1
|
Shaikh T, Nguyen D, Dugal JK, DiCaro MV, Yee B, Houshmand N, Lei K, Namazi A. Arrhythmogenic Right Ventricular Cardiomyopathy: A Comprehensive Review. J Cardiovasc Dev Dis 2025; 12:71. [PMID: 39997505 PMCID: PMC11855979 DOI: 10.3390/jcdd12020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is characterized by structural abnormalities, arrhythmias, and a spectrum of genetic and clinical manifestations. Clinically, ARVC is structurally distinguished by right ventricular dilation due to increased adiposity and fibrosis in the ventricular walls, and it manifests as cardiac arrhythmias ranging from non-sustained ventricular tachycardia to sudden cardiac death. Its prevalence has been estimated to range from 1 in every 1000 to 5000 people, with its large range being attributed to the variability in genetic penetrance from asymptomatic to significant burden. It is even suggested that the prevalence is underestimated, as the presence of genotypic mutations does not always lead to clinical manifestations that would facilitate diagnosis. Additionally, while set criteria have been in place since the 1990s, newer understanding of this condition and advancements in cardiac technology have prompted multiple revisions in the diagnostic criteria for ARVC. Novel discoveries of gene variants predisposing patients to ARVC have led to established screening techniques while providing insight into genetic counseling and management. This review aims to provide an overview of the genetics, pathophysiology, and clinical approach to ARVC. It will also focus on clinical presentation, ARVC diagnostic criteria, electrophysiological findings, including electrocardiogram characteristics, and imaging findings from cardiac MRI, 2D, and 3D echocardiogram. Current management options-including anti-arrhythmic medications, device indications, and ablation techniques-and the effectiveness of treatment will also be reviewed.
Collapse
Affiliation(s)
- Taha Shaikh
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Darren Nguyen
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Jasmine K. Dugal
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Michael V. DiCaro
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Brianna Yee
- Department of Internal Medicine, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (T.S.); (D.N.); (J.K.D.); (B.Y.)
| | - Nazanin Houshmand
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| | - KaChon Lei
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| | - Ali Namazi
- Department of Internal Medicine, Division of Cardiology, University of Nevada Las Vegas, Las Vegas, NV 89154, USA; (N.H.); (K.L.); (A.N.)
| |
Collapse
|
2
|
Yin H, Li X, Lu D, Zhao X, Yang Z, Wang Z, Xu F, Chen Y, Li C. Myofibrillogenesis Regulator-1 in Smooth Muscle Cells Modulates Inflammation Signaling Pathways via Regulating ROCK1 Ubiquitination and Degradation to Impact Aortic Dissection. J Inflamm Res 2025; 18:1719-1738. [PMID: 39931165 PMCID: PMC11808051 DOI: 10.2147/jir.s485163] [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] [Received: 08/28/2024] [Accepted: 11/29/2024] [Indexed: 02/13/2025] Open
Abstract
Background Aortic dissection (AD) is a life-threatening cardiovascular emergency and currently lacks effective drug treatment. Inflammation is a critical mechanism in the development of AD, and identifying specific molecular targets to regulate inflammation is crucial for stopping its progression. This study aimed to investigate the role of MR-1 and ROCK1 in the regulation of inflammation in AD and their potentialities as therapeutic targets. Methods Researchers performed protein immunoblotting on aortic wall tissue from 10 patients who underwent aortic arch replacement and 10 patients who underwent coronary artery bypass grafting to examine the expression levels of MR-1, ROCK1, and inflammatory pathways in the aortas. In vitro experiments, human aortic smooth muscle cells were extracted, and an in vitro dissection model was constructed with angiotensin II. siRNA silencing studies were performed to investigate the effects of MR-1 and ROCK1 on the development of AD and their interconnections. Results Analysis of aortic tissues revealed significantly elevated levels of MR-1 and ROCK1 in AD patients, and meanwhile the inflammatory indexes showed the same trend. Furthermore, it was observed that overexpression of MR-1 and ROCK1 facilitated smooth muscle cell phenotypic transformation and augmented matrix metalloproteinase release in in vitro settings through inflammatory pathway activation. The relationship between MR-1 and ROCK1 was elucidated, too. Conclusion MR-1 and ROCK1 overexpression is associated with the occurrence and development of AD through inflammation. This study highlights the role of inflammation in AD development and tap the potentiality of using MR-1 and ROCK1 as targets to alleviate AD development.
Collapse
Affiliation(s)
- Hang Yin
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Xiaoxing Li
- Department of Geriatrics, Qilu Hospital, Shandong University, Jinan, Shandong, People’s Republic of China
| | - Dazhou Lu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Xin Zhao
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, People’s Republic of China
| | - Zeyu Yang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Zerui Wang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Feng Xu
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Yuguo Chen
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| | - Chuanbao Li
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Chest Pain Center, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Shandong Provincial Engineering Laboratory for Emergency and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- Shandong Key Laboratory: Magnetic Field-Free Medicine & Functional Imaging (MF), Qilu Hospital of Shandong University, Jinan, People’s Republic of China
- NMPA Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Qilu Hospital of Shandong University, Jinan, People’s Republic of China
| |
Collapse
|
3
|
Sedmera D, Olejnickova V, Sankova B, Kolesova H, Bartos M, Kvasilova A, Phillips LC, Bamforth SD, Phillips HM. Morphological, electrophysiological, and molecular alterations in foetal noncompacted cardiomyopathy induced by disruption of ROCK signalling. Front Cell Dev Biol 2024; 12:1471751. [PMID: 39435333 PMCID: PMC11491540 DOI: 10.3389/fcell.2024.1471751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Left ventricular noncompaction cardiomyopathy is associated with heart failure, arrhythmia, and sudden cardiac death. The developmental mechanism underpinning noncompaction in the adult heart is still not fully understood, with lack of trabeculae compaction, hypertrabeculation, and loss of proliferation cited as possible causes. To study this, we utilised a mouse model of aberrant Rho kinase (ROCK) signalling in cardiomyocytes, which led to a noncompaction phenotype during embryogenesis, and monitored how this progressed after birth and into adulthood. The cause of the early noncompaction at E15.5 was attributed to a decrease in proliferation in the developing ventricular wall. By E18.5, the phenotype became patchy, with regions of noncompaction interspersed with thick compacted areas of ventricular wall. To study how this altered myoarchitecture of the heart influenced impulse propagation in the developing and adult heart, we used histology with immunohistochemistry for gap junction protein expression, optical mapping, and electrocardiography. At the prenatal stages, a clear reduction in left ventricular wall thickness, accompanied by abnormal conduction of the ectopically paced beat in that area, was observed in mutant hearts. This correlated with increased expression of connexin-40 and connexin-43 in noncompacted trabeculae. In postnatal stages, left ventricular noncompaction was resolved, but the right ventricular wall remained structurally abnormal through to adulthood with cardiomyocyte hypertrophy and retention of myocardial crypts. Thus, this is a novel model of self-correcting embryonic hypertrabeculation cardiomyopathy, but it highlights that remodelling potential differs between the left and right ventricles. We conclude that disruption of ROCK signalling induces both morphological and electrophysiological changes that evolve over time, highlighting the link between myocyte proliferation and noncompaction phenotypes and electrophysiological differentiation.
Collapse
Affiliation(s)
- David Sedmera
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Laboratory of Developmental Cardiology, Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| | - Veronika Olejnickova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Barbora Sankova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Hana Kolesova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Martin Bartos
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
- Institute of Dental Medicine, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Alena Kvasilova
- Institute of Anatomy, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Lauren C. Phillips
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon D. Bamforth
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen M. Phillips
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Fan X, Yang G, Duru F, Grilli M, Akin I, Zhou X, Saguner AM, Ei-Battrawy I. Arrhythmogenic Cardiomyopathy: from Preclinical Models to Genotype-phenotype Correlation and Pathophysiology. Stem Cell Rev Rep 2023; 19:2683-2708. [PMID: 37731079 PMCID: PMC10661732 DOI: 10.1007/s12015-023-10615-0] [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] [Accepted: 08/23/2023] [Indexed: 09/22/2023]
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a hereditary myocardial disease characterized by the replacement of the ventricular myocardium with fibrous fatty deposits. ACM is usually inherited in an autosomal dominant pattern with variable penetrance and expressivity, which is mainly related to ventricular tachyarrhythmia and sudden cardiac death (SCD). Importantly, significant progress has been made in determining the genetic background of ACM due to the development of new techniques for genetic analysis. The exact molecular pathomechanism of ACM, however, is not completely clear and the genotype-phenotype correlations have not been fully elucidated, which are useful to predict the prognosis and treatment of ACM patients. Different gene-targeted and transgenic animal models, human-induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) models, and heterologous expression systems have been developed. Here, this review aims to summarize preclinical ACM models and platforms promoting our understanding of the pathogenesis of ACM and assess their value in elucidating the ACM genotype-phenotype relationship.
Collapse
Affiliation(s)
- Xuehui Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Guoqiang Yang
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- Department of Acupuncture and Rehabilitation, the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- Research Unit of Molecular Imaging Probes, Department of Radiologic Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Firat Duru
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Maurizio Grilli
- Faculty of Medicine, University Medical Centre Mannheim (UMM), University of Heidelberg, Mannheim, Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany
| | - Xiaobo Zhou
- Key Laboratory of Medical Electrophysiology, Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention of Cardiovascular Diseases, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- First Department of Medicine, University Medical Centre Mannheim, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany.
| | - Ardan Muammer Saguner
- Department of Cardiology, University Heart Centre, University Hospital Zurich, Zurich, Switzerland
| | - Ibrahim Ei-Battrawy
- European Center for AngioScience (ECAS), German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/ Mannheim, and Centre for Cardiovascular Acute Medicine Mannheim (ZKAM), Medical Centre Mannheim, Heidelberg University, Partner Site, Heidelberg-Mannheim, Germany.
- Department of Cardiology and Angiology, Ruhr University, Bochum, Germany; Institute of Physiology, Department of Cellular and Translational Physiology and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr- University Bochum, Bochum, Germany.
| |
Collapse
|
5
|
Shoykhet M, Dervishi O, Menauer P, Hiermaier M, Moztarzadeh S, Osterloh C, Ludwig RJ, Williams T, Gerull B, Kääb S, Clauss S, Schüttler D, Waschke J, Yeruva S. EGFR inhibition leads to enhanced desmosome assembly and cardiomyocyte cohesion via ROCK activation. JCI Insight 2023; 8:163763. [PMID: 36795511 PMCID: PMC10070108 DOI: 10.1172/jci.insight.163763] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/15/2023] [Indexed: 02/17/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is a familial heart disease partly caused by impaired desmosome turnover. Thus, stabilization of desmosome integrity may provide new treatment options. Desmosomes, apart from cellular cohesion, provide the structural framework of a signaling hub. Here, we investigated the role of the epidermal growth factor receptor (EGFR) in cardiomyocyte cohesion. We inhibited EGFR under physiological and pathophysiological conditions using the murine plakoglobin-KO AC model, in which EGFR was upregulated. EGFR inhibition enhanced cardiomyocyte cohesion. Immunoprecipitation showed an interaction of EGFR and desmoglein 2 (DSG2). Immunostaining and atomic force microscopy (AFM) revealed enhanced DSG2 localization and binding at cell borders upon EGFR inhibition. Enhanced area composita length and desmosome assembly were observed upon EGFR inhibition, confirmed by enhanced DSG2 and desmoplakin (DP) recruitment to cell borders. PamGene Kinase assay performed in HL-1 cardiomyocytes treated with erlotinib, an EGFR inhibitor, revealed upregulation of Rho-associated protein kinase (ROCK). Erlotinib-mediated desmosome assembly and cardiomyocyte cohesion were abolished upon ROCK inhibition. Thus, inhibiting EGFR and, thereby, stabilizing desmosome integrity via ROCK might provide treatment options for AC.
Collapse
Affiliation(s)
- Maria Shoykhet
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Orsela Dervishi
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Philipp Menauer
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Matthias Hiermaier
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sina Moztarzadeh
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Colin Osterloh
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Ralf J Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, Lübeck, Germany
| | - Tatjana Williams
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Brenda Gerull
- Comprehensive Heart Failure Center and Department of Medicine I, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Dominik Schüttler
- Medizinische Klinik und Poliklinik I, LMU Hospital, LMU, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modeling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, LMU Hospital, LMU, Munich, Germany
| | - Jens Waschke
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Sunil Yeruva
- Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| |
Collapse
|
6
|
Kopecny LR, Lee BWH, Coroneo MT. A systematic review on the effects of ROCK inhibitors on proliferation and/or differentiation in human somatic stem cells: A hypothesis that ROCK inhibitors support corneal endothelial healing via acting on the limbal stem cell niche. Ocul Surf 2023; 27:16-29. [PMID: 36586668 DOI: 10.1016/j.jtos.2022.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022]
Abstract
Rho kinase inhibitors (ROCKi) have attracted growing multidisciplinary interest, particularly in Ophthalmology where the question as to how they promote corneal endothelial healing remains unresolved. Concurrently, stem cell biology has rapidly progressed in unravelling drivers of stem cell (SC) proliferation and differentiation, where mechanical niche factors and the actin cytoskeleton are increasingly recognized as key players. There is mounting evidence from the study of the peripheral corneal endothelium that supports the likelihood of an internal limbal stem cell niche. The possibility that ROCKi stimulate the endothelial SC niche has not been addressed. Furthermore, there is currently a paucity of data that directly evaluates whether ROCKi promotes corneal endothelial healing by acting on this limbal SC niche located near the transition zone. Therefore, we performed a systematic review examining the effects ROCKi on the proliferation and differentiation of human somatic SC, to provide insight into its effects on various human SC populations. An appraisal of electronic searches of four databases identified 1 in vivo and 58 in vitro studies (36 evaluated proliferation while 53 examined differentiation). Types of SC studied included mesenchymal (n = 32), epithelial (n = 11), epidermal (n = 8), hematopoietic and other (n = 8). The ROCK 1/2 selective inhibitor Y-27632 was used in almost all studies (n = 58), while several studies evaluated ≥2 ROCKi (n = 4) including fasudil, H-1152, and KD025. ROCKi significantly influenced human somatic SC proliferation in 81% of studies (29/36) and SC differentiation in 94% of studies (50/53). The present systemic review highlights that ROCKi are influential in regulating human SC proliferation and differentiation, and provides evidence to support the hypothesis that ROCKi promotes corneal endothelial division and maintenance via acting on the inner limbal SC niche.
Collapse
Affiliation(s)
- Lloyd R Kopecny
- School of Clinical Medicine, University of New South Wales, Sydney, Australia.
| | - Brendon W H Lee
- Department of Ophthalmology, School of Clinical Medicine, University of New South Wales, Level 2 South Wing, Edmund Blacket Building, Prince of Wales Hospital, Randwick, NSW, 2031, Australia
| | - Minas T Coroneo
- Department of Ophthalmology, Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
7
|
Yeruva S, Waschke J. Structure and regulation of desmosomes in intercalated discs: Lessons from epithelia. J Anat 2022; 242:81-90. [PMID: 35128661 PMCID: PMC9773171 DOI: 10.1111/joa.13634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
For electromechanical coupling of cardiomyocytes, intercalated discs (ICDs) are pivotal as highly specialized intercellular contact areas. ICD consists of adhesive contacts, such as desmosomes and adherens junctions (AJs) that are partially intermingled and thereby form an area composita to provide mechanical strength, as well as gap junctions (GJ) and sodium channels for excitation propagation. In contrast, in epithelia, mixed junctions with features of desmosomes and AJs are regarded as transitory primarily during the formation of desmosomes. The anatomy of desmosomes is defined by a typical ultrastructure with dense intracellular plaques anchoring the cadherin-type adhesion molecules to the intermediate filament cytoskeleton. Desmosomal diseases characterized by impaired adhesive and signalling functions of desmosomal contacts lead to arrhythmogenic cardiomyopathy when affecting cardiomyocytes and cause pemphigus when manifesting in keratinocytes or present as cardiocutaneous syndromes when both cell types are targeted by the disease, which underscores the high biomedical relevance of these cell contacts. Therefore, comparative analyses regarding the structure and regulation of desmosomal contacts in cardiomyocytes and epithelial cells are helpful to better understand disease pathogenesis. In this brief review, we describe the structural properties of ICD compared to epithelial desmosomes and suggest that mechanisms regulating adhesion may at least in part be comparable. Also, we discuss whether phenomena such as hyperadhesion or the bidirectional regulation of desmosomes to serve as signalling hubs in epithelial cells may also be relevant for ICD.
Collapse
Affiliation(s)
- Sunil Yeruva
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| | - Jens Waschke
- Ludwig‐Maximilian‐Universität München, Anatomische Anstalt, Lehrstuhl Anatomie I – Vegetative AnatomieMunichGermany
| |
Collapse
|
8
|
Shi J, Wei L. Rho Kinases in Embryonic Development and Stem Cell Research. Arch Immunol Ther Exp (Warsz) 2022; 70:4. [PMID: 35043239 PMCID: PMC8766376 DOI: 10.1007/s00005-022-00642-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022]
Abstract
The Rho-associated coiled-coil containing kinases (ROCKs or Rho kinases) belong to the AGC (PKA/PKG/PKC) family of serine/threonine kinases and are major downstream effectors of small GTPase RhoA, a key regulator of actin-cytoskeleton reorganization. The ROCK family contains two members, ROCK1 and ROCK2, which share 65% overall identity and 92% identity in kinase domain. ROCK1 and ROCK2 were assumed to be functionally redundant, based largely on their major common activators, their high degree kinase domain homology, and study results from overexpression with kinase constructs or chemical inhibitors. ROCK signaling research has expanded to all areas of biology and medicine since its discovery in 1996. The rapid advance is befitting ROCK’s versatile functions in modulating various cell behavior, such as contraction, adhesion, migration, proliferation, polarity, cytokinesis, and differentiation. The rapid advance is noticeably driven by an extensive linking with clinical medicine, including cardiovascular abnormalities, aberrant immune responsive, and cancer development and metastasis. The rapid advance during the past decade is further powered by novel biotechnologies including CRISPR-Cas and single cell omics. Current consensus, derived mainly from gene targeting and RNA interference approaches, is that the two ROCK isoforms have overlapping and distinct cellular, physiological and pathophysiology roles. In this review, we present an overview of the milestone discoveries in ROCK research. We then focus on the current understanding of ROCK signaling in embryonic development, current research status using knockout and knockin mouse models, and stem cell research.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of Medicine, Indiana University, 1044 West Walnut Street, R4-370, Indianapolis, IN, 46202-5225, USA.
| |
Collapse
|
9
|
Xu Z, Hu Z, Xu H, Zhang L, Li L, Wang Y, Zhu Y, Yang L, Hu D. Liquiritigenin alleviates doxorubicin-induced chronic heart failure via promoting ARHGAP18 and suppressing RhoA/ROCK1 pathway. Exp Cell Res 2022; 411:113008. [PMID: 34990617 DOI: 10.1016/j.yexcr.2022.113008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/30/2021] [Accepted: 01/01/2022] [Indexed: 12/01/2022]
Abstract
Chronic heart failure (CHF) is one of the most common chronic diseases with increasing incidence and mortality. Liquiritigenin (LQG) is shown to protect mice from cardiotoxicity. However, its underlying mechanism remains unclear. Our study aimed to reveal the role of ARHGAP18 in LQG-mediated cardioprotective effects in CHF. In the current study, CHF cell model and rat model were established by the application of doxorubicin (DOX). The reactive oxygen species (ROS) level and cell apoptosis were determined by flow cytometry. The cardiac function of rats was evaluated by measuring left ventricular systolic pressure, left ventricular end diastolic pressure, and serum level of lactate dehydrogenase and brain natriuretic peptide. The expression of active RhoA was elevated and that of ARHGAP18 was decreased in DOX-induced CHF cell model. ARHGAP18 could reduce DOX-induced RhoA activation, ROS elevation, and cell apoptosis. Meanwhile, the knockdown of ARHGAP18 could promote the activation of RhoA, the level of ROS, and the rate of cell apoptosis, which could be reversed by the application of RhoA inhibitor. LQG promoted the expression of ARHGAP18 and exerted similar effects of ARHGAP18 in CHF cell model. The application of LQG could also reverse the effects mediated by ARHGAP18 knockdown. Moreover, LQG significantly improved cardiac function and ameliorated DOX-induced cardiotoxicity of CHF rats. In conclusion, LQG could alleviate DOX-induced CHF via promoting ARHGAP18 and suppressing RhoA/ROCK1 pathway. LQG was a potential agent for CHF treatment.
Collapse
Affiliation(s)
- Zhibing Xu
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Zongde Hu
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China
| | - Hanchen Xu
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, China
| | - Lifen Zhang
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Liang Li
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Yi Wang
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Yuanqing Zhu
- Department of Emergency, Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, China
| | - Limeng Yang
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China.
| | - Dan Hu
- Department of Traditional Chinese Medicine, Shanghai Pudong New Area Hospital of Traditional Chinese Medicine, China.
| |
Collapse
|
10
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
11
|
Deciphering the Role of Wnt and Rho Signaling Pathway in iPSC-Derived ARVC Cardiomyocytes by In Silico Mathematical Modeling. Int J Mol Sci 2021; 22:ijms22042004. [PMID: 33670616 PMCID: PMC7923182 DOI: 10.3390/ijms22042004] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/29/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Arrhythmogenic Right Ventricular cardiomyopathy (ARVC) is an inherited cardiac muscle disease linked to genetic deficiency in components of the desmosomes. The disease is characterized by progressive fibro-fatty replacement of the right ventricle, which acts as a substrate for arrhythmias and sudden cardiac death. The molecular mechanisms underpinning ARVC are largely unknown. Here we propose a mathematical model for investigating the molecular dynamics underlying heart remodeling and the loss of cardiac myocytes identity during ARVC. Our methodology is based on three computational models: firstly, in the context of the Wnt pathway, we examined two different competition mechanisms between β-catenin and Plakoglobin (PG) and their role in the expression of adipogenic program. Secondly, we investigated the role of RhoA-ROCK pathway in ARVC pathogenesis, and thirdly we analyzed the interplay between Wnt and RhoA-ROCK pathways in the context of the ARVC phenotype. We conclude with the following remark: both Wnt/β-catenin and RhoA-ROCK pathways must be inactive for a significant increase of PPARγ expression, suggesting that a crosstalk mechanism might be responsible for mediating ARVC pathogenesis.
Collapse
|
12
|
Satoh K. Drug discovery focused on novel pathogenic proteins for pulmonary arterial hypertension. J Cardiol 2021; 78:1-11. [PMID: 33563508 DOI: 10.1016/j.jjcc.2021.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 10/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease in which the wall thickening and narrowing of pulmonary microvessels progress due to complicated interactions among processes such as endothelial dysfunction, the proliferation of pulmonary artery smooth muscle cells (PASMCs) and adventitial fibrocytes, and inflammatory cell infiltration. Early diagnosis of patients with PAH is difficult and lung transplantation is the only last choice to save severely ill patients. However, the number of donors is limited. Many patients with PAH show rapid progression and a high degree of pulmonary arterial remodeling characterized by the abnormal proliferation of PASMCs, which makes treatment difficult even with multidrug therapy comprising pulmonary vasodilators. Thus, it is important to develop novel therapy targeting factors other than vasodilation, such as PASMC proliferation. In the development of PAH, inflammation and oxidative stress are deeply involved in its pathogenesis. Excessive proliferation and apoptosis resistance in PASMCs are key mechanisms underlying PAH. Based on those characteristics, we recently screened novel pathogenic proteins and have performed drug discovery targeting those proteins. To confirm the clinical significance of this, we used patient-derived blood samples to evaluate biomarker potential for diagnosis and prognosis. Moreover, we conducted high throughput screening and found several inhibitors of the pathogenic proteins. In this review, we introduce the recent progress on basic and clinical PAH research, focusing on the screening of pathogenic proteins and drug discovery.
Collapse
Affiliation(s)
- Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan.
| |
Collapse
|
13
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
14
|
Yu B, Sladojevic N, Blair JE, Liao JK. Targeting Rho-associated coiled-coil forming protein kinase (ROCK) in cardiovascular fibrosis and stiffening. Expert Opin Ther Targets 2020; 24:47-62. [PMID: 31906742 PMCID: PMC7662835 DOI: 10.1080/14728222.2020.1712593] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 01/04/2020] [Indexed: 02/07/2023]
Abstract
Introduction: Pathological cardiac fibrosis, through excessive extracellular matrix protein deposition from fibroblasts and pro-fibrotic immune responses and vascular stiffening is associated with most forms of cardiovascular disease. Pathological cardiac fibrosis and stiffening can lead to heart failure and arrythmias and vascular stiffening may lead to hypertension. ROCK, a serine/threonine kinase downstream of the Rho-family of GTPases, may regulate many pro-fibrotic and pro-stiffening signaling pathways in numerous cell types.Areas covered: This article outlines the molecular mechanisms by which ROCK in fibroblasts, T helper cells, endothelial cells, vascular smooth muscle cells, and macrophages mediate fibrosis and stiffening. We speculate on how ROCK could be targeted to inhibit cardiovascular fibrosis and stiffening.Expert opinion: Critical gaps in knowledge must be addressed if ROCK inhibitors are to be used in the clinic. Numerous studies indicate that each ROCK isoform may play differential roles in regulating fibrosis and may have opposing roles in specific tissues. Future work needs to highlight the isoform- and tissue-specific contributions of ROCK in fibrosis, and how isoform-specific ROCK inhibitors in murine models and in clinical trials affect the pathophysiology of cardiac fibrosis and stiffening. This could progress knowledge regarding new treatments for heart failure, arrythmias and hypertension and the repair processes after myocardial infarction.
Collapse
Affiliation(s)
- Brian Yu
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Nikola Sladojevic
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - John E Blair
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - James K Liao
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
15
|
Satoh K, Shimokawa H. Recent Advances in the Development of Cardiovascular Biomarkers. Arterioscler Thromb Vasc Biol 2019; 38:e61-e70. [PMID: 29695533 DOI: 10.1161/atvbaha.118.310226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| |
Collapse
|
16
|
Bailey KE, MacGowan GA, Tual-Chalot S, Phillips L, Mohun TJ, Henderson DJ, Arthur HM, Bamforth SD, Phillips HM. Disruption of embryonic ROCK signaling reproduces the sarcomeric phenotype of hypertrophic cardiomyopathy. JCI Insight 2019; 5:125172. [PMID: 30835717 PMCID: PMC6538384 DOI: 10.1172/jci.insight.125172] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Sarcomeric disarray is a hallmark of gene mutations in patients with hypertrophic cardiomyopathy (HCM). However, it is unknown when detrimental sarcomeric changes first occur and whether they originate in the developing embryonic heart. Furthermore, Rho kinase (ROCK) is a serine/threonine protein kinase that is critical for regulating the function of several sarcomeric proteins, and therefore, our aim was to determine whether disruption of ROCK signaling during the earliest stages of heart development would disrupt the integrity of sarcomeres, altering heart development and function. Using a mouse model in which the function of ROCK is specifically disrupted in embryonic cardiomyocytes, we demonstrate a progressive cardiomyopathy that first appeared as sarcomeric disarray during cardiogenesis. This led to abnormalities in the structure of the embryonic ventricular wall and compensatory cardiomyocyte hypertrophy during fetal development. This sarcomeric disruption and hypertrophy persisted throughout adult life, triggering left ventricular concentric hypertrophy with systolic dysfunction, and reactivation of fetal gene expression and cardiac fibrosis, all typical features of HCM. Taken together, our findings establish a mechanism for the developmental origin of the sarcomeric phenotype of HCM and suggest that variants in the ROCK genes or disruption of ROCK signaling could, in part, contribute to its pathogenesis. Disruption of ROCK activity in embryonic cardiomyocytes revealed a developmental origin for hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Kate E Bailey
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Guy A MacGowan
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon Tual-Chalot
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lauren Phillips
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Deborah J Henderson
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen M Arthur
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Simon D Bamforth
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Helen M Phillips
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
17
|
Identification of Novel Therapeutic Targets for Pulmonary Arterial Hypertension. Int J Mol Sci 2018; 19:ijms19124081. [PMID: 30562953 PMCID: PMC6321293 DOI: 10.3390/ijms19124081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 12/29/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) are fatal diseases; however, their pathogenesis still remains to be elucidated. We have recently screened novel pathogenic molecules and have performed drug discovery targeting those molecules. Pulmonary artery smooth muscle cells (PASMCs) in patients with PAH (PAH-PASMCs) have high proliferative properties like cancer cells, which leads to thickening and narrowing of distal pulmonary arteries. Thus, we conducted a comprehensive analysis of PAH-PASMCs and lung tissues to search for novel pathogenic proteins. We validated the pathogenic role of the selected proteins by using tissue-specific knockout mice. To confirm its clinical significance, we used patient-derived blood samples to evaluate the potential as a biomarker for diagnosis and prognosis. Finally, we conducted a high throughput screening and found inhibitors for the pathogenic proteins.
Collapse
|
18
|
Abstract
Desmosomes are junctional protein complexes that confer strong adhesive capacity to adjacent host cells. In a recent study, we showed that enteropathogenic Escherichia coli (EPEC) disrupts desmosomes, weakens cell-cell adhesion and perturbs barrier function of intestinal epithelial (C2BBe) cells. Desmosomal damage was dependent on the EPEC effector protein EspH and its inhibitory effect on Rho GTPases. EspH-mediated Rho inactivation resulted in retraction of keratin intermediate filaments and degradation of desmosomal cadherins. Immunofluorescence studies of EPEC-infected C2BBe cells revealed keratin retraction towards the nucleus coincident with significant cytoplasmic redistribution of the desmosomal cadherin desmoglein-2 (DSG2). In this addendum, we expand on how EPEC-induced keratin retraction leads to loss of DSG2 anchoring at the junctions, and show that maturity of the epithelial cell monolayer impacts the fate of desmosomes during infection.
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA
| | - Gayatri Vedantam
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA,Department of Immunobiology, University of Arizona, Tucson, AZ, USA,BIO5 Institute for Collaborative Research, University of Arizona, Tucson, AZ, USA,Southern Arizona VA Healthcare System, Tucson, AZ, USA
| | - V.K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA,Department of Immunobiology, University of Arizona, Tucson, AZ, USA,BIO5 Institute for Collaborative Research, University of Arizona, Tucson, AZ, USA,CONTACT V.K. Viswanathan, Ph.D. School of Animal & Comparative Biomedical Sciences, University of Arizona, 1007 E. Lowell, Building 106, Rm. 231, Tucson, AZ 85721, USA
| |
Collapse
|
19
|
Different roles of myocardial ROCK1 and ROCK2 in cardiac dysfunction and postcapillary pulmonary hypertension in mice. Proc Natl Acad Sci U S A 2018; 115:E7129-E7138. [PMID: 29987023 DOI: 10.1073/pnas.1721298115] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although postcapillary pulmonary hypertension (PH) is an important prognostic factor for patients with heart failure (HF), its pathogenesis remains to be fully elucidated. To elucidate the different roles of Rho-kinase isoforms, ROCK1 and ROCK2, in cardiomyocytes in response to chronic pressure overload, we performed transverse aortic constriction (TAC) in cardiac-specific ROCK1-deficient (cROCK1-/-) and ROCK2-deficient (cROCK2-/-) mice. Cardiomyocyte-specific ROCK1 deficiency promoted pressure-overload-induced cardiac dysfunction and postcapillary PH, whereas cardiomyocyte-specific ROCK2 deficiency showed opposite results. Histological analysis showed that pressure-overload-induced cardiac hypertrophy and fibrosis were enhanced in cROCK1-/- mice compared with controls, whereas cardiac hypertrophy was attenuated in cROCK2-/- mice after TAC. Consistently, the levels of oxidative stress were up-regulated in cROCK1-/- hearts and down-regulated in cROCK2-/- hearts compared with controls after TAC. Furthermore, cyclophilin A (CyPA) and basigin (Bsg), both of which augment oxidative stress, enhanced cardiac dysfunction and postcapillary PH in cROCK1-/- mice, whereas their expressions were significantly lower in cROCK2-/- mice. In clinical studies, plasma levels of CyPA were significantly increased in HF patients and were higher in patients with postcapillary PH compared with those without it. Finally, high-throughput screening demonstrated that celastrol, an antioxidant and antiinflammatory agent, reduced the expressions of CyPA and Bsg in the heart and the lung, ameliorating cardiac dysfunction and postcapillary PH induced by TAC. Thus, by differentially affecting CyPA and Bsg expressions, ROCK1 protects and ROCK2 jeopardizes the heart from pressure-overload HF with postcapillary PH, for which celastrol may be a promising agent.
Collapse
|
20
|
Dorn T, Kornherr J, Parrotta EI, Zawada D, Ayetey H, Santamaria G, Iop L, Mastantuono E, Sinnecker D, Goedel A, Dirschinger RJ, My I, Laue S, Bozoglu T, Baarlink C, Ziegler T, Graf E, Hinkel R, Cuda G, Kääb S, Grace AA, Grosse R, Kupatt C, Meitinger T, Smith AG, Laugwitz KL, Moretti A. Interplay of cell-cell contacts and RhoA/MRTF-A signaling regulates cardiomyocyte identity. EMBO J 2018; 37:e98133. [PMID: 29764980 PMCID: PMC6003642 DOI: 10.15252/embj.201798133] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 03/29/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Cell-cell and cell-matrix interactions guide organ development and homeostasis by controlling lineage specification and maintenance, but the underlying molecular principles are largely unknown. Here, we show that in human developing cardiomyocytes cell-cell contacts at the intercalated disk connect to remodeling of the actin cytoskeleton by regulating the RhoA-ROCK signaling to maintain an active MRTF/SRF transcriptional program essential for cardiomyocyte identity. Genetic perturbation of this mechanosensory pathway activates an ectopic fat gene program during cardiomyocyte differentiation, which ultimately primes the cells to switch to the brown/beige adipocyte lineage in response to adipogenesis-inducing signals. We also demonstrate by in vivo fate mapping and clonal analysis of cardiac progenitors that cardiac fat and a subset of cardiac muscle arise from a common precursor expressing Isl1 and Wt1 during heart development, suggesting related mechanisms of determination between the two lineages.
Collapse
Affiliation(s)
- Tatjana Dorn
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Jessica Kornherr
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elvira I Parrotta
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Dorota Zawada
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Harold Ayetey
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Laura Iop
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elisa Mastantuono
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Daniel Sinnecker
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Alexander Goedel
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Ralf J Dirschinger
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Ilaria My
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Svenja Laue
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Tarik Bozoglu
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | | | - Tilman Ziegler
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Rabea Hinkel
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
- IPEK Institute for Cardiovascular Prevention, Klinikum der Universität München - Ludwig-Maximillians-Universität, Munich, Germany
| | - Giovanni Cuda
- Department of Experimental and Clinical Medicine, Medical School, University of Magna Grecia, Catanzaro, Italy
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München - Ludwig-Maximillians-Universität, Munich, Germany
| | - Andrew A Grace
- Papworth Hospital NHS Foundation Trust, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Robert Grosse
- Pharmacology Institute, Philipps University Marburg, Marburg, Germany
| | - Christian Kupatt
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
| | - Austin G Smith
- Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Karl-Ludwig Laugwitz
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| | - Alessandra Moretti
- Klinik und Poliklinik Innere Medizin I, Klinikum rechts der Isar - Technical University of Munich, Munich, Germany
- DZHK (German Centre for Cardiovascular Research) - partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
21
|
Hoorntje ET, Te Rijdt WP, James CA, Pilichou K, Basso C, Judge DP, Bezzina CR, van Tintelen JP. Arrhythmogenic cardiomyopathy: pathology, genetics, and concepts in pathogenesis. Cardiovasc Res 2018; 113:1521-1531. [PMID: 28957532 DOI: 10.1093/cvr/cvx150] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 08/03/2017] [Indexed: 02/06/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is a rare, heritable heart disease characterized by fibro-fatty replacement of the myocardium and a high degree of electric instability. It was first thought to be a congenital disorder, but is now regarded as a dystrophic heart muscle disease that develops over time. There is no curative treatment and current treatment strategies focus on attenuating the symptoms, slowing disease progression, and preventing life-threatening arrhythmias and sudden cardiac death. Identification of mutations in genes encoding desmosomal proteins and in other genes has led to insights into the disease pathogenesis and greatly facilitated identification of family members at risk. The disease phenotype is, however, highly variable and characterized by incomplete penetrance. Although the reasons are still poorly understood, sex, endurance exercise and a gene-dosage effect seem to play a role in these phenomena. The discovery of the genes and mutations implicated in ACM has allowed animal and cellular models to be generated, enabling researchers to start unravelling it's underlying molecular mechanisms. Observations in humans and in animal models suggest that reduced cell-cell adhesion affects gap junction and ion channel remodelling at the intercalated disc, and along with impaired desmosomal function, these can lead to perturbations in signalling cascades like the Wnt/β-catenin and Hippo/YAP pathways. Perturbations of these pathways are also thought to lead to fibro-fatty replacement. A better understanding of the molecular processes may lead to new therapies that target specific pathways involved in ACM.
Collapse
Affiliation(s)
- Edgar T Hoorntje
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.,Netherlands Heart Institute, Moreelsepark 1, 3511 EP, Utrecht, The Netherlands
| | - Wouter P Te Rijdt
- Department of Genetics, University of Groningen, University Medical Centre Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Cynthia A James
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD, USA
| | - Kalliopi Pilichou
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua 35121, Italy
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Padua 35121, Italy
| | - Daniel P Judge
- Department of Medicine, Division of Cardiology, Johns Hopkins University School of Medicine, 1800 Orleans Street, Baltimore, MD, USA
| | - Connie R Bezzina
- Department of Clinical and Experimental Cardiology, Heart Centre, Academic Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - J Peter van Tintelen
- Netherlands Heart Institute, Moreelsepark 1, 3511 EP, Utrecht, The Netherlands.,Department of Clinical Genetics, Academic Medical Centre Amsterdam, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
22
|
Roxas JL, Monasky RC, Roxas BAP, Agellon AB, Mansoor A, Kaper JB, Vedantam G, Viswanathan V. Enteropathogenic Escherichia coli EspH-Mediated Rho GTPase Inhibition Results in Desmosomal Perturbations. Cell Mol Gastroenterol Hepatol 2018; 6:163-180. [PMID: 30003123 PMCID: PMC6039986 DOI: 10.1016/j.jcmgh.2018.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 04/20/2018] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS The diarrheagenic pathogen, enteropathogenic Escherichia coli (EPEC), uses a type III secretion system to deliver effector molecules into intestinal epithelial cells (IECs). While exploring the basis for the lateral membrane separation of EPEC-infected IECs, we observed infection-induced loss of the desmosomal cadherin desmoglein-2 (DSG2). We sought to identify the molecule(s) involved in, and delineate the mechanisms and consequences of, EPEC-induced DSG2 loss. METHODS DSG2 abundance and localization was monitored via immunoblotting and immunofluorescence, respectively. Junctional perturbations were visualized by electron microscopy, and cell-cell adhesion was assessed using dispase assays. EspH alanine-scan mutants as well as pharmacologic agents were used to evaluate impacts on desmosomal alterations. EPEC-mediated DSG2 loss, and its impact on bacterial colonization in vivo, was assessed using a murine model. RESULTS The secreted virulence protein EspH mediates EPEC-induced DSG2 degradation, and contributes to desmosomal perturbation, loss of cell junction integrity, and barrier disruption in infected IECs. EspH sequesters Rho guanine nucleotide exchange factors and inhibits Rho guanosine triphosphatase signaling; EspH mutants impaired for Rho guanine nucleotide exchange factor interaction failed to inhibit RhoA or deplete DSG2. Cytotoxic necrotizing factor 1, which locks Rho guanosine triphosphatase in the active state, jasplakinolide, a molecule that promotes actin polymerization, and the lysosomal inhibitor bafilomycin A, respectively, rescued infected cells from EPEC-induced DSG2 loss. Wild-type EPEC, but not an espH-deficient strain, colonizes mouse intestines robustly, widens paracellular junctions, and induces DSG2 re-localization in vivo. CONCLUSIONS Our studies define the mechanism and consequences of EPEC-induced desmosomal alterations in IECs. These perturbations contribute to the colonization and virulence of EPEC, and likely related pathogens.
Collapse
Key Words
- A/E, attaching and effacing
- BSA, bovine serum albumin
- CM, calcium and magnesium
- DMEM, Dulbecco's modified Eagle medium
- DSC, desmocollin
- DSG, desmoglein
- DSG2
- Desmoglein
- EPEC
- EPEC, enteropathogenic Escherichia coli
- GEF, guanine nucleotide exchange factors
- GTPase, guanosine triphosphatase
- Host–Pathogen Interaction
- IEC, intestinal epithelial cell
- IF, intermediate filament
- PBS, phosphate-buffered saline
- T3SS, type 3 secretion system
- TER, transepithelial electrical resistance
- TJ, tight junction
- WT, wild-type
Collapse
Affiliation(s)
- Jennifer Lising Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Ross Calvin Monasky
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Bryan Angelo P. Roxas
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - Al B. Agellon
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
| | - Asad Mansoor
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
| | - James B. Kaper
- University of Maryland School of Medicine, Baltimore, Maryland
| | - Gayatri Vedantam
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- Southern Arizona VA Healthcare System, Tucson, Arizona
| | - V.K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, Arizona
- BIO5 Institute for Collaborative Research, University of Arizona, Tucson, Arizona
- Department of Immunobiology, University of Arizona, Tucson, Arizona
- Correspondence Address correspondence to: V. K. Viswanathan, PhD, School of Animal and Comparative Biomedical Sciences, 1006 E. Lowell, Building 106, Room 231, University of Arizona, Tucson, Arizona 85721. fax: (520) 621-6366.
| |
Collapse
|
23
|
Abstract
Rho kinases (ROCKs) are the first discovered RhoA effectors that are now widely known for their effects on actin organization. Recent studies have shown that ROCKs play important roles in cardiac physiology. Abnormal activation of ROCKs participate in multiple cardiovascular pathological processes, including cardiac hypertrophy, apoptosis, fibrosis, systemic hypertension, and pulmonary hypertension. ROCK inhibitors, fasudil and statins, have shown beneficial cardiovascular effects in many animal studies, clinical trials, and applications. Here, we mainly discuss the current understanding of the physiological roles of Rho kinase signaling in the heart, and briefly summarize the roles of ROCKs in cardiac-related vascular dysfunctions. We will also discuss the clinical application of ROCK inhibitors.
Collapse
Affiliation(s)
- Yuan Dai
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Weijia Luo
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| | - Jiang Chang
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, USA
| |
Collapse
|
24
|
Abstract
Cyclophilin A (CyPA) is secreted from vascular smooth muscle cells, inflammatory cells, activated platelets, and cardiac fibroblasts in response to oxidative stress. Excessive and continuous activation of the RhoA/Rho-kinase system promotes the secretion of CyPA, resulting in the development of multiple cardiovascular diseases. Basigin (Bsg), a transmembrane glycoprotein that activates matrix metalloproteinases, is an extracellular receptor for CyPA that promotes cell proliferation and inflammation. Thus, the CyPA/Bsg system is potentially a novel therapeutic target for cardiovascular diseases. Importantly, plasma CyPA levels are increased in patients with coronary artery disease, abdominal aortic aneurysms, pulmonary hypertension, and heart failure. Moreover, plasma CyPA levels can predict all-cause death in patients with coronary artery disease and pulmonary hypertension. Additionally, plasma soluble Bsg levels are increased and predict all-cause death in patients with heart failure, suggesting that CyPA and Bsg are novel biomarkers for cardiovascular diseases. To discover further novel molecules targeting the CyPA/Bsg system, high-throughput screening of compounds found molecules that ameliorate the development of cardiovascular diseases. In addition to CyPA and Bsg, novel therapeutic targets and their inhibitors for patients with pulmonary arterial hypertension have been recently screened and identified. Ultimately, the final goal is to develop novel biomarkers and medications that will be useful for improving the prognosis and quality of life in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Kimio Satoh
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine
| |
Collapse
|
25
|
Marian AJ, van Rooij E, Roberts R. Genetics and Genomics of Single-Gene Cardiovascular Diseases: Common Hereditary Cardiomyopathies as Prototypes of Single-Gene Disorders. J Am Coll Cardiol 2017; 68:2831-2849. [PMID: 28007145 DOI: 10.1016/j.jacc.2016.09.968] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/14/2016] [Accepted: 09/19/2016] [Indexed: 01/05/2023]
Abstract
This is the first of 2 review papers on genetics and genomics appearing as part of the series on "omics." Genomics pertains to all components of an organism's genes, whereas genetics involves analysis of a specific gene or genes in the context of heredity. The paper provides introductory comments, describes the basis of human genetic diversity, and addresses the phenotypic consequences of genetic variants. Rare variants with large effect sizes are responsible for single-gene disorders, whereas complex polygenic diseases are typically due to multiple genetic variants, each exerting a modest effect size. To illustrate the clinical implications of genetic variants with large effect sizes, 3 common forms of hereditary cardiomyopathies are discussed as prototypic examples of single-gene disorders, including their genetics, clinical manifestations, pathogenesis, and treatment. The genetic basis of complex traits is discussed in a separate paper.
Collapse
Affiliation(s)
- Ali J Marian
- Center for Cardiovascular Genetics, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, and Texas Heart Institute, Houston, Texas.
| | - Eva van Rooij
- Hubrecht Institute, KNAW and University Medical Center Utrecht, Utrecht, the Netherlands; Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Robert Roberts
- University of Arizona College of Medicine, Phoenix, Arizona
| |
Collapse
|
26
|
TRPM8 downregulation by angiotensin II in vascular smooth muscle cells is involved in hypertension. Mol Med Rep 2017; 15:1900-1908. [PMID: 28138709 DOI: 10.3892/mmr.2017.6158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 12/08/2016] [Indexed: 11/05/2022] Open
Abstract
Angiotensin II (Ang II)-induced injury of vascular smooth muscle cells (VSMCs) serves an important role in hypertension and other cardiovascular disorders. Transient receptor potential melastatin 8 (TRPM8) is a thermally‑regulated Ca2+‑permeable channel that is activated by reduced body temperature. Although several recent studies have revealed the regulatory effect of TRPM8 in vascular tone and hypertension, the precise role of TRPM8 in dysfunction of vascular smooth muscle cells (VSMCs) induced by Ang II remains elusive. In the present study, the possible function of TRPM8 in Ang II‑induced VSMCs malfunction in vivo and in vitro was investigated. In the aortae from rats that had undergone a two‑kidney one‑clip operation, which is a widely‑used renovascular hypertension model, the mRNA and protein levels of TRPM8 were reduced. In addition, exogenous Ang II treatment decreased TRPM8 mRNA and protein expression levels in primary cultures of rat VSMCs. TRPM8 activation by menthol, a pharmacological agonist, in VSMCs, significantly attenuated the Ang II‑induced increase in reactive oxygen species and H2O2 production. In addition, TRPM8 activation reduced the Ang II‑induced upregulation of NADPH oxidase (NOX) 1 and NOX4 in VSMCs. Furthermore, TRPM8 activation relieved the Ang II‑induced activation of ras homolog gene family, member A‑rho associated protein kinase 2 and janus kinase 2 signaling pathways in VSMCs. In conclusion, the results presented in the current study indicated that TRPM8 downregulation by Ang II in VSMCs may be involved in hypertension.
Collapse
|
27
|
Abstract
PURPOSE OF REVIEW The study of cardiac development is critical to inform management strategies for congenital and acquired heart disease. This review serves to highlight some of the advances in this field over the past year. RECENT FINDINGS Three main areas of study are included that have been particularly innovative and progressive. These include more precise gene targeting in animal models of disease and in moving from animal models to human disease, more precise in-vitro models including three-dimensional structuring and inclusion of hemodynamic components, and expanding the concepts of genetic regulation of heart development and disease. SUMMARY Targeted genetics in animal models are able to make use of tissue and time-specific promotors that drive gene expression or knockout with high specificity. In-vitro models can recreate flow patterns in blood vessels and across cardiac valves. Noncoding RNAs, once thought to be of no consequence to gene transcription and translation, prove to be key regulators of genetic function in health and disease.
Collapse
|
28
|
Choy L, Yeo JM, Tse V, Chan SP, Tse G. Cardiac disease and arrhythmogenesis: Mechanistic insights from mouse models. IJC HEART & VASCULATURE 2016; 12:1-10. [PMID: 27766308 PMCID: PMC5064289 DOI: 10.1016/j.ijcha.2016.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 05/02/2016] [Indexed: 12/19/2022]
Abstract
The mouse is the second mammalian species, after the human, in which substantial amount of the genomic information has been analyzed. With advances in transgenic technology, mutagenesis is now much easier to carry out in mice. Consequently, an increasing number of transgenic mouse systems have been generated for the study of cardiac arrhythmias in ion channelopathies and cardiomyopathies. Mouse hearts are also amenable to physical manipulation such as coronary artery ligation and transverse aortic constriction to induce heart failure, radiofrequency ablation of the AV node to model complete AV block and even implantation of a miniature pacemaker to induce cardiac dyssynchrony. Last but not least, pharmacological models, despite being simplistic, have enabled us to understand the physiological mechanisms of arrhythmias and evaluate the anti-arrhythmic properties of experimental agents, such as gap junction modulators, that may be exert therapeutic effects in other cardiac diseases. In this article, we examine these in turn, demonstrating that primary inherited arrhythmic syndromes are now recognized to be more complex than abnormality in a particular ion channel, involving alterations in gene expression and structural remodelling. Conversely, in cardiomyopathies and heart failure, mutations in ion channels and proteins have been identified as underlying causes, and electrophysiological remodelling are recognized pathological features. Transgenic techniques causing mutagenesis in mice are extremely powerful in dissecting the relative contributions of different genes play in producing disease phenotypes. Mouse models can serve as useful systems in which to explore how protein defects contribute to arrhythmias and direct future therapy.
Collapse
Affiliation(s)
- Lois Choy
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Jie Ming Yeo
- School of Medicine, Imperial College London, SW7 2AZ, UK
| | - Vivian Tse
- Department of Physiology, McGill University, Canada
| | - Shing Po Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| | - Gary Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong
| |
Collapse
|
29
|
Abstract
Twenty years ago, Rho-kinase was identified as an important downstream effector of the small GTP-binding protein, RhoA. Thereafter, a series of studies demonstrated the important roles of Rho-kinase in the cardiovascular system. The RhoA/Rho-kinase pathway is now widely known to play important roles in many cellular functions, including contraction, motility, proliferation, and apoptosis, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Furthermore, the important role of Rho-kinase has been demonstrated in the pathogenesis of vasospasm, arteriosclerosis, ischemia/reperfusion injury, hypertension, pulmonary hypertension, and heart failure. Cyclophilin A is secreted by vascular smooth muscle cells and inflammatory cells and activated platelets in a Rho-kinase-dependent manner, playing important roles in a wide range of cardiovascular diseases. Thus, the RhoA/Rho-kinase pathway plays crucial roles under both physiological and pathological conditions and is an important therapeutic target in cardiovascular medicine. Recently, functional differences between ROCK1 and ROCK2 have been reported in vitro. ROCK1 is specifically cleaved by caspase-3, whereas granzyme B cleaves ROCK2. However, limited information is available on the functional differences and interactions between ROCK1 and ROCK2 in the cardiovascular system in vivo. Herein, we will review the recent advances about the importance of RhoA/Rho-kinase in the cardiovascular system.
Collapse
Affiliation(s)
- Hiroaki Shimokawa
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Shinichiro Sunamura
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kimio Satoh
- From the Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
30
|
Abstract
Hypertensive cardiac remodeling is characterized by left ventricular hypertrophy and interstitial fibrosis, which can lead to heart failure with preserved ejection fraction. The Rho-associated coiled-coil containing kinases (ROCKs) are members of the serine/threonine protein kinase family, which mediates the downstream effects of the small GTP-binding protein RhoA. There are 2 isoforms: ROCK1 and ROCK2. They have different functions in different types of cells and tissues. There is growing evidence that ROCKs contribute to the development of cardiovascular diseases, including cardiac fibrosis, hypertrophy, and subsequent heart failure. Recent experimental studies using ROCK inhibitors, such as fasudil, have shown the benefits of ROCK inhibition in cardiac remodeling. Mice lacking each ROCK isoform also exhibit reduced myocardial fibrosis in a variety of pathological models of cardiac remodeling. Indeed, clinical studies with fasudil have suggested that ROCKs could be potential novel therapeutic targets for cardiovascular diseases. In this review, we summarize the current understanding of the roles of ROCKs in the development of cardiac fibrosis and hypertrophy and discuss their therapeutic potential for deleterious cardiac remodeling. (Circ J 2016; 80: 1491-1498).
Collapse
Affiliation(s)
- Toru Shimizu
- Section of Cardiology, Department of Medicine, University of Chicago
| | | |
Collapse
|