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
|
Fujita K, Desmond P, Blondelle J, Soták M, Rajan MR, Clark M, Estève É, Chan Y, Gu Y, Actis Dato V, Marrocco V, Dalton ND, Ghassemian M, Do A, Klos M, Peterson KL, Sheikh F, Cho Y, Börgeson E, Lange S. Combined Loss of Obsc and Obsl1 in Murine Hearts Results in Diastolic Dysfunction, Altered Metabolism, and Deregulated Mitophagy. Circ Heart Fail 2025; 18:e011867. [PMID: 40066567 PMCID: PMC11995854 DOI: 10.1161/circheartfailure.124.011867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/09/2025] [Indexed: 04/03/2025]
Abstract
BACKGROUND Muscle proteins of the obscurin protein family play important roles in sarcomere organization and sarcoplasmic reticulum and T-tubule architecture and function. However, their precise molecular functions and redundancies between protein family members as well as their involvement in cardiac diseases remain to be fully understood. METHODS To investigate the functional roles of Obsc (obscurin) and its close homolog Obsl1 (obscurin-like 1) in the heart, we generated and analyzed knockout mice for Obsc, Obsl1, as well as Obsc/Obsl1 double knockouts. RESULTS We show that double-knockout mice are viable but show postnatal deficits in cardiac muscle sarcoplasmic reticulum and mitochondrial architecture and function at the microscopic, biochemical, and cellular levels. Altered sarcoplasmic reticulum structure resulted in perturbed calcium cycling, whereas mitochondrial ultrastructure deficits were linked to decreased levels of Chchd3 (coiled-coil-helix-coiled-coil-helix domain containing 3), a Micos (mitochondrial contact site and cristae organizing system) complex protein. Hearts of double-knockout mice also show altered levels of Atg4 proteins, novel Obsl1 interactors, resulting in abnormal mitophagy, and increased unfolded protein response. At the physiological level, loss of obscurin and Obsl1 resulted in a profound delay of cardiac relaxation, associated with metabolic signs of heart failure. CONCLUSIONS Taken together, our data suggest that Obsc and Obsl1 play crucial roles in cardiac sarcoplasmic reticulum structure, calcium cycling, mitochondrial function, turnover, and metabolism.
Collapse
Affiliation(s)
- Kyohei Fujita
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women’s Medical University, Japan (K.F.)
| | - Patrick Desmond
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Jordan Blondelle
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Matúš Soták
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
| | - Meenu Rohini Rajan
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
| | - Madison Clark
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| | - Éric Estève
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- PhyMedExp, University of Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Regionale Universitaire (CHRU) Montpellier, France (E.E.)
| | - Yunghang Chan
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Yusu Gu
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Virginia Actis Dato
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Valeria Marrocco
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Nancy D. Dalton
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Majid Ghassemian
- Department of Chemistry and Biochemistry (M.G.), University of California San Diego, La Jolla
| | - Aryanne Do
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Matthew Klos
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Kirk L. Peterson
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Farah Sheikh
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Yoshitake Cho
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
| | - Emma Börgeson
- Clinical Immunology and Transfusion Medicine, Sahlgrenska University Hospital, University of Gothenburg, Sweden (M.S., M.R.R., E.B.)
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| | - Stephan Lange
- Division of Cardiovascular Medicine, School of Medicine (K.F., P.D., J.B., M.C., E.E., Y. Chan, Y.G., V.A.D., V.M., N.D.D., A.D., M.K., K.L.P., F.S., Y. Cho, S.L.), University of California San Diego, La Jolla
- Department of Biomedicine, Aarhus University, Denmark (M.C., E.B., S.L.)
- STENO Diabetes Center Aarhus, Denmark (M.C., E.B., S.L.)
| |
Collapse
|
3
|
Sun X, Chen Y, Zhong J, Chen H, Xie J, Wang R. Identification of Compound Heterozygous Variants in OBSCN Gene Associated With Rhabdomyolysis: A Case Report. Mol Genet Genomic Med 2025; 13:e70094. [PMID: 40186404 PMCID: PMC11971531 DOI: 10.1002/mgg3.70094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/10/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The obscurin protein encoded by the OBSCN gene is an important structural protein in the regulation of myocyte sarcoplasmic nodule stability and sarcoplasmic reticulum function and is particularly closely associated with calcium ion (Ca2+) signaling. With increasing genomic studies, pathogenic variants in the OBSCN gene have been shown to be associated with a variety of inherited diseases, such as cardiomyopathy. However, case reports of its variants causing rhabdomyolysis are more limited. METHODS We performed whole exome sequencing on a patient with exercise-induced rhabdomyolysis to identify possible causative gene variants. In addition, functional prediction of the pathogenicity of the variants was performed by combining multiple bioinformatics analysis tools and in-depth analyses with clinical phenotypes and family history. RESULTS The patient carried compound heterozygous variants, including c.21184C>T (nonsense variant) and c.15610+12C>T (intronic splicing variant). The c.21184C>T variant resulted in a premature termination of the protein, was not included in population-based databases, and was supported by multiple prediction tools as a potentially pathogenic variant. The c.15610+12C>T variant was also absent in the gnomAD_EAS database and predicted to disturb normal splicing, potentially creating a novel donor site. The pathogenicity of the variant is further supported by the fact that the patient's mother, with a homozygous OBSCN variant, also exhibited exercise-induced myalgia. Clinically, the patient presented with exercise-induced rhabdomyolysis accompanied by significant serum creatine kinase elevation, muscle pain, and MRI-demonstrated muscle edema of both lower limbs without significant muscle weakness or cardiac abnormalities. CONCLUSION We report the first case of rhabdomyolysis in China caused by OBSCN gene variants. This finding further extends the spectrum of the OBSCN gene variants. It also provides an important basis for genetic counseling and helps in the early diagnosis and management of similar cases.
Collapse
Affiliation(s)
- Xiaolan Sun
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| | - Yong Chen
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| | - Jianmin Zhong
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| | - Hui Chen
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| | - Jihua Xie
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| | - Ruiyan Wang
- Department of NeurologyJiangxi Provincial Children's HospitalNanchangChina
| |
Collapse
|
4
|
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
|
5
|
Kahsay A, Dennhag N, Liu JX, Nord H, Rönnbäck H, Thorell AE, von Hofsten J, Pedrosa Domellöf F. Obscurin Maintains Myofiber Identity in Extraocular Muscles. Invest Ophthalmol Vis Sci 2024; 65:19. [PMID: 38334702 PMCID: PMC10860686 DOI: 10.1167/iovs.65.2.19] [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: 11/24/2023] [Accepted: 01/12/2024] [Indexed: 02/10/2024] Open
Abstract
Purpose The cytoskeleton of the extraocular muscles (EOMs) is significantly different from that of other muscles. We aimed to investigate the role of obscurin, a fundamental cytoskeletal protein, in the EOMs. Methods The distribution of obscurin in human and zebrafish EOMs was compared using immunohistochemistry. The two obscurin genes in zebrafish, obscna and obscnb, were knocked out using CRISPR/Cas9, and the EOMs were investigated using immunohistochemistry, qPCR, and in situ hybridization. The optokinetic reflex (OKR) in five-day-old larvae and adult obscna-/-;obscnb-/- and sibling control zebrafish was analyzed. Swimming distance was recorded at the same age. Results The obscurin distribution pattern was similar in human and zebrafish EOMs. The proportion of slow and fast myofibers was reduced in obscna-/-;obscnb-/- zebrafish EOMs but not in trunk muscle, whereas the number of myofibers containing cardiac myosin myh7 was significantly increased in EOMs of obscurin double mutants. Loss of obscurin resulted in less OKRs in zebrafish larvae but not in adult zebrafish. Conclusions Obscurin expression is conserved in normal human and zebrafish EOMs. Loss of obscurin induces a myofiber type shift in the EOMs, with upregulation of cardiac myosin heavy chain, myh7, showing an adaptation strategy in EOMs. Our model will facilitate further studies in conditions related to obscurin.
Collapse
Affiliation(s)
- Abraha Kahsay
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Nils Dennhag
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | - Jing-Xia Liu
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Hanna Nord
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Hugo Rönnbäck
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| | | | - Jonas von Hofsten
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
| | - Fatima Pedrosa Domellöf
- Department of Integrative Medical Biology (IMB), Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
| |
Collapse
|
6
|
Grogan A, Huang W, Brong A, Kane MA, Kontrogianni-Konstantopoulos A. Alterations in cytoskeletal and Ca 2+ cycling regulators in atria lacking the obscurin Ig58/59 module. Front Cardiovasc Med 2023; 10:1085840. [PMID: 37304957 PMCID: PMC10251194 DOI: 10.3389/fcvm.2023.1085840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/26/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Obscurin (720-870 kDa) is a giant cytoskeletal and signaling protein that possesses both structural and regulatory functions in striated muscles. Immunoglobulin domains 58/59 (Ig58/59) of obscurin bind to a diverse set of proteins that are essential for the proper structure and function of the heart, including giant titin, novex-3, and phospholamban (PLN). Importantly, the pathophysiological significance of the Ig58/59 module has been further underscored by the discovery of several mutations within Ig58/59 that are linked to various forms of myopathy in humans. We previously generated a constitutive deletion mouse model, Obscn-ΔIg58/59, that expresses obscurin lacking Ig58/59, and characterized the effects of this deletion on cardiac morphology and function through aging. Our findings demonstrated that Obscn-ΔIg58/59 male animals develop severe arrhythmia, primarily manifesting as episodes of junctional escape and spontaneous loss of regular p-waves, reminiscent of human atrial fibrillation, accompanied by significant atrial enlargement that progresses in severity with aging. Methods and Results To comprehensively characterize the molecular alterations responsible for these pathologies, we performed proteomic and phospho-proteomic analyses in aging Obscn-ΔIg58/59 atria. Our studies revealed extensive and novel alterations in the expression and phosphorylation profile of major cytoskeletal proteins, Ca2+ regulators, and Z-disk associated protein complexes in the Obscn-ΔIg58/59 atria through aging. Discussion These studies implicate obscurin, particularly the Ig58/59 module, as an essential regulator of the Z-disk associated cytoskeleton and Ca2+ cycling in the atria and provide new molecular insights into the development of atrial fibrillation and remodeling.
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, United States
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | - Annie Brong
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, MD, United States
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD, United States
| | | |
Collapse
|
7
|
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
|
8
|
The necessity of incorporating non-genetic risk factors into polygenic risk score models. Sci Rep 2023; 13:1351. [PMID: 36807592 PMCID: PMC9941118 DOI: 10.1038/s41598-023-27637-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 01/05/2023] [Indexed: 02/22/2023] Open
Abstract
The growing public interest in genetic risk scores for various health conditions can be harnessed to inspire preventive health action. However, current commercially available genetic risk scores can be deceiving as they do not consider other, easily attainable risk factors, such as sex, BMI, age, smoking habits, parental disease status and physical activity. Recent scientific literature shows that adding these factors can improve PGS based predictions significantly. However, implementation of existing PGS based models that also consider these factors requires reference data based on a specific genotyping chip, which is not always available. In this paper, we offer a method naïve to the genotyping chip used. We train these models using the UK Biobank data and test these externally in the Lifelines cohort. We show improved performance at identifying the 10% most at-risk individuals for type 2 diabetes (T2D) and coronary artery disease (CAD) by including common risk factors. Incidence in the highest risk group increases from 3.0- and 4.0-fold to 5.8 for T2D, when comparing the genetics-based model, common risk factor-based model and combined model, respectively. Similarly, we observe an increase from 2.4- and 3.0-fold to 4.7-fold risk for CAD. As such, we conclude that it is paramount that these additional variables are considered when reporting risk, unlike current practice with current available genetic tests.
Collapse
|
9
|
Parker F, Tang AAS, Rogers B, Carrington G, dos Remedios C, Li A, Tomlinson D, Peckham M. Affimers targeting proteins in the cardiomyocyte Z-disc: Novel tools that improve imaging of heart tissue. Front Cardiovasc Med 2023; 10:1094563. [PMID: 36865889 PMCID: PMC9971620 DOI: 10.3389/fcvm.2023.1094563] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Dilated Cardiomyopathy is a common form of heart failure. Determining how this disease affects the structure and organization of cardiomyocytes in the human heart is important in understanding how the heart becomes less effective at contraction. Here we isolated and characterised Affimers (small non-antibody binding proteins) to Z-disc proteins ACTN2 (α-actinin-2), ZASP (also known as LIM domain binding protein 3 or LDB3) and the N-terminal region of the giant protein titin (TTN Z1-Z2). These proteins are known to localise in both the sarcomere Z-discs and the transitional junctions, found close to the intercalated discs that connect adjacent cardiomyocytes. We use cryosections of left ventricles from two patients diagnosed with end-stage Dilated Cardiomyopathy who underwent Orthotopic Heart Transplantation and were whole genome sequenced. We describe how Affimers substantially improve the resolution achieved by confocal and STED microscopy compared to conventional antibodies. We quantified the expression of ACTN2, ZASP and TTN proteins in two patients with dilated cardiomyopathy and compared them with a sex- and age-matched healthy donor. The small size of the Affimer reagents, combined with a small linkage error (the distance from the epitope to the dye label covalently bound to the Affimer) revealed new structural details in Z-discs and intercalated discs in the failing samples. Affimers are thus useful for analysis of changes to cardiomyocyte structure and organisation in diseased hearts.
Collapse
Affiliation(s)
- Francine Parker
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Anna A. S. Tang
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Brendan Rogers
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Glenn Carrington
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Cris dos Remedios
- Mechanobiology Laboratory, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
| | - Amy Li
- Sydney Heart Bank, The University of Sydney, Sydney, NSW, Australia
- Department of Pharmacy & Biomedical Sciences, La Trobe University, Bendigo, VIC, Australia
- Centre for Healthy Futures, Torrens University Australia, Surrey Hills, NSW, Australia
| | - Darren Tomlinson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Michelle Peckham
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
10
|
Hu H, Geng Z, Zhang S, Xu Y, Wang Q, Chen S, Zhang B, Sun K, Lu Y. Rare copy number variation analysis identifies disease-related variants in atrioventricular septal defect patients. Front Genet 2023; 14:1075349. [PMID: 36816019 PMCID: PMC9936062 DOI: 10.3389/fgene.2023.1075349] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Atrioventricular septal defect (AVSD) is a deleterious subtype of congenital heart diseases (CHD) characterized by atrioventricular canal defect. The pathogenic genetic changes of AVSD remain elusive, particularly for copy number variation (CNV), a large segment variation of the genome, which is one of the major forms of genetic variants resulting in congenital heart diseases. In the present study, we recruited 150 AVSD cases and 100 healthy subjects as controls for whole exome sequencing (WES). We identified total 4255 rare CNVs using exon Hidden Markov model (XHMM) and screened rare CNVs by eliminating common CNVs based on controls and Database of Genomic Variants (DGV). Each patient contained at least 9 CNVs, and the CNV burden was prominently presented in chromosomes 19,22,21&16. Small CNVs (<500 kb) were frequently observed. By leveraging gene-based burden test, we further identified 20 candidate AVSD-risk genes. Among them, DYRK1A, OBSCN and TTN were presented in the core disease network of CHD and highly and dynamically expressed in the heart during the development, which indicated they possessed the high potency to be AVSD-susceptible genes. These findings not only provided a roadmap for finally unveiling the genetic cause of AVSD, but also provided more resources and proofs for clinical genetics.
Collapse
Affiliation(s)
- Huan Hu
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zilong Geng
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shasha Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuejuan Xu
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qingjie Wang
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Sun Chen
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bing Zhang
- Key Laboratory of Systems Biomedicine, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Bing Zhang, ; Kun Sun, ; Yanan Lu,
| | - Kun Sun
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Bing Zhang, ; Kun Sun, ; Yanan Lu,
| | - Yanan Lu
- Department of Pediatric Cardiology, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China,*Correspondence: Bing Zhang, ; Kun Sun, ; Yanan Lu,
| |
Collapse
|
11
|
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
|
12
|
Subramaniam J, Yamankurt G, Cunha SR. Obscurin regulates ankyrin macromolecular complex formation. J Mol Cell Cardiol 2022; 168:44-57. [PMID: 35447147 PMCID: PMC11057898 DOI: 10.1016/j.yjmcc.2022.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 03/28/2022] [Accepted: 04/12/2022] [Indexed: 10/18/2022]
Abstract
Obscurin is a large scaffolding protein in striated muscle that maintains sarcolemmal integrity and aligns the sarcoplasmic reticulum with the underlying contractile machinery. Ankyrins are a family of adaptor proteins with some isoforms that interact with obscurin. Previous studies have examined obscurin interacting with individual ankyrins. In this study, we demonstrate that two different ankyrins interact with obscurin's carboxyl terminus via independent ankyrin-binding domains (ABDs). Using in-vitro binding assays, co-precipitation assays, and FLIM-FRET analysis, we show that obscurin interacts with small ankyrin 1.5 (sAnk1.5) and the muscle-specific ankyrin-G isoform (AnkG107). While there is no direct interaction between sAnk1.5 and AnkG107, obscurin connects the two ankyrins both in vitro and in cells. Moreover, AnkG107 recruits β-spectrin to this macromolecular protein complex and mutating obscurin's ABDs disrupts complex formation. To further characterize AnkG107 interaction with obscurin, we measure obscurin-binding to different AnkG107 isoforms expressed in the heart and find that the first obscurin-binding domain in AnkG107 principally mediates this interaction. We also find that AnkG107 does not bind to filamin-C and displays minimal binding to plectin-1 compared to obscurin. Finally, both sAnk1.5-GFP and AnkG107-CTD-RFP are targeted to the M-lines of ventricular cardiomyocytes and mutating their obscurin-binding domains disrupts the M-line localization of these ankyrin constructs. Altogether, these findings support a model in which obscurin can interact via independent binding domains with two different ankyrin protein complexes to target them to the sarcomeric M-line of ventricular cardiomyocytes.
Collapse
Affiliation(s)
- Janani Subramaniam
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - Gokay Yamankurt
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America
| | - Shane R Cunha
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030, United States of America.
| |
Collapse
|
13
|
Lunardi M, Al-Habbaa A, Abdelshafy M, Davey MG, Elkoumy A, Ganly S, Elzomor H, Cawley C, Sharif F, Crowley J, Kerin M, Wijns W, Lowery A, Soliman O. Genetic and RNA-related molecular markers of trastuzumab-chemotherapy-associated cardiotoxicity in HER2 positive breast cancer: a systematic review. BMC Cancer 2022; 22:396. [PMID: 35413811 PMCID: PMC9004047 DOI: 10.1186/s12885-022-09437-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 03/16/2022] [Indexed: 11/19/2022] Open
Abstract
Cancer-therapy related cardiotoxicity (CTRCT) is a significant and frequent complication of monoclonal antibody directed therapy, especially Trastuzumab, for human epidermal growth factor receptor 2 (HER2) overexpressing breast cancers. Reliable, clinically available molecular predictive markers of CTRCT have not yet been developed. Identifying specific genetic variants and their molecular markers, which make the host susceptible to this complication is key to personalised risk stratification. A systematic review was conducted until April 2021, using the Medline, Embase databases and Google Scholar, to identify studies genetic and RNA-related markers associated with CTRCT in HER2 positive breast cancer patients. So far, researchers have mainly focused on HER2 related polymorphisms, revealing codons 655 and 1170 variants as the most likely SNPs associated with cardiotoxicity, despite some contradictory results. More recently, new potential genetic markers unrelated to the HER2 gene, and linked to known cardiomyopathy genes or to genes regulating cardiomyocytes apoptosis and metabolism, have been detected. Moreover, microRNAs are gaining increasing recognition as additional potential molecular markers in the cardio-oncology field, supported by encouraging preliminary data about their relationship with cardiotoxicity in breast cancers. In this review, we sought to synthesize evidence for genetic variants and RNA-related molecular markers associated with cardiotoxicity in HER2-positive breast cancer.
Collapse
Affiliation(s)
- Mattia Lunardi
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
- Division of Cardiology, Department of Medicine, University of Verona, Verona, Italy
- The Lambe Institute for Translational Medicine and CURAM, National University of Ireland Galway (NUIG), Galway, Ireland
| | - Ahmed Al-Habbaa
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
- Department of Cardiology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Mahmoud Abdelshafy
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - Matthew G Davey
- Discipline of Surgery, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
| | - Ahmed Elkoumy
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - Sandra Ganly
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
- The Lambe Institute for Translational Medicine and CURAM, National University of Ireland Galway (NUIG), Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), National University of Ireland, Galway, Ireland
| | - Hesham Elzomor
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - Christian Cawley
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - Faisal Sharif
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - James Crowley
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
| | - Michael Kerin
- Discipline of Surgery, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), National University of Ireland, Galway, Ireland
| | - William Wijns
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland
- The Lambe Institute for Translational Medicine and CURAM, National University of Ireland Galway (NUIG), Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), National University of Ireland, Galway, Ireland
| | - Aoife Lowery
- Discipline of Surgery, School of Medicine, Lambe Institute for Translational Research, National University of Ireland Galway, Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), National University of Ireland, Galway, Ireland
| | - Osama Soliman
- Discipline of Cardiology, Saolta Group, Galway University Hospital, Health Service Executive and CORRIB Core Lab, National University of Ireland Galway (NUIG), Galway, H91 TK33, Ireland.
- Precision Cardio-Oncology Research Enterprise (P-CORE), National University of Ireland, Galway, Ireland.
| |
Collapse
|
14
|
Grosser M, Lin H, Wu M, Zhang Y, Tipper S, Venter D, Lu J, dos Remedios CG. A bibliometric review of peripartum cardiomyopathy compared to other cardiomyopathies using artificial intelligence and machine learning. Biophys Rev 2022; 14:381-401. [PMID: 35340600 PMCID: PMC8921361 DOI: 10.1007/s12551-022-00933-x] [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: 12/21/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022] Open
Abstract
As developments in artificial intelligence and machine learning become more widespread in healthcare, their potential to transform clinical outcomes also increases. Peripartum cardiomyopathy is a rare and poorly-characterised condition that presents as heart failure in the last trimester prior to delivery or within 5-6 months postpartum. The lack of a definitive understanding of the molecular causes and clinical progress of this condition suggests that bibliometrics will be well-suited to creating new insights into this serious clinical problem. We examine similarities and differences between peripartum and its closely related familial dilated cardiomyopathy and idiopathic dilated cardiomyopathy. Using PubMed as the source of bibliometric data, we apply artificial intelligence-supported natural language processing to compare extracted data and genes association with these cardiomyopathies. Gene data were enhanced with additional metadata from third-party datasets and then analysed for their impact and specificity for peripartum cardiomyopathy. Artificial intelligence identified 14 genes that distinguished peripartum from both dilated and familial dilated cardiomyopathy. They are as follows: CTSD, RLN2, MMP23B*, SLC17A5, ST2*, PTHLH, CFH*, CFI, GPT, MR1, Rln1, SRI, STAT5A* and THBD. We then used the Human Protein Atlas website that uses affinity-purified rabbit polyclonal antibodies to identify genes that are expressed at the protein level (bold), or as RNA transcripts (*) in healthy human left ventricles. Additional analysis focussed on the full set of peripartum genes on linkage and specificity to cardiomyopathy yielded a different set of thirteen genes (bold font indicates those expressed in cardiomyocytes: PRL, RLN2, PLN, ST2, CTSD, F2, ACE, STAT3, TTN, SPP1, LGALS3, miR-146a, GNB3, SRI). This type of analysis can highlight new avenues for research, aimed at improving genomics-driven peripartum cardiomyopathy diagnosis as well as potential pathological and clinical sub-classification. We expect that this will allow for future improvements in identification, treatment and management of this condition. The first step in the application of these bibliometric-based artificial intelligence methods is to understand the current knowledge, and it is the aim of this paper to show how this might be achieved.
Collapse
Affiliation(s)
- M. Grosser
- 23 Strands Pty Ltd, 107, 26 Pirrama Rd, Pyrmont, NSW Australia
| | - H. Lin
- 23 Strands Pty Ltd, 107, 26 Pirrama Rd, Pyrmont, NSW Australia
| | - M. Wu
- University Technology Sydney, 15 Broadway, Ultimo, NSW Australia
| | - Y. Zhang
- University Technology Sydney, 15 Broadway, Ultimo, NSW Australia
| | - S. Tipper
- 23 Strands Pty Ltd, 107, 26 Pirrama Rd, Pyrmont, NSW Australia
| | - D. Venter
- 23 Strands Pty Ltd, 107, 26 Pirrama Rd, Pyrmont, NSW Australia
| | - J. Lu
- University Technology Sydney, 15 Broadway, Ultimo, NSW Australia
| | - C. G. dos Remedios
- Victor Chang Cardiac Research Institute, 405 Liverpool St, Darlinghurst, Australia
- Sydney Heart Bank, University of Sydney, Sydney, Australia
| |
Collapse
|
15
|
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
|
16
|
Peripartum cardiomyopathy: a global effort to find the cause and cure for the rare and little understood disease. Biophys Rev 2022; 14:369-379. [PMID: 35340597 PMCID: PMC8921403 DOI: 10.1007/s12551-022-00930-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/06/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
In this review, we present our current understanding of peripartum cardiomyopathy (PPCM) based on reports of the incidence, diagnosis and current treatment options. We summarise opinions on whether PPCM is triggered by vascular and/or hormonal causes and examine the influence of comorbidities such as preeclampsia. Two articles published in 2021 strongly support the hypothesis that PPCM may be a familial disease. Using large cohorts of PPCM patients, they summarised the available genomic DNA sequence data that are expressed in human cardiomyocytes. While PPCM is considered a disease predominately affecting the left ventricle, there are data to suggest that some cases also involve right ventricular failure. Finally, we conclude that there is sufficient evidence to warrant an RNAseq investigation and that this would be most informative if performed at the cardiomyocytes level rather than analysing genomic DNA from the peripheral circulation. Given the rarity of PPCM, the combined resources of international human heart tissue biobanks have assembled 30 ventricular tissue samples from PPCM patients, and we are actively seeking to enlarge this patient base by collaborating with human heart tissue banks and research laboratories who would like to join this endeavour.
Collapse
|
17
|
Pierantozzi E, Szentesi P, Paolini C, Dienes B, Fodor J, Oláh T, Colombini B, Rassier DE, Rubino EM, Lange S, Rossi D, Csernoch L, Bagni MA, Reggiani C, Sorrentino V. Impaired Intracellular Ca 2+ Dynamics, M-Band and Sarcomere Fragility in Skeletal Muscles of Obscurin KO Mice. Int J Mol Sci 2022; 23:1319. [PMID: 35163243 PMCID: PMC8835721 DOI: 10.3390/ijms23031319] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Obscurin is a giant sarcomeric protein expressed in striated muscles known to establish several interactions with other proteins of the sarcomere, but also with proteins of the sarcoplasmic reticulum and costameres. Here, we report experiments aiming to better understand the contribution of obscurin to skeletal muscle fibers, starting with a detailed characterization of the diaphragm muscle function, which we previously reported to be the most affected muscle in obscurin (Obscn) KO mice. Twitch and tetanus tension were not significantly different in the diaphragm of WT and Obscn KO mice, while the time to peak (TTP) and half relaxation time (HRT) were prolonged. Differences in force-frequency and force-velocity relationships and an enhanced fatigability are observed in an Obscn KO diaphragm with respect to WT controls. Voltage clamp experiments show that a sarcoplasmic reticulum's Ca2+ release and SERCA reuptake rates were decreased in muscle fibers from Obscn KO mice, suggesting that an impairment in intracellular Ca2+ dynamics could explain the observed differences in the TTP and HRT in the diaphragm. In partial contrast with previous observations, Obscn KO mice show a normal exercise tolerance, but fiber damage, the altered sarcomere ultrastructure and M-band disarray are still observed after intense exercise.
Collapse
Affiliation(s)
- Enrico Pierantozzi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Péter Szentesi
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Cecilia Paolini
- Department of Neuroscience, Imaging and Clinical Sciences, University Gabriele d’ Annunzio of Chieti, 66100 Chieti, Italy;
| | - Beatrix Dienes
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - János Fodor
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Tamás Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Barbara Colombini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Dilson E. Rassier
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC H2W 1S4, Canada;
| | - Egidio Maria Rubino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - Stephan Lange
- Biomedical Research Facility 2, School of Medicine, University of California, La Jolla, CA 92093, USA;
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, 413 45 Gothenburg, Sweden
| | - Daniela Rossi
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| | - László Csernoch
- Department of Physiology, Faculty of Medicine, University of Debrecen, H-4002 Debrecen, Hungary; (P.S.); (B.D.); (J.F.); (T.O.); (L.C.)
| | - Maria Angela Bagni
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (B.C.); (M.A.B.)
| | - Carlo Reggiani
- Department of Biomedical Science, University of Padova, 35121 Padova, Italy;
- Science and Research Center Koper, Institute for Kinesiology Research, 6000 Koper, Slovenia
| | - Vincenzo Sorrentino
- Department of Molecular and Developmental Medicine, Molecular Medicine Section, University of Siena, 53100 Siena, Italy; (E.P.); (E.M.R.); (D.R.)
| |
Collapse
|
18
|
Ren J, Jiang L, Liu X, Liao Y, Zhao X, Tang F, Yu H, Shao Y, Wang J, Wen L, Song L. Heart-specific DNA methylation analysis in plasma for the investigation of myocardial damage. J Transl Med 2022; 20:36. [PMID: 35062960 PMCID: PMC8780310 DOI: 10.1186/s12967-022-03234-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/05/2022] [Indexed: 11/10/2022] Open
Abstract
Background Circulating cell-free DNA (cfDNA) can be released when myocardial damage occurs. Methods Here, we used the methylated CpG tandem amplification and sequencing (MCTA-seq) method for analyzing dynamic changes in heart-derived DNA in plasma samples from myocardial infarction (MI) patients. Results We identified six CGCGCGG loci showing heart-specific hypermethylation patterns. MCTA-seq deconvolution analysis combining these loci detected heart-released cfDNA in MI patients at hospital admission, and showed that the prominently elevated total cfDNA level after percutaneous coronary intervention (PCI) was derived from both the heart and white blood cells. Furthermore, for the top marker CORO6, we developed a digital droplet PCR (ddPCR) assay that clearly detected heart damage signals in cfDNA of MI patients at hospital admission. Conclusions Our study provides insights into MI pathologies and developed a new ddPCR assay for detecting myocardial damage in clinical applications. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03234-9.
Collapse
Affiliation(s)
- Jie Ren
- Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.,Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lin Jiang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China.,National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China
| | - Xiaomeng Liu
- Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.,Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuhan Liao
- Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.,Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, 100871, China
| | - Xueyan Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China.,National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.,Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Huimin Yu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yibing Shao
- Department of Cardiology, Qingdao Municipal Hospital, Qingdao, Shandong, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China. .,National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China.
| | - Lu Wen
- Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China. .,Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, 100871, China.
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China. .,National Clinical Research Center for Cardiovascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100871, China.
| |
Collapse
|
19
|
Cabrera-Serrano M, Caccavelli L, Savarese M, Vihola A, Jokela M, Johari M, Capiod T, Madrange M, Bugiardini E, Brady S, Quinlivan R, Merve A, Scalco R, Hilton-Jones D, Houlden H, Ibrahim Aydin H, Ceylaner S, Vockley J, Taylor RL, Folland C, Kelly A, Goullee H, Ylikallio E, Auranen M, Tyynismaa H, Udd B, Forrest ARR, Davis MR, Bratkovic D, Manton N, Robertson T, McCombe P, Laing NG, Phillips L, de Lonlay P, Ravenscroft G. Bi-allelic loss-of-function OBSCN variants predispose individuals to severe recurrent rhabdomyolysis. Brain 2021; 145:3985-3998. [DOI: 10.1093/brain/awab484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 12/04/2021] [Accepted: 12/10/2021] [Indexed: 11/14/2022] Open
Abstract
Abstract
Rhabdomyolysis is the acute breakdown of skeletal myofibres in response to an initiating factor, most commonly toxins and over exertion. A variety of genetic disorders predispose to rhabdomyolysis through different pathogenic mechanisms, particularly in patients with recurrent episodes. However, most cases remain without a genetic diagnosis. Here we present six patients who presented with severe and recurrent rhabdomyolysis, usually with onset in the teenage years; other features included a history of myalgia and muscle cramps. We identified ten bi-allelic loss-of-function variants in the gene encoding obscurin (OBSCN) predisposing individuals to recurrent rhabdomyolysis. We show reduced expression of OBSCN and loss of obscurin protein in patient muscle. Obscurin is proposed to be involved in SR function and Ca2+ handling. Patient cultured myoblasts appear more susceptible to starvation as evidenced by a greater decreased in SR Ca2+ content compared to control myoblasts. This likely reflects a lower efficiency when pumping Ca2+ back into the SR and/or a decrease in Ca2+ SR storage ability when metabolism is diminished. OSBCN variants have previously been associated with cardiomyopathies. None of the patients presented with a cardiomyopathy and cardiac examinations were normal in all cases in which cardiac function was assessed. There was also no history of cardiomyopathy in first degree relatives, in particular in any of the carrier parents. This cohort is relatively young, thus follow-up studies and the identification of additional cases with bi-allelic null OBSCN variants will further delineate OBSCN-related disease and the clinical course of disease.
Collapse
Affiliation(s)
- Macarena Cabrera-Serrano
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
- Unidad de Enfermedades Neuromusculares. Servicio de Neurologia y Neurofisiologia. Hospital Virgen del Rocio, Sevilla, Spain
| | - Laure Caccavelli
- Inserm U1151, Institut Necker Enfants-Malades, Reference Center of Inherited Metabolic Diseases and MetabERN, Necker-Enfants-Malades Hospital, Paris University, Paris, France
| | - Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland and Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Anna Vihola
- Folkhälsan Research Center, Helsinki, Finland and Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
- Tampere Neuromuscular Center, Tampere University Hospital, Tampere, Finland
| | - Manu Jokela
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland
- Neurocenter, Department of Neurology, Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland
| | - Mridul Johari
- Folkhälsan Research Center, Helsinki, Finland and Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Thierry Capiod
- Inserm U1151, Institut Necker Enfants-Malades, Reference Center of Inherited Metabolic Diseases and MetabERN, Necker-Enfants-Malades Hospital, Paris University, Paris, France
| | - Marine Madrange
- Inserm U1151, Institut Necker Enfants-Malades, Reference Center of Inherited Metabolic Diseases and MetabERN, Necker-Enfants-Malades Hospital, Paris University, Paris, France
| | - Enrico Bugiardini
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | - Stefen Brady
- Department of Neurology, Southmead Hospital, Bristol, UK
| | - Rosaline Quinlivan
- MRC Centre for Neuromuscular Diseases, University College Hospitals, London, UK
| | - Ashirwad Merve
- MRC Centre for Neuromuscular Diseases, University College Hospitals, London, UK
| | - Renata Scalco
- MRC Centre for Neuromuscular Diseases, University College Hospitals, London, UK
| | - David Hilton-Jones
- Neurosciences Group, Nuffield Department of Clinical Neurosciences, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Henry Houlden
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, London, UK
| | | | - Serdar Ceylaner
- Intergen Genetic Diagnosis and Research Center, Ankara, Turkey
| | - Jerry Vockley
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Rhonda L. Taylor
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Chiara Folland
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Aasta Kelly
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
| | - Hayley Goullee
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Emil Ylikallio
- Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Mari Auranen
- Clinical Neurosciences, Neurology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Henna Tyynismaa
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland and Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
- Tampere Neuromuscular Center, Tampere University Hospital, Tampere, Finland
| | - Alistair R. R. Forrest
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| | - Mark R. Davis
- Department of Diagnostic Genomics, PathWest Laboratory Medicine WA, Nedlands, WA, Australia
| | - Drago Bratkovic
- Metabolic Clinic, Women and Children’s Hospital, North Adelaide, SA, Australia
| | - Nicholas Manton
- SA Pathology, Women and Children’s Hospital, North Adelaide, SA, Australia
| | - Thomas Robertson
- Anatomical Pathology, Queensland Pathology, Brisbane, Queensland, Australia
| | - Pamela McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Centre for Clinical Research, The University of Queensland Centre for Clinical Research, Brisbane, Queensland, Australia
| | - Nigel G. Laing
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
- Department of Diagnostic Genomics, PathWest Laboratory Medicine WA, Nedlands, WA, Australia
| | - Liza Phillips
- SA Pathology, Women and Children’s Hospital, North Adelaide, SA, Australia
- The University of Adelaide, Adelaide, SA, Australia
| | - Pascale de Lonlay
- Inserm U1151, Institut Necker Enfants-Malades, Reference Center of Inherited Metabolic Diseases and MetabERN, Necker-Enfants-Malades Hospital, Paris University, Paris, France
| | - Gianina Ravenscroft
- Harry Perkins Institute of Medical Research, Nedlands, WA, Australia
- Centre of Medical Research, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
20
|
Louadi Z, Elkjaer ML, Klug M, Lio CT, Fenn A, Illes Z, Bongiovanni D, Baumbach J, Kacprowski T, List M, Tsoy O. Functional enrichment of alternative splicing events with NEASE reveals insights into tissue identity and diseases. Genome Biol 2021; 22:327. [PMID: 34857024 PMCID: PMC8638120 DOI: 10.1186/s13059-021-02538-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/10/2021] [Indexed: 01/27/2023] Open
Abstract
Alternative splicing (AS) is an important aspect of gene regulation. Nevertheless, its role in molecular processes and pathobiology is far from understood. A roadblock is that tools for the functional analysis of AS-set events are lacking. To mitigate this, we developed NEASE, a tool integrating pathways with structural annotations of protein-protein interactions to functionally characterize AS events. We show in four application cases how NEASE can identify pathways contributing to tissue identity and cell type development, and how it highlights splicing-related biomarkers. With a unique view on AS, NEASE generates unique and meaningful biological insights complementary to classical pathways analysis.
Collapse
Affiliation(s)
- Zakaria Louadi
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Notkestrasse 9, 22607, Hamburg, Germany
| | - Maria L Elkjaer
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Melissa Klug
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Department of Internal Medicine I, School of Medicine, University hospital rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Chit Tong Lio
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Notkestrasse 9, 22607, Hamburg, Germany
| | - Amit Fenn
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Notkestrasse 9, 22607, Hamburg, Germany
| | - Zsolt Illes
- Department of Neurology, Odense University Hospital, Odense, Denmark
- Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
- Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Dario Bongiovanni
- Department of Internal Medicine I, School of Medicine, University hospital rechts der Isar, Technical University of Munich, Munich, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Department of Cardiovascular Medicine, Humanitas Clinical and Research Center IRCCS and Humanitas University, Rozzano, Milan, Italy
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Notkestrasse 9, 22607, Hamburg, Germany
- Institute of Mathematics and Computer Science, University of Southern Denmark, Campusvej 55, 5000, Odense, Denmark
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, 85354, Freising, Germany.
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, Notkestrasse 9, 22607, Hamburg, Germany.
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Fukuzawa A, Koch D, Grover S, Rees M, Gautel M. When is an obscurin variant pathogenic? The impact of Arg4344Gln and Arg4444Trp variants on protein-protein interactions and protein stability. Hum Mol Genet 2021; 30:1131-1141. [PMID: 33438037 PMCID: PMC8188405 DOI: 10.1093/hmg/ddab010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/17/2020] [Accepted: 01/04/2021] [Indexed: 11/13/2022] Open
Abstract
Obscurin is a giant muscle protein that connects the sarcomere with the sarcoplasmic reticulum, and has poorly understood structural and signalling functions. Increasingly, obscurin variants are implicated in the pathophysiology of cardiovascular diseases. The Arg4344Gln variant (R4344Q) in obscurin domain Ig58, initially discovered in a patient with hypertrophic cardiomyopathy, has been reported to reduce binding to titin domains Z8-Z9, impairing obscurin’s Z-disc localization. An R4344Q knock-in mouse developed a cardiomyopathy-like phenotype with abnormal Ca2+-handling and arrhythmias, which were attributed to an enhanced affinity of a putative interaction between obscurin Ig58 and phospholamban (PLN) due to the R4344Q variant. However, the R4344Q variant is found in 15% of African Americans, arguing against its pathogenicity. To resolve this apparent paradox, we quantified the influence of the R4344Q variant (alongside another potentially pathogenic variant: Arg4444Trp (R4444W)) on binding to titin Z8-Z9, novex-3 and PLN using pull-down assays and microscale thermophoresis and characterized the influence on domain stability using differential scanning fluorimetry. We found no changes in titin binding and thermostability for both variants and modestly increased affinities of PLN for R4344Q and R4444W. While we could not confirm the novex-3/obscurin interaction, the PLN/obscurin interaction relies on the transmembrane region of PLN and is not reproducible in mammalian cells, suggesting it is an in vitro artefact. Without clear clinical evidence for disease involvement, we advise against classifying these obscurin variants as pathogenic.
Collapse
Affiliation(s)
- Atsushi Fukuzawa
- Randall Centre for Cell & Molecular Biophysics, King's College London, 18-20 Newcomen Street, SE1 1UL, UK
| | - Daniel Koch
- Randall Centre for Cell & Molecular Biophysics, King's College London, 18-20 Newcomen Street, SE1 1UL, UK
| | - Sarah Grover
- Randall Centre for Cell & Molecular Biophysics, King's College London, 18-20 Newcomen Street, SE1 1UL, UK
| | - Martin Rees
- Randall Centre for Cell & Molecular Biophysics, King's College London, 18-20 Newcomen Street, SE1 1UL, UK
| | - Mathias Gautel
- Randall Centre for Cell & Molecular Biophysics, King's College London, 18-20 Newcomen Street, SE1 1UL, UK
| |
Collapse
|
24
|
Auxerre-Plantié E, Nielsen T, Grunert M, Olejniczak O, Perrot A, Özcelik C, Harries D, Matinmehr F, Dos Remedios C, Mühlfeld C, Kraft T, Bodmer R, Vogler G, Sperling SR. Identification of MYOM2 as a candidate gene in hypertrophic cardiomyopathy and Tetralogy of Fallot, and its functional evaluation in the Drosophila heart. Dis Model Mech 2020; 13:dmm045377. [PMID: 33033063 PMCID: PMC7758640 DOI: 10.1242/dmm.045377] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 10/01/2020] [Indexed: 01/11/2023] Open
Abstract
The causal genetic underpinnings of congenital heart diseases, which are often complex and multigenic, are still far from understood. Moreover, there are also predominantly monogenic heart defects, such as cardiomyopathies, with known disease genes for the majority of cases. In this study, we identified mutations in myomesin 2 (MYOM2) in patients with Tetralogy of Fallot (TOF), the most common cyanotic heart malformation, as well as in patients with hypertrophic cardiomyopathy (HCM), who do not exhibit any mutations in the known disease genes. MYOM2 is a major component of the myofibrillar M-band of the sarcomere, and a hub gene within interactions of sarcomere genes. We show that patient-derived cardiomyocytes exhibit myofibrillar disarray and reduced passive force with increasing sarcomere lengths. Moreover, our comprehensive functional analyses in the Drosophila animal model reveal that the so far uncharacterized fly gene CG14964 [herein referred to as Drosophila myomesin and myosin binding protein (dMnM)] may be an ortholog of MYOM2, as well as other myosin binding proteins. Its partial loss of function or moderate cardiac knockdown results in cardiac dilation, whereas more severely reduced function causes a constricted phenotype and an increase in sarcomere myosin protein. Moreover, compound heterozygous combinations of CG14964 and the sarcomere gene Mhc (MYH6/7) exhibited synergistic genetic interactions. In summary, our results suggest that MYOM2 not only plays a critical role in maintaining robust heart function but may also be a candidate gene for heart diseases such as HCM and TOF, as it is clearly involved in the development of the heart.This article has an associated First Person interview with Emilie Auxerre-Plantié and Tanja Nielsen, joint first authors of the paper.
Collapse
Affiliation(s)
- Emilie Auxerre-Plantié
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Tanja Nielsen
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Marcel Grunert
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Olga Olejniczak
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| | - Andreas Perrot
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Cemil Özcelik
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
| | - Dennis Harries
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Faramarz Matinmehr
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Cristobal Dos Remedios
- Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Camperdown, Sydney, New South Wales 2006, Australia
| | - Christian Mühlfeld
- Institute of Functional and Applied Anatomy, Hannover Medical School, 30625 Hannover, Germany
| | - Theresia Kraft
- Medical School of Hannover, Institute of Molecular and Cell Physiology, 30625 Hannover, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Georg Vogler
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Silke R Sperling
- Cardiovascular Genetics, Charité - Universitätsmedizin Berlin, 13125 Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
| |
Collapse
|
25
|
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
|
26
|
Vikhorev PG, Vikhoreva NN, Yeung W, Li A, Lal S, dos Remedios CG, Blair CA, Guglin M, Campbell KS, Yacoub MH, de Tombe P, Marston SB. Titin-truncating mutations associated with dilated cardiomyopathy alter length-dependent activation and its modulation via phosphorylation. Cardiovasc Res 2020; 118:241-253. [PMID: 33135063 PMCID: PMC8752363 DOI: 10.1093/cvr/cvaa316] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 10/20/2020] [Indexed: 12/18/2022] Open
Abstract
Abstract
Aims
Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants.
Methods and results
We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants.
Conclusion
Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Natalia N Vikhoreva
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
| | - WaiChun Yeung
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Amy Li
- Department of Pharmacy and Biomedical Sciences, La Trobe University, Bendigo, VIC 3550, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, NSW 2006, Australia
| | - Cristobal G dos Remedios
- Division of Molecular Cardiology and Biophysics, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Cheavar A Blair
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Maya Guglin
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Magdi H Yacoub
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Pieter de Tombe
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
- Heart Science Centre, Magdi Yacoub Institute, Harefield Hospital, London UB9 6JH, UK
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
27
|
Liu B, Zhu L, Yuan P, Marsboom G, Hong Z, Liu J, Zhang P, Hu Q. Comprehensive identification of signaling pathways for idiopathic pulmonary arterial hypertension. Am J Physiol Cell Physiol 2020; 318:C913-C930. [PMID: 32159364 DOI: 10.1152/ajpcell.00382.2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Whole exome sequencing (WES) was used in the research of familial pulmonary arterial hypertension (FPAH). CAV1 and KCNK3 were found as two novel candidate genes of FPAH. However, few pathogenic genes were identified in idiopathic pulmonary arterial hypertension (IPAH). We conducted WES in 20 unrelated IPAH patients who did not carry the known PAH-pathogenic variants among BMPR2, CAV1, KCNK3, SMAD9, ALK1, and ENG. We found a total of 4,950 variants in 3,534 genes, including 4,444 single-nucleotide polymorphisms and 506 insertions/deletions (InDels). Through the comprehensive and multilevel analysis, we disclosed several novel signaling cascades significantly connected to IPAH, including variants related to cadherin signaling pathway, dilated cardiomyopathy, glucose metabolism, immune response, mucin-type O-glycosylation, phospholipase C (PLC)-activating G protein-coupled receptor (GPCR) signaling pathway, vascular contraction and generation, and voltage-dependent Ca2+ channels. We also conducted validation studies in five mutant genes related to PLC-activating GPCR signaling pathway potentially involved in intracellular calcium regulation through Sanger sequencing for mutation accuracy, qRT-PCR for mRNA stability, immunofluorescence for subcellular localization, Western blotting for protein level, Fura-2 imaging for intracellular calcium, and proliferation analysis for cell function. The validation experiments showed that those variants in CCR5 and C3AR1 significantly increased the rise of intracellular calcium and the variant in CCR5 profoundly enhanced proliferative capacity of human pulmonary artery smooth muscle cells. Thus, our study suggests that multiple genetically affected signaling pathways take effect together to cause the formation of IPAH and the development of right heart failure and may further provide new therapy targets or putative clues for the present treatments such as limited therapeutic effectiveness of Ca2+ channel blockers.
Collapse
Affiliation(s)
- Bingxun Liu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zhu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Yuan
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Glenn Marsboom
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Zhigang Hong
- Department of Pharmacology, University of Illinois College of Medicine, Chicago, Illinois
| | - Jinming Liu
- Department of Cardiopulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinghua Hu
- Department of Pathophysiology, School of Basic Medicine, and Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
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
|
29
|
Jaouadi H, Bouyacoub Y, Chabrak S, Kraoua L, Zaroui A, Elouej S, Nagara M, Dallali H, Delague V, Levy N, Benkhalifa R, Mechmeche R, Zaffran S, Abdelhak S. Multiallelic rare variants support an oligogenic origin of sudden cardiac death in the young. Herz 2020; 46:94-102. [PMID: 31970460 DOI: 10.1007/s00059-019-04883-1] [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: 08/15/2019] [Revised: 09/20/2019] [Accepted: 12/06/2019] [Indexed: 12/16/2022]
Abstract
Unexplained sudden death in the young is cardiovascular in most cases. Structural and conduction defects in cardiac-related genes can conspire to underlie sudden cardiac death. Here we report a clinical investigation and an extensive genetic assessment of a Tunisian family with sudden cardiac death in young members. In order to identify the family-genetic basis of sudden cardiac death, we performed Whole Exome Sequencing (WES), read depth copy-number-variation (CNV) screening and segregation analysis. We identify 6 ultra-rare pathogenic heterozygous variants in OBSCN, RYR2, DSC2, AKAP9, CACNA1C and RBM20 genes, and one homozygous splicing variant in TECRL gene consistent with an oligogenic model of inheritance. CNV analysis did not reveal any causative CNV consistent with the family phenotype. Overall, our results are highly suggestive for a cumulative effect of heterozygous missense variants as disease causation and to account for a greater disease severity among offspring. Our study further confirms the complexity of the inheritance of sudden cardiac death and highlights the utility of family-based WES and segregation analysis in the identification of family specific mutations within different cardiac genes pathways.
Collapse
Affiliation(s)
- Hager Jaouadi
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, 13 Place Pasteur, BP74-1002, Tunis, belvédère, Tunisia.
| | - Yosra Bouyacoub
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, 13 Place Pasteur, BP74-1002, Tunis, belvédère, Tunisia
| | - Sonia Chabrak
- Department of Cardiology, La Rabta Hospital, Tunis, Tunisia.,Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Lilia Kraoua
- Department of Congenital and Hereditary Diseases, Charles Nicolle Hospital, Tunis, Tunisia
| | - Amira Zaroui
- Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Sahar Elouej
- Aix Marseille University, INSERM, U1251, Marseille Medical Genetics, Marseille, France
| | - Majdi Nagara
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, 13 Place Pasteur, BP74-1002, Tunis, belvédère, Tunisia
| | - Hamza Dallali
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, 13 Place Pasteur, BP74-1002, Tunis, belvédère, Tunisia
| | - Valérie Delague
- Aix Marseille University, INSERM, U1251, Marseille Medical Genetics, Marseille, France
| | - Nicolas Levy
- Aix Marseille University, INSERM, U1251, Marseille Medical Genetics, Marseille, France
| | - Rym Benkhalifa
- Venoms and Therapeutic Biomolecules Laboratory LR16IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Rachid Mechmeche
- Department of Cardiology, La Rabta Hospital, Tunis, Tunisia.,Faculty of Medicine of Tunis, Université Tunis El Manar, Tunis, Tunisia
| | - Stéphane Zaffran
- Aix Marseille University, INSERM, U1251, Marseille Medical Genetics, Marseille, France
| | - Sonia Abdelhak
- Biomedical Genomics and Oncogenetics Laboratory LR16IPT05, Institut Pasteur de Tunis, Université Tunis El Manar, 13 Place Pasteur, BP74-1002, Tunis, belvédère, Tunisia
| |
Collapse
|
30
|
Yu Y, Choi K, Wu J, Andreassen PR, Dexheimer PJ, Keddache M, Brems H, Spinner RJ, Cancelas JA, Martin LJ, Wallace MR, Legius E, Vogel KS, Ratner N. NF1 patient missense variants predict a role for ATM in modifying neurofibroma initiation. Acta Neuropathol 2020; 139:157-174. [PMID: 31664505 PMCID: PMC7243727 DOI: 10.1007/s00401-019-02086-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 10/11/2019] [Accepted: 10/15/2019] [Indexed: 01/01/2023]
Abstract
In Neurofibromatosis type 1, NF1 gene mutations in Schwann cells (SC) drive benign plexiform neurofibroma (PNF), and no additional SC changes explain patient-to-patient variability in tumor number. Evidence from twin studies suggests that variable expressivity might be caused by unidentified modifier genes. Whole exome sequencing of SC and fibroblast DNA from the same resected PNFs confirmed biallelic SC NF1 mutations; non-NF1 somatic SC variants were variable and present at low read number. We identified frequent germline variants as possible neurofibroma modifier genes. Genes harboring variants were validated in two additional cohorts of NF1 patients and by variant burden test. Genes including CUBN, CELSR2, COL14A1, ATR and ATM also showed decreased gene expression in some neurofibromas. ATM-relevant DNA repair defects were also present in a subset of neurofibromas with ATM variants, and in some neurofibroma SC. Heterozygous ATM G2023R or homozygous S707P variants reduced ATM protein expression in heterologous cells. In mice, genetic Atm heterozygosity promoted Schwann cell precursor self-renewal and increased tumor formation in vivo, suggesting that ATM variants contribute to neurofibroma initiation. We identify germline variants, rare in the general population, overrepresented in NF1 patients with neurofibromas. ATM and other identified genes are candidate modifiers of PNF pathogenesis.
Collapse
Affiliation(s)
- Yanan Yu
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA
- Graduate Program in Cancer and Cell Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Kwangmin Choi
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA
| | - Jianqiang Wu
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA
| | - Paul R Andreassen
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA
| | - Phillip J Dexheimer
- Division of Biomedical Informatics, Cincinnati Children's Hospital, Cincinnati, OH, USA
| | - Mehdi Keddache
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and the Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Robert J Spinner
- Department of Neurological Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jose A Cancelas
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, OH, USA
| | - Lisa J Martin
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and the Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology, UF Genetics Institute, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| | - Eric Legius
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Kristine S Vogel
- Department of Cell Systems and Anatomy, UT Health San Antonio, San Antonio, TX, USA
| | - Nancy Ratner
- Department of Experimental Hematology and Cancer Biology, Cincinati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinanti School of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
31
|
Liu L, Shi X, Yue P, Zheng X, Hua Y, Zhou K, Li Y. Is genomic screening necessary for fetuses who suffer moderate to severe tricuspid regurgitation?: A case report. Medicine (Baltimore) 2019; 98:e17771. [PMID: 31770195 PMCID: PMC6890352 DOI: 10.1097/md.0000000000017771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Tricuspid regurgitation (TR) is a frequent finding during echocardiography screening in fetal or neonatal life, which reveals a weak association between TR and cardiac malformation. Except for structural abnormalities, dilated cardiomyopathy (DCM) ranks as the top reason for early child morbidity and mortality among all kinds of cardiomyopathy. In the early fetal stage, cardiac abnormalities detected by early fetal genetic testing followed by abnormalities on ultrasound would provide more valuable information for parents and physicians to make a better therapeutic schedule. PATIENT CONCERNS A case of severe TR was found via the fetal ultrasound screening. After birth, this child suffered severe heart dysfunction, and echocardiography confirmed a DCM phenotype within a very short time. DIAGNOSIS AND INTERVENTION A 40-year-old female received routine fetal echocardiographic screening, which demonstrated that the fetus presented severe TR. Six months after birth, the baby experienced severe heart failure, as the EF dropped to 22% with an extremely large LV chamber. The genomic sequence had been determined, and 3 pathogenic gene mutations located in 2 genes, cardiac troponin T (TNNT2) c.548G>A, desmoplakin (DSP) c.3146C>T, and DSP c.5213G>A, were identified. Finally, the patient was diagnosed with DCM. This child received digoxin, hydrochlorothiazide, spironolactone diuresis, captopril, and L-carnitine, and the symptoms of heart failure had been controlled as the patient waited for heart transplantation. OUTCOMES During the follow-up, the patient still suffered from poor heart function and an enlarged left ventricle. Concomitantly, the parents placed her on a waiting list for heart transplantation. LESSONS Fetal TR is a common phenomenon, and many studies have indicated that isolated TR is not an appropriate predictor of chromosomal abnormalities or congenital heart defects. However, according to this case, it is urgent to recommend that the mother should take advantage of free fetal DNA analysis in a maternal blood sample to obtain further molecular evidence once fetal echocardiography reveals moderate to severe TR with any maternal high-risk factors for birth defects.
Collapse
Affiliation(s)
- Lei Liu
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoqing Shi
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peng Yue
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaolan Zheng
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yimin Hua
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaiyu Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yifei Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University
- Ministry of Education Key Laboratory of Women and Children's Diseases and Birth Defects, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
32
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
33
|
ALFARIH MASHAEL, SYRRIS PETROS, ARBUSTINI ELOISA, AUGUSTO JOÃOB, HUGHES ALUN, LLOYD GUY, LOPES LUISR, MOON JAMESC, MOHIDDIN SAIDI, CAPTUR GABRIELLA. Familial cardiomyopathy caused by a novel heterozygous mutation in the gene LMNA (c.1434dupG): a cardiac MRI-augmented segregation study. ACTA MYOLOGICA : MYOPATHIES AND CARDIOMYOPATHIES : OFFICIAL JOURNAL OF THE MEDITERRANEAN SOCIETY OF MYOLOGY 2019; 38:159-162. [PMID: 31788659 PMCID: PMC6859410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In a five-generation family carrying a novel frameshift LMNA variant (c.1434dupG, p.Leu479AlafsX72), imaging-augmented segregation analysis supports its association with lamin heart disease. Affected members exhibit conduction abnormalities, supraventricular and ventricular arrythmias, dilated cardiomyopathy with non-infarct pattern midwall septal fibrosis, heart failure and thromboembolic complications.
Collapse
Affiliation(s)
- MASHAEL ALFARIH
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK,Department of Cardiac Technology, College of Applied Medial Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - PETROS SYRRIS
- Institute of Cardiovascular Science, University College London, Gower Street, London, UK
| | - ELOISA ARBUSTINI
- Center for Inherited Cardiovascular Diseases, Foundation IRCCS Policlinico San Matteo, University of Pavia, Italy
| | - JOÃO B. AUGUSTO
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK
| | - ALUN HUGHES
- Institute of Cardiovascular Science, University College London, Gower Street, London, UK,UCL MRC Unit for Lifelong Health and Ageing, 33 Bedford Place, London, UK
| | - GUY LLOYD
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK
| | - LUIS R. LOPES
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK
| | - JAMES C. MOON
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK
| | - SAIDI MOHIDDIN
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK
| | - GABRIELLA CAPTUR
- Barts Heart Center, The Cardiovascular Magnetic Resonance Imaging Unit, St Bartholomew’s Hospital, West Smithfield, London, UK,Institute of Cardiovascular Science, University College London, Gower Street, London, UK,UCL MRC Unit for Lifelong Health and Ageing, 33 Bedford Place, London, UK, Cardiology Department, Royal Free Hospital NHS Trust, Pond St, Hampstead, London, UK,Address for correspondence: Gabriella Captur, Institute of Cardiovascular Science, University College London, Gower Street, London WC1E 6BT, UK. Tel. + 44 7809621264. E-mail:
| |
Collapse
|
34
|
Ahrens-Nicklas RC, Pappas CT, Farman GP, Mayfield RM, Larrinaga TM, Medne L, Ritter A, Krantz ID, Murali C, Lin KY, Berger JH, Yum SW, Carreon CK, Gregorio CC. Disruption of cardiac thin filament assembly arising from a mutation in LMOD2: A novel mechanism of neonatal dilated cardiomyopathy. SCIENCE ADVANCES 2019; 5:eaax2066. [PMID: 31517052 PMCID: PMC6726455 DOI: 10.1126/sciadv.aax2066] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 07/31/2019] [Indexed: 05/10/2023]
Abstract
Neonatal heart failure is a rare, poorly-understood presentation of familial dilated cardiomyopathy (DCM). Exome sequencing in a neonate with severe DCM revealed a homozygous nonsense variant in leiomodin 2 (LMOD2, p.Trp398*). Leiomodins (Lmods) are actin-binding proteins that regulate actin filament assembly. While disease-causing mutations in smooth (LMOD1) and skeletal (LMOD3) muscle isoforms have been described, the cardiac (LMOD2) isoform has not been previously associated with human disease. Like our patient, Lmod2-null mice have severe early-onset DCM and die before weaning. The infant's explanted heart showed extraordinarily short thin filaments with isolated cardiomyocytes displaying a large reduction in maximum calcium-activated force production. The lack of extracardiac symptoms in Lmod2-null mice, and remarkable morphological and functional similarities between the patient and mouse model informed the decision to pursue cardiac transplantation in the patient. To our knowledge, this is the first report of aberrant cardiac thin filament assembly associated with human cardiomyopathy.
Collapse
Affiliation(s)
- Rebecca C. Ahrens-Nicklas
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Christopher T. Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Gerrie P. Farman
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Rachel M. Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Tania M. Larrinaga
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Livija Medne
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alyssa Ritter
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ian D. Krantz
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Chaya Murali
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly Y. Lin
- Division of Pediatric Cardiology, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Justin H. Berger
- Division of Pediatric Cardiology, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Sabrina W. Yum
- Division of Pediatric Neurology, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Chrystalle Katte Carreon
- Department of Pathology, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
35
|
Kagemoto T, Oyama K, Yamane M, Tsukamoto S, Kobirumaki-Shimozawa F, Li A, Dos Remedios C, Fukuda N, Ishiwata S. Sarcomeric Auto-Oscillations in Single Myofibrils From the Heart of Patients With Dilated Cardiomyopathy. Circ Heart Fail 2019; 11:e004333. [PMID: 29980594 DOI: 10.1161/circheartfailure.117.004333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 05/31/2018] [Indexed: 01/30/2023]
Abstract
BACKGROUND Left ventricular wall motion is depressed in patients with dilated cardiomyopathy (DCM). However, whether or not the depressed left ventricular wall motion is caused by impairment of sarcomere dynamics remains to be fully clarified. METHODS AND RESULTS We analyzed the mechanical properties of single sarcomere dynamics during sarcomeric auto-oscillations (calcium spontaneous oscillatory contractions [Ca-SPOC]) that occurred at partial activation under the isometric condition in myofibrils from donor hearts and from patients with severe DCM (New York Heart Association classification III-IV). Ca-SPOC reproducibly occurred in the presence of 1 μmol/L free Ca2+ in both nonfailing and DCM myofibrils, and sarcomeres exhibited a saw-tooth waveform along single myofibrils composed of quick lengthening and slow shortening. The period of Ca-SPOC was longer in DCM myofibrils than in nonfailing myofibrils, in association with prolonged shortening time. Lengthening time was similar in both groups. Then, we performed Tn (troponin) exchange in myofibrils with a DCM-causing homozygous mutation (K36Q) in cTnI (cardiac TnI). On exchange with the Tn complex from healthy porcine ventricles, period, shortening time, and shortening velocity in cTnI-K36Q myofibrils became similar to those in Tn-reconstituted nonfailing myofibrils. Protein kinase A abbreviated period in both Tn-reconstituted nonfailing and cTnI-K36Q myofibrils, demonstrating acceleration of cross-bridge kinetics. CONCLUSIONS Sarcomere dynamics was found to be depressed under loaded conditions in DCM myofibrils because of impairment of thick-thin filament sliding. Thus, microscopic analysis of Ca-SPOC in human cardiac myofibrils is beneficial to systematically unveil the kinetic properties of single sarcomeres in various types of heart disease.
Collapse
Affiliation(s)
- Tatsuya Kagemoto
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.)
| | - Kotaro Oyama
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Mitsunori Yamane
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.)
| | - Seiichi Tsukamoto
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Fuyu Kobirumaki-Shimozawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.)
| | - Amy Li
- School of Medical Sciences, Bosch Institute, The University of Sydney, Australia (A.L., C.D.R.)
| | - Cristobal Dos Remedios
- School of Medical Sciences, Bosch Institute, The University of Sydney, Australia (A.L., C.D.R.)
| | - Norio Fukuda
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan (K.O., S.T., F.K.-S., N.F.).
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Tokyo, Japan (T.K., M.Y., S.I.).
| |
Collapse
|
36
|
Deelen P, van Dam S, Herkert JC, Karjalainen JM, Brugge H, Abbott KM, van Diemen CC, van der Zwaag PA, Gerkes EH, Zonneveld-Huijssoon E, Boer-Bergsma JJ, Folkertsma P, Gillett T, van der Velde KJ, Kanninga R, van den Akker PC, Jan SZ, Hoorntje ET, Te Rijdt WP, Vos YJ, Jongbloed JDH, van Ravenswaaij-Arts CMA, Sinke R, Sikkema-Raddatz B, Kerstjens-Frederikse WS, Swertz MA, Franke L. Improving the diagnostic yield of exome- sequencing by predicting gene-phenotype associations using large-scale gene expression analysis. Nat Commun 2019; 10:2837. [PMID: 31253775 PMCID: PMC6599066 DOI: 10.1038/s41467-019-10649-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/23/2019] [Indexed: 02/06/2023] Open
Abstract
The diagnostic yield of exome and genome sequencing remains low (8-70%), due to incomplete knowledge on the genes that cause disease. To improve this, we use RNA-seq data from 31,499 samples to predict which genes cause specific disease phenotypes, and develop GeneNetwork Assisted Diagnostic Optimization (GADO). We show that this unbiased method, which does not rely upon specific knowledge on individual genes, is effective in both identifying previously unknown disease gene associations, and flagging genes that have previously been incorrectly implicated in disease. GADO can be run on www.genenetwork.nl by supplying HPO-terms and a list of genes that contain candidate variants. Finally, applying GADO to a cohort of 61 patients for whom exome-sequencing analysis had not resulted in a genetic diagnosis, yields likely causative genes for ten cases.
Collapse
Affiliation(s)
- Patrick Deelen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Sipko van Dam
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Johanna C Herkert
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Juha M Karjalainen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Harm Brugge
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Kristin M Abbott
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Cleo C van Diemen
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Paul A van der Zwaag
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Erica H Gerkes
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Evelien Zonneveld-Huijssoon
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Jelkje J Boer-Bergsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Pytrik Folkertsma
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Tessa Gillett
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - K Joeri van der Velde
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Roan Kanninga
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Peter C van den Akker
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Sabrina Z Jan
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Edgar T Hoorntje
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,Netherlands Heart Institute, 3511 EP, Utrecht, The Netherlands
| | - Wouter P Te Rijdt
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,Netherlands Heart Institute, 3511 EP, Utrecht, The Netherlands
| | - Yvonne J Vos
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Jan D H Jongbloed
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Conny M A van Ravenswaaij-Arts
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Richard Sinke
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | - Birgit Sikkema-Raddatz
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands
| | | | - Morris A Swertz
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.,University of Groningen, University Medical Center Groningen, Genomics Coordination Center, 9700 VB, Groningen, The Netherlands
| | - Lude Franke
- University of Groningen, University Medical Center Groningen, Department of Genetics, 9700 VB, Groningen, The Netherlands.
| |
Collapse
|
37
|
Li A, Lal S, Dos Remedios CG. A step towards understanding the molecular nature of human heart failure: advances using the Sydney Heart Bank collection. Biophys Rev 2019; 11:241-244. [PMID: 30891671 DOI: 10.1007/s12551-019-00514-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Amy Li
- The University of Sydney, Sydney, Australia
| | - Sean Lal
- The University of Sydney, Sydney, Australia
| | | |
Collapse
|
38
|
Whitley JA, Ex-Willey AM, Marzolf DR, Ackermann MA, Tongen AL, Kokhan O, Wright NT. Obscurin is a semi-flexible molecule in solution. Protein Sci 2019; 28:717-726. [PMID: 30666746 DOI: 10.1002/pro.3578] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 01/14/2019] [Indexed: 01/10/2023]
Abstract
Obscurin, a giant modular cytoskeletal protein, is comprised mostly of tandem immunoglobulin-like (Ig-like) domains. This architecture allows obscurin to connect distal targets within the cell. The linkers connecting the Ig domains are usually short (3-4 residues). The physical effect arising from these short linkers is not known; such linkers may lead to a stiff elongated molecule or, conversely, may lead to a more compact and dynamic structure. In an effort to better understand how linkers affect obscurin flexibility, and to better understand the physical underpinnings of this flexibility, here we study the structure and dynamics of four representative sets of dual obscurin Ig domains using experimental and computational techniques. We find in all cases tested that tandem obscurin Ig domains interact at the poles of each domain and tend to stay relatively extended in solution. NMR, SAXS, and MD simulations reveal that while tandem domains are elongated, they also bend and flex significantly. By applying this behavior to a simplified model, it becomes apparent obscurin can link targets more than 200 nm away. However, as targets get further apart, obscurin begins acting as a spring and requires progressively more energy to further elongate.
Collapse
Affiliation(s)
- Jacob A Whitley
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, 22807
| | - Aidan M Ex-Willey
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, 22807.,Department of Physiology and Cell Biology, Wexner Medical Center, Ohio State University, Columbus, Ohio, 43210
| | - Daniel R Marzolf
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, 22807
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, Wexner Medical Center, Ohio State University, Columbus, Ohio, 43210
| | - Anthony L Tongen
- Department of Mathematics and Statistics, James Madison University, Harrisonburg, Virginia, 22807
| | - Oleksandr Kokhan
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, 22807
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, 22807
| |
Collapse
|
39
|
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
|
40
|
Sedaghat-Hamedani F, Haas J, Zhu F, Geier C, Kayvanpour E, Liss M, Lai A, Frese K, Pribe-Wolferts R, Amr A, Li DT, Samani OS, Carstensen A, Bordalo DM, Müller M, Fischer C, Shao J, Wang J, Nie M, Yuan L, Haßfeld S, Schwartz C, Zhou M, Zhou Z, Shu Y, Wang M, Huang K, Zeng Q, Cheng L, Fehlmann T, Ehlermann P, Keller A, Dieterich C, Streckfuß-Bömeke K, Liao Y, Gotthardt M, Katus HA, Meder B. Clinical genetics and outcome of left ventricular non-compaction cardiomyopathy. Eur Heart J 2018; 38:3449-3460. [PMID: 29029073 DOI: 10.1093/eurheartj/ehx545] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 09/20/2017] [Indexed: 12/30/2022] Open
Abstract
Aims In this study, we aimed to clinically and genetically characterize LVNC patients and investigate the prevalence of variants in known and novel LVNC disease genes. Introduction Left ventricular non-compaction cardiomyopathy (LVNC) is an increasingly recognized cause of heart failure, arrhythmia, thromboembolism, and sudden cardiac death. We sought here to dissect its genetic causes, phenotypic presentation and outcome. Methods and results In our registry with follow-up of in the median 61 months, we analysed 95 LVNC patients (68 unrelated index patients and 27 affected relatives; definite familial LVNC = 23.5%) by cardiac phenotyping, molecular biomarkers and exome sequencing. Cardiovascular events were significantly more frequent in LVNC patients compared with an age-matched group of patients with non-ischaemic dilated cardiomyopathy (hazard ratio = 2.481, P = 0.002). Stringent genetic classification according to ACMG guidelines revealed that TTN, LMNA, and MYBPC3 are the most prevalent disease genes (13 patients are carrying a pathogenic truncating TTN variant, odds ratio = 40.7, Confidence interval = 21.6-76.6, P < 0.0001, percent spliced in 76-100%). We also identified novel candidate genes for LVNC. For RBM20, we were able to perform detailed familial, molecular and functional studies. We show that the novel variant p.R634L in the RS domain of RBM20 co-segregates with LVNC, leading to titin mis-splicing as revealed by RNA sequencing of heart tissue in mutation carriers, protein analysis, and functional splice-reporter assays. Conclusion Our data demonstrate that the clinical course of symptomatic LVNC can be severe. The identified pathogenic variants and distribution of disease genes-a titin-related pathomechanism is found in every fourth patient-should be considered in genetic counselling of patients. Pathogenic variants in the nuclear proteins Lamin A/C and RBM20 were associated with worse outcome.
Collapse
Affiliation(s)
- Farbod Sedaghat-Hamedani
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Jan Haas
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Feng Zhu
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Christian Geier
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité Medical Faculty and Max Delbrück Center for Molecular Medicine (MDC), Augustenburger Platz 1, 13353 Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Elham Kayvanpour
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Martin Liss
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Alan Lai
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Karen Frese
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Regina Pribe-Wolferts
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Ali Amr
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Daniel Tian Li
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Omid Shirvani Samani
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Avisha Carstensen
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Diana Martins Bordalo
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Marion Müller
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Christine Fischer
- Department of Human Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | - Jing Shao
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Ming Nie
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Li Yuan
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Sabine Haßfeld
- Department of Cardiology, Virchow Klinikum, Charité University Medicine Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christine Schwartz
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation of Charité Medical Faculty and Max Delbrück Center for Molecular Medicine (MDC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Min Zhou
- Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Yanwen Shu
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Min Wang
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Kai Huang
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Qiutang Zeng
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Longxian Cheng
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Tobias Fehlmann
- Department of Bioinformatics, University of Saarland, Building E2.1, 66123 Saarbrücken, Germany
| | - Philipp Ehlermann
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Andreas Keller
- Department of Bioinformatics, University of Saarland, Building E2.1, 66123 Saarbrücken, Germany
| | - Christoph Dieterich
- DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany.,Department of Medicine III, Klaus Tschira Institute for Computational Cardiology, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | - Katrin Streckfuß-Bömeke
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany.,DZHK (German Centre for Cardiovascular Research), Göttingen, Germany
| | - Yuhua Liao
- Department of Cardiology, Institute of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022 Wuhan, China
| | - Michael Gotthardt
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany.,Neuromuscular and Cardiovascular Cell Biology, Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092 Berlin, Germany
| | - Hugo A Katus
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| | - Benjamin Meder
- Department of Medicine III, Institute for Cardiomyopathies Heidelberg (ICH), University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg, Germany
| |
Collapse
|
41
|
The Molecular Mechanisms of Mutations in Actin and Myosin that Cause Inherited Myopathy. Int J Mol Sci 2018; 19:ijms19072020. [PMID: 29997361 PMCID: PMC6073311 DOI: 10.3390/ijms19072020] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 07/06/2018] [Accepted: 07/08/2018] [Indexed: 12/23/2022] Open
Abstract
The discovery that mutations in myosin and actin genes, together with mutations in the other components of the muscle sarcomere, are responsible for a range of inherited muscle diseases (myopathies) has revolutionized the study of muscle, converting it from a subject of basic science to a relevant subject for clinical study and has been responsible for a great increase of interest in muscle studies. Myopathies are linked to mutations in five of the myosin heavy chain genes, three of the myosin light chain genes, and three of the actin genes. This review aims to determine to what extent we can explain disease phenotype from the mutant genotype. To optimise our chances of finding the right mechanism we must study a myopathy where there are a large number of different mutations that cause a common phenotype and so are likely to have a common mechanism: a corollary to this criterion is that if any mutation causes the disease phenotype but does not correspond to the proposed mechanism, then the whole mechanism is suspect. Using these criteria, we consider two cases where plausible genotype-phenotype mechanisms have been proposed: the actin “A-triad” and the myosin “mesa/IHD” models.
Collapse
|
42
|
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
|
43
|
An historical perspective of the discovery of titin filaments -Part 2. Biophys Rev 2018; 10:1201-1203. [PMID: 29332246 DOI: 10.1007/s12551-017-0393-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 12/18/2017] [Indexed: 10/18/2022] Open
Abstract
In 2017, a Special Issue of Biophysical Reviews was devoted to "Titin and Its Binding Partners. The issue contained a review: "An historical perspective of the discovery of titin filaments" by dos Remedios and Gilmour that was intended to be a history of the discovery of the giant protein titin, previously named connectin. The review took readers back to the earliest discovery of the so-called third filament component of skeletal and cardiac muscle sarcomeres and ended in 1969. Recently, my colleague Shin'ichi Ishiwata gently reminded me of two papers published in 1990 and 1993 that were unwittingly omitted from the original historical perspective. In the first paper (J Cell Biol 110:53-62, 1990), Funatsu et al. examined the elastic filaments in skeletal muscle using a combination of light and electron microscopy, but they also measured resting as well as passive stiffness mechanical measurements to establish that connectin (titin) is responsible for both stiffness and fiber tension. In the second paper (J Cell Biol 120:711-724, 1993), Funatsu et al. used permeabilised cardiac muscle myocytes (from rabbit papillary muscles) and focussed on filament ultrastructure using either freeze-substitution or deep-etched replica methods to visualise connectin/titin filaments in fibers with and without actin and myosin filaments.
Collapse
|
44
|
Rajendran BK, Deng CX. A comprehensive genomic meta-analysis identifies confirmatory role of OBSCN gene in breast tumorigenesis. Oncotarget 2017; 8:102263-102276. [PMID: 29254242 PMCID: PMC5731952 DOI: 10.18632/oncotarget.20404] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
The giant multifunctional protein "OBSCURIN" is encoded by OBSCN gene and is mostly expressed in cardiac and other skeletal muscles responsible for myofibrils organization. Loss of OBSCURIN affects the entire downstream pathway proteins vital for various cellular functions including cell integration and cell adhesion. The OBSCN gene mutations are more frequently observed in various muscular diseases, and cancers. Nevertheless, the direct role of OBSCN in tumorigenesis remains elusive. Interestingly, in clinical breast cancer samples a significant number of function changing mutations have been identified in OBSCN gene. In this study, we identified a significant role of OBSCN by conducting an integrative analysis of copy number alterations, functional mutations, gene methylation and expression data from various BRCA cancer projects data available on cBioPortal and TCGA firebrowse portal. Finally, we carried out genetic network analysis, which revealed that OBSCN gene plays a significant role in GPCR, RAS, p75 or Wnt signaling pathways. Similarly, OBSCN gene interacts with many cancer-associated genes involved in breast tumorigenesis. The OBSCN gene probably regulates breast cancer progression and metastasis and the prognostic molecular signatures such as copy number alterations and gene expression of OBSCN may serve as a tool to identify breast tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Barani Kumar Rajendran
- Cancer Research Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Chu-Xia Deng
- Cancer Research Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
45
|
Vikhorev PG, Smoktunowicz N, Munster AB, Copeland O, Kostin S, Montgiraud C, Messer AE, Toliat MR, Li A, Dos Remedios CG, Lal S, Blair CA, Campbell KS, Guglin M, Richter M, Knöll R, Marston SB. Abnormal contractility in human heart myofibrils from patients with dilated cardiomyopathy due to mutations in TTN and contractile protein genes. Sci Rep 2017; 7:14829. [PMID: 29093449 PMCID: PMC5665940 DOI: 10.1038/s41598-017-13675-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 09/26/2017] [Indexed: 11/29/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is an important cause of heart failure. Single gene mutations in at least 50 genes have been proposed to account for 25–50% of DCM cases and up to 25% of inherited DCM has been attributed to truncating mutations in the sarcomeric structural protein titin (TTNtv). Whilst the primary molecular mechanism of some DCM-associated mutations in the contractile apparatus has been studied in vitro and in transgenic mice, the contractile defect in human heart muscle has not been studied. In this study we isolated cardiac myofibrils from 3 TTNtv mutants, and 3 with contractile protein mutations (TNNI3 K36Q, TNNC1 G159D and MYH7 E1426K) and measured their contractility and passive stiffness in comparison with donor heart muscle as a control. We found that the three contractile protein mutations but not the TTNtv mutations had faster relaxation kinetics. Passive stiffness was reduced about 38% in all the DCM mutant samples. However, there was no change in maximum force or the titin N2BA/N2B isoform ratio and there was no titin haploinsufficiency. The decrease in myofibril passive stiffness was a common feature in all hearts with DCM-associated mutations and may be causative of DCM.
Collapse
Affiliation(s)
- Petr G Vikhorev
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom.
| | - Natalia Smoktunowicz
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom
| | - Alex B Munster
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom
| | - O'Neal Copeland
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom
| | - Sawa Kostin
- Max-Planck-Institute for Heart and Lung Research, Ludwigstrasse 43, 61231, Bad Nauheim, 61231, Germany
| | - Cecile Montgiraud
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom
| | - Andrew E Messer
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom
| | - Mohammad R Toliat
- Cologne Center for Genomics, University of Cologne, Cologne, 50931, Germany
| | - Amy Li
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Cristobal G Dos Remedios
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Sean Lal
- Discipline of Anatomy and Histology, Bosch Institute, University of Sydney, Sydney, NSW, 2006, Australia
| | - Cheavar A Blair
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Kenneth S Campbell
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Maya Guglin
- Division of Cardiovascular Medicine, Department of Physiology, University of Kentucky, Lexington, Kentucky, USA
| | - Manfred Richter
- Department of Cardiac Surgery, Kerckhoff-Clinic, Benekestrasse 2-8, Bad Nauheim, 61231, Germany
| | - Ralph Knöll
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom.,ICMC (Integrated Cardio Metabolic Centre), Myocardial Genetics, Karolinska Institutet, University Hospital, Heart and Vascular Theme, Novum, Hiss A, våning 7, Hälsovägen 7-9, Huddinge, 141 57, Sweden.,AstraZeneca R&D Gothenburg, R&D, Innovative Medicines & Early Development, Cardiovascular, Renal and Metabolic Diseases (CVRM), Pepparedsleden 1, SE-431 83, Mölndal, Sweden
| | - Steven B Marston
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, United Kingdom.
| |
Collapse
|
46
|
Rossi D, Palmio J, Evilä A, Galli L, Barone V, Caldwell TA, Policke RA, Aldkheil E, Berndsen CE, Wright NT, Malfatti E, Brochier G, Pierantozzi E, Jordanova A, Guergueltcheva V, Romero NB, Hackman P, Eymard B, Udd B, Sorrentino V. A novel FLNC frameshift and an OBSCN variant in a family with distal muscular dystrophy. PLoS One 2017; 12:e0186642. [PMID: 29073160 PMCID: PMC5657976 DOI: 10.1371/journal.pone.0186642] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/04/2017] [Indexed: 11/30/2022] Open
Abstract
A novel FLNC c.5161delG (p.Gly1722ValfsTer61) mutation was identified in two members of a French family affected by distal myopathy and in one healthy relative. This FLNC c.5161delG mutation is one nucleotide away from a previously reported FLNC mutation (c.5160delC) that was identified in patients and in asymptomatic carriers of three Bulgarian families with distal muscular dystrophy, indicating a low penetrance of the FLNC frameshift mutations. Given these similarities, we believe that the two FLNC mutations alone can be causative of distal myopathy without full penetrance. Moreover, comparative analysis of the clinical manifestations indicates that patients of the French family show an earlier onset and a complete segregation of the disease. As a possible explanation of this, the two French patients also carry a OBSCN c.13330C>T (p.Arg4444Trp) mutation. The p.Arg4444Trp variant is localized within the OBSCN Ig59 domain that, together with Ig58, binds to the ZIg9/ZIg10 domains of titin at Z-disks. Structural and functional studies indicate that this OBSCN p.Arg4444Trp mutation decreases titin binding by ~15-fold. On this line, we suggest that the combination of the OBSCN p.Arg4444Trp variant and of the FLNC c.5161delG mutation, can cooperatively affect myofibril stability and increase the penetrance of muscular dystrophy in the French family.
Collapse
Affiliation(s)
- Daniela Rossi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Johanna Palmio
- Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
| | - Anni Evilä
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Lucia Galli
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Virginia Barone
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Tracy A. Caldwell
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Rachel A. Policke
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Esraa Aldkheil
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Christopher E. Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Nathan T. Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Edoardo Malfatti
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Guy Brochier
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Albena Jordanova
- Molecular Neurogenomics Group, University of Antwerp, Antwerp, Belgium
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University-Sofia, Sofia, Bulgaria
| | | | - Norma Beatriz Romero
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Peter Hackman
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Bruno Eymard
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Bjarne Udd
- Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
- * E-mail:
| |
Collapse
|
47
|
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
|
48
|
Khalil A, Al-Haddad C, Hariri H, Shibbani K, Bitar F, Kurban M, Nemer G, Arabi M. A Novel Mutation in FOXC1 in a Lebanese Family with Congenital Heart Disease and Anterior Segment Dysgenesis: Potential Roles for NFATC1 and DPT in the Phenotypic Variations. Front Cardiovasc Med 2017; 4:58. [PMID: 28979898 PMCID: PMC5611365 DOI: 10.3389/fcvm.2017.00058] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/31/2017] [Indexed: 01/06/2023] Open
Abstract
Congenital heart diseases (CHDs) are still the leading cause of death in neonates. Anterior segment dysgenesis is a broad clinical phenotype that affects the normal development of the eye, leading in most of the cases to glaucoma which is still a major cause of blindness for children and adolescents. Despite tremendous insights gained from genetic studies, a clear genotype–phenotype correlation is still difficult to draw. In Lebanon, a small country with still a high rate of consanguineous marriages, there are little data on the epidemiology of glaucoma amongst children with or without CHD. We carried out whole exome sequencing (WES) on a family with anterior segment dysgenesis, and CHD composed of three affected children with glaucoma, two of them with structural cardiac defects and three healthy siblings. The results unravel a novel mutation in FOXC1 (p. R127H) segregating with the phenotype and inherited from the mother, who did not develop glaucoma. We propose a digenic model for glaucoma in this family by combining the FOXC1 variant with a missense variant inherited from the father in the dermatopontin (DPT) gene. We also unravel a novel NFATC1 missense mutation predicted to be deleterious and present only in the patient with a severe ocular and cardiac phenotype. This is the first report on FOXC1 using WES to genetically characterize a family with both ocular and cardiac malformations. Our results support the usage of such technology to have a better genotype–phenotype picture for Mendelian-inherited diseases for which expressivity and penetrance are still not answered.
Collapse
Affiliation(s)
- Athar Khalil
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | | | - Hadla Hariri
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Kamel Shibbani
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Fadi Bitar
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| | - Mazen Kurban
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, American University of Beirut, Beirut, Lebanon.,Department of Dermatology, Columbia University, New York, NY, United States
| | - Georges Nemer
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Mariam Arabi
- Department of Pediatrics and Adolescent Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
49
|
Dos Remedios CG, Lal SP, Li A, McNamara J, Keogh A, Macdonald PS, Cooke R, Ehler E, Knöll R, Marston SB, Stelzer J, Granzier H, Bezzina C, van Dijk S, De Man F, Stienen GJM, Odeberg J, Pontén F, Linke WA, Linke W, van der Velden J. The Sydney Heart Bank: improving translational research while eliminating or reducing the use of animal models of human heart disease. Biophys Rev 2017; 9:431-441. [PMID: 28808947 DOI: 10.1007/s12551-017-0305-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 01/09/2023] Open
Abstract
The Sydney Heart Bank (SHB) is one of the largest human heart tissue banks in existence. Its mission is to provide high-quality human heart tissue for research into the molecular basis of human heart failure by working collaboratively with experts in this field. We argue that, by comparing tissues from failing human hearts with age-matched non-failing healthy donor hearts, the results will be more relevant than research using animal models, particularly if their physiology is very different from humans. Tissue from heart surgery must generally be used soon after collection or it significantly deteriorates. Freezing is an option but it raises concerns that freezing causes substantial damage at the cellular and molecular level. The SHB contains failing samples from heart transplant patients and others who provided informed consent for the use of their tissue for research. All samples are cryopreserved in liquid nitrogen within 40 min of their removal from the patient, and in less than 5-10 min in the case of coronary arteries and left ventricle samples. To date, the SHB has collected tissue from about 450 failing hearts (>15,000 samples) from patients with a wide range of etiologies as well as increasing numbers of cardiomyectomy samples from patients with hypertrophic cardiomyopathy. The Bank also has hearts from over 120 healthy organ donors whose hearts, for a variety of reasons (mainly tissue-type incompatibility with waiting heart transplant recipients), could not be used for transplantation. Donor hearts were collected by the St Vincent's Hospital Heart and Lung transplantation team from local hospitals or within a 4-h jet flight from Sydney. They were flushed with chilled cardioplegic solution and transported to Sydney where they were quickly cryopreserved in small samples. Failing and/or donor samples have been used by more than 60 research teams around the world, and have resulted in more than 100 research papers. The tissues most commonly requested are from donor left ventricles, but right ventricles, atria, interventricular system, and coronary arteries vessels have also been reported. All tissues are stored for long-term use in liquid N or vapor (170-180 °C), and are shipped under nitrogen vapor to avoid degradation of sensitive molecules such as RNAs and giant proteins. We present evidence that the availability of these human heart samples has contributed to a reduction in the use of animal models of human heart failure.
Collapse
Affiliation(s)
- C G Dos Remedios
- Sydney Heart Bank, Discipline of Anatomy & Histology, University of Sydney, Sydney, Australia.
| | - S P Lal
- Sydney Heart Bank, Discipline of Anatomy & Histology, University of Sydney, Sydney, Australia
| | - A Li
- Sydney Heart Bank, Discipline of Anatomy & Histology, University of Sydney, Sydney, Australia.,Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - J McNamara
- Sydney Heart Bank, Discipline of Anatomy & Histology, University of Sydney, Sydney, Australia
| | - A Keogh
- Heart Transplant Unit, St Vincent's Hospital, Sydney, Australia
| | - P S Macdonald
- Heart Transplant Unit, St Vincent's Hospital, Sydney, Australia
| | - R Cooke
- Cardiovascular Research Institute, University of California San Francisco, California, USA
| | - E Ehler
- Cardiovascular Division, Randall Division of Cell and Molecular Biophysics, London, UK
| | - R Knöll
- Department of Medicine, Karolinska Institutet, Huddinge, Sweden
| | - S B Marston
- National Heart and Lung Institute, Imperial College London, London, UK
| | - J Stelzer
- Department of Physiology & Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - H Granzier
- Molecular Cardiovascular Research Program, University of Arizona, Tucson, USA
| | - C Bezzina
- Department of Experimental Cardiology, Heart Failure Research Center, Amsterdam, The Netherlands
| | - S van Dijk
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - F De Man
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - G J M Stienen
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - J Odeberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - F Pontén
- Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm, Sweden
| | | | - W Linke
- Ruhr University, Bochum, Germany
| | - J van der Velden
- Department of Physiology, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
50
|
Randazzo D, Pierantozzi E, Rossi D, Sorrentino V. The potential of obscurin as a therapeutic target in muscle disorders. Expert Opin Ther Targets 2017; 21:897-910. [DOI: 10.1080/14728222.2017.1361931] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Davide Randazzo
- Light Imaging Section, Office of Science and Technology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Daniela Rossi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| |
Collapse
|