation of the Ras-mitogen-activated protein kinase, ubiquitin-mediated proteolysis, angiogenesis-related (eg, hypoxia-inducible factor-1, vascular endothelial growth factor), and Hippo pathways. Pathways known to be dysregulated in HCM, including metabolic, inflammatory, and extracellular matrix pathways, were also dysregulated. Pathway analysis of proteins associated with clinical indicators of severe HCM and of differentially expressed genes supported these findings. CONCLUSIONS The present study represents the most comprehensive (>7,000 proteins) and largest-scale (n = 99 HCM cases) proteomic profiling of human HCM myocardium to date. Proteomic profiling and confirmatory transcriptomic profiling elucidate dysregulation of both newly recognized (eg, Ras-mitogen-activated protein kinase) and known pathways associated with pathogenesis and progression to severe forms of HCM.
Collapse
Affiliation(s)
- Heidi S Lumish
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mark V Sherrid
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Giovanni Ferrari
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA; Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Estibaliz Castillero
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, New York, USA
| | - Elizabeth Adlestein
- Leon Charney Division of Cardiology, New York University Langone Health, New York, New York, USA
| | - Daniel G Swistel
- Department of Cardiothoracic Surgery, New York University Langone Health, New York, New York, USA
| | - Veli K Topkara
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Mathew S Maurer
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA; Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| | - Yuichi J Shimada
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA.
| |
Collapse
|
4
|
Amanda B, Pragasta R, Cakrasana H, Mustika A, Faizah Z, Oceandy D. The Hippo Signaling Pathway, Reactive Oxygen Species Production, and Oxidative Stress: A Two-Way Traffic Regulation. Cells 2024; 13:1868. [PMID: 39594616 PMCID: PMC11592687 DOI: 10.3390/cells13221868] [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: 09/11/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
The Hippo signaling pathway is recognized for its significant role in cell differentiation, proliferation, survival, and tissue regeneration. Recently, the Hippo signaling pathway was also found to be associated with oxidative stress and reactive oxygen species (ROS) regulation, which are important in the regulation of cell survival. Studies indicate a correlation between components of the Hippo signaling pathway, including MST1, YAP, and TAZ, and the generation of ROS. On the other hand, ROS and oxidative stress can activate key components of the Hippo signaling pathway. For example, ROS production activates MST1, which subsequently phosphorylates FOXO3, leading to apoptotic cell death. ROS was also found to regulate YAP, in addition to MST1/2. Oxidative stress and ROS formation can impair lipids, proteins, and DNA, leading to many disorders, including aging, neurodegeneration, atherosclerosis, and diabetes. Consequently, understanding the interplay between the Hippo signaling pathway, ROS, and oxidative stress is crucial for developing future disease management strategies. This paper aimed to review the association between the Hippo signaling pathway, regulation of ROS production, and oxidative stress to provide beneficial information in understanding cell function and pathological processes.
Collapse
Affiliation(s)
- Bella Amanda
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
- Airlangga University Teaching Hospital, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Rangga Pragasta
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
- Faculty of Medicine, Universitas Islam Malang, Malang 65144, Indonesia
| | - Haris Cakrasana
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
| | - Arifa Mustika
- Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia;
| | - Zakiyatul Faizah
- Andrology Study Program, Department of Biomedical Sciences, Faculty of Medicine, Universitas Airlangga, Surabaya 60132, Indonesia; (R.P.); (H.C.); (Z.F.)
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK;
| |
Collapse
|
5
|
Miyamoto S. Untangling the role of RhoA in the heart: protective effect and mechanism. Cell Death Dis 2024; 15:579. [PMID: 39122698 PMCID: PMC11315981 DOI: 10.1038/s41419-024-06928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
RhoA (ras homolog family member A) is a small G-protein that transduces intracellular signaling to regulate a broad range of cellular functions such as cell growth, proliferation, migration, and survival. RhoA serves as a proximal downstream effector of numerous G protein-coupled receptors (GPCRs) and is also responsive to various stresses in the heart. Upon its activation, RhoA engages multiple downstream signaling pathways. Rho-associated coiled-coil-containing protein kinase (ROCK) is the first discovered and best characterized effector or RhoA, playing a major role in cytoskeletal arrangement. Many other RhoA effectors have been identified, including myocardin-related transcription factor A (MRTF-A), Yes-associated Protein (YAP) and phospholipase Cε (PLCε) to regulate transcriptional and post-transcriptional processes. The role of RhoA signaling in the heart has been increasingly studied in last decades. It was initially suggested that RhoA signaling pathway is maladaptive in the heart, but more recent studies using cardiac-specific expression or deletion of RhoA have revealed that RhoA activation provides cardioprotection against stress through various mechanisms including the novel role of RhoA in mitochondrial quality control. This review summarizes recent advances in understanding the role of RhoA in the heart and its signaling pathways to prevent progression of heart disease.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
6
|
Zhang Y, Ren Y, Li X, Li M, Fu M, Zhou W, Yu Y, Xiong Y. A review on decoding the roles of YAP/TAZ signaling pathway in cardiovascular diseases: Bridging molecular mechanisms to therapeutic insights. Int J Biol Macromol 2024; 271:132473. [PMID: 38795886 DOI: 10.1016/j.ijbiomac.2024.132473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/02/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) serve as transcriptional co-activators that dynamically shuttle between the cytoplasm and nucleus, resulting in either the suppression or enhancement of their downstream gene expression. Recent emerging evidence demonstrates that YAP/TAZ is strongly implicated in the pathophysiological processes that contribute to cardiovascular diseases (CVDs). In the cardiovascular system, YAP/TAZ is involved in the orchestration of a range of biological processes such as oxidative stress, inflammation, proliferation, and autophagy. Furthermore, YAP/TAZ has been revealed to be closely associated with the initiation and development of various cardiovascular diseases, including atherosclerosis, pulmonary hypertension, myocardial fibrosis, cardiac hypertrophy, and cardiomyopathy. In this review, we delve into recent studies surrounding YAP and TAZ, along with delineating their roles in contributing to the pathogenesis of CVDs with a link to various physiological processes in the cardiovascular system. Additionally, we highlight the current potential drugs targeting YAP/TAZ for CVDs therapy and discuss their challenges for translational application. Overall, this review may offer novel insights for understanding and treating cardiovascular disorders.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yuanyuan Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Xiaofang Li
- Department of Gastroenterology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Man Li
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi 710018, PR China
| | - Mingdi Fu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Wenjing Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China
| | - Yi Yu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China.
| | - Yuyan Xiong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, Shaanxi, PR China; Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, 710018 Xi'an, Shaanxi, PR China.
| |
Collapse
|
7
|
Rawson R, Duong L, Tkachenko E, Chiang AWT, Okamoto K, Dohil R, Lewis NE, Kurten R, Abud EM, Aceves SS. Mechanotransduction-induced interplay between phospholamban and yes-activated protein induces smooth muscle cell hypertrophy. Mucosal Immunol 2024; 17:315-322. [PMID: 38423390 PMCID: PMC11195688 DOI: 10.1016/j.mucimm.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
The gastrointestinal system is a hollow organ affected by fibrostenotic diseases that cause volumetric compromise of the lumen via smooth muscle hypertrophy and fibrosis. Many of the driving mechanisms remain unclear. Yes-associated protein-1 (YAP) is a critical mechanosensory transcriptional regulator that mediates cell hypertrophy in response to elevated extracellular rigidity. In the type 2 inflammatory disorder, eosinophilic esophagitis (EoE), phospholamban (PLN) can induce smooth muscle cell hypertrophy. We used EoE as a disease model for understanding a mechanistic pathway in which PLN and YAP interact in response to rigid extracellular substrate to induce smooth muscle cell hypertrophy. PLN-induced YAP nuclear sequestration in a feed-forward loop caused increased cell size in response to a rigid substrate. This mechanism of rigidity sensing may have previously unappreciated clinical implications for PLN-expressing hollow systems such as the esophagus and heart.
Collapse
Affiliation(s)
- Renee Rawson
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Loan Duong
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Eugene Tkachenko
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; Department of Medicine, University of California, San Diego, California
| | - Austin W T Chiang
- Department of Pediatrics, University of California, San Diego, California; Department of Bioengineering, University of California, San Diego, California
| | - Kevin Okamoto
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Ranjan Dohil
- Department of Pediatrics, University of California, San Diego, California; Division of Gastroenterology, University of California, San Diego, California; XXX, Rady Children's Hospital, San Diego, California
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, California; Department of Bioengineering, University of California, San Diego, California
| | - Richard Kurten
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Arkansas Children's Hospital Research Institute, Little Rock, Arkansas; Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California
| | - Edsel M Abud
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; XXX, Scripps Research Translational Institute, Scripps Research, San Diego, California
| | - Seema S Aceves
- Department of Pediatrics, University of California, San Diego, California; Division of Allergy Immunology, University of California, San Diego, California; XXX, Rady Children's Hospital, San Diego, California; Department of Medicine, University of California, San Diego, California.
| |
Collapse
|
8
|
Cheng X, Liu Y, Qi B, Wang Y, Zheng Y, Liang X, Chang Y, Ning M, Gao W, Li T. Glycyrrhizic acid alleviated MI/R-induced injuries by inhibiting Hippo/YAP signaling pathways. Cell Signal 2024; 115:111036. [PMID: 38185229 DOI: 10.1016/j.cellsig.2024.111036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/22/2023] [Accepted: 01/02/2024] [Indexed: 01/09/2024]
Abstract
BACKGROUND Previous research has demonstrated that glycyrrhizic acid (GA) exhibits antioxidant, anti-inflammatory, and antiapoptotic characteristics. Using myocardial ischemia/reperfusion injury as a case study, this study aims to clarify the functional significance of GA and to elucidate the mechanisms involved. MATERIALS AND METHODS In this study, an MI/R injury model was established both in vivo and in vitro to investigate the impact of GA on MI/R injury. The viability of H9c2 cells was evaluated using the Cell Counting Kit-8. Myocardial damage was assessed through the measurement of creatine kinase myocardial band (CK-MB) levels and lactate dehydrogenase (LDH), HE staining, and MASSON staining. Inflammatory cytokine levels (IL-6, IL-1β, IL-10, and TNF-α) were measured to determine the presence of inflammation. Cellular oxidative stress was evaluated by measuring ROS and MMP levels, while cardiac function was assessed using cardiac color Doppler ultrasound. Immunofluorescence staining to determine the nuclear translocation of YAP, TUNEL to determine apoptosis, and western blotting to determine gene expression. RESULTS GA treatment effectively alleviated myocardial injury induced by MI/R, as evidenced by reduced levels of inflammatory cytokines (IL-1β, IL-6, IL-10, and TNF-α) and cardiac biomarkers (CK-MB, LDH) in MI/R rats. Moreover, There was a significant increase in cell viability in vitro after GA treatment and inhibited reactive oxygen species (ROS) during oxidative stress, while also increasing mitochondrial membrane potential (MMP) in vitro. The Western blot findings indicate that GA treatment effectively suppressed apoptosis in both in vivo and in vitro settings. Additionally, GA demonstrated inhibitory effects on the activation of the Hippo/YAP signaling pathway triggered by MI/R and facilitated YAP nuclear translocation both in vitro and in vivo. It has been found, however, in vitro, that silencing the YAP gene negates GA's protective effect against hypoxia/reoxygenation-induced myocardial injury. CONCLUSION This study suggests that GA regulates YAP nuclear translocation by inhibiting the Hippo/YAP signaling pathway, which protects ists against MI/R injury. This finding may present a novel therapeutic approach for the treatment of MI/R.
Collapse
Affiliation(s)
- Xian Cheng
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Yanwu Liu
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Bingcai Qi
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yuchao Wang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yue Zheng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoyu Liang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yun Chang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Meng Ning
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Tong Li
- The Third Central Clinical College of Tianjin Medical University, Tianjin 300170, China; Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin 300170, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; School of Medicine, Nankai University, Tianjin 300071, China; Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin 300170, China; Tianjin ECMO Treatment and Training Base, Tianjin 300170, China; Artificial Cell Engineering Technology Research Center, Tianjin, China.
| |
Collapse
|
9
|
Ahmed DW, Eiken MK, DePalma SJ, Helms AS, Zemans RL, Spence JR, Baker BM, Loebel C. Integrating mechanical cues with engineered platforms to explore cardiopulmonary development and disease. iScience 2023; 26:108472. [PMID: 38077130 PMCID: PMC10698280 DOI: 10.1016/j.isci.2023.108472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024] Open
Abstract
Mechanical forces provide critical biological signals to cells during healthy and aberrant organ development as well as during disease processes in adults. Within the cardiopulmonary system, mechanical forces, such as shear, compressive, and tensile forces, act across various length scales, and dysregulated forces are often a leading cause of disease initiation and progression such as in bronchopulmonary dysplasia and cardiomyopathies. Engineered in vitro models have supported studies of mechanical forces in a number of tissue and disease-specific contexts, thus enabling new mechanistic insights into cardiopulmonary development and disease. This review first provides fundamental examples where mechanical forces operate at multiple length scales to ensure precise lung and heart function. Next, we survey recent engineering platforms and tools that have provided new means to probe and modulate mechanical forces across in vitro and in vivo settings. Finally, the potential for interdisciplinary collaborations to inform novel therapeutic approaches for a number of cardiopulmonary diseases are discussed.
Collapse
Affiliation(s)
- Donia W. Ahmed
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Madeline K. Eiken
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Samuel J. DePalma
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Adam S. Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Rachel L. Zemans
- Department of Internal Medicine, Division of Pulmonary Sciences and Critical Care Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Jason R. Spence
- Department of Internal Medicine – Gastroenterology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA
| | - Brendon M. Baker
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Lurie Biomedical Engineering Building, 1101 Beal Avenue, Ann Arbor, MI 48109, USA
- Department of Materials Science & Engineering, University of Michigan, North Campus Research Complex, 2800 Plymouth Road, Ann Arbor, MI 48109, USA
| |
Collapse
|
10
|
Wang J, Wang Z, Xia F, Duan Q, Peng X. Atorvastatin reduces renal interstitial fibrosis caused by unilateral ureteral obstruction through inhibiting the transcriptional activity of YAP. Biochem Biophys Res Commun 2023; 678:109-114. [PMID: 37633180 DOI: 10.1016/j.bbrc.2023.08.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/28/2023]
Abstract
Renal interstitial fibrosis is the primary pathological basis for the progression and development of various chronic kidney diseases, ultimately leading to renal failure. Obstructive kidney disease caused by conditions such as kidney stones, is a common cause of renal fibrosis. The Hippo pathway is a crucial signaling pathway that senses mechanical forces and is involved in the pathophysiology of fibrosis. In this study, we established a mouse model of obstructive kidney disease induced by unilateral ureteral obstruction (UUO). The UUO procedure significantly upregulated YAP and fibrosis-related gene expression in a time-dependent manner. Morphologically, the renal fibrotic lesions associated with hydronephrosis progressively worsened over time in the UUO group. Atorvastatin, which is widely used to lower blood cholesterol levels, has recently been shown to inhibit Yes1 associated protein (YAP). We treated UUO mice with atorvastatin for 3 and 10 days and observed a decrease in the expression of YAP and fibrosis-related genes at the mRNA and protein levels, along with a reduction in the renal fibrosis analyzed by Masson's staining. These findings suggest that atorvastatin may serve as a preventive agent for fibrosis associated with obstructive kidney disease.
Collapse
Affiliation(s)
- Jun Wang
- First Clinical Medical College, Nanchang University, Nanchang, 330006, China
| | - Zhichao Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China
| | - Fan Xia
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China
| | - Qiong Duan
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China.
| | - Xiaoping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China; Jiangxi Hypertension Research Institute, Nanchang, 330006, China.
| |
Collapse
|
11
|
Langa P, Shafaattalab S, Goldspink PH, Wolska BM, Fernandes AA, Tibbits GF, Solaro RJ. A perspective on Notch signalling in progression and arrhythmogenesis in familial hypertrophic and dilated cardiomyopathies. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220176. [PMID: 37122209 PMCID: PMC10150215 DOI: 10.1098/rstb.2022.0176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/13/2022] [Indexed: 05/02/2023] Open
Abstract
In this perspective, we discussed emerging data indicating a role for Notch signalling in inherited disorders of the heart failure with focus on hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) linked to variants of genes encoding mutant proteins of the sarcomere. We recently reported an upregulation of elements in the Notch signalling cascade in cardiomyocytes derived from human inducible pluripotent stem cells expressing a TNNT2 variant encoding cardiac troponin T (cTnT-I79N+/-), which induces hypertrophy, remodelling, abnormalities in excitation-contraction coupling and electrical instabilities (Shafaattalab S et al. 2021 Front. Cell Dev. Biol. 9, 787581. (doi:10.3389/fcell.2021.787581)). Our search of the literature revealed the novelty of this finding and stimulated us to discuss potential connections between the Notch signalling pathway and familial cardiomyopathies. Our considerations focused on the potential role of these interactions in arrhythmias, microvascular ischaemia, and fibrosis. This finding underscored a need to consider the role of Notch signalling in familial cardiomyopathies which are trigged by sarcomere mutations engaging mechano-signalling pathways for which there is evidence of a role for Notch signalling with crosstalk with Hippo signalling. Our discussion included a role for both cardiac myocytes and non-cardiac myocytes in progression of HCM and DCM. This article is part of the theme issue 'The heartbeat: its molecular basis and physiological mechanisms'.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Sanam Shafaattalab
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - Paul H. Goldspink
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Beata M. Wolska
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Aurelia A. Fernandes
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| | - Glen F. Tibbits
- Molecular Biology and Biochemistry; BC Children’s Hospital Research Institute, Vancouver, BC, V5Z 4H4; Simon Fraser University Burnaby, British Columbia, V5A 4H4, Canada
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, Chicago, IL, 60612, USA
| |
Collapse
|
12
|
He X, Wang J, Xue Y, Wang S, Dong Y, Zhang H, Wang M. Mammalian sterile 20-like kinase 1 acts as a candidate biomarker of mortality of emergency surgical repair for acute type a aortic dissection. BMC Cardiovasc Disord 2023; 23:188. [PMID: 37038132 PMCID: PMC10088138 DOI: 10.1186/s12872-023-03144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 02/23/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Acute type A aortic dissection (ATAAD) is a life-threatening pathological change of the aorta. Patients who have undergone aortic surgery are usually at high risk of mortality. AIM We investigated the predictive value of serum Mammalian sterile 20-like kinase 1 (MST1) as a biomarker for the risk of mortality of ATAAD patients. METHODS In this retrospective cohort study, we analyzed 160 consecutive ATAAD patients who had undergone emergency surgery from July 2016 to April 2017. Medical records and blood samples were collected and analyzed. ELISA assays were performed to detect the concentrations of several proteins including MST1. The relationship between these potential biomarkers and the primary endpoint of death was evaluated using Cox proportional hazard regression analysis. RESULTS Compared with a low level (< 1330.8 ng/L), high serum MST1 level (≥ 1330.8 ng/L) was positively associated with the 30-day mortality (OR = 5.233, 95%CI, 1.843-14.862, P < 0.01) and retained predictive after adjustment for sex, age, BMI, nasopharyngeal temperature and deep hypothermia circulatory arrest time (OR = 4.628 95% CI, 1.572-13.625, P < 0.01). A pre-existing basic clinical prediction model was improved with the inclusion of preoperative serum MST1. Specifically, the area under the ROC curve for base model (history of cerebrovascular disease, creatinine, time of operation) was 0.708 (95%CI, 0.546-0.836) and markedly increased to 0.823 when taking MST1 into consideration (95%CI, 0.700-0.912, P = 0.02). CONCLUSION Our study suggests that high preoperative circulating MST1, with a concentration greater than 1330.8 ng/L, was correlated with the 30-day mortality of ATAAD patients who underwent emergency surgery.
Collapse
Affiliation(s)
- Xiaohui He
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Jing Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Yuan Xue
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shipan Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yanjun Dong
- College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Beijing Lab for Cardiovascular Precision Medicine, Beijing, China.
| | - Meili Wang
- Beijing Lab for Cardiovascular Precision Medicine, Beijing, China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
13
|
Langa P, Marszalek RJ, Warren CM, Chowdhury SK, Halas M, Batra A, Rafael-Clyke K, Bacon A, Goldspink PH, Solaro RJ, Wolska BM. Altered coronary artery function, arteriogenesis and endothelial YAP signaling in postnatal hypertrophic cardiomyopathy. Front Physiol 2023; 14:1136852. [PMID: 37064918 PMCID: PMC10102353 DOI: 10.3389/fphys.2023.1136852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
Introduction: Hypertrophic cardiomyopathy (HCM) is a cardiovascular genetic disease caused largely by sarcomere protein mutations. Gaps in our understanding exist as to how maladaptive sarcomeric biophysical signals are transduced to intra- and extracellular compartments leading to HCM progression. To investigate early HCM progression, we focused on the onset of myofilament dysfunction during neonatal development and examined cardiac dynamics, coronary vascular structure and function, and mechano-transduction signaling in mice harboring a thin-filament HCM mutation. Methods: We studied postnatal days 7-28 (P7-P28) in transgenic (TG) TG-cTnT-R92Q and non-transgenic (NTG) mice using skinned fiber mechanics, echocardiography, biochemistry, histology, and immunohistochemistry. Results: At P7, skinned myofiber bundles exhibited an increased Ca2+-sensitivity (pCa50 TG: 5.97 ± 0.04, NTG: 5.84 ± 0.01) resulting from cTnT-R92Q expression on a background of slow skeletal (fetal) troponin I and α/β myosin heavy chain isoform expression. Despite the transition to adult isoform expressions between P7-P14, the increased Ca2+- sensitivity persisted through P28 with no apparent differences in gross morphology among TG and NTG hearts. At P7 significant diastolic dysfunction was accompanied by coronary flow perturbation (mean diastolic velocity, TG: 222.5 ± 18.81 mm/s, NTG: 338.7 ± 28.07 mm/s) along with localized fibrosis (TG: 4.36% ± 0.44%, NTG: 2.53% ± 0.47%). Increased phosphorylation of phospholamban (PLN) was also evident indicating abnormalities in Ca2+ homeostasis. By P14 there was a decline in arteriolar cross-sectional area along with an expansion of fibrosis (TG: 9.72% ± 0.73%, NTG: 2.72% ± 0.2%). In comparing mechano-transduction signaling in the coronary arteries, we uncovered an increase in endothelial YAP expression with a decrease in its nuclear to cytosolic ratio at P14 in TG hearts, which was reversed by P28. Conclusion: We conclude that those early mechanisms that presage hypertrophic remodeling in HCM include defective biophysical signals within the sarcomere that drive diastolic dysfunction, impacting coronary flow dynamics, defective arteriogenesis and fibrosis. Changes in mechano-transduction signaling between the different cellular compartments contribute to the pathogenesis of HCM.
Collapse
Affiliation(s)
- Paulina Langa
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Richard J. Marszalek
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Chad M. Warren
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Shamim K. Chowdhury
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Monika Halas
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ashley Batra
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Koreena Rafael-Clyke
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Angelie Bacon
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Paul H. Goldspink
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - R. John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Beata M. Wolska
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
- Department of Medicine, Division of Cardiology, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
14
|
Genome Editing and Cardiac Regeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1396:37-52. [DOI: 10.1007/978-981-19-5642-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
15
|
Zheng M, Li RG, Song J, Zhao X, Tang L, Erhardt S, Chen W, Nguyen BH, Li X, Li M, Wang J, Evans SM, Christoffels VM, Li N, Wang J. Hippo-Yap Signaling Maintains Sinoatrial Node Homeostasis. Circulation 2022; 146:1694-1711. [PMID: 36317529 PMCID: PMC9897204 DOI: 10.1161/circulationaha.121.058777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/20/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND The sinoatrial node (SAN) functions as the pacemaker of the heart, initiating rhythmic heartbeats. Despite its importance, the SAN is one of the most poorly understood cardiac entities because of its small size and complex composition and function. The Hippo signaling pathway is a molecular signaling pathway fundamental to heart development and regeneration. Although abnormalities of the Hippo pathway are associated with cardiac arrhythmias in human patients, the role of this pathway in the SAN is unknown. METHODS We investigated key regulators of the Hippo pathway in SAN pacemaker cells by conditionally inactivating the Hippo signaling kinases Lats1 and Lats2 using the tamoxifen-inducible, cardiac conduction system-specific Cre driver Hcn4CreERT2 with Lats1 and Lats2 conditional knockout alleles. In addition, the Hippo-signaling effectors Yap and Taz were conditionally inactivated in the SAN. To determine the function of Hippo signaling in the SAN and other cardiac conduction system components, we conducted a series of physiological and molecular experiments, including telemetry ECG recording, echocardiography, Masson Trichrome staining, calcium imaging, immunostaining, RNAscope, cleavage under targets and tagmentation sequencing using antibodies against Yap1 or H3K4me3, quantitative real-time polymerase chain reaction, and Western blotting. We also performed comprehensive bioinformatics analyses of various datasets. RESULTS We found that Lats1/2 inactivation caused severe sinus node dysfunction. Compared with the controls, Lats1/2 conditional knockout mutants exhibited dysregulated calcium handling and increased fibrosis in the SAN, indicating that Lats1/2 function through both cell-autonomous and non-cell-autonomous mechanisms. It is notable that the Lats1/2 conditional knockout phenotype was rescued by genetic deletion of Yap and Taz in the cardiac conduction system. These rescued mice had normal sinus rhythm and reduced fibrosis of the SAN, indicating that Lats1/2 function through Yap and Taz. Cleavage Under Targets and Tagmentation sequencing data showed that Yap potentially regulates genes critical for calcium homeostasis such as Ryr2 and genes encoding paracrine factors important in intercellular communication and fibrosis induction such as Tgfb1 and Tgfb3. Consistent with this, Lats1/2 conditional knockout mutants had decreased Ryr2 expression and increased Tgfb1 and Tgfb3 expression compared with control mice. CONCLUSIONS We reveal, for the first time to our knowledge, that the canonical Hippo-Yap pathway plays a pivotal role in maintaining SAN homeostasis.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Rich G Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Jia Song
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Xiaolei Zhao
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Li Tang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| | - Wen Chen
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
| | - Bao H Nguyen
- Department of Molecular Physiology and Biophysics (B.H.N.)
| | - Xiao Li
- Texas Heart Institute, Houston (R.G.L., X.L.)
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Jianxin Wang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha, Hunan, China (L.T., M.L., Jianxin Wang)
| | - Sylvia M Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Departments of Pharmacology and Medicine, University of California at San Diego, La Jolla (S.M.E.)
| | - Vincent M Christoffels
- Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, The Netherlands (V.M.C.)
| | - Na Li
- Department of Medicine (Section of Cardiovascular Research), Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX (J.S., N.L.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (M.Z., X.Z., S.E., W.C., Jun Wang)
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston (S.E., Jun Wang)
| |
Collapse
|
16
|
Fu M, Hu Y, Lan T, Guan KL, Luo T, Luo M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct Target Ther 2022; 7:376. [PMID: 36347846 PMCID: PMC9643504 DOI: 10.1038/s41392-022-01191-9] [Citation(s) in RCA: 196] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/11/2022] Open
Abstract
As an evolutionarily conserved signalling network, the Hippo pathway plays a crucial role in the regulation of numerous biological processes. Thus, substantial efforts have been made to understand the upstream signals that influence the activity of the Hippo pathway, as well as its physiological functions, such as cell proliferation and differentiation, organ growth, embryogenesis, and tissue regeneration/wound healing. However, dysregulation of the Hippo pathway can cause a variety of diseases, including cancer, eye diseases, cardiac diseases, pulmonary diseases, renal diseases, hepatic diseases, and immune dysfunction. Therefore, therapeutic strategies that target dysregulated Hippo components might be promising approaches for the treatment of a wide spectrum of diseases. Here, we review the key components and upstream signals of the Hippo pathway, as well as the critical physiological functions controlled by the Hippo pathway. Additionally, diseases associated with alterations in the Hippo pathway and potential therapies targeting Hippo components will be discussed.
Collapse
Affiliation(s)
- Minyang Fu
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Yuan Hu
- Department of Pediatric Nephrology Nursing, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Tianxia Lan
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Ting Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| | - Min Luo
- Breast Disease Center, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, South of Renmin Road, 610041, Chengdu, China.
| |
Collapse
|
17
|
Depletion of VGLL4 Causes Perinatal Lethality without Affecting Myocardial Development. Cells 2022; 11:cells11182832. [PMID: 36139407 PMCID: PMC9496954 DOI: 10.3390/cells11182832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/31/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital heart disease is one of the leading causes of pediatric morbidity and mortality, thus highlighting the importance of deciphering the molecular mechanisms that control heart development. As the terminal transcriptional effectors of the Hippo-YAP pathway, YAP and TEAD1 form a transcriptional complex that regulates the target gene expression and depletes either of these two genes in cardiomyocytes, thus resulting in cardiac hypoplasia. Vestigial-like 4 (VGLL4) is a transcriptional co-factor that interacts with TEAD and suppresses the YAP/TEAD complex by competing against YAP for TEAD binding. To understand the VGLL4 function in the heart, we generated two VGLL4 loss-of-function mouse lines: a germline Vgll4 depletion allele and a cardiomyocyte-specific Vgll4 depletion allele. The whole-body deletion of Vgll4 caused defective embryo development and perinatal lethality. The analysis of the embryos at day 16.5 revealed that Vgll4 knockout embryos had reduced body size, malformed tricuspid valves, and normal myocardium. Few whole-body Vgll4 knockout pups could survive up to 10 days, and none of them showed body weight gain. In contrast to the whole-body Vgll4 knockout mutants, cardiomyocyte-specific Vgll4 knockout mice had no noticeable heart growth defects and had normal heart function. In summary, our data suggest that VGLL4 is required for embryo development but dispensable for myocardial growth.
Collapse
|
18
|
Zheng A, Chen Q, Zhang L. The Hippo-YAP pathway in various cardiovascular diseases: Focusing on the inflammatory response. Front Immunol 2022; 13:971416. [PMID: 36059522 PMCID: PMC9433876 DOI: 10.3389/fimmu.2022.971416] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/02/2022] [Indexed: 11/25/2022] Open
Abstract
The Hippo pathway was initially discovered in Drosophila melanogaster and mammals as a key regulator of tissue growth both in physiological and pathological states. Numerous studies depict the vital role of the Hippo pathway in cardiovascular development, heart regeneration, organ size and vascular remodeling through the regulation of YAP (yes-associated protein) translocation. Recently, an increasing number of studies have focused on the Hippo-YAP pathway in inflammation and immunology. Although the Hippo-YAP pathway has been revealed to play controversial roles in different contexts and cell types in the cardiovascular system, the mechanisms regulating tissue inflammation and the immune response remain to be clarified. In this review, we summarize findings from the past decade on the function and mechanism of the Hippo-YAP pathway in CVDs (cardiovascular diseases) such as myocardial infarction, cardiomyopathy and atherosclerosis. In particular, we emphasize the role of the Hippo-YAP pathway in regulating inflammatory cell infiltration and inflammatory cytokine activation.
Collapse
Affiliation(s)
| | | | - Li Zhang
- *Correspondence: Li Zhang, ; Qishan Chen,
| |
Collapse
|
19
|
Structural basis and molecular mechanism of biased GPBAR signaling in regulating NSCLC cell growth via YAP activity. Proc Natl Acad Sci U S A 2022; 119:e2117054119. [PMID: 35858343 PMCID: PMC9303995 DOI: 10.1073/pnas.2117054119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The G protein-coupled bile acid receptor (GPBAR) is the membrane receptor for bile acids and a driving force of the liver-bile acid-microbiota-organ axis to regulate metabolism and other pathophysiological processes. Although GPBAR is an important therapeutic target for a spectrum of metabolic and neurodegenerative diseases, its activation has also been found to be linked to carcinogenesis, leading to potential side effects. Here, via functional screening, we found that two specific GPBAR agonists, R399 and INT-777, demonstrated strikingly different regulatory effects on the growth and apoptosis of non-small cell lung cancer (NSCLC) cells both in vitro and in vivo. Further mechanistic investigation showed that R399-induced GPBAR activation displayed an obvious bias for β-arrestin 1 signaling, thus promoting YAP signaling activation to stimulate cell proliferation. Conversely, INT-777 preferentially activated GPBAR-Gs signaling, thus inactivating YAP to inhibit cell proliferation and induce apoptosis. Phosphorylation of GPBAR by GRK2 at S310/S321/S323/S324 sites contributed to R399-induced GPBAR-β-arrestin 1 association. The cryoelectron microscopy (cryo-EM) structure of the R399-bound GPBAR-Gs complex enabled us to identify key interaction residues and pivotal conformational changes in GPBAR responsible for the arrestin signaling bias and cancer cell proliferation. In summary, we demonstrate that different agonists can regulate distinct functions of cell growth and apoptosis through biased GPBAR signaling and control of YAP activity in a NSCLC cell model. The delineated mechanism and structural basis may facilitate the rational design of GPBAR-targeting drugs with both metabolic and anticancer benefits.
Collapse
|
20
|
Tsai CR, Martin JF. Hippo signaling in cardiac fibroblasts during development, tissue repair, and fibrosis. Curr Top Dev Biol 2022; 149:91-121. [PMID: 35606063 PMCID: PMC10898347 DOI: 10.1016/bs.ctdb.2022.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The evolutionarily conserved Hippo signaling pathway plays key roles in regulating the balance between cell proliferation and apoptosis, cell differentiation, organ size control, tissue repair, and regeneration. Recently, the Hippo pathway has been shown to regulate heart fibrosis, defined as excess extracellular matrix (ECM) deposition and increased tissue stiffness. Cardiac fibroblasts (CFs) are the primary cell type that produces, degrades, and remodels the ECM during homeostasis, aging, inflammation, and tissue repair and regeneration. Here, we review the available evidence from the current literature regarding how the Hippo pathway regulates the formation and function of CFs during heart development and tissue repair.
Collapse
Affiliation(s)
- Chang-Ru Tsai
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Cardiomyocyte Renewal Laboratory, Texas Heart Institute, Houston, TX, United States.
| |
Collapse
|
21
|
Ramaccini D, Pedriali G, Perrone M, Bouhamida E, Modesti L, Wieckowski MR, Giorgi C, Pinton P, Morciano G. Some Insights into the Regulation of Cardiac Physiology and Pathology by the Hippo Pathway. Biomedicines 2022; 10:biomedicines10030726. [PMID: 35327528 PMCID: PMC8945338 DOI: 10.3390/biomedicines10030726] [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: 01/28/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 11/16/2022] Open
Abstract
The heart is one of the most fascinating organs in living beings. It beats up to 100,000 times a day throughout the lifespan, without resting. The heart undergoes profound anatomical, biochemical, and functional changes during life, from hypoxemic fetal stages to a completely differentiated four-chambered cardiac muscle. In the middle, many biological events occur after and intersect with each other to regulate development, organ size, and, in some cases, regeneration. Several studies have defined the essential roles of the Hippo pathway in heart physiology through the regulation of apoptosis, autophagy, cell proliferation, and differentiation. This molecular route is composed of multiple components, some of which were recently discovered, and is highly interconnected with multiple known prosurvival pathways. The Hippo cascade is evolutionarily conserved among species, and in addition to its regulatory roles, it is involved in disease by drastically changing the heart phenotype and its function when its components are mutated, absent, or constitutively activated. In this review, we report some insights into the regulation of cardiac physiology and pathology by the Hippo pathway.
Collapse
Affiliation(s)
- Daniela Ramaccini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Gaia Pedriali
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| | - Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| |
Collapse
|
22
|
Mondal A, Das S, Samanta J, Chakraborty S, sengupta A. YAP1 induces hyperglycemic stress-mediated cardiac hypertrophy and fibrosis in an AKT-FOXM1 dependent signaling pathway. Arch Biochem Biophys 2022; 722:109198. [DOI: 10.1016/j.abb.2022.109198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 11/28/2022]
|
23
|
Abstract
ABSTRACT Cardiovascular disease (CVD) remains the leading cause of death worldwide. Therefore, exploring the mechanism of CVDs and critical regulatory factors is of great significance for promoting heart repair, reversing cardiac remodeling, and reducing adverse cardiovascular events. Recently, significant progress has been made in understanding the function of protein kinases and their interactions with other regulatory proteins in myocardial biology. Protein kinases are positioned as critical regulators at the intersection of multiple signals and coordinate nearly every aspect of myocardial responses, regulating contractility, metabolism, transcription, and cellular death. Equally, reconstructing the disrupted protein kinases regulatory network will help reverse pathological progress and stimulate cardiac repair. This review summarizes recent researches concerning the function of protein kinases in CVDs, discusses their promising clinical applications, and explores potential targets for future treatments.
Collapse
|
24
|
Specific Deletion of the FHA Domain Containing SLMAP3 Isoform in Postnatal Myocardium Has No Impact on Structure or Function. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11040018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Sarcolemmal membrane-associated proteins (SLMAPs) belong to the superfamily of tail-anchored membrane proteins known to regulate diverse biological processes, including protein trafficking and signal transduction. Mutations in SLMAP have been linked to Brugada and defective sodium channel Nav1.5 shuttling. The SLMAP gene is alternatively spliced to generate numerous isoforms, broadly defined as SLMAP1 (~35 kDa), SLMAP2 (~45 kDa) and SLMAP3 (~80–95 kDa), which are highly expressed in the myocardium. The SLMAP3 isoform exhibits ubiquitous expression carrying an FHA domain and is believed to negatively regulate Hippo signaling to dictate cell growth/death and differentiation. Using the αMHC-MerCreMer-flox system to target the SLMAP gene, we specifically deleted the SLMAP3 isoform in postnatal mouse hearts without any changes in the expression of SLMAP1/SLMAP2 isoforms. The in vivo analysis of mice with SLMAP3 cardiac deficiency revealed no significant changes to heart structure or function in young or aged mice without or with isoproterenol-induced stress. SLMAP3-deficient hearts revealed no obvious differences in cardiac size, function or hypertrophic response. Further, the molecular analysis indicated that SLMAP3 loss had a minor impact on sodium channel (Nav1.5) expression without affecting cardiac electrophysiology in postnatal myocardium. Surprisingly, the loss of SLMAP3 did not impact Hippo signaling in postnatal myocardium. We conclude that the FHA domain-containing SLMAP3 isoform has no impact on Hippo signaling or sodium channels in postnatal myocardium, which is able to function and respond normally to stress in its absence. Whether SLMAP1/SMAP2 isoforms can compensate for the loss of SLMAP3 in the affairs of the postnatal heart remains to be determined.
Collapse
|
25
|
Meng F, Xie B, Martin JF. Targeting the Hippo pathway in heart repair. Cardiovasc Res 2021; 118:2402-2414. [PMID: 34528077 DOI: 10.1093/cvr/cvab291] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 12/17/2022] Open
Abstract
The Hippo pathway is an evolutionarily and functionally conserved signaling pathway that controls organ size by regulating cell proliferation, apoptosis, and differentiation. Emerging evidence has shown that the Hippo pathway plays critical roles in cardiac development, homeostasis, disease, and regeneration. Targeting the Hippo pathway has tremendous potential as a therapeutic strategy for treating intractable cardiovascular diseases such as heart failure. In this review, we summarize the function of the Hippo pathway in the heart. Particularly, we highlight the posttranslational modification of Hippo pathway components, including the core kinases LATS1/2 and their downstream effectors YAP/TAZ, in different contexts, which has provided new insights and avenues in cardiac research.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - Bing Xie
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas, 77030.,Texas Heart Institute, Houston, Texas, 77030
| |
Collapse
|
26
|
Valizadeh A, Asghari S, Mansouri P, Alemi F, Majidinia M, Mahmoodpoor A, Yousefi B. The roles of signaling pathways in cardiac regeneration. Curr Med Chem 2021; 29:2142-2166. [PMID: 34521319 DOI: 10.2174/0929867328666210914115411] [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] [Received: 03/03/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
In recent years, knowledge of cardiac regeneration mechanisms has dramatically expanded. Regeneration can replace lost parts of organs, common among animal species. The heart is commonly considered an organ with terminal development, which has no reparability potential during post-natal life; however, some intrinsic regeneration capacity has been reported for cardiac muscle, which opens novel avenues in cardiovascular disease treatment. Different endogenous mechanisms were studied for cardiac repairing and regeneration in recent decades. Survival, proliferation, inflammation, angiogenesis, cell-cell communication, cardiomyogenesis, and anti-aging pathways are the most important mechanisms that have been studied in this regard. Several in vitro and animal model studies focused on proliferation induction for cardiac regeneration reported promising results. These studies have mainly focused on promoting proliferation signaling pathways and demonstrated various signaling pathways such as Wnt, PI3K/Akt, IGF-1, TGF-β, Hippo, and VEGF signaling cardiac regeneration. Therefore, in this review, we intended to discuss the connection between different critical signaling pathways in cardiac repair and regeneration.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Samira Asghari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Parinaz Mansouri
- Students Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Forough Alemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia. Iran
| | - Ata Mahmoodpoor
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
27
|
Yes-Associated Protein (Yap) Is Up-Regulated in Heart Failure and Promotes Cardiac Fibroblast Proliferation. Int J Mol Sci 2021; 22:ijms22116164. [PMID: 34200497 PMCID: PMC8201133 DOI: 10.3390/ijms22116164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 05/04/2021] [Accepted: 05/11/2021] [Indexed: 01/01/2023] Open
Abstract
Left ventricular (LV) heart failure (HF) is a significant and increasing cause of death worldwide. HF is characterized by myocardial remodeling and excessive fibrosis. Transcriptional co-activator Yes-associated protein (Yap), the downstream effector of HIPPO signaling pathway, is an essential factor in cardiomyocyte survival; however, its status in human LV HF is not entirely elucidated. Here, we report that Yap is elevated in LV tissue of patients with HF, and is associated with down-regulation of its upstream inhibitor HIPPO component large tumor suppressor 1 (LATS1) activation as well as upregulation of the fibrosis marker connective tissue growth factor (CTGF). Applying the established profibrotic combined stress of TGFβ and hypoxia to human ventricular cardiac fibroblasts in vitro increased Yap protein levels, down-regulated LATS1 activation, increased cell proliferation and collagen I production, and decreased ribosomal protein S6 and S6 kinase phosphorylation, a hallmark of mTOR activation, without any significant effect on mTOR and raptor protein expression or phosphorylation of mTOR or 4E-binding protein 1 (4EBP1), a downstream effector of mTOR pathway. As previously reported in various cell types, TGFβ/hypoxia also enhanced cardiac fibroblast Akt and ERK1/2 phosphorylation, which was similar to our observation in LV tissues from HF patients. Further, depletion of Yap reduced TGFβ/hypoxia-induced cardiac fibroblast proliferation and Akt phosphorylation at Ser 473 and Thr308, without any significant effect on TGFβ/hypoxia-induced ERK1/2 activation or reduction in S6 and S6 kinase activities. Taken together, these data demonstrate that Yap is a mediator that promotes human cardiac fibroblast proliferation and suggest its possible contribution to remodeling of the LV, opening the door to further studies to decipher the cell-specific roles of Yap signaling in human HF.
Collapse
|
28
|
Xiong J, Dong X, Li S, Jiang F, Chen J, Yu S, Dong B, Su Q. Effects of (Pro)renin Receptor on Diabetic Cardiomyopathy Pathological Processes in Rats via the PRR-AMPK-YAP Pathway. Front Physiol 2021; 12:657378. [PMID: 34122131 PMCID: PMC8191636 DOI: 10.3389/fphys.2021.657378] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/08/2021] [Indexed: 01/12/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a common complication associated with diabetes. The (pro)renin receptor (PRR) is an important member of the local tissue renin-angiotensin system and plays a vital role in many cardiovascular diseases. Yes-associated protein (YAP) also plays a crucial role in many cardiovascular diseases. However, the mechanism responsible for the effects of PRR and YAP on DCM remains unclear. The purpose of this study was to determine the role of PRR in the pathological progression of DCM and whether PRR influences the pathological processes of diabetic cardiomyopathy through YAP. We first established diabetic cardiomyopathy rats model, downregulated the expression of PRR, and upregulated and downregulated the expression of YAP. The levels of myocardial inflammation and fibrosis were then measured and cardiac function was evaluated. In vitro, primary rat cardiac fibroblasts (CFs) were cultured with high glucose, with or without transfection with recombinant adenovirus expressing PRR, and GSK621 was used to observe the effect of AMPK. The levels of inflammation and fibrosis were measured in vitro. The results showed that PRR and YAP silencing alleviated myocardial inflammation and fibrosis. GSK621 blocked the effect of PRR on AMPK and YAP and improved CF inflammation and fibrosis. The inhibition of PRR expression offers a new therapeutic strategy for the treatment of DCM. The effects of PRR on the pathological process of DCM in rats may be mediated via the PRR-AMPK-YAP pathway.
Collapse
Affiliation(s)
- Jie Xiong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital of Shandong University, Jinan, China
| | - Xuefei Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,Department of Sport, Health and Exercise Science, University of Hull, Hull, United Kingdom
| | - Shengnan Li
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital of Shandong University, Jinan, China
| | - Fan Jiang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital of Shandong University, Jinan, China
| | - Jing Chen
- Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - Shiran Yu
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital of Shandong University, Jinan, China
| | - Bo Dong
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China.,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Qilu Hospital of Shandong University, Jinan, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
29
|
Okuyama M, Jiang W, Yang L, Subramanian V. Mst1/2 Kinases Inhibitor, XMU-MP-1, Attenuates Angiotensin II-Induced Ascending Aortic Expansion in Hypercholesterolemic Mice. Circ Rep 2021; 3:259-266. [PMID: 34007939 PMCID: PMC8099673 DOI: 10.1253/circrep.cr-20-0104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Background:
Ascending and abdominal aortic aneurysms (AAs) are asymptomatic, permanent dilations of the aorta with surgical intervention as the currently available therapy. Hippo-Yap signaling cascade plays a critical role in stem cell self-renewal, tissue regeneration and organ size control. By using XMU-MP-1, a pharmacological inhibitor of the key component of Hippo-Yap signaling, MST1/2, we examined the functional contribution of Hippo-Yap in the development of AAs in Angiotensin II (AngII)-infused hypercholesterolemic mice. Methods and Results:
MST, p-MST, p-YAP, p-MOB and TAZ proteins in AngII-infused ascending and abdominal aortas were assessed by immunohistochemical and western blot analyses. To examine the effect of MST1/2 inhibition on AAs, western diet-fed low density lipoprotein (LDL) receptor −/− mice infused with AngII were administered with either vehicle or XMU-MP-1 for 5 weeks. Hippo-YAP signaling proteins were significantly elevated in AngII infused ascending and abdominal aortas. XMU-MP-1 administration resulted in the attenuation of AngII-induced ascending AAs without influencing abdominal AAs and aortic atherosclerosis. Inhibition of Hippo-YAP signaling also resulted in the suppression of AngII-induced matrix metalloproteinase 2 (MMP2) activity, macrophage accumulation, aortic medial hypertrophy and elastin breaks in the ascending aorta. Conclusions:
The present study demonstrates a pivotal role for the Hippo-YAP signaling pathway in AngII-induced ascending AA development.
Collapse
Affiliation(s)
- Michihiro Okuyama
- Saha Cardiovascular Research Center, University of Kentucky Lexington, KY USA.,Department of Physiology, University of Kentucky Lexington, KY USA
| | - Weihua Jiang
- Saha Cardiovascular Research Center, University of Kentucky Lexington, KY USA
| | - Lihua Yang
- Saha Cardiovascular Research Center, University of Kentucky Lexington, KY USA
| | - Venkateswaran Subramanian
- Saha Cardiovascular Research Center, University of Kentucky Lexington, KY USA.,Department of Physiology, University of Kentucky Lexington, KY USA
| |
Collapse
|
30
|
Manno G, Filorizzo C, Fanale D, Brando C, Di Lisi D, Lunetta M, Bazan V, Russo A, Novo G. Role of the HIPPO pathway as potential key player in the cross talk between oncology and cardiology. Crit Rev Oncol Hematol 2021; 159:103246. [PMID: 33545354 DOI: 10.1016/j.critrevonc.2021.103246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
The HIPPO pathway (HP) is a highly conserved kinase cascade that affects organ size by regulating proliferation, cell survival and differentiation. Discovered in Drosophila melanogaster to early 2000, it immediately opened wide frontiers in the field of research. Over the last years the field of knowledge on HP is quickly expanding and it is thought will offer many answers on complex pathologies. Here, we summarized the results of several studies that have investigated HP signaling both in oncology than in cardiology field, with an overview on future perspectives in cardiology research.
Collapse
Affiliation(s)
- Girolamo Manno
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Clarissa Filorizzo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Chiara Brando
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Daniela Di Lisi
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Monica Lunetta
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, Palermo, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy.
| | - Giuseppina Novo
- Cardiology Unit, University Hospital P. Giaccone, Department of Excellence of Sciences for Health Promotion and Mothernal-Child Care, Internal Medicine and Specialities (ProMISE) "G. D'Alessandro", Palermo, Italy
| |
Collapse
|
31
|
Zhang HX, Zhang CS, Huang RZ, Cao X, Dai XQ, Zuo CY, Lv P, Zhu LJ, Yu SG. Oral administration of MnCl 2 attenuated hyperlipidemia-related cardiac remodeling in ApoE -/- mice. J Pharmacol Sci 2021; 145:167-174. [PMID: 33451751 DOI: 10.1016/j.jphs.2020.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/19/2020] [Accepted: 11/20/2020] [Indexed: 12/29/2022] Open
Abstract
Manganese chloride (MnCl2) has been shown to inhibit the Yes-associated protein (YAP) in high-fat diet-fed ApoE-/- mice. Although YAP has been implicated in atherogenesis, there are limited data on the effects of MnCl2 on cardiac remodeling. In this study, we discovered, by electrocardiography, that hyperlipidemia led to spontaneous supraventricular arrhythmia (SVA) in ApoE-/- (KO) mice, with 3 of 9 KO + MnCl2 mice (33%) exhibiting lower incidence of spontaneous SVA than KO mice (6 of 10 mice, 60%). Echocardiography revealed that reduced systolic function in KO mice was reversed by MnCl2 treatment. Oil Red O staining of the aortas and biochemical analysis of lipid levels showed that MnCl2 inhibited plaque formation in a lipid metabolism-independent manner. MnCl2 inhibited inflammatory cell infiltration and reduced fibrosis, as evidenced by hematoxylin and eosin, immunohistochemical and Masson's trichrome staining, respectively. Our findings demonstrate that spontaneous SVA and reduced systolic function were blocked by MnCl2. Our findings show that MnCl2 was useful in delaying cardiac remodeling and reducing susceptibility to spontaneous SVA in a mouse model of hyperlipidemia.
Collapse
Affiliation(s)
- Han-Xiao Zhang
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Cheng-Shun Zhang
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Rui-Zhen Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, 39 Shi-er-qiao Road, Jinniu District, Chengdu, 610072, Sichuan, China
| | - Xin Cao
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China.
| | - Xiao-Qin Dai
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| | - Chuan-Yi Zuo
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Peng Lv
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Li-Juan Zhu
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| | - Shu-Guang Yu
- Acupuncture and Tuina School/Third Teaching Hospital, Chengdu University of Traditional Chinese Medicine, 37 Shi-er-qiao Road, Jinniu District, Chengdu, 610075, Sichuan, China
| |
Collapse
|
32
|
Jiang W, Xue Y, Li H, Zhang H, Zhao Y. TAZ Is Related to Postoperative In-Hospital Mortality of Acute Type A Aortic Dissection. Front Cardiovasc Med 2020; 7:587996. [PMID: 33240939 PMCID: PMC7670070 DOI: 10.3389/fcvm.2020.587996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/29/2020] [Indexed: 01/16/2023] Open
Abstract
Background: Surgical repair of acute type A aortic dissection (ATAAD) has high risk and mortality, and there are few biomarkers of postoperative in-hospital mortality until now. This study investigated the association between WW domain–containing transcription regulator protein 1 (TAZ) and the postoperative in-hospital mortality of ATAAD patients. Methods: This is a retrospective cohort study. Data and blood samples were collected from 95 consecutive patients with ATAAD who underwent surgeries in our hospital from July 1, 2016, to December 31, 2016. The data collection included all the risk factors introduced by the modified EuroSCORE (European System for Cardiac Operative Risk Evaluation). The predictors of postoperative in-hospital death were confirmed by univariate regression analysis. Multivariable logistic regressions were used to analyze the association of the preoperative plasma level of TAZ and the postoperative in-hospital mortality of ATAAD patients. In addition, we used the generalized additive model to identify non-linear relationships. Results: Three models were used in the multivariable logistic regression analysis of the relationship between the preoperative plasma level of TAZ and postoperative in-hospital death. In the crude model, the preoperative plasma level of TAZ showed a positive correlation with postoperative in-hospital death [odds ratio (OR) = 1.33, 95% confidence interval (CI): 1.01–1.74, P = 0.04]. In adjusted model I and adjusted model II, similar results were found (OR = 1.35, 95% CI: 1.01–1.80, P = 0.04 and OR = 1.35, 95% CI: 1.01–1.81, P = 0.04). The risk of postoperative in-hospital death in the preoperative plasma level of the TAZ≥12.70 ng/mL group was 10.08 times (OR = 10.08, 95% CI: 1.63–62.37; P = 0.01) that of the preoperative plasma level of the TAZ <12.70 ng/mL group. Conclusions: The high preoperative plasma level of TAZ suggested poor surgical prognosis for ATAAD patients. The patients with a preoperative plasma level of TAZ ≥ 12.7 ng/ml had much higher postoperative in-hospital mortality.
Collapse
Affiliation(s)
- Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Yuan Xue
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Haibin Li
- Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, China.,Beijing Lab for Cardiovascular Precision Medicine, Beijing, China
| | - Yuanfei Zhao
- Centre for Transplant and Renal Research, The Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Yu Y, Su X, Qin Q, Hou Y, Zhang X, Zhang H, Jia M, Chen Y. Yes-associated protein and transcriptional coactivator with PDZ-binding motif as new targets in cardiovascular diseases. Pharmacol Res 2020; 159:105009. [DOI: 10.1016/j.phrs.2020.105009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/14/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022]
|
34
|
Common Regulatory Pathways Mediate Activity of MicroRNAs Inducing Cardiomyocyte Proliferation. Cell Rep 2020; 27:2759-2771.e5. [PMID: 31141697 PMCID: PMC6547019 DOI: 10.1016/j.celrep.2019.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 05/03/2018] [Accepted: 04/30/2019] [Indexed: 01/04/2023] Open
Abstract
Loss of functional cardiomyocytes is a major determinant of heart failure after myocardial infarction. Previous high throughput screening studies have identified a few microRNAs (miRNAs) that can induce cardiomyocyte proliferation and stimulate cardiac regeneration in mice. Here, we show that all of the most effective of these miRNAs activate nuclear localization of the master transcriptional cofactor Yes-associated protein (YAP) and induce expression of YAP-responsive genes. In particular, miR-199a-3p directly targets two mRNAs coding for proteins impinging on the Hippo pathway, the upstream YAP inhibitory kinase TAOK1, and the E3 ubiquitin ligase β-TrCP, which leads to YAP degradation. Several of the pro-proliferative miRNAs (including miR-199a-3p) also inhibit filamentous actin depolymerization by targeting Cofilin2, a process that by itself activates YAP nuclear translocation. Thus, activation of YAP and modulation of the actin cytoskeleton are major components of the pro-proliferative action of miR-199a-3p and other miRNAs that induce cardiomyocyte proliferation. A few microRNAs can stimulate cardiac myocyte proliferation The most effective of these microRNAs activate YAP Several pro-proliferative microRNAs also inhibit actin depolymerization miR-199a-3p directly targets TAOK1, b-TrCP, and Cofilin2 to achieve its effects
Collapse
|
35
|
Zheng M, Jacob J, Hung SH, Wang J. The Hippo Pathway in Cardiac Regeneration and Homeostasis: New Perspectives for Cell-Free Therapy in the Injured Heart. Biomolecules 2020; 10:biom10071024. [PMID: 32664346 PMCID: PMC7407108 DOI: 10.3390/biom10071024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 12/19/2022] Open
Abstract
Intractable cardiovascular diseases are leading causes of mortality around the world. Adult mammalian hearts have poor regenerative capacity and are not capable of self-repair after injury. Recent studies of cell-free therapeutics such as those designed to stimulate endogenous cardiac regeneration have uncovered new feasible therapeutic avenues for cardiac repair. The Hippo pathway, a fundamental pathway with pivotal roles in cell proliferation, survival and differentiation, has tremendous potential for therapeutic manipulation in cardiac regeneration. In this review, we summarize the most recent studies that have revealed the function of the Hippo pathway in heart regeneration and homeostasis. In particular, we discuss the molecular mechanisms of how the Hippo pathway maintains cardiac homeostasis by directing cardiomyocyte chromatin remodeling and regulating the cell-cell communication between cardiomyocytes and non-cardiomyocytes in the heart.
Collapse
Affiliation(s)
- Mingjie Zheng
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
| | - Joan Jacob
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Shao-Hsi Hung
- Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center and UTHealth, Houston, TX 77030, USA; (J.J.); (S.-H.H.)
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA;
- Correspondence: ; Tel.: +1-7135-005-723
| |
Collapse
|
36
|
Gong L, Wang S, Shen L, Liu C, Shenouda M, Li B, Liu X, Shaw JA, Wineman AL, Yang Y, Xiong D, Eichmann A, Evans SM, Weiss SJ, Si MS. SLIT3 deficiency attenuates pressure overload-induced cardiac fibrosis and remodeling. JCI Insight 2020; 5:136852. [PMID: 32644051 PMCID: PMC7406261 DOI: 10.1172/jci.insight.136852] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 05/06/2020] [Indexed: 01/28/2023] Open
Abstract
In pulmonary hypertension and certain forms of congenital heart disease, ventricular pressure overload manifests at birth and is an obligate hemodynamic abnormality that stimulates myocardial fibrosis, which leads to ventricular dysfunction and poor clinical outcomes. Thus, an attractive strategy is to attenuate the myocardial fibrosis to help preserve ventricular function. Here, by analyzing RNA-sequencing databases and comparing the transcript and protein levels of fibrillar collagen in WT and global-knockout mice, we found that slit guidance ligand 3 (SLIT3) was present predominantly in fibrillar collagen-producing cells and that SLIT3 deficiency attenuated collagen production in the heart and other nonneuronal tissues. We then performed transverse aortic constriction or pulmonary artery banding to induce left and right ventricular pressure overload, respectively, in WT and knockout mice. We discovered that SLIT3 deficiency abrogated fibrotic and hypertrophic changes and promoted long-term ventricular function and overall survival in both left and right ventricular pressure overload. Furthermore, we found that SLIT3 stimulated fibroblast activity and fibrillar collagen production, which coincided with the transcription and nuclear localization of the mechanotransducer yes-associated protein 1. These results indicate that SLIT3 is important for regulating fibroblast activity and fibrillar collagen synthesis in an autocrine manner, making it a potential therapeutic target for fibrotic diseases, especially myocardial fibrosis and adverse remodeling induced by persistent afterload elevation.
Collapse
Affiliation(s)
- Lianghui Gong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuyun Wang
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Li Shen
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Catherine Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mena Shenouda
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Baolei Li
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Xiaoxiao Liu
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Alan L. Wineman
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Yifeng Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Dingding Xiong
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA.,Paris Cardiovascular Research Center, INSERM U970, Paris, France.,Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sylvia M. Evans
- Skaggs School of Pharmacy and Pharmaceutical Sciences,,Department of Medicine, and,Department of Pharmacology, UCSD, La Jolla, California, USA
| | - Stephen J. Weiss
- Division of Genetic Medicine,,Department of Internal Medicine,,Life Sciences Institute,,Cellular and Molecular Biology Graduate Program, and,Rogel Cancer Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Ming-Sing Si
- Section of Pediatric Cardiovascular Surgery, Department of Cardiac Surgery, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
37
|
Liu J, Xu L, Zhan X. LncRNA MALAT1 regulates diabetic cardiac fibroblasts through the Hippo-YAP signaling pathway. Biochem Cell Biol 2020; 98:537-547. [PMID: 32069074 DOI: 10.1139/bcb-2019-0434] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Diabetic cardiomyopathy (DCM) is a major diabetes-related microvascular disease. LncRNA MALAT1 is widely expressed in cardiomyocytes responding to hypoxia and high levels of glucose (high glucose). In this study, cardiac fibroblasts (CFs) were transfected with si-MALAT1 and exposed to high glucose. CFs in the high glucose groups were treated with 30 mmol/L glucose, and the control CFs were treated with 5.5 mmol/L glucose. The expression of MALAT1 in the nucleus and cytoplasm of CFs was detected. The biological behavior of CFs, as well as collagen production, activity of the Hippo-YAP pathway, and nuclear localization of YAP were measured. Mouse models of DCM were established to observe the pathological changes to myocardium and determine the levels of collagen I, Bax, and Bcl-2. The interaction between MALAT1 and YAP was analyzed, and CREB expression in the high-glucose treated CFs was detected. MALAT1 was upregulated in high-glucose CFs and located in the nucleus. High-glucose increased collagen production, inflammation, cell proliferation, cell invasiveness, and phosphorylation of MST1 and LATS1, and also promoted nuclear translocation of YAP. These trends in high-glucose treated CFs and the DCM mice were reversed by transfection with si-MALAT1. MALAT1 positively regulated the nuclear translocation of YAP by binding to CREB. CREB levels were increased in the high-glucose CFs, but decreased after silencing MALAT1. These results indicate that si-MALAT1 reduces inflammation and collagen accumulation in high-glucose CFs and DCM mice via the Hippo-YAP pathway and CREB.
Collapse
Affiliation(s)
- Jiangwen Liu
- Endocrine and Metabolic Diseases, Harbin Medical University, Harbin 150001, Heilongjiang, P.R. China
| | - Liang Xu
- Department of Cardiology, The First Hospital of Harbin, Harbin 150001, Heilongjiang, P.R. China
| | - Xiaorong Zhan
- Department of Endocrinology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, P.R. China
| |
Collapse
|
38
|
Li HR, Wang C, Sun P, Liu DD, Du GQ, Tian JW. Melatonin attenuates doxorubicin-induced cardiotoxicity through preservation of YAP expression. J Cell Mol Med 2020; 24:3634-3646. [PMID: 32068341 PMCID: PMC7131936 DOI: 10.1111/jcmm.15057] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 01/10/2020] [Accepted: 01/22/2020] [Indexed: 12/21/2022] Open
Abstract
There are increasing concerns related to the cardiotoxicity of doxorubicin in the clinical setting. Recently, melatonin has been shown to exert a cardioprotective effect in various cardiovascular diseases, including cardiotoxic conditions. In this study, we examined the possible protective effects of melatonin on doxorubicin‐induced cardiotoxicity and explored the underlying mechanisms related to this process. We found that in vitro doxorubicin treatment significantly decreased H9c2 cell viability and induced apoptosis as manifested by increased TUNEL‐positive cells, down‐regulation of anti‐apoptotic protein Bcl‐2, as well as up‐regulation of pro‐apoptotic protein Bax. This was associated with increased reactive oxygen species (ROS) levels and decreased mitochondrial membrane potentials (MMP). In vivo, five weeks of doxorubicin treatment significantly decreased cardiac function, as evaluated by echocardiography. TUNEL staining results confirmed the increased apoptosis caused by doxorubicin. On the other hand, combinational treatment of doxorubicin with melatonin decreased cardiomyocyte ROS and apoptosis levels, along with increasing MMP. Such doxorubicin‐melatonin co‐treatment alleviated in vivo doxorubicin‐induced cardiac injury. Western Blots, along with in vitro immunofluorescence and in vivo immunohistochemical staining confirmed that doxorubicin treatment significantly down‐regulated Yes‐associated protein (YAP) expression, while YAP levels were maintained under co‐treatment of doxorubicin and melatonin. YAP inhibition by siRNA abolished the protective effects of melatonin on doxorubicin‐treated cardiomyocytes, with reversed ROS level and apoptosis. Our findings suggested that melatonin treatment attenuated doxorubicin‐induced cardiotoxicity through preserving YAP levels, which in turn decreases oxidative stress and apoptosis.
Collapse
Affiliation(s)
- Hai-Ru Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Chao Wang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Ping Sun
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Dan-Dan Liu
- Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guo-Qing Du
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| | - Jia-Wei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,Key Laboratories of Myocardial Ischemia Mechanism and Treatment, Harbin Medical University, Ministry of Education, Harbin, China
| |
Collapse
|
39
|
Ikeda S, Mizushima W, Sciarretta S, Abdellatif M, Zhai P, Mukai R, Fefelova N, Oka SI, Nakamura M, Del Re DP, Farrance I, Park JY, Tian B, Xie LH, Kumar M, Hsu CP, Sadayappan S, Shimokawa H, Lim DS, Sadoshima J. Hippo Deficiency Leads to Cardiac Dysfunction Accompanied by Cardiomyocyte Dedifferentiation During Pressure Overload. Circ Res 2019; 124:292-305. [PMID: 30582455 DOI: 10.1161/circresaha.118.314048] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
RATIONALE The Hippo pathway plays an important role in determining organ size through regulation of cell proliferation and apoptosis. Hippo inactivation and consequent activation of YAP (Yes-associated protein), a transcription cofactor, have been proposed as a strategy to promote myocardial regeneration after myocardial infarction. However, the long-term effects of Hippo deficiency on cardiac function under stress remain unknown. OBJECTIVE We investigated the long-term effect of Hippo deficiency on cardiac function in the presence of pressure overload (PO). METHODS AND RESULTS We used mice with cardiac-specific homozygous knockout of WW45 (WW45cKO), in which activation of Mst1 (Mammalian sterile 20-like 1) and Lats2 (large tumor suppressor kinase 2), the upstream kinases of the Hippo pathway, is effectively suppressed because of the absence of the scaffolding protein. We used male mice at 3 to 4 month of age in all animal experiments. We subjected WW45cKO mice to transverse aortic constriction for up to 12 weeks. WW45cKO mice exhibited higher levels of nuclear YAP in cardiomyocytes during PO. Unexpectedly, the progression of cardiac dysfunction induced by PO was exacerbated in WW45cKO mice, despite decreased apoptosis and activated cardiomyocyte cell cycle reentry. WW45cKO mice exhibited cardiomyocyte sarcomere disarray and upregulation of TEAD1 (transcriptional enhancer factor) target genes involved in cardiomyocyte dedifferentiation during PO. Genetic and pharmacological inactivation of the YAP-TEAD1 pathway reduced the PO-induced cardiac dysfunction in WW45cKO mice and attenuated cardiomyocyte dedifferentiation. Furthermore, the YAP-TEAD1 pathway upregulated OSM (oncostatin M) and OSM receptors, which played an essential role in mediating cardiomyocyte dedifferentiation. OSM also upregulated YAP and TEAD1 and promoted cardiomyocyte dedifferentiation, indicating the existence of a positive feedback mechanism consisting of YAP, TEAD1, and OSM. CONCLUSIONS Although activation of YAP promotes cardiomyocyte regeneration after cardiac injury, it induces cardiomyocyte dedifferentiation and heart failure in the long-term in the presence of PO through activation of the YAP-TEAD1-OSM positive feedback mechanism.
Collapse
Affiliation(s)
- Shohei Ikeda
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Wataru Mizushima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.).,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S. Sciarretta).,Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S. Sciarretta)
| | - Maha Abdellatif
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Risa Mukai
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Nadezhda Fefelova
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Shin-Ichi Oka
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Michinari Nakamura
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Dominic P Del Re
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | | | - Ji Yeon Park
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Bin Tian
- Department of Microbiology, Biochemistry, and Molecular Genetics, New Jersey Medical School, Rutgers University, Newark (J.Y.P., B.T.)
| | - Lai-Hua Xie
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, National Yang-Ming University School of Medicine, Taiwan (C.-P.H.)
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, OH (M.K., S. Sadayappan)
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan (S.I., H.S.)
| | - Dae-Sik Lim
- Department of Biological Science, National Creative Research Initiatives Center for Cell Division and Differentiation, Korea Advanced Institute of Science and Technology, Daejeon (D.-S.L.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (S.I., W.M., S. Sciarretta, M.A., P.Z., R.M., N.F., S.-i.O., M.N., D.P.D.R., L.-H.X., J.S.)
| |
Collapse
|
40
|
Mia MM, Singh MK. The Hippo Signaling Pathway in Cardiac Development and Diseases. Front Cell Dev Biol 2019; 7:211. [PMID: 31632964 PMCID: PMC6779857 DOI: 10.3389/fcell.2019.00211] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/12/2019] [Indexed: 01/07/2023] Open
Abstract
Heart disease continues to be the leading cause of morbidity and mortality worldwide. Cardiac malformation during development could lead to embryonic or postnatal death. However, matured heart tissue has a very limited regenerative capacity. Thus, loss of cardiomyocytes from injury or diseases in adults could lead to heart failure. The Hippo signaling pathway is a newly identified signaling cascade that modulates regenerative response by regulating cardiomyocyte proliferation in the embryonic heart, as well as in postnatal hearts after injury. In this review, we summarize recent findings highlighting the function and regulation of the Hippo signaling pathway in cardiac development and diseases.
Collapse
Affiliation(s)
- Masum M Mia
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Manvendra K Singh
- Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore.,National Heart Research Institute Singapore, National Heart Center, Singapore, Singapore
| |
Collapse
|
41
|
Abstract
The Hippo-YAP (Yes-associated protein) pathway is an evolutionarily and functionally conserved regulator of organ size and growth with crucial roles in cell proliferation, apoptosis, and differentiation. This pathway has great potential for therapeutic manipulation in different disease states and to promote organ regeneration. In this Review, we summarize findings from the past decade revealing the function and regulation of the Hippo-YAP pathway in cardiac development, growth, homeostasis, disease, and regeneration. In particular, we highlight the roles of the Hippo-YAP pathway in endogenous heart muscle renewal, including the pivotal role of the Hippo-YAP pathway in regulating cardiomyocyte proliferation and differentiation, stress response, and mechanical signalling. The human heart lacks the capacity to self-repair; therefore, the loss of cardiomyocytes after injury such as myocardial infarction can result in heart failure and death. Despite substantial advances in the treatment of heart failure, an enormous unmet clinical need exists for alternative treatment options. Targeting the Hippo-YAP pathway has tremendous potential for developing therapeutic strategies for cardiac repair and regeneration for currently intractable cardiovascular diseases such as heart failure. The lessons learned from cardiac repair and regeneration studies will also bring new insights into the regeneration of other tissues with limited regenerative capacity.
Collapse
|
42
|
Hu X, Shen G, Lu X, Ding G, Shen L. Identification of key proteins and lncRNAs in hypertrophic cardiomyopathy by integrated network analysis. Arch Med Sci 2019; 15:484-497. [PMID: 30899302 PMCID: PMC6425197 DOI: 10.5114/aoms.2018.75593] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/02/2018] [Indexed: 01/16/2023] Open
Abstract
INTRODUCTION Hypertrophic cardiomyopathy (HCM), a genetically heterogeneous disorder of cardiac myocytes, is one of the main causes of sudden cardiac death of young people. However, the molecular mechanism involved in HCM has remained largely unclear. Of note, non-coding RNAs were reported to play an important role in human diseases. In this study, we focused on identifying differentially expressed long non-coding RNA (lncRNAs) and mRNAs in HCM by analyzing a public dataset (GSE36961). MATERIAL AND METHODS We performed bioinformatics analysis to explore key pathways underlying HCM progression. Gene Ontology (GO) analysis was first performed to evaluate the potential roles of differentially expressed genes and lncRNAs in HCM. Moreover, protein-protein interaction (PPI) networks were constructed to reveal interactions among differentially expressed proteins. Specifically, co-expression networks were also constructed to identify hub lncRNAs in HCM. RESULTS A total of 6147 mRNAs (p < 0.001) and 126 lncRNAs (p < 0.001) were found to be dysregulated in HCM. Gene Ontology (GO) analysis showed that these differentially expressed genes and lncRNAs were associated with metabolism, energy pathways, signal transduction, and cell communication. Moreover, TSPYL3, LOC401431, LOC158376, LOC606724, PDIA3P and LOH3CR2A (p < 0.001) were identified as key lncRNAs in HCM progression. CONCLUSIONS Taken together, our analysis revealed a series of lncRNAs and mRNAs that were differentially expressed in HCM and which were involved in HCM progression by regulating pathways, such as metabolism, energy pathways, signal transduction, and cell communication. This study will provide useful information to explore the mechanisms underlying HCM progression and to provide potential candidate biomarkers for diagnosis in HCM.
Collapse
Affiliation(s)
- Xiaofeng Hu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| | - Guilin Shen
- Department of Cardiology, Anji People’s Hospital, Huzhou, Zhejiang Province, China
| | - Xiaoli Lu
- Department of Cardiology, Anji People’s Hospital, Huzhou, Zhejiang Province, China
| | - Guomin Ding
- Department of Cardiology, Anji People’s Hospital, Huzhou, Zhejiang Province, China
| | - Lishui Shen
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang Province, China
| |
Collapse
|
43
|
A positive feedback regulation of Heme oxygenase 1 by CELF1 in cardiac myoblast cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:209-218. [PMID: 30508596 DOI: 10.1016/j.bbagrm.2018.11.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 11/22/2018] [Accepted: 11/22/2018] [Indexed: 11/23/2022]
Abstract
As an RNA binding protein, CUG-BP Elav-like family (CELF) has been shown to be critical for heart biological functions. However, no reports have revealed the function of CELF1 in hypertrophic cardiomyopathy (HCM). Hinted by RNA immunoprecipitation-sequencing (RIP-seq) data, the influence of the CELF protein on heme oxygenase-1 (HO-1) expression was tested by modulating CELF1 levels. Cardiac hypertrophy is related to oxidative stress-induced damage. Hence, the cardiovascular system may be protected against further injury by upregulating the expression of antioxidant enzymes, such as HO-1. During the past two decades, research has demonstrated the central role of HO-1 in the protection against diseases. Thus, understanding the molecular mechanisms underlying the modulation of HO-1 expression is profoundly important for developing new strategies to prevent cardiac hypertrophy. To elucidate the molecular mechanisms underlying HO-1 regulation by the CELF protein, we performed RNA immunoprecipitation (RIP), biotin pull-down analysis, luciferase reporter and mRNA stability assays. We found that the expression of HO-1 was downregulated by CELF1 through the conserved GU-rich elements (GREs) in HO-1 3'UTR transcripts. Correspondingly, CELF1 expression was regulated by controlling the release of carbon monoxide (CO) in H9C2 cells. The CELF1-HO-1-CO regulation axis constituted a novel positive feedback circuit. In addition, we detected the potential involvement of CELF1 and HO-1 in samples from HCM patients. We found that CELF1 and CELF2, but not HO-1, were highly expressed in HCM heart samples. Thus, a manipulation targeting CELF1 could be developed as a potential therapeutic option for cardiac hypertrophy.
Collapse
|
44
|
|
45
|
Understanding the role of mammalian sterile 20-like kinase 1 (MST1) in cardiovascular disorders. J Mol Cell Cardiol 2018; 114:141-149. [DOI: 10.1016/j.yjmcc.2017.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 12/27/2022]
|
46
|
Jiang WJ, Ren WH, Liu XJ, Liu Y, Wu FJ, Sun LZ, Lan F, Du J, Zhang HJ. Disruption of mechanical stress in extracellular matrix is related to Stanford type A aortic dissection through down-regulation of Yes-associated protein. Aging (Albany NY) 2017; 8:1923-1939. [PMID: 27608489 PMCID: PMC5076445 DOI: 10.18632/aging.101033] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 09/06/2016] [Indexed: 01/19/2023]
Abstract
In this study, we assessed whether the down-regulation of Yes-associated protein (YAP) is involved in the pathogenesis of extracellular matrix (ECM) mechanical stress-induced Stanford type A aortic dissection (STAAD). Human aortic samples were obtained from heart transplantation donors as normal controls and from STAAD patients undergoing surgical replacement of the ascending aorta. Decreased maximum aortic wall velocity, ECM disorders, increased VSMC apoptosis, and YAP down-regulation were identified in STAAD samples. In a mouse model of STAAD, YAP was down-regulated over time during the development of ECM damage, and increased VSMC apoptosis was also observed. YAP knockdown induced VSMC apoptosis under static conditions in vitro, and the change in mechanical stress induced YAP down-regulation and VSMC apoptosis. This study provides evidence that YAP down-regulation caused by the disruption of mechanical stress is associated with the development of STAAD via the induction of apoptosis in aortic VSMCs. As STAAD is among the most elusive and life-threatening vascular diseases, better understanding of the molecular pathogenesis of STAAD is critical to improve clinical outcome.
Collapse
Affiliation(s)
- Wen-Jian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| | - Wei-Hong Ren
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Xu-Jie Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Yan Liu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Fu-Jian Wu
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Li-Zhong Sun
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| | - Feng Lan
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Jie Du
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China
| | - Hong-Jia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 10029, China.,Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 10029, China.,Beijing Laboratory for Cardiovascular Precision Medicine, Beijing 10029, China.,The Key Laboratory of Remodeling-related Cardiovascular Disease, Ministry of Education, Beijing 10029, China.,Beijing Aortic Disease Center, Cardiovascular Surgery Center, Beijing 10029, China.,Beijing Engineering Research Center for Vascular Prostheses, Beijing, China, 10029
| |
Collapse
|
47
|
Hou N, Wen Y, Yuan X, Xu H, Wang X, Li F, Ye B. Activation of Yap1/Taz signaling in ischemic heart disease and dilated cardiomyopathy. Exp Mol Pathol 2017; 103:267-275. [PMID: 29154888 DOI: 10.1016/j.yexmp.2017.11.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 01/20/2023]
Abstract
Genetic manipulation of key components of the evolutionally conserved Hippo pathway has shown that the precise control of these signaling molecules is critical to cardiac development and response to stresses. However, how this pathway is involved in the progression of cardiac dysfunction in different heart diseases remains unclear. We investigated the expressional levels and subcellular localization of Yap1, Taz, and Tead1 and determined Hippo target gene expression in failing human hearts with ischemic heart disease (IHD) and idiopathic dilated cardiomyopathy (IDC) and mouse desmin-related cardiomyopathy (DES). Our results demonstrated that Yap1, Taz, and Tead1 were significantly increased in failing human and DES hearts compared with the non-failing controls (NFH) or wild type (WT) mouse hearts at both mRNA and protein levels. Interestingly, adult human and mouse hearts had more Taz than Yap1 by mRNA and protein expression and their increases in diseased hearts were proportional and did not change Yap1/Taz ratio. Yap1, Taz, and Tead1 were accumulated in the nuclear fraction and cardiomyocyte nuclei of diseased hearts. The ratio of Yap1 phosphorylated at serine 127 (human) or serine 112 (mouse) to the total Yap1 (pYap1/Yap1) was significantly lower in the nuclear fraction of diseased hearts than that in normal controls. More importantly, Hippo downstream targets Ankrd1, Ctgf, and Cyr61 were transcriptionally elevated in the diseased hearts. These results suggest that Yap1/Taz signaling is activated in human and mouse dysfunctional hearts. Further investigation with relevant animal models will determine whether this pathway is a potential target for preventing and reversing abnormal remodeling during the progression of different cardiac disorders.
Collapse
Affiliation(s)
- Ning Hou
- Department of Pharmacology, School of Pharmaceutical Sciences, and The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ying Wen
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Xun Yuan
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Haodong Xu
- Department of Pathology, Center for Cardiovascular Biology and Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD 57069, USA
| | - Faqian Li
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA; Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA; Lillehei Heart Institute, Cancer & Cardiovascular Research Center, 2231 6th Street SE, Minneapolis, MN 55455, USA.
| | - Bo Ye
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
48
|
Pfleger CM. The Hippo Pathway: A Master Regulatory Network Important in Development and Dysregulated in Disease. Curr Top Dev Biol 2017; 123:181-228. [PMID: 28236967 DOI: 10.1016/bs.ctdb.2016.12.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Hippo Pathway is a master regulatory network that regulates proliferation, cell growth, stemness, differentiation, and cell death. Coordination of these processes by the Hippo Pathway throughout development and in mature organisms in response to diverse external and internal cues plays a role in morphogenesis, in controlling organ size, and in maintaining organ homeostasis. Given the importance of these processes, the Hippo Pathway also plays an important role in organismal health and has been implicated in a variety of diseases including eye disease, cardiovascular disease, neurodegeneration, and cancer. This review will focus on Drosophila reports that identified the core components of the Hippo Pathway revealing specific downstream biological outputs of this complicated network. A brief description of mammalian reports will complement review of the Drosophila studies. This review will also survey upstream regulation of the core components with a focus on feedback mechanisms.
Collapse
Affiliation(s)
- Cathie M Pfleger
- The Icahn School of Medicine at Mount Sinai, New York, NY, United States; The Graduate School of Biomedical Sciences, The Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
49
|
Xin Z, Ma Z, Jiang S, Wang D, Fan C, Di S, Hu W, Li T, She J, Yang Y. FOXOs in the impaired heart: New therapeutic targets for cardiac diseases. Biochim Biophys Acta Mol Basis Dis 2016; 1863:486-498. [PMID: 27890702 DOI: 10.1016/j.bbadis.2016.11.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 10/24/2016] [Accepted: 11/23/2016] [Indexed: 01/17/2023]
Abstract
Cardiac diseases have a high morbidity and mortality and affect the global population. Based on recent accumulating evidence, Forkhead box O (FOXOs) play important roles in cardiac diseases. Therefore, a summary of the current literature on the molecular mechanisms and roles of FOXOs in the heart will provide valuable information. In this review, we first briefly introduce the molecular features of FOXOs. Then, we discuss the regulation and cardiac actions of the FOXO pathways. Based on this background, we expand our discussion to the roles of FOXOs in several major cardiac diseases, such as ischemic cardiac diseases, diabetic cardiomyopathy and myocardial hypertrophy. Then, we describe some methodological problems associated with the FOXO gene-modified animal models. Finally, we discuss potential future directions. The information reviewed here may be significant for the design of future studies and may increase the potential of FOXOs as therapeutic targets.
Collapse
Affiliation(s)
- Zhenlong Xin
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shuai Jiang
- Department of Aerospace Medicine, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an 710061, China.
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China; Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China.
| |
Collapse
|
50
|
Ren CW, Liu JJ, Li JH, Li JW, Dai J, Lai YQ. RNA‑seq profiling of mRNA associated with hypertrophic cardiomyopathy. Mol Med Rep 2016; 14:5573-5586. [PMID: 27840985 DOI: 10.3892/mmr.2016.5931] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 09/20/2016] [Indexed: 11/06/2022] Open
Abstract
The mechanisms of hypertrophic cardiomyopathy (HCM) pathogenesis can be investigated by determining the differences between healthy and disease states at the molecular level. In the present study, large‑scale transcriptome sequencing was performed to compare mRNA expression in patients with HCM and control groups using an Illumina sequencing platform. Compared with the genome background, 257 differentially expressed genes (DEGs) were identified in which 62 genes were downregulated and 195 genes were upregulated. Reverse transcription‑quantitative polymerase chain reaction was performed to validate the expression pattern of certain mRNAs. Gene ontology enrichment and KEGG analysis of mRNAs was conducted to identify the biological modules and pathological pathways associated with the DEGs. To the best of our knowledge, this is the first time study to investigate the differences in mRNA between patients with HCM and normal controls at the transcriptome level. The results of the study will contributed to the understanding of the important molecular mechanisms involved in HCM and aid the selection of key genes to investigate in the future.
Collapse
Affiliation(s)
- Chang-Wei Ren
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jia-Ji Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jin-Hua Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jing-Wei Li
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Jiang Dai
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Yong-Qiang Lai
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| |
Collapse
|