1
|
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
|
2
|
Essandoh K, Teuber JP, Brody MJ. Regulation of cardiomyocyte intracellular trafficking and signal transduction by protein palmitoylation. Biochem Soc Trans 2024; 52:41-53. [PMID: 38385554 PMCID: PMC10903464 DOI: 10.1042/bst20221296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/23/2024]
Abstract
Despite the well-established functions of protein palmitoylation in fundamental cellular processes, the roles of this reversible post-translational lipid modification in cardiomyocyte biology remain poorly studied. Palmitoylation is catalyzed by a family of 23 zinc finger and Asp-His-His-Cys domain-containing S-acyltransferases (zDHHC enzymes) and removed by select thioesterases of the lysophospholipase and α/β-hydroxylase domain (ABHD)-containing families of serine hydrolases. Recently, studies utilizing genetic manipulation of zDHHC enzymes in cardiomyocytes have begun to unveil essential functions for these enzymes in regulating cardiac development, homeostasis, and pathogenesis. Palmitoylation co-ordinates cardiac electrophysiology through direct modulation of ion channels and transporters to impact their trafficking or gating properties as well as indirectly through modification of regulators of channels, transporters, and calcium handling machinery. Not surprisingly, palmitoylation has roles in orchestrating the intracellular trafficking of proteins in cardiomyocytes, but also dynamically fine-tunes cardiomyocyte exocytosis and natriuretic peptide secretion. Palmitoylation has emerged as a potent regulator of intracellular signaling in cardiomyocytes, with recent studies uncovering palmitoylation-dependent regulation of small GTPases through direct modification and sarcolemmal targeting of the small GTPases themselves or by modification of regulators of the GTPase cycle. In addition to dynamic control of G protein signaling, cytosolic DNA is sensed and transduced into an inflammatory transcriptional output through palmitoylation-dependent activation of the cGAS-STING pathway, which has been targeted pharmacologically in preclinical models of heart disease. Further research is needed to fully understand the complex regulatory mechanisms governed by protein palmitoylation in cardiomyocytes and potential emerging therapeutic targets.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - James P. Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
| | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, U.S.A
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, U.S.A
| |
Collapse
|
3
|
Baldwin TA, Teuber JP, Kuwabara Y, Subramani A, Lin SCJ, Kanisicak O, Vagnozzi RJ, Zhang W, Brody MJ, Molkentin JD. Palmitoylation-dependent regulation of cardiomyocyte Rac1 signaling activity and minor effects on cardiac hypertrophy. J Biol Chem 2023; 299:105426. [PMID: 37926281 PMCID: PMC10716590 DOI: 10.1016/j.jbc.2023.105426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
S-palmitoylation is a reversible lipid modification catalyzed by 23 S-acyltransferases with a conserved zinc finger aspartate-histidine-histidine-cysteine (zDHHC) domain that facilitates targeting of proteins to specific intracellular membranes. Here we performed a gain-of-function screen in the mouse and identified the Golgi-localized enzymes zDHHC3 and zDHHC7 as regulators of cardiac hypertrophy. Cardiomyocyte-specific transgenic mice overexpressing zDHHC3 show cardiac disease, and S-acyl proteomics identified the small GTPase Rac1 as a novel substrate of zDHHC3. Notably, cardiomyopathy and congestive heart failure in zDHHC3 transgenic mice is preceded by enhanced Rac1 S-palmitoylation, membrane localization, activity, downstream hypertrophic signaling, and concomitant induction of all Rho family small GTPases whereas mice overexpressing an enzymatically dead zDHHC3 mutant show no discernible effect. However, loss of Rac1 or other identified zDHHC3 targets Gαq/11 or galectin-1 does not diminish zDHHC3-induced cardiomyopathy, suggesting multiple effectors and pathways promoting decompensation with sustained zDHHC3 activity. Genetic deletion of Zdhhc3 in combination with Zdhhc7 reduces cardiac hypertrophy during the early response to pressure overload stimulation but not over longer time periods. Indeed, cardiac hypertrophy in response to 2 weeks of angiotensin-II infusion is not diminished by Zdhhc3/7 deletion, again suggesting other S-acyltransferases or signaling mechanisms compensate to promote hypertrophic signaling. Taken together, these data indicate that the activity of zDHHC3 and zDHHC7 at the cardiomyocyte Golgi promote Rac1 signaling and maladaptive cardiac remodeling, but redundant signaling effectors compensate to maintain cardiac hypertrophy with sustained pathological stimulation in the absence of zDHHC3/7.
Collapse
Affiliation(s)
- Tanya A Baldwin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - James P Teuber
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yasuhide Kuwabara
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Araskumar Subramani
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
| | - Suh-Chin J Lin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Onur Kanisicak
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pathology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ronald J Vagnozzi
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Division of Cardiology, Department of Medicine, Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Weiqi Zhang
- Laboratory of Molecular Psychiatry, Department of Mental Health, University of Münster, Münster, Germany
| | - Matthew J Brody
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA; Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA; Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
4
|
Perike S, Gonzalez-Gonzalez FJ, Abu-Taha I, Damen FW, Hanft LM, Lizama KS, Aboonabi A, Capote AE, Aguilar-Sanchez Y, Levin B, Han Z, Sridhar A, Grand J, Martin J, Akar JG, Warren CM, Solaro RJ, Sang-Ging O, Darbar D, McDonald KS, Goergen CJ, Wolska BM, Dobrev D, Wehrens XH, McCauley MD. PPP1R12C Promotes Atrial Hypocontractility in Atrial Fibrillation. Circ Res 2023; 133:758-771. [PMID: 37737016 PMCID: PMC10616980 DOI: 10.1161/circresaha.123.322516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 09/14/2023] [Indexed: 09/23/2023]
Abstract
BACKGROUND Atrial fibrillation (AF)-the most common sustained cardiac arrhythmia-increases thromboembolic stroke risk 5-fold. Although atrial hypocontractility contributes to stroke risk in AF, the molecular mechanisms reducing myofilament contractile function remain unknown. We tested the hypothesis that increased expression of PPP1R12C (protein phosphatase 1 regulatory subunit 12C)-the PP1 (protein phosphatase 1) regulatory subunit targeting MLC2a (atrial myosin light chain 2)-causes hypophosphorylation of MLC2a and results in atrial hypocontractility. METHODS Right atrial appendage tissues were isolated from human patients with AF versus sinus rhythm controls. Western blots, coimmunoprecipitation, and phosphorylation studies were performed to examine how the PP1c (PP1 catalytic subunit)-PPP1R12C interaction causes MLC2a dephosphorylation. In vitro studies of pharmacological MRCK (myotonic dystrophy kinase-related Cdc42-binding kinase) inhibitor (BDP5290) in atrial HL-1 cells were performed to evaluate PP1 holoenzyme activity on MLC2a. Cardiac-specific lentiviral PPP1R12C overexpression was performed in mice to evaluate atrial remodeling with atrial cell shortening assays, echocardiography, and AF inducibility with electrophysiology studies. RESULTS In human patients with AF, PPP1R12C expression was increased 2-fold versus sinus rhythm controls (P=2.0×10-2; n=12 and 12 in each group) with >40% reduction in MLC2a phosphorylation (P=1.4×10-6; n=12 and 12 in each group). PPP1R12C-PP1c binding and PPP1R12C-MLC2a binding were significantly increased in AF (P=2.9×10-2 and 6.7×10-3, respectively; n=8 and 8 in each group). In vitro studies utilizing drug BDP5290, which inhibits T560-PPP1R12C phosphorylation, demonstrated increased PPP1R12C binding with both PP1c and MLC2a and dephosphorylation of MLC2a. Mice treated with lentiviral PPP1R12C vector demonstrated a 150% increase in left atrial size versus controls (P=5.0×10-6; n=12, 8, and 12), with reduced atrial strain and atrial ejection fraction. Pacing-induced AF in mice treated with lentiviral PPP1R12C vector was significantly higher than in controls (P=1.8×10-2 and 4.1×10-2, respectively; n=6, 6, and 5). CONCLUSIONS Patients with AF exhibit increased levels of PPP1R12C protein compared with controls. PPP1R12C overexpression in mice increases PP1c targeting to MLC2a and causes MLC2a dephosphorylation, which reduces atrial contractility and increases AF inducibility. These findings suggest that PP1 regulation of sarcomere function at MLC2a is a key determinant of atrial contractility in AF.
Collapse
Affiliation(s)
- Srikanth Perike
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
- Jesse Brown VA Medical Center, Chicago, IL
| | - Francisco J. Gonzalez-Gonzalez
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
- Jesse Brown VA Medical Center, Chicago, IL
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
| | - Frederick W. Damen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Laurin M. Hanft
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia
| | - Ken S. Lizama
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
- Jesse Brown VA Medical Center, Chicago, IL
| | - Anahita Aboonabi
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
| | - Andrielle E. Capote
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
| | - Yuriana Aguilar-Sanchez
- Department of Integrative Physiology and The Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| | | | - Zhenbo Han
- Department of Pharmacology and Regenerative Medicine, College of Medicine,University of Illinois at Chicago
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
| | - Jacob Grand
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
| | | | | | - Chad M. Warren
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
| | - R. John Solaro
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
| | - Ong Sang-Ging
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Pharmacology and Regenerative Medicine, College of Medicine,University of Illinois at Chicago
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
- Jesse Brown VA Medical Center, Chicago, IL
| | - Kerry S. McDonald
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia
| | - Craig J. Goergen
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN
| | - Beata M. Wolska
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Germany
- Department of Integrative Physiology and The Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
- Department of Medicine, Montréal Heart Institute and Université de Montréal, Montréal, Canada
| | - Xander H.T. Wehrens
- Department of Integrative Physiology and The Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX
| | - Mark D. McCauley
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, University of Illinois at Chicago
- Jesse Brown VA Medical Center, Chicago, IL
| |
Collapse
|
5
|
Dai Y, Ignatyeva N, Xu H, Wali R, Toischer K, Brandenburg S, Lenz C, Pronto J, Fakuade FE, Sossalla S, Zeisberg EM, Janshoff A, Kutschka I, Voigt N, Urlaub H, Rasmussen TB, Mogensen J, Lehnart SE, Hasenfuss G, Ebert A. An Alternative Mechanism of Subcellular Iron Uptake Deficiency in Cardiomyocytes. Circ Res 2023; 133:e19-e46. [PMID: 37313752 DOI: 10.1161/circresaha.122.321157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 05/26/2023] [Indexed: 06/15/2023]
Abstract
BACKGROUND Systemic defects in intestinal iron absorption, circulation, and retention cause iron deficiency in 50% of patients with heart failure. Defective subcellular iron uptake mechanisms that are independent of systemic absorption are incompletely understood. The main intracellular route for iron uptake in cardiomyocytes is clathrin-mediated endocytosis. METHODS We investigated subcellular iron uptake mechanisms in patient-derived and CRISPR/Cas-edited induced pluripotent stem cell-derived cardiomyocytes as well as patient-derived heart tissue. We used an integrated platform of DIA-MA (mass spectrometry data-independent acquisition)-based proteomics and signaling pathway interrogation. We employed a genetic induced pluripotent stem cell model of 2 inherited mutations (TnT [troponin T]-R141W and TPM1 [tropomyosin 1]-L185F) that lead to dilated cardiomyopathy (DCM), a frequent cause of heart failure, to study the underlying molecular dysfunctions of DCM mutations. RESULTS We identified a druggable molecular pathomechanism of impaired subcellular iron deficiency that is independent of systemic iron metabolism. Clathrin-mediated endocytosis defects as well as impaired endosome distribution and cargo transfer were identified as a basis for subcellular iron deficiency in DCM-induced pluripotent stem cell-derived cardiomyocytes. The clathrin-mediated endocytosis defects were also confirmed in the hearts of patients with DCM with end-stage heart failure. Correction of the TPM1-L185F mutation in DCM patient-derived induced pluripotent stem cells, treatment with a peptide, Rho activator II, or iron supplementation rescued the molecular disease pathway and recovered contractility. Phenocopying the effects of the TPM1-L185F mutation into WT induced pluripotent stem cell-derived cardiomyocytes could be ameliorated by iron supplementation. CONCLUSIONS Our findings suggest that impaired endocytosis and cargo transport resulting in subcellular iron deficiency could be a relevant pathomechanism for patients with DCM carrying inherited mutations. Insight into this molecular mechanism may contribute to the development of treatment strategies and risk management in heart failure.
Collapse
Affiliation(s)
- Yuanyuan Dai
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Nadezda Ignatyeva
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Hang Xu
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Ruheen Wali
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Karl Toischer
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Sören Brandenburg
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Christof Lenz
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Department of Clinical Chemistry, University Medical Center Goettingen, (C.L., H.U.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen (C.L., H.U.)
| | - Julius Pronto
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
| | - Funsho E Fakuade
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Samuel Sossalla
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
- Department for Internal Medicine II, University Medical Center Regensburg (S.S.)
| | - Elisabeth M Zeisberg
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| | - Andreas Janshoff
- Institute for Physical Chemistry (A.J.), University of Goettingen, Germany
| | - Ingo Kutschka
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Department of Thoracic and Cardiovascular Surgery, University Medical Center Göttingen (I.K.)
| | - Niels Voigt
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, (J.P., F.E.F., N.V.), University of Goettingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Henning Urlaub
- Department of Clinical Chemistry, University Medical Center Goettingen, (C.L., H.U.), University of Goettingen, Germany
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen (C.L., H.U.)
| | | | - Jens Mogensen
- Department of Cardiology, Aalborg University Hospital, Denmark (J.M.)
| | - Stephan E Lehnart
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC; C.L., F.E.F., N.V., S.E.L.), University of Goettingen, Germany
| | - Gerd Hasenfuss
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
- Heart Center, Clinic for Cardiology and Pneumology, University Medical Center Goettingen (K.T., S.B., S.S., G.H.), University of Goettingen, Germany
| | - Antje Ebert
- Heart Research Center Goettingen, Clinic for Cardiology and Pneumology, University Medical Center Goettingen, Georg-August University of Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., S.S., E.M.Z., S.E.L., G.H., A.E.)
- DZHK (German Center for Cardiovascular Research), partner site Goettingen, Germany (Y.D., N.I., H.X., R.W., K.T., S.B., C.L., J.P., F.E.F., E.M.Z., I.K., N.V., S.E.L., G.H., A.E.)
| |
Collapse
|
6
|
Keefe JA, Hulsurkar MM, Reilly S, Wehrens XHT. Mouse models of spontaneous atrial fibrillation. Mamm Genome 2023; 34:298-311. [PMID: 36173465 PMCID: PMC10898345 DOI: 10.1007/s00335-022-09964-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022]
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in adults, with a prevalence increasing with age. Current clinical management of AF is focused on tertiary prevention (i.e., treating the symptoms and sequelae) rather than addressing the underlying molecular pathophysiology. Robust animal models of AF, particularly those that do not require supraphysiologic stimuli to induce AF (i.e., showing spontaneous AF), enable studies that can uncover the underlying mechanisms of AF. Several mouse models of AF have been described to exhibit spontaneous AF, but pathophysiologic drivers of AF differ among models. Here, we describe relevant AF mechanisms and provide an overview of large and small animal models of AF. We then provide an in-depth review of the spontaneous mouse models of AF, highlighting the relevant AF mechanisms for each model.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Mohit M Hulsurkar
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, One Baylor Plaza, BCM335, Houston, TX, 77030, USA.
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Perike S, Gonzalez-Gonzalez FJ, Abu-Taha I, Damen FW, Lizama KS, Aboonabi A, Capote AE, Aguilar-Sanchez Y, Levin B, Han Z, Sridhar A, Grand J, Martin J, Akar JG, Warren CM, Solaro RJ, Ong SG, Darbar D, Goergen CJ, Wolska BM, Dobrev D, Wehrens XHT, McCauley MD. Myosin Light Chain Dephosphorylation by PPP1R12C Promotes Atrial Hypocontractility in Atrial Fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537590. [PMID: 37131731 PMCID: PMC10153354 DOI: 10.1101/2023.04.19.537590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background Atrial fibrillation (AF), the most common sustained cardiac arrhythmia, increases thromboembolic stroke risk five-fold. Although atrial hypocontractility contributes to stroke risk in AF, the molecular mechanisms reducing myofilament contractile function remain unknown. We tested the hypothesis that increased expression of PPP1R12C, the PP1 regulatory subunit targeting atrial myosin light chain 2 (MLC2a), causes hypophosphorylation of MLC2a and results in atrial hypocontractility. Methods Right atrial appendage tissues were isolated from human AF patients versus sinus rhythm (SR) controls. Western blots, co-immunoprecipitation, and phosphorylation studies were performed to examine how the PP1c-PPP1R12C interaction causes MLC2a de-phosphorylation. In vitro studies of pharmacologic MRCK inhibitor (BDP5290) in atrial HL-1 cells were performed to evaluate PP1 holoenzyme activity on MLC2a. Cardiac-specific lentiviral PPP1R12C overexpression was performed in mice to evaluate atrial remodeling with atrial cell shortening assays, echocardiography, and AF inducibility with EP studies. Results In human patients with AF, PPP1R12C expression was increased two-fold versus SR controls ( P =2.0×10 -2 , n=12,12 in each group) with > 40% reduction in MLC2a phosphorylation ( P =1.4×10 -6 , n=12,12 in each group). PPP1R12C-PP1c binding and PPP1R12C-MLC2a binding were significantly increased in AF ( P =2.9×10 -2 and 6.7×10 -3 respectively, n=8,8 in each group). In vitro studies utilizing drug BDP5290, which inhibits T560-PPP1R12C phosphorylation, demonstrated increased PPP1R12C binding with both PP1c and MLC2a, and dephosphorylation of MLC2a. Lenti-12C mice demonstrated a 150% increase in LA size versus controls ( P =5.0×10 -6 , n=12,8,12), with reduced atrial strain and atrial ejection fraction. Pacing-induced AF in Lenti-12C mice was significantly higher than controls ( P =1.8×10 -2 and 4.1×10 -2 respectively, n= 6,6,5). Conclusions AF patients exhibit increased levels of PPP1R12C protein compared to controls. PPP1R12C overexpression in mice increases PP1c targeting to MLC2a and causes MLC2a dephosphorylation, which reduces atrial contractility and increases AF inducibility. These findings suggest that PP1 regulation of sarcomere function at MLC2a is a key determinant of atrial contractility in AF.
Collapse
|
8
|
Wilson C, Zi M, Smith M, Hussain M, D’Souza A, Dobrzynski H, Boyett MR. Atrioventricular node dysfunction in pressure overload-induced heart failure—Involvement of the immune system and transcriptomic remodelling. Front Pharmacol 2023; 14:1083910. [PMID: 37081960 PMCID: PMC10110994 DOI: 10.3389/fphar.2023.1083910] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Heart failure is associated with atrioventricular (AV) node dysfunction, and AV node dysfunction in the setting of heart failure is associated with an increased risk of mortality and heart failure hospitalisation. This study aims to understand the causes of AV node dysfunction in heart failure by studying changes in the whole nodal transcriptome. The mouse transverse aortic constriction model of pressure overload-induced heart failure was studied; functional changes were assessed using electrocardiography and echocardiography and the transcriptome of the AV node was quantified using RNAseq. Heart failure was associated with a significant increase in the PR interval, indicating a slowing of AV node conduction and AV node dysfunction, and significant changes in 3,077 transcripts (5.6% of the transcriptome). Many systems were affected: transcripts supporting AV node conduction were downregulated and there were changes in transcripts identified by GWAS as determinants of the PR interval. In addition, there was evidence of remodelling of the sarcomere, a shift from fatty acid to glucose metabolism, remodelling of the extracellular matrix, and remodelling of the transcription and translation machinery. There was evidence of the causes of this widespread remodelling of the AV node: evidence of dysregulation of multiple intracellular signalling pathways, dysregulation of 109 protein kinases and 148 transcription factors, and an immune response with a proliferation of neutrophils, monocytes, macrophages and B lymphocytes and a dysregulation of 40 cytokines. In conclusion, inflammation and a widespread transcriptional remodelling of the AV node underlies AV node dysfunction in heart failure.
Collapse
Affiliation(s)
- Claire Wilson
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Min Zi
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Matthew Smith
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Munir Hussain
- Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Alicia D’Souza
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
- Department of Anatomy, Jagiellonian University Medical College, Kraków, Poland
- *Correspondence: Halina Dobrzynski, ; Mark R. Boyett,
| | - Mark R. Boyett
- Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
- *Correspondence: Halina Dobrzynski, ; Mark R. Boyett,
| |
Collapse
|
9
|
Ezeani M, Prabhu S. PI3K(p110α) as a determinant and gene therapy for atrial enlargement in atrial fibrillation. Mol Cell Biochem 2023; 478:471-490. [PMID: 35900667 PMCID: PMC9938077 DOI: 10.1007/s11010-022-04526-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is an irregular heart rhythm, characterised by chaotic atrial activation, which is promoted by remodelling. Once initiated, AF can also propagate the progression of itself in the so-called ''AF begets AF''. Several lines of investigation have shown that signalling molecules, including reactive oxygen species, angiotensin II, and phosphoinositide 3-kinases (PI3Ks), in presence or absence of cardiovascular disease risk factors, stabilise and promote AF maintenance. In particular, reduced cardiac-specific PI3K activity that is not associated with oncology is cardiotoxic and increases susceptibility to AF. Atrial-specific PI3K(p110α) transgene can cause pathological atrial enlargement. Highlighting the crucial importance of the p110α protein in a clinical problem that currently challenges the professional health care practice, in over forty (40) transgenic mouse models of AF (Table1), currently existing, of which some of the models are models of human genetic disorders, including PI3K(p110α) transgenic mouse model, over 70% of them reporting atrial size showed enlarged, greater atrial size. Individuals with minimal to severely dilated atria develop AF more likely. Left atrial diameter and volume stratification are an assessment for follow-up surveillance to detect AF. Gene therapy to reduce atrial size will be associated with a reduction in AF burden. In this overview, PI3K(p110α), a master regulator of organ size, was investigated in atrial enlargement and in physiological determinants that promote AF.
Collapse
Affiliation(s)
- Martin Ezeani
- NanoBiotechnology Laboratory, Central Clinical School, Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, 3004, Australia.
| | - Sandeep Prabhu
- The Alfred, and Baker Heart and Diabetes Institute, Melbourne, Australia
- The University of Melbourne, Melbourne, Australia
| |
Collapse
|
10
|
Murphy MB, Kannankeril PJ, Murray KT. Overview of programmed electrical stimulation to assess atrial fibrillation susceptibility in mice. Front Physiol 2023; 14:1149023. [PMID: 37113690 PMCID: PMC10126433 DOI: 10.3389/fphys.2023.1149023] [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: 01/20/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common human arrhythmia and is associated with increased risk of stroke, dementia, heart failure, and death. Among several animal models that have been used to investigate the molecular determinants of AF, mouse models have become the most prevalent due to low cost, ease of genetic manipulation, and similarity to human disease. Programmed electrical stimulation (PES) using intracardiac or transesophageal atrial pacing is used to induce AF as most mouse models do not develop spontaneous AF. However, there is a lack of standardized methodology resulting in numerous PES protocols in the literature that differ with respect to multiple parameters, including pacing protocol and duration, stimulus amplitude, pulse width, and even the definition of AF. Given this complexity, the selection of the appropriate atrial pacing protocol for a specific model has been arbitrary. Herein we review the development of intracardiac and transesophageal PES, including commonly used protocols, selected experimental models, and advantages and disadvantages of both techniques. We also emphasize detection of artifactual AF induction due to unintended parasympathetic stimulation, which should be excluded from results. We recommend that the optimal pacing protocol to elicit an AF phenotype should be individualized to the specific model of genetic or acquired risk factors, with an analysis using several definitions of AF as an endpoint.
Collapse
|
11
|
Li W, Cologna SM. Mass spectrometry-based proteomics in neurodegenerative lysosomal storage disorders. Mol Omics 2022; 18:256-278. [PMID: 35343995 PMCID: PMC9098683 DOI: 10.1039/d2mo00004k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The major function of the lysosome is to degrade unwanted materials such as lipids, proteins, and nucleic acids; therefore, deficits of the lysosomal system can result in improper degradation and trafficking of these biomolecules. Diseases associated with lysosomal failure can be lethal and are termed lysosomal storage disorders (LSDs), which affect 1 in 5000 live births collectively. LSDs are inherited metabolic diseases caused by mutations in single lysosomal and non-lysosomal proteins and resulting in the subsequent accumulation of macromolecules within. Most LSD patients present with neurodegenerative clinical symptoms, as well as damage in other organs. The discovery of new biomarkers is necessary to understand and monitor these diseases and to track therapeutic progress. Over the past ten years, mass spectrometry (MS)-based proteomics has flourished in the biomarker studies in many diseases, including neurodegenerative, and more specifically, LSDs. In this review, biomarkers of disease pathophysiology and monitoring of LSDs revealed by MS-based proteomics are discussed, including examples from Niemann-Pick disease type C, Fabry disease, neuronal ceroid-lipofuscinoses, mucopolysaccharidosis, Krabbe disease, mucolipidosis, and Gaucher disease.
Collapse
Affiliation(s)
- Wenping Li
- Department of Chemistry, University of Illinois at Chicago, USA.
| | | |
Collapse
|
12
|
Signaling Pathways Involved in Myocardial Ischemia-Reperfusion Injury and Cardioprotection: A Systematic Review of Transcriptomic Studies in Sus scrofa. J Cardiovasc Dev Dis 2022; 9:jcdd9050132. [PMID: 35621843 PMCID: PMC9145716 DOI: 10.3390/jcdd9050132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/17/2022] Open
Abstract
Myocardial damage in acute myocardial infarctions (AMI) is primarily the result of ischemia−reperfusion injury (IRI). Recognizing the timing of transcriptional events and their modulation by cardioprotective strategies is critical to address the pathophysiology of myocardial IRI. Despite the relevance of pigs for translational studies of AMI, only a few have identified how transcriptomic changes shape cellular signaling pathways in response to injury. We systematically reviewed transcriptomic studies of myocardial IRI and cardioprotection in Sus scrofa. Gene expression datasets were analyzed for significantly enriched terms using the Enrichr analysis tool, and statistically significant results (adjusted p-values of <0.05) for Signaling Pathways, Transcription Factors, Molecular Functions, and Biological Processes were compared between eligible studies to describe how these dynamic changes transform the myocardium from an injured and inflamed tissue into a scar. Then, we address how cardioprotective interventions distinctly modulate the myocardial transcriptome and discuss the implications of uncovering gene regulatory networks for cardiovascular pathologies and translational applications.
Collapse
|
13
|
Inhibition of RhoA and Cdc42 by miR-133a Modulates Retinoic Acid Signalling during Early Development of Posterior Cardiac Tube Segment. Int J Mol Sci 2022; 23:ijms23084179. [PMID: 35456995 PMCID: PMC9025022 DOI: 10.3390/ijms23084179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/15/2022] Open
Abstract
It is well known that multiple microRNAs play crucial roles in cardiovascular development, including miR-133a. Additionally, retinoic acid regulates atrial marker expression. In order to analyse the role of miR-133a as a modulator of retinoic acid signalling during the posterior segment of heart tube formation, we performed functional experiments with miR-133a and retinoic acid by means of microinjections into the posterior cardiac precursors of both primitive endocardial tubes in chick embryos. Subsequently, we subjected embryos to whole mount in situ hybridisation, immunohistochemistry and qPCR analysis. Our results demonstrate that miR-133a represses RhoA and Cdc42, as well as Raldh2/Aldh1a2, and the specific atrial markers Tbx5 and AMHC1, which play a key role during differentiation. Furthermore, we observed that miR-133a upregulates p21 and downregulates cyclin A by repressing RhoA and Cdc42, respectively, thus functioning as a cell proliferation inhibitor. Additionally, retinoic acid represses miR-133a, while it increases Raldh2, Tbx5 and AMHC1. Given that RhoA and Cdc42 are involved in Raldh2 expression and that they are modulated by miR-133a, which is influenced by retinoic acid signalling, our results suggest the presence of a negative feedback mechanism between miR-133a and retinoic acid during early development of the posterior cardiac tube segment. Despite additional unexplored factors being possible contributors to this negative feedback mechanism, miR-133a might also be considered as a potential therapeutic tool for the diagnosis, therapy and prognosis of cardiac diseases.
Collapse
|
14
|
Essandoh K, Auchus RJ, Brody MJ. Cardiac decompensation and promiscuous prenylation of small GTPases in cardiomyocytes in response to local mevalonate pathway disruption. J Pathol 2022; 256:249-252. [PMID: 34783037 PMCID: PMC8825694 DOI: 10.1002/path.5837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 11/12/2021] [Indexed: 11/09/2022]
Abstract
Investigations of major mevalonate pathway enzymes have demonstrated the importance of local isoprenoid synthesis in cardiac homeostasis. Farnesyl diphosphate synthase (FPPS) synthesizes isoprenoid precursors needed for cholesterol biosynthesis and protein prenylation. Wang, Zhang, Chen et al, in a recently published article in The Journal of Pathology, elegantly elucidated the pathological outcomes of FPPS deficiency in cardiomyocytes, which paradoxically resulted in increased prenylation of the small GTPases Ras and Rheb. Cardiomyocyte FPPS depletion caused severe dilated cardiomyopathy that was associated with enhanced GTP-loading and abundance of Ras and Rheb in lipidated protein-enriched cardiac fractions and robust activation of downstream hypertrophic ERK1/2 and mTOR signaling pathways. Cardiomyopathy and activation of ERK1/2 and mTOR caused by loss of FPPS were ameliorated by inhibition of farnesyltransferase, suggesting that impairment of FPPS activity results in promiscuous activation of Ras and Rheb through non-canonical actions of farnesyltransferase. Here, we discuss the findings and adaptive signaling mechanisms in response to disruption of local cardiomyocyte mevalonate pathway activity, highlighting how alteration in a key branch point in the mevalonate pathway affects cardiac biology and function and perturbs protein prenylation, which might unveil novel strategies and intricacies of targeting the mevalonate pathway to treat cardiovascular diseases. © 2021 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kobina Essandoh
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Richard J. Auchus
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Matthew J. Brody
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA,Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA,Correspondence to: Matthew J. Brody, PhD. Department of Pharmacology, University of Michigan Medical School, 1150 W. Medical Center Dr., Ann Arbor, MI 48109-5632, USA.
| |
Collapse
|
15
|
Sakabe M, Thompson M, Chen N, Verba M, Hassan A, Lu R, Xin M. Inhibition of β1-AR/Gαs signaling promotes cardiomyocyte proliferation in juvenile mice through activation of RhoA-YAP axis. eLife 2022; 11:74576. [PMID: 36479975 PMCID: PMC9767473 DOI: 10.7554/elife.74576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
The regeneration potential of the mammalian heart is incredibly limited, as cardiomyocyte proliferation ceases shortly after birth. β-adrenergic receptor (β-AR) blockade has been shown to improve heart functions in response to injury; however, the underlying mechanisms remain poorly understood. Here, we inhibited β-AR signaling in the heart using metoprolol, a cardio-selective β blocker for β1-adrenergic receptor (β1-AR) to examine its role in heart maturation and regeneration in postnatal mice. We found that metoprolol enhanced cardiomyocyte proliferation and promoted cardiac regeneration post myocardial infarction, resulting in reduced scar formation and improved cardiac function. Moreover, the increased cardiomyocyte proliferation was also induced by the genetic deletion of Gnas, the gene encoding G protein alpha subunit (Gαs), a downstream effector of β-AR. Genome wide transcriptome analysis revealed that the Hippo-effector YAP, which is associated with immature cardiomyocyte proliferation, was upregulated in the cardiomyocytes of β-blocker treated and Gnas cKO hearts. Moreover, the increased YAP activity is modulated by RhoA signaling. Our pharmacological and genetic studies reveal that β1-AR-Gαs-YAP signaling axis is involved in regulating postnatal cardiomyocyte proliferation. These results suggest that inhibiting β-AR-Gαs signaling promotes the regenerative capacity and extends the cardiac regenerative window in juvenile mice by activating YAP-mediated transcriptional programs.
Collapse
Affiliation(s)
- Masahide Sakabe
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Michael Thompson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Nong Chen
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mark Verba
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Aishlin Hassan
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Richard Lu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical CenterCincinnatiUnited States,Department of Pediatrics, College of Medicine, University of CincinnatiCincinnatiUnited States
| |
Collapse
|
16
|
Molecular Signaling to Preserve Mitochondrial Integrity against Ischemic Stress in the Heart: Rescue or Remove Mitochondria in Danger. Cells 2021; 10:cells10123330. [PMID: 34943839 PMCID: PMC8699551 DOI: 10.3390/cells10123330] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases are one of the leading causes of death and global health problems worldwide, and ischemic heart disease is the most common cause of heart failure (HF). The heart is a high-energy demanding organ, and myocardial energy reserves are limited. Mitochondria are the powerhouses of the cell, but under stress conditions, they become damaged, release necrotic and apoptotic factors, and contribute to cell death. Loss of cardiomyocytes plays a significant role in ischemic heart disease. In response to stress, protective signaling pathways are activated to limit mitochondrial deterioration and protect the heart. To prevent mitochondrial death pathways, damaged mitochondria are removed by mitochondrial autophagy (mitophagy). Mitochondrial quality control mediated by mitophagy is functionally linked to mitochondrial dynamics. This review provides a current understanding of the signaling mechanisms by which the integrity of mitochondria is preserved in the heart against ischemic stress.
Collapse
|
17
|
Wang L, Lu F, Xu J. Identification of Potential miRNA-mRNA Regulatory Network Contributing to Hypertrophic Cardiomyopathy (HCM). Front Cardiovasc Med 2021; 8:660372. [PMID: 34136543 PMCID: PMC8200816 DOI: 10.3389/fcvm.2021.660372] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/23/2021] [Indexed: 01/14/2023] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is a myocardial disease with unidentified pathogenesis. Increasing evidence indicated the potential role of microRNA (miRNA)-mRNA regulatory network in disease development. This study aimed to explore the miRNA-mRNA axis in HCM. Methods: The miRNA and mRNA expression profiles obtained from the Gene Expression Omnibus (GEO) database were used to identify differentially expressed miRNAs (DEMs) and genes (DEGs) between HCM and normal samples. Target genes of DEMs were determined by miRTarBase. Gene ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were conducted to identify biological functions of the DEGs and DEMs. miRNA-mRNA regulatory network was constructed to identify the hub genes and miRNAs. Logistic regression model for HCM prediction was established basing on the network. Results: A total of 224 upregulated and 366 downregulated DEGs and 10 upregulated and 14 downregulated DEMs were determined. We identified 384 DEM-targeted genes, and 20 of them were overlapped with the DEGs. The enriched functions include extracellular structure organization, organ growth, and phagosome and melanoma pathways. The four miRNAs and three mRNAs, including hsa-miR-373, hsa-miR-371-3p, hsa-miR-34b, hsa-miR-452, ARHGDIA, SEC61A1, and MYC, were identified through miRNA-mRNA regulatory network to construct the logistic regression model. The area under curve (AUC) values over 0.9 suggested the good performance of the model. Conclusion: The potential miRNA-mRNA regulatory network and established logistic regression model in our study may provide promising diagnostic methods for HCM.
Collapse
Affiliation(s)
- Lin Wang
- Cardiology Department, Tianjin Chest Hospital, Tianjin, China
| | | | | |
Collapse
|
18
|
Kilian LS, Voran J, Frank D, Rangrez AY. RhoA: a dubious molecule in cardiac pathophysiology. J Biomed Sci 2021; 28:33. [PMID: 33906663 PMCID: PMC8080415 DOI: 10.1186/s12929-021-00730-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/23/2021] [Indexed: 02/08/2023] Open
Abstract
The Ras homolog gene family member A (RhoA) is the founding member of Rho GTPase superfamily originally studied in cancer cells where it was found to stimulate cell cycle progression and migration. RhoA acts as a master switch control of actin dynamics essential for maintaining cytoarchitecture of a cell. In the last two decades, however, RhoA has been coined and increasingly investigated as an essential molecule involved in signal transduction and regulation of gene transcription thereby affecting physiological functions such as cell division, survival, proliferation and migration. RhoA has been shown to play an important role in cardiac remodeling and cardiomyopathies; underlying mechanisms are however still poorly understood since the results derived from in vitro and in vivo experiments are still inconclusive. Interestingly its role in the development of cardiomyopathies or heart failure remains largely unclear due to anomalies in the current data available that indicate both cardioprotective and deleterious effects. In this review, we aimed to outline the molecular mechanisms of RhoA activation, to give an overview of its regulators, and the probable mechanisms of signal transduction leading to RhoA activation and induction of downstream effector pathways and corresponding cellular responses in cardiac (patho)physiology. Furthermore, we discuss the existing studies assessing the presented results and shedding light on the often-ambiguous data. Overall, we provide an update of the molecular, physiological and pathological functions of RhoA in the heart and its potential in cardiac therapeutics.
Collapse
Affiliation(s)
- Lucia Sophie Kilian
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Jakob Voran
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany
| | - Derk Frank
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Internal Medicine III (Cardiology, Angiology, Intensive Care), University Medical Center Kiel, Rosalind-Franklin Str. 12, 24105, Kiel, Germany. .,DZHK (German Centre for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, 24105, Kiel, Germany. .,Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
19
|
Xu H, Shen Y, Liang C, Wang H, Huang J, Xue P, Luo M. Inhibition of the mevalonate pathway improves myocardial fibrosis. Exp Ther Med 2021; 21:224. [PMID: 33603833 PMCID: PMC7851600 DOI: 10.3892/etm.2021.9655] [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] [Received: 06/17/2020] [Accepted: 12/10/2020] [Indexed: 12/15/2022] Open
Abstract
The mevalonate (MVA) pathway serves an important role in ventricular remodeling. Targeting the MVA pathway has protective effects against myocardial fibrosis. The present study aimed to investigate the mechanism behind these effects. Primary cultured cardiac fibroblasts from C57BL/6 mice were treated in vitro in 5 groups: i) negative control; ii) angiotensin II (Ang II) model (1x10-5 mol/l); iii) Ang II + rosuvastatin (ROS); iv) Ang II + alendronate (ALE); and v) Ang II + fasudil (FAS). Collagen and crystal violet staining were used to assess morphological changes in cardiac fibroblasts. Reverse transcription quantitative PCR and western blotting were used to analyze the expression of key signaling molecules involved in the MVA pathway. Collagen staining in the ALE, FAS, and ROS groups was weak compared with the Ang II group, while the rate of cell proliferation in the ROS, ALE, and FAS groups was slower compared with that in the Ang II group. In addition, the expression of key signaling molecules in the MVA pathway, including transforming growth factor-β1 (TGF-β1), heat shock protein 47 (HSP47), collagen type I α1 (COL1A1), vascular endothelial growth factor 2 (VEGF2) and fibroblast growth factor 2 (FGF2), was decreased in the FAS and ROS groups compared with the Ang II model. Compared with the Ang II group, 3-Hydroxy-3-Methylglutaryl-CoA reductase (HMGCR) gene expression was significantly lowered in the drug intervention groups, whereas farnesyl pyrophosphate synthase (FDPS) expression was downregulated in the ALE group, but elevated in the FAS and ROS groups. Compared with that in the Ang II group, ras homolog family member A (RhoA) expression was downregulated in the FAS and ROS groups, whilst mevalonate kinase expression was reduced in the ROS group. Protein expression of TGF-β1, COL1A1 and HSP47 were decreased following intervention with each of the three drugs compared with the Ang II group. Overall, rosuvastatin, aledronate and fasudil decreased the proliferation of myocardial fibroblasts and inhibited collagen synthesis. Rosuvastatin had the strongest protective effects against myocardial fibrosis compared with the other drugs tested, suggesting this to be a potential agent for the clinical treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Huifeng Xu
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Yi Shen
- Department of Geriatrics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Chenyu Liang
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Haifeng Wang
- Department of Geriatrics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Junling Huang
- Department of Geriatrics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Pengcheng Xue
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ming Luo
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| |
Collapse
|
20
|
Kakinuma Y. Characteristic Effects of the Cardiac Non-Neuronal Acetylcholine System Augmentation on Brain Functions. Int J Mol Sci 2021; 22:ijms22020545. [PMID: 33430415 PMCID: PMC7826949 DOI: 10.3390/ijms22020545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/05/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Since the discovery of non-neuronal acetylcholine in the heart, this specific system has drawn scientific interest from many research fields, including cardiology, immunology, and pharmacology. This system, acquired by cardiomyocytes independent of the parasympathetic nervous system of the autonomic nervous system, helps us to understand unsolved issues in cardiac physiology and to realize that the system may be more pivotal for cardiac homeostasis than expected. However, it has been shown that the effects of this system may not be restricted to the heart, but rather extended to cover extra-cardiac organs. To this end, this system intriguingly influences brain function, specifically potentiating blood brain barrier function. Although the results reported appear to be unusual, this novel characteristic can provide us with another research interest and therapeutic application mode for central nervous system diseases. In this review, we discuss our recent studies and raise the possibility of application of this system as an adjunctive therapeutic modality.
Collapse
Affiliation(s)
- Yoshihiko Kakinuma
- Department of Bioregulatory Science, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
21
|
van Wijk SW, Ramos KS, Brundel BJJM. Cardioprotective Role of Heat Shock Proteins in Atrial Fibrillation: From Mechanism of Action to Therapeutic and Diagnostic Target. Int J Mol Sci 2021; 22:ijms22010442. [PMID: 33466228 PMCID: PMC7795054 DOI: 10.3390/ijms22010442] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
Atrial fibrillation (AF) is the most common age-related cardiac arrhythmia worldwide and is associated with ischemic stroke, heart failure, and substantial morbidity and mortality. Unfortunately, current AF therapy is only moderately effective and does not prevent AF progression from recurrent intermittent episodes (paroxysmal) to persistent and finally permanent AF. It has been recognized that AF persistence is related to the presence of electropathology. Electropathology is defined as structural damage, including degradation of sarcomere structures, in the atrial tissue which, in turn, impairs electrical conduction and subsequently the contractile function of atrial cardiomyocytes. Recent research findings indicate that derailed proteostasis underlies structural damage and, consequently, electrical conduction impairment. A healthy proteostasis is of vital importance for proper function of cells, including cardiomyocytes. Cells respond to a loss of proteostatic control by inducing a heat shock response (HSR), which results in heat shock protein (HSP) expression. Emerging clinical evidence indicates that AF-induced proteostasis derailment is rooted in exhaustion of HSPs. Cardiomyocytes lose defense against structural damage-inducing pathways, which drives progression of AF and induction of HSP expression. In particular, small HSPB1 conserves sarcomere structures by preventing their degradation by proteases, and overexpression of HSPB1 accelerates recovery from structural damage in experimental AF model systems. In this review, we provide an overview of the mechanisms of action of HSPs in preventing AF and discuss the therapeutic potential of HSP-inducing compounds in clinical AF, as well as the potential of HSPs as biomarkers to discriminate between the various stages of AF and recurrence of AF after treatment.
Collapse
Affiliation(s)
- Stan W. van Wijk
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (K.S.R.); (B.J.J.M.B.)
- Correspondence:
| | - Kennedy S. Ramos
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (K.S.R.); (B.J.J.M.B.)
- Erasmus Medical Center, Department of Cardiology, 3015 GD Rotterdam, The Netherlands
| | - Bianca J. J. M. Brundel
- Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HZ Amsterdam, The Netherlands; (K.S.R.); (B.J.J.M.B.)
| |
Collapse
|
22
|
Small G-protein RhoA is a potential inhibitor of cardiac fast sodium current. J Physiol Biochem 2020; 77:13-23. [PMID: 33145656 DOI: 10.1007/s13105-020-00774-w] [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: 06/26/2020] [Accepted: 10/27/2020] [Indexed: 12/30/2022]
Abstract
Small G-proteins of Rho family modulate the activity of several classes of ion channels, including K+ channels Kv1.2, Kir2.1, and ERG; Ca2+ channels; and epithelial Na+ channels. The present study was aimed to check the RhoA potential regulatory effects on Na+ current (INa) transferred by Na+ channel cardiac isoform NaV1.5 in heterologous expression system and in native rat cardiomyocytes. Whole-cell patch-clamp experiments showed that coexpression of NaV1.5 with the wild-type RhoA in CHO-K1 cell line caused 2.7-fold decrease of INa density with minimal influence on steady-state activation and inactivation. This effect was reproduced by the coexpression with a constitutively active RhoA, but not with a dominant negative RhoA. In isolated ventricular rat cardiomyocytes, a 5-h incubation with the RhoA activator narciclasine (5 × 10-6 M) reduced the maximal INa density by 38.8%. The RhoA-selective inhibitor rhosin (10-5 M) increased the maximal INa density by 25.3%. Experiments with sharp microelectrode recordings in isolated right ventricular wall preparations showed that 5 × 10-6 M narciclasine induced a significant reduction of action potential upstroke velocity after 2 h of incubation. Thus, RhoA might be considered as a potential negative regulator of sodium channels cardiac isoform NaV1.5.
Collapse
|
23
|
Soppert J, Lehrke M, Marx N, Jankowski J, Noels H. Lipoproteins and lipids in cardiovascular disease: from mechanistic insights to therapeutic targeting. Adv Drug Deliv Rev 2020; 159:4-33. [PMID: 32730849 DOI: 10.1016/j.addr.2020.07.019] [Citation(s) in RCA: 145] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
With cardiovascular disease being the leading cause of morbidity and mortality worldwide, effective and cost-efficient therapies to reduce cardiovascular risk are highly needed. Lipids and lipoprotein particles crucially contribute to atherosclerosis as underlying pathology of cardiovascular disease and influence inflammatory processes as well as function of leukocytes, vascular and cardiac cells, thereby impacting on vessels and heart. Statins form the first-line therapy with the aim to block cholesterol synthesis, but additional lipid-lowering drugs are sometimes needed to achieve low-density lipoprotein (LDL) cholesterol target values. Furthermore, beyond LDL cholesterol, also other lipid mediators contribute to cardiovascular risk. This review comprehensively discusses low- and high-density lipoprotein cholesterol, lipoprotein (a), triglycerides as well as fatty acids and derivatives in the context of cardiovascular disease, providing mechanistic insights into their role in pathological processes impacting on cardiovascular disease. Also, an overview of applied as well as emerging therapeutic strategies to reduce lipid-induced cardiovascular burden is provided.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Nikolaus Marx
- Medical Clinic I, University Hospital Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, Maastricht University, the Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, the Netherlands.
| |
Collapse
|
24
|
Ren YS, Li HH, Yao JC, Tan YJ, Pan LH, Peng T, Zhao LL, Zhang GM, Yue J, Hu XM, Liu Z, Li J. Application quantitative proteomics approach to identify differentially expressed proteins associated with cardiac protection mediated by cycloastragenol in acute myocardial infarction rats. J Proteomics 2020; 222:103691. [PMID: 32068187 DOI: 10.1016/j.jprot.2020.103691] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/10/2020] [Accepted: 02/14/2020] [Indexed: 01/18/2023]
Abstract
Acute myocardial infarction (AMI) is an acute heart disease. Cycloastragenol, as a natural product, inhibits inflammation and protects cardiomyocytes. Cycloastragenol (Y006) modulates inflammation in AMI is not known. To explore the function of Cycloastragenol in AMI, this study investigated the effect of Y006 and its mechanisms both in vitro and in vivo. Y006 influences the concentration of 11 proteins, as shown by a proteomics analysis, immunohistochemistry and western blotting. Among these 11 proteins, Erk1/2, PLCG1, IKBKG, and ZEB1 are related to inflammatory regulation. BAX, COX2, and GSK3β are involved in modulating cardiomyocyte apoptosis, and RhoA and DSC2 are directly associated with myocardial function. However, the functions of ARHGAP17 and Rit2 in heart are less well established. Additionally, Y006 suppressed TNF-α, IFN-γ and IL-17 production in PBMCs (peripheral blood monocytes) from patients with acute myocardial infarction and enhanced IL-10 and IL-4 expression. Similar results were obtained in a rat model of AMI by flow cytometry detection and ELISA. Our findings indicate that Y006 protects rats from AMI through direct or indirect inhibition of inflammation and cardiomyocyte apoptosis. However, the specific mechanism of Y006's protective function requires further study. Nonetheless, this research revealed a novel aspect for the treatment of myocardial infarction. SIGNIFICANCE: In the present study, we undertook the first proteomic evaluation of Cycloastragenol (Y006) function in acute myocardial infarction (AMI). Y006 significantly improved myocardial function in vivo by regulating multiple molecular expressions. Hypoxia is a direct reason for AMI. And our data support a role of Y006 in gene expression, cell apoptosis under hypoxia. The conclusions of this research assist to explain the potential molecular mechanism in Cycloastragenol treating AMI and supply a new method for ameliorating AMI.
Collapse
Affiliation(s)
- Yu-Shan Ren
- Department of Immunology, Binzhou Medical University, Yantai 264003, China
| | - Hong-Hua Li
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Jing-Chun Yao
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Yu-Jun Tan
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Li-Hong Pan
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Tao Peng
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Li-Li Zhao
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; National Engineering & Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Gui-Min Zhang
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; National Engineering & Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; School of Pharmacy, Linyi University, Linyi, China
| | - Jiang Yue
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
| | - Xue-Mei Hu
- Department of Immunology, Binzhou Medical University, Yantai 264003, China
| | - Zhong Liu
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; National Engineering & Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China
| | - Jie Li
- National Engineering Laboratory of High Level Expression in Mammalian Cells, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Lunan Pharmaceutical Group Co., Ltd., Linyi, China; National Engineering & Technology Research Center of Chirality Pharmaceutical, Lunan Pharmaceutical Group Co., Ltd., Linyi, China.
| |
Collapse
|
25
|
Scholz B, Schulte JS, Hamer S, Himmler K, Pluteanu F, Seidl MD, Stein J, Wardelmann E, Hammer E, Völker U, Müller FU. HDAC (Histone Deacetylase) Inhibitor Valproic Acid Attenuates Atrial Remodeling and Delays the Onset of Atrial Fibrillation in Mice. Circ Arrhythm Electrophysiol 2019; 12:e007071. [PMID: 30879335 PMCID: PMC6426346 DOI: 10.1161/circep.118.007071] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Supplemental Digital Content is available in the text. Background: A structural, electrical and metabolic atrial remodeling is central in the development of atrial fibrillation (AF) contributing to its initiation and perpetuation. In the heart, HDACs (histone deacetylases) control remodeling associated processes like hypertrophy, fibrosis, and energy metabolism. Here, we analyzed, whether the HDAC class I/IIa inhibitor valproic acid (VPA) is able to attenuate atrial remodeling in CREM-IbΔC-X (cAMP responsive element modulator isoform IbΔC-X) transgenic mice, a mouse model of extensive atrial remodeling with age-dependent progression from spontaneous atrial ectopy to paroxysmal and finally long-lasting AF. Methods: VPA was administered for 7 or 25 weeks to transgenic and control mice. Atria were analyzed macroscopically and using widefield and electron microscopy. Action potentials were recorded from atrial cardiomyocytes using patch-clamp technique. ECG recordings documented the onset of AF. A proteome analysis with consecutive pathway mapping identified VPA-mediated proteomic changes and related pathways. Results: VPA attenuated many components of atrial remodeling that are present in transgenic mice, animal AF models, and human AF. VPA significantly (P<0.05) reduced atrial dilatation, cardiomyocyte enlargement, atrial fibrosis, and the disorganization of myocyte’s ultrastructure. It significantly reduced the occurrence of atrial thrombi, reversed action potential alterations, and finally delayed the onset of AF by 4 to 8 weeks. Increased histone H4-acetylation in atria from VPA-treated transgenic mice verified effective in vivo HDAC inhibition. Cardiomyocyte-specific genetic inactivation of HDAC2 in transgenic mice attenuated the ultrastructural disorganization of myocytes comparable to VPA. Finally, VPA restrained dysregulation of proteins in transgenic mice that are involved in a multitude of AF relevant pathways like oxidative phosphorylation or RhoA (Ras homolog gene family, member A) signaling and disease functions like cardiac fibrosis and apoptosis of muscle cells. Conclusions: Our results suggest that VPA, clinically available, well-tolerated, and prescribed to many patients for years, has the therapeutic potential to delay the development of atrial remodeling and the onset of AF in patients at risk.
Collapse
Affiliation(s)
- Beatrix Scholz
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Jan Sebastian Schulte
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Sabine Hamer
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Kirsten Himmler
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Florentina Pluteanu
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Matthias Dodo Seidl
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Juliane Stein
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| | - Eva Wardelmann
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, Germany (E.W.)
| | - Elke Hammer
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Uwe Völker
- Interfaculty Institute of Genetics und Functional Genomics, University Medicine Greifswald, Germany (E.H., U.V.).,DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Germany (E.H., U.V.)
| | - Frank Ulrich Müller
- Institute of Pharmacology and Toxicology, University of Münster, Germany (B.S., J.S.S., S.H., K.H., F.P., M.D.S., J.S., F.U.M.)
| |
Collapse
|
26
|
Strassheim D, Gerasimovskaya E, Irwin D, Dempsey EC, Stenmark K, Karoor V. RhoGTPase in Vascular Disease. Cells 2019; 8:E551. [PMID: 31174369 PMCID: PMC6627336 DOI: 10.3390/cells8060551] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/24/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022] Open
Abstract
Ras-homologous (Rho)A/Rho-kinase pathway plays an essential role in many cellular functions, including contraction, motility, proliferation, and apoptosis, inflammation, and its excessive activity induces oxidative stress and promotes the development of cardiovascular diseases. Given its role in many physiological and pathological functions, targeting can result in adverse effects and limit its use for therapy. In this review, we have summarized the role of RhoGTPases with an emphasis on RhoA in vascular disease and its impact on endothelial, smooth muscle, and heart and lung fibroblasts. It is clear from the various studies that understanding the regulation of RhoGTPases and their regulators in physiology and pathological conditions is required for effective targeting of Rho.
Collapse
Affiliation(s)
- Derek Strassheim
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Evgenia Gerasimovskaya
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - David Irwin
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Edward C Dempsey
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA.
| | - Kurt Stenmark
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Department of Pediatrics, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| | - Vijaya Karoor
- Cardiovascular and Pulmonary Research Lab, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
- Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045, USA.
| |
Collapse
|
27
|
Brand CS, Lighthouse JK, Trembley MA. Protective transcriptional mechanisms in cardiomyocytes and cardiac fibroblasts. J Mol Cell Cardiol 2019; 132:1-12. [PMID: 31042488 DOI: 10.1016/j.yjmcc.2019.04.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/19/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Heart failure is the leading cause of morbidity and mortality worldwide. Several lines of evidence suggest that physical activity and exercise can pre-condition the heart to improve the response to acute cardiac injury such as myocardial infarction or ischemia/reperfusion injury, preventing the progression to heart failure. It is becoming more apparent that cardioprotection is a concerted effort between multiple cell types and converging signaling pathways. However, the molecular mechanisms of cardioprotection are not completely understood. What is clear is that the mechanisms underlying this protection involve acute activation of transcriptional activators and their corresponding gene expression programs. Here, we review the known stress-dependent transcriptional programs that are activated in cardiomyocytes and cardiac fibroblasts to preserve function in the adult heart after injury. Focus is given to prominent transcriptional pathways such as mechanical stress or reactive oxygen species (ROS)-dependent activation of myocardin-related transcription factors (MRTFs) and transforming growth factor beta (TGFβ), and gene expression that positively regulates protective PI3K/Akt signaling. Together, these pathways modulate both beneficial and pathological responses to cardiac injury in a cell-specific manner.
Collapse
Affiliation(s)
- Cameron S Brand
- Department of Pharmacology, School of Medicine, University of California - San Diego, 9500 Gilman Drive, Biomedical Sciences Building, La Jolla, CA 92093, USA.
| | - Janet K Lighthouse
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Box CVRI, Rochester, NY 14624, USA.
| | - Michael A Trembley
- Department of Cardiology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Song HY, Chien CS, Yarmishyn AA, Chou SJ, Yang YP, Wang ML, Wang CY, Leu HB, Yu WC, Chang YL, Chiou SH. Generation of GLA-Knockout Human Embryonic Stem Cell Lines to Model Autophagic Dysfunction and Exosome Secretion in Fabry Disease-Associated Hypertrophic Cardiomyopathy. Cells 2019; 8:cells8040327. [PMID: 30965672 PMCID: PMC6523555 DOI: 10.3390/cells8040327] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022] Open
Abstract
Fabry disease (FD) is a rare inherited disorder characterized by a wide range of systemic symptoms; it is particularly associated with cardiovascular and renal problems. Enzyme replacement therapy and pharmacological chaperone migalastat are the only approved and effective treatment strategies for FD patients. It is well documented that alpha-galactosidase A (GLA) enzyme activity deficiency causes globotriaosylceramide (Gb3) accumulation, which plays a crucial role in the etiology of FD. However, the detailed mechanisms remain unclear, and the lack of a reliable and powerful disease model is an obstacle. In this study, we created such a model by using CRISPR/Cas9-mediated editing of GLA gene to knockout its expression in human embryonic stem cells (hESCs). The cardiomyocytes differentiated from these hESCs (GLA-null CMs) were characterized by the accumulation of Gb3 and significant increases of cell surface area, the landmarks of FD-associated cardiomyopathy. Furthermore, we used mass spectrometry to compare the proteomes of GLA-null CMs and parental wild type CMs and found that the Rab GTPases involved in exocytotic vesicle release were significantly downregulated. This caused impairment of autophagic flux and protein turnover, resulting in an increase of reactive oxygen species and apoptosis. To summarize, we established a FD model which can be used as a promising tool to study human hypertrophic cardiomyopathy in a physiologically and pathologically relevant manner and to develop new therapies by targeting Rab GTPases signaling-related exosomal vesicles transportation.
Collapse
Affiliation(s)
- Hui-Yung Song
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Chian-Shiu Chien
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Aliaksandr A Yarmishyn
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Shih-Jie Chou
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Mong-Lien Wang
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
| | - Chien-Ying Wang
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Emergent Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Hsin-Bang Leu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Division of Cardiology & Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Wen-Chung Yu
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Division of Cardiology & Department of Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Yuh-Lih Chang
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Pharmacy, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
| | - Shih-Hwa Chiou
- Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan.
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan.
- School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Genomics Research Center, Academia Sinica, Taipei 11574, Taiwan.
| |
Collapse
|
29
|
Kant S, Freytag B, Herzog A, Reich A, Merkel R, Hoffmann B, Krusche CA, Leube RE. Desmoglein 2 mutation provokes skeletal muscle actin expression and accumulation at intercalated discs in murine hearts. J Cell Sci 2019; 132:jcs.199612. [PMID: 30659114 DOI: 10.1242/jcs.199612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/30/2018] [Indexed: 01/05/2023] Open
Abstract
Arrhythmogenic cardiomyopathy (AC) is an incurable progressive disease that is linked to mutations in genes coding for components of desmosomal adhesions that are localized to the intercalated disc region, which electromechanically couples adjacent cardiomyocytes. To date, the underlying molecular dysfunctions are not well characterized. In two murine AC models, we find an upregulation of the skeletal muscle actin gene (Acta1), which is known to be a compensatory reaction to compromised heart function. Expression of this gene is elevated prior to visible morphological alterations and clinical symptoms, and persists throughout pathogenesis with an additional major rise during the chronic disease stage. We provide evidence that the increased Acta1 transcription is initiated through nuclear activation of the serum response transcription factor (SRF) by its transcriptional co-activator megakaryoblastic leukemia 1 protein (MKL1, also known as MRTFA). Our data further suggest that perturbed desmosomal adhesion causes Acta1 overexpression during the early stages of the disease, which is amplified by transforming growth factor β (TGFβ) release from fibrotic lesions and surrounding cardiomyocytes during later disease stages. These observations highlight a hitherto unknown molecular AC pathomechanism.
Collapse
Affiliation(s)
- Sebastian Kant
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Benjamin Freytag
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Antonia Herzog
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Anna Reich
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf Merkel
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Bernd Hoffmann
- Forschungszentrum Jülich, Institute of Complex Systems, ICS-7, Biomechanics, 52428 Jülich, Germany
| | - Claudia A Krusche
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| | - Rudolf E Leube
- Institute of Molecular and Cellular Anatomy, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
30
|
Disruption of RHOA‐ROCK Signaling Results in Atrioventricular Block and Disturbed Development of the Putative Atrioventricular Node. Anat Rec (Hoboken) 2018; 302:83-92. [DOI: 10.1002/ar.23912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/07/2018] [Accepted: 02/25/2018] [Indexed: 12/27/2022]
|
31
|
Adriaens ME, Lodder EM, Moreno‐Moral A, Šilhavý J, Heinig M, Glinge C, Belterman C, Wolswinkel R, Petretto E, Pravenec M, Remme CA, Bezzina CR. Systems Genetics Approaches in Rat Identify Novel Genes and Gene Networks Associated With Cardiac Conduction. J Am Heart Assoc 2018; 7:e009243. [PMID: 30608189 PMCID: PMC6404199 DOI: 10.1161/jaha.118.009243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/03/2018] [Indexed: 01/20/2023]
Abstract
Background Electrocardiographic ( ECG ) parameters are regarded as intermediate phenotypes of cardiac arrhythmias. Insight into the genetic underpinnings of these parameters is expected to contribute to the understanding of cardiac arrhythmia mechanisms. Here we used HXB / BXH recombinant inbred rat strains to uncover genetic loci and candidate genes modulating ECG parameters. Methods and Results RR interval, PR interval, QRS duration, and QT c interval were measured from ECG s obtained in 6 male rats from each of the 29 available HXB / BXH recombinant inbred strains. Genes at loci displaying significant quantitative trait loci (QTL) effects were prioritized by assessing the presence of protein-altering variants, and by assessment of cis expression QTL ( eQTL ) effects and correlation of transcript abundance to the respective trait in the heart. Cardiac RNA -seq data were additionally used to generate gene co-expression networks. QTL analysis of ECG parameters identified 2 QTL for PR interval, respectively, on chromosomes 10 and 17. At the chromosome 10 QTL , cis- eQTL effects were identified for Acbd4, Cd300lg, Fam171a2, and Arhgap27; the transcript abundance in the heart of these 4 genes was correlated with PR interval. At the chromosome 17 QTL , a cis- eQTL was uncovered for Nhlrc1 candidate gene; the transcript abundance of this gene was also correlated with PR interval. Co-expression analysis furthermore identified 50 gene networks, 6 of which were correlated with PR interval or QRS duration, both parameters of cardiac conduction. Conclusions These newly identified genetic loci and gene networks associated with the ECG parameters of cardiac conduction provide a starting point for future studies with the potential of identifying novel mechanisms underlying cardiac electrical function.
Collapse
Affiliation(s)
- Michiel E. Adriaens
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
- Maastricht Centre for Systems BiologyMaastricht UniversityMaastrichtThe Netherlands
| | - Elisabeth M. Lodder
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| | | | - Jan Šilhavý
- Institute of PhysiologyAcademy of Sciences of the Czech RepublicPragueCzech Republic
| | - Matthias Heinig
- Institute of Computational BiologyHelmholtz Zentrum MünchenMünchenGermany
| | - Charlotte Glinge
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| | - Charly Belterman
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| | - Rianne Wolswinkel
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| | - Enrico Petretto
- The MRC London Institute of Medical SciencesImperial College LondonLondonUnited Kingdom
- Duke‐NUS Medical SchoolSingapore
| | - Michal Pravenec
- Institute of PhysiologyAcademy of Sciences of the Czech RepublicPragueCzech Republic
| | - Carol Ann Remme
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| | - Connie R. Bezzina
- Department of Experimental CardiologyHeart CentreAcademic Medical Center AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
32
|
Frank DU, Sutcliffe MD, Saucerman JJ. Network-based predictions of in vivo cardiac hypertrophy. J Mol Cell Cardiol 2018; 121:180-189. [PMID: 30030017 DOI: 10.1016/j.yjmcc.2018.07.243] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/12/2018] [Accepted: 07/16/2018] [Indexed: 12/13/2022]
Abstract
Cardiac hypertrophy is a common response of cardiac myocytes to stress and a predictor of heart failure. While in vitro cell culture studies have identified numerous molecular mechanisms driving hypertrophy, it is unclear to what extent these mechanisms can be integrated into a consistent framework predictive of in vivo phenotypes. To address this question, we investigate the degree to which an in vitro-based, manually curated computational model of the hypertrophy signaling network is able to predict in vivo hypertrophy of 52 cardiac-specific transgenic mice. After minor revisions motivated by in vivo literature, the model concordantly predicts the qualitative responses of 78% of output species and 69% of signaling intermediates within the network model. Analysis of four double-transgenic mouse models reveals that the computational model robustly predicts hypertrophic responses in mice subjected to multiple, simultaneous perturbations. Thus the model provides a framework with which to mechanistically integrate data from multiple laboratories and experimental systems to predict molecular regulation of cardiac hypertrophy.
Collapse
Affiliation(s)
- Deborah U Frank
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States; Department of Pediatrics, University of Virginia, HSC Box 800386, Charlottesville 22908-0386, VA, United States.
| | - Matthew D Sutcliffe
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States; Department of Pediatrics, University of Virginia, HSC Box 800386, Charlottesville 22908-0386, VA, United States.
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Box 800759, Charlottesville 22908, VA, United States.
| |
Collapse
|
33
|
Vicente-Steijn R, Kelder TP, Tertoolen LG, Wisse LJ, Pijnappels DA, Poelmann RE, Schalij MJ, deRuiter MC, Gittenberger-de Groot AC, Jongbloed MRM. RHOA-ROCK signalling is necessary for lateralization and differentiation of the developing sinoatrial node. Cardiovasc Res 2018; 113:1186-1197. [PMID: 28899000 DOI: 10.1093/cvr/cvx104] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 07/03/2017] [Indexed: 01/16/2023] Open
Abstract
Aims RHOA-ROCK signalling regulates cell migration, proliferation, differentiation, and transcription. RHOA is expressed in the developing cardiac conduction system in chicken and mice. In early development, the entire sinus venosus myocardium, including both the transient left-sided and the definitive sinoatrial node (SAN), has pacemaker potential. Later, pacemaker potential is restricted to the right-sided SAN. Disruption of RHOA expression in adult mice causes arrhythmias including bradycardia and atrial fibrillation, the mechanism of which is unknown but presumed to affect the SAN. The aim of this study is to assess the role of RHOA-ROCK signalling in SAN development in the chicken heart. Methods and results ROCK signalling was inhibited chemically in embryonic chicken hearts using Y-27632. This prolonged the immature state of the sinus venosus myocardium, evidenced by up-regulation of the transcription factor ISL1, wide distribution of pacemaker potential, and significantly reduced heart rate. Furthermore ROCK inhibition caused aberrant expression of typical SAN genes: ROCK1, ROCK2, SHOX2, TBX3, TBX5, ISL1, HCN4, CX40, CAV3.1, and NKX2.5 and left-right asymmetry genes: PITX2C and NODAL. Anatomical abnormalities in pulmonary vein development were also observed. Patch clamp electrophysiology confirmed the immature phenotype of the SAN cells and a residual left-sided sinus venosus myocardium pacemaker-like potential. Conclusions RHOA-ROCK signalling is involved in establishing the right-sided SAN as the definitive pacemaker of the heart and restricts typical pacemaker gene expression to the right side of the sinus venosus myocardium.
Collapse
Affiliation(s)
- Rebecca Vicente-Steijn
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.,ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| | - Tim P Kelder
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Leon G Tertoolen
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lambertus J Wisse
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Daniël A Pijnappels
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert E Poelmann
- Sylvius Laboratory, Institute of Biology Leiden (IBL), Leiden University, Leiden, The Netherlands
| | - Martin J Schalij
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C deRuiter
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Adriana C Gittenberger-de Groot
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Monique R M Jongbloed
- Department of Anatomy & Embryology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
34
|
Zafirovic S, Sudar-Milovanovic E, Obradovic M, Djordjevic J, Jasnic N, Borovic ML, Isenovic ER. Involvement of PI3K, Akt and RhoA in Oestradiol Regulation of Cardiac iNOS Expression. Curr Vasc Pharmacol 2018; 17:307-318. [PMID: 29437011 DOI: 10.2174/1570161116666180212142414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/06/2018] [Accepted: 02/07/2018] [Indexed: 01/12/2023]
Abstract
BACKGROUND Oestradiol is an important regulatory factor with several positive effects on the cardiovascular (CV) system. We evaluated the molecular mechanism of the in vivo effects of oestradiol on the regulation of cardiac inducible nitric oxide (NO) synthase (iNOS) expression and activity. METHODS Male Wistar rats were treated with oestradiol (40 mg/kg, intraperitoneally) and after 24 h the animals were sacrificed. The concentrations of NO and L-Arginine (L-Arg) were determined spectrophotometrically. For protein expressions of iNOS, p65 subunit of nuclear factor-κB (NFκB-p65), Ras homolog gene family-member A (RhoA), angiotensin II receptor type 1 (AT1R), insulin receptor substrate 1 (IRS-1), p85, p110 and protein kinase B (Akt), Western blot method was used. Coimmunoprecipitation was used for measuring the association of IRS-1 with the p85 subunit of phosphatidylinositol- 3-kinase (PI3K). The expression of iNOS messenger ribonucleic acid (mRNA) was measured with the quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemical analysis of the tissue was used to detect localization and expression of iNOS in heart tissue. RESULTS Oestradiol treatment reduced L-Arg concentration (p<0.01), iNOS mRNA (p<0.01) and protein (p<0.001) expression, level of RhoA (p<0.05) and AT1R (p<0.001) protein. In contrast, plasma NO (p<0.05), Akt phosphorylation at Thr308 (p<0.05) and protein level of p85 (p<0.001) increased after oestradiol treatment. CONCLUSION Our results suggest that oestradiol in vivo regulates cardiac iNOS expression via the PI3K/Akt signaling pathway, through attenuation of RhoA and AT1R.
Collapse
Affiliation(s)
- Sonja Zafirovic
- Institute of Nuclear Sciences "Vinca", Department of Radiobiology and Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Emina Sudar-Milovanovic
- Institute of Nuclear Sciences "Vinca", Department of Radiobiology and Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Milan Obradovic
- Institute of Nuclear Sciences "Vinca", Department of Radiobiology and Molecular Genetics, University of Belgrade, Belgrade, Serbia
| | - Jelena Djordjevic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Nebojsa Jasnic
- Institute of Physiology and Biochemistry, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Milica Labudovic Borovic
- Institute of Histology and Embryology "Aleksandar D. Kostic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Esma R Isenovic
- Institute of Nuclear Sciences "Vinca", Department of Radiobiology and Molecular Genetics, University of Belgrade, Belgrade, Serbia.,Faculty of Stomatology, Pancevo, University Business Academy, Novi Sad, Serbia
| |
Collapse
|
35
|
Abreu Velez AM, Howard MS, Velazquez-Velez JE. Cardiac rhythm and pacemaking abnormalities in patients affected by endemic pemphigus in Colombia may be the result of deposition of autoantibodies, complement, fibrinogen, and other molecules. Heart Rhythm 2017; 15:725-731. [PMID: 29277685 DOI: 10.1016/j.hrthm.2017.12.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND We previously showed that one-third of patients affected by endemic pemphigus foliaceus in El Bagre, Colombia (El Bagre-EPF), display autoreactivity to the heart. OBJECTIVE The purpose of this study was to investigate rhythm disturbances with the presence of autoantibodies and correlate them with ECG changes in these patients. METHODS We performed a study comparing 30 patients and 30 controls from the endemic area, matched by demographics, including age, sex, weight, work activities, and comorbidities. ECG as well as direct and indirect immunofluorescence, immunohistochemistry, and confocal microscopic studies focusing on cardiac node abnormalities were performed. Autopsies of 7 patients also were reviewed. RESULTS The main ECG abnormalities seen in the El Bagre-EPF patients were sinus bradycardia (in one-half), followed by left bundle branch block, left posterior fascicular block, and left anterior fascicular block compared with the controls. One-third of the patients displayed polyclonal autoantibodies against the sinoatrial and/or AV nodes and the His bundle correlating with rhythm anomalies and delays in the cardiac conduction system (P <.01). The patient antibodies colocalized with commercial antibodies to desmoplakins I and II, p0071, armadillo repeat gene deleted in velo-cardio-facial syndrome (ARVCF), and myocardium-enriched zonula occludens-1-associated protein (MYZAP; Progen Biotechnik) (P <.01). CONCLUSION One-third of the patients affected by El Bagre-EPF have rhythm abnormalities that slow the conduction of impulses in cardiac nodes and the cardiac conduction system. These abnormalities likely occur as a result of deposition of autoantibodies, complement, and other inflammatory molecules. We show for the first time that MYZAP is present in cardiac nodes.
Collapse
|
36
|
van Eldik W, den Adel B, Monshouwer-Kloots J, Salvatori D, Maas S, van der Made I, Creemers EE, Frank D, Frey N, Boontje N, van der Velden J, Steendijk P, Mummery C, Passier R, Beqqali A. Z-disc protein CHAPb induces cardiomyopathy and contractile dysfunction in the postnatal heart. PLoS One 2017; 12:e0189139. [PMID: 29206857 PMCID: PMC5716575 DOI: 10.1371/journal.pone.0189139] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 11/20/2017] [Indexed: 12/17/2022] Open
Abstract
Aims The Z-disc is a crucial structure of the sarcomere and is implicated in mechanosensation/transduction. Dysregulation of Z-disc proteins often result in cardiomyopathy. We have previously shown that the Z-disc protein Cytoskeletal Heart-enriched Actin-associated Protein (CHAP) is essential for cardiac and skeletal muscle development. Furthermore, the CHAP gene has been associated with atrial fibrillation in humans. Here, we studied the misregulated expression of CHAP isoforms in heart disease. Methods and results Mice that underwent transverse aortic constriction and calcineurin transgenic (Tg) mice, both models of experimental heart failure, displayed a significant increase in cardiac expression of fetal isoform CHAPb. To investigate whether increased expression of CHAPb postnatally is sufficient to induce cardiomyopathy, we generated CHAPb Tg mice under the control of the cardiac-specific αMHC promoter. CHAPb Tg mice displayed cardiac hypertrophy, interstitial fibrosis and enlargement of the left atrium at three months, which was more pronounced at the age of six months. Hypertrophy and fibrosis were confirmed by evidence of activation of the hypertrophic gene program (Nppa, Nppb, Myh7) and increased collagen expression, respectively. Connexin40 and 43 were downregulated in the left atrium, which was associated with delayed atrioventricular conduction. Tg hearts displayed both systolic and diastolic dysfunction partly caused by impaired sarcomere function evident from a reduced force generating capacity of single cardiomyocytes. This co-incided with activation of the actin signalling pathway leading to the formation of stress fibers. Conclusion This study demonstrated that the fetal isoform CHAPb initiates progression towards cardiac hypertrophy, which is accompanied by delayed atrioventricular conduction and diastolic dysfunction. Moreover, CHAP may be a novel therapeutic target or candidate gene for screening in cardiomyopathies and atrial fibrillation.
Collapse
Affiliation(s)
- Willemijn van Eldik
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Interuniversity Cardiology Institute of the Netherlands (ICIN), Utrecht, The Netherlands
| | - Brigit den Adel
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Daniela Salvatori
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Central Laboratory Animal Facility, Leiden University Medical Center, Leiden, The Netherlands
| | - Saskia Maas
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Central Laboratory Animal Facility, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingeborg van der Made
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Esther E. Creemers
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Derk Frank
- Department of Cardiology and Angiology, Universitätsklinikum Schleswig-Holstein (UKSH), University of Kiel, Kiel, Germany
| | - Norbert Frey
- Department of Cardiology and Angiology, Universitätsklinikum Schleswig-Holstein (UKSH), University of Kiel, Kiel, Germany
| | - Nicky Boontje
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Paul Steendijk
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert Passier
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Abdelaziz Beqqali
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Experimental Cardiology, Academic Medical Center, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
37
|
Antonopoulos AS, Margaritis M, Shirodaria C, Antoniades C. Translating the effects of statins: from redox regulation to suppression of vascular wall inflammation. Thromb Haemost 2017; 108:840-8. [PMID: 22872079 DOI: 10.1160/th12-05-0337] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/24/2012] [Indexed: 12/21/2022]
Abstract
Vascular oxidative stress is a key feature of atherogenesis, and targeting vascular redox signalling is a rational therapeutic goal in vascular disease pathogenesis. 3-hydroxy-3-methyl-glutaryl-CoA reductase inhibitors or statins are potent lipid-lowering drugs that improve cardiovascular outcomes. It is now widely accepted that cardiovascular disease prevention by statins is dependent not only on their lipid lowering effects, but also on their beneficial effects on vascular redox signalling. Cell culture and animal models have provided important findings on the effects of statins on vascular redox and nitric oxide bioavailability. Recent evidence from studies on human vessels has further enhanced our understanding of the "pleiotropic" effects of statins on vascular wall. Reversal of endothelial dysfunction in human vessels by statins is dependent on the mevalonate pathway and Rac1 inhibition. These critical steps are responsible for reducing NADPH-oxidase activity and improving tetrahydrobiopterin bioavailability and nitric oxide synthase (NOS) coupling in human vessels. However, mevalonate pathway inhibition has been also held responsible for some of the side effects observed after statin treatment. In this review we summarise the existing knowledge on the effects of statins on vascular biology by discussing key findings from basic science as well as recent evidence from translational studies in humans. Finally, we discuss emerging aspects of statin pleiotropy, such as their effects on adipose tissue biology and adipokine synthesis that may light additional mechanistic links between statin treatment and improvement of clinical outcome in primary and secondary prevention.
Collapse
|
38
|
Olgar Y, Celen MC, Yamasan BE, Ozturk N, Turan B, Ozdemir S. Rho-kinase inhibition reverses impaired Ca 2+ handling and associated left ventricular dysfunction in pressure overload-induced cardiac hypertrophy. Cell Calcium 2017; 67:81-90. [PMID: 29029794 DOI: 10.1016/j.ceca.2017.09.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/24/2017] [Accepted: 09/09/2017] [Indexed: 10/18/2022]
|
39
|
Abstract
Myocardial injury, mechanical stress, neurohormonal activation, inflammation, and/or aging all lead to cardiac remodeling, which is responsible for cardiac dysfunction and arrhythmogenesis. Of the key histological components of cardiac remodeling, fibrosis either in the form of interstitial, patchy, or dense scars, constitutes a key histological substrate of arrhythmias. Here we discuss current research findings focusing on the role of fibrosis, in arrhythmogenesis. Numerous studies have convincingly shown that patchy or interstitial fibrosis interferes with myocardial electrophysiology by slowing down action potential propagation, initiating reentry, promoting after-depolarizations, and increasing ectopic automaticity. Meanwhile, there has been increasing appreciation of direct involvement of myofibroblasts, the activated form of fibroblasts, in arrhythmogenesis. Myofibroblasts undergo phenotypic changes with expression of gap-junctions and ion channels thereby forming direct electrical coupling with cardiomyocytes, which potentially results in profound disturbances of electrophysiology. There is strong evidence that systemic and regional inflammatory processes contribute to fibrogenesis (i.e., structural remodeling) and dysfunction of ion channels and Ca2+ homeostasis (i.e., electrical remodeling). Recognizing the pivotal role of fibrosis in the arrhythmogenesis has promoted clinical research on characterizing fibrosis by means of cardiac imaging or fibrosis biomarkers for clinical stratification of patients at higher risk of lethal arrhythmia, as well as preclinical research on the development of antifibrotic therapies. At the end of this review, we discuss remaining key questions in this area and propose new research approaches. © 2017 American Physiological Society. Compr Physiol 7:1009-1049, 2017.
Collapse
Affiliation(s)
- My-Nhan Nguyen
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Helen Kiriazis
- Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Xiao-Ming Gao
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| | - Xiao-Jun Du
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Central Clinical School, Monash University, Melbourne, Australia
| |
Collapse
|
40
|
Zhou Q, Wei SS, Wang H, Wang Q, Li W, Li G, Hou JW, Chen XM, Chen J, Xu WP, Li YG, Wang YP. Crucial Role of ROCK2-Mediated Phosphorylation and Upregulation of FHOD3 in the Pathogenesis of Angiotensin II-Induced Cardiac Hypertrophy. Hypertension 2017; 69:1070-1083. [PMID: 28438902 DOI: 10.1161/hypertensionaha.116.08662] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/12/2016] [Accepted: 03/21/2017] [Indexed: 01/01/2023]
Abstract
Cardiac hypertrophy is characterized by increased myofibrillogenesis. Angiotensin II (Ang-II) is an essential mediator of the pressure overload-induced cardiac hypertrophy in part through RhoA/ROCK (small GTPase/Rho-associated coiled-coil containing protein kinase) pathway. FHOD3 (formin homology 2 domain containing 3), a cardiac-restricted member of diaphanous-related formins, is crucial in regulating myofibrillogenesis in cardiomyocytes. FHOD3 maintains inactive through autoinhibition by an intramolecular interaction between its C- and N-terminal domains. Phosphorylation of the 3 highly conserved residues (1406S, 1412S, and 1416T) within the C terminus (CT) of FHOD3 by ROCK1 is sufficient for its activation. However, it is unclear whether ROCK-mediated FHOD3 activation plays a role in the pathogenesis of Ang-II-induced cardiac hypertrophy. In this study, we detected increases in FHOD3 expression and phosphorylation in cardiomyocytes from Ang-II-induced rat cardiac hypertrophy models. Valsartan attenuated such increases. In cultured neonate rat cardiomyocytes, overexpression of phosphor-mimetic mutant FHOD3-DDD, but not wild-type FHOD3, resulted in myofibrillogenesis and cardiomyocyte hypertrophy. Expression of a phosphor-resistant mutant FHOD3-AAA completely abolished myofibrillogenesis and attenuated Ang-II-induced cardiomyocyte hypertrophy. Pretreatment of neonate rat cardiomyocytes with ROCK inhibitor Y27632 reduced Ang-II-induced FHOD3 activation and upregulation, suggesting the involvement of ROCK activities. Silencing of ROCK2, but not ROCK1, in neonate rat cardiomyocytes, significantly lessened Ang-II-induced cardiomyocyte hypertrophy. ROCK2 can directly phosphorylate FHOD3 at both 1412S and 1416T in vitro and is more potent than ROCK1. Both kinases failed to phosphorylate 1406S. Coexpression of FHOD3 with constitutively active ROCK2 induced more stress fiber formation than that with constitutively active ROCK1. Collectively, our results demonstrated the importance of ROCK2 regulated FHOD3 expression and activation in Ang-II-induced myofibrillogenesis, thus provided a novel mechanism for the pathogenesis of Ang-II-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Qing Zhou
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Si-Si Wei
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Hong Wang
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Qian Wang
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Wei Li
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Gang Li
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Jian-Wen Hou
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Xiao-Meng Chen
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Jie Chen
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Wei-Ping Xu
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China
| | - Yi-Gang Li
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China.
| | - Yue-Peng Wang
- From the Molecular Cardiology Research Laboratory, Department of Cardiology (Q.Z., H.W., Q.W., W.L., G.L., J.-W.H., X.-M.C., J.C., W.-P.X., Y.-G.L., Y.-P.W.) and Department of Pediatrics (S.-S.W.), Affiliated Xinhua Hospital, Shanghai Jiaotong University (SJTU) School of Medicine, China.
| |
Collapse
|
41
|
Abstract
Cardiac arrhythmias can follow disruption of the normal cellular electrophysiological processes underlying excitable activity and their tissue propagation as coherent wavefronts from the primary sinoatrial node pacemaker, through the atria, conducting structures and ventricular myocardium. These physiological events are driven by interacting, voltage-dependent, processes of activation, inactivation, and recovery in the ion channels present in cardiomyocyte membranes. Generation and conduction of these events are further modulated by intracellular Ca2+ homeostasis, and metabolic and structural change. This review describes experimental studies on murine models for known clinical arrhythmic conditions in which these mechanisms were modified by genetic, physiological, or pharmacological manipulation. These exemplars yielded molecular, physiological, and structural phenotypes often directly translatable to their corresponding clinical conditions, which could be investigated at the molecular, cellular, tissue, organ, and whole animal levels. Arrhythmogenesis could be explored during normal pacing activity, regular stimulation, following imposed extra-stimuli, or during progressively incremented steady pacing frequencies. Arrhythmic substrate was identified with temporal and spatial functional heterogeneities predisposing to reentrant excitation phenomena. These could arise from abnormalities in cardiac pacing function, tissue electrical connectivity, and cellular excitation and recovery. Triggering events during or following recovery from action potential excitation could thereby lead to sustained arrhythmia. These surface membrane processes were modified by alterations in cellular Ca2+ homeostasis and energetics, as well as cellular and tissue structural change. Study of murine systems thus offers major insights into both our understanding of normal cardiac activity and its propagation, and their relationship to mechanisms generating clinical arrhythmias.
Collapse
Affiliation(s)
- Christopher L-H Huang
- Physiological Laboratory and the Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
42
|
Signaling Pathways in Cardiac Myocyte Apoptosis. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9583268. [PMID: 28101515 PMCID: PMC5215135 DOI: 10.1155/2016/9583268] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/20/2016] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases, the number 1 cause of death worldwide, are frequently associated with apoptotic death of cardiac myocytes. Since cardiomyocyte apoptosis is a highly regulated process, pharmacological intervention of apoptosis pathways may represent a promising therapeutic strategy for a number of cardiovascular diseases and disorders including myocardial infarction, ischemia/reperfusion injury, chemotherapy cardiotoxicity, and end-stage heart failure. Despite rapid growth of our knowledge in apoptosis signaling pathways, a clinically applicable treatment targeting this cellular process is currently unavailable. To help identify potential innovative directions for future research, it is necessary to have a full understanding of the apoptotic pathways currently known to be functional in cardiac myocytes. Here, we summarize recent progress in the regulation of cardiomyocyte apoptosis by multiple signaling molecules and pathways, with a focus on the involvement of these pathways in the pathogenesis of heart disease. In addition, we provide an update regarding bench to bedside translation of this knowledge and discuss unanswered questions that need further investigation.
Collapse
|
43
|
Li Z, Shen Z, Du L, He J, Chen S, Zhang J, Luan Y, Fu G. Fn14 is regulated via the RhoA pathway and mediates nuclear factor-kappaB activation by Angiotensin II. Am J Transl Res 2016; 8:5386-5398. [PMID: 28078010 PMCID: PMC5209490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/06/2016] [Indexed: 06/06/2023]
Abstract
Angiotesin II (Ang II) plays an important role in cardiac remodeling. Fibroblast growth factor inducible-14 (Fn14) is the smallest member of the tumor necrosis factor superfamily of receptors. Currently, little is known about the functional role of Fn14 in the heart. Chiefly, we observe the up-regulation of extracellular matrix in in vivo model. We therefore assess the expression and regulation of Fn14 in cardiomyocytes and in vivo models induced by Ang II. In order to study the regulation of Fn14, cardiac remodeling was established in rats and neonatal cardiomyocytes were used in in vitro model. As well, Ang II is able to strongly induce Fn14 expression in in vivo and in vitro models. Fn14 is mediated via RhoA pathways, since siRNA against RhoA prevented the expression of Fn14 in cardiomyocytes. Pretreatment of cardiomyoctes with siRNA against NF-κB and IκBα also decreased Fn14 expression induced by Ang II. We here describe for the first time Ang II regulation of Fn14 in in vivo and in vitro models via RhoA, NF-κB and NF-κB driven gene signaling pathway. In conclusion, Fn14 may be important in regulating the process of cardiac remodeling induced by Ang II.
Collapse
Affiliation(s)
- Zhengwei Li
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Lailing Du
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Jialin He
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Shengyu Chen
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Jiefang Zhang
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Yi Luan
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University No. 3 East Qingchun Road, Hangzhou 310016, Zhejiang Province, PR China
| |
Collapse
|
44
|
Liu LJ, Yao FJ, Lu GH, Xu CG, Xu Z, Tang K, Cheng YJ, Gao XR, Wu SH. The Role of the Rho/ROCK Pathway in Ang II and TGF-β1-Induced Atrial Remodeling. PLoS One 2016; 11:e0161625. [PMID: 27611832 PMCID: PMC5017578 DOI: 10.1371/journal.pone.0161625] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 08/09/2016] [Indexed: 02/05/2023] Open
Abstract
Objectives To study the role of the Rho/ROCK pathway in Ang II and TGF-β1-induced atrial remodeling. Methods and Results A canine atrial fibrillation (AF) model was established by rapid atrial pacing (RAP) of the left atrium. The roles of TGF-β1, the RhoA/ROCK signaling pathway and connective tissue growth factor (CTGF) in atrial remodeling were studied via both in vitro and in vivo experiments. Each of the dogs that received RAP developed persistent AF within 4 weeks. The mRNA expression levels of TGF-β1 (1.32±0.38), Collagen-I(1.33±0.91), CTGF(5.83±3.71), RhoA(1.23±0.57) and ROCK-1 (1.02±0.27) in the left atrium were significantly increased following 4 weeks of RAP. Angiotensin II (Ang II) induced the proliferation of atrial fibroblasts and up-regulated the expression of both CTGF and ROCK-1 in a dose-dependent manner. Simvastatin and Y27632 reversed Ang II-induced CFs proliferation, as well as ROCK-1(0.89±0.05 and 1.27±0.03, respectively) and CTGF (0.87±0.04 and 0.91±0.02, respectively) expression. The expression mRNA of ROCK-1(1.74±0.13) and CTGF (2.28±0.11) can upregulated by TGF-β1, and down-regulated by Simvastatin (1.22±0.03 vs 2.27±0.11), Y27632 (1.01±0.04 vs 1.64±0.03), Los (1.04±0.11 vs 1.26±0.05), respectively. Losartan and Simvastatin attenuated the effects of TGF-β1, inhibited RhoA activity as opposed to RhoA protein expression. Y27632 had no effect on either the expression or the activity of RhoA. Conclusions The increased expression of profibrotic factors (CTGF, ROCK1 and Smad2/3) played an important role in our RAP-induced AF model. Increased atrial profibrotic factors involve the activation of either the TGF-β1/RhoA/ROCK-1 or the TGF-β1/Smad2/3 signaling pathway.
Collapse
Affiliation(s)
- Li-Juan Liu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Feng-Juan Yao
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gui-Hua Lu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cheng-Gui Xu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhe Xu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kai Tang
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yun-Jiu Cheng
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiu-Ren Gao
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (SW); (XG)
| | - Su-Hua Wu
- Department of Cardiology and Department of Ultrasonography (Feng-Juan Yao), the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- * E-mail: (SW); (XG)
| |
Collapse
|
45
|
Hanovice NJ, McMains E, Gross JM. A GAL4-inducible transgenic tool kit for the in vivo modulation of Rho GTPase activity in zebrafish. Dev Dyn 2016; 245:844-53. [PMID: 27105927 PMCID: PMC4946996 DOI: 10.1002/dvdy.24412] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 03/21/2016] [Accepted: 03/29/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Rho GTPases are small monomeric G-proteins that play key roles in many cellular processes. Due to Rho GTPases' widespread expression and broad functions, analyses of their function during late development require tissue-specific modulation of activity. The GAL4/UAS system provides an excellent tool for investigating the function of Rho GTPases in vivo. With this in mind, we created a transgenic tool kit enabling spatial and temporal modulation of Rho GTPase activity in zebrafish. RESULTS Transgenic constructs were assembled driving dominant-negative, constitutively active, and wild-type versions of Cdc42, RhoA, and Rac1 under 10XUAS control. The self-cleaving viral peptide F2A was utilized to allow bicistronic expression of a fluorescent reporter and Rho GTPase. Global heat shock of hsp70l:gal4(+) transgenic embryos confirmed GAL4-specific construct expression. Western blot analysis indicated myc-tagged Rho GTPases were expressed only in the presence of GAL4. Construct expression was confined to proper cells when combined with pou4f3:gal4 or ptf1a:gal4. Finally, transgene expression resulted in reproducible defects in lens formation, indicating that the transgenes are functional in vivo. CONCLUSIONS We generated and validated 10 transgenic lines, creating a versatile tool kit for the temporal-spatial modulation of Cdc42, RhoA, and Rac1 activity in vivo. These lines will enable systematic analysis of Rho GTPase function in any tissue of interest. Developmental Dynamics 245:844-853, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nicholas J Hanovice
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Emily McMains
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
| | - Jeffrey M Gross
- Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas
- Department of Ophthalmology, Louis J. Fox Center for Vision Restoration, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
46
|
Oligophrenin1 protects mice against myocardial ischemia and reperfusion injury by modulating inflammation and myocardial apoptosis. Cell Signal 2016; 28:967-78. [DOI: 10.1016/j.cellsig.2016.04.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 04/11/2016] [Accepted: 04/21/2016] [Indexed: 12/12/2022]
|
47
|
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
|
48
|
Kelder TP, Vicente-Steijn R, Poelmann RE, Mummery CL, DeRuiter MC, Jongbloed MRM. The avian embryo to study development of the cardiac conduction system. Differentiation 2016; 91:90-103. [PMID: 26856662 DOI: 10.1016/j.diff.2016.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 01/26/2016] [Indexed: 11/17/2022]
Abstract
The avian embryo has long been a popular model system in developmental biology. The easy accessibility of the embryo makes it particularly suitable for in ovo microsurgery and manipulation. Re-incubation of the embryo allows long-term follow-up of these procedures. The current review focuses on the variety of techniques available to study development of the cardiac conduction system in avian embryos. Based on the large amount of relevant data arising from experiments in avian embryos, we conclude that the avian embryo has and will continue to be a powerful model system to study development in general and the developing cardiac conduction system in particular.
Collapse
Affiliation(s)
- Tim P Kelder
- Anatomy & Embryology, Leiden University Medical Center, The Netherlands
| | - Rebecca Vicente-Steijn
- Anatomy & Embryology, Leiden University Medical Center, The Netherlands; Cardiology, Leiden University Medical Center, The Netherlands; ICIN Netherlands Heart Institute, Utrecht, The Netherlands
| | - Robert E Poelmann
- Cardiology, Leiden University Medical Center, The Netherlands; Integrative Zoology, Institute Biology, University Leiden, The Netherlands
| | | | - Marco C DeRuiter
- Anatomy & Embryology, Leiden University Medical Center, The Netherlands
| | - Monique R M Jongbloed
- Anatomy & Embryology, Leiden University Medical Center, The Netherlands; Cardiology, Leiden University Medical Center, The Netherlands.
| |
Collapse
|
49
|
Pellman J, Sheikh F. Atrial fibrillation: mechanisms, therapeutics, and future directions. Compr Physiol 2016; 5:649-65. [PMID: 25880508 DOI: 10.1002/cphy.c140047] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, affecting 1% to 2% of the general population. It is characterized by rapid and disorganized atrial activation leading to impaired atrial function, which can be diagnosed on an EKG by lack of a P-wave and irregular QRS complexes. AF is associated with increased morbidity and mortality and is a risk factor for embolic stroke and worsening heart failure. Current research on AF support and explore the hypothesis that initiation and maintenance of AF require pathophysiological remodeling of the atria, either specifically as in lone AF or secondary to other heart disease as in heart failure-associated AF. Remodeling in AF can be grouped into three categories that include: (i) electrical remodeling, which includes modulation of L-type Ca(2+) current, various K(+) currents and gap junction function; (ii) structural remodeling, which includes changes in tissues properties, size, and ultrastructure; and (iii) autonomic remodeling, including altered sympathovagal activity and hyperinnervation. Electrical, structural, and autonomic remodeling all contribute to creating an AF-prone substrate which is able to produce AF-associated electrical phenomena including a rapidly firing focus, complex multiple reentrant circuit or rotors. Although various remodeling events occur in AF, current AF therapies focus on ventricular rate and rhythm control strategies using pharmacotherapy and surgical interventions. Recent progress in the field has started to focus on the underlying substrate that drives and maintains AF (termed upstream therapies); however, much work is needed in this area. Here, we review current knowledge of AF mechanisms, therapies, and new areas of investigation.
Collapse
Affiliation(s)
- Jason Pellman
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | |
Collapse
|
50
|
Bernardo BC, Sapra G, Patterson NL, Cemerlang N, Kiriazis H, Ueyama T, Febbraio MA, McMullen JR. Long-Term Overexpression of Hsp70 Does Not Protect against Cardiac Dysfunction and Adverse Remodeling in a MURC Transgenic Mouse Model with Chronic Heart Failure and Atrial Fibrillation. PLoS One 2015; 10:e0145173. [PMID: 26660322 PMCID: PMC4680216 DOI: 10.1371/journal.pone.0145173] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 11/30/2015] [Indexed: 12/19/2022] Open
Abstract
Previous animal studies had shown that increasing heat shock protein 70 (Hsp70) using a transgenic, gene therapy or pharmacological approach provided cardiac protection in models of acute cardiac stress. Furthermore, clinical studies had reported associations between Hsp70 levels and protection against atrial fibrillation (AF). AF is the most common cardiac arrhythmia presenting in cardiology clinics and is associated with increased rates of heart failure and stroke. Improved therapies for AF and heart failure are urgently required. Despite promising observations in animal studies which targeted Hsp70, we recently reported that increasing Hsp70 was unable to attenuate cardiac dysfunction and pathology in a mouse model which develops heart failure and intermittent AF. Given our somewhat unexpected finding and the extensive literature suggesting Hsp70 provides cardiac protection, it was considered important to assess whether Hsp70 could provide protection in another mouse model of heart failure and AF. The aim of the current study was to determine whether increasing Hsp70 could attenuate adverse cardiac remodeling, cardiac dysfunction and episodes of arrhythmia in a mouse model of heart failure and AF due to overexpression of Muscle-Restricted Coiled-Coil (MURC). Cardiac function and pathology were assessed in mice at approximately 12 months of age. We report here, that chronic overexpression of Hsp70 was unable to provide protection against cardiac dysfunction, conduction abnormalities, fibrosis or characteristic molecular markers of the failing heart. In summary, elevated Hsp70 may provide protection in acute cardiac stress settings, but appears insufficient to protect the heart under chronic cardiac disease conditions.
Collapse
Affiliation(s)
| | - Geeta Sapra
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | | | - Nelly Cemerlang
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Tomomi Ueyama
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 602–8566, Japan
| | - Mark A. Febbraio
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
- Garvan Institute of Medical Research, Darlinghurst, 2010, Australia
| | - Julie R. McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
- * E-mail:
| |
Collapse
|