1
|
Desai D, Song T, Singh RR, Baby A, McNamara J, Green LC, Nabavizadeh P, Ericksen M, Bazrafshan S, Natesan S, Sadayappan S. MYBPC3 D389V Variant Induces Hypercontractility in Cardiac Organoids. Cells 2024; 13:1913. [PMID: 39594661 PMCID: PMC11592734 DOI: 10.3390/cells13221913] [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: 10/03/2024] [Revised: 10/27/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
MYBPC3, encoding cardiac myosin binding protein-C (cMyBP-C), is the most mutated gene known to cause hypertrophic cardiomyopathy (HCM). However, since little is known about the underlying etiology, additional in vitro studies are crucial to defining the underlying molecular mechanisms. Accordingly, this study aimed to investigate the molecular mechanisms underlying the pathogenesis of HCM associated with a polymorphic variant (D389V) in MYBPC3 by using isogenic human-induced pluripotent stem cell (hiPSC)-derived cardiac organoids (hCOs). The hiPSC-derived cardiomyocytes (hiPSC-CMs) and hCOs were generated from human subjects to define the molecular, cellular, functional, and energetic changes caused by the MYBPC3D389V variant, which is associated with increased fractional shortening and highly prevalent in South Asian descendants. Recombinant C0-C2, N' region of cMyBP-C (wild-type and D389V), and myosin S2 proteins were also utilized to perform binding and motility assays in vitro. Confocal and electron microscopic analyses of hCOs generated from noncarriers (NC) and carriers of the MYBPC3D389V variant revealed the presence of highly organized sarcomeres. Furthermore, functional experiments showed hypercontractility, faster calcium cycling, and faster contractile kinetics in hCOs expressing MYBPC3D389V than NC hCOs. Interestingly, significantly increased cMyBP-C phosphorylation in MYBPC3D389V hCOs was observed, but without changes in total protein levels, in addition to higher oxidative stress and lower mitochondrial membrane potential (ΔΨm). Next, spatial mapping revealed the presence of endothelial cells, fibroblasts, macrophages, immune cells, and cardiomyocytes in the hCOs. The hypercontractile function was significantly improved after the treatment of the myosin inhibitor mavacamten (CAMZYOS®) in MYBPC3D389V hCOs. Lastly, various vitro binding assays revealed a significant loss of affinity in the presence of MYBPC3D389V with myosin S2 region as a likely mechanism for hypercontraction. Conceptually, we showed the feasibility of assessing the functional and molecular mechanisms of HCM using highly translatable hCOs through pragmatic experiments that led to determining the MYBPC3D389V hypercontractile phenotype, which was rescued by the administration of a myosin inhibitor.
Collapse
Affiliation(s)
- Darshini Desai
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Taejeong Song
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Rohit R. Singh
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Akhil Baby
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - James McNamara
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Lisa C. Green
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Pooneh Nabavizadeh
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Mark Ericksen
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Sholeh Bazrafshan
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Sakthivel Sadayappan
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; (T.S.); (R.R.S.); (A.B.); (J.M.); (P.N.)
| |
Collapse
|
2
|
Cizauskas HE, Burnham HV, Panni A, Peña A, Alvarez-Arce A, Davis MT, Araujo KN, Delligatti CE, Edassery S, Kirk JA, Arora R, Barefield DY. Proteolytic degradation of atrial sarcomere proteins underlies contractile defects in atrial fibrillation. Am J Physiol Heart Circ Physiol 2024; 327:H460-H472. [PMID: 38940916 PMCID: PMC11442024 DOI: 10.1152/ajpheart.00148.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 05/24/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
Atrial fibrillation (AFib) is the most common cardiac rhythm disturbance, often treated via electrical cardioversion. Following rhythm restoration, a period of depressed mechanical function known as atrial stunning occurs, suggesting that defects in contractility occur in AFib and are revealed upon restoration of rhythm. This project aims to define the contractile remodeling that occurs in AFib. To assess contractile function, we used a canine atrial tachypacing model of induced AFib. Mass spectrometry analysis showed dysregulation of contractile proteins in samples from AFib compared with sinus rhythm atria. Atrial cardiomyocytes show reduced force of contraction, decreased resting tension, and increased calcium sensitivity in skinned single cardiomyocyte studies. These alterations correlated with degradation of myofilament proteins including myosin heavy chain altering force of contraction, titin altering resting tension, and troponin I altering calcium sensitivity. We measured degradation of other myofilament proteins, including cardiac myosin binding protein C and actinin, that show degradation products in the AFib samples that are absent in the sinus rhythm atria. Many of the degradation products appeared as discrete cleavage products that are generated by calpain proteolysis. We assessed calpain activity and found it to be significantly increased. These results provide an understanding of the contractile remodeling that occurs in AFib and provide insight into the molecular explanation for atrial stunning and the increased risk of atrial thrombus and stroke in AFib.NEW & NOTEWORTHY Atrial fibrillation is the most common cardiac rhythm disorder, and remodeling during atrial fibrillation is highly variable between patients. This study has defined the biophysical changes in contractility that occur in atrial fibrillation along with identifying potential molecular mechanisms that may drive this remodeling. This includes proteolysis of several myofilament proteins including titin, troponin I, myosin heavy chain, myosin binding protein C, and actinin, which is consistent with the observed contractile deficits.
Collapse
Affiliation(s)
- Hannah E Cizauskas
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Hope V Burnham
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Azaria Panni
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - Alexandra Peña
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Alejandro Alvarez-Arce
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - M Therese Davis
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Kelly N Araujo
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Christine E Delligatti
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Seby Edassery
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Jonathan A Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| | - Rishi Arora
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States
| | - David Y Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Chicago, Illinois, United States
| |
Collapse
|
3
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Substrate mechanics unveil early structural and functional pathology in iPSC micro-tissue models of hypertrophic cardiomyopathy. iScience 2024; 27:109954. [PMID: 38827401 PMCID: PMC11141149 DOI: 10.1016/j.isci.2024.109954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/22/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024] Open
Abstract
Hypertension is a major cause of morbidity and mortality in patients with hypertrophic cardiomyopathy (HCM), suggesting a potential role for mechanics in HCM pathogenesis. Here, we developed an in vitro physiological model to investigate how mechanics acts together with HCM-linked myosin binding protein C (MYBPC3) mutations to trigger disease. Micro-heart muscles (μHM) were engineered from induced pluripotent stem cell (iPSC)-derived cardiomyocytes bearing MYBPC3+/- mutations and challenged to contract against substrates of different elasticity. μHMs that worked against substrates with stiffness at or exceeding the stiffness of healthy adult heart muscle exhibited several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in MYBPC3+/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca2+ intake through membrane-embedded channels underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease with iPSC technology.
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - David Schuftan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan D. Moreno
- Division of Cardiology, Department of Medicine, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Ghiska Ramahdita
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Druv Bhagavan
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Jonathan Silva
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University in Saint Louis, Saint Louis, MO 63130, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, MO 63130, USA
| |
Collapse
|
4
|
Desai D, Song T, Singh RR, Baby A, McNamara J, Green L, Nabavizadeh P, Ericksen M, Bazrafshan S, Natesan S, Sadayappan S. MYBPC3 D389V Variant Induces Hypercontractility in Cardiac Organoids. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596463. [PMID: 38853909 PMCID: PMC11160759 DOI: 10.1101/2024.05.29.596463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
BACKGROUND MYBPC3 , encoding cardiac myosin binding protein-C (cMyBP-C), is the most mutated gene known to cause hypertrophic cardiomyopathy (HCM). However, since little is known about the underlying etiology, additional in vitro studies are crucial to defining the underlying molecular mechanisms. Accordingly, this study aimed to investigate the molecular mechanisms underlying the pathogenesis of HCM associated with a polymorphic variant (D389V) in MYBPC3 by using human-induced pluripotent stem cell (hiPSC)-derived cardiac organoids (hCOs). METHODS The hiPSC-derived cardiomyocytes (hiPSC-CMs) and hCOs were generated from human subjects to define the molecular, cellular, and functional changes caused by the MYBPC3 D389V variant. This variant is associated with increased fractional shortening and is highly prevalent in South Asian descendants. Recombinant C0-C2, N'-region of cMyBP-C (wildtype and D389V), and myosin S2 proteins were also utilized to perform binding and motility assays in vitro . RESULTS Confocal and electron microscopic analyses of hCOs generated from noncarriers (NC) and carriers of the MYBPC3 D389V variant revealed the presence of highly organized sarcomeres. Furthermore, functional experiments showed hypercontractility with increased contraction velocity, faster calcium cycling, and faster contractile kinetics in hCOs expressing MYBPC3 D389V than NC hCOs. Interestingly, significantly increased cMyBP-C phosphorylation in MYBPC3 D389V hCOs was observed, but without changes in total protein levels, in addition to higher oxidative stress and lower mitochondrial membrane potential (ΔΨm). Next, spatial mapping revealed the presence of endothelial cells, fibroblasts, macrophages, immune cells, and cardiomyocytes in the hCOs. The hypercontractile function was significantly improved after treatment with the myosin inhibitor mavacamten (CAMZYOS®) in MYBPC3 D389V hCOs. Lastly, various in vitro binding assays revealed a significant loss of affinity in the presence of MYBPC3 D389V with myosin S2 region as a likely mechanism for hypercontraction. CONCLUSIONS Conceptually, we showed the feasibility of assessing the functional and molecular mechanisms of HCM using highly translatable hCOs through pragmatic experiments that led to determining the MYBPC3 D389V hypercontractile phenotype, which was rescued by administration of a myosin inhibitor. Novelty and Significance: What Is Known?: MYBPC3 mutations have been implicated in hypertrophic cardiomyopathy. D389V is a polymorphic variant of MYBPC3 predicted to be present in 53000 US South Asians owing to the founder effect. D389V carriers have shown evidence of hyperdynamic heart, and human-induced pluripotent stem cells (hiPSC)-derived cardiomyocytes with D389V show cellular hypertrophy and irregular calcium transients. The molecular mechanism by which the D389V variant develops pathological cardiac dysfunction remains to be conclusively determined.What New Information Does This Article Contribute ?: The authors leveraged a highly translational cardiac organoid model to explore the role of altered cardiac calcium handling and cardiac contractility as a common pathway leading to pathophysiological phenotypes in patients with early HCM. The MYBPC3 D389V -mediated pathological pathway is first studied here by comparing functional properties using three-dimensional cardiac organoids differentiated from hiPSC and determining the presence of hypercontraction. Our data demonstrate that faster sarcomere kinetics resulting from lower binding affinity between D389V-mutated cMyBP-C protein and myosin S2, as evidenced by in vitro studies, could cause hypercontractility which was rescued by administration of mavacamten (CAMZYOS®), a myosin inhibitor. In addition, hypercontractility causes secondary mitochondrial defects such as higher oxidative stress and lower mitochondrial membrane potential (ΔΨm), highlighting a possible early adaptive response to primary sarcomeric changes. Early treatment of MYBPC3 D389V carriers with mavacamten may prevent or reduce early HCM-related pathology. GRAPHICAL ABSTRACT: A graphical abstract is available for this article.
Collapse
|
5
|
Schuftan D, Kooh YKG, Guo J, Sun Y, Aryan L, Stottlemire B, Berkland C, Genin GM, Huebsch N. Dynamic control of contractile resistance to iPSC-derived micro-heart muscle arrays. J Biomed Mater Res A 2024; 112:534-548. [PMID: 37952251 PMCID: PMC10922390 DOI: 10.1002/jbm.a.37642] [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: 05/31/2023] [Revised: 09/25/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023]
Abstract
Many types of cardiovascular disease are linked to the mechanical forces placed on the heart. However, our understanding of how mechanical forces exactly affect the cellular biology of the heart remains incomplete. In vitro models based on cardiomyocytes derived from human induced pluripotent stem cells (iPSC-CM) enable researchers to develop medium to high-throughput systems to study cardiac mechanobiology at the cellular level. Previous models have been developed to enable the study of mechanical forces, such as cardiac afterload. However, most of these models require exogenous extracellular matrix (ECM) to form cardiac tissues. Recently, a system was developed to simulate changes in afterload by grafting ECM-free micro-heart muscle arrays to elastomeric substrates of discrete stiffnesses. In the present study, we extended this system by combining the elastomer-grafted tissue arrays with a magnetorheological elastomeric substrate. This system allows iPSC-CM based micro-heart muscle arrays to experience dynamic changes in contractile resistance to mimic dynamically altered afterload. Acute changes in substrate stiffness led to acute changes in the calcium dynamics and contractile forces, illustrating the system's ability to dynamically elicit changes in tissue mechanics by dynamically changing contractile resistance.
Collapse
Affiliation(s)
- David Schuftan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yasaman Kargar Gaz Kooh
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Jingxuan Guo
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Yuwen Sun
- Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Lavanya Aryan
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Bryce Stottlemire
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
| | - Cory Berkland
- Department of Chemical and Petroleum Engineering, Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, Kansas, USA
| | - Guy M. Genin
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri, USA
- NSF Center for Engineering Mechanobiology, St. Louis, Missouri, USA
| |
Collapse
|
6
|
Barefield DY, Tonino P, Woulfe KC, Rahmanseresht S, O’Leary TS, Burnham HV, Wasserstrom JA, Kirk JA, Previs MJ, Granzier HL, McNally EM. Myosin-binding protein H-like regulates myosin-binding protein distribution and function in atrial cardiomyocytes. Proc Natl Acad Sci U S A 2023; 120:e2314920120. [PMID: 38091294 PMCID: PMC10741380 DOI: 10.1073/pnas.2314920120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/25/2023] [Indexed: 12/18/2023] Open
Abstract
Mutations in atrial-enriched genes can cause a primary atrial myopathy that can contribute to overall cardiovascular dysfunction. MYBPHL encodes myosin-binding protein H-like (MyBP-HL), an atrial sarcomere protein that shares domain homology with the carboxy-terminus of cardiac myosin-binding protein-C (cMyBP-C). The function of MyBP-HL and the relationship between MyBP-HL and cMyBP-C is unknown. To decipher the roles of MyBP-HL, we used structured illumination microscopy, immuno-electron microscopy, and mass spectrometry to establish the localization and stoichiometry of MyBP-HL. We found levels of cMyBP-C, a major regulator of myosin function, were half as abundant compared to levels in the ventricle. In genetic mouse models, loss of MyBP-HL doubled cMyBP-C abundance in the atria, and loss of cMyBP-C doubled MyBP-HL abundance in the atria. Structured illumination microscopy showed that both proteins colocalize in the C-zone of the A-band, with MyBP-HL enriched closer to the M-line. Immuno-electron microscopy of mouse atria showed MyBP-HL strongly localized 161 nm from the M-line, consistent with localization to the third 43 nm repeat of myosin heads. Both cMyBP-C and MyBP-HL had less-defined sarcomere localization in the atria compared to ventricle, yet areas with the expected 43 nm repeat distance were observed for both proteins. Isometric force measurements taken from control and Mybphl null single atrial myofibrils revealed that loss of Mybphl accelerated the linear phase of relaxation. These findings support a mechanism where MyBP-HL regulates cMyBP-C abundance to alter the kinetics of sarcomere relaxation in atrial sarcomeres.
Collapse
Affiliation(s)
- David Y. Barefield
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL60153
| | - Paola Tonino
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, AZ85724
| | - Kathleen C. Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO80045
| | - Sheema Rahmanseresht
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT01655
| | - Thomas S. O’Leary
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT01655
| | - Hope V. Burnham
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL60153
| | - J. Andrew Wasserstrom
- Department of Medicine and The Feinberg Cardiovascular and Renal Institute, Northwestern University Feinberg School of Medicine, Chicago, IL60611
| | - Jonathan A. Kirk
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL60153
| | - Michael J. Previs
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT01655
| | - Henk L. Granzier
- Department of Cell and Molecular Medicine, University of Arizona, Tucson, AZ85724
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL60611
| |
Collapse
|
7
|
Song T, McNamara JW, Baby A, Ma W, Landim-Vieira M, Natesan S, Pinto JR, Lorenz JN, Irving TC, Sadayappan S. Unlocking the Role of sMyBP-C: A Key Player in Skeletal Muscle Development and Growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.23.563591. [PMID: 38076858 PMCID: PMC10705270 DOI: 10.1101/2023.10.23.563591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Skeletal muscle is the largest organ in the body, responsible for gross movement and metabolic regulation. Recently, variants in the MYBPC1 gene have been implicated in a variety of developmental muscle diseases, such as distal arthrogryposis. How MYBPC1 variants cause disease is not well understood. Here, through a collection of novel gene-edited mouse models, we define a critical role for slow myosin binding protein-C (sMyBP-C), encoded by MYBPC1, across muscle development, growth, and maintenance during prenatal, perinatal, postnatal and adult stages. Specifically, Mybpc1 knockout mice exhibited early postnatal lethality and impaired skeletal muscle formation and structure, skeletal deformity, and respiratory failure. Moreover, a conditional knockout of Mybpc1 in perinatal, postnatal and adult stages demonstrates impaired postnatal muscle growth and function secondary to disrupted actomyosin interaction and sarcomere structural integrity. These findings confirm the essential role of sMyBP-C in skeletal muscle and reveal specific functions in both prenatal embryonic musculoskeletal development and postnatal muscle growth and function.
Collapse
Affiliation(s)
- Taejeong Song
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - James W. McNamara
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Akhil Baby
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Weikang Ma
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL, USA
| | - John N. Lorenz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Thomas C. Irving
- BioCAT, Department of Biology, Illinois Institute of Technology, Chicago, IL, USA
| | - Sakthivel Sadayappan
- Center for Cardiovascular Research, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
8
|
Guo J, Jiang H, Schuftan D, Moreno JD, Ramahdita G, Aryan L, Bhagavan D, Silva J, Huebsch N. Mechanical Resistance to Micro-Heart Tissue Contractility unveils early Structural and Functional Pathology in iPSC Models of Hypertrophic Cardiomyopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.30.564856. [PMID: 37961198 PMCID: PMC10634965 DOI: 10.1101/2023.10.30.564856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Hypertrophic cardiomyopathy is the most common cause of sudden death in the young. Because the disease exhibits variable penetrance, there are likely nongenetic factors that contribute to the manifestation of the disease phenotype. Clinically, hypertension is a major cause of morbidity and mortality in patients with HCM, suggesting a potential synergistic role for the sarcomeric mutations associated with HCM and mechanical stress on the heart. We developed an in vitro physiological model to investigate how the afterload that the heart muscle works against during contraction acts together with HCM-linked MYBPC3 mutations to trigger a disease phenotype. Micro-heart muscle arrays (μHM) were engineered from iPSC-derived cardiomyocytes bearing MYBPC3 loss-of-function mutations and challenged to contract against mechanical resistance with substrates stiffnesses ranging from the of embryonic hearts (0.4 kPa) up to the stiffness of fibrotic adult hearts (114 kPa). Whereas MYBPC3 +/- iPSC-cardiomyocytes showed little signs of disease pathology in standard 2D culture, μHMs that included components of afterload revealed several hallmarks of HCM, including cellular hypertrophy, impaired contractile energetics, and maladaptive calcium handling. Remarkably, we discovered changes in troponin C and T localization in the MYBPC3 +/- μHM that were entirely absent in 2D culture. Pharmacologic studies suggested that excessive Ca 2+ intake through membrane-embedded channels, rather than sarcoplasmic reticulum Ca 2+ ATPase (SERCA) dysfunction or Ca 2+ buffering at myofilaments underlie the observed electrophysiological abnormalities. These results illustrate the power of physiologically relevant engineered tissue models to study inherited disease mechanisms with iPSC technology.
Collapse
|
9
|
Thompson AD, Wagner MJ, Rodriguez J, Malhotra A, Vander Roest S, Lilienthal U, Shao H, Vignesh M, Weber K, Yob JM, Prosser BL, Helms AS, Gestwicki JE, Ginsburg D, Day SM. An Unbiased Screen Identified the Hsp70-BAG3 Complex as a Regulator of Myosin-Binding Protein C3. JACC Basic Transl Sci 2023; 8:1198-1211. [PMID: 37791314 PMCID: PMC10544073 DOI: 10.1016/j.jacbts.2023.04.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 10/05/2023]
Abstract
Variants in the gene myosin-binding protein C3 (MYBPC3) account for approximately 50% of familial hypertrophic cardiomyopathy (HCM), leading to reduced levels of myosin-binding protein C3 (MyBP-C), the protein product made by gene MYBPC3. Elucidation of the pathways that regulate MyBP-C protein homeostasis could uncover new therapeutic strategies. Toward this goal, we screened a library of 2,426 bioactive compounds and identified JG98, an allosteric modulator of heat shock protein 70 that inhibits interaction with Bcl-2-associated athanogene (BAG) domain co-chaperones. JG98 reduces MyBP-C protein levels. Furthermore, genetic reduction of BAG3 phenocopies treatment with JG-98 by reducing MYBP-C protein levels.. Thus, an unbiased compound screen identified the heat shock protein 70-BAG3 complex as a regulator of MyBP-C stability.
Collapse
Affiliation(s)
- Andrea D. Thompson
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Marcus J. Wagner
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Juliani Rodriguez
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Alok Malhotra
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Steve Vander Roest
- Center for Chemical Genomics, Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Ulla Lilienthal
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Hao Shao
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - Mathav Vignesh
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Keely Weber
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jaime M. Yob
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Benjamin L. Prosser
- Department of Physiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adam S. Helms
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason E. Gestwicki
- Institute for Neurodegenerative Diseases and Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, California, USA
| | - David Ginsburg
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, USA
- Howard Hughes Medical Institute, Chevy Chase, Maryland, USA
- The Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharlene M. Day
- Department of Internal Medicine, Division of Cardiovascular Medicine and Cardiovascular Institute, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
10
|
Chou C, Martin GL, Perera G, Awata J, Larson A, Blanton R, Chin MT. A novel αB-crystallin R123W variant drives hypertrophic cardiomyopathy by promoting maladaptive calcium-dependent signal transduction. Front Cardiovasc Med 2023; 10:1223244. [PMID: 37435054 PMCID: PMC10331725 DOI: 10.3389/fcvm.2023.1223244] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 06/13/2023] [Indexed: 07/13/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disorder affecting 1 in 500 people in the general population. Characterized by asymmetric left ventricular hypertrophy, cardiomyocyte disarray and cardiac fibrosis, HCM is a highly complex disease with heterogenous clinical presentation, onset and complication. While mutations in sarcomere genes can account for a substantial proportion of familial cases of HCM, 40%-50% of HCM patients do not carry such sarcomere variants and the causal mutations for their diseases remain elusive. Recently, we identified a novel variant of the alpha-crystallin B chain (CRYABR123W) in a pair of monozygotic twins who developed concordant HCM phenotypes that manifested over a nearly identical time course. Yet, how CRYABR123W promotes the HCM phenotype remains unclear. Here, we generated mice carrying the CryabR123W knock-in allele and demonstrated that hearts from these animals exhibit increased maximal elastance at young age but reduced diastolic function with aging. Upon transverse aortic constriction, mice carrying the CryabR123W allele developed pathogenic left ventricular hypertrophy with substantial cardiac fibrosis and progressively decreased ejection fraction. Crossing of mice with a Mybpc3 frame-shift model of HCM did not potentiate pathological hypertrophy in compound heterozygotes, indicating that the pathological mechanisms in the CryabR123W model are independent of the sarcomere. In contrast to another well-characterized CRYAB variant (R120G) which induced Desmin aggregation, no evidence of protein aggregation was observed in hearts expressing CRYABR123W despite its potent effect on driving cellular hypertrophy. Mechanistically, we uncovered an unexpected protein-protein interaction between CRYAB and calcineurin. Whereas CRYAB suppresses maladaptive calcium signaling in response to pressure-overload, the R123W mutation abolished this effect and instead drove pathologic NFAT activation. Thus, our data establish the CryabR123W allele as a novel genetic model of HCM and unveiled additional sarcomere-independent mechanisms of cardiac pathological hypertrophy.
Collapse
Affiliation(s)
- Chun Chou
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| | - Gregory L. Martin
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Gayani Perera
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Junya Awata
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Amy Larson
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Robert Blanton
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| | - Michael T. Chin
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
11
|
Tudurachi BS, Zăvoi A, Leonte A, Țăpoi L, Ureche C, Bîrgoan SG, Chiuariu T, Anghel L, Radu R, Sascău RA, Stătescu C. An Update on MYBPC3 Gene Mutation in Hypertrophic Cardiomyopathy. Int J Mol Sci 2023; 24:10510. [PMID: 37445689 PMCID: PMC10341819 DOI: 10.3390/ijms241310510] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent genetically inherited cardiomyopathy that follows an autosomal dominant inheritance pattern. The majority of HCM cases can be attributed to mutation of the MYBPC3 gene, which encodes cMyBP-C, a crucial structural protein of the cardiac muscle. The manifestation of HCM's morphological, histological, and clinical symptoms is subject to the complex interplay of various determinants, including genetic mutation and environmental factors. Approximately half of MYBPC3 mutations give rise to truncated protein products, while the remaining mutations cause insertion/deletion, frameshift, or missense mutations of single amino acids. In addition, the onset of HCM may be attributed to disturbances in the protein and transcript quality control systems, namely, the ubiquitin-proteasome system and nonsense-mediated RNA dysfunctions. The aforementioned genetic modifications, which appear to be associated with unfavorable lifelong outcomes and are largely influenced by the type of mutation, exhibit a unique array of clinical manifestations ranging from asymptomatic to arrhythmic syncope and even sudden cardiac death. Although the current understanding of the MYBPC3 mutation does not comprehensively explain the varied phenotypic manifestations witnessed in patients with HCM, patients with pathogenic MYBPC3 mutations can exhibit an array of clinical manifestations ranging from asymptomatic to advanced heart failure and sudden cardiac death, leading to a higher rate of adverse clinical outcomes. This review focuses on MYBPC3 mutation and its characteristics as a prognostic determinant for disease onset and related clinical consequences in HCM.
Collapse
Affiliation(s)
- Bogdan-Sorin Tudurachi
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Alexandra Zăvoi
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Andreea Leonte
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Laura Țăpoi
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Carina Ureche
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Silviu Gabriel Bîrgoan
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Traian Chiuariu
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Larisa Anghel
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Rodica Radu
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Radu Andy Sascău
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| | - Cristian Stătescu
- Department of Internal Medicine, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy of Iasi, 16 University Street, 700115 Iasi, Romania; (B.-S.T.); (L.Ț.); (C.U.); (L.A.); (R.R.); (R.A.S.); (C.S.)
- Prof. Dr. George I.M. Georgescu Institute of Cardiovascular Diseases, Carol I Boulevard, No. 50, 700503 Iasi, Romania; (A.L.); (S.G.B.); (T.C.)
| |
Collapse
|
12
|
Barefield DY, Alvarez-Arce A, Araujo KN. Mechanisms of Sarcomere Protein Mutation-Induced Cardiomyopathies. Curr Cardiol Rep 2023; 25:473-484. [PMID: 37060436 PMCID: PMC11141690 DOI: 10.1007/s11886-023-01876-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 04/16/2023]
Abstract
PURPOSE OF REVIEW The pace of identifying cardiomyopathy-associated mutations and advances in our understanding of sarcomere function that underlies many cardiomyopathies has been remarkable. Here, we aim to synthesize how these advances have led to the promising new treatments that are being developed to treat cardiomyopathies. RECENT FINDINGS The genomics era has identified and validated many genetic causes of hypertrophic and dilated cardiomyopathies. Recent advances in our mechanistic understanding of sarcomere pathophysiology include high-resolution molecular models of sarcomere components and the identification of the myosin super-relaxed state. The advances in our understanding of sarcomere function have yielded several therapeutic agents that are now in development and clinical use to correct contractile dysfunction-mediated cardiomyopathy. New genes linked to cardiomyopathy include targets with limited clinical evidence and require additional investigation. Large portions of cardiomyopathy with family history remain genetically undiagnosed and may be due to polygenic disease.
Collapse
Affiliation(s)
- David Y Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Alejandro Alvarez-Arce
- Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA
| | - Kelly N Araujo
- Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA
| |
Collapse
|
13
|
De Lange WJ, Farrell ET, Hernandez JJ, Stempien A, Kreitzer CR, Jacobs DR, Petty DL, Moss RL, Crone WC, Ralphe JC. cMyBP-C ablation in human engineered cardiac tissue causes progressive Ca2+-handling abnormalities. J Gen Physiol 2023; 155:e202213204. [PMID: 36893011 PMCID: PMC10038829 DOI: 10.1085/jgp.202213204] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 01/02/2023] [Accepted: 02/14/2023] [Indexed: 03/10/2023] Open
Abstract
Truncation mutations in cardiac myosin binding protein C (cMyBP-C) are common causes of hypertrophic cardiomyopathy (HCM). Heterozygous carriers present with classical HCM, while homozygous carriers present with early onset HCM that rapidly progress to heart failure. We used CRISPR-Cas9 to introduce heterozygous (cMyBP-C+/-) and homozygous (cMyBP-C-/-) frame-shift mutations into MYBPC3 in human iPSCs. Cardiomyocytes derived from these isogenic lines were used to generate cardiac micropatterns and engineered cardiac tissue constructs (ECTs) that were characterized for contractile function, Ca2+-handling, and Ca2+-sensitivity. While heterozygous frame shifts did not alter cMyBP-C protein levels in 2-D cardiomyocytes, cMyBP-C+/- ECTs were haploinsufficient. cMyBP-C-/- cardiac micropatterns produced increased strain with normal Ca2+-handling. After 2 wk of culture in ECT, contractile function was similar between the three genotypes; however, Ca2+-release was slower in the setting of reduced or absent cMyBP-C. At 6 wk in ECT culture, the Ca2+-handling abnormalities became more pronounced in both cMyBP-C+/- and cMyBP-C-/- ECTs, and force production became severely depressed in cMyBP-C-/- ECTs. RNA-seq analysis revealed enrichment of differentially expressed hypertrophic, sarcomeric, Ca2+-handling, and metabolic genes in cMyBP-C+/- and cMyBP-C-/- ECTs. Our data suggest a progressive phenotype caused by cMyBP-C haploinsufficiency and ablation that initially is hypercontractile, but progresses to hypocontractility with impaired relaxation. The severity of the phenotype correlates with the amount of cMyBP-C present, with more severe earlier phenotypes observed in cMyBP-C-/- than cMyBP-C+/- ECTs. We propose that while the primary effect of cMyBP-C haploinsufficiency or ablation may relate to myosin crossbridge orientation, the observed contractile phenotype is Ca2+-mediated.
Collapse
Affiliation(s)
- Willem J. De Lange
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Emily T. Farrell
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Jonathan J. Hernandez
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Alana Stempien
- Departments of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline R. Kreitzer
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Derek R. Jacobs
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Dominique L. Petty
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard L. Moss
- Cell and Regenerative Biology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Wendy C. Crone
- Departments of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, USA
- Engineering Physics, University of Wisconsin-Madison, Madison, WI, USA
- Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - J. Carter Ralphe
- Departments of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
14
|
Barefield DY. Is haploinsufficiency a sufficient mechanism for MYBPC3 truncating mutations? J Gen Physiol 2023; 155:e202313351. [PMID: 36946992 PMCID: PMC10072154 DOI: 10.1085/jgp.202313351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
Reduced expression of MYBPC3 causes early dysfunction in human cell culture models prior to reduced cMyBP-C levels.
Collapse
Affiliation(s)
- David Y. Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
15
|
Timmermans S, Vandewalle J, Libert C. Mousepost 2.0, a major expansion of the resource. Nucleic Acids Res 2023; 51:1652-1661. [PMID: 36762471 PMCID: PMC9976886 DOI: 10.1093/nar/gkad064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 02/11/2023] Open
Abstract
The Mousepost 1.0 online search tool, launched in 2017, allowed to search for variations in all protein-coding gene sequences of 36 sequenced mouse inbred strains, compared to the reference strain C57BL/6J, which could be linked to strain-specific phenotypes and modifier effects. Because recently these genome sequences have been significantly updated and sequences of 16 extra strains added by the Mouse Genomes Project, a profound update, correction and expansion of the Mousepost 1.0 database has been performed and is reported here. Moreover, we have added a new class of protein disturbing sequence polymorphisms (besides stop codon losses, stop codon gains, small insertions and deletions, and missense mutations), namely start codon mutations. The current version, Mousepost 2.0 (https://mousepost.be), therefore is a significantly updated and invaluable tool available to the community and is described here and foreseen by multiple examples.
Collapse
Affiliation(s)
- Steven Timmermans
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jolien Vandewalle
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Claude Libert
- VIB Center for Inflammation Research, 9052 Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Da’as SI, Hasan W, Salem R, Younes N, Abdelrahman D, Mohamed IA, Aldaalis A, Temanni R, Mathew LS, Lorenz S, Yacoub M, Nomikos M, Nasrallah GK, Fakhro KA. Transcriptome Profile Identifies Actin as an Essential Regulator of Cardiac Myosin Binding Protein C3 Hypertrophic Cardiomyopathy in a Zebrafish Model. Int J Mol Sci 2022; 23:ijms23168840. [PMID: 36012114 PMCID: PMC9408294 DOI: 10.3390/ijms23168840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 01/15/2023] Open
Abstract
Variants in cardiac myosin-binding protein C (cMyBP-C) are the leading cause of inherited hypertrophic cardiomyopathy (HCM), demonstrating the key role that cMyBP-C plays in the heart’s contractile machinery. To investigate the c-MYBPC3 HCM-related cardiac impairment, we generated a zebrafish mypbc3-knockout model. These knockout zebrafish displayed significant morphological heart alterations related to a significant decrease in ventricular and atrial diameters at systolic and diastolic states at the larval stages. Immunofluorescence staining revealed significant hyperplasia in the mutant’s total cardiac and ventricular cardiomyocytes. Although cardiac contractility was similar to the wild-type control, the ejection fraction was significantly increased in the mypbc3 mutants. At later stages of larval development, the mutants demonstrated an early cardiac phenotype of myocardium remodeling, concurrent cardiomyocyte hyperplasia, and increased ejection fraction as critical processes in HCM initiation to counteract the increased ventricular myocardial wall stress. The examination of zebrafish adults showed a thickened ventricular cardiac wall with reduced heart rate, swimming speed, and endurance ability in both the mypbc3 heterozygous and homozygous groups. Furthermore, heart transcriptome profiling showed a significant downregulation of the actin-filament-based process, indicating an impaired actin cytoskeleton organization as the main dysregulating factor associated with the early ventricular cardiac hypertrophy in the zebrafish mypbc3 HCM model.
Collapse
Affiliation(s)
- Sahar Isa Da’as
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Correspondence:
| | - Waseem Hasan
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Rola Salem
- Health Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Nadine Younes
- Department of Biomedical Sciences, College of Health Science, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Doua Abdelrahman
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Iman A. Mohamed
- Australian Regenerative Medicine Institute, Monash University, Melbourne 3168, Australia
| | - Arwa Aldaalis
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | - Ramzi Temanni
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Lisa Sara Mathew
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Stephan Lorenz
- Integrated Genomics Services, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | | | - Michail Nomikos
- College of Medicine, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
| | - Gheyath K. Nasrallah
- Department of Biomedical Sciences, College of Health Science, Member of QU Health, Qatar University, Doha P.O. Box 2713, Qatar
- Biomedical Research Center, Qatar University, Doha P.O. Box 2713, Qatar
| | - Khalid A. Fakhro
- Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Australian Regenerative Medicine Institute, College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- Weill Cornell Medical College, Doha P.O. Box 24811, Qatar
| |
Collapse
|
17
|
Moore J, Emili A. Mass-Spectrometry-Based Functional Proteomic and Phosphoproteomic Technologies and Their Application for Analyzing Ex Vivo and In Vitro Models of Hypertrophic Cardiomyopathy. Int J Mol Sci 2021; 22:13644. [PMID: 34948439 PMCID: PMC8709159 DOI: 10.3390/ijms222413644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an autosomal dominant disease thought to be principally caused by mutations in sarcomeric proteins. Despite extensive genetic analysis, there are no comprehensive molecular frameworks for how single mutations in contractile proteins result in the diverse assortment of cellular, phenotypic, and pathobiological cascades seen in HCM. Molecular profiling and system biology approaches are powerful tools for elucidating, quantifying, and interpreting dynamic signaling pathways and differential macromolecule expression profiles for a wide range of sample types, including cardiomyopathy. Cutting-edge approaches combine high-performance analytical instrumentation (e.g., mass spectrometry) with computational methods (e.g., bioinformatics) to study the comparative activity of biochemical pathways based on relative abundances of functionally linked proteins of interest. Cardiac research is poised to benefit enormously from the application of this toolkit to cardiac tissue models, which recapitulate key aspects of pathogenesis. In this review, we evaluate state-of-the-art mass-spectrometry-based proteomic and phosphoproteomic technologies and their application to in vitro and ex vivo models of HCM for global mapping of macromolecular alterations driving disease progression, emphasizing their potential for defining the components of basic biological systems, the fundamental mechanistic basis of HCM pathogenesis, and treating the ensuing varied clinical outcomes seen among affected patient cohorts.
Collapse
Affiliation(s)
- Jarrod Moore
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
- MD-PhD Program, Boston University School of Medicine, Boston, MA 02118, USA
| | - Andrew Emili
- Center for Network Systems Biology, Boston University School of Medicine, Boston, MA 02118, USA;
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
18
|
Schwäbe FV, Peter EK, Taft MH, Manstein DJ. Assessment of the Contribution of a Thermodynamic and Mechanical Destabilization of Myosin-Binding Protein C Domain C2 to the Pathomechanism of Hypertrophic Cardiomyopathy-Causing Double Mutation MYBPC3Δ25bp/D389V. Int J Mol Sci 2021; 22:ijms222111949. [PMID: 34769381 PMCID: PMC8584774 DOI: 10.3390/ijms222111949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 01/01/2023] Open
Abstract
Mutations in the gene encoding cardiac myosin-binding protein-C (MyBPC), a thick filament assembly protein that stabilizes sarcomeric structure and regulates cardiac function, are a common cause for the development of hypertrophic cardiomyopathy. About 10% of carriers of the Δ25bp variant of MYBPC3, which is common in individuals from South Asia, are also carriers of the D389V variant on the same allele. Compared with noncarriers and those with MYBPC3Δ25bp alone, indicators for the development of hypertrophic cardiomyopathy occur with increased frequency in MYBPC3Δ25bp/D389V carriers. Residue D389 lies in the IgI-like C2 domain that is part of the N-terminal region of MyBPC. To probe the effects of mutation D389V on structure, thermostability, and protein–protein interactions, we produced and characterized wild-type and mutant constructs corresponding to the isolated 10 kDa C2 domain and a 52 kDa N-terminal fragment that includes subdomains C0 to C2. Our results show marked reductions in the melting temperatures of D389V mutant constructs. Interactions of construct C0–C2 D389V with the cardiac isoforms of myosin-2 and actin remain unchanged. Molecular dynamics simulations reveal changes in the stiffness and conformer dynamics of domain C2 caused by mutation D389V. Our results suggest a pathomechanism for the development of HCM based on the toxic buildup of misfolded protein in young MYBPC3Δ25bp/D389V carriers that is supplanted and enhanced by C-zone haploinsufficiency at older ages.
Collapse
Affiliation(s)
- Frederic V. Schwäbe
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Emanuel K. Peter
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Manuel H. Taft
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
| | - Dietmar J. Manstein
- Fritz Hartmann Centre for Medical Research, Institute for Biophysical Chemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany; (F.V.S.); (E.K.P.); (M.H.T.)
- Division for Structural Biochemistry, Hannover Medical School, Carl Neuberg Str. 1, D-30625 Hannover, Germany
- Correspondence:
| |
Collapse
|
19
|
Pradeep R, Akram A, Proute MC, Kothur NR, Georgiou P, Serhiyenia T, Shi W, Kerolos ME, Mostafa JA. Understanding the Genetic and Molecular Basis of Familial Hypertrophic Cardiomyopathy and the Current Trends in Gene Therapy for Its Management. Cureus 2021; 13:e17548. [PMID: 34646605 PMCID: PMC8481153 DOI: 10.7759/cureus.17548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/28/2021] [Indexed: 01/16/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetically acquired disease of cardiac myocytes. Studies show that 70% of this disease is a result of different mutations in various sarcomere genes. This review aims to discuss several genetic mutations, epigenetic factors, and signal transduction pathways leading to the development of HCM. In addition, this article elaborates on recent advances in gene therapies and their implications for managing this condition. We start by discussing the founding mutations in HCM and their effect on power stroke generation. The less explored field of epigenetics including methylation, acetylation, and the role of different micro RNAs in the development of cardiac muscle hypertrophy has been highlighted in this article. The signal transduction pathways that lead to gene transcription, which in turn lead to increased protein synthesis of cardiac muscle fibers are elaborated. Finally, the microscopic events leading to the pathophysiologic macro events of cardiac failure, and the current experimental trials of gene therapy models, and the clustered regularly interspaced short palindromic repeats (CRISPR) type 2 system proteins, are discussed. We have concluded our discussion by emphasizing the need for more studies on epigenomics and experimental designs for gene therapy in HCM patients. This review focuses on the process of HCM from initial mutation to the development of phenotypic expression and various points of intervention in cardiac myocardial hypertrophy development.
Collapse
Affiliation(s)
- Roshini Pradeep
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Aqsa Akram
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Matthew C Proute
- Family Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nageshwar R Kothur
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Petros Georgiou
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Tatsiana Serhiyenia
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Wangpan Shi
- Pathology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Mina E Kerolos
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Jihan A Mostafa
- Psychiatry/Cognitive Behavioural Psychotherapy, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
20
|
Guo J, Jiang H, Oguntuyo K, Rios B, Boodram Z, Huebsch N. Interplay of Genotype and Substrate Stiffness in Driving the Hypertrophic Cardiomyopathy Phenotype in iPSC-Micro-Heart Muscle Arrays. Cell Mol Bioeng 2021; 14:409-425. [PMID: 34777601 PMCID: PMC8548480 DOI: 10.1007/s12195-021-00684-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/04/2021] [Indexed: 10/21/2022] Open
Abstract
INTRODUCTION In clinical and animal studies, Hypertrophic Cardiomyopathy (HCM) shares many similarities with non-inherited cardiac hypertrophy induced by pressure overload (hypertension). This suggests a potential role for mechanical stress in priming tissues with mutation-induced changes in the sarcomere to develop phenotypes associated with HCM, including hypercontractility and aberrant calcium handling. Here, we tested the hypothesis that heterozygous loss of function of Myosin Binding Protein C (MYBCP3 +/- , mutations in which account for almost 50% of inherited HCM) combines with environmental stiffness to drive HCM phenotypes. METHODS We differentiated isogenic control (WTC) and MYBPC3 +/- iPSC into cardiomyocytes using small molecule manipulation of Wnt signaling, and then purified them using lactate media. The purified cardiomyocytes were seeded into "dog bone" shaped stencil molds to form micro-heart muscle arrays (μHM). To mimic changes in myocardial stiffness stemming from pressure overload, we varied the rigidity of the substrates μHM contract against. Stiffness levels ranged from those corresponding to fetal (5 kPa), healthy (15 kPa), pre-fibrotic (30 kPa) to fibrotic (65 kPa) myocardium. Substrates were embedded with a thin layer of fluorescent beads to track contractile force, and parent iPSC were engineered to express the genetic calcium indicator, GCaMP6f. High speed video microscopy and image analysis were used to quantify calcium handling and contractility of μHM. RESULTS Substrate rigidity triggered physiological adaptation for both genotypes. However, MYBPC3 +/- μHM showed a lower tolerance to substrate stiffness with the peak traction on 15 kPa, while WTC μHM had peak traction on 30 kPa. MYBPC3 +/- μHM exhibited hypercontractility, which was exaggerated by substrate rigidity. MYBPC3 +/- μHM hypercontractility was associated with longer rise times for calcium uptake and force development, along with higher overall Ca2+ intake. CONCLUSION We found MYBPC3 +/- mutations cause iPSC-μHM to exhibit hypercontractility, and also a lower tolerance for mechanical stiffness. Understanding how genetics work in combination with mechanical stiffness to trigger and/or exacerbate pathophysiology may lead to more effective therapies for HCM. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at (10.1007/s12195-021-00684-x).
Collapse
Affiliation(s)
- Jingxuan Guo
- Department of Mechanical Engineering and Material Science, Washington University in Saint Louis, Saint Louis, USA
| | - Huanzhu Jiang
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Kasoorelope Oguntuyo
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Brandon Rios
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Zoë Boodram
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
| | - Nathaniel Huebsch
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, USA
- NSF Science and Technology Center for Engineering Mechanobiology, McKelvey School of Engineering, Saint Louis, USA
- Center for Cardiovascular Research, Center for Regenerative Medicine, Center for Investigation of Membrane Excitability Diseases, Washington University in Saint Louis, Saint Louis, USA
| |
Collapse
|
21
|
Glavaški M, Velicki L. Shared Molecular Mechanisms of Hypertrophic Cardiomyopathy and Its Clinical Presentations: Automated Molecular Mechanisms Extraction Approach. Life (Basel) 2021; 11:life11080785. [PMID: 34440529 PMCID: PMC8398249 DOI: 10.3390/life11080785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/23/2021] [Accepted: 07/30/2021] [Indexed: 12/30/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiovascular disease with a prevalence of 1 in 500 people and varying clinical presentations. Although there is much research on HCM, underlying molecular mechanisms are poorly understood, and research on the molecular mechanisms of its specific clinical presentations is scarce. Our aim was to explore the molecular mechanisms shared by HCM and its clinical presentations through the automated extraction of molecular mechanisms. Molecular mechanisms were congregated by a query of the INDRA database, which aggregates knowledge from pathway databases and combines it with molecular mechanisms extracted from abstracts and open-access full articles by multiple machine-reading systems. The molecular mechanisms were extracted from 230,072 articles on HCM and 19 HCM clinical presentations, and their intersections were found. Shared molecular mechanisms of HCM and its clinical presentations were represented as networks; the most important elements in the intersections’ networks were found, centrality scores for each element of each network calculated, networks with reduced level of noise generated, and cooperatively working elements detected in each intersection network. The identified shared molecular mechanisms represent possible mechanisms underlying different HCM clinical presentations. Applied methodology produced results consistent with the information in the scientific literature.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Correspondence: or
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia;
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| |
Collapse
|
22
|
Suay-Corredera C, Pricolo MR, Velázquez-Carreras D, Pathak D, Nandwani N, Pimenta-Lopes C, Sánchez-Ortiz D, Urrutia-Irazabal I, Vilches S, Dominguez F, Frisso G, Monserrat L, García-Pavía P, de Sancho D, Spudich JA, Ruppel KM, Herrero-Galán E, Alegre-Cebollada J. Nanomechanical Phenotypes in Cardiac Myosin-Binding Protein C Mutants That Cause Hypertrophic Cardiomyopathy. ACS NANO 2021; 15:10203-10216. [PMID: 34060810 PMCID: PMC8514129 DOI: 10.1021/acsnano.1c02242] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a disease of the myocardium caused by mutations in sarcomeric proteins with mechanical roles, such as the molecular motor myosin. Around half of the HCM-causing genetic variants target contraction modulator cardiac myosin-binding protein C (cMyBP-C), although the underlying pathogenic mechanisms remain unclear since many of these mutations cause no alterations in protein structure and stability. As an alternative pathomechanism, here we have examined whether pathogenic mutations perturb the nanomechanics of cMyBP-C, which would compromise its modulatory mechanical tethers across sliding actomyosin filaments. Using single-molecule atomic force spectroscopy, we have quantified mechanical folding and unfolding transitions in cMyBP-C domains targeted by HCM mutations that do not induce RNA splicing alterations or protein thermodynamic destabilization. Our results show that domains containing mutation R495W are mechanically weaker than wild-type at forces below 40 pN and that R502Q mutant domains fold faster than wild-type. None of these alterations are found in control, nonpathogenic variants, suggesting that nanomechanical phenotypes induced by pathogenic cMyBP-C mutations contribute to HCM development. We propose that mutation-induced nanomechanical alterations may be common in mechanical proteins involved in human pathologies.
Collapse
Affiliation(s)
| | - Maria Rosaria Pricolo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Naples, Italy
| | | | - Divya Pathak
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Neha Nandwani
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | | | - David Sánchez-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | | | - Silvia Vilches
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
| | - Fernando Dominguez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
| | - Giulia Frisso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131, Naples, Italy
- CEINGE Biotecnologie Avanzate, scarl, 80145, Naples, Italy
| | | | - Pablo García-Pavía
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, 28222, Madrid, Spain
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN GUARD-HEART, http://guardheart.ern-net.eu/), 28222, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), 28029, Madrid, Spain
- Universidad Francisco de Vitoria (UFV), 28223, Pozuelo de Alarcón, Madrid, Spain
| | - David de Sancho
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika Fakultatea, Euskal Herriko Unibertsitatea UPV/EHU, 20018, Donostia-San Sebastián, Spain
- Donostia International Physics Center (DIPC), 20018, Donostia-San Sebastián, Spain
| | - James A Spudich
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Kathleen M Ruppel
- Department of Biochemistry, Stanford University School of Medicine, Stanford, California 94305, United States
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Elías Herrero-Galán
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | | |
Collapse
|
23
|
Niborski LL, Paces-Fessy M, Ricci P, Bourgeois A, Magalhães P, Kuzma-Kuzniarska M, Lesaulnier C, Reczko M, Declercq E, Zürbig P, Doucet A, Umbhauer M, Cereghini S. Hnf1b haploinsufficiency differentially affects developmental target genes in a new renal cysts and diabetes mouse model. Dis Model Mech 2021; 14:dmm047498. [PMID: 33737325 PMCID: PMC8126479 DOI: 10.1242/dmm.047498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/09/2021] [Indexed: 12/26/2022] Open
Abstract
Heterozygous mutations in HNF1B cause the complex syndrome renal cysts and diabetes (RCAD), characterized by developmental abnormalities of the kidneys, genital tracts and pancreas, and a variety of renal, pancreas and liver dysfunctions. The pathogenesis underlying this syndrome remains unclear as mice with heterozygous null mutations have no phenotype, while constitutive/conditional Hnf1b ablation leads to more severe phenotypes. We generated a novel mouse model carrying an identified human mutation at the intron-2 splice donor site. Unlike heterozygous mice previously characterized, mice heterozygous for the splicing mutation exhibited decreased HNF1B protein levels and bilateral renal cysts from embryonic day 15, originated from glomeruli, early proximal tubules (PTs) and intermediate nephron segments, concurrently with delayed PT differentiation, hydronephrosis and rare genital tract anomalies. Consistently, mRNA sequencing showed that most downregulated genes in embryonic kidneys were primarily expressed in early PTs and the loop of Henle and involved in ion/drug transport, organic acid and lipid metabolic processes, while the expression of previously identified targets upon Hnf1b ablation, including cystic disease genes, was weakly or not affected. Postnatal analyses revealed renal abnormalities, ranging from glomerular cysts to hydronephrosis and, rarely, multicystic dysplasia. Urinary proteomics uncovered a particular profile predictive of progressive decline in kidney function and fibrosis, and displayed common features with a recently reported urine proteome in an RCAD pediatric cohort. Altogether, our results show that reduced HNF1B levels lead to developmental disease phenotypes associated with the deregulation of a subset of HNF1B targets. They further suggest that this model represents a unique clinical/pathological viable model of the RCAD disease.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cell Polarity
- Central Nervous System Diseases/genetics
- Central Nervous System Diseases/pathology
- Cilia/pathology
- Dental Enamel/abnormalities
- Dental Enamel/pathology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Disease Models, Animal
- Embryo, Mammalian/pathology
- Gene Dosage
- Gene Expression Profiling
- Genes, Developmental
- Haploinsufficiency/genetics
- Hepatocyte Nuclear Factor 1-beta/genetics
- Heterozygote
- Humans
- Hydronephrosis/complications
- Kidney Diseases, Cystic/genetics
- Kidney Diseases, Cystic/pathology
- Kidney Glomerulus/pathology
- Kidney Tubules/pathology
- Mice, Inbred C57BL
- Mutation/genetics
- Nephrons/pathology
- RNA Splicing/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Severity of Illness Index
- Mice
Collapse
Affiliation(s)
- Leticia L. Niborski
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Mélanie Paces-Fessy
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Pierbruno Ricci
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Adeline Bourgeois
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Pedro Magalhães
- Mosaiques Diagnostics, 30659 Hannover, Germany
- Department of Pediatric Nephrology, Hannover Medical School, 30625 Hannover, Germany
| | - Maria Kuzma-Kuzniarska
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Celine Lesaulnier
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Martin Reczko
- Biomedical Sciences Research Center Alexander Fleming, Institute for Fundamental Biomedical Science, 16672 Athens, Greece
| | - Edwige Declercq
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | | | - Alain Doucet
- Sorbonne Université, Université Paris Descartes, UMRS 1138, CNRS, ERL 8228, Centre de Recherche des Cordeliers, F-75006 Paris, France
| | - Muriel Umbhauer
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| | - Silvia Cereghini
- Sorbonne Université, CNRS, Institut de Biologie Paris Seine, Laboratoire de Biologie du Développement, IBPS, UMR7622, F-75005 Paris, France
| |
Collapse
|
24
|
Xu F, Chen Y, Tillman KA, Cui Y, Williams RW, Bhattacharya SK, Lu L, Sun Y. Characterizing modifier genes of cardiac fibrosis phenotype in hypertrophic cardiomyopathy. Int J Cardiol 2021; 330:135-141. [PMID: 33529666 PMCID: PMC8105878 DOI: 10.1016/j.ijcard.2021.01.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Clinical phenotypes of hypertrophic cardiomyopathy (HCM) vary greatly even among patients with the same gene mutations. This variability is largely regulated by unidentified modifier loci. The purpose of the study is to identify modifier genes for cardiac fibrosis-a major phenotype of HCM-using the BXD family, a murine cohort. METHODS The relative severity of cardiac fibrosis was estimated by quantitation of cardiac collagen volume fraction (CCVF) across 66 members of the BXD family. Quantitative trait locus (QTL) mapping for cardiac fibrosis was done using GeneNetwork. Candidate modifier loci and genes associated with fibrosis were prioritized based on an explicit scoring system. Networks of correlation between fibrosis and cardiac transcriptomes were evaluated to generate causal models of disease susceptibility. RESULTS CCVF levels varied greatly within this family. Interval mapping identified a significant CCVF-related QTL on chromosome (Chr) 2 in males, and a significant QTL on Chr 4 Mb in females. The scoring system highlighted two strong candidate genes in the Chr 2 locus-Nek6 and Nr6a1. Both genes are highly expressed in the heart. Cardiac Nek6 mRNA levels are significantly correlated with CCVF. Nipsnap3b and Fktn are lead candidate genes for the Chr 4 locus, and both are also highly expressed in heart. Cardiac Nipsnap3b gene expression correlates well with CCVF. CONCLUSION Our study demonstrated that candidate modifier genes of cardiac fibrosis phenotype in HCM are different in males and females. Nek6 and Nr6a1 are strong candidates in males, while Nipsnap3b and Fktn are top candidates in females.
Collapse
Affiliation(s)
- Fuyi Xu
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Kaitlin A Tillman
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Yan Cui
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Robert W Williams
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America
| | - Lu Lu
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America.
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine, Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN, United States of America.
| |
Collapse
|
25
|
Song T, McNamara JW, Ma W, Landim-Vieira M, Lee KH, Martin LA, Heiny JA, Lorenz JN, Craig R, Pinto JR, Irving T, Sadayappan S. Fast skeletal myosin-binding protein-C regulates fast skeletal muscle contraction. Proc Natl Acad Sci U S A 2021; 118:e2003596118. [PMID: 33888578 PMCID: PMC8092462 DOI: 10.1073/pnas.2003596118] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fast skeletal myosin-binding protein-C (fMyBP-C) is one of three MyBP-C paralogs and is predominantly expressed in fast skeletal muscle. Mutations in the gene that encodes fMyBP-C, MYBPC2, are associated with distal arthrogryposis, while loss of fMyBP-C protein is associated with diseased muscle. However, the functional and structural roles of fMyBP-C in skeletal muscle remain unclear. To address this gap, we generated a homozygous fMyBP-C knockout mouse (C2-/-) and characterized it both in vivo and in vitro compared to wild-type mice. Ablation of fMyBP-C was benign in terms of muscle weight, fiber type, cross-sectional area, and sarcomere ultrastructure. However, grip strength and plantar flexor muscle strength were significantly decreased in C2-/- mice. Peak isometric tetanic force and isotonic speed of contraction were significantly reduced in isolated extensor digitorum longus (EDL) from C2-/- mice. Small-angle X-ray diffraction of C2-/- EDL muscle showed significantly increased equatorial intensity ratio during contraction, indicating a greater shift of myosin heads toward actin, while MLL4 layer line intensity was decreased at rest, indicating less ordered myosin heads. Interfilament lattice spacing increased significantly in C2-/- EDL muscle. Consistent with these findings, we observed a significant reduction of steady-state isometric force during Ca2+-activation, decreased myofilament calcium sensitivity, and sinusoidal stiffness in skinned EDL muscle fibers from C2-/- mice. Finally, C2-/- muscles displayed disruption of inflammatory and regenerative pathways, along with increased muscle damage upon mechanical overload. Together, our data suggest that fMyBP-C is essential for maximal speed and force of contraction, sarcomere integrity, and calcium sensitivity in fast-twitch muscle.
Collapse
Affiliation(s)
- Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH 45267
| | - James W McNamara
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH 45267
| | - Weikang Ma
- Biophysics Collaborative Access Team, Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616
| | - Maicon Landim-Vieira
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306
| | - Kyoung Hwan Lee
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Lisa A Martin
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH 45267
| | - Judith A Heiny
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - John N Lorenz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Jose Renato Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306
| | - Thomas Irving
- Biophysics Collaborative Access Team, Department of Biological Sciences, Illinois Institute of Technology, Chicago, IL 60616
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, OH 45267;
| |
Collapse
|
26
|
Oldt RF, Bussey KJ, Settles ML, Fass JN, Roberts JA, Reader JR, Komandoor S, Abrich VA, Kanthaswamy S. MYBPC3 Haplotype Linked to Hypertrophic Cardiomyopathy in Rhesus Macaques ( Macaca mulatta). Comp Med 2020; 70:358-367. [PMID: 32753092 PMCID: PMC7574221 DOI: 10.30802/aalas-cm-19-000108] [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: 11/07/2019] [Revised: 01/13/2020] [Accepted: 02/07/2020] [Indexed: 11/05/2022]
Abstract
In humans, abnormal thickening of the left ventricle of the heart clinically defines hypertrophic cardiomyopathy (HCM), a common inherited cardiovascular disorder that can precede a sudden cardiac death event. The wide range of clinical presentations in HCM obscures genetic variants that may influence an individual's susceptibility to sudden cardiac death. Although exon sequencing of major sarcomere genes can be used to detect high-impact causal mutations, this strategy is successful in only half of patient cases. The incidence of left ventricular hypertrophy (LVH) in a managed research colony of rhesus macaques provides an excellent comparative model in which to explore the genomic etiology of severe HCM and sudden cardiac death. Because no rhesus HCM-associated mutations have been reported, we used a next-generation genotyping assay that targets 7 sarcomeric rhesus genes within 63 genomic sites that are orthologous to human genomic regions known to harbor HCM disease variants. Amplicon sequencing was performed on 52 macaques with confirmed LVH and 42 unrelated, unaffected animals representing both the Indian and Chinese rhesus macaque subspecies. Bias-reduced logistic regression uncovered a risk haplotype in the rhesus MYBPC3 gene, which is frequently disrupted in both human and feline HCM; this haplotype implicates an intronic variant strongly associated with disease in either homozygous or carrier form. Our results highlight that leveraging evolutionary genomic data provides a unique, practical strategy for minimizing population bias in complex disease studies.
Collapse
Affiliation(s)
- Robert F Oldt
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; Evolutionary Biology Graduate Program, School of Life Sciences, Arizona State University at the West Campus, Glendale, Arizona;,
| | - Kimberly J Bussey
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; BEYOND Center for Fundamental Concepts in Science, Arizona State University at the West Campus, Glendale, Arizona
| | - Matthew L Settles
- Bioinformatics Core, UC Davis Genome Center, University of California, Davis, California
| | - Joseph N Fass
- Bioinformatics Core, UC Davis Genome Center, University of California, Davis, California
| | - Jeffrey A Roberts
- California National Primate Research Center, University of California, Davis, California
| | - J Rachel Reader
- California National Primate Research Center, University of California, Davis, California
| | | | - Victor A Abrich
- Division of Cardiovascular Diseases, Mayo Clinic, Scottsdale, Arizona
| | - Sreetharan Kanthaswamy
- School of Mathematical and Natural Sciences, Arizona State University at the West Campus, Glendale, Arizona; Evolutionary Biology Graduate Program, School of Life Sciences, Arizona State University at the West Campus, Glendale, Arizona; California National Primate Research Center, University of California, Davis, California
| |
Collapse
|
27
|
Arif M, Nabavizadeh P, Song T, Desai D, Singh R, Bazrafshan S, Kumar M, Wang Y, Gilbert RJ, Dhandapany PS, Becker RC, Kranias EG, Sadayappan S. Genetic, clinical, molecular, and pathogenic aspects of the South Asian-specific polymorphic MYBPC3 Δ25bp variant. Biophys Rev 2020; 12:1065-1084. [PMID: 32656747 PMCID: PMC7429610 DOI: 10.1007/s12551-020-00725-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by ventricular enlargement, diastolic dysfunction, and increased risk for sudden cardiac death. Sarcomeric genetic defects are the predominant known cause of HCM. In particular, mutations in the myosin-binding protein C gene (MYBPC3) are associated with ~ 40% of all HCM cases in which a genetic basis has been established. A decade ago, our group reported a 25-base pair deletion in intron 32 of MYBPC3 (MYBPC3Δ25bp) that is uniquely prevalent in South Asians and is associated with autosomal dominant cardiomyopathy. Although our studies suggest that this deletion results in left ventricular dysfunction, cardiomyopathies, and heart failure, the precise mechanism by which this variant predisposes to heart disease remains unclear. Increasingly appreciated, however, is the contribution of secondary risk factors, additional mutations, and lifestyle choices in augmenting or modifying the HCM phenotype in MYBPC3Δ25bp carriers. Therefore, the goal of this review article is to summarize the current research dedicated to understanding the molecular pathophysiology of HCM in South Asians with the MYBPC3Δ25bp variant. An emphasis is to review the latest techniques currently applied to explore the MYBPC3Δ25bp pathogenesis and to provide a foundation for developing new diagnostic strategies and advances in therapeutics.
Collapse
Affiliation(s)
- Mohammed Arif
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA.
| | - Pooneh Nabavizadeh
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Taejeong Song
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Darshini Desai
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Rohit Singh
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Sholeh Bazrafshan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Mohit Kumar
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Richard J Gilbert
- Research Service, Providence VA Medical Center, Providence, RI, 02908, USA
| | - Perundurai S Dhandapany
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Biology and Regenerative Medicine (inStem), Bangalore, India
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
- Department of Medicine, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR, USA
| | - Richard C Becker
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati, College of Medicine, Cincinnati, OH, 45267, USA
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, Heart, Lung and Vascular Institute, University of Cincinnati, College of Medicine, 231 Albert Sabin Way, Cincinnati, OH, 45267-0575, USA
| |
Collapse
|
28
|
Chen Y, Xu F, Munkhsaikhan U, Boyle C, Borcky T, Zhao W, Purevjav E, Towbin JA, Liao F, Williams RW, Bhattacharya SK, Lu L, Sun Y. Identifying modifier genes for hypertrophic cardiomyopathy. J Mol Cell Cardiol 2020; 144:119-126. [DOI: 10.1016/j.yjmcc.2020.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
|
29
|
Becker RC, Phillip Owens A, Sadayappan S. The potential roles of Von Willebrand factor and neutrophil extracellular traps in the natural history of hypertrophic and hypertensive cardiomyopathy. Thromb Res 2020; 192:78-87. [PMID: 32460175 DOI: 10.1016/j.thromres.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 02/09/2023]
Abstract
Inflammation is often applied broadly to human disease. Despite its general familiarity, inflammation is highly complex. There are numerous injurious, immune and infectious determinants, functional elements and signaling pathways, ranging from genetic to epigenetic, environmental, racial, molecular and cellular that participate in disease onset and progression, phenotypic heterogeneity, and treatment selection and response. In addition, inflammation can be tissue and organ specific, adding a layer of complexity to achieving a detailed and translatable understanding of its role in health and disease. The following review takes a close look at inflammation in the context of two common heart diseases, hypertrophic cardiomyopathy and hypertensive cardiomyopathy.
Collapse
Affiliation(s)
- Richard C Becker
- Division of Cardiovascular Health and Disease, Heart, Lung and Blood Institute, University of Cincinnati College of Medicine, United States of America.
| | - A Phillip Owens
- Division of Cardiovascular Health and Disease, Heart, Lung and Blood Institute, University of Cincinnati College of Medicine, United States of America
| | - Sakthivel Sadayappan
- Division of Cardiovascular Health and Disease, Heart, Lung and Blood Institute, University of Cincinnati College of Medicine, United States of America
| |
Collapse
|
30
|
Helms AS, Tang VT, O'Leary TS, Friedline S, Wauchope M, Arora A, Wasserman AH, Smith ED, Lee LM, Wen XW, Shavit JA, Liu AP, Previs MJ, Day SM. Effects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy. JCI Insight 2020; 5:133782. [PMID: 31877118 DOI: 10.1172/jci.insight.133782] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022] Open
Abstract
Mutations in cardiac myosin binding protein C (MyBP-C, encoded by MYBPC3) are the most common cause of hypertrophic cardiomyopathy (HCM). Most MYBPC3 mutations result in premature termination codons (PTCs) that cause RNA degradation and a reduction of MyBP-C in HCM patient hearts. However, a reduction in MyBP-C has not been consistently observed in MYBPC3-mutant induced pluripotent stem cell cardiomyocytes (iPSCMs). To determine early MYBPC3 mutation effects, we used patient and genome-engineered iPSCMs. iPSCMs with frameshift mutations were compared with iPSCMs with MYBPC3 promoter and translational start site deletions, revealing that allelic loss of function is the primary inciting consequence of mutations causing PTCs. Despite a reduction in wild-type mRNA in all heterozygous iPSCMs, no reduction in MyBP-C protein was observed, indicating protein-level compensation through what we believe is a previously uncharacterized mechanism. Although homozygous mutant iPSCMs exhibited contractile dysregulation, heterozygous mutant iPSCMs had normal contractile function in the context of compensated MyBP-C levels. Agnostic RNA-Seq analysis revealed differential expression in genes involved in protein folding as the only dysregulated gene set. To determine how MYBPC3-mutant iPSCMs achieve compensated MyBP-C levels, sarcomeric protein synthesis and degradation were measured with stable isotope labeling. Heterozygous mutant iPSCMs showed reduced MyBP-C synthesis rates but a slower rate of MyBP-C degradation. These findings indicate that cardiomyocytes have an innate capacity to attain normal MyBP-C stoichiometry despite MYBPC3 allelic loss of function due to truncating mutations. Modulating MyBP-C degradation to maintain MyBP-C protein levels may be a novel treatment approach upstream of contractile dysfunction for HCM.
Collapse
Affiliation(s)
- Adam S Helms
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Vi T Tang
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas S O'Leary
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, Vermont, USA
| | - Sabrina Friedline
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mick Wauchope
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Akul Arora
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Eric D Smith
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | - Allen P Liu
- Mechanical Engineering.,Biophysics, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael J Previs
- Department of Molecular Physiology & Biophysics, University of Vermont, Burlington, Vermont, USA
| | - Sharlene M Day
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Departments of Molecular and Integrative Physiology
| |
Collapse
|
31
|
Seeger T, Shrestha R, Lam CK, Chen C, McKeithan WL, Lau E, Wnorowski A, McMullen G, Greenhaw M, Lee J, Oikonomopoulos A, Lee S, Yang H, Mercola M, Wheeler M, Ashley EA, Yang F, Karakikes I, Wu JC. A Premature Termination Codon Mutation in MYBPC3 Causes Hypertrophic Cardiomyopathy via Chronic Activation of Nonsense-Mediated Decay. Circulation 2019; 139:799-811. [PMID: 30586709 DOI: 10.1161/circulationaha.118.034624] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) is frequently caused by mutations in myosin-binding protein C3 ( MYBPC3) resulting in a premature termination codon (PTC). The underlying mechanisms of how PTC mutations in MYBPC3 lead to the onset and progression of HCM are poorly understood. This study's aim was to investigate the molecular mechanisms underlying the pathogenesis of HCM associated with MYBPC3 PTC mutations by utilizing human isogenic induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). METHODS Isogenic iPSC lines were generated from HCM patients harboring MYBPC3 PTC mutations (p.R943x; p.R1073P_Fsx4) using genome editing. Comprehensive phenotypic and transcriptome analyses were performed in the iPSC-CMs. RESULTS We observed aberrant calcium handling properties with prolonged decay kinetics and elevated diastolic calcium levels in the absence of structural abnormalities or contracile dysfunction in HCM iPSC-CMs as compared to isogenic controls. The mRNA expression levels of MYBPC3 were significantly reduced in mutant iPSC-CMs, but the protein levels were comparable among isogenic iPSC-CMs, suggesting that haploinsufficiency of MYBPC3 does not contribute to the pathogenesis of HCM in vitro. Furthermore, truncated MYBPC3 peptides were not detected. At the molecular level, the nonsense-mediated decay pathway was activated, and a set of genes involved in major cardiac signaling pathways was dysregulated in HCM iPSC-CMs, indicating an HCM gene signature in vitro. Specific inhibition of the nonsense-mediated decay pathway in mutant iPSC-CMs resulted in reversal of the molecular phenotype and normalization of calcium-handling abnormalities. CONCLUSIONS iPSC-CMs carrying MYBPC3 PTC mutations displayed aberrant calcium signaling and molecular dysregulations in the absence of significant haploinsufficiency of MYBPC3 protein. Here we provided the first evidence of the direct connection between the chronically activated nonsense-mediated decay pathway and HCM disease development.
Collapse
Affiliation(s)
- Timon Seeger
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Rajani Shrestha
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Caressa Chen
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Wesley L McKeithan
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Edward Lau
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Alexa Wnorowski
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Bioengineering (A.W., S.L., F.Y.), Stanford University School of Medicine, CA
| | - George McMullen
- Department of Cardiothoracic Surgery (G.M., M.G., I.K.), Stanford University School of Medicine, CA
| | - Matthew Greenhaw
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Cardiothoracic Surgery (G.M., M.G., I.K.), Stanford University School of Medicine, CA
| | - Jaecheol Lee
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Angelos Oikonomopoulos
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Soah Lee
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA.,Department of Bioengineering (A.W., S.L., F.Y.), Stanford University School of Medicine, CA.,Department of Orthopedic Surgery (S.L.), Stanford University School of Medicine, CA
| | - Huaxiao Yang
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Mark Mercola
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Matthew Wheeler
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Euan A Ashley
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA
| | - Fan Yang
- Department of Bioengineering (A.W., S.L., F.Y.), Stanford University School of Medicine, CA
| | - Ioannis Karakikes
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA
| | - Joseph C Wu
- Stanford Cardiovascular Institute (T.S., R.S., C.K.L., C.C., W.L.M., E.L., A.W., M.G, J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., I.K., J.C.W.), Stanford University School of Medicine, CA.,Department of Medicine, Division of Cardiology (T.S., R.S., C.K.L., C.C., W.L.M., E.L., J.L., A.O., S.L., H.Y., M.M., M.W., E.A.A., J.C.W.), Stanford University School of Medicine, CA.,Institute for Stem Cell Biology and Regenerative Medicine (J.C.W.) Stanford University School of Medicine, CA
| |
Collapse
|
32
|
Kuster DWD, Lynch TL, Barefield DY, Sivaguru M, Kuffel G, Zilliox MJ, Lee KH, Craig R, Namakkal-Soorappan R, Sadayappan S. Altered C10 domain in cardiac myosin binding protein-C results in hypertrophic cardiomyopathy. Cardiovasc Res 2019; 115:1986-1997. [PMID: 31050699 PMCID: PMC6872972 DOI: 10.1093/cvr/cvz111] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/04/2019] [Accepted: 04/25/2019] [Indexed: 12/12/2022] Open
Abstract
AIMS A 25-base pair deletion in the cardiac myosin binding protein-C (cMyBP-C) gene (MYBPC3), proposed to skip exon 33, modifies the C10 domain (cMyBP-CΔC10mut) and is associated with hypertrophic cardiomyopathy (HCM) and heart failure, affecting approximately 100 million South Asians. However, the molecular mechanisms underlying the pathogenicity of cMyBP-CΔC10mutin vivo are unknown. We hypothesized that expression of cMyBP-CΔC10mut exerts a poison polypeptide effect leading to improper assembly of cardiac sarcomeres and the development of HCM. METHODS AND RESULTS To determine whether expression of cMyBP-CΔC10mut is sufficient to cause HCM and contractile dysfunction in vivo, we generated transgenic (TG) mice having cardiac-specific protein expression of cMyBP-CΔC10mut at approximately half the level of endogenous cMyBP-C. At 12 weeks of age, significant hypertrophy was observed in TG mice expressing cMyBP-CΔC10mut (heart weight/body weight ratio: 4.43 ± 0.11 mg/g non-transgenic (NTG) vs. 5.34 ± 0.25 mg/g cMyBP-CΔC10mut, P < 0.05). Furthermore, haematoxylin and eosin, Masson's trichrome staining, as well as second-harmonic generation imaging revealed the presence of significant fibrosis and a greater relative nuclear area in cMyBP-CΔC10mut hearts compared with NTG controls. M-mode echocardiography analysis revealed hypercontractile hearts (EF: 53.4%±2.9% NTG vs. 66.4% ± 4.7% cMyBP-CΔC10mut; P < 0.05) and early diastolic dysfunction (E/E': 28.7 ± 3.7 NTG vs. 46.3 ± 8.4 cMyBP-CΔC10mut; P < 0.05), indicating the presence of an HCM phenotype. To assess whether these changes manifested at the myofilament level, contractile function of single skinned cardiomyocytes was measured. Preserved maximum force generation and increased Ca2+-sensitivity of force generation were observed in cardiomyocytes from cMyBP-CΔC10mut mice compared with NTG controls (EC50: 3.6 ± 0.02 µM NTG vs. 2.90 ± 0.01 µM cMyBP-CΔC10mut; P < 0.0001). CONCLUSION Expression of cMyBP-C protein with a modified C10 domain is sufficient to cause contractile dysfunction and HCM in vivo.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/pathology
- Cardiomyopathy, Hypertrophic/physiopathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Disease Models, Animal
- Fibrosis
- Gene Expression Regulation
- Gene Regulatory Networks
- Genetic Predisposition to Disease
- Mice, Transgenic
- Mutation
- Myocardial Contraction
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Protein Domains
- Sarcomeres/genetics
- Sarcomeres/metabolism
- Sarcomeres/pathology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Remodeling
Collapse
Affiliation(s)
- Diederik W D Kuster
- Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
- Department of Physiology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam Cardiovascular Sciences, Amsterdam, Netherlands
| | - Thomas L Lynch
- Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - David Y Barefield
- Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - Mayandi Sivaguru
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Gina Kuffel
- Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | | | - Kyoung Hwan Lee
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Roger Craig
- Division of Cell Biology and Imaging, Department of Radiology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Rajasekaran Namakkal-Soorappan
- Molecular and Cellular Pathology, Department of Pathology, School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sakthivel Sadayappan
- Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, 231 Albert Sabin Way, Cincinnati, OH, USA
| |
Collapse
|
33
|
Tabish AM, Arif M, Song T, Elbeck Z, Becker RC, Knöll R, Sadayappan S. Association of intronic DNA methylation and hydroxymethylation alterations in the epigenetic etiology of dilated cardiomyopathy. Am J Physiol Heart Circ Physiol 2019; 317:H168-H180. [PMID: 31026178 PMCID: PMC6692731 DOI: 10.1152/ajpheart.00758.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/15/2019] [Accepted: 04/22/2019] [Indexed: 01/03/2023]
Abstract
In this study, we investigated the role of DNA methylation [5-methylcytosine (5mC)] and 5-hydroxymethylcytosine (5hmC), epigenetic modifications that regulate gene activity, in dilated cardiomyopathy (DCM). A MYBPC3 mutant mouse model of DCM was compared with wild type and used to profile genomic 5mC and 5hmC changes by Chip-seq, and gene expression levels were analyzed by RNA-seq. Both 5mC-altered genes (957) and 5hmC-altered genes (2,022) were identified in DCM hearts. Diverse gene ontology and KEGG pathways were enriched for DCM phenotypes, such as inflammation, tissue fibrosis, cell death, cardiac remodeling, cardiomyocyte growth, and differentiation, as well as sarcomere structure. Hierarchical clustering of mapped genes affected by 5mC and 5hmC clearly differentiated DCM from wild-type phenotype. Based on these data, we propose that genomewide 5mC and 5hmC contents may play a major role in DCM pathogenesis. NEW & NOTEWORTHY Our data demonstrate that development of dilated cardiomyopathy in mice is associated with significant epigenetic changes, specifically in intronic regions, which, when combined with gene expression profiling data, highlight key signaling pathways involved in pathological cardiac remodeling and heart contractile dysfunction.
Collapse
Affiliation(s)
- Ali M Tabish
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Mohammed Arif
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Taejeong Song
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Zaher Elbeck
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
| | - Richard C Becker
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| | - Ralph Knöll
- Integrated Cardio-Metabolic Centre, Karolinska Institutet , Stockholm , Sweden
- Cardiovascular and Metabolic Disease Innovative Medicines and Early Development Unit, AstraZeneca R&D, Gothenburg , Sweden
| | - Sakthivel Sadayappan
- Heart, Lung, Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati , Cincinnati, Ohio
| |
Collapse
|
34
|
Tonino P, Kiss B, Gohlke J, Smith JE, Granzier H. Fine mapping titin's C-zone: Matching cardiac myosin-binding protein C stripes with titin's super-repeats. J Mol Cell Cardiol 2019; 133:47-56. [PMID: 31158359 DOI: 10.1016/j.yjmcc.2019.05.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/22/2019] [Accepted: 05/30/2019] [Indexed: 01/04/2023]
Abstract
Titin is largely comprised of serially-linked immunoglobulin (Ig) and fibronectin type-III (Fn3) domains. Many of these domains are arranged in an 11 domain super-repeat pattern that is repeated 11 times, forming the so-named titin C-zone in the A-band region of the sarcomere. Each super-repeat is thought to provide binding sites for thick filament proteins, such as cMyBP-C (cardiac myosin-binding protein C). However, it remains to be established which of titin's 11 C-zone super-repeats anchor cMyBP-C as titin contains 11 super-repeats and cMyBP-C is found in 9 stripes only. To study the layout of titin's C-zone in relation to MyBP-C, immunolabeling studies were performed on mouse skinned myocardium with antibodies to titin and cMyBP-C, using both immuno-electron microscopy and super-resolution optical microscopy. Results indicate that cMyBP-C locates near the interface between titin's C-zone super-repeats. Studies on a mouse model in which two of titin's C-zone repeats have been genetically deleted support that the first Ig domain of a super-repeat is important for anchoring cMyBP-C but also Fn3 domains located at the end of the preceding repeat. Furthermore, not all super-repeat interfaces are equal as the interface between super-repeat 1 and 2 (close to titin's D-zone) does not contain cMyBP-C. Finally, titin's C-zone does not extend all the way to the bare zone but instead terminates at the level of the second myosin crown. This study enhances insights in the molecular layout of the C-zone of titin, its relation to cMyBP-C, and its possible roles in cardiomyopathies.
Collapse
Affiliation(s)
- Paola Tonino
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Balazs Kiss
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - John E Smith
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA; Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
35
|
Barefield DY, McNamara JW, Lynch TL, Kuster DWD, Govindan S, Haar L, Wang Y, Taylor EN, Lorenz JN, Nieman ML, Zhu G, Luther PK, Varró A, Dobrev D, Ai X, Janssen PML, Kass DA, Jones WK, Gilbert RJ, Sadayappan S. Ablation of the calpain-targeted site in cardiac myosin binding protein-C is cardioprotective during ischemia-reperfusion injury. J Mol Cell Cardiol 2019; 129:236-246. [PMID: 30862451 PMCID: PMC7222036 DOI: 10.1016/j.yjmcc.2019.03.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/07/2019] [Accepted: 03/08/2019] [Indexed: 12/31/2022]
Abstract
Cardiac myosin binding protein-C (cMyBP-C) phosphorylation is essential for normal heart function and protects the heart from ischemia-reperfusion (I/R) injury. It is known that protein kinase-A (PKA)-mediated phosphorylation of cMyBP-C prevents I/R-dependent proteolysis, whereas dephosphorylation of cMyBP-C at PKA sites correlates with its degradation. While sites on cMyBP-C associated with phosphorylation and proteolysis co-localize, the mechanisms that link cMyBP-C phosphorylation and proteolysis during cardioprotection are not well understood. Therefore, we aimed to determine if abrogation of cMyBP-C proteolysis in association with calpain, a calcium-activated protease, confers cardioprotection during I/R injury. Calpain is activated in both human ischemic heart samples and ischemic mouse myocardium where cMyBP-C is dephosphorylated and undergoes proteolysis. Moreover, cMyBP-C is a substrate for calpain proteolysis and cleaved by calpain at residues 272-TSLAGAGRR-280, a domain termed as the calpain-target site (CTS). Cardiac-specific transgenic (Tg) mice in which the CTS motif was ablated were bred into a cMyBP-C null background. These Tg mice were conclusively shown to possess a normal basal structure and function by analysis of histology, electron microscopy, immunofluorescence microscopy, Q-space MRI of tissue architecture, echocardiography, and hemodynamics. However, the genetic ablation of the CTS motif conferred resistance to calpain-mediated proteolysis of cMyBP-C. Following I/R injury, the loss of the CTS reduced infarct size compared to non-transgenic controls. Collectively, these findings demonstrate the physiological significance of calpain-targeted cMyBP-C proteolysis and provide a rationale for studying inhibition of calpain-mediated proteolysis of cMyBP-C as a therapeutic target for cardioprotection.
Collapse
Affiliation(s)
- David Y Barefield
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA; Center for Genetic Medicine, Northwestern University, Chicago, IL, USA.
| | - James W McNamara
- Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Thomas L Lynch
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA; Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Diederik W D Kuster
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA; Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, the Netherlands
| | - Suresh Govindan
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA
| | - Lauren Haar
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Yang Wang
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Erik N Taylor
- Department of Physiology and Biophysics, Boston University, Boston, MA, USA
| | - John N Lorenz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michelle L Nieman
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Pradeep K Luther
- Molecular Medicine Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Andras Varró
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Xun Ai
- Department of Physiology and Biophysics, Rush University, Chicago, IL, USA
| | - Paul M L Janssen
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Walter Keith Jones
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Richard J Gilbert
- Research Service, Providence VA Medical Center and Brown University, Providence, RI, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Loyola University Chicago, Maywood, IL, USA; Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
36
|
Thompson BR, Soller KJ, Vetter A, Yang J, Veglia G, Bowser MT, Metzger JM. Cytoplasmic nucleic acid-based XNAs directly enhance live cardiac cell function by a Ca 2+ cycling-independent mechanism via the sarcomere. J Mol Cell Cardiol 2019; 130:1-9. [PMID: 30849419 DOI: 10.1016/j.yjmcc.2019.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/05/2019] [Accepted: 02/27/2019] [Indexed: 11/19/2022]
Abstract
Nucleic acid - protein interactions are critical for regulating gene activation in the nucleus. In the cytoplasm, however, potential nucleic acid-protein functional interactions are less clear. The emergence of a large and expanding number of non-coding RNAs and DNA fragments raises the possibility that the cytoplasmic nucleic acids may interact with cytoplasmic cellular components to directly alter key biological processes within the cell. We now show that both natural and synthetic nucleic acids, collectively XNAs, when introduced to the cytoplasm of live cell cardiac myocytes, markedly enhance contractile function via a mechanism that is independent of new translation, activation of the TLR-9 pathway or by altered intracellular Ca2+ cycling. Findings show a steep XNA oligo length-dependence, but not sequence dependence or nucleic acid moiety dependence, for cytoplasmic XNAs to hasten myocyte relaxation. XNAs localized to the sarcomere in a striated pattern and bound the cardiac troponin regulatory complex with high affinity in an electrostatic-dependent manner. Mechanistically, XNAs phenocopy PKA-based modified troponin to cause faster relaxation. Collectively, these data support a new role for cytoplasmic nucleic acids in directly modulating live cell cardiac performance and raise the possibility that cytoplasmic nucleic acid - protein interactions may alter functionally relevant pathways in other cell types.
Collapse
Affiliation(s)
- Brian R Thompson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Kailey J Soller
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Anthony Vetter
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Jing Yang
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Michael T Bowser
- Department of Chemistry, University of Minnesota, Minneapolis, MN, United States of America
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN, United States of America.
| |
Collapse
|
37
|
O'Leary TS, Snyder J, Sadayappan S, Day SM, Previs MJ. MYBPC3 truncation mutations enhance actomyosin contractile mechanics in human hypertrophic cardiomyopathy. J Mol Cell Cardiol 2018; 127:165-173. [PMID: 30550750 DOI: 10.1016/j.yjmcc.2018.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 01/15/2023]
Abstract
RATIONALE Truncation mutations in the MYBPC3 gene, encoding for cardiac myosin-binding protein C (MyBP-C), are the leading cause of hypertrophic cardiomyopathy (HCM). Whole heart, fiber and molecular studies demonstrate that MyBP-C is a potent modulator of cardiac contractility, but how these mutations contribute to HCM is unresolved. OBJECTIVES To readdress whether MYBPC3 truncation mutations result in loss of MyBP-C content and/or the expression of truncated MyBP-C from the mutant allele and determine how these mutations effect myofilament sliding in human myocardium. METHODS AND RESULTS Septal wall tissue samples were obtained from HCM patients undergoing myectomy (n = 18) and donor controls (n = 8). The HCM samples contained 40% less MyBP-C and reduced levels of MyBP-C phosphorylation, when compared to the donor control samples using quantitative mass spectrometry. These differences occurred in the absence of changes in the stoichiometry of other myofilament proteins or production of truncated MyBP-C from the mutant MYBPC3 allele. The functional impact of MYBPC3 truncation mutations on myofilament sliding was determined using a total internal reflection microscopy (TIRFM) single particle assay. Myosin-thick filaments containing their native complement of MyBP-C, and actin-thin filaments decorated with the troponin/tropomyosin calcium regulatory proteins, were isolated from a subgroup of the HCM (n = 4) and donor (n = 5) heart samples. The maximal sliding velocity of native thin filaments was enhanced within the C-zones of the native thick filaments isolated from the HCM samples, when compared to velocity within the C-zones of thick filaments isolated from the donor samples. Analytical modeling demonstrated that the 40% reduction in MyBP-C content was sufficient to enhance the myofilament sliding velocity, as observed in the TIRFM assay. CONCLUSIONS HCM-causing MYBPC3 truncation mutations result in a loss of MyBP-C content that enhances maximal myofilament sliding velocities, only where MyBP-C is localized within the C-zone. These findings support therapeutic rationale for restoring normal levels of MyBP-C and/or dampening maximal contractile velocities for the treatment of human HCM.
Collapse
Affiliation(s)
- Thomas S O'Leary
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute of Vermont, University of Vermont, Burlington, VT, United States
| | - Julia Snyder
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute of Vermont, University of Vermont, Burlington, VT, United States
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Division of Cardiovascular Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Sharlene M Day
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Michael J Previs
- Department of Molecular Physiology and Biophysics, Cardiovascular Research Institute of Vermont, University of Vermont, Burlington, VT, United States.
| |
Collapse
|
38
|
Allelic imbalance and haploinsufficiency in MYBPC3-linked hypertrophic cardiomyopathy. Pflugers Arch 2018; 471:781-793. [PMID: 30456444 DOI: 10.1007/s00424-018-2226-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/04/2018] [Accepted: 10/17/2018] [Indexed: 01/04/2023]
Abstract
Mutations in cardiac myosin binding protein C (MYBPC3) represent the most frequent cause of familial hypertrophic cardiomyopathy (HCM), making up approximately 50% of identified HCM mutations. MYBPC3 is distinct among other sarcomere genes associated with HCM in that truncating mutations make up the vast majority, whereas nontruncating mutations predominant in other sarcomere genes. Several studies using myocardial tissue from HCM patients have found reduced abundance of wild-type MYBPC3 compared to control hearts, suggesting haploinsufficiency of full-length MYBPC3. Further, decreased mutant versus wild-type mRNA and lack of truncated mutant MYBPC3 protein has been demonstrated, highlighting the presence of allelic imbalance. In this review, we will begin by introducing allelic imbalance and haploinsufficiency, highlighting the broad role each plays within the spectrum of human disease. We will subsequently focus on the roles allelic imbalance and haploinsufficiency play within MYBPC3-linked HCM. Finally, we will explore the implications of these findings on future directions of HCM research. An improved understanding of allelic imbalance and haploinsufficiency may help us better understand genotype-phenotype relationships in HCM and develop novel targeted therapies, providing exciting future research opportunities.
Collapse
|
39
|
Dvornikov AV, de Tombe PP, Xu X. Phenotyping cardiomyopathy in adult zebrafish. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2018; 138:116-125. [PMID: 29884423 PMCID: PMC6269218 DOI: 10.1016/j.pbiomolbio.2018.05.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 04/26/2018] [Accepted: 05/29/2018] [Indexed: 12/21/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is usually manifested by increased myofilament Ca2+ sensitivity, excessive contractility, and impaired relaxation. In contrast, dilated cardiomyopathy (DCM) originates from insufficient sarcomere contractility and reduced cardiac pump function, subsequently resulting in heart failure. The zebrafish has emerged as a new model of human cardiomyopathy with high-throughput screening, which will facilitate the discovery of novel genetic factors and the development of new therapies. Given the small hearts of zebrafish, better phenotyping tools are needed to discern different types of cardiomyopathy, such as HCM and DCM. This article reviews the existing models of cardiomyopathy, available morphologic and functional methods, and current understanding of the different types of cardiomyopathy in adult zebrafish.
Collapse
Affiliation(s)
- Alexey V Dvornikov
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Pieter P de Tombe
- University of Illinois at Chicago, Department of Physiology and Biophysics, Chicago, IL, USA; Magdi Yacoub Institute, Cardiac Biophysics Division, Harefield, UK; Imperial College, Heart and Lung Institute, London, UK; Freiburg University, Institute for Experimental Cardiovascular Medicine, Germany
| | - Xiaolei Xu
- Department of Biochemistry and Molecular Biology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
40
|
Abstract
Hypertrophic cardiomyopathy (HCM) is most commonly transmitted as an autosomal dominant trait, caused by mutations in genes encoding cardiac sarcomere proteins1–3. Other inheritable causes of the disease include mutations in genes coding for proteins important in calcium handling or that form part of the cytoskeleton4–6. At present, the primary clinical role of genetic testing in HCM is to facilitate familial screening to allow the identification of individuals at risk of developing the disease7,8. It is also used to diagnose genocopies, such as lysosomal9–11 and glycogen storage disease which have different treatment strategies, rates of disease progression and prognosis12–14. The role of genetic testing in predicting prognosis is limited at present, but emerging data suggest that knowledge of the genetic basis of disease will assume an important role in disease stratification15–17 and offer potential targets for disease-modifying therapy in the near future18.
Collapse
Affiliation(s)
- Mohammed Akhtar
- Bart's Heart Centre, St Bartholomew's Hospital, London & University College London
| | - Perry Elliott
- Bart's Heart Centre, St Bartholomew's Hospital, London & University College London
| |
Collapse
|
41
|
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: 48] [Impact Index Per Article: 6.9] [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
|
42
|
Grimes KM, Barefield DY, Kumar M, McNamara JW, Weintraub ST, de Tombe PP, Sadayappan S, Buffenstein R. The naked mole-rat exhibits an unusual cardiac myofilament protein profile providing new insights into heart function of this naturally subterranean rodent. Pflugers Arch 2017; 469:1603-1613. [PMID: 28780592 PMCID: PMC5856255 DOI: 10.1007/s00424-017-2046-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/27/2017] [Accepted: 07/23/2017] [Indexed: 02/08/2023]
Abstract
The long-lived, hypoxic-tolerant naked mole-rat well-maintains cardiac function over its three-decade-long lifespan and exhibits many cardiac features atypical of similar-sized laboratory rodents. For example, they exhibit low heart rates and resting cardiac contractility, yet have a large cardiac reserve. These traits are considered ecophysiological adaptations to their dank subterranean atmosphere of low oxygen and high carbon dioxide levels and may also contribute to negligible declines in cardiac function during aging. We asked if naked mole-rats had a different myofilament protein signature to that of similar-sized mice that commonly show both high heart rates and high basal cardiac contractility. Adult mouse ventricles predominantly expressed α-myosin heavy chain (97.9 ± 0.4%). In contrast, and more in keeping with humans, β myosin heavy chain was the dominant isoform (79.0 ± 2.0%) in naked mole-rat ventricles. Naked mole-rat ventricles diverged from those of both humans and mice, as they expressed both cardiac and slow skeletal isoforms of troponin I. This myofilament protein profile is more commonly observed in mice in utero and during cardiomyopathies. There were no species differences in phosphorylation of cardiac myosin binding protein-C or troponin I. Phosphorylation of both ventricular myosin light chain 2 and cardiac troponin T in naked mole-rats was approximately half that observed in mice. Myofilament function was also compared between the two species using permeabilized cardiomyocytes. Together, these data suggest a cardiac myofilament protein signature that may contribute to the naked mole-rat's suite of adaptations to its natural subterranean habitat.
Collapse
Affiliation(s)
- Kelly M Grimes
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - David Y Barefield
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
- Center for Genetic Medicine, Northwestern University, Chicago, IL, USA
| | - Mohit Kumar
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| | - James W McNamara
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Susan T Weintraub
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Pieter P de Tombe
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, IL, USA
- Heart, Lung and Vascular Institute, University of Cincinnati, Cincinnati, OH, USA
| | - Rochelle Buffenstein
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Sam and Ann Barshop Institute for Aging and Longevity Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
- Calico Life Sciences, 1170 Veterans Blvd, South San Francisco, CA, 94080, USA.
| |
Collapse
|
43
|
Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 2017; 121:749-770. [PMID: 28912181 DOI: 10.1161/circresaha.117.311059] [Citation(s) in RCA: 809] [Impact Index Per Article: 101.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disorder that is characterized by left ventricular hypertrophy unexplained by secondary causes and a nondilated left ventricle with preserved or increased ejection fraction. It is commonly asymmetrical with the most severe hypertrophy involving the basal interventricular septum. Left ventricular outflow tract obstruction is present at rest in about one third of the patients and can be provoked in another third. The histological features of HCM include myocyte hypertrophy and disarray, as well as interstitial fibrosis. The hypertrophy is also frequently associated with left ventricular diastolic dysfunction. In the majority of patients, HCM has a relatively benign course. However, HCM is also an important cause of sudden cardiac death, particularly in adolescents and young adults. Nonsustained ventricular tachycardia, syncope, a family history of sudden cardiac death, and severe cardiac hypertrophy are major risk factors for sudden cardiac death. This complication can usually be averted by implantation of a cardioverter-defibrillator in appropriate high-risk patients. Atrial fibrillation is also a common complication and is not well tolerated. Mutations in over a dozen genes encoding sarcomere-associated proteins cause HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin-binding protein C, respectively, are the 2 most common genes involved, together accounting for ≈50% of the HCM families. In ≈40% of HCM patients, the causal genes remain to be identified. Mutations in genes responsible for storage diseases also cause a phenotype resembling HCM (genocopy or phenocopy). The routine applications of genetic testing and preclinical identification of family members represents an important advance. The genetic discoveries have enhanced understanding of the molecular pathogenesis of HCM and have stimulated efforts designed to identify new therapeutic agents.
Collapse
Affiliation(s)
- Ali J Marian
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.).
| | - Eugene Braunwald
- From the Center for Cardiovascular Genetics, Institute of Molecular Medicine, Department of Medicine, University of Texas Health Sciences Center at Houston (A.J.M.); Texas Heart Institute, Houston (A.J.M.); and TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (E.B.)
| |
Collapse
|
44
|
Barefield DY, Puckelwartz MJ, Kim EY, Wilsbacher LD, Vo AH, Waters EA, Earley JU, Hadhazy M, Dellefave-Castillo L, Pesce LL, McNally EM. Experimental Modeling Supports a Role for MyBP-HL as a Novel Myofilament Component in Arrhythmia and Dilated Cardiomyopathy. Circulation 2017; 136:1477-1491. [PMID: 28778945 DOI: 10.1161/circulationaha.117.028585] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 07/21/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Cardiomyopathy and arrhythmias are under significant genetic influence. Here, we studied a family with dilated cardiomyopathy and associated conduction system disease in whom prior clinical cardiac gene panel testing was unrevealing. METHODS Whole-genome sequencing and induced pluripotent stem cells were used to examine a family with dilated cardiomyopathy and atrial and ventricular arrhythmias. We also characterized a mouse model with heterozygous and homozygous deletion of Mybphl. RESULTS Whole-genome sequencing identified a premature stop codon, R255X, in the MYBPHL gene encoding MyBP-HL (myosin-binding protein-H like), a novel member of the myosin-binding protein family. MYBPHL was found to have high atrial expression with low ventricular expression. We determined that MyBP-HL protein was myofilament associated in the atria, and truncated MyBP-HL protein failed to incorporate into the myofilament. Human cell modeling demonstrated reduced expression from the mutant MYBPHL allele. Echocardiography of Mybphl heterozygous and null mouse hearts exhibited a 36% reduction in fractional shortening and an increased diastolic ventricular chamber size. Atria weight normalized to total heart weight was significantly increased in Mybphl heterozygous and null mice. Using a reporter system, we detected robust expression of Mybphl in the atria, and in discrete puncta throughout the right ventricular wall and septum, as well. Telemetric electrocardiogram recordings in Mybphl mice revealed cardiac conduction system abnormalities with aberrant atrioventricular conduction and an increased rate of arrhythmia in heterozygous and null mice. CONCLUSIONS The findings of reduced ventricular function and conduction system defects in Mybphl mice support that MYBPHL truncations may increase risk for human arrhythmias and cardiomyopathy.
Collapse
Affiliation(s)
- David Y Barefield
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Megan J Puckelwartz
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Ellis Y Kim
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Lisa D Wilsbacher
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Andy H Vo
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Emily A Waters
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Judy U Earley
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Michele Hadhazy
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Lisa Dellefave-Castillo
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Lorenzo L Pesce
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.)
| | - Elizabeth M McNally
- From Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL (D.Y.B., M.J.P., J.U.E., M.H., L.D.-C., E.M.M.); Molecular Pathogenesis and Molecular Medicine, University of Chicago, IL (E.Y.K.); Feinberg Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL (L.D.W.); Committee on Development, Regeneration and Stem Cell Biology, University of Chicago, IL (A.H.V.); Northwestern University Center for Advanced Molecular Imaging, Evanston, IL (E.A.W.); and Computation Institute, University of Chicago, IL (L.L.P.).
| |
Collapse
|
45
|
Burke MA, Cook SA, Seidman JG, Seidman CE. Clinical and Mechanistic Insights Into the Genetics of Cardiomyopathy. J Am Coll Cardiol 2017; 68:2871-2886. [PMID: 28007147 DOI: 10.1016/j.jacc.2016.08.079] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 08/01/2016] [Accepted: 08/02/2016] [Indexed: 12/19/2022]
Abstract
Over the last quarter-century, there has been tremendous progress in genetics research that has defined molecular causes for cardiomyopathies. More than a thousand mutations have been identified in many genes with varying ontologies, therein indicating the diverse molecules and pathways that cause hypertrophic, dilated, restrictive, and arrhythmogenic cardiomyopathies. Translation of this research to the clinic via genetic testing can precisely group affected patients according to molecular etiology, and identify individuals without evidence of disease who are at high risk for developing cardiomyopathy. These advances provide insights into the earliest manifestations of cardiomyopathy and help to define the molecular pathophysiological basis for cardiac remodeling. Although these efforts remain incomplete, new genomic technologies and analytic strategies provide unparalleled opportunities to fully explore the genetic architecture of cardiomyopathies. Such data hold the promise that mutation-specific pathophysiology will uncover novel therapeutic targets, and herald the beginning of precision therapy for cardiomyopathy patients.
Collapse
Affiliation(s)
- Michael A Burke
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia; Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Stuart A Cook
- National Heart & Lung Institute, Imperial College London, London, United Kingdom; National Heart Centre Singapore, Singapore; Duke-National University of Singapore, Singapore
| | | | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase, Maryland.
| |
Collapse
|
46
|
Mohamed IA, Krishnamoorthy NT, Nasrallah GK, Da'as SI. The Role of Cardiac Myosin Binding Protein C3 in Hypertrophic Cardiomyopathy-Progress and Novel Therapeutic Opportunities. J Cell Physiol 2017; 232:1650-1659. [PMID: 27731493 DOI: 10.1002/jcp.25639] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/07/2016] [Indexed: 11/11/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a common autosomal dominant genetic cardiovascular disorder marked by genetic and phenotypic heterogeneity. Mutations in the gene encodes the cardiac myosin-binding protein C, cMYBPC3 is amongst the various sarcomeric genes that are associated with HCM. These mutations produce mutated mRNAs and truncated cMyBP-C proteins. In this review, we will discuss the implications and molecular mechanisms involved in MYBPC3 different mutations. Further, we will highlight the novel targets that can be developed into potential therapeutics for the treatment of HMC. J. Cell. Physiol. 232: 1650-1659, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Iman A Mohamed
- Department of Biomedical Science, Zewail City of Science and Technology, Giza, Egypt
| | - Navaneethakrishnan T Krishnamoorthy
- Division of Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Heart Science Centre, National Heart and Lung Institute, Imperial College London, London, UK
| | - Gheyath K Nasrallah
- Department of Biomedical Science, College of Health Science, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar
| | - Sahar I Da'as
- Division of Experimental Genetics, Sidra Medical and Research Center, Doha, Qatar.,Department of Biomedical and Biological Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
47
|
Monteiro da Rocha A, Guerrero-Serna G, Helms A, Luzod C, Mironov S, Russell M, Jalife J, Day SM, Smith GD, Herron TJ. Deficient cMyBP-C protein expression during cardiomyocyte differentiation underlies human hypertrophic cardiomyopathy cellular phenotypes in disease specific human ES cell derived cardiomyocytes. J Mol Cell Cardiol 2016; 99:197-206. [PMID: 27620334 PMCID: PMC5609478 DOI: 10.1016/j.yjmcc.2016.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/18/2016] [Accepted: 09/08/2016] [Indexed: 02/07/2023]
Abstract
AIMS Mutations of cardiac sarcomere genes have been identified to cause HCM, but the molecular mechanisms that lead to cardiomyocyte hypertrophy and risk for sudden death are uncertain. The aim of this study was to examine HCM disease mechanisms at play during cardiac differentiation of human HCM specific pluripotent stem cells. METHODS AND RESULTS We generated a human embryonic stem cell (hESC) line carrying a naturally occurring mutation of MYPBC3 (c.2905 +1 G >A) to study HCM pathogenesis during cardiac differentiation. HCM-specific hESC-derived cardiomyocytes (hESC-CMs) displayed hallmark aspects of HCM including sarcomere disarray, hypertrophy and impaired calcium impulse propagation. HCM hESC-CMs presented a transient haploinsufficiency of cMyBP-C during cardiomyocyte differentiation, but by day 30 post-differentiation cMyBP-C levels were similar to control hESC-CMs. Gene transfer of full-length MYBPC3 during differentiation prevented hypertrophy, sarcomere disarray and improved calcium impulse propagation in HCM hESC-CMs. CONCLUSION(S) These findings point to the critical role of MYBPC3 during sarcomere assembly in cardiac myocyte differentiation and suggest developmental influences of MYBPC3 truncating mutations on the mature hypertrophic phenotype.
Collapse
Affiliation(s)
- Andre Monteiro da Rocha
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States; Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Guadalupe Guerrero-Serna
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Adam Helms
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Carly Luzod
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Sergey Mironov
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Mark Russell
- Department of Pediatrics, University of Michigan, Ann Arbor, MI 48109, United States
| | - José Jalife
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Sharlene M Day
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States
| | - Gary D Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, United States.
| | - Todd J Herron
- Department of Internal Medicine, Center for Arrhythmia Research, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
48
|
Geist J, Kontrogianni-Konstantopoulos A. MYBPC1, an Emerging Myopathic Gene: What We Know and What We Need to Learn. Front Physiol 2016; 7:410. [PMID: 27683561 PMCID: PMC5021714 DOI: 10.3389/fphys.2016.00410] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022] Open
Abstract
Myosin Binding Protein-C (MyBP-C) comprises a family of accessory proteins that includes the cardiac, slow skeletal, and fast skeletal isoforms. The three isoforms share structural and sequence homology, and localize at the C-zone of the sarcomeric A-band where they interact with thick and thin filaments to regulate the cycling of actomyosin crossbridges. The cardiac isoform, encoded by MYBPC3, has been extensively studied over the last several decades due to its high mutational rate in congenital hypertrophic and dilated cardiomyopathy. It is only recently, however, that the MYBPC1 gene encoding the slow skeletal isoform (sMyBP-C) has gained attention. Accordingly, during the last 5 years it has been shown that MYBPC1 undergoes extensive exon shuffling resulting in the generation of multiple slow variants, which are co-expressed in different combinations and amounts in both slow and fast skeletal muscles. The sMyBP-C variants are subjected to PKA- and PKC-mediated phosphorylation in constitutive and alternatively spliced sites. More importantly, missense, and nonsense mutations in MYBPC1 have been directly linked with the development of severe and lethal forms of distal arthrogryposis myopathy and muscle tremors. Currently, there is no mammalian animal model of sMyBP-C, but new technologies including CRISPR/Cas9 and xenografting of human biopsies into immunodeficient mice could provide unique ways to study the regulation and roles of sMyBP-C in health and disease.
Collapse
Affiliation(s)
- Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine Baltimore, MD, USA
| | | |
Collapse
|
49
|
Taylor EN, Hoffman MP, Barefield DY, Aninwene GE, Abrishamchi AD, Lynch TL, Govindan S, Osinska H, Robbins J, Sadayappan S, Gilbert RJ. Alterations in Multi-Scale Cardiac Architecture in Association With Phosphorylation of Myosin Binding Protein-C. J Am Heart Assoc 2016; 5:e002836. [PMID: 27068630 PMCID: PMC4943261 DOI: 10.1161/jaha.115.002836] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Background The geometric organization of myocytes in the ventricular wall comprises the structural underpinnings of cardiac mechanical function. Cardiac myosin binding protein‐C (MYBPC3) is a sarcomeric protein, for which phosphorylation modulates myofilament binding, sarcomere morphology, and myocyte alignment in the ventricular wall. To elucidate the mechanisms by which MYBPC3 phospho‐regulation affects cardiac tissue organization, we studied ventricular myoarchitecture using generalized Q‐space imaging (GQI). GQI assessed geometric phenotype in excised hearts that had undergone transgenic (TG) modification of phospho‐regulatory serine sites to nonphosphorylatable alanines (MYBPC3AllP−/(t/t)) or phospho‐mimetic aspartic acids (MYBPC3AllP+/(t/t)). Methods and Results Myoarchitecture in the wild‐type (MYBPC3WT) left‐ventricle (LV) varied with transmural position, with helix angles ranging from −90/+90 degrees and contiguous circular orientation from the LV mid‐myocardium to the right ventricle (RV). Whereas MYBPC3AllP+/(t/t) hearts were not architecturally distinct from MYBPC3WT, MYBPC3AllP−/(t/t) hearts demonstrated a significant reduction in LV transmural helicity. Null MYBPC3(t/t) hearts, as constituted by a truncated MYBPC3 protein, demonstrated global architectural disarray and loss in helicity. Electron microscopy was performed to correlate the observed macroscopic architectural changes with sarcomere ultrastructure and demonstrated that impaired phosphorylation of MYBPC3 resulted in modifications of the sarcomere aspect ratio and shear angle. The mechanical effect of helicity loss was assessed through a geometric model relating cardiac work to ejection fraction, confirming the mechanical impairments observed with echocardiography. Conclusions We conclude that phosphorylation of MYBPC3 contributes to the genesis of ventricular wall geometry, linking myofilament biology with multiscale cardiac mechanics and myoarchitecture.
Collapse
Affiliation(s)
- Erik N Taylor
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA
| | - Matthew P Hoffman
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA
| | - David Y Barefield
- Health Sciences Division, Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, IL
| | - George E Aninwene
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA
| | - Aurash D Abrishamchi
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA
| | - Thomas L Lynch
- Health Sciences Division, Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, IL
| | - Suresh Govindan
- Health Sciences Division, Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, IL
| | - Hanna Osinska
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jeffrey Robbins
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sakthivel Sadayappan
- Health Sciences Division, Department of Cell and Molecular Physiology, Loyola University of Chicago, Maywood, IL
| | - Richard J Gilbert
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA
| |
Collapse
|
50
|
Sivaguru M, Fried G, Sivaguru BS, Sivaguru VA, Lu X, Choi KH, Saif MTA, Lin B, Sadayappan S. Cardiac muscle organization revealed in 3-D by imaging whole-mount mouse hearts using two-photon fluorescence and confocal microscopy. Biotechniques 2015; 59:295-308. [DOI: 10.2144/000114356] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 09/03/2015] [Indexed: 11/23/2022] Open
Abstract
The ability to image the entire adult mouse heart at high resolution in 3-D would provide enormous advantages in the study of heart disease. However, a technique for imaging nuclear/cellular detail as well as the overall structure of the entire heart in 3-D with minimal effort is lacking. To solve this problem, we modified the benzyl alcohol:benzyl benzoate (BABB) clearing technique by labeling mouse hearts with periodic acid Schiff (PAS) stain. We then imaged the hearts with a combination of two-photon fluorescence microscopy and automated tile-scan imaging/stitching. Utilizing the differential spectral properties of PAS, we could identify muscle and nuclear compartments in the heart. We were also able to visualize the differences between a 3-month-old normal mouse heart and a mouse heart that had undergone heart failure due to the expression of cardiac myosin binding protein-C (cMyBP-C) gene mutation (t/t). Using 2-D and 3-D morphometric analysis, we found that the t/t heart had anomalous ventricular shape, volume, and wall thickness, as well as a disrupted sarcomere pattern. We further validated our approach using decellularized hearts that had been cultured with 3T3 fibroblasts, which were tracked using a nuclear label. We were able to detect the 3T3 cells inside the decellularized intact heart tissue, achieving nuclear/cellular resolution in 3-D. The combination of labeling, clearing, and two-photon microscopy together with tiling eliminates laborious and time-consuming physical sectioning, alignment, and 3-D reconstruction.
Collapse
Affiliation(s)
- Mayandi Sivaguru
- Microscopy and Imaging Core Facility, Carl R. Woese Institute for Genomic Biology
| | - Glenn Fried
- Microscopy and Imaging Core Facility, Carl R. Woese Institute for Genomic Biology
| | | | | | - Xiaochen Lu
- Department of Cell and Developmental Biology and Carl R. Woese Institute for Genomic Biology
| | - Kyung Hwa Choi
- Department of Mechanical Science and Engineering, University of Illinois at Urbana Champaign, Urbana
| | - M Taher A Saif
- Department of Mechanical Science and Engineering, University of Illinois at Urbana Champaign, Urbana
| | - Brian Lin
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, IL
| | - Sakthivel Sadayappan
- Cell and Molecular Physiology, Stritch School of Medicine, Loyola University, Maywood, IL
| |
Collapse
|