1
|
Pioner JM, Pierantozzi E, Coppini R, Rubino EM, Biasci V, Vitale G, Laurino A, Santini L, Scardigli M, Randazzo D, Olianti C, Serano M, Rossi D, Tesi C, Cerbai E, Lange S, Reggiani C, Sacconi L, Poggesi C, Ferrantini C, Sorrentino V. Obscurin deficiency leads to compensated dilated cardiomyopathy and increased arrhythmias. J Gen Physiol 2025; 157:e202413696. [PMID: 40366302 PMCID: PMC12077377 DOI: 10.1085/jgp.202413696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 03/17/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Obscurin is a large muscle protein whose multiple functions include providing mechanical strength to the M-band and linking the sarcomere to the sarcoplasmic reticulum. Mutations in obscurin are linked to various forms of muscle diseases. This study compares cardiac function in a murine model of obscurin deletion (KO) with wild-type (WT) in vivo and ex vivo. Echocardiography showed that KO hearts had larger (+20%) end-diastolic and end-systolic volumes, reduced fractional shortening, and impaired ejection fraction, consistent with dilated cardiomyopathy. However, stroke volume and cardiac output were preserved due to increased end-diastolic volume. Morphological analyses revealed reduced sarcoplasmic reticulum volume, with preserved T-tubule network. While myofilament function was preserved in isolated myofibrils and skinned trabeculae, experiments in intact trabeculae revealed that Obscn KO hearts compared with WT displayed (1) reduced active tension at high frequencies and during resting-state contractions, (2) impaired positive inotropic and lusitropic response to β-adrenergic stimulation (isoproterenol 0.1 μM), and (3) faster mechanical restitution, suggesting reduced sarcoplasmic reticulum refractoriness. Intracellular [Ca2+]i measurements showed reduced peak systolic and increased diastolic levels in KO versus WT cardiomyocytes. Western blot experiments revealed lower SERCA and phospholamban (PLB) expression and reduced PLB phosphorylation in KO mice. While action potential parameters and conduction velocity were unchanged, β-adrenergic stimulation induced more frequent spontaneous Ca2+ waves and increased arrhythmia susceptibility in KO compared with WT. Taken together, these findings suggest that obscurin deletion, in adult mice, is linked to compensated dilated cardiomyopathy, altered E-C coupling, impaired response to inotropic agents, and increased propensity to arrhythmias.
Collapse
Affiliation(s)
| | - Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | | | - Egidio Maria Rubino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Valentina Biasci
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Florence, Italy
| | - Giulia Vitale
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Annunziatina Laurino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Lorenzo Santini
- Department NeuroFarBa, University of Florence, Florence, Italy
| | - Marina Scardigli
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Davide Randazzo
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Camilla Olianti
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Florence, Italy
| | - Matteo Serano
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Stephan Lange
- Institute of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Science and Research Center Koper, Institute for Kinesiology Research, Koper, Slovenia
| | - Leonardo Sacconi
- Institute of Clinical Physiology, National Research Council (IFC-CNR), Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
- Program of Molecular Diagnosis of Rare Genetic Diseases, Azienda Ospedaliero Universitaria Senese, Siena, Italy
| |
Collapse
|
2
|
Noureddine M, Mikolajek H, Morgan NV, Denning C, Loughna S, Gehmlich K, Mohammed F. Structural and functional insights into α-actinin isoforms and their implications in cardiovascular disease. J Gen Physiol 2025; 157:e202413684. [PMID: 39918740 PMCID: PMC11804879 DOI: 10.1085/jgp.202413684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/11/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
α-actinin (ACTN) is a pivotal member of the actin-binding protein family, crucial for the anchoring and organization of actin filaments within the cytoskeleton. Four isoforms of α-actinin exist: two non-muscle isoforms (ACTN1 and ACTN4) primarily associated with actin stress fibers and focal adhesions, and two muscle-specific isoforms (ACTN2 and ACTN3) localized to the Z-disk of the striated muscle. Although these isoforms share structural similarities, they exhibit distinct functional characteristics that reflect their specialized roles in various tissues. Genetic variants in α-actinin isoforms have been implicated in a range of pathologies, including cardiomyopathies, thrombocytopenia, and non-cardiovascular diseases, such as nephropathy. However, the precise impact of these genetic variants on the α-actinin structure and their contribution to disease pathogenesis remains poorly understood. This review provides a comprehensive overview of the structural and functional attributes of the four α-actinin isoforms, emphasizing their roles in actin crosslinking and sarcomere stabilization. Furthermore, we present detailed structural modeling of select ACTN1 and ACTN2 variants to elucidate mechanisms underlying disease pathogenesis, with a particular focus on macrothrombocytopenia and hypertrophic cardiomyopathy. By advancing our understanding of α-actinin's role in both normal cellular function and disease states, this review lays the groundwork for future research and the development of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Maya Noureddine
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
| | - Halina Mikolajek
- Diamond Light Source Ltd., Harwell Science and Innovation Campus, Didcot, UK
| | - Neil V. Morgan
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
| | - Chris Denning
- Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Siobhan Loughna
- School of Life Sciences, Faculty of Medicine and Health Sciences, University of Nottingham, Nottingham, UK
| | - Katja Gehmlich
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health University of Birmingham, Birmingham, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, UK
| | - Fiyaz Mohammed
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
3
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
4
|
Xu J, He K, Ji Y, Liu X, Dai Q. Downregulation of HHATL promotes cardiac hypertrophy via activation of SHH/DRP1. Exp Cell Res 2024; 439:114072. [PMID: 38719175 DOI: 10.1016/j.yexcr.2024.114072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/13/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
HHATL, previously implicated in cardiac hypertrophy in the zebrafish model, has emerged as a prioritized HCM risk gene. We identified six rare mutations in HHATL, present in 6.94 % of nonsarcomeric HCM patients (5/72). Moreover, a decrease of HHATL in the heart tissue from HCM patients and cardiac hypertrophy mouse model using transverse aortic constriction was observed. Despite this, the precise pathogenic mechanisms underlying HHATL-associated cardiac hypertrophy remain elusive. In this study, we observed that HHATL downregulation in H9C2 cells resulted in elevated expression of hypertrophic markers and reactive oxygen species (ROS), culminating in cardiac hypertrophy and mitochondrial dysfunction. Notably, the bioactive form of SHH, SHHN, exhibited a significant increase, while the mitochondrial fission protein dynamin-like GTPase (DRP1) decreased upon HHATL depletion. Intervention with the SHH inhibitor RU-SKI 43 or DRP1 overexpression effectively prevented Hhatl-depletion-induced cardiac hypertrophy, mitigating disruptions in mitochondrial morphology and membrane potential through the SHH/DRP1 axis. In summary, our findings suggest that HHATL depletion activates SHH signaling, reducing DRP1 levels and thereby promoting the expression of hypertrophic markers, ROS generation, and mitochondrial dysfunction, ultimately leading to cardiac hypertrophy. This study provides additional compelling evidence supporting the association of HHATL with cardiac hypertrophy.
Collapse
Affiliation(s)
- Jing Xu
- Department of Clinical Laboratory, ZhongDa Hospital, Southeast University, Nanjing, China; School of Medicine, Southeast University, Nanjing, China.
| | - Keyu He
- Department of Clinical Laboratory, ZhongDa Hospital, Southeast University, Nanjing, China
| | - Yichen Ji
- School of Medicine, Southeast University, Nanjing, China
| | - Xiangdong Liu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Qiming Dai
- Department of Cardiology, ZhongDa Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
5
|
Koutsofti C, Ioannides M, Polydorou C, Papagregoriou G, Malatras A, Michael G, Hadjiioannou I, Pieri S, Loizidou EM, Eftychiou C, Papasavvas E, Christophides T, Alkelai A, Kapoor M, Shuldiner AR, Avraamides P, Deltas C. Massive Parallel DNA Sequencing of Patients with Inherited Cardiomyopathies in Cyprus and Suggestion of Digenic or Oligogenic Inheritance. Genes (Basel) 2024; 15:319. [PMID: 38540378 PMCID: PMC10970479 DOI: 10.3390/genes15030319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 06/14/2024] Open
Abstract
Inherited cardiomyopathies represent a highly heterogeneous group of cardiac diseases. DNA variants in genes expressed in cardiomyocytes cause a diverse spectrum of cardiomyopathies, ultimately leading to heart failure, arrythmias, and sudden cardiac death. We applied massive parallel DNA sequencing using a 72-gene panel for studying inherited cardiomyopathies. We report on variants in 25 families, where pathogenicity was predicted by different computational approaches, databases, and an in-house filtering analysis. All variants were validated using Sanger sequencing. Familial segregation was tested when possible. We identified 41 different variants in 26 genes. Analytically, we identified fifteen variants previously reported in the Human Gene Mutation Database: twelve mentioned as disease-causing mutations (DM) and three as probable disease-causing mutations (DM?). Additionally, we identified 26 novel variants. We classified the forty-one variants as follows: twenty-eight (68.3%) as variants of uncertain significance, eight (19.5%) as likely pathogenic, and five (12.2%) as pathogenic. We genetically characterized families with a cardiac phenotype. The genetic heterogeneity and the multiplicity of candidate variants are making a definite molecular diagnosis challenging, especially when there is a suspicion of incomplete penetrance or digenic-oligogenic inheritance. This is the first systematic study of inherited cardiac conditions in Cyprus, enabling us to develop a genetic baseline and precision cardiology.
Collapse
Affiliation(s)
- Constantina Koutsofti
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Marios Ioannides
- Department of Cardiology, Nicosia General Hospital, Nicosia 2029, Cyprus; (M.I.); (C.E.); (T.C.)
| | - Christiana Polydorou
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Gregory Papagregoriou
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Apostolos Malatras
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - George Michael
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Irene Hadjiioannou
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Stylianos Pieri
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Eleni M. Loizidou
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
| | - Christos Eftychiou
- Department of Cardiology, Nicosia General Hospital, Nicosia 2029, Cyprus; (M.I.); (C.E.); (T.C.)
| | | | - Theodoros Christophides
- Department of Cardiology, Nicosia General Hospital, Nicosia 2029, Cyprus; (M.I.); (C.E.); (T.C.)
| | - Anna Alkelai
- Regeneron Genetics Center, Tarrytown, NY 10591, USA; (A.A.); (M.K.); (A.R.S.)
| | - Manav Kapoor
- Regeneron Genetics Center, Tarrytown, NY 10591, USA; (A.A.); (M.K.); (A.R.S.)
| | - Alan R. Shuldiner
- Regeneron Genetics Center, Tarrytown, NY 10591, USA; (A.A.); (M.K.); (A.R.S.)
| | - Panayiotis Avraamides
- Department of Cardiology, Nicosia General Hospital, Nicosia 2029, Cyprus; (M.I.); (C.E.); (T.C.)
| | - Constantinos Deltas
- Molecular Medicine Research Center, biobank.cy Center of Excellence in Biobanking and Biomedical Research, University of Cyprus, Nicosia 2109, Cyprus; (C.K.); (C.P.); (G.P.); (A.M.); (G.M.); (I.H.); (S.P.); (E.M.L.)
- School of Medicine, University of Cyprus, Nicosia 2109, Cyprus
| |
Collapse
|
6
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
7
|
Stathopoulou K, Schnittger J, Raabe J, Fleischer F, Mangels N, Piasecki A, Findlay J, Hartmann K, Krasemann S, Schlossarek S, Uebeler J, Wixler V, Blake DJ, Baillie GS, Carrier L, Ehler E, Cuello F. CMYA5 is a novel interaction partner of FHL2 in cardiac myocytes. FEBS J 2022; 289:4622-4645. [PMID: 35176204 DOI: 10.1111/febs.16402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/13/2022] [Accepted: 02/15/2022] [Indexed: 11/27/2022]
Abstract
Four-and-a-half LIM domains protein 2 (FHL2) is an anti-hypertrophic adaptor protein that regulates cardiac myocyte signalling and function. Herein, we identified cardiomyopathy-associated 5 (CMYA5) as a novel FHL2 interaction partner in cardiac myocytes. In vitro pull-down assays demonstrated interaction between FHL2 and the N- and C-terminal regions of CMYA5. The interaction was verified in adult cardiac myocytes by proximity ligation assays. Immunofluorescence and confocal microscopy demonstrated co-localisation in the same subcellular compartment. The binding interface between FHL2 and CMYA5 was mapped by peptide arrays. Exposure of neonatal rat ventricular myocytes to a CMYA5 peptide covering one of the FHL2 interaction sites led to an increase in cell area at baseline, but a blunted response to chronic phenylephrine treatment. In contrast to wild-type hearts, loss or reduced FHL2 expression in Fhl2-targeted knockout mouse hearts or in a humanised mouse model of hypertrophic cardiomyopathy led to redistribution of CMYA5 into the perinuclear and intercalated disc region. Taken together, our results indicate a direct interaction of the two adaptor proteins FHL2 and CMYA5 in cardiac myocytes, which might impact subcellular compartmentation of CMYA5.
Collapse
Affiliation(s)
- Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Josef Schnittger
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Frederic Fleischer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Nils Mangels
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Angelika Piasecki
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Jane Findlay
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Kristin Hartmann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Krasemann
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - June Uebeler
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Viktor Wixler
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Germany
| | - Derek J Blake
- Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, UK
| | - George S Baillie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| | - Elisabeth Ehler
- School of Cardiovascular Medicine and Sciences, BHF Research Excellence Centre, King's College London, UK.,Randall Centre for Cell and Molecular Biophysics (School of Basic and Medical Biosciences), King's College London, UK
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Germany.,DZHK (German Center for Cardiovascular Research), partner site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
8
|
A novel missense mutation in obscurin gene in a Chinese consanguineous family with left ventricular noncompaction. J Geriatr Cardiol 2022; 19:531-538. [PMID: 35975021 PMCID: PMC9361159 DOI: 10.11909/j.issn.1671-5411.2022.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Left ventricular noncompaction (LVNC) is an increasingly recognised cardiomyopathy of which a significant percentage are genetic in origin. The purpose of the present study was to identify potential pathogenic mutation leading to disease in a Chinese LVNC family. METHODS A 3-generation family affected by LVNC was recruited. Clinical assessments were performed on available family members, with clinical examination, ECG, echocardiography and cardiac MRI. The proband (I-2), the proband's daughter (II-1, affected) and mother (III-1, unaffected) were selected for WGS. Sanger sequencing were performed in all of the 4 surviving family members. RESULTS Combined whole genome sequencing with linkage analysis identified a novel missense mutation in the giant protein obscurin (OBSCN NM_001098623, c.C19063T), as the only plausible disease-causing variant that segregates with disease among the four surviving individuals, with interrogation of the entire genome excluding other potential causes. This c.C19063T missense mutation resulted in p.R6355W in the encoded OBSCN protein. It affected a highly conserved residue in the C terminus of the obscurin-B-like isoform between the PH and STKc domains, which was predicted to affect the function of the protein by different bioinformatics tools. CONCLUSIONS Here we present clinical and genetic evidence implicating the novel R6355W missense mutation in obscurin as the cause of familial LVNC. This expands the spectrum of obscurin's roles in cardiomyopathies. It furthermore highlights that rare obscurin missense variants, currently often ignored or left uninterpreted, should be considered to be relevant for cardiomyopathies and can be identified by the approach presented here. This study also provided new insights into the molecular basis of OBSCN mutation positive LVNC.
Collapse
|
9
|
Wang Y, Zhang C, Zhou B, Hui L, Zheng L, Chen X, Wang S, Yang L, Hao S, Zhang Q. Three Variants Affecting Exon 1 of Ectodysplasin A Cause X-Linked Hypohidrotic Ectodermal Dysplasia: Clinical and Molecular Characteristics. Front Genet 2022; 13:916340. [PMID: 35873474 PMCID: PMC9298733 DOI: 10.3389/fgene.2022.916340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/23/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Ectodysplasin A (EDA) variations are major pathogenic factors for hypohidrotic ectodermal dysplasia (HED), the most common form of ectodermal dysplasia (ED), characterized by hypotrichosis, hypohidrosis, hypodontia, and other oral features. Methods: Molecular genetic defects in three HED families were detected by whole-exome sequencing and confirmed by Sanger sequencing or multiplex ligation-dependent probe amplification. The effect of splicing variant was further verified by EDA minigene in vitro analysis. De novo deletion was confirmed by chromosomal microarray analysis. Results: Three variants (c.396 + 1 G > C, c.171-173 del GTT, and exon 1 deletion) were identified, all affecting exon 1 of the EDA gene. Variants c.396 + 1 G > C and c.171-173 del GTT were first identified. Minigene analysis of the splicing variant (c.396 + 1 G > C) displayed a prolonged EDA-A1 transcript containing extra 699 bp at the start of intron 1, representing a functional cryptic splice site formation in vitro. Combining the results of chromosomal microarray analysis and whole-exome sequencing, the deletion variant was over 87 kb. Three variants were predicted to affect protein function to differing degrees, and were responsible for X-linked HED with varying phenotype. Conclusion: Investigating the clinical and molecular characteristics of these variations broadens our understanding of EDA gene variants, supporting clinical diagnosis, genetic counseling, and prenatal diagnosis of HED.
Collapse
Affiliation(s)
- Yupei Wang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Chuan Zhang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Bingbo Zhou
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Ling Hui
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Lei Zheng
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Xue Chen
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Shifan Wang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Lan Yang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Shengju Hao
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| | - Qinghua Zhang
- Medical Genetics Center, Gansu Provincial Maternity and Child-care Hospital, Lanzhou, China.,Gansu Provincial Clinical Research Center for Birth Defects and Rare Diseases, Lanzhou, China
| |
Collapse
|
10
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
11
|
Abstract
Junctophilins (JPHs) comprise a family of structural proteins that connect the plasma membrane to intracellular organelles such as the endo/sarcoplasmic reticulum. Tethering of these membrane structures results in the formation of highly organized subcellular junctions that play important signaling roles in all excitable cell types. There are four JPH isoforms, expressed primarily in muscle and neuronal cell types. Each JPH protein consists of 6 'membrane occupation and recognition nexus' (MORN) motifs, a joining region connecting these to another set of 2 MORN motifs, a putative alpha-helical region, a divergent region exhibiting low homology between JPH isoforms, and a carboxy-terminal transmembrane region anchoring into the ER/SR membrane. JPH isoforms play essential roles in developing and maintaining subcellular membrane junctions. Conversely, inherited mutations in JPH2 cause hypertrophic or dilated cardiomyopathy, while trinucleotide expansions in the JPH3 gene cause Huntington Disease-Like 2. Loss of JPH1 protein levels can cause skeletal myopathy, while loss of cardiac JPH2 levels causes heart failure and atrial fibrillation, among other disease. This review will provide a comprehensive overview of the JPH gene family, phylogeny, and evolutionary analysis of JPH genes and other MORN domain proteins. JPH biogenesis, membrane tethering, and binding partners will be discussed, as well as functional roles of JPH isoforms in excitable cells. Finally, potential roles of JPH isoform deficits in human disease pathogenesis will be reviewed.
Collapse
Affiliation(s)
- Stephan E Lehnart
- Cellular Biophysics and Translational Cardiology Section, Heart Research Center Göttingen, University Medical Center Göttingen, Department of Cardiology and Pneumology, Georg-August University Göttingen, Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, Texas, United States; Departments of Molecular Physiology and Biophysics, Medicine (Cardiology), Pediatrics (Cardiology), Neuroscience, and Center for Space Medicine, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
12
|
Chaklader M, Rothermel BA. Calcineurin in the heart: New horizons for an old friend. Cell Signal 2021; 87:110134. [PMID: 34454008 PMCID: PMC8908812 DOI: 10.1016/j.cellsig.2021.110134] [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] [Received: 05/13/2021] [Revised: 08/10/2021] [Accepted: 08/23/2021] [Indexed: 01/20/2023]
Abstract
Calcineurin, also known as PP2B or PPP3, is a member of the PPP family of protein phosphatases that also includes PP1 and PP2A. Together these three phosphatases carryout the majority of dephosphorylation events in the heart. Calcineurin is distinct in that it is activated by the binding of calcium/calmodulin (Ca2+/CaM) and therefore acts as a node for integrating Ca2+ signals with changes in phosphorylation, two fundamental intracellular signaling cascades. In the heart, calcineurin is primarily thought of in the context of pathological cardiac remodeling, acting through the Nuclear Factor of Activated T-cell (NFAT) family of transcription factors. However, calcineurin activity is also essential for normal heart development and homeostasis in the adult heart. Furthermore, it is clear that NFAT-driven changes in transcription are not the only relevant processes initiated by calcineurin in the setting of pathological remodeling. There is a growing appreciation for the diversity of calcineurin substrates that can impact cardiac function as well as the diversity of mechanisms for targeting calcineurin to specific sub-cellular domains in cardiomyocytes and other cardiac cell types. Here, we will review the basics of calcineurin structure, regulation, and function in the context of cardiac biology. Particular attention will be given to: the development of improved tools to identify and validate new calcineurin substrates; recent studies identifying new calcineurin isoforms with unique properties and targeting mechanisms; and the role of calcineurin in cardiac development and regeneration.
Collapse
Affiliation(s)
- Malay Chaklader
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA
| | - Beverly A Rothermel
- Departments of Internal Medicine (Division of Cardiology) and Molecular Biology, University of Texas Southwestern Medical Centre, Dallas, TX, USA.
| |
Collapse
|
13
|
Wu G, Liu J, Liu M, Huang Q, Ruan J, Zhang C, Wang D, Sun X, Jiang W, Kang L, Wang J, Song L. Truncating Variants in OBSCN Gene Associated With Disease-Onset and Outcomes of Hypertrophic Cardiomyopathy. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2021; 14:e003401. [PMID: 34601892 DOI: 10.1161/circgen.121.003401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The presence of variants in OBSCN was identified to be linked to hypertrophic cardiomyopathy (HCM), but whether OBSCN truncating variants were associated with HCM remained unknown. METHODS Whole-exome sequencing was performed in 986 patients with HCM and 761 non-HCM controls to search for OBSCN truncating variants, and the result was tested in a replication cohort consisting of 529 patients with HCM and 307 controls. The association of the OBSCN truncating variants with baseline characteristics and prognosis of patients with HCM were ascertained. RESULTS There were 28 qualifying truncating variants in the OBSCN gene detected in 26 (2.6%) patients with HCM and 6 (0.8%) controls. The OBSCN truncating variants were more prevalent in patients with HCM than controls (odds ratio, 3.4, P=0.004). This association was confirmed in the replication cohort (odds ratio, 3.8, P=0.024). The combined effects of the two cohorts estimated the odds ratio to be 3.58 (P<0.001). Patients with or without OBSCN truncating variants shared similar demographic and echocardiographic variables at baseline. During 3.3±2.4 years (4795 patient-years) follow-up, the patients with OBSCN truncating variants were more likely to experience cardiovascular death (adjusted hazard ratio, 3.1 [95% CI, 1.40-6.70], P=0.005) and all-cause death (adjusted hazard ratio, 2.63 [95% CI, 1.21-5.71], P=0.015). CONCLUSIONS Our data indicated that OBSCN truncating variants contributed to the disease-onset of HCM, and increased the risk of malignant events in patients with HCM.
Collapse
Affiliation(s)
- Guixin Wu
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Liu
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minghao Liu
- Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiya Huang
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jieyun Ruan
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Channa Zhang
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dong Wang
- Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaolu Sun
- Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Jiang
- Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lianming Kang
- Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease (G.W., J.L., Q.H., J.R., C.Z., J.W., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Cardiomyopathy Ward (G.W., J.L., M.L., Q.H., J.R., D.W., X.S., W.J., L.K., L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,National Clinical Research Center of Cardiovascular Diseases (L.S.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Biquand A, Spinozzi S, Tonino P, Cosette J, Strom J, Elbeck Z, Knöll R, Granzier H, Lostal W, Richard I. Titin M-line insertion sequence 7 is required for proper cardiac function in mice. J Cell Sci 2021; 134:271843. [PMID: 34401916 DOI: 10.1242/jcs.258684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 08/06/2021] [Indexed: 11/20/2022] Open
Abstract
Titin is a giant sarcomeric protein that is involved in a large number of functions, with a primary role in skeletal and cardiac sarcomere organization and stiffness. The titin gene (TTN) is subject to various alternative splicing events, but in the region that is present at the M-line, the only exon that can be spliced out is Mex5, which encodes for the insertion sequence 7 (is7). Interestingly, in the heart, the majority of titin isoforms are Mex5+, suggesting a cardiac role for is7. Here, we performed comprehensive functional, histological, transcriptomic, microscopic and molecular analyses of a mouse model lacking the Ttn Mex5 exon (ΔMex5), and revealed that the absence of the is7 is causative for dilated cardiomyopathy. ΔMex5 mice showed altered cardiac function accompanied by increased fibrosis and ultrastructural alterations. Abnormal expression of excitation-contraction coupling proteins was also observed. The results reported here confirm the importance of the C-terminal region of titin in cardiac function and are the first to suggest a possible relationship between the is7 and excitation-contraction coupling. Finally, these findings give important insights for the identification of new targets in the treatment of titinopathies.
Collapse
Affiliation(s)
- Ariane Biquand
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Simone Spinozzi
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Paola Tonino
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | | | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - Zaher Elbeck
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institutet, 141 57 Huddinge, Sweden
| | - Ralph Knöll
- Department of Medicine, Integrated Cardio Metabolic Centre (ICMC), Heart and Vascular Theme, Karolinska Institutet, 141 57 Huddinge, Sweden.,Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 50 Gothenburg, Sweden
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85721, USA
| | - William Lostal
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| | - Isabelle Richard
- Genethon, 91000 Evry, France.,Université Paris-Saclay, Univ Evry, Inserm, Généthon, Integrare research unit UMR_S951, 91000 Evry-Courcouronnes, France
| |
Collapse
|
15
|
Wang S, Wang E, Chen Q, Yang Y, Xu L, Zhang X, Wu R, Hu X, Wu Z. Uncovering Potential lncRNAs and mRNAs in the Progression From Acute Myocardial Infarction to Myocardial Fibrosis to Heart Failure. Front Cardiovasc Med 2021; 8:664044. [PMID: 34336943 PMCID: PMC8322527 DOI: 10.3389/fcvm.2021.664044] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Morbidity and mortality of heart failure (HF) post-myocardial infarction (MI) remain elevated. The aim of this study was to find potential long non-coding RNAs (lncRNAs) and mRNAs in the progression from acute myocardial infarction (AMI) to myocardial fibrosis (MF) to HF. Methods: Firstly, blood samples from AMI, MF, and HF patients were used for RNA sequencing. Secondly, differentially expressed lncRNAs and mRNAs were obtained in MF vs. AMI and HF vs. MF, followed by functional analysis of shared differentially expressed mRNAs between two groups. Thirdly, interaction networks of lncRNA-nearby targeted mRNA and lncRNA-co-expressed mRNA were constructed in MF vs. AMI and HF vs. MF. Finally, expression validation and diagnostic capability analysis of selected lncRNAs and mRNAs were performed. Results: Several lncRNA-co-expressed/nearby targeted mRNA pairs including AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 were identified. Several signaling pathways including TNF and cytokine–cytokine receptor interaction, fructose and mannose metabolism and HIF-1, hematopoietic cell lineage and fluid shear stress, and atherosclerosis and estrogen were selected. IL1R2, IRAK3, LRG1, and PLAC4 had a potential diagnostic value for both AMI and HF. Conclusion: Identified AC005392.3/AC007278.2-IL18R1, AL356356.1/AL137145.2-PFKFB3, and MKNK1-AS1/LINC01127-IL1R2 lncRNA-co-expressed/nearby targeted mRNA pairs may play crucial roles in the development of AMI, MF, and HF.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Enmao Wang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Qincong Chen
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Yan Yang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Lei Xu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaolei Zhang
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Rubing Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xitian Hu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Zhihong Wu
- Department of Cardiovasology, Shijiazhuang People's Hospital, Shijiazhuang, China
| |
Collapse
|
16
|
Xu J, Liu X, Dai Q. Integration of transcriptomic data identifies key hallmark genes in hypertrophic cardiomyopathy. BMC Cardiovasc Disord 2021; 21:330. [PMID: 34225646 PMCID: PMC8259117 DOI: 10.1186/s12872-021-02147-7] [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: 10/19/2020] [Accepted: 06/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypertrophic cardiomyopathy (HCM) represents one of the most common inherited heart diseases. To identify key molecules involved in the development of HCM, gene expression patterns of the heart tissue samples in HCM patients from multiple microarray and RNA-seq platforms were investigated. METHODS The significant genes were obtained through the intersection of two gene sets, corresponding to the identified differentially expressed genes (DEGs) within the microarray data and within the RNA-Seq data. Those genes were further ranked using minimum-Redundancy Maximum-Relevance feature selection algorithm. Moreover, the genes were assessed by three different machine learning methods for classification, including support vector machines, random forest and k-Nearest Neighbor. RESULTS Outstanding results were achieved by taking exclusively the top eight genes of the ranking into consideration. Since the eight genes were identified as candidate HCM hallmark genes, the interactions between them and known HCM disease genes were explored through the protein-protein interaction (PPI) network. Most candidate HCM hallmark genes were found to have direct or indirect interactions with known HCM diseases genes in the PPI network, particularly the hub genes JAK2 and GADD45A. CONCLUSIONS This study highlights the transcriptomic data integration, in combination with machine learning methods, in providing insight into the key hallmark genes in the genetic etiology of HCM.
Collapse
Affiliation(s)
- Jing Xu
- Department of Clinical Laboratory, ZhongDa Hospital, Southeast University, Nanjing, China
| | - Xiangdong Liu
- Institute of Life Science, Southeast University, Nanjing, China
| | - Qiming Dai
- Department of Cardiology, ZhongDa Hospital, Southeast University, Nanjing, China.
| |
Collapse
|
17
|
Myospryn deficiency leads to impaired cardiac structure and function and schizophrenia-associated symptoms. Cell Tissue Res 2021; 385:675-696. [PMID: 34037836 DOI: 10.1007/s00441-021-03447-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/02/2021] [Indexed: 12/25/2022]
Abstract
The desmin-associated protein myospryn, encoded by the cardiomyopathy-associated gene 5 (CMYA5), is a TRIM-like protein associated to the BLOC-1 (Biogenesis of Lysosomes Related Organelles Complex 1) protein dysbindin. Human myospryn mutations are linked to both cardiomyopathy and schizophrenia; however, there is no evidence of a direct causative link of myospryn to these diseases. Therefore, we sought to unveil the role of myospryn in heart and brain. We have genetically inactivated the myospryn gene by homologous recombination and demonstrated that myospryn null hearts have dilated phenotype and compromised cardiac function. Ultrastructural analyses revealed that the sarcomere organization is not obviously affected; however, intercalated disk (ID) integrity is impaired, along with mislocalization of ID and sarcoplasmic reticulum (SR) protein components. Importantly, cardiac and skeletal muscles of myospryn null mice have severe mitochondrial defects with abnormal internal vacuoles and extensive cristolysis. In addition, swollen SR and T-tubules often accompany the mitochondrial defects, strongly implying a potential link of myospryn together with desmin to SR- mitochondrial physical and functional cross-talk. Furthermore, given the reported link of human myospryn mutations to schizophrenia, we performed behavioral studies, which demonstrated that myospryn-deficient male mice display disrupted startle reactivity and prepulse inhibition, asocial behavior, decreased exploratory behavior, and anhedonia. Brain neurochemical and ultrastructural analyses revealed prefrontal-striatal monoaminergic neurotransmitter defects and ultrastructural degenerative aberrations in cerebellar cytoarchitecture, respectively, in myospryn-deficient mice. In conclusion, myospryn is essential for both cardiac and brain structure and function and its deficiency leads to cardiomyopathy and schizophrenia-associated symptoms.
Collapse
|
18
|
Narumanchi S, Wang H, Perttunen S, Tikkanen I, Lakkisto P, Paavola J. Zebrafish Heart Failure Models. Front Cell Dev Biol 2021; 9:662583. [PMID: 34095129 PMCID: PMC8173159 DOI: 10.3389/fcell.2021.662583] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/06/2021] [Indexed: 01/02/2023] Open
Abstract
Heart failure causes significant morbidity and mortality worldwide. The understanding of heart failure pathomechanisms and options for treatment remain incomplete. Zebrafish has proven useful for modeling human heart diseases due to similarity of zebrafish and mammalian hearts, fast easily tractable development, and readily available genetic methods. Embryonic cardiac development is rapid and cardiac function is easy to observe and quantify. Reverse genetics, by using morpholinos and CRISPR-Cas9 to modulate gene function, make zebrafish a primary animal model for in vivo studies of candidate genes. Zebrafish are able to effectively regenerate their hearts following injury. However, less attention has been given to using zebrafish models to increase understanding of heart failure and cardiac remodeling, including cardiac hypertrophy and hyperplasia. Here we discuss using zebrafish to study heart failure and cardiac remodeling, and review zebrafish genetic, drug-induced and other heart failure models, discussing the advantages and weaknesses of using zebrafish to model human heart disease. Using zebrafish models will lead to insights on the pathomechanisms of heart failure, with the aim to ultimately provide novel therapies for the prevention and treatment of heart failure.
Collapse
Affiliation(s)
- Suneeta Narumanchi
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Hong Wang
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Sanni Perttunen
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| | - Ilkka Tikkanen
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland.,Abdominal Center Nephrology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Päivi Lakkisto
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland.,Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Jere Paavola
- Unit of Cardiovascular Research, Minerva Foundation Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland
| |
Collapse
|
19
|
Shi X, Zhang Y, Gong Y, Chen M, Brand-Arzamendi K, Liu X, Wen XY. Zebrafish hhatla is involved in cardiac hypertrophy. J Cell Physiol 2021; 236:3700-3709. [PMID: 33052609 DOI: 10.1002/jcp.30106] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/24/2020] [Accepted: 10/01/2020] [Indexed: 01/26/2023]
Abstract
Cardiac hypertrophy is a compensatory response to pathological stimuli, ultimately progresses to cardiomyopathy, heart failure, or sudden death. Although many signaling pathways have been reported to be involved in the hypertrophic process, it is still not fully clear about the underlying molecular mechanisms for cardiac hypertrophy. Hedgehog acyltransferase-like (Hhatl), a sarcoplasmic reticulum-resident protein, exhibits high expression in the heart and muscle. However, the biological role of Hhatl in the heart remains unknown. In this study, we first found that the expression level of Hhatl is markedly decreased in cardiac hypertrophy. We further studied the role of hhatla, homolog of Hhatl with the zebrafish model. The depletion of hhatla in zebrafish leads to cardiac defects, as well as an enhanced level of hypertrophic markers. Besides, we found that calcineurin signaling participates in hhatla depletion-induced cardiac hypertrophy. Together, these results demonstrate a critical role for hhatla in cardiac hypertrophy.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yijie Gong
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Mengying Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Koroboshka Brand-Arzamendi
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada
- Department of Medicine, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Xiangdong Liu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Li Ka Shing Knowledge Institute, Toronto, Ontario, Canada
- Department of Medicine, Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Mori AA, Castro LRD, Bortolin RH, Bastos GM, Oliveira VFD, Ferreira GM, Hirata TDC, Fajardo CM, Sampaio MF, Moreira DAR, Pachón-Mateos JC, Correia EDB, Sousa AGDMR, Brión M, Carracedo A, Hirata RDC, Hirata MH. Association of variants in MYH7, MYBPC3 and TNNT2 with sudden cardiac death-related risk factors in Brazilian patients with hypertrophic cardiomyopathy. Forensic Sci Int Genet 2021; 52:102478. [PMID: 33588347 DOI: 10.1016/j.fsigen.2021.102478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is characterized by unexplained left ventricular hypertrophy (LVH) and is one of the major causes of sudden cardiac death (SCD). An exon-targeted gene sequencing strategy was used to investigate the association of functional variants in sarcomeric genes (MYBPC3, MYH7 and TNNT2) with severe LVH and other SCD-related risk factors in Brazilian HCM patients. Clinical data of 55 HCM patients attending a Cardiology Hospital (Sao Paulo city, Brazil) were recorded. Severe LVH, aborted SCD, family history of SCD, syncope, non-sustained ventricular tachycardia and abnormal blood pressure in response to exercise were evaluated as SCD risk factors. Blood samples were obtained for genomic DNA extraction and the exons and untranslated regions of the MYH7, MYBPC3 and TNNT2 were sequenced using Nextera® and MiSEq® reagents. Variants were identified and annotated using in silico tools, and further classified as pathogenic or benign according to the American College of Medical Genetics and Genomics guidelines. Variants with functional effects were identified in MYBPC3 (n = 9), MYH7 (n = 6) and TNNT2 (n = 4). The benign variants MYBPC3 p.Val158Met and TNNT2 p.Lys263Arg were associated with severe LVH (p < 0.05), and the MYH7 p.Val320Met (pathogenic) was associated with family history of SCD (p = 0.037). Increased risk for severe LVH was found in carriers of MYBPC3 Met158 (c.472 A allele, OR = 13.5, 95% CI = 1.80-101.12, p = 0.011) or combined variants (MYBPC3, MYH7 and TNNT2: OR = 12.39, 95% CI = 2.14-60.39, p = 0.004). Carriers of TNNT2 p.Lys263Arg and combined variants had higher values of septum thickness than non-carriers (p < 0.05). Molecular modeling analysis showed that MYBPC3 158Met reduces the interaction of cardiac myosin-binding protein C (cMyBP-C) RASK domain (amino acids Arg215-Ala216-Ser217-Lys218) with tropomyosin. In conclusion, the variants MYBPC3 p.Val158Met, TNNT2 p.Lys263Arg and MYH7 p.Val320Met individually or combined contribute to the risk of sudden cardiac death and other outcomes of hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Augusto Akira Mori
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | | | | | - Gisele Medeiros Bastos
- Institute Dante Pazzanese of Cardiology, Sao Paulo, Brazil; Real e Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo, Brazil
| | | | - Glaucio Monteiro Ferreira
- School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil; Institute Dante Pazzanese of Cardiology, Sao Paulo, Brazil
| | | | | | - Marcelo Ferraz Sampaio
- Institute Dante Pazzanese of Cardiology, Sao Paulo, Brazil; Real e Benemerita Associação Portuguesa de Beneficiencia, Sao Paulo, Brazil
| | | | | | | | | | - Maria Brión
- Genetica Cardiovascular, Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, A Coruña, Spain; Grupo de Medicina Genômica, Universidad de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain
| | - Angel Carracedo
- Grupo de Medicina Genômica, Universidad de Santiago de Compostela, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Santiago de Compostela, Spain; Centro Nacional de Genotipado-CeGen-USC-PRB3-ISCIII, Santiago de Compostela, Spain
| | | | | |
Collapse
|
21
|
Dhandapany PS, Kang S, Kashyap DK, Rajagopal R, Sundaresan NR, Singh R, Thangaraj K, Jayaprakash S, Manjunath CN, Shenthar J, Lebeche D. Adiponectin receptor 1 variants contribute to hypertrophic cardiomyopathy that can be reversed by rapamycin. SCIENCE ADVANCES 2021; 7:eabb3991. [PMID: 33523960 PMCID: PMC7787482 DOI: 10.1126/sciadv.abb3991] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 11/13/2020] [Indexed: 06/12/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is a heterogeneous genetic heart muscle disease characterized by hypertrophy with preserved or increased ejection fraction in the absence of secondary causes. However, recent studies have demonstrated that a substantial proportion of individuals with HCM also have comorbid diabetes mellitus (~10%). Whether genetic variants may contribute a combined phenotype of HCM and diabetes mellitus is not known. Here, using next-generation sequencing methods, we identified novel and ultrarare variants in adiponectin receptor 1 (ADIPOR1) as risk factors for HCM. Biochemical studies showed that ADIPOR1 variants dysregulate glucose and lipid metabolism and cause cardiac hypertrophy through the p38/mammalian target of rapamycin and/or extracellular signal-regulated kinase pathways. A transgenic mouse model expressing an ADIPOR1 variant displayed cardiomyopathy that recapitulated the cellular findings, and these features were rescued by rapamycin. Our results provide the first evidence that ADIPOR1 variants can cause HCM and provide new insights into ADIPOR1 regulation.
Collapse
Affiliation(s)
- Perundurai S Dhandapany
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India.
- The Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR 97239, USA
- Departments of Medicine, Molecular, and Medical Genetics, Oregon Health and Science University, Portland, OR 97239, USA
| | - Soojeong Kang
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Deepak K Kashyap
- Centre for Cardiovascular Biology and Disease, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, India
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India
| | - Raksha Rajagopal
- Department of Microbiology and Cell Biology, Indian Institute of Science, CV Raman Avenue, Bangalore, India
| | - Nagalingam R Sundaresan
- Department of Microbiology and Cell Biology, Indian Institute of Science, CV Raman Avenue, Bangalore, India
| | - Rajvir Singh
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Kumarasamy Thangaraj
- CSIR-Center for Cellular and Molecular Biology, Hyderabad, India
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | - Shilpa Jayaprakash
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, India
| | - Cholenahally N Manjunath
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, India
| | - Jayaprakash Shenthar
- Department of Cardiology, Sri Jayadeva Institute of Cardiovascular Sciences and Research, Bengaluru, India
| | - Djamel Lebeche
- Cardiovascular Research Center, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
- Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
22
|
T-tubule remodeling in human hypertrophic cardiomyopathy. J Muscle Res Cell Motil 2020; 42:305-322. [PMID: 33222034 PMCID: PMC8332592 DOI: 10.1007/s10974-020-09591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/22/2020] [Indexed: 11/17/2022]
Abstract
The highly organized transverse T-tubule membrane system represents the ultrastructural substrate for excitation–contraction coupling in ventricular myocytes. While the architecture and function of T-tubules have been well described in animal models, there is limited morpho-functional data on T-tubules in human myocardium. Hypertrophic cardiomyopathy (HCM) is a primary disease of the heart muscle, characterized by different clinical presentations at the various stages of its progression. Most HCM patients, indeed, show a compensated hypertrophic disease (“non-failing hypertrophic phase”), with preserved left ventricular function, and only a small subset of individuals evolves into heart failure (“end stage HCM”). In terms of T-tubule remodeling, the “end-stage” disease does not differ from other forms of heart failure. In this review we aim to recapitulate the main structural features of T-tubules during the “non-failing hypertrophic stage” of human HCM by revisiting data obtained from human myectomy samples. Moreover, by comparing pathological changes observed in myectomy samples with those introduced by acute (experimentally induced) detubulation, we discuss the role of T-tubular disruption as a part of the complex excitation–contraction coupling remodeling process that occurs during disease progression. Lastly, we highlight how T-tubule morpho-functional changes may be related to patient genotype and we discuss the possibility of a primitive remodeling of the T-tubule system in rare HCM forms associated with genes coding for proteins implicated in T-tubule structural integrity, formation and maintenance.
Collapse
|
23
|
Shi X, Zhang Y, Chen R, Gong Y, Zhang M, Guan R, Rotstein OD, Liu X, Wen XY. ndufa7 plays a critical role in cardiac hypertrophy. J Cell Mol Med 2020; 24:13151-13162. [PMID: 32989924 PMCID: PMC7701565 DOI: 10.1111/jcmm.15921] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/22/2022] Open
Abstract
Cardiac hypertrophy is a common pathological change in patients with progressive cardiac function failure, which can be caused by hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) or arterial hypertension. Despite years of study, there is still limited knowledge about the underlying molecular mechanisms for cardiac hypertrophy. NDUFA7, a subunit of NADH:ubiquinone oxidoreductase (complex I), has been reported to be a novel HCM associated gene. However, the biological role of NDUFA7 in heart remains unknown. In this study, we found that NDUFA7 exhibited high expression in the heart, and its level was significantly decreased in mice model of cardiac hypertrophy. Moreover, we demonstrated that ndufa7 knockdown in developing zebrafish embryos resulted in cardiac development and functional defects, associated with increased expression of pathological hypertrophy biomarkers nppa (ANP) and nppb (BNP). Mechanistic study demonstrated that ndufa7 depletion promoted ROS production and calcineurin signalling activation. Moreover, NDUFA7 depletion contributed to cardiac cell hypertrophy. Together, these results report for the first time that ndufa7 is implicated in pathological cardiac hypertrophy.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Ru Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yijie Gong
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Mingming Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Rui Guan
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, & Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Ori D Rotstein
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, & Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Xiangdong Liu
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada.,Department of Medicine, & Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
24
|
Grogan A, Tsakiroglou P, Kontrogianni-Konstantopoulos A. Double the trouble: giant proteins with dual kinase activity in the heart. Biophys Rev 2020; 12:1019-1029. [PMID: 32638332 DOI: 10.1007/s12551-020-00715-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022] Open
Abstract
Obscurin and its homolog, striated muscle preferentially expressed gene (SPEG), constitute a unique group of proteins abundantly expressed in striated muscles that contain two tandemly arranged MLCK-like kinases. The physiological significance of the dual kinase motifs is largely understudied; however, a collection of recent studies characterizing their binding interactions, putative targets, and disease-linked mutations have begun to shed light on their potential roles in muscle pathophysiology. Specifically, obscurin kinase 1 is proposed to regulate cardiomyocyte adhesion via phosphorylating N-cadherin, whereas SPEG kinases 1 and 2 regulate Ca2+ cycling by phosphorylating junctophilin-2 and the sarcoendoplasmic Ca2+ ATPase 2 (SERCA2). Herein, we review what is currently known regarding the potential substrates, physiological roles, and disease associations of obscurin and SPEG tandem kinase domains and provide future directions that have yet to be investigated.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD, 21201, USA
| | - Panagiotis Tsakiroglou
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St, Baltimore, MD, 21201, USA
| | | |
Collapse
|
25
|
Hawley MH, Almontashiri N, Biesecker LG, Berger N, Chung WK, Garcia J, Grebe TA, Kelly MA, Lebo MS, Macaya D, Mei H, Platt J, Richard G, Ryan A, Thomson KL, Vatta M, Walsh R, Ware JS, Wheeler M, Zouk H, Mason-Suares H, Funke B. An assessment of the role of vinculin loss of function variants in inherited cardiomyopathy. Hum Mutat 2020; 41:1577-1587. [PMID: 32516855 DOI: 10.1002/humu.24061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/11/2020] [Accepted: 05/24/2020] [Indexed: 11/05/2022]
Abstract
The ACMG/AMP variant classification framework was intended for highly penetrant Mendelian conditions. While it is appreciated that clinically relevant variants exhibit a wide spectrum of penetrance, accurately assessing and expressing the pathogenicity of variants with lower penetrance can be challenging. The vinculin (VCL) gene illustrates these challenges. Model organism data provide evidence that loss of function of VCL may play a role in cardiomyopathy and aggregate case-control studies suggest low penetrance. VCL loss of function variants, however, are rarely identified in affected probands and therefore there is a paucity of family studies clarifying the clinical significance of individual variants. This study, which aggregated data from >18,000 individuals who underwent gene panel or exome testing for inherited cardiomyopathies, identified 32 probands with VCL loss-of-function variants and confirmed enrichment in probands with dilated cardiomyopathy (odds ratio [OR] = 9.01; confidence interval [CI] = 4.93-16.45). Our data revealed that the majority of these individuals (89.5%) had pediatric onset of disease. Family studies demonstrated that heterozygous loss of function of VCL alone is insufficient to cause cardiomyopathy but that these variants do contribute to disease risk. In conclusion, VCL loss-of-function variants should be reported in a diagnostic setting but need to be clearly distinguished as having lower penetrance.
Collapse
Affiliation(s)
- Megan H Hawley
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, Massachusetts
| | - Naif Almontashiri
- Faculty of Applied Medical Sciences, Center for Genetics and Inherited Diseases, Taibah University, Almadinah Almunwarah, Saudi Arabia
| | - Leslie G Biesecker
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Natalie Berger
- Department of Maternal Fetal Medicine, SSM Health St Mary's Hospital, Madison, Wisconsin
| | - Wendy K Chung
- Departments of Pediatrics and Medicine, Columbia University Irving Medical Center, New York, New York
| | - John Garcia
- Invitae Corporation, San Francisco, California
| | - Theresa A Grebe
- Division of Genetics and Metabolism, Department of Child Health, Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona
| | - Melissa A Kelly
- Genomic Medicine Institute, Geisinger, Danville, Pennsylvania
| | - Matthew S Lebo
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, Massachusetts
| | | | - Hui Mei
- GeneDx, Inc, Gaithersburg, Maryland
| | - Julia Platt
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | | | - Ashley Ryan
- Division of Genetics and Metabolism, Department of Child Health, Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona
| | - Kate L Thomson
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - Roddy Walsh
- Department of Clinical and Experimental Cardiology, Heart Centre, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherland
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, UK.,Cardiovascular Research Centre, Royal Brompton and Harefield Hospitals NHS Foundation Trust, Harefield, UK
| | - Matthew Wheeler
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Hana Zouk
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, Massachusetts
| | - Heather Mason-Suares
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, Massachusetts
| | - Birgit Funke
- Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine, Cambridge, Massachusetts
| |
Collapse
|
26
|
Lange S, Pinotsis N, Agarkova I, Ehler E. The M-band: The underestimated part of the sarcomere. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118440. [PMID: 30738787 PMCID: PMC7023976 DOI: 10.1016/j.bbamcr.2019.02.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/16/2019] [Accepted: 02/05/2019] [Indexed: 12/20/2022]
Abstract
The sarcomere is the basic unit of the myofibrils, which mediate skeletal and cardiac Muscle contraction. Two transverse structures, the Z-disc and the M-band, anchor the thin (actin and associated proteins) and thick (myosin and associated proteins) filaments to the elastic filament system composed of titin. A plethora of proteins are known to be integral or associated proteins of the Z-disc and its structural and signalling role in muscle is better understood, while the molecular constituents of the M-band and its function are less well defined. Evidence discussed here suggests that the M-band is important for managing force imbalances during active muscle contraction. Its molecular composition is fine-tuned, especially as far as the structural linkers encoded by members of the myomesin family are concerned and depends on the specific mechanical characteristics of each particular muscle fibre type. Muscle activity signals from the M-band to the nucleus and affects transcription of sarcomeric genes, especially via serum response factor (SRF). Due to its important role as shock absorber in contracting muscle, the M-band is also more and more recognised as a contributor to muscle disease.
Collapse
Affiliation(s)
- Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, San Diego, Medical Sciences Research Bldg, 9500 Gilman Drive, La Jolla, CA 92093-0613C, USA; University of Gothenburg, Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg, Sweden
| | - Nikos Pinotsis
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, Malet Street, London WC1E 7HX, UK
| | - Irina Agarkova
- InSphero, Wagistrasse 27, CH-8952 Schlieren, Switzerland
| | - Elisabeth Ehler
- Randall Centre for Cell and Molecular Biophysics, School of Basic and Medical Biosciences, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK; School of Cardiovascular Medicine and Sciences, British Heart Foundation Research Excellence Centre, King's College London, New Hunt's House, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
27
|
Xu J, Wang L, Liu X, Dai Q. A novel LAMP2 p.G93R mutation associated with mild Danon disease presenting with familial hypertrophic cardiomyopathy. Mol Genet Genomic Med 2019; 7:e00941. [PMID: 31464081 PMCID: PMC6785429 DOI: 10.1002/mgg3.941] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Danon disease (DD) is an X-linked dominant multisystem disorder that is associated with cardiomyopathy, skeletal myopathy, and varying degrees of intellectual disability. It results from mutations in the lysosome-associated membrane protein 2 (LAMP2) gene. METHODS Herein, a proband with a mild DD case presenting with a familial hypertrophic cardiomyopathy (HCM) phenotype and additional family members were evaluated. Exome sequencing and Sanger sequencing were performed to explore the genetic basis of DD in the proband. Segregation, in silico, and functional analyses were carried out to explore potential pathogenicity in the candidate mutation. RESULTS Exome sequencing and Sanger sequencing identified one novel missense mutation (p.G93R) in the LAMP2 gene in the proband, and this mutation was also identified in three other family members. In silico analysis of LAMP2 predicted that the mutation causes a conformational change and subsequent protein destabilization. Furthermore, functional examination showed that mutation carriers have a significant reduction in LAMP2 expression, which supports that the mutation is pathogenic. Moreover, skewed X chromosome inactivation (XCI) was identified in one female mutation carrier, thus suggesting that skewed XCI may be the reason why this individual escaped the pathogenic influence of the mutation. CONCLUSION These findings will aid in diagnosing DD patients carrying this LAMP2 mutation that presents with a HCM phenotype. Furthermore, this study illustrates the importance of utilizing a molecular diagnostic approach in HCM patients and is the first study to report a LAMP2 p.G93R mutation associated with mild DD and identify that XCI serves a protective role in DD etiology.
Collapse
Affiliation(s)
- Jing Xu
- Department of Clinical Laboratory, ZhongDa Hospital, Southeast University, Nanjing, P. R. China.,Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Lu Wang
- Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Xiangdong Liu
- Institute of Life Science, Southeast University, Nanjing, P. R. China
| | - Qiming Dai
- Department of Cardiology, ZhongDa Hospital, Southeast University, Nanjing, P. R. China
| |
Collapse
|
28
|
Refaat MM, Hassanieh S, Ballout JA, Zakka P, Hotait M, Khalil A, Bitar F, Arabi M, Arnaout S, Skouri H, Abchee A, Abi-Saleh B, Khoury M, Massouras A, Nemer G. Non-familial cardiomyopathies in Lebanon: exome sequencing results for five idiopathic cases. BMC Med Genomics 2019; 12:33. [PMID: 30764827 PMCID: PMC6375196 DOI: 10.1186/s12920-019-0478-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Accepted: 01/29/2019] [Indexed: 12/21/2022] Open
Abstract
Background Cardiomyopathies affect more than 0.5% of the general population. They are associated with high risk of sudden cardiac death, which can result from either heart failure or electrical abnormalities. Although different mechanisms underlie the various types of cardiomyopathies, a principal pathology is common to all and is usually at the level of the cardiac muscle. With a relatively high incidence rate in most countries, and a subsequent major health burden on both the families and governments, cardiomyopathies are gaining more attention by researchers and pharmaceutical companies as well as health government bodies. In Lebanon, there is no official data about the spectrum of the diseases in terms of their respective prevalence, clinical, or genetic profiles. Methods We used exome sequencing to unravel the genetic basis of idiopathic cases of cardiomyopathies in Lebanon, a relatively small country with high rates of consanguineous marriages. Results Five cases were diagnosed with different forms of cardiomyopathies, and exome sequencing revealed the presence of already documented or novel mutations in known genes in three cases: LMNA for an Emery Dreifuss Muscular Dystrophy case, PKP2 for an arrhythmogenic right ventricle dysplasia case, and MYPN for a dilated cardiomyopathy case. Interestingly two brothers with hypertrophic cardiomyopathy have a novel missense variation in NPR1, the gene encoding the natriuretic peptides receptor type I, not reported previously to be causing cardiomyopathies. Conclusion Our results unravel novel mutations in known genes implicated in cardiomyopathies in Lebanon. Changes in clinical management however, require genetic profiling of a larger cohort of patients. Electronic supplementary material The online version of this article (10.1186/s12920-019-0478-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marwan M Refaat
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon. .,Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon.
| | - Sylvana Hassanieh
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Jad A Ballout
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Patrick Zakka
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Mostafa Hotait
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Athar Khalil
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Samir Arnaout
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Hadi Skouri
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Antoine Abchee
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Bernard Abi-Saleh
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | - Maurice Khoury
- Department of Internal Medicine, Cardiology Division, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| | | | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, American University of Beirut Faculty of Medicine and Medical Center (AUBMC), Phase I, 8th floor, Room C-823, PO Box 11-0236, Riad El-Solh, Beirut, 1107 2020, Lebanon
| |
Collapse
|
29
|
Grogan A, Kontrogianni-Konstantopoulos A. Unraveling obscurins in heart disease. Pflugers Arch 2018; 471:735-743. [PMID: 30099631 DOI: 10.1007/s00424-018-2191-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/01/2018] [Indexed: 12/18/2022]
Abstract
Obscurins, expressed from the single OBSCN gene, are a family of giant, modular, cytoskeletal proteins that play key structural and regulatory roles in striated muscles. They were first implicated in the development of heart disease in 2007 when two missense mutations were found in a patient diagnosed with hypertrophic cardiomyopathy (HCM). Since then, the discovery of over a dozen missense, frameshift, and splicing mutations that are linked to various forms of cardiomyopathy, including HCM, dilated cardiomyopathy (DCM), and left ventricular non-compaction (LVNC), has highlighted OBSCN as a potential disease-causing gene. At this time, the functional consequences of the identified mutations remain largely elusive, and much work has yet to be done to characterize the disease mechanisms of pathological OBSCN variants. Herein, we describe the OBSCN mutations known to date, discuss their potential impact on disease development, and provide future directions in order to better understand the involvement of obscurins in heart disease.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene St., Baltimore, MD, 21201, USA
| | | |
Collapse
|
30
|
Mak TSH, Lee YK, Tang CS, Hai JSH, Ran X, Sham PC, Tse HF. Coverage and diagnostic yield of Whole Exome Sequencing for the Evaluation of Cases with Dilated and Hypertrophic Cardiomyopathy. Sci Rep 2018; 8:10846. [PMID: 30022097 PMCID: PMC6052112 DOI: 10.1038/s41598-018-29263-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 07/04/2018] [Indexed: 12/14/2022] Open
Abstract
Targeted next generation sequencing of gene panels has become a popular tool for the genetic diagnosis of hypertrophic (HCM) and dilated cardiomyopathy (DCM). However, it is uncertain whether the use of Whole Exome Sequencing (WES) represents a more effective approach for diagnosis of cases with HCM and DCM. In this study, we performed indirect comparisons of the coverage and diagnostic yield of WES on genes and variants related to HCM and DCM versus 4 different commercial gene panels using 40 HCM and DCM patients, assuming perfect coverage in those panels. We identified 6 pathogenic or likely pathogenic among 14 HCM patients (diagnostic yield 43%). 3 pathogenic or likely pathogenic were found among the 26 DCM patients (diagnostic yield 12%). The coverage was similar to that of four existing commercial gene panels due to the clustering of mutation within MYH7, MYBPC3, TPM1, TNT2, and TTN. Moreover, the coverage of WES appeared inadequate for TNNI3 and PLN. We conclude that most of the pathogenic variants for HCM and DCM can be found within a small number of genes which were covered by all the commercial gene panels, and the application of WES did not increase diagnostic yield.
Collapse
Affiliation(s)
- Timothy Shin Heng Mak
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yee-Ki Lee
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Clara S Tang
- Department of Surgery, the University of Hong Kong, Hong Kong, China
- The University of Hong Kong - Karolinska Institutet Collaboration in Regenerative Medicine, Hong Kong, China
| | - JoJo S H Hai
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Xinru Ran
- Department of Medicine, the University of Hong Kong, Hong Kong, China
| | - Pak-Chung Sham
- Centre for Genomic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Department of Psychiatry, the University of Hong Kong, Hong Kong, China.
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.
| | - Hung-Fat Tse
- Department of Medicine, the University of Hong Kong, Hong Kong, China.
- Department of Medicine, Shenzhen Hong Kong University Hospital, Shenzhen, China.
- Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, the University of Hong Kong, Hong Kong, China.
- Shenzhen Institutes of Research and Innovation, the University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
31
|
Jumaah S, Çelekli A, Sucu M. The role of human urotensin-II in patients with hypertrophic cardiomyopathy. J Immunoassay Immunochem 2018; 39:150-162. [PMID: 28686108 DOI: 10.1080/15321819.2017.1344130] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) is a genetic condition with the hallmark feature of left ventricular hypertrophy. Human Urotensin-II (hUT-II) is regarded as a cardiovascular autacoid/hormone, and it has cardiac inotropic and hypertrophic properties. Aims of this study were to elucidate the clinical significance of serum hUT-II levels as a potential new biomarker in patients with HCM. METHODS This study included 40 HCM patients (60% males and 40% females) and were compared to 30 healthy control subjects (47% males and 53% females. All patients underwent extensive clinical, laboratory, and echocardiographic. Blood samples were taken to test for serum hUT-II levels by commercial ELISA Kit. RESULTS Serum hUT-II was significantly higher (p < 0.01) in patients with HCM (15.8 ± 2.1 pmol/L) compared with healthy controls (3.3 ± 1.7 pmol/L). With regard to HCM patient, Serum hUT-II levels were significantly higher in the female with 16.3 ± 1.9 pmol/L than the male with 15.4 ± 2.2 pmol/L (p < 0.05). Among echocardiographic parameters, hUT-II was negatively associated with ejection fraction (r = -0.160, p = 0.324). CONCLUSION Results of the first study indicated that serum hUT-II levels were markedly elevated in patients with HCM. Serum hUT-II is a novel biomarker parameter that has clinical use in patients with the severity of LVH.
Collapse
Affiliation(s)
- Saman Jumaah
- a Institute of Natural and Applied Sciences, Department of Biochemistry Science and Technology , Gaziantep University , Gaziantep , Turkey
| | - Abuzer Çelekli
- b Faculty of Arts and Science, Department of Biology , Gaziantep University , Gaziantep , Turkey
| | - Murat Sucu
- c Medical Faculty Cardiology , Gaziantep University , Gaziantep , Turkey
| |
Collapse
|
32
|
Li L, Liu H, Hu X, Huang Y, Wang Y, He Y, Lei Q. Identification of key genes in non‑alcoholic fatty liver disease progression based on bioinformatics analysis. Mol Med Rep 2018; 17:7708-7720. [PMID: 29620197 PMCID: PMC5983972 DOI: 10.3892/mmr.2018.8852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/22/2018] [Indexed: 12/18/2022] Open
Abstract
Due to economic development and lifestyle changes, the incidence of non-alcoholic fatty liver disease (NAFLD) has gradually increased in recent years. However, the pathogenesis of NAFLD is not yet fully understood. To identify candidate genes that contribute to the development and progression of NAFLD, two microarray datasets were downloaded from the Gene Expression Omnibus database. The differentially expressed genes (DEGs) were identified and functional enrichment analyses were performed. A protein-protein interaction network was constructed and modules were extracted using the Search Tool for the Retrieval of Interacting Genes and Cytoscape. The enriched functions and pathways of the DEGs included ‘cellular macromolecule biosynthetic process’, ‘cellular response to chemical stimulus’, ‘extracellular matrix organization’, ‘metabolic pathways’, ‘insulin resistance’ and ‘forkhead box protein O1 signaling pathway’. The DEGs, including type-1 angiotensin II receptor, formin-binding protein 1-like, RNA-binding protein with serine-rich domain 1, Ras-related C3 botulinum toxin substrate 1 and polyubiquitin-C, were identified using multiple bioinformatics methods and validated in vitro with reverse transcription-quantitative polymerase chain reaction analysis. In conclusion, five hub genes were identified in the present study, and they may aid in understanding of the molecular mechanisms underlying the development and progression of NAFLD.
Collapse
Affiliation(s)
- Lin Li
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Huabao Liu
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Xiaoyu Hu
- Department of Infectious Disease, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Yi Huang
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Yanan Wang
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Yansha He
- Department of Liver Disease, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400021, P.R. China
| | - Qingsong Lei
- Department of Infectious Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
33
|
Wang L, Geist J, Grogan A, Hu LYR, Kontrogianni-Konstantopoulos A. Thick Filament Protein Network, Functions, and Disease Association. Compr Physiol 2018; 8:631-709. [PMID: 29687901 PMCID: PMC6404781 DOI: 10.1002/cphy.c170023] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sarcomeres consist of highly ordered arrays of thick myosin and thin actin filaments along with accessory proteins. Thick filaments occupy the center of sarcomeres where they partially overlap with thin filaments. The sliding of thick filaments past thin filaments is a highly regulated process that occurs in an ATP-dependent manner driving muscle contraction. In addition to myosin that makes up the backbone of the thick filament, four other proteins which are intimately bound to the thick filament, myosin binding protein-C, titin, myomesin, and obscurin play important structural and regulatory roles. Consistent with this, mutations in the respective genes have been associated with idiopathic and congenital forms of skeletal and cardiac myopathies. In this review, we aim to summarize our current knowledge on the molecular structure, subcellular localization, interacting partners, function, modulation via posttranslational modifications, and disease involvement of these five major proteins that comprise the thick filament of striated muscle cells. © 2018 American Physiological Society. Compr Physiol 8:631-709, 2018.
Collapse
Affiliation(s)
- Li Wang
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Janelle Geist
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | | |
Collapse
|
34
|
Mechanical dyssynchrony and diastolic dysfunction are common in LVH: a pilot correlation study using Doppler echocardiography and CZT gated-SPECT MPI. Sci Rep 2018. [PMID: 29520004 PMCID: PMC5843584 DOI: 10.1038/s41598-018-22213-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an often under-diagnosed cause of left ventricular hypertrophy (LVH). It affects 1/500 of the population, is the most commonly inherited cardiovascular disorder, and can present in apical, concentric, or septal forms. Although most patients are asymptomatic, sudden cardiac death can be the initial presentation of HCM. By retrospectively enrolling patients suspected of having three different types of HCM in the absence of epicardial coronary stenosis, we aimed to examine systolic and diastolic dysfunction and perfusion abnormalities using both Doppler echocardiography and state-of-the-art gated single-photon emission computerized tomography (SPECT) myocardial perfusion imaging (MPI) with a cadmium-zinc-telluride camera and thallium-201. Both regional perfusion and gated SPECT parameters were collected in addition to diastolic parameters from Doppler echocardiography. The results showed that mild ischemia was common in patients suspected of having HCM, with a mean summed stress score of 4.7 ± 4.9 (score 0–4 in 17-segment model). The patients with HCM were associated with discernible left ventricular mechanical dyssynchrony, especially those with the apical form. In addition, diastolic dysfunction was prevalent and early to late ventricular filling velocity ratios were significantly different between groups. By combining gated-MPI and Doppler data, the trivial functional changes in HCM may be identified.
Collapse
|
35
|
Jin ZB, Li Z, Liu Z, Jiang Y, Cai XB, Wu J. Identification of de novo germline mutations and causal genes for sporadic diseases using trio-based whole-exome/genome sequencing. Biol Rev Camb Philos Soc 2017; 93:1014-1031. [PMID: 29154454 DOI: 10.1111/brv.12383] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 12/14/2022]
Abstract
Whole-genome or whole-exome sequencing (WGS/WES) of the affected proband together with normal parents (trio) is commonly adopted to identify de novo germline mutations (DNMs) underlying sporadic cases of various genetic disorders. However, our current knowledge of the occurrence and functional effects of DNMs remains limited and accurately identifying the disease-causing DNM from a group of irrelevant DNMs is complicated. Herein, we provide a general-purpose discussion of important issues related to pathogenic gene identification based on trio-based WGS/WES data. Specifically, the relevance of DNMs to human sporadic diseases, current knowledge of DNM biogenesis mechanisms, and common strategies or software tools used for DNM detection are reviewed, followed by a discussion of pathogenic gene prioritization. In addition, several key factors that may affect DNM identification accuracy and causal gene prioritization are reviewed. Based on recent major advances, this review both sheds light on how trio-based WGS/WES technologies can play a significant role in the identification of DNMs and causal genes for sporadic diseases, and also discusses existing challenges.
Collapse
Affiliation(s)
- Zi-Bing Jin
- Division of Ophthalmic Genetics, The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China.,State Key Laboratory of Ophthalmology Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhongshan Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhenwei Liu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yi Jiang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xue-Bi Cai
- Division of Ophthalmic Genetics, The Eye Hospital, School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, 325027, China.,State Key Laboratory of Ophthalmology Optometry and Vision Science, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jinyu Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
36
|
Ackermann MA, King B, Lieberman NAP, Bobbili PJ, Rudloff M, Berndsen CE, Wright NT, Hecker PA, Kontrogianni-Konstantopoulos A. Novel obscurins mediate cardiomyocyte adhesion and size via the PI3K/AKT/mTOR signaling pathway. J Mol Cell Cardiol 2017; 111:27-39. [PMID: 28826662 PMCID: PMC5694667 DOI: 10.1016/j.yjmcc.2017.08.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 08/02/2017] [Accepted: 08/03/2017] [Indexed: 12/29/2022]
Abstract
The intercalated disc of cardiac muscle embodies a highly-ordered, multifunctional network, essential for the synchronous contraction of the heart. Over 200 known proteins localize to the intercalated disc. The challenge now lies in their characterization as it relates to the coupling of neighboring cells and whole heart function. Using molecular, biochemical and imaging techniques, we characterized for the first time two small obscurin isoforms, obscurin-40 and obscurin-80, which are enriched at distinct locations of the intercalated disc. Both proteins bind specifically and directly to select phospholipids via their pleckstrin homology (PH) domain. Overexpression of either isoform or the PH-domain in cardiomyocytes results in decreased cell adhesion and size via reduced activation of the PI3K/AKT/mTOR pathway that is intimately linked to cardiac hypertrophy. In addition, obscurin-80 and obscurin-40 are significantly reduced in acute (myocardial infarction) and chronic (pressure overload) murine cardiac-stress models underscoring their key role in maintaining cardiac homeostasis. Our novel findings implicate small obscurins in the maintenance of cardiomyocyte size and coupling, and the development of heart failure by antagonizing the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Maegen A Ackermann
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States; Department of Physiology and Cell Biology, Wexner College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States.
| | - Brendan King
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States
| | - Nicole A P Lieberman
- Department of Biochemistry and Molecular Biology, University of Maryland, School of Medicine, Baltimore, MD 21201, United States
| | - Prameela J Bobbili
- Department of Physiology and Cell Biology, Wexner College of Medicine, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, United States
| | - Michael Rudloff
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Christopher E Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, United States
| | - Peter A Hecker
- Division of Cardiology and Department of Medicine, University of Maryland, Baltimore, MD 20201, United States
| | | |
Collapse
|
37
|
Ryanodine receptors are part of the myospryn complex in cardiac muscle. Sci Rep 2017; 7:6312. [PMID: 28740084 PMCID: PMC5524797 DOI: 10.1038/s41598-017-06395-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/12/2017] [Indexed: 01/28/2023] Open
Abstract
The Cardiomyopathy-associated gene 5 (Cmya5) encodes myospryn, a large tripartite motif (TRIM)-related protein found predominantly in cardiac and skeletal muscle. Cmya5 is an expression biomarker for a number of diseases affecting striated muscle and may also be a schizophrenia risk gene. To further understand the function of myospryn in striated muscle, we searched for additional myospryn paralogs. Here we identify a novel muscle-expressed TRIM-related protein minispryn, encoded by Fsd2, that has extensive sequence similarity with the C-terminus of myospryn. Cmya5 and Fsd2 appear to have originated by a chromosomal duplication and are found within evolutionarily-conserved gene clusters on different chromosomes. Using immunoaffinity purification and mass spectrometry we show that minispryn co-purifies with myospryn and the major cardiac ryanodine receptor (RyR2) from heart. Accordingly, myospryn, minispryn and RyR2 co-localise at the junctional sarcoplasmic reticulum of isolated cardiomyocytes. Myospryn redistributes RyR2 into clusters when co-expressed in heterologous cells whereas minispryn lacks this activity. Together these data suggest a novel role for the myospryn complex in the assembly of ryanodine receptor clusters in striated muscle.
Collapse
|
38
|
Hu LYR, Ackermann MA, Hecker PA, Prosser BL, King B, O’Connell KA, Grogan A, Meyer LC, Berndsen CE, Wright NT, Jonathan Lederer W, Kontrogianni-Konstantopoulos A. Deregulated Ca 2+ cycling underlies the development of arrhythmia and heart disease due to mutant obscurin. SCIENCE ADVANCES 2017; 3:e1603081. [PMID: 28630914 PMCID: PMC5462502 DOI: 10.1126/sciadv.1603081] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/17/2017] [Indexed: 05/05/2023]
Abstract
Obscurins are cytoskeletal proteins with structural and regulatory roles encoded by OBSCN. Mutations in OBSCN are associated with the development of hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Specifically, the R4344Q mutation present in immunoglobulin domain 58 (Ig58) was the first to be linked with the development of HCM. To assess the effects of R4344Q in vivo, we generated the respective knock-in mouse model. Mutant obscurins are expressed and incorporated normally into sarcomeres. The expression patterns of sarcomeric and Ca2+-cycling proteins are unaltered in sedentary 1-year-old knock-in myocardia, with the exception of sarco/endoplasmic reticulum Ca2+ adenosine triphosphatase 2 (SERCA2) and pentameric phospholamban whose levels are significantly increased and decreased, respectively. Isolated cardiomyocytes from 1-year-old knock-in hearts exhibit increased Ca2+-transients and Ca2+-load in the sarcoplasmic reticulum and faster contractility kinetics. Moreover, sedentary 1-year-old knock-in animals develop tachycardia accompanied by premature ventricular contractions, whereas 2-month-old knock-in animals subjected to pressure overload develop a DCM-like phenotype. Structural analysis revealed that the R4344Q mutation alters the distribution of electrostatic charges over the Ig58 surface, thus interfering with its binding capabilities. Consistent with this, wild-type Ig58 interacts with phospholamban modestly, and this interaction is markedly enhanced in the presence of R4344Q. Together, our studies demonstrate that under sedentary conditions, the R4344Q mutation results in Ca2+ deregulation and spontaneous arrhythmia, whereas in the presence of chronic, pathological stress, it leads to cardiac remodeling and dilation. We postulate that enhanced binding between mutant obscurins and phospholamban leads to SERCA2 disinhibition, which may underlie the observed pathological alterations.
Collapse
Affiliation(s)
- Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Maegen A. Ackermann
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Peter A. Hecker
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Benjamin L. Prosser
- Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brendan King
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Kelly A. O’Connell
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Logan C. Meyer
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Christopher E. Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Nathan T. Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - W. Jonathan Lederer
- Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
39
|
Abstract
The inherited cardiomyopathies, hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) and left ventricular non-compaction (LVNC), have been frequently associated with mutations in sarcomeric proteins. In recent years, advances in DNA sequencing technology has allowed the study of the giant proteins of the sarcomere, such as titin and nebulin. Obscurin has been somewhat neglected in these studies, largely because its functional role is far from clear, although there was an isolated report in 2007 of obscurin mutations associated with HCM. Recently, whole exome sequencing methodology (WES) has been used to address mutations in OBSCN, the gene for obscurin, and OBSCN variants were found to be relatively common in inherited cardiomyopathies. In different studies, 5 OBSCN unique variants have been found in a group of 30 end-stage failing hearts, 6 OBSCN unique variants in 74 HCM cases and 3 OBSCN unique variants in 10 LVNC patients. As yet, the number of known potentially disease-causing OBSCN variants is quite small. The reason for this is that mutations in the OBSCN gene have not been recognised as potentially disease-causing until recently, and were not included in large-scale genetic surveys. OBSCN mutations may be causative of HCM, DCM and LVNC and other cardiomyopathies, or they may work in concert with other variants in the same or other genes to initiate the pathology. Currently, the function of obscurin is not well understood, but we anticipate that many more OBSCN variants linked to cardiomyopathy will be found when the large cohorts of patient sequences available are tested. It is to be hoped that the establishment of the importance of obscurin in pathology will stimulate a thorough investigation of obscurin function.
Collapse
|
40
|
Pillar N, Pleniceanu O, Fang M, Ziv L, Lahav E, Botchan S, Cheng L, Dekel B, Shomron N. A rare variant in the FHL1 gene associated with X-linked recessive hypoparathyroidism. Hum Genet 2017; 136:835-845. [PMID: 28444561 PMCID: PMC5487855 DOI: 10.1007/s00439-017-1804-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 04/17/2017] [Indexed: 12/12/2022]
Abstract
Isolated familial hypoparathyroidism is an extremely rare disorder, which to date has been linked to several loci including mutations in CASR, GCM2, and PTH, as well as a rare condition defined as X-linked recessive hypoparathyroidism, previously associated with a 1.5 Mb region on Xq26-q27. Here, we report a patient with hypocalcemia-induced seizures leading to the diagnosis of primary hypoparathyroidism. Mutations in CASR, GCM2, and PTH were ruled out, while whole exome sequencing of the family suggested FHL1, located on chromosome Xq26, as the most likely causative gene variant (FHL1, exon 4, c.C283T, p.R95W). Since FHL1 has not been linked to calcium regulation before, we provide evidence for its functional role in hypoparathyroidism by: (i) bioinformatics analysis coupling its action to known modulators of PTH function; (ii) observing strong expression of fhl1b in Corpuscles of Stannius, gland-like aggregates in zebrafish that function in calcium regulation similar to mammalian PTH; and (iii) implicating fhl1b and FHL1 as regulators of calcium homeostasis in zebrafish and human cells, respectively. Altogether, our data suggest that FHL1 is a novel regulator of calcium homeostasis and implicate it as the causative gene for X-linked recessive hypoparathyroidism.
Collapse
Affiliation(s)
- Nir Pillar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Stem Cell Research Institute & Division of Pediatric Nephrology, Edmond & Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | | | - Limor Ziv
- Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Einat Lahav
- Pediatric Stem Cell Research Institute & Division of Pediatric Nephrology, Edmond & Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel
| | - Shay Botchan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Benjamin Dekel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Pediatric Stem Cell Research Institute & Division of Pediatric Nephrology, Edmond & Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel.
| | - Noam Shomron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
41
|
Alonso-Montes C, Rodríguez-Reguero J, Martín M, Gómez J, Coto E, Naves-Díaz M, Morís C, Cannata-Andía JB, Rodríguez I. Rare genetic variants in GATA transcription factors in patients with hypertrophic cardiomyopathy. J Investig Med 2017; 65:926-934. [PMID: 28381408 DOI: 10.1136/jim-2016-000364] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2017] [Indexed: 11/03/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) is a very heterogeneous disease. Although primarily caused by mutations in genes encoding sarcomeric proteins, other genes might explain that heterogeneity. Potential candidate genes are GATA transcription factors that regulate the expression of proteins associated with HCM. Exons of GATA2, GATA4, and GATA6 genes were sequenced in 212 patients with unrelated HCM previously analyzed for genes encoding the most frequently mutated sarcomeric proteins. Functional effects of variants were predicted by in silico analyses. 3 potentially pathogenic variants were identified: c.-77G>A in GATA2, p.Ala343Thr (rs370588269) in GATA4, and p.Pro555Ala (rs146243018) in GATA6 Multivariate analyses showed that angina was more frequent in patients carrying sarcomeric and GATA rare variants (55% vs 23.2% in non-carriers of GATA rare variants, OR (95% CI) 7.12 (1.23 to 41.27), p=0.029). Among patients without a known causal mutation, GATA rare variants were associated with a greater maximum posterior wall thickness (16.4±4.4 vs 14.0±3.1 mm in non-carriers, p=0.021). Thus, variants having a putative effect on GATA genes would alter the expression of their target genes and could modify the hypertrophic response. Therefore, although relatively infrequent in patients with HCM, they may represent a novel insight into the molecular mechanisms related to the pathogenesis of HCM.
Collapse
Affiliation(s)
- Cristina Alonso-Montes
- Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain.,Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain
| | - Julián Rodríguez-Reguero
- Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Cardiology Department, Fundación Asturcor, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - María Martín
- Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Cardiology Department, Fundación Asturcor, Hospital Universitario Central de Asturias, Oviedo, Spain.,Molecular Genetics Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Juan Gómez
- Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Universidad de Oviedo, Oviedo, Spain
| | - Eliecer Coto
- Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Molecular Genetics Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain.,Universidad de Oviedo, Oviedo, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain.,Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain
| | - César Morís
- Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Cardiology Department, Fundación Asturcor, Hospital Universitario Central de Asturias, Oviedo, Spain.,Molecular Genetics Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain.,Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain.,Molecular Genetics Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Isabel Rodríguez
- Bone and Mineral Research Unit, Instituto Reina Sofía de Investigación, Hospital Universitario Central de Asturias, Oviedo, Spain.,Red de Investigación Renal REDinREN from Instituto de Salud Carlos III, Oviedo, Spain
| |
Collapse
|
42
|
Manring HR, Carter OA, Ackermann MA. Obscure functions: the location-function relationship of obscurins. Biophys Rev 2017; 9:245-258. [PMID: 28510116 DOI: 10.1007/s12551-017-0254-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/05/2017] [Indexed: 12/18/2022] Open
Abstract
The obscurin family of polypeptides is essential for normal striated muscle function and contributes to the pathogenesis of fatal diseases, including cardiomyopathies and cancers. The single mammalian obscurin gene, OBSCN, gives rise to giant (∼800 kDa) and smaller (∼40-500 kDa) proteins that are composed of tandem adhesion and signaling motifs. Mammalian obscurin proteins are expressed in a variety of cell types, including striated muscles, and localize to distinct subcellular compartments where they contribute to diverse cellular processes. Obscurin homologs in Caenorhabditis elegans and Drosophila possess a similar domain architecture and are also expressed in striated muscles. The long sought after question, "what does obscurin do?" is complex and cannot be addressed without taking into consideration the subcellular distribution of these proteins and local isoform concentration. Herein, we present an overview of the functions of obscurins and begin to define the intricate relationship between their subcellular distributions and functions in striated muscles.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Olivia A Carter
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA. .,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
43
|
Popa MO, Irimia AM, Papagheorghe MN, Vasile EM, Tircol SA, Negulescu RA, Toader C, Adam R, Dorobantu L, Caldararu C, Alexandrescu M, Onciul S. The mechanisms, diagnosis and management of mitral regurgitation in mitral valve prolapse and hypertrophic cardiomyopathy. Discoveries (Craiova) 2016; 4:e61. [PMID: 32309580 PMCID: PMC7159827 DOI: 10.15190/d.2016.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Valvular disease is a frequent cardiac pathology leading to heart failure and, ultimately, death. Mitral regurgitation, defined as the inability of the two mitral leaflets to coapt, is a common valvular disease and a self sustained pathology. A better understanding of the mitral valve histological layers provides a better understanding of the leaflet and chordae changes in mitral valve prolapse.
Mitral valve prolapse may occur in myxomatous degenerative abnormalities, connective tissue disorders or in sporadic isolated cases. It is the most common mitral abnormality of non-ischemic cause leading to severe surgery-requiring mitral regurgitation. In addition to standard echocardiographic investigations, newly implemented three-dimensional techniques are being used and they permit a better visualisation, from the so-called ‘surgical view’, and an improved evaluation of the mitral valve.
Hypertrophic cardiomyopathy is the most frequent inherited myocardial disease caused by mutations in various genes encoding proteins of the cardiac sarcomere, leading to a marked left ventricular hypertrophy unexplained by other comorbidities. The pathological echocardiographic hallmarks of hypertrophic cardiomyopathy are left ventricular hypertrophy, left ventricular outflow tract obstruction and systolic anterior motion of the mitral valve. The systolic anterior motion of the mitral valve contributes to the development of mitral regurgitation and further narrows the left ventricular outflow tract, leading to more severe symptomatology. Cardiac magnetic resonance imaging accurately measures the left ventricular mass, the degree of diastolic function and it may also be used to distinguish phenotypic variants.
The clinical outcome of patients with these pathologies is mostly determined by the selected option of treatment. The purpose of surgical correction regarding mitral valve involvement is to restore valvular competence. Surgery has proven to be the only useful treatment in preventing heart failure, improving symptomatology and reducing mortality. Our approach wishes to enhance the understanding of the mitral valve’s involvement in hypertrophic cardiomyopathy and mitral valve prolapse from genetic, haemodynamic and clinical perspectives, as well as to present novelties in the grand field of treatment.
Collapse
Affiliation(s)
| | - Ana Maria Irimia
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | | | | | | | | | - Catalina Toader
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Robert Adam
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Lucian Dorobantu
- Department of Cardiovascular Surgery, Monza Hospital, Bucharest, Romania
| | | | - Maria Alexandrescu
- Department of Radiology and Imaging Sciences, Monza Hospital, Bucharest, Romania
| | - Sebastian Onciul
- Department of Cardiology, Floreasca Clinical Emergency Hospital, Bucharest, Romania
| |
Collapse
|