1
|
Lopez-Pier MA, Marino VA, Vazquez-Loreto AC, Skaria RS, Cannon DK, Hoyer-Kimura CH, Solomon AE, Lipovka Y, Doubleday K, Pier M, Chu M, Mayfield R, Behunin SM, Hu T, Langlais PR, McKinsey TA, Konhilas JP. Myocardial transcriptomic and proteomic landscapes across the menopausal continuum in a murine model of chemically induced accelerated ovarian failure. Physiol Genomics 2025; 57:409-430. [PMID: 40266891 DOI: 10.1152/physiolgenomics.00133.2024] [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: 08/27/2024] [Revised: 09/17/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Risk of cardiovascular disease (CVD) in women increases with the menopausal transition. Using a chemical model (4-vinylcyclohexene diepoxide; VCD) of accelerated ovarian failure, we previously demonstrated that menopausal females are more susceptible to CVD compared with peri- or premenopausal females like humans. Yet, the cellular and molecular mechanisms underlying this shift in CVD susceptibility across the pre- to peri- to menopause continuum remain understudied. In this work using the VCD mouse model, we phenotyped cellular and molecular signatures from hearts at each hormonally distinct stage that included transcriptomic, proteomic, and cell biological analyses. The transcriptional profile of premenopausal hearts clustered separately from perimenopausal and menopausal hearts, which clustered more similarly. Proteomics also revealed hormonal clustering; perimenopausal hearts grouped more closely with premenopausal than menopausal hearts. Both proteomes and transcriptomes showed similar trends in genes associated with atherothrombosis, contractility, and impaired nuclear signaling between pre-, peri-, and menopausal murine hearts. Further analysis of posttranslational modifications (PTMs) showed hormone-dependent shifts in the phosphoproteome and acetylome. To further interrogate these findings, we triggered pathological remodeling using angiotensin II (Ang II). Phosphorylation of AMP-activated protein kinase (AMPK) signaling and histone deacetylase (HDAC) activity were found to be dependent on hormonal status and Ang II stimulation. Finally, knockdown of anti-inflammatory regulatory T cells (Treg) exacerbated Ang II-dependent fibrosis implicating HDAC-mediated epigenetic suppression of Treg activity. Taken together, we demonstrated unique cellular and molecular profiles underlying the cardiac phenotype of pre-, peri-, and menopausal mice supporting the necessity to study CVD in females across the hormonal transition.NEW & NOTEWORTHY Cycling and perimenopausal females are protected from cardiovascular disease (CVD) whereas menopausal females are more susceptible to CVD and other pathological sequalae. The cellular and molecular mechanisms underlying loss of CVD protection across the pre- to peri- to menopause transition remain understudied. Using the murine 4-vinylcyclohexene diepoxide (VCD) model of menopause we highlight cellular and molecular signatures from hearts at each hormonally distinct stage that included transcriptomic, proteomic, and cell biological analyses.
Collapse
Affiliation(s)
- Marissa A Lopez-Pier
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, United States
| | - Vito A Marino
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | | | - Rinku S Skaria
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- College of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Danielle K Cannon
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
| | | | - Alice E Solomon
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, United States
| | - Yulia Lipovka
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
- Department of Chemistry-Biology, University of Sonora, Hermosillo, Mexico
| | - Kevin Doubleday
- College of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Maricela Pier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, United States
| | - Meinsung Chu
- Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, United States
| | - Rachel Mayfield
- Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, United States
| | - Samantha M Behunin
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| | - Tianjing Hu
- Division of Cardiology and Consortium for Fibrosis Research & Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Paul R Langlais
- Department of Endocrinology, University of Arizona, Tucson, Arizona, United States
- College of Medicine, University of Arizona, Tucson, Arizona, United States
| | - Timothy A McKinsey
- Division of Cardiology and Consortium for Fibrosis Research & Translation, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - John P Konhilas
- Department of Physiology, University of Arizona, Tucson, Arizona, United States
- Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States
| |
Collapse
|
2
|
Wang Y, Zhao M, Liu X, Xu B, Reddy GR, Jovanovic A, Wang Q, Zhu C, Xu H, Bayne EF, Xiang W, Tilley DG, Ge Y, Tate CG, Feil R, Chiu JC, Bers DM, Xiang YK. Carvedilol Activates a Myofilament Signaling Circuitry to Restore Cardiac Contractility in Heart Failure. JACC Basic Transl Sci 2024; 9:982-1001. [PMID: 39297139 PMCID: PMC11405995 DOI: 10.1016/j.jacbts.2024.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 09/21/2024]
Abstract
Phosphorylation of myofilament proteins critically regulates beat-to-beat cardiac contraction and is typically altered in heart failure (HF). β-Adrenergic activation induces phosphorylation in numerous substrates at the myofilament. Nevertheless, how cardiac β-adrenoceptors (βARs) signal to the myofilament in healthy and diseased hearts remains poorly understood. The aim of this study was to uncover the spatiotemporal regulation of local βAR signaling at the myofilament and thus identify a potential therapeutic target for HF. Phosphoproteomic analysis of substrate phosphorylation induced by different βAR ligands in mouse hearts was performed. Genetically encoded biosensors were used to characterize cyclic adenosine and guanosine monophosphate signaling and the impacts on excitation-contraction coupling induced by β1AR ligands at both the cardiomyocyte and whole-heart levels. Myofilament signaling circuitry was identified, including protein kinase G1 (PKG1)-dependent phosphorylation of myosin light chain kinase, myosin phosphatase target subunit 1, and myosin light chain at the myofilaments. The increased phosphorylation of myosin light chain enhances cardiac contractility, with a minimal increase in calcium (Ca2+) cycling. This myofilament signaling paradigm is promoted by carvedilol-induced β1AR-nitric oxide synthetase 3 (NOS3)-dependent cyclic guanosine monophosphate signaling, drawing a parallel to the β1AR-cyclic adenosine monophosphate-protein kinase A pathway. In patients with HF and a mouse HF model of myocardial infarction, increasing expression and association of NOS3 with β1AR were observed. Stimulating β1AR-NOS3-PKG1 signaling increased cardiac contraction in the mouse HF model. This research has characterized myofilament β1AR-PKG1-dependent signaling circuitry to increase phosphorylation of myosin light chain and enhance cardiac contractility, with a minimal increase in Ca2+ cycling. The present findings raise the possibility of targeting this myofilament signaling circuitry for treatment of patients with HF.
Collapse
Affiliation(s)
- Ying Wang
- Department of Pharmacology, University of California-Davis, Davis, California, USA
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Meimi Zhao
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Xianhui Liu
- Department of Entomology and Nematology, University of California-Davis, Davis, California, USA
| | - Bing Xu
- Department of Pharmacology, University of California-Davis, Davis, California, USA
- VA Northern California Health Care System, Mather, California, USA
| | - Gopireddy R. Reddy
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Aleksandra Jovanovic
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Qingtong Wang
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Chaoqun Zhu
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Heli Xu
- Department of Cardiovascular Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Elizabeth F. Bayne
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Wenjing Xiang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Douglas G. Tilley
- Department of Cardiovascular Sciences, Temple University, Philadelphia, Pennsylvania, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | | | - Robert Feil
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Joanna C. Chiu
- Department of Entomology and Nematology, University of California-Davis, Davis, California, USA
| | - Donald M. Bers
- Department of Pharmacology, University of California-Davis, Davis, California, USA
| | - Yang K. Xiang
- Department of Pharmacology, University of California-Davis, Davis, California, USA
- VA Northern California Health Care System, Mather, California, USA
| |
Collapse
|
3
|
Tsisanova E, Nobles M, Sebastian S, Ng KE, Thomas A, Weinstein LS, Munroe PB, Tinker A. The ric-8b protein (resistance to inhibitors of cholinesterase 8b) is key to preserving contractile function in the adult heart. J Biol Chem 2024; 300:107470. [PMID: 38879012 PMCID: PMC11277413 DOI: 10.1016/j.jbc.2024.107470] [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: 02/12/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 07/09/2024] Open
Abstract
Resistance to inhibitors of cholinesterases (ric-8 proteins) are involved in modulating G-protein function, but little is known of their potential physiological importance in the heart. In the present study, we assessed the role of resistance to inhibitors of cholinesterase 8b (Ric-8b) in determining cardiac contractile function. We developed a murine model in which it was possible to conditionally delete ric-8b in cardiac tissue in the adult animal after the addition of tamoxifen. Deletion of ric-8b led to severely reduced contractility as measured using echocardiography days after administration of tamoxifen. Histological analysis of the ventricular tissue showed highly variable myocyte size, prominent fibrosis, and an increase in cellular apoptosis. RNA sequencing revealed transcriptional remodeling in response to cardiac ric-8b deletion involving the extracellular matrix and inflammation. Phosphoproteomic analysis revealed substantial downregulation of phosphopeptides related to myosin light chain 2. At the cellular level, the deletion of ric-8b led to loss of activation of the L-type calcium channel through the β-adrenergic pathways. Using fluorescence resonance energy transfer-based assays, we showed ric-8b protein selectively interacts with the stimulatory G-protein, Gαs. We explored if deletion of Gnas (the gene encoding Gαs) in cardiac tissue using a similar approach in the mouse led to an equivalent phenotype. The conditional deletion of the Gαs gene in the ventricle led to comparable effects on contractile function and cardiac histology. We conclude that ric-8b is essential to preserve cardiac contractile function likely through an interaction with the stimulatory G-protein and downstream phosphorylation of myosin light chain 2.
Collapse
Affiliation(s)
- Elena Tsisanova
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Muriel Nobles
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sonia Sebastian
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Keat-Eng Ng
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Alison Thomas
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Patricia B Munroe
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrew Tinker
- Department of Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
4
|
Chen Z, Pan Z, Huang C, Zhu X, Li N, Huynh H, Xu J, Huang L, Vaz FM, Liu J, Han Z, Ouyang K. Cardiac lipidomic profiles in mice undergo changes from fetus to adult. Life Sci 2024; 341:122484. [PMID: 38311219 DOI: 10.1016/j.lfs.2024.122484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/20/2024] [Accepted: 01/30/2024] [Indexed: 02/09/2024]
Abstract
AIMS Lipids are essential cellular components with many important biological functions. Disturbed lipid biosynthesis and metabolism has been shown to cause cardiac developmental abnormality and cardiovascular diseases. In this study, we aimed to investigate the composition and the molecular profiles of lipids in mammalian hearts between embryonic and adult stages and uncover the underlying links between lipid and cardiac development and maturation. MATERIALS AND METHODS We collected mouse hearts at the embryonic day 11.5 (E11.5), E15.5, and the age of 2 months, 4 months and 10 months, and performed lipidomic analysis to determine the changes of the composition, molecular species, and relative abundance of cardiac lipids between embryonic and adult stages. Additionally, we also performed the electronic microscopy and RNA sequencing in both embryonic and adult mouse hearts. KEY FINDINGS The relative abundances of certain phospholipids and sphingolipids including cardiolipin, phosphatidylglycerol, phosphatidylethanolamine, and ceramide, are different between embryonic and adult hearts. Such lipidomic changes are accompanied with increased densities of mitochondrial membranes and elevated expression of genes related to mitochondrial formation in adult mouse hearts. We also analyzed individual molecular species of phospholipids and sphingolipids, and revealed that the composition and distribution of lipid molecular species in hearts also change with development. SIGNIFICANCE Our study provides not only a lipidomic view of mammalian hearts when developing from the embryonic to the adult stage, but also a potential pool of lipid indicators for cardiac cell development and maturation.
Collapse
Affiliation(s)
- Ze'e Chen
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Zhixiang Pan
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Can Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Xiangbin Zhu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Na Li
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Helen Huynh
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, United States of America
| | - Junjie Xu
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Lei Huang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China
| | - Frédéric M Vaz
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Departments of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, the Netherlands
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, Shenzhen, China
| | - Zhen Han
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong Province, China.
| |
Collapse
|
5
|
Burnham HV, Cizauskas HE, Barefield DY. Fine tuning contractility: atrial sarcomere function in health and disease. Am J Physiol Heart Circ Physiol 2024; 326:H568-H583. [PMID: 38156887 PMCID: PMC11221815 DOI: 10.1152/ajpheart.00252.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The molecular mechanisms of sarcomere proteins underlie the contractile function of the heart. Although our understanding of the sarcomere has grown tremendously, the focus has been on ventricular sarcomere isoforms due to the critical role of the ventricle in health and disease. However, atrial-specific or -enriched myofilament protein isoforms, as well as isoforms that become expressed in disease, provide insight into ways this complex molecular machine is fine-tuned. Here, we explore how atrial-enriched sarcomere protein composition modulates contractile function to fulfill the physiological requirements of atrial function. We review how atrial dysfunction negatively affects the ventricle and the many cardiovascular diseases that have atrial dysfunction as a comorbidity. We also cover the pathophysiology of mutations in atrial-enriched contractile proteins and how they can cause primary atrial myopathies. Finally, we explore what is known about contractile function in various forms of atrial fibrillation. The differences in atrial function in health and disease underscore the importance of better studying atrial contractility, especially as therapeutics currently in development to modulate cardiac contractility may have different effects on atrial sarcomere function.
Collapse
Affiliation(s)
- Hope V Burnham
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, United States
| | - Hannah E Cizauskas
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, United States
| | - David Y Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, Illinois, United States
| |
Collapse
|
6
|
Lee E, May H, Kazmierczak K, Liang J, Nguyen N, Hill JA, Gillette TG, Szczesna-Cordary D, Chang AN. The MYPT2-regulated striated muscle-specific myosin light chain phosphatase limits cardiac myosin phosphorylation in vivo. J Biol Chem 2024; 300:105652. [PMID: 38224947 PMCID: PMC10851227 DOI: 10.1016/j.jbc.2024.105652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/17/2024] Open
Abstract
The physiological importance of cardiac myosin regulatory light chain (RLC) phosphorylation by its dedicated cardiac myosin light chain kinase has been established in both humans and mice. Constitutive RLC-phosphorylation, regulated by the balanced activities of cardiac myosin light chain kinase and myosin light chain phosphatase (MLCP), is fundamental to the biochemical and physiological properties of myofilaments. However, limited information is available on cardiac MLCP. In this study, we hypothesized that the striated muscle-specific MLCP regulatory subunit, MYPT2, targets the phosphatase catalytic subunit to cardiac myosin, contributing to the maintenance of cardiac function in vivo through the regulation of RLC-phosphorylation. To test this hypothesis, we generated a floxed-PPP1R12B mouse model crossed with a cardiac-specific Mer-Cre-Mer to conditionally ablate MYPT2 in adult cardiomyocytes. Immunofluorescence microscopy using the gene-ablated tissue as a control confirmed the localization of MYPT2 to regions where it overlaps with a subset of RLC. Biochemical analysis revealed an increase in RLC-phosphorylation in vivo. The loss of MYPT2 demonstrated significant protection against pressure overload-induced hypertrophy, as evidenced by heart weight, qPCR of hypertrophy-associated genes, measurements of myocyte diameters, and expression of β-MHC protein. Furthermore, mantATP chase assays revealed an increased ratio of myosin heads distributed to the interfilament space in MYPT2-ablated heart muscle fibers, confirming that RLC-phosphorylation regulated by MLCP, enhances cardiac performance in vivo. Our findings establish MYPT2 as the regulatory subunit of cardiac MLCP, distinct from the ubiquitously expressed canonical smooth muscle MLCP. Targeting MYPT2 to increase cardiac RLC-phosphorylation in vivo may improve baseline cardiac performance, thereby attenuating pathological hypertrophy.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Herman May
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Nhu Nguyen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph A Hill
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Thomas G Gillette
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Audrey N Chang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Pak Center for Mineral Metabolism and Clinical Research, UTSW Medical Center, Dallas, Texas, USA.
| |
Collapse
|
7
|
Tanner BCW. Design Principles and Benefits of Spatially Explicit Models of Myofilament Function. Methods Mol Biol 2024; 2735:43-62. [PMID: 38038843 DOI: 10.1007/978-1-0716-3527-8_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Spatially explicit models of muscle contraction include fine-scale details about the spatial, kinetic, and/or mechanical properties of the biological processes being represented within the model network. Over the past 25 years, this has primarily consisted of a set of mathematical and computational algorithms representing myosin cross-bridge activity, Ca2+-activation of contraction, and ensemble force production within a half-sarcomere representation of the myofilament network. Herein we discuss basic design principles associated with creating spatially explicit models of myofilament function, as well as model assumptions underlying model development. A brief overview of computational approaches is introduced. Opportunities for new model directions that could investigate coupled regulatory pathways between the thick-filament and thin-filaments are also presented. Given the modular design and flexibility associated with spatially explicit models, we highlight some advantages of this approach compared to other model formulations.
Collapse
Affiliation(s)
- Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA.
| |
Collapse
|
8
|
Tsukamoto O, Takashima S. Response by Tsukamoto and Takashima to Letter Regarding Article, "Restoration of Cardiac Myosin Light Chain Kinase Ameliorates Systolic Dysfunction by Reducing Super-Relaxed Myosin". Circulation 2023; 148:2074-2075. [PMID: 38109341 DOI: 10.1161/circulationaha.123.066938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Affiliation(s)
- Osamu Tsukamoto
- Department of Biochemistry, Hyogo Medical University, Nishinomiya, Japan (O.T.)
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Japan (O.T., S.T.)
| | - Seiji Takashima
- Department of Biochemistry, Hyogo Medical University, Nishinomiya, Japan (O.T.)
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Japan (O.T., S.T.)
| |
Collapse
|
9
|
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
|
10
|
Holmes JB, Lemieux ME, Stelzer JE. Torsional and strain dysfunction precede overt heart failure in a mouse model of dilated cardiomyopathy pathogenesis. Am J Physiol Heart Circ Physiol 2023; 325:H449-H467. [PMID: 37417875 PMCID: PMC10538988 DOI: 10.1152/ajpheart.00130.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 05/24/2023] [Accepted: 06/28/2023] [Indexed: 07/08/2023]
Abstract
Detailed assessments of whole heart mechanics are crucial for understanding the consequences of sarcomere perturbations that lead to cardiomyopathy in mice. Echocardiography offers an accessible and cost-effective method of obtaining metrics of cardiac function, but the most routine imaging and analysis protocols might not identify subtle mechanical deficiencies. This study aims to use advanced echocardiography imaging and analysis techniques to identify previously unappreciated mechanical deficiencies in a mouse model of dilated cardiomyopathy (DCM) before the onset of overt systolic heart failure (HF). Mice lacking muscle LIM protein expression (MLP-/-) were used to model DCM-linked HF pathogenesis. Left ventricular (LV) function of MLP-/- and wild-type (WT) controls were studied at 3, 6, and 10 wk of age using conventional and four-dimensional (4-D) echocardiography, followed by speckle-tracking analysis to assess torsional and strain mechanics. Mice were also studied with RNA-seq. Although 3-wk-old MLP-/- mice showed normal LV ejection fraction (LVEF), these mice displayed abnormal torsional and strain mechanics alongside reduced β-adrenergic reserve. Transcriptome analysis showed that these defects preceded most molecular markers of HF. However, these markers became upregulated as MLP-/- mice aged and developed overt systolic dysfunction. These findings indicate that subtle deficiencies in LV mechanics, undetected by LVEF and conventional molecular markers, may act as pathogenic stimuli in DCM-linked HF. Using these analyses in future studies will further help connect in vitro measurements of the sarcomere function to whole heart function.NEW & NOTEWORTHY A detailed study of how perturbations to sarcomere proteins impact whole heart mechanics in mouse models is a major yet challenging step in furthering our understanding of cardiovascular pathophysiology. This study uses advanced echocardiographic imaging and analysis techniques to reveal previously unappreciated subclinical whole heart mechanical defects in a mouse model of cardiomyopathy. In doing so, it offers an accessible set of measurements for future studies to use when connecting sarcomere and whole heart function.
Collapse
Affiliation(s)
- Joshua B Holmes
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| | | | - Julian E Stelzer
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, Ohio, United States
| |
Collapse
|
11
|
Hitsumoto T, Tsukamoto O, Matsuoka K, Li J, Liu L, Kuramoto Y, Higo S, Ogawa S, Fujino N, Yoshida S, Kioka H, Kato H, Hakui H, Saito Y, Okamoto C, Inoue H, Hyejin J, Ueda K, Segawa T, Nishimura S, Asano Y, Asanuma H, Tani A, Imamura R, Komagawa S, Kanai T, Takamura M, Sakata Y, Kitakaze M, Haruta JI, Takashima S. Restoration of Cardiac Myosin Light Chain Kinase Ameliorates Systolic Dysfunction by Reducing Superrelaxed Myosin. Circulation 2023; 147:1902-1918. [PMID: 37128901 PMCID: PMC10270284 DOI: 10.1161/circulationaha.122.062885] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 04/05/2023] [Indexed: 05/03/2023]
Abstract
BACKGROUND Cardiac-specific myosin light chain kinase (cMLCK), encoded by MYLK3, regulates cardiac contractility through phosphorylation of ventricular myosin regulatory light chain. However, the pathophysiological and therapeutic implications of cMLCK in human heart failure remain unclear. We aimed to investigate whether cMLCK dysregulation causes cardiac dysfunction and whether the restoration of cMLCK could be a novel myotropic therapy for systolic heart failure. METHODS We generated the knock-in mice (Mylk3+/fs and Mylk3fs/fs) with a familial dilated cardiomyopathy-associated MYLK3 frameshift mutation (MYLK3+/fs) that had been identified previously by us (c.1951-1G>T; p.P639Vfs*15) and the human induced pluripotent stem cell-derived cardiomyocytes from the carrier of the mutation. We also developed a new small-molecule activator of cMLCK (LEUO-1154). RESULTS Both mice (Mylk3+/fs and Mylk3fs/fs) showed reduced cMLCK expression due to nonsense-mediated messenger RNA decay, reduced MLC2v (ventricular myosin regulatory light chain) phosphorylation in the myocardium, and systolic dysfunction in a cMLCK dose-dependent manner. Consistent with this result, myocardium from the mutant mice showed an increased ratio of cardiac superrelaxation/disordered relaxation states that may contribute to impaired cardiac contractility. The phenotypes observed in the knock-in mice were rescued by cMLCK replenishment through the AAV9_MYLK3 vector. Human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation reduced cMLCK expression by 50% and contractile dysfunction, accompanied by an increased superrelaxation/disordered relaxation ratio. CRISPR-mediated gene correction, or cMLCK replenishment by AAV9_MYLK3 vector, successfully recovered cMLCK expression, the superrelaxation/disordered relaxation ratio, and contractile dysfunction. LEUO-1154 increased human cMLCK activity ≈2-fold in the Vmax for ventricular myosin regulatory light chain phosphorylation without affecting the Km. LEUO-1154 treatment of human induced pluripotent stem cell-derived cardiomyocytes with MYLK3+/fs mutation restored the ventricular myosin regulatory light chain phosphorylation level and superrelaxation/disordered relaxation ratio and improved cardiac contractility without affecting calcium transients, indicating that the cMLCK activator acts as a myotrope. Finally, human myocardium from advanced heart failure with a wide variety of causes had a significantly lower MYLK3/PPP1R12B messenger RNA expression ratio than control hearts, suggesting an altered balance between myosin regulatory light chain kinase and phosphatase in the failing myocardium, irrespective of the causes. CONCLUSIONS cMLCK dysregulation contributes to the development of cardiac systolic dysfunction in humans. Our strategy to restore cMLCK activity could form the basis of a novel myotropic therapy for advanced systolic heart failure.
Collapse
Affiliation(s)
- Tatsuro Hitsumoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Ken Matsuoka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Junjun Li
- Department of Cardiovascular Surgery (J.L., L.L.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Li Liu
- Department of Cardiovascular Surgery (J.L., L.L.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yuki Kuramoto
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Shuichiro Higo
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Shou Ogawa
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Noboru Fujino
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Shohei Yoshida
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Hidetaka Kioka
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Hisakazu Kato
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Hideyuki Hakui
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yuki Saito
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Chisato Okamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Hijiri Inoue
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Jo Hyejin
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Kyoko Ueda
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Takatsugu Segawa
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| | - Shunsuke Nishimura
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | - Yoshihiro Asano
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
- Department of Genomic Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan (Y.A.)
| | - Hiroshi Asanuma
- Department of Internal Medicine, Meiji University of Integrative Medicine, Nantan, Kyoto, Japan (H.A.)
| | - Akiyoshi Tani
- Compound Library Screening Center (A.T.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Riyo Imamura
- Drug Discovery Initiative, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan (R.I.)
| | - Shinsuke Komagawa
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Toshio Kanai
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Masayuki Takamura
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kanazawa University. Kanazawa, Ishikawa, Japan (N.F., S.Y., M.T.)
| | - Yasushi Sakata
- Department of Cardiology (Y.K., S.H., S.O., H. Kioka, HY.H., S.N., Y.A., Y.S.), Osaka University Graduate School of Medicine. Suita, Osaka, Japan
| | | | - Jun-ichi Haruta
- Lead Explorating Units (S.K., T.K., J.-i.H.), Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine/Frontier Biosciences, Suita, Osaka, Japan (T.H., O.T., K.M., H. Kioka, H. Kato, H.H., Y.S., C.O., H.I., J.H., K.U., T.S., S.N., S.T.)
| |
Collapse
|
12
|
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
|
13
|
Turner KL, Morris HS, Awinda PO, Fitzsimons DP, Tanner BCW. RLC phosphorylation amplifies Ca2+ sensitivity of force in myocardium from cMyBP-C knockout mice. J Gen Physiol 2023; 155:213841. [PMID: 36715675 PMCID: PMC9930131 DOI: 10.1085/jgp.202213250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/11/2022] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the leading genetic cause of heart disease. The heart comprises several proteins that work together to properly facilitate force production and pump blood throughout the body. Cardiac myosin binding protein-C (cMyBP-C) is a thick-filament protein, and mutations in cMyBP-C are frequently linked with clinical cases of HCM. Within the sarcomere, the N-terminus of cMyBP-C likely interacts with the myosin regulatory light chain (RLC); RLC is a subunit of myosin located within the myosin neck region that modulates contractile dynamics via its phosphorylation state. Phosphorylation of RLC is thought to influence myosin head position along the thick-filament backbone, making it more favorable to bind the thin filament of actin and facilitate force production. However, little is known about how these two proteins interact. We tested the effects of RLC phosphorylation on Ca2+-regulated contractility using biomechanical assays on skinned papillary muscle strips isolated from cMyBP-C KO mice and WT mice. RLC phosphorylation increased Ca2+ sensitivity of contraction (i.e., pCa50) from 5.80 ± 0.02 to 5.95 ± 0.03 in WT strips, whereas RLC phosphorylation increased Ca2+ sensitivity of contraction from 5.86 ± 0.02 to 6.15 ± 0.03 in cMyBP-C KO strips. These data suggest that the effects of RLC phosphorylation on Ca2+ sensitivity of contraction are amplified when cMyBP-C is absent from the sarcomere. This implies that cMyBP-C and RLC act in concert to regulate contractility in healthy hearts, and mutations to these proteins that lead to HCM (or a loss of phosphorylation with disease progression) may disrupt important interactions between these thick-filament regulatory proteins.
Collapse
Affiliation(s)
- Kyrah L Turner
- School of Molecular Biosciences & Neuroscience, Washington State University , Pullman, WA, USA
| | - Haley S Morris
- School of Molecular Biosciences & Neuroscience, Washington State University , Pullman, WA, USA
| | - Peter O Awinda
- Department of Integrative Physiology & Neuroscience, Washington State University , Pullman, WA, USA
| | - Daniel P Fitzsimons
- Department of Animal, Veterinary and Food Sciences, University of Idaho , Moscow, ID, USA
| | - Bertrand C W Tanner
- Department of Integrative Physiology & Neuroscience, Washington State University , Pullman, WA, USA
| |
Collapse
|
14
|
Hu T, Kalyanaraman H, Pilz RB, Casteel DE. Phosphatase regulatory subunit MYPT2 knock-out partially compensates for the cardiac dysfunction in mice caused by lack of myosin light chain kinase 3. J Biol Chem 2023; 299:104584. [PMID: 36889588 PMCID: PMC10124902 DOI: 10.1016/j.jbc.2023.104584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 02/26/2023] [Accepted: 02/27/2023] [Indexed: 03/08/2023] Open
Abstract
Cardiac contraction is modulated by the phosphorylation state of myosin regulatory light chain 2 (MLC-2v). The level of MLC-2v phosphorylation is dependent on the opposing activities of MLC kinases and phosphatases. The predominant MLC phosphatase found in cardiac myocytes contains Myosin Phosphatase Targeting Subunit 2 (MYPT2). Overexpression of MYPT2 in cardiac myocytes results in a decreased level of MLC phosphorylation, reduced left ventricular contraction and induction of hypertrophy; however, the effect of knocking out MYPT2 on cardiac function is unknown. We obtained heterozygous mice containing a MYPT2 null allele from the Mutant Mouse Resource Center. These mice were produced in a C57BL/6N background which lack MLCK3, the main regulatory light chain kinase in cardiac myocytes. We found that mice null for MYPT2 were viable and had no obvious phenotypic abnormality when compared to wild-type mice. Additionally, we determined that wild-type C57BL/6N mice had a low basal level of MLC-2v phosphorylation which was significantly increased when MYPT2 was absent. At 12-weeks, MYPT2 knock-out mice had smaller hearts and showed down-regulation of genes involved in cardiac remodeling. Using cardiac echo, we found that 24-week-old male MYPT2 knock-out mice had decreased heart size with increased fractional shortening compared to their MYPT2 wild-type littermates. Collectively, these studies highlight the important role that MYPT2 plays in cardiac function in vivo and demonstrate that its deletion can partially compensate for the lack of MLCK3.
Collapse
Affiliation(s)
- Tingfei Hu
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Hema Kalyanaraman
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Renate B Pilz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Darren E Casteel
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093.
| |
Collapse
|
15
|
Schwarz EL, Pegolotti L, Pfaller MR, Marsden AL. Beyond CFD: Emerging methodologies for predictive simulation in cardiovascular health and disease. BIOPHYSICS REVIEWS 2023; 4:011301. [PMID: 36686891 PMCID: PMC9846834 DOI: 10.1063/5.0109400] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 12/12/2022] [Indexed: 01/15/2023]
Abstract
Physics-based computational models of the cardiovascular system are increasingly used to simulate hemodynamics, tissue mechanics, and physiology in evolving healthy and diseased states. While predictive models using computational fluid dynamics (CFD) originated primarily for use in surgical planning, their application now extends well beyond this purpose. In this review, we describe an increasingly wide range of modeling applications aimed at uncovering fundamental mechanisms of disease progression and development, performing model-guided design, and generating testable hypotheses to drive targeted experiments. Increasingly, models are incorporating multiple physical processes spanning a wide range of time and length scales in the heart and vasculature. With these expanded capabilities, clinical adoption of patient-specific modeling in congenital and acquired cardiovascular disease is also increasing, impacting clinical care and treatment decisions in complex congenital heart disease, coronary artery disease, vascular surgery, pulmonary artery disease, and medical device design. In support of these efforts, we discuss recent advances in modeling methodology, which are most impactful when driven by clinical needs. We describe pivotal recent developments in image processing, fluid-structure interaction, modeling under uncertainty, and reduced order modeling to enable simulations in clinically relevant timeframes. In all these areas, we argue that traditional CFD alone is insufficient to tackle increasingly complex clinical and biological problems across scales and systems. Rather, CFD should be coupled with appropriate multiscale biological, physical, and physiological models needed to produce comprehensive, impactful models of mechanobiological systems and complex clinical scenarios. With this perspective, we finally outline open problems and future challenges in the field.
Collapse
Affiliation(s)
- Erica L. Schwarz
- Departments of Pediatrics and Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Luca Pegolotti
- Departments of Pediatrics and Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Martin R. Pfaller
- Departments of Pediatrics and Bioengineering, Stanford University, Stanford, California 94305, USA
| | - Alison L. Marsden
- Departments of Pediatrics and Bioengineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
16
|
Martin AA, Thompson BR, Hahn D, Angulski ABB, Hosny N, Cohen H, Metzger JM. Cardiac Sarcomere Signaling in Health and Disease. Int J Mol Sci 2022; 23:16223. [PMID: 36555864 PMCID: PMC9782806 DOI: 10.3390/ijms232416223] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
The cardiac sarcomere is a triumph of biological evolution wherein myriad contractile and regulatory proteins assemble into a quasi-crystalline lattice to serve as the central point upon which cardiac muscle contraction occurs. This review focuses on the many signaling components and mechanisms of regulation that impact cardiac sarcomere function. We highlight the roles of the thick and thin filament, both as necessary structural and regulatory building blocks of the sarcomere as well as targets of functionally impactful modifications. Currently, a new focus emerging in the field is inter-myofilament signaling, and we discuss here the important mediators of this mechanism, including myosin-binding protein C and titin. As the understanding of sarcomere signaling advances, so do the methods with which it is studied. This is reviewed here through discussion of recent live muscle systems in which the sarcomere can be studied under intact, physiologically relevant conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Joseph M. Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
17
|
Robinett JC, Hanft LM, Biesiadecki B, McDonald KS. Molecular regulation of stretch activation. Am J Physiol Cell Physiol 2022; 323:C1728-C1739. [PMID: 36280392 PMCID: PMC9744651 DOI: 10.1152/ajpcell.00101.2022] [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: 03/07/2022] [Revised: 09/27/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Stretch activation is defined as a delayed increase in force after rapid stretches. Although there is considerable evidence for stretch activation in isolated cardiac myofibrillar preparations, few studies have measured mechanisms of stretch activation in mammalian skeletal muscle fibers. We measured stretch activation following rapid step stretches [∼1%-4% sarcomere length (SL)] during submaximal Ca2+ activations of rat permeabilized slow-twitch skeletal muscle fibers before and after protein kinase A (PKA), which phosphorylates slow myosin binding protein-C. PKA significantly increased stretch activation during low (∼25%) Ca2+ activation and accelerated rates of delayed force development (kef) during both low and half-maximal Ca2+ activation. Following the step stretches and subsequent force development, fibers were rapidly shortened to original sarcomere length, which often elicited a shortening-induced transient force overshoot. After PKA, step shortening-induced transient force overshoot increased ∼10-fold following an ∼4% SL shortening during low Ca2+ activation levels. kdf following step shortening also increased after PKA during low and half-maximal Ca2+ activations. We next investigated thin filament regulation of stretch activation. We tested the interplay between cardiac troponin I (cTnI) phosphorylation at the canonical PKA and novel tyrosine kinase sites on stretch activation. Native slow-skeletal Tn complexes were exchanged with recombinant human cTn complex with different human cTnI N-terminal pseudo-phosphorylation molecules: 1) nonphosphorylated wild type (WT), 2) the canonical S22/23D PKA sites, 3) the tyrosine kinase Y26E site, and 4) the combinatorial S22/23D + Y26E cTnI. All three pseudo-phosphorylated cTnIs elicited greater stretch activation than WT. Following stretch activation, a new, elevated stretch-induced steady-state force was reached with pseudo-phosphorylated cTnI. Combinatorial S22/23D + Y26E pseudo-phosphorylated cTnI increased kdf. These results suggest that slow-skeletal myosin binding protein-C (sMyBP-C) phosphorylation modulates stretch activation by a combination of cross-bridge recruitment and faster cycling kinetics, whereas cTnI phosphorylation regulates stretch activation by both redundant and synergistic mechanisms; and, taken together, these sarcomere phosphoproteins offer precision targets for enhanced contractility.
Collapse
Affiliation(s)
- Joel C Robinett
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Laurin M Hanft
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| | - Brandon Biesiadecki
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio
| | - Kerry S McDonald
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, Columbia, Missouri
| |
Collapse
|
18
|
Ahmed RE, Tokuyama T, Anzai T, Chanthra N, Uosaki H. Sarcomere maturation: function acquisition, molecular mechanism, and interplay with other organelles. Philos Trans R Soc Lond B Biol Sci 2022; 377:20210325. [PMID: 36189811 PMCID: PMC9527934 DOI: 10.1098/rstb.2021.0325] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 06/15/2022] [Indexed: 12/31/2022] Open
Abstract
During postnatal cardiac development, cardiomyocytes mature and turn into adult ones. Hence, all cellular properties, including morphology, structure, physiology and metabolism, are changed. One of the most important aspects is the contractile apparatus, of which the minimum unit is known as a sarcomere. Sarcomere maturation is evident by enhanced sarcomere alignment, ultrastructural organization and myofibrillar isoform switching. Any maturation process failure may result in cardiomyopathy. Sarcomere function is intricately related to other organelles, and the growing evidence suggests reciprocal regulation of sarcomere and mitochondria on their maturation. Herein, we summarize the molecular mechanism that regulates sarcomere maturation and the interplay between sarcomere and other organelles in cardiomyocyte maturation. This article is part of the theme issue 'The cardiomyocyte: new revelations on the interplay between architecture and function in growth, health, and disease'.
Collapse
Affiliation(s)
- Razan E. Ahmed
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Takeshi Tokuyama
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Tatsuya Anzai
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Department of Pediatrics, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Nawin Chanthra
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hideki Uosaki
- Division of Regenerative Medicine, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
19
|
Müller M, Eghbalian R, Boeckel JN, Frese KS, Haas J, Kayvanpour E, Sedaghat-Hamedani F, Lackner MK, Tugrul OF, Ruppert T, Tappu R, Martins Bordalo D, Kneuer JM, Piekarek A, Herch S, Schudy S, Keller A, Grammes N, Bischof C, Klinke A, Cardoso-Moreira M, Kaessmann H, Katus HA, Frey N, Steinmetz LM, Meder B. NIMA-related kinase 9 regulates the phosphorylation of the essential myosin light chain in the heart. Nat Commun 2022; 13:6209. [PMID: 36266340 PMCID: PMC9585074 DOI: 10.1038/s41467-022-33658-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 09/28/2022] [Indexed: 12/24/2022] Open
Abstract
To adapt to changing hemodynamic demands, regulatory mechanisms modulate actin-myosin-kinetics by calcium-dependent and -independent mechanisms. We investigate the posttranslational modification of human essential myosin light chain (ELC) and identify NIMA-related kinase 9 (NEK9) to interact with ELC. NEK9 is highly expressed in the heart and the interaction with ELC is calcium-dependent. Silencing of NEK9 results in blunting of calcium-dependent ELC-phosphorylation. CRISPR/Cas9-mediated disruption of NEK9 leads to cardiomyopathy in zebrafish. Binding to ELC is mediated via the protein kinase domain of NEK9. A causal relationship between NEK9 activity and ELC-phosphorylation is demonstrated by genetic sensitizing in-vivo. Finally, we observe significantly upregulated ELC-phosphorylation in dilated cardiomyopathy patients and provide a unique map of human ELC-phosphorylation-sites. In summary, NEK9-mediated ELC-phosphorylation is a calcium-dependent regulatory system mediating cardiac contraction and inotropy.
Collapse
Affiliation(s)
- Marion Müller
- Kardiogenetikzentrum Heidelberg, University Hospital of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Rose Eghbalian
- Kardiogenetikzentrum Heidelberg, University Hospital of Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
| | - Jes-Niels Boeckel
- Clinic and Polyclinic for Cardiology, University of Leipzig, Leipzig, Germany
| | - Karen S Frese
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Jan Haas
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Elham Kayvanpour
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Farbod Sedaghat-Hamedani
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Maximilian K Lackner
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Oguz F Tugrul
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Thomas Ruppert
- CFMP, Core Facility for Mass Spectrometry & Proteomics at ZMBH, Heidelberg University, Heidelberg, Germany
- ZMBH, Center for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Rewati Tappu
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Diana Martins Bordalo
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Jasmin M Kneuer
- Clinic and Polyclinic for Cardiology, University of Leipzig, Leipzig, Germany
| | - Annika Piekarek
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Sabine Herch
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Sarah Schudy
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
- Department of Neurology and Neurological Sciences, Stanford University Medical School, Stanford, CA, USA
| | - Nadja Grammes
- Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
- Department of Neurology and Neurological Sciences, Stanford University Medical School, Stanford, CA, USA
| | - Cornelius Bischof
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Anna Klinke
- Clinic for General and Interventional Cardiology/ Angiology, Herz- und Diabeteszentrum NRW, University Hospital of the Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | | | - Henrik Kaessmann
- ZMBH, Center for Molecular Biology, Heidelberg University, Heidelberg, Germany
| | - Hugo A Katus
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Norbert Frey
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Department of Medicine III, University of Heidelberg, Heidelberg, Germany
| | - Lars M Steinmetz
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany
- Stanford Genome Technology Center, Stanford University Medical School, Stanford, CA, USA
| | - Benjamin Meder
- Kardiogenetikzentrum Heidelberg, University Hospital of Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Berlin, Germany.
- Stanford Genome Technology Center, Stanford University Medical School, Stanford, CA, USA.
| |
Collapse
|
20
|
Kazmierczak K, Liang J, Gomez-Guevara M, Szczesna-Cordary D. Functional comparison of phosphomimetic S15D and T160D mutants of myosin regulatory light chain exchanged in cardiac muscle preparations of HCM and WT mice. Front Cardiovasc Med 2022; 9:988066. [PMID: 36204565 PMCID: PMC9530205 DOI: 10.3389/fcvm.2022.988066] [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: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 12/30/2022] Open
Abstract
In this study, we investigated the rescue potential of two phosphomimetic mutants of the myosin regulatory light chain (RLC, MYL2 gene), S15D, and T160D RLCs. S15D-RLC mimics phosphorylation of the established serine-15 site of the human cardiac RLC. T160D-RLC mimics the phosphorylation of threonine-160, identified by computational analysis as a high-score phosphorylation site of myosin RLC. Cardiac myosin and left ventricular papillary muscle (LVPM) fibers were isolated from a previously generated model of hypertrophic cardiomyopathy (HCM), Tg-R58Q, and Tg-wild-type (WT) mice. Muscle specimens were first depleted of endogenous RLC and then reconstituted with recombinant human cardiac S15D and T160D phosphomimetic RLCs. Preparations reconstituted with recombinant human cardiac WT-RLC and R58Q-RLC served as controls. Mouse myosins were then tested for the actin-activated myosin ATPase activity and LVPM fibers for the steady-state force development and Ca2+-sensitivity of force. The data showed that S15D-RLC significantly increased myosin ATPase activity compared with T160D-RLC or WT-RLC reconstituted preparations. The two S15D and T160D phosphomimetic RLCs were able to rescue Vmax of Tg-R58Q myosin reconstituted with recombinant R58Q-RLC, but the effect of S15D-RLC was more pronounced than T160D-RLC. Low tension observed for R58Q-RLC reconstituted LVPM from Tg-R58Q mice was equally rescued by both phosphomimetic RLCs. In the HCM Tg-R58Q myocardium, the S15D-RLC caused a shift from the super-relaxed (SRX) state to the disordered relaxed (DRX) state, and the number of heads readily available to interact with actin and produce force was increased. At the same time, T160D-RLC stabilized the SRX state at a level similar to R58Q-RLC reconstituted fibers. We report here on the functional superiority of the established S15 phospho-site of the human cardiac RLC vs. C-terminus T160-RLC, with S15D-RLC showing therapeutic potential in mitigating a non-canonical HCM behavior underlined by hypocontractile behavior of Tg-R58Q myocardium.
Collapse
|
21
|
Lee E, Liu Z, Nguyen N, Nairn A, Chang AN. Myosin light chain phosphatase catalytic subunit dephosphorylates cardiac myosin via mechanisms dependent and independent of the MYPT regulatory subunits. J Biol Chem 2022; 298:102296. [PMID: 35872014 PMCID: PMC9418503 DOI: 10.1016/j.jbc.2022.102296] [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: 12/09/2021] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022] Open
Abstract
Cardiac muscle myosin regulatory light chain (RLC) is constitutively phosphorylated at ∼0.4 mol phosphate/mol RLC in normal hearts, and phosphorylation is maintained by balanced activities of dedicated cardiac muscle–specific myosin light chain kinase and myosin light chain phosphatase (MLCP). Previously, the identity of the cardiac-MLCP was biochemically shown to be similar to the smooth muscle MLCP, which is a well-characterized trimeric protein comprising the regulatory subunit (MYPT1), catalytic subunit PP1cβ, and accessory subunit M20. In smooth muscles in vivo, MYPT1 and PP1cβ co-stabilize each other and are both necessary for normal smooth muscle contractions. In the cardiac muscle, MYPT1 and MYPT2 are both expressed, but contributions to physiological regulation of cardiac myosin dephosphorylation are unclear. We hypothesized that the main catalytic subunit for cardiac-MLCP is PP1cβ, and maintenance of RLC phosphorylation in vivo is dependent on regulation by striated muscle–specific MYPT2. Here, we used PP1cβ conditional knockout mice to biochemically define cardiac-MLCP proteins and developed a cardiac myofibrillar phosphatase assay to measure the direct contribution of MYPT-regulated and MYPT-independent phosphatase activities toward phosphorylated cardiac myosin. We report that (1) PP1cβ is the main isoform expressed in the cardiac myocyte, (2) cardiac muscle pathogenesis in PP1cβ knockout animals involve upregulation of total PP1cα in myocytes and non-muscle cells, (3) the stability of cardiac MYPT1 and MYPT2 proteins in vivo is not dependent on the PP1cβ expression, and (4) phosphorylated myofibrillar cardiac myosin is dephosphorylated by both myosin-targeted and soluble MYPT-independent PP1cβ activities. These results contribute to our understanding of the cardiac-MLCP in vivo.
Collapse
Affiliation(s)
- Eunyoung Lee
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas Texas 75390 USA
| | - Zhenan Liu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas Texas 75390 USA
| | - Nhu Nguyen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas Texas 75390 USA
| | - Angus Nairn
- Department of Psychiatry, Yale University School of Medicine, New Haven CT 06508 USA
| | - Audrey N Chang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas Texas 75390 USA; Pak Center for Mineral Metabolism and Clinical Research, UTSW Medical Center, Dallas Texas 75390 USA.
| |
Collapse
|
22
|
Bartolucci C, Forouzandehmehr M, Severi S, Paci M. A Novel In Silico Electromechanical Model of Human Ventricular Cardiomyocyte. Front Physiol 2022; 13:906146. [PMID: 35721558 PMCID: PMC9198403 DOI: 10.3389/fphys.2022.906146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/10/2022] [Indexed: 11/25/2022] Open
Abstract
Contractility has become one of the main readouts in computational and experimental studies on cardiomyocytes. Following this trend, we propose a novel mathematical model of human ventricular cardiomyocytes electromechanics, BPSLand, by coupling a recent human contractile element to the BPS2020 model of electrophysiology. BPSLand is the result of a hybrid optimization process and it reproduces all the electrophysiology experimental indices captured by its predecessor BPS2020, simultaneously enabling the simulation of realistic human active tension and its potential abnormalities. The transmural heterogeneity in both electrophysiology and contractility departments was simulated consistent with previous computational and in vitro studies. Furthermore, our model could capture delayed afterdepolarizations (DADs), early afterdepolarizations (EADs), and contraction abnormalities in terms of aftercontractions triggered by either drug action or special pacing modes. Finally, we further validated the mechanical results of the model against previous experimental and in silico studies, e.g., the contractility dependence on pacing rate. Adding a new level of applicability to the normative models of human cardiomyocytes, BPSLand represents a robust, fully-human in silico model with promising capabilities for translational cardiology.
Collapse
Affiliation(s)
- Chiara Bartolucci
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | | | - Stefano Severi
- Computational Physiopathology Unit, Department of Electrical, Electronic and Information Engineering "Guglielmo Marconi", University of Bologna, Bologna, Italy
| | - Michelangelo Paci
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
23
|
Riaz M, Park J, Sewanan LR, Ren Y, Schwan J, Das SK, Pomianowski PT, Huang Y, Ellis MW, Luo J, Liu J, Song L, Chen IP, Qiu C, Yazawa M, Tellides G, Hwa J, Young LH, Yang L, Marboe CC, Jacoby DL, Campbell SG, Qyang Y. Muscle LIM Protein Force-Sensing Mediates Sarcomeric Biomechanical Signaling in Human Familial Hypertrophic Cardiomyopathy. Circulation 2022; 145:1238-1253. [PMID: 35384713 PMCID: PMC9109819 DOI: 10.1161/circulationaha.121.056265] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Familial hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease and is typically caused by mutations in genes encoding sarcomeric proteins that regulate cardiac contractility. HCM manifestations include left ventricular hypertrophy and heart failure, arrythmias, and sudden cardiac death. How dysregulated sarcomeric force production is sensed and leads to pathological remodeling remains poorly understood in HCM, thereby inhibiting the efficient development of new therapeutics. METHODS Our discovery was based on insights from a severe phenotype of an individual with HCM and a second genetic alteration in a sarcomeric mechanosensing protein. We derived cardiomyocytes from patient-specific induced pluripotent stem cells and developed robust engineered heart tissues by seeding induced pluripotent stem cell-derived cardiomyocytes into a laser-cut scaffold possessing native cardiac fiber alignment to study human cardiac mechanobiology at both the cellular and tissue levels. Coupled with computational modeling for muscle contraction and rescue of disease phenotype by gene editing and pharmacological interventions, we have identified a new mechanotransduction pathway in HCM, shown to be essential in modulating the phenotypic expression of HCM in 5 families bearing distinct sarcomeric mutations. RESULTS Enhanced actomyosin crossbridge formation caused by sarcomeric mutations in cardiac myosin heavy chain (MYH7) led to increased force generation, which, when coupled with slower twitch relaxation, destabilized the MLP (muscle LIM protein) stretch-sensing complex at the Z-disc. Subsequent reduction in the sarcomeric muscle LIM protein level caused disinhibition of calcineurin-nuclear factor of activated T-cells signaling, which promoted cardiac hypertrophy. We demonstrate that the common muscle LIM protein-W4R variant is an important modifier, exacerbating the phenotypic expression of HCM, but alone may not be a disease-causing mutation. By mitigating enhanced actomyosin crossbridge formation through either genetic or pharmacological means, we alleviated stress at the Z-disc, preventing the development of hypertrophy associated with sarcomeric mutations. CONCLUSIONS Our studies have uncovered a novel biomechanical mechanism through which dysregulated sarcomeric force production is sensed and leads to pathological signaling, remodeling, and hypertrophic responses. Together, these establish the foundation for developing innovative mechanism-based treatments for HCM that stabilize the Z-disc MLP-mechanosensory complex.
Collapse
Affiliation(s)
- Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jinkyu Park
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Lorenzo R. Sewanan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Yongming Ren
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Jonas Schwan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Subhash K. Das
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | | | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew W. Ellis
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Juli Liu
- Department of Pediatrics, Anatomy and Cell Biology, Indiana University, Indianapolis, IN, USA
| | - Loujin Song
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | - I-Ping Chen
- Department of Oral Health and Diagnostic Sciences, University of Connecticut Health, Farmington, CT, USA
| | | | - Masayuki Yazawa
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY, USA
| | | | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| | - Lawrence H. Young
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Lei Yang
- Department of Pediatrics, Anatomy and Cell Biology, Indiana University, Indianapolis, IN, USA
| | - Charles C. Marboe
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Daniel L. Jacoby
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Stuart G. Campbell
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Stem Cell Center, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
24
|
Wu Y, Hu Q, Wang S, Liu C, Shan Y, Guo W, Jiang R, Wang X, Gu J. Highly Regional Genes: graph-based gene selection for single cell RNA-seq data. J Genet Genomics 2022; 49:891-899. [PMID: 35144027 DOI: 10.1016/j.jgg.2022.01.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 12/26/2022]
Abstract
Gene selection is an indispensable step for analyzing noisy and high-dimensional single cell RNA-seq (scRNA-seq) data. Compared with the commonly used variance-based methods, by mimicking the human maker selection in the 2D visualization of cells, a new feature selection method called HRG (Highly Regional Genes) is proposed to find the informative genes which show regional expression patterns in the cell-cell similarity network. We mathematically find the optimal expression patterns which can maximize the proposed scoring function. In comparison with several unsupervised methods, HRG shows high accuracy and robustness, and can increase the performance of downstream cell clustering and gene correlation analysis. Also, it is applicable for selecting informative genes of sequencing-based spatial transcriptomic data.
Collapse
Affiliation(s)
- Yanhong Wu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Qifan Hu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Shicheng Wang
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Changyi Liu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yiran Shan
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wenbo Guo
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Rui Jiang
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xiaowo Wang
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
25
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
26
|
Powers JD, Kirkland NJ, Liu C, Razu SS, Fang X, Engler AJ, Chen J, McCulloch AD. Subcellular Remodeling in Filamin C Deficient Mouse Hearts Impairs Myocyte Tension Development during Progression of Dilated Cardiomyopathy. Int J Mol Sci 2022; 23:871. [PMID: 35055055 PMCID: PMC8779483 DOI: 10.3390/ijms23020871] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/28/2021] [Accepted: 01/11/2022] [Indexed: 01/15/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a life-threatening form of heart disease that is typically characterized by progressive thinning of the ventricular walls, chamber dilation, and systolic dysfunction. Multiple mutations in the gene encoding filamin C (FLNC), an actin-binding cytoskeletal protein in cardiomyocytes, have been found in patients with DCM. However, the mechanisms that lead to contractile impairment and DCM in patients with FLNC variants are poorly understood. To determine how FLNC regulates systolic force transmission and DCM remodeling, we used an inducible, cardiac-specific FLNC-knockout (icKO) model to produce a rapid onset of DCM in adult mice. Loss of FLNC reduced systolic force development in single cardiomyocytes and isolated papillary muscles but did not affect twitch kinetics or calcium transients. Electron and immunofluorescence microscopy showed significant defects in Z-disk alignment in icKO mice and altered myofilament lattice geometry. Moreover, a loss of FLNC induces a softening myocyte cortex and structural adaptations at the subcellular level that contribute to disrupted longitudinal force production during contraction. Spatially explicit computational models showed that these structural defects could be explained by a loss of inter-myofibril elastic coupling at the Z-disk. Our work identifies FLNC as a key regulator of the multiscale ultrastructure of cardiomyocytes and therefore plays an important role in maintaining systolic mechanotransmission pathways, the dysfunction of which may be key in driving progressive DCM.
Collapse
Affiliation(s)
- Joseph D. Powers
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
| | - Natalie J. Kirkland
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
| | - Canzhao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.L.); (X.F.)
| | - Swithin S. Razu
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
| | - Xi Fang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.L.); (X.F.)
| | - Adam J. Engler
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
| | - Ju Chen
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.L.); (X.F.)
| | - Andrew D. McCulloch
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093, USA; (N.J.K.); (S.S.R.); (A.J.E.); (J.C.); (A.D.M.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (C.L.); (X.F.)
- Institute for Engineering in Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
27
|
Papadaki M, Kampaengsri T, Barrick SK, Campbell SG, von Lewinski D, Rainer PP, Harris SP, Greenberg MJ, Kirk JA. Myofilament glycation in diabetes reduces contractility by inhibiting tropomyosin movement, is rescued by cMyBPC domains. J Mol Cell Cardiol 2022; 162:1-9. [PMID: 34487755 PMCID: PMC8766917 DOI: 10.1016/j.yjmcc.2021.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/21/2021] [Accepted: 08/19/2021] [Indexed: 01/17/2023]
Abstract
Diabetes doubles the risk of developing heart failure (HF). As the prevalence of diabetes grows, so will HF unless the mechanisms connecting these diseases can be identified. Methylglyoxal (MG) is a glycolysis by-product that forms irreversible modifications on lysine and arginine, called glycation. We previously found that myofilament MG glycation causes sarcomere contractile dysfunction and is increased in patients with diabetes and HF. The aim of this study was to discover the molecular mechanisms by which MG glycation of myofilament proteins cause sarcomere dysfunction and to identify therapeutic avenues to compensate. In humans with type 2 diabetes without HF, we found increased glycation of sarcomeric actin compared to non-diabetics and it correlated with decreased calcium sensitivity. Depressed calcium sensitivity is pathogenic for HF, therefore myofilament glycation represents a promising therapeutic target to inhibit the development of HF in diabetics. To identify possible therapeutic targets, we further defined the molecular actions of myofilament glycation. Skinned myocytes exposed to 100 μM MG exhibited decreased calcium sensitivity, maximal calcium-activated force, and crossbridge kinetics. Replicating MG's functional affects using a computer simulation of sarcomere function predicted simultaneous decreases in tropomyosin's blocked-to-closed rate transition and crossbridge duty cycle were consistent with all experimental findings. Stopped-flow experiments and ATPase activity confirmed MG decreased the blocked-to-closed transition rate. Currently, no therapeutics target tropomyosin, so as proof-of-principal, we used a n-terminal peptide of myosin-binding protein C, previously shown to alter tropomyosin's position on actin. C0C2 completely rescued MG-induced calcium desensitization, suggesting a possible treatment for diabetic HF.
Collapse
Affiliation(s)
- Maria Papadaki
- Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, Illinois, USA
| | - Theerachat Kampaengsri
- Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, Illinois, USA
| | - Samantha K. Barrick
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, St Louis, Missouri, USA
| | - Stuart G. Campbell
- Department of Bioengineering, Yale University, New Haven, Connecticut, USA
| | | | - Peter P. Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Samantha P. Harris
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Michael J. Greenberg
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, St Louis, Missouri, USA
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, Illinois, USA,Corresponding Author: Jonathan A. Kirk, Ph.D., Department of Cell and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Center for Translational Research and Education, Room 522, 2160 S. First Ave., Maywood, IL 60153, Ph: 708-216-6348,
| |
Collapse
|
28
|
Pathophysiology of heart failure and an overview of therapies. Cardiovasc Pathol 2022. [DOI: 10.1016/b978-0-12-822224-9.00025-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
29
|
Regulation of myosin light-chain phosphorylation and its roles in cardiovascular physiology and pathophysiology. Hypertens Res 2022; 45:40-52. [PMID: 34616031 DOI: 10.1038/s41440-021-00733-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/19/2021] [Accepted: 07/08/2021] [Indexed: 01/22/2023]
Abstract
The regulation of muscle contraction is a critical function in the cardiovascular system, and abnormalities may be life-threatening or cause illness. The common basic mechanism in muscle contraction is the interaction between the protein filaments myosin and actin. Although this interaction is primarily regulated by intracellular Ca2+, the primary targets and intracellular signaling pathways differ in vascular smooth muscle and cardiac muscle. Phosphorylation of the myosin regulatory light chain (RLC) is a primary molecular switch for smooth muscle contraction. The equilibrium between phosphorylated and unphosphorylated RLC is dynamically achieved through two enzymes, myosin light chain kinase, a Ca2+-dependent enzyme, and myosin phosphatase, which modifies the Ca2+ sensitivity of contractions. In cardiac muscle, the primary target protein for Ca2+ is troponin C on thin filaments; however, RLC phosphorylation also plays a modulatory role in contraction. This review summarizes recent advances in our understanding of the regulation, physiological function, and pathophysiological involvement of RLC phosphorylation in smooth and cardiac muscles.
Collapse
|
30
|
Forouzandehmehr M, Koivumäki JT, Hyttinen J, Paci M. A mathematical model of hiPSC cardiomyocytes electromechanics. Physiol Rep 2021; 9:e15124. [PMID: 34825519 PMCID: PMC8617339 DOI: 10.14814/phy2.15124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 01/21/2023] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are becoming instrumental in cardiac research, human-based cell level cardiotoxicity tests, and developing patient-specific care. As one of the principal functional readouts is contractility, we propose a novel electromechanical hiPSC-CM computational model named the hiPSC-CM-CE. This model comprises a reparametrized version of contractile element (CE) by Rice et al., 2008, with a new passive force formulation, integrated into a hiPSC-CM electrophysiology formalism by Paci et al. in 2020. Our simulated results were validated against in vitro data reported for hiPSC-CMs at matching conditions from different labs. Specifically, key action potential (AP) and calcium transient (CaT) biomarkers simulated by the hiPSC-CM-CE model were within the experimental ranges. On the mechanical side, simulated cell shortening, contraction-relaxation kinetic indices (RT50 and RT25 ), and the amplitude of tension fell within the experimental intervals. Markedly, as an inter-scale analysis, correct classification of the inotropic effects due to non-cardiomyocytes in hiPSC-CM tissues was predicted on account of the passive force expression introduced to the CE. Finally, the physiological inotropic effects caused by Verapamil and Bay-K 8644 and the aftercontractions due to the early afterdepolarizations (EADs) were simulated and validated against experimental data. In the future, the presented model can be readily expanded to take in pharmacological trials and genetic mutations, such as those involved in hypertrophic cardiomyopathy, and study arrhythmia trigger mechanisms.
Collapse
Affiliation(s)
| | - Jussi T. Koivumäki
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Jari Hyttinen
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Michelangelo Paci
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| |
Collapse
|
31
|
Kovács Á, Herwig M, Budde H, Delalat S, Kolijn D, Bódi B, Hassoun R, Tangos M, Zhazykbayeva S, Balogh Á, Czuriga D, Van Linthout S, Tschöpe C, Dhalla NS, Mügge A, Tóth A, Papp Z, Barta J, Hamdani N. Interventricular Differences of Signaling Pathways-Mediated Regulation of Cardiomyocyte Function in Response to High Oxidative Stress in the Post-Ischemic Failing Rat Heart. Antioxidants (Basel) 2021; 10:antiox10060964. [PMID: 34208541 PMCID: PMC8234177 DOI: 10.3390/antiox10060964] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 01/09/2023] Open
Abstract
Standard heart failure (HF) therapies have failed to improve cardiac function or survival in HF patients with right ventricular (RV) dysfunction suggesting a divergence in the molecular mechanisms of RV vs. left ventricular (LV) failure. Here we aimed to investigate interventricular differences in sarcomeric regulation and function in experimental myocardial infarction (MI)-induced HF with reduced LV ejection fraction (HFrEF). MI was induced by LAD ligation in Sprague-Dawley male rats. Sham-operated animals served as controls. Eight weeks after intervention, post-ischemic HFrEF and Sham animals were euthanized. Heart tissue samples were deep-frozen stored (n = 3-5 heart/group) for ELISA, kinase activity assays, passive stiffness and Ca2+-sensitivity measurements on isolated cardiomyocytes, phospho-specific Western blot, and PAGE of contractile proteins, as well as for collagen gene expressions. Markers of oxidative stress and inflammation showed interventricular differences in post-ischemic rats: TGF-β1, lipid peroxidation, and 3-nitrotyrosine levels were higher in the LV than RV, while hydrogen peroxide, VCAM-1, TNFα, and TGF-β1 were increased in both ventricles. In addition, nitric oxide (NO) level was significantly decreased, while FN-1 level was significantly increased only in the LV, but both were unchanged in RV. CaMKII activity showed an 81.6% increase in the LV, in contrast to a 38.6% decrease in the RV of HFrEF rats. Cardiomyocyte passive stiffness was higher in the HFrEF compared to the Sham group as evident from significantly steeper Fpassive vs. sarcomere length relationships. In vitro treatment with CaMKIIδ, however, restored cardiomyocyte passive stiffness only in the HFrEF RV, but had no effect in the HFrEF LV. PKG activity was lower in both ventricles in the HFrEF compared to the Sham group. In vitro PKG administration decreased HFrEF cardiomyocyte passive stiffness; however, the effect was more pronounced in the HFrEF LV than HFrEF RV. In line with this, we observed distinct changes of titin site-specific phosphorylation in the RV vs. LV of post-ischemic rats, which may explain divergent cardiomyocyte stiffness modulation observed. Finally, Ca2+-sensitivity of RV cardiomyocytes was unchanged, while LV cardiomyocytes showed increased Ca2+-sensitivity in the HFrEF group. This could be explained by decreased Ser-282 phosphorylation of cMyBP-C by 44.5% in the RV, but without any alteration in the LV, while Ser-23/24 phosphorylation of cTnI was decreased in both ventricles in the HFrEF vs. the Sham group. Our data pointed to distinct signaling pathways-mediated phosphorylations of sarcomeric proteins for the RV and LV of the post-ischemic failing rat heart. These results implicate divergent responses for oxidative stress and open a new avenue in targeting the RV independently of the LV.
Collapse
Affiliation(s)
- Árpád Kovács
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.K.); (B.B.); (A.T.); (Z.P.)
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Melissa Herwig
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Simin Delalat
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Detmar Kolijn
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Beáta Bódi
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.K.); (B.B.); (A.T.); (Z.P.)
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Melina Tangos
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Saltanat Zhazykbayeva
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Ágnes Balogh
- Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.B.); (D.C.); (J.B.)
| | - Dániel Czuriga
- Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.B.); (D.C.); (J.B.)
| | - Sophie Van Linthout
- Berlin Institute of Health at Charite (BIH)-Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany; (S.V.L.); (C.T.)
| | - Carsten Tschöpe
- Berlin Institute of Health at Charite (BIH)-Universitätmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany; (S.V.L.); (C.T.)
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Department of Physiology and Pathophysiology, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R2H 2A6, Canada;
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
| | - Attila Tóth
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.K.); (B.B.); (A.T.); (Z.P.)
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary
| | - Zoltán Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.K.); (B.B.); (A.T.); (Z.P.)
- HAS-UD Vascular Biology and Myocardial Pathophysiology Research Group, Hungarian Academy of Sciences, H-4032 Debrecen, Hungary
| | - Judit Barta
- Department of Cardiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (Á.B.); (D.C.); (J.B.)
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL) Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany; (M.H.); (H.B.); (S.D.); (D.K.); (R.H.); (M.T.); (S.Z.); (A.M.)
- Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, 44801 Bochum, Germany
- Correspondence: ; Tel.: +49-234-5095-9053
| |
Collapse
|
32
|
Complex functionality of protein phosphatase 1 isoforms in the heart. Cell Signal 2021; 85:110059. [PMID: 34062239 DOI: 10.1016/j.cellsig.2021.110059] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/21/2021] [Accepted: 05/28/2021] [Indexed: 02/04/2023]
Abstract
Protein phosphatase 1(PP1) is a key regulator of cardiac function through dephosphorylating serine/threonine residues within target proteins to oppose the function of protein kinases. Studies from failing hearts of animal models and human patients have demonstrated significant increase of PP1 activity in myocardium, while elevated PP1 activity in transgenic mice leads to cardiac dysfunction, suggesting that PP1 might be a therapeutic target to ameliorate cardiac dysfunction in failing hearts. In fact, cardiac overexpression of inhibitor 1, the endogenous inhibitor of PP1, increases cardiac contractility and suppresses heart failure progression. However, this notion of PP1 inhibition for heart failure treatment has been challenged by recent studies on the isoform-specific roles of PP1 in the heart. PP1 is a holoenzyme composed of catalytic subunits (PP1α, PP1β, or PP1γ) and regulatory proteins that target them to distinct subcellular locations for functional specificity. This review will summarize how PP1 regulates phosphorylation of some of the key cardiac proteins involved in Ca2+ handling and cardiac contraction, and the potential role of PP1 isoforms in controlling cardiac physiology and pathophysiology.
Collapse
|
33
|
Cao L, Wang Z, Zhang D, Li X, Hou C, Ren C. Phosphorylation of myosin regulatory light chain at Ser17 regulates actomyosin dissociation. Food Chem 2021; 356:129655. [PMID: 33831832 DOI: 10.1016/j.foodchem.2021.129655] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 11/19/2022]
Abstract
Phosphorylation of myosin regulatory light chain (MRLC) can regulate muscle contraction and thus affect actomyosin dissociation and meat quality. The objective of this study was to explore the mechanism by how MRLC phosphorylation regulates actomyosin dissociation and thus develop strategies for improving meat quality. Here, the phosphorylation status of MRLC was modulated by myosin light chain kinase and myosin light chain kinase inhibitor. MRLC phosphorylation at Ser17 decreased the kinetic energy and total energy of actomyosin, thus stabilized the structure, facilitating the interaction between myosin and actin; this was one possible way that MRLC phosphorylation at Ser17 negatively affects actomyosin dissociation. Moreover, MRLC phosphorylation at Ser17 was beneficial to the formation of ionic bonds, hydrogen bonds, and hydrophobic interaction between myosin and actin, and was the second possible way that MRLC phosphorylation at Ser17 negatively affects actomyosin dissociation.
Collapse
Affiliation(s)
- Lichuang Cao
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| | - Zhenyu Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China.
| | - Dequan Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China.
| | - Xin Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China
| | - Chengli Hou
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China
| | - Chi Ren
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China
| |
Collapse
|
34
|
Zhang J, Wang L, Kazmierczak K, Yun H, Szczesna-Cordary D, Kawai M. Hypertrophic cardiomyopathy associated E22K mutation in myosin regulatory light chain decreases calcium-activated tension and stiffness and reduces myofilament Ca 2+ sensitivity. FEBS J 2021; 288:4596-4613. [PMID: 33548158 DOI: 10.1111/febs.15753] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/11/2020] [Accepted: 02/04/2021] [Indexed: 11/30/2022]
Abstract
We investigated the mechanisms associated with E22K mutation in myosin regulatory light chain (RLC), found to cause hypertrophic cardiomyopathy (HCM) in humans and mice. Specifically, we characterized the mechanical profiles of papillary muscle fibers from transgenic mice expressing human ventricular RLC wild-type (Tg-WT) or E22K mutation (Tg-E22K). Because the two mouse models expressed different amounts of transgene, the B6SJL mouse line (NTg) was used as an additional control. Mechanical experiments were carried out on Ca2+ - and ATP-activated fibers and in rigor. Sinusoidal analysis was performed to elucidate the effect of E22K on tension and stiffness during activation/rigor, tension-pCa, and myosin cross-bridge (CB) kinetics. We found significant reductions in active tension (by 54%) and stiffness (active by 40% and rigor by 54%). A decrease in the Ca2+ sensitivity of tension (by ∆pCa ~ 0.1) was observed in Tg-E22K compared with Tg-WT fibers. The apparent (=measured) rate constant of exponential process B (2πb: force generation step) was not affected by E22K, but the apparent rate constant of exponential process C (2πc: CB detachment step) was faster in Tg-E22K compared with Tg-WT fibers. Both 2πb and 2πc were smaller in NTg than in Tg-WT fibers, suggesting a kinetic difference between the human and mouse RLC. Our results of E22K-induced reduction in myofilament stiffness and tension suggest that the main effect of this mutation was to disturb the interaction of RLC with the myosin heavy chain and impose structural abnormalities in the lever arm of myosin CB. When placed in vivo, the E22K mutation is expected to result in reduced contractility and decreased cardiac output whereby leading to HCM. SUB-DISCIPLINE Bioenergetics. DATABASE The data that support the findings of this study are available from the corresponding authors upon reasonable request. ANIMAL PROTOCOL BK20150353 (Soochow University). RESEARCH GOVERNANCE School of Nursing: Hua-Gang Hu: seuboyh@163.com; Soochow University: Chen Ge chge@suda.edu.cn.
Collapse
Affiliation(s)
- Jiajia Zhang
- School of Nursing, Medical College, Soochow University, Suzhou, China
| | - Li Wang
- School of Nursing, Medical College, Soochow University, Suzhou, China
| | | | - Hang Yun
- School of Nursing, Medical College, Soochow University, Suzhou, China
| | | | - Masataka Kawai
- Department of Anatomy and Cell Biology, University of Iowa, IA, USA
| |
Collapse
|
35
|
Awinda PO, Watanabe M, Bishaw Y, Huckabee AM, Agonias KB, Kazmierczak K, Szczesna-Cordary D, Tanner BCW. Mavacamten decreases maximal force and Ca 2+ sensitivity in the N47K-myosin regulatory light chain mouse model of hypertrophic cardiomyopathy. Am J Physiol Heart Circ Physiol 2021; 320:H881-H890. [PMID: 33337957 PMCID: PMC8082789 DOI: 10.1152/ajpheart.00345.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 01/12/2023]
Abstract
Morbidity and mortality associated with heart disease is a growing threat to the global population, and novel therapies are needed. Mavacamten (formerly called MYK-461) is a small molecule that binds to cardiac myosin and inhibits myosin ATPase. Mavacamten is currently in clinical trials for the treatment of obstructive hypertrophic cardiomyopathy (HCM), and it may provide benefits for treating other forms of heart disease. We investigated the effect of mavacamten on cardiac muscle contraction in two transgenic mouse lines expressing the human isoform of cardiac myosin regulatory light chain (RLC) in their hearts. Control mice expressed wild-type RLC (WT-RLC), and HCM mice expressed the N47K RLC mutation. In the absence of mavacamten, skinned papillary muscle strips from WT-RLC mice produced greater isometric force than strips from N47K mice. Adding 0.3 µM mavacamten decreased maximal isometric force and reduced Ca2+ sensitivity of contraction for both genotypes, but this reduction in pCa50 was nearly twice as large for WT-RLC versus N47K. We also used stochastic length-perturbation analysis to characterize cross-bridge kinetics. The cross-bridge detachment rate was measured as a function of [MgATP] to determine the effect of mavacamten on myosin nucleotide handling rates. Mavacamten increased the MgADP release and MgATP binding rates for both genotypes, thereby contributing to faster cross-bridge detachment, which could speed up myocardial relaxation during diastole. Our data suggest that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction via decreased strong cross-bridge binding. Mavacamten may become a useful therapy for patients with heart disease, including some forms of HCM.NEW & NOTEWORTHY Mavacamten is a pharmaceutical that binds to myosin, and it is under investigation as a therapy for some forms of heart disease. We show that mavacamten reduces isometric tension and Ca2+ sensitivity of contraction in skinned myocardial strips from a mouse model of hypertrophic cardiomyopathy that expresses the N47K mutation in cardiac myosin regulatory light chain. Mavacamten reduces contractility by decreasing strong cross-bridge binding, partially due to faster cross-bridge nucleotide handling rates that speed up myosin detachment.
Collapse
Affiliation(s)
- Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Marissa Watanabe
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Yemeserach Bishaw
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Anna M Huckabee
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Keinan B Agonias
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida
| | - Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| |
Collapse
|
36
|
Li X, Zhang D, Ren C, Bai Y, Ijaz M, Hou C, Chen L. Effects of protein posttranslational modifications on meat quality: A review. Compr Rev Food Sci Food Saf 2020; 20:289-331. [PMID: 33443799 DOI: 10.1111/1541-4337.12668] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/14/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023]
Abstract
Meat quality plays an important role in the purchase decision of consumers, affecting producers and retailers. The formation mechanisms determining meat quality are intricate, as several endogenous and exogenous factors contribute during antemortem and postmortem periods. Abundant research has been performed on meat quality; however, unexpected variation in meat quality remains an issue in the meat industry. Protein posttranslational modifications (PTMs) regulate structures and functions of proteins in living tissues, and recent reports confirmed their importance in meat quality. The objective of this review was to provide a summary of the research on the effects of PTMs on meat quality. The effects of four common PTMs, namely, protein phosphorylation, acetylation, S-nitrosylation, and ubiquitination, on meat quality were discussed, with emphasis on the effects of protein phosphorylation on meat tenderness, color, and water holding capacity. The mechanisms and factors that may affect the function of protein phosphorylation are also discussed. The current research confirms that meat quality traits are regulated by multiple PTMs. Cross talk between different PTMs and interactions of PTMs with postmortem biochemical processes need to be explored to improve our understanding on factors affecting meat quality.
Collapse
Affiliation(s)
- Xin Li
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dequan Zhang
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chi Ren
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yuqiang Bai
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Muawuz Ijaz
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chengli Hou
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Li Chen
- Key Laboratory of Agro-Products Processing, Ministry of Agriculture and Rural Affairs, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
37
|
Wang S, Wang H, Su X, Liu B, Wang L, Yan H, Mao S, Huang H, Huang C, Cheng M, Wu G. β-adrenergic activation may promote myosin light chain kinase degradation through calpain in pressure overload-induced cardiac hypertrophy: β-adrenergic activation results in MLCK degradation. Biomed Pharmacother 2020; 129:110438. [PMID: 32768940 DOI: 10.1016/j.biopha.2020.110438] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/13/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND β-adrenergic activation is able to exacerbate cardiac hypertrophy. Myosin light chain kinase (MLCK) and its phosphorylated substrate, phospho-myosin light chain 2 (p-MLC2), play vital roles in regulating cardiac hypertrophy. However, it is not yet clear whether there is a relationship between β-adrenergic activation and MLCK in the progression of cardiac hypertrophy. Therefore, we explored this relationship and the underlying mechanisms in this work. METHODS Cardiac hypertrophy and cardiomyocyte hypertrophy were induced by pressure overload and isoproterenol (ISO) stimulation, respectively. Echocardiography, histological analysis, immunofluorescence and qRT-PCR were used to confirm the successful establishment of the models. A β-blocker (metoprolol) and a calpain inhibitor (calpeptin) were administered to inhibit β-adrenergic activity in rats and calpain in cardiomyocytes, respectively. The protein expression levels of MLCK, myosin light chain 2 (MLC2), p-MLC2, myosin phosphatase 2 (MYPT2), calmodulin (CaM) and calpain were measured using western blotting. A cleavage assay was performed to assess the degradation of recombinant human MLCK by recombinant human calpain. RESULTS The β-blocker alleviated cardiac hypertrophy and dysfunction, increased MLCK and MLC2 phosphorylation and decreased calpain expression in pressure overload-induced cardiac hypertrophy. Additionally, the calpain inhibitor calpeptin attenuated cardiomyocyte hypertrophy, upregulated MLCK and p-MLC2 and reduced MLCK degradation in ISO-induced cardiomyocyte hypertrophy. Recombinant human calpain degraded recombinant human MLCK in vitro in concentration- and time-dependent manners, and this degradation was inhibited by the calpain inhibitor calpeptin. CONCLUSION Our study suggested that β-adrenergic activation may promote the degradation of MLCK through calpain in pressure overload-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Shun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Haixiong Wang
- Department of Cardiology, Shanxi Cardiovascular Hospital, Taiyuan, 030001, China
| | - Xiaoling Su
- Department of Cardiology, Qinghai Provincial People's Hospital, Xining, 810007, China
| | - Beilei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Le Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Hui Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Shuai Mao
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China
| | - Mian Cheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Hubei Key Laboratory of Cardiology, Wuhan, 430060, China; Department of Cardiology, Ezhou Hospital, Renmin Hospital of Wuhan University, Ezhou, 436000, China.
| |
Collapse
|
38
|
Williams JL, Paudyal A, Awad S, Nicholson J, Grzesik D, Botta J, Meimaridou E, Maharaj AV, Stewart M, Tinker A, Cox RD, Metherell LA. Mylk3 null C57BL/6N mice develop cardiomyopathy, whereas Nnt null C57BL/6J mice do not. Life Sci Alliance 2020; 3:3/4/e201900593. [PMID: 32213617 PMCID: PMC7103425 DOI: 10.26508/lsa.201900593] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/10/2020] [Accepted: 03/10/2020] [Indexed: 12/30/2022] Open
Abstract
The C57BL/6J and C57BL/6N mice have well-documented phenotypic and genotypic differences, including the infamous nicotinamide nucleotide transhydrogenase (Nnt) null mutation in the C57BL/6J substrain, which has been linked to cardiovascular traits in mice and cardiomyopathy in humans. To assess whether Nnt loss alone causes a cardiovascular phenotype, we investigated the C57BL/6N, C57BL/6J mice and a C57BL/6J-BAC transgenic rescuing NNT expression, at 3, 12, and 18 mo. We identified a modest dilated cardiomyopathy in the C57BL/6N mice, absent in the two B6J substrains. Immunofluorescent staining of cardiomyocytes revealed eccentric hypertrophy in these mice, with defects in sarcomere organisation. RNAseq analysis identified differential expression of a number of cardiac remodelling genes commonly associated with cardiac disease segregating with the phenotype. Variant calling from RNAseq data identified a myosin light chain kinase 3 (Mylk3) mutation in C57BL/6N mice, which abolishes MYLK3 protein expression. These results indicate the C57BL/6J Nnt-null mice do not develop cardiomyopathy; however, we identified a null mutation in Mylk3 as a credible cause of the cardiomyopathy phenotype in the C57BL/6N.
Collapse
Affiliation(s)
- Jack L Williams
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anju Paudyal
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Sherine Awad
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - James Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Dominika Grzesik
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joaquin Botta
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Eirini Meimaridou
- School of Human Sciences, London Metropolitan University, London, UK
| | - Avinaash V Maharaj
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Michelle Stewart
- Medical Research Council Harwell Institute, Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Andrew Tinker
- William Harvey Heart Centre, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Roger D Cox
- Medical Research Council Harwell Institute, Mammalian Genetics Unit, Harwell Campus, Oxfordshire, UK
| | - Lou A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Charterhouse Square, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
39
|
Sevrieva IR, Brandmeier B, Ponnam S, Gautel M, Irving M, Campbell KS, Sun YB, Kampourakis T. Cardiac myosin regulatory light chain kinase modulates cardiac contractility by phosphorylating both myosin regulatory light chain and troponin I. J Biol Chem 2020; 295:4398-4410. [PMID: 32086378 PMCID: PMC7135997 DOI: 10.1074/jbc.ra119.011945] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
Heart muscle contractility and performance are controlled by posttranslational modifications of sarcomeric proteins. Although myosin regulatory light chain (RLC) phosphorylation has been studied extensively in vitro and in vivo, the precise role of cardiac myosin light chain kinase (cMLCK), the primary kinase acting upon RLC, in the regulation of cardiomyocyte contractility remains poorly understood. In this study, using recombinantly expressed and purified proteins, various analytical methods, in vitro and in situ kinase assays, and mechanical measurements in isolated ventricular trabeculae, we demonstrate that human cMLCK is not a dedicated kinase for RLC but can phosphorylate other sarcomeric proteins with well-characterized regulatory functions. We show that cMLCK specifically monophosphorylates Ser23 of human cardiac troponin I (cTnI) in isolation and in the trimeric troponin complex in vitro and in situ in the native environment of the muscle myofilament lattice. Moreover, we observed that human cMLCK phosphorylates rodent cTnI to a much smaller extent in vitro and in situ, suggesting species-specific adaptation of cMLCK. Although cMLCK treatment of ventricular trabeculae exchanged with rat or human troponin increased their cross-bridge kinetics, the increase in sensitivity of myofilaments to calcium was significantly blunted by human TnI, suggesting that human cTnI phosphorylation by cMLCK modifies the functional consequences of RLC phosphorylation. We propose that cMLCK-mediated phosphorylation of TnI is functionally significant and represents a critical signaling pathway that coordinates the regulatory states of thick and thin filaments in both physiological and potentially pathophysiological conditions of the heart.
Collapse
Affiliation(s)
- Ivanka R Sevrieva
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Birgit Brandmeier
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Saraswathi Ponnam
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Mathias Gautel
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Malcolm Irving
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Kenneth S Campbell
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0298
| | - Yin-Biao Sun
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| | - Thomas Kampourakis
- Randall Centre for Cell and Molecular Biophysics and British Heart Foundation Centre of Research Excellence, King's College London, London SE1 1UL, United Kingdom
| |
Collapse
|
40
|
Parsanathan R, Jain SK. Novel Invasive and Noninvasive Cardiac-Specific Biomarkers in Obesity and Cardiovascular Diseases. Metab Syndr Relat Disord 2020; 18:10-30. [PMID: 31618136 PMCID: PMC7041332 DOI: 10.1089/met.2019.0073] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of fatality and disability worldwide regardless of gender. Obesity has reached epidemic proportions in population across different regions. According to epidemiological studies, CVD risk markers in childhood obesity are one of the significant risk factors for adulthood CVD, but have received disproportionally little attention. This review has examined the evidence for the presence of traditional cardiac biomarkers (nonspecific; lactate dehydrogenase, alanine aminotransferase, aspartate aminotransferase, creatine kinase, myoglobulin, glycogen phosphorylase isoenzyme BB, myosin light chains, ST2, and ischemia-modified albumin) and novel emerging cardiac-specific biomarkers (cardiac troponins, natriuretic peptides, heart-type fatty acid-binding protein, and miRNAs). Besides, noninvasive anatomical and electrophysiological markers (carotid intima-media thickness, coronary artery calcification, and heart rate variability) in CVDs and obesity are also discussed. Modifiable and nonmodifiable risk factors associated with metabolic syndrome in the progression of CVD, such as obesity, diabetes, hypertension, dyslipidemia, oxidative stress, inflammation, and adipocytokines are also outlined. These underlying prognostic risk factors predict the onset of future microvascular and macrovascular complications. The understanding of invasive and noninvasive cardiac-specific biomarkers and the risk factors may yield valuable insights into the pathophysiology and prevention of CVD in a high-risk obese population at an early stage.
Collapse
Affiliation(s)
- Rajesh Parsanathan
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| | - Sushil K. Jain
- Department of Pediatrics and Center for Cardiovascular Diseases and Sciences, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana
| |
Collapse
|
41
|
X Cai L, Tanada Y, D Bello G, C Fleming J, F Alkassis F, Ladd T, Golde T, Koh J, Chen S, Kasahara H. Cardiac MLC2 kinase is localized to the Z-disc and interacts with α-actinin2. Sci Rep 2019; 9:12580. [PMID: 31467300 PMCID: PMC6715661 DOI: 10.1038/s41598-019-48884-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiac contractility is enhanced by phosphorylation of myosin light chain 2 (MLC2) by cardiac-specific MLC kinase (cMLCK), located at the neck region of myosin heavy chain. In normal mouse and human hearts, the level of phosphorylation is maintained relatively constant, at around 30-40% of total MLC2, likely by well-balanced phosphorylation and phosphatase-dependent dephosphorylation. Overexpression of cMLCK promotes sarcomere organization, while the loss of cMLCK leads to cardiac atrophy in vitro and in vivo. In this study, we showed that cMLCK is predominantly expressed at the Z-disc with additional diffuse cytosolic expression in normal adult mouse and human hearts. cMLCK interacts with the Z-disc protein, α-actinin2, with a high-affinity kinetic value of 13.4 ± 0.1 nM through the N-terminus region of cMLCK unique to cardiac-isoform. cMLCK mutant deficient for interacting with α-actinin2 did not promote sarcomeric organization and reduced cardiomyocyte cell size. In contrast, a cMLCK kinase-deficient mutant showed effects similar to wild-type cMLCK on sarcomeric organization and cardiomyocyte cell size. Our results suggest that cMLCK plays a role in sarcomere organization, likely distinct from its role in phosphorylating MLC2, both of which will contribute to the enhancement of cardiac contractility.
Collapse
Affiliation(s)
- Lawrence X Cai
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Yohei Tanada
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Gregory D Bello
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - James C Fleming
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Fariz F Alkassis
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Thomas Ladd
- Department of Neuroscience, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Todd Golde
- Department of Neuroscience, University of Florida, College of Medicine, Gainesville, FL, 32610, USA
| | - Jin Koh
- Proteomics and Mass Spectrometry, Interdisciplinary Center for Biotechnology Research (ICBR), University of Florida, Gainesville, FL, 32610, USA
| | - Sixue Chen
- Proteomics and Mass Spectrometry, Interdisciplinary Center for Biotechnology Research (ICBR), University of Florida, Gainesville, FL, 32610, USA.,Department of Biology, Genetics Institute, Plant Molecular and Cellular Biology Program, University of Florida, Gainesville, FL, 32610, USA
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, University of Florida, College of Medicine, Gainesville, FL, 32610, USA.
| |
Collapse
|
42
|
Fan F, Duan Y, Yang F, Trexler C, Wang H, Huang L, Li Y, Tang H, Wang G, Fang X, Liu J, Jia N, Chen J, Ouyang K. Deletion of heat shock protein 60 in adult mouse cardiomyocytes perturbs mitochondrial protein homeostasis and causes heart failure. Cell Death Differ 2019; 27:587-600. [PMID: 31209364 PMCID: PMC7205885 DOI: 10.1038/s41418-019-0374-x] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/10/2019] [Accepted: 06/05/2019] [Indexed: 01/22/2023] Open
Abstract
To maintain healthy mitochondrial enzyme content and function, mitochondria possess a complex protein quality control system, which is composed of different endogenous sets of chaperones and proteases. Heat shock protein 60 (HSP60) is one of these mitochondrial molecular chaperones and has been proposed to play a pivotal role in the regulation of protein folding and the prevention of protein aggregation. However, the physiological function of HSP60 in mammalian tissues is not fully understood. Here we generated an inducible cardiac-specific HSP60 knockout mouse model, and demonstrated that HSP60 deletion in adult mouse hearts altered mitochondrial complex activity, mitochondrial membrane potential, and ROS production, and eventually led to dilated cardiomyopathy, heart failure, and lethality. Proteomic analysis was performed in purified control and mutant mitochondria before mutant hearts developed obvious cardiac abnormalities, and revealed a list of mitochondrial-localized proteins that rely on HSP60 (HSP60-dependent) for correctly folding in mitochondria. We also utilized an in vitro system to assess the effects of HSP60 deletion on mitochondrial protein import and protein stability after import, and found that both HSP60-dependent and HSP60-independent mitochondrial proteins could be normally imported in mutant mitochondria. However, the former underwent degradation in mutant mitochondria after import, suggesting that the protein exhibited low stability in mutant mitochondria. Interestingly, the degradation could be almost fully rescued by a non-specific LONP1 and proteasome inhibitor, MG132, in mutant mitochondria. Therefore, our results demonstrated that HSP60 plays an essential role in maintaining normal cardiac morphology and function by regulating mitochondrial protein homeostasis and mitochondrial function.
Collapse
Affiliation(s)
- Feifei Fan
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Yaoyun Duan
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Feili Yang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Christa Trexler
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Hong Wang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Lei Huang
- Shenzhen Peking University Hospital, 518055, Shenzhen, China
| | - Yali Li
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Huayuan Tang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China
| | - Gang Wang
- Department of Pathophysiology, School of Medicine, Shenzhen University, 518055, Shenzhen, China
| | - Xi Fang
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jie Liu
- Department of Pathophysiology, School of Medicine, Shenzhen University, 518055, Shenzhen, China
| | - Nan Jia
- Shenzhen People's Hospital, 518055, Shenzhen, China
| | - Ju Chen
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Kunfu Ouyang
- School of Chemical Biology and Biotechnology, State Key Laboratory of Chemical Oncogenomics, Peking University Shenzhen Graduate School, 518055, Shenzhen, China.
| |
Collapse
|
43
|
Tougas CL, Grindrod T, Cai LX, Alkassis FF, Kasahara H. Heterozygous Mylk3 Knockout Mice Partially Recapitulate Human DCM With Heterozygous MYLK3 Mutations. Front Physiol 2019; 10:696. [PMID: 31244672 PMCID: PMC6563786 DOI: 10.3389/fphys.2019.00696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/16/2019] [Indexed: 11/29/2022] Open
Abstract
Backgrounds: Recent studies identified heterozygous variants in MYLK3 gene that encodes cardiac myosin light chain kinase (cMLCK) are related to familial dilated cardiomyopathy (DCM) for the first time. Autosomal dominant traits suggest that pathogenesis of DCM could be related to heterozygous MYLK3 loss-of-function variants (haploinsufficiency). We previously generated and examined homozygous Mylk3 knockout mice that lead to heart failure. It had yet to be examined whether heterozygous Mylk3 knockout mice represent a DCM-like phenotype. Methods and Results: Heterozygous knockout (Mylk3wild/-) mice were examined regarding cardiac function, heart histology and expression of cMLCK protein and mRNA relative to age-matched wild-type controls (Mylk3wild/wild). At 4 months of age, cardiac contractility in heterozygous knockout mice was reduced with percent fractional shortening of 23.3 ± 1.2% compared to 30.1 ± 1.8% in control (Mylk3wild/- vs. Mylk3wild/wild, n = 9 each). In 4-month-old heterozygous knockout hearts, expression of cMLCK mRNA was expectedly reduced by almost half, however, protein expression was reduced by approximately 75% relative to the control wild-type (Mylk3wild/- vs. Mylk3wild/wild, n = 9 each). Isolated ventricular cardiomyocytes from heterozygous knockout mice were larger with increase of short-axis length relative to the cardiomyocytes from control mice. However, increase of heart failure markers as well as interstitial fibrosis were not evident in heterozygous knockout mice compared to controls. Conclusion: Heterozygous Mylk3 knockout mice show mild reduction of cardiac contractility by 4 months of age, and proteins reduced by approximately 75% relative to the control wild-type mice. These mice partly resemble human with the heterozygous MYLK3 mutation, but the reduction in cardiac contractility was milder.
Collapse
Affiliation(s)
- Carson L Tougas
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Tabor Grindrod
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Lawrence X Cai
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Fariz F Alkassis
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Hideko Kasahara
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
44
|
Yadav S, Yuan CC, Kazmierczak K, Liang J, Huang W, Takeuchi LM, Kanashiro-Takeuchi RM, Szczesna-Cordary D. Therapeutic potential of AAV9-S15D-RLC gene delivery in humanized MYL2 mouse model of HCM. J Mol Med (Berl) 2019; 97:1033-1047. [PMID: 31101927 DOI: 10.1007/s00109-019-01791-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/24/2019] [Accepted: 05/01/2019] [Indexed: 12/15/2022]
Abstract
Familial hypertrophic cardiomyopathy (HCM) is an autosomal dominant disorder characterized by ventricular hypertrophy, myofibrillar disarray, and fibrosis, and is primarily caused by mutations in sarcomeric genes. With no definitive cure for HCM, there is an urgent need for the development of novel preventive and reparative therapies. This study is focused on aspartic acid-to-valine (D166V) mutation in the myosin regulatory light chain, RLC (MYL2 gene), associated with a malignant form of HCM. Since myosin RLC phosphorylation is critical for normal cardiac function, we aimed to exploit this post-translational modification via phosphomimetic-RLC gene therapy. We hypothesized that mimicking/modulating cardiac RLC phosphorylation in non-phosphorylatable D166V myocardium would improve heart function of HCM-D166V mice. Adeno-associated virus, serotype-9 (AAV9) was used to deliver phosphomimetic human RLC variant with serine-to-aspartic acid substitution at Ser15-RLC phosphorylation site (S15D-RLC) into the hearts of humanized HCM-D166V mice. Improvement of heart function was monitored by echocardiography, invasive hemodynamics (PV-loops) and muscle contractile mechanics. A significant increase in cardiac output and stroke work and a decrease in relaxation constant, Tau, shown to be prolonged in HCM mice, were observed in AAV- vs. PBS-injected HCM mice. Strain analysis showed enhanced myocardial longitudinal shortening in AAV-treated vs. control mice. In addition, increased maximal contractile force was observed in skinned papillary muscles from AAV-injected HCM hearts. Our data suggest that myosin RLC phosphorylation may have important translational implications for the treatment of RLC mutations-induced HCM and possibly play a role in other disease settings accompanied by depressed Ser15-RLC phosphorylation. KEY MESSAGES: HCM-D166V mice show decreased RLC phosphorylation and decompensated function. AAV9-S15D-RLC gene therapy in HCM-D166V mice, but not in WT-RLC, results in improved heart performance. Global longitudinal strain analysis shows enhanced contractility in AAV vs controls. Increased systolic and diastolic function is paralleled by higher contractile force. Phosphomimic S15D-RLC has a therapeutic potential for HCM.
Collapse
Affiliation(s)
- Sunil Yadav
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Chen-Ching Yuan
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Wenrui Huang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Lauro M Takeuchi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Rosemeire M Kanashiro-Takeuchi
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.,Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
45
|
Breithaupt JJ, Pulcastro HC, Awinda PO, DeWitt DC, Tanner BCW. Regulatory light chain phosphorylation augments length-dependent contraction in PTU-treated rats. J Gen Physiol 2018; 151:66-76. [PMID: 30523115 PMCID: PMC6314387 DOI: 10.1085/jgp.201812158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022] Open
Abstract
Contraction of cardiac muscle is regulated by sarcomere length and proteins that comprise the sarcomeric filaments. Breithaupt et al. find that phosphorylation of myosin regulatory light chain augments length-dependent activation of contraction when β-cardiac myosin heavy chain predominates. Force production by actin–myosin cross-bridges in cardiac muscle is regulated by thin-filament proteins and sarcomere length (SL) throughout the heartbeat. Prior work has shown that myosin regulatory light chain (RLC), which binds to the neck of myosin heavy chain, increases cardiac contractility when phosphorylated. We recently showed that cross-bridge kinetics slow with increasing SLs, and that RLC phosphorylation amplifies this effect, using skinned rat myocardial strips predominantly composed of the faster α-cardiac myosin heavy chain isoform. In the present study, to assess how RLC phosphorylation influences length-dependent myosin function as myosin motor speed varies, we used a propylthiouracil (PTU) diet to induce >95% expression of the slower β-myosin heavy chain isoform in rat cardiac ventricles. We measured the effect of RLC phosphorylation on Ca2+-activated isometric contraction and myosin cross-bridge kinetics (via stochastic length perturbation analysis) in skinned rat papillary muscle strips at 1.9- and 2.2-µm SL. Maximum tension and Ca2+ sensitivity increased with SL, and RLC phosphorylation augmented this response at 2.2-µm SL. Subtle increases in viscoelastic myocardial stiffness occurred with RLC phosphorylation at 2.2-µm SL, but not at 1.9-µm SL, thereby suggesting that RLC phosphorylation increases β-myosin heavy chain binding or stiffness at longer SLs. The cross-bridge detachment rate slowed as SL increased, providing a potential mechanism for prolonged cross-bridge attachment to augment length-dependent activation of contraction at longer SLs. Length-dependent slowing of β-myosin heavy chain detachment rate was not affected by RLC phosphorylation. Together with our previous studies, these data suggest that both α- and β-myosin heavy chain isoforms show a length-dependent activation response and prolonged myosin attachment as SL increases in rat myocardial strips, and that RLC phosphorylation augments length-dependent activation at longer SLs. In comparing cardiac isoforms, however, we found that β-myosin heavy chain consistently showed greater length-dependent sensitivity than α-myosin heavy chain. Our work suggests that RLC phosphorylation is a vital contributor to the regulation of myocardial contractility in both cardiac myosin heavy chain isoforms.
Collapse
Affiliation(s)
- Jason J Breithaupt
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| | - Hannah C Pulcastro
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| | - Peter O Awinda
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| | - David C DeWitt
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| | - Bertrand C W Tanner
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA
| |
Collapse
|
46
|
Yadav S, Kazmierczak K, Liang J, Sitbon YH, Szczesna-Cordary D. Phosphomimetic-mediated in vitro rescue of hypertrophic cardiomyopathy linked to R58Q mutation in myosin regulatory light chain. FEBS J 2018; 286:151-168. [PMID: 30430732 DOI: 10.1111/febs.14702] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 10/03/2018] [Accepted: 11/13/2018] [Indexed: 12/16/2022]
Abstract
Myosin regulatory light chain (RLC) phosphorylation is important for cardiac muscle mechanics/function as well as for the Ca2+ -troponin/tropomyosin regulation of muscle contraction. This study focuses on the arginine to glutamine (R58Q) substitution in the human ventricular RLC (MYL2 gene), linked to malignant hypertrophic cardiomyopathy in humans and causing severe functional abnormalities in transgenic (Tg) R58Q mice, including inhibition of cardiac RLC phosphorylation. Using a phosphomimic recombinant RLC variant where Ser-15 at the phosphorylation site was substituted with aspartic acid (S15D) and placed in the background of R58Q, we aimed to assess whether we could rescue/mitigate R58Q-induced structural/functional abnormalities in vitro. We show rescue of several R58Q-exerted adverse phenotypes in S15D-R58Q-reconstituted porcine cardiac muscle preparations. A low level of maximal isometric force observed for R58Q- versus WT-reconstituted fibers was restored by S15D-R58Q. Significant beneficial effects were also observed on the Vmax of actin-activated myosin ATPase activity in S15D-R58Q versus R58Q-reconstituted myosin, along with its binding to fluorescently labeled actin. We also report that R58Q promotes the OFF state of myosin, both in reconstituted porcine fibers and in Tg mouse papillary muscles, thereby stabilizing the super-relaxed state (SRX) of myosin, characterized by a very low ATP turnover rate. Experiments in S15D-R58Q-reconstituted porcine fibers showed a mild destabilization of the SRX state, suggesting an S15D-mediated shift in disordered-relaxed (DRX)↔SRX equilibrium toward the DRX state of myosin. Our study shows that S15D-phosphomimic can be used as a potential rescue strategy to abrogate/alleviate the RLC mutation-induced phenotypes and is a likely candidate for therapeutic intervention in HCM patients.
Collapse
Affiliation(s)
- Sunil Yadav
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, FL, USA
| | - Katarzyna Kazmierczak
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, FL, USA
| | - Jingsheng Liang
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, FL, USA
| | - Yoel H Sitbon
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, FL, USA
| | - Danuta Szczesna-Cordary
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, FL, USA
| |
Collapse
|
47
|
Cai W, Hite ZL, Lyu B, Wu Z, Lin Z, Gregorich ZR, Messer AE, McIlwain SJ, Marston SB, Kohmoto T, Ge Y. Temperature-sensitive sarcomeric protein post-translational modifications revealed by top-down proteomics. J Mol Cell Cardiol 2018; 122:11-22. [PMID: 30048711 DOI: 10.1016/j.yjmcc.2018.07.247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/11/2018] [Accepted: 07/21/2018] [Indexed: 10/28/2022]
Abstract
Despite advancements in symptom management for heart failure (HF), this devastating clinical syndrome remains the leading cause of death in the developed world. Studies using animal models have greatly advanced our understanding of the molecular mechanisms underlying HF; however, differences in cardiac physiology and the manifestation of HF between animals, particularly rodents, and humans necessitates the direct interrogation of human heart tissue samples. Nevertheless, an ever-present concern when examining human heart tissue samples is the potential for artefactual changes related to temperature changes during tissue shipment or sample processing. Herein, we examined the effects of temperature on the post-translational modifications (PTMs) of sarcomeric proteins, the proteins responsible for muscle contraction, under conditions mimicking those that might occur during tissue shipment or sample processing. Using a powerful top-down proteomics method, we found that sarcomeric protein PTMs were differentially affected by temperature. Specifically, cardiac troponin I and enigma homolog isoform 2 showed robust increases in phosphorylation when tissue was incubated at either 4 °C or 22 °C. The observed increase is likely due to increased cyclic AMP levels and activation of protein kinase A in the tissue. On the contrary, cardiac troponin T and myosin regulatory light chain phosphorylation decreased when tissue was incubated at 4 °C or 22 °C. Furthermore, significant protein degradation was also observed after incubation at 4 °C or 22 °C. Overall, these results indicate that temperature exerts various effects on sarcomeric protein PTMs and careful tissue handling is critical for studies involving human heart samples. Moreover, these findings highlight the power of top-down proteomics for examining the integrity of cardiac tissue samples.
Collapse
Affiliation(s)
- Wenxuan Cai
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachary L Hite
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Beini Lyu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zhijie Wu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Ziqing Lin
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Zachery R Gregorich
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Andrew E Messer
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sean J McIlwain
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA; UW Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Steve B Marston
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Takushi Kohmoto
- Department of Surgery, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ying Ge
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA; Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
48
|
Stöhr EJ, Takayama H, Ferrari G. Stretch your heart-but not too far: The role of titin mutations in dilated cardiomyopathy. J Thorac Cardiovasc Surg 2018; 156:209-214. [PMID: 29685583 PMCID: PMC6724204 DOI: 10.1016/j.jtcvs.2017.10.160] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Eric J Stöhr
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, NY; School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom.
| | - Hiroo Takayama
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY
| | - Giovanni Ferrari
- Division of Cardiothoracic and Vascular Surgery, Department of Surgery, Columbia University Irving Medical Center, New York, NY
| |
Collapse
|
49
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
50
|
Stroud MJ, Fang X, Zhang J, Guimarães-Camboa N, Veevers J, Dalton ND, Gu Y, Bradford WH, Peterson KL, Evans SM, Gerace L, Chen J. Luma is not essential for murine cardiac development and function. Cardiovasc Res 2018; 114:378-388. [PMID: 29040414 PMCID: PMC6019056 DOI: 10.1093/cvr/cvx205] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/11/2017] [Indexed: 12/13/2022] Open
Abstract
AIMS Luma is a recently discovered, evolutionarily conserved protein expressed in mammalian heart, which is associated with the LInker of Nucleoskeleton and Cytoskeleton (LINC) complex. The LINC complex structurally integrates the nucleus and the cytoplasm and plays a critical role in mechanotransduction across the nuclear envelope. Mutations in several LINC components in both humans and mice result in various cardiomyopathies, implying they play essential, non-redundant roles. A single amino acid substitution of serine 358 to leucine (S358L) in Luma is the unequivocal cause of a distinct form of arrhythmogenic cardiomyopathy. However, the role of Luma in heart has remained obscure. In addition, it also remains to be determined how the S358L mutation in Luma leads to cardiomyopathy. METHODS AND RESULTS To determine the role of Luma in the heart, we first determined the expression pattern of Luma in mouse heart. Luma was sporadically expressed in cardiomyocytes throughout the heart, but was highly and uniformly expressed in cardiac fibroblasts and vascular smooth muscle cells. We also generated germline null Luma mice and discovered that germline null mutants were viable and exhibited normal cardiac function. Luma null mice also responded normally to pressure overload induced by transverse aortic constriction. In addition, localization and expression of other LINC complex components in both cardiac myocytes and fibroblasts was unaffected by global loss of Luma. Furthermore, we also generated and characterized Luma S358L knock-in mice, which displayed normal cardiac function and morphology. CONCLUSION Our data suggest that Luma is dispensable for murine cardiac development and function and that the Luma S358L mutation alone may not cause cardiomyopathy in mice.
Collapse
MESH Headings
- Animals
- Arrhythmogenic Right Ventricular Dysplasia/genetics
- Arrhythmogenic Right Ventricular Dysplasia/metabolism
- Cells, Cultured
- Cytoskeleton/metabolism
- Female
- Fibroblasts/metabolism
- Gene Expression Regulation, Developmental
- Genetic Predisposition to Disease
- Heart/embryology
- Heart/physiopathology
- Humans
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mechanotransduction, Cellular
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Morphogenesis
- Myocardium/metabolism
- Myocardium/pathology
- Myocytes, Cardiac/metabolism
- Myocytes, Smooth Muscle/metabolism
- Nuclear Matrix/metabolism
Collapse
Affiliation(s)
- Matthew J Stroud
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Xi Fang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Jianlin Zhang
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Nuno Guimarães-Camboa
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jennifer Veevers
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Nancy D Dalton
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Yusu Gu
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - William H Bradford
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Kirk L Peterson
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Sylvia M Evans
- Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Larry Gerace
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ju Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
- Corresponding author. Tel: 858 822 4276; fax: 858 822 3027, E-mail:
| |
Collapse
|