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
|
Oghabian A, Jonson PH, Gayathri SN, Johari M, Nippala E, Andres DG, Munell F, Soriano JC, Duran MAS, Sinisalo J, Tolppanen H, Tolva J, Hackman P, Savarese M, Udd B. OBSCN undergoes extensive alternative splicing during human cardiac and skeletal muscle development. Skelet Muscle 2025; 15:5. [PMID: 40025502 PMCID: PMC11871629 DOI: 10.1186/s13395-025-00374-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/27/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Highly expressed in skeletal muscles, the gene Obscurin (i.e. OBSCN) has 121 non-overlapping exons and codes for some of the largest known mRNAs in the human genome. Furthermore, it plays an essential role in muscle development and function. Mutations in OBSCN are associated with several hypertrophic cardiomyopathies and muscular disorders. OBSCN undergoes extensive and complex alternative splicing, which is the main reason that its splicing regulation associated with skeletal and cardiac muscle development has not previously been thoroughly studied. METHODS We analyzed RNA-Seq data from skeletal and cardiac muscles extracted from 44 postnatal individuals and six fetuses. We applied the intron/exon level splicing analysis software IntEREst to study the splicing of OBSCN in the studied samples. The differential splicing analysis was adjusted for batch effects. Our comparisons revealed the splicing variations in OBSCN between the human skeletal and cardiac muscle, as well as between post-natal muscle (skeletal and cardiac) and the pre-natal equivalent muscle. RESULTS We detected several splicing regulations located in the 5'end, 3' end, and the middle of OBSCN that are associated with human cardiac or skeletal muscle development. Many of these alternative splicing events have not previously been reported. Our results also suggest that many of these muscle-development associated splicing events may be regulated by BUB3. CONCLUSIONS We conclude that the splicing of OBSCN is extensively regulated during the human skeletal/cardiac muscle development. We developed an interactive visualization tool that can be used by clinicians and researchers to study the inclusion of specific OBSCN exons in pre- and postnatal cardiac and skeletal muscles and access the statistics for the differential inclusion of the exons across the studied sample groups. The OBSCN exon inclusion map related to the human cardiac and skeletal muscle development is available at http://psivis.it.helsinki.fi:3838/OBSCN_PSIVIS/ . These findings are essential for an accurate pre- and postnatal clinical interpretation of the OBSCN exonic variants.
Collapse
Affiliation(s)
- Ali Oghabian
- Folkhälsan Research Center, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.
| | - Per Harald Jonson
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Swethaa Natraj Gayathri
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Mridul Johari
- Folkhälsan Research Center, Helsinki, Finland
- Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands, WA, Australia
| | | | - David Gomez Andres
- Pediatric Neuromuscular Unit. Child Neurology Department. Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR) ES, Barcelona, Spain
| | - Francina Munell
- Pediatric Neuromuscular Unit. Child Neurology Department. Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR) ES, Barcelona, Spain
| | | | | | - Juha Sinisalo
- Department of Obstetrics, Maternal Fetal Medicine Unit, Universitat Autònoma de Barcelona, Hospital Vall D'hebron, Barcelona, Spain
| | | | - Johanna Tolva
- Department of Pathology, Transplantation Laboratory, University of Helsinki, Helsinki, Finland
| | - Peter Hackman
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland
- Department of Medical Genetics, University of Helsinki, Helsinki, Finland
- Pediatric Neuromuscular Unit. Child Neurology Department. Hospital Universitari Vall d'Hebron, Vall d'Hebron Research Institute (VHIR) ES, Barcelona, Spain
- Department of Neurology, Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
| |
Collapse
|
3
|
Isaacs A, Zeemering S, Winters J, Batlle M, Bidar E, Boukens B, Casadei B, Chua W, Crijns HJGM, Fabritz L, Guasch E, Hatem SN, Hermans B, Kääb S, Kawczynski M, Maesen B, Maessen J, Mont L, Sinner MF, Wakili R, Verheule S, Kirchhof P, Schotten U, Stoll M. Lateral Atrial Expression Patterns Provide Insights into Local Transcription Disequilibrium Contributing to Disease Susceptibility. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2025; 18:e004594. [PMID: 39846178 DOI: 10.1161/circgen.124.004594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 11/19/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND Transcriptional dysregulation, possibly affected by genetic variation, contributes to disease etiology. Due to dissimilarities in development, function, and remodeling during disease progression, transcriptional differences between the left atrium (LA) and right atrium (RA) may provide insight into diseases such as atrial fibrillation. METHODS Lateral differences in atrial transcription were evaluated in CATCH ME (Characterizing Atrial fibrillation by Translating its Causes into Health Modifiers in the Elderly) using a 2-stage discovery and replication design. The design took advantage of the availability of 32 paired samples, for which both LA and RA tissue were obtained, as a discovery cohort, and 98 LA and 69 RA unpaired samples utilized as a replication cohort. RESULTS A total of 714 transcripts were identified and replicated as differentially expressed (DE) between LA and RA, as well as 98 exons in 55 genes. Approximately 50% of DE transcripts were colocated with another frequently correlated DE transcript (PFDR ≤0.05 for 579 regions). These "transcription disequilibrium" blocks contained examples including side-specific differential exon usage, such as the PITX2 locus, where ENPEP showed evidence of differential exon usage. Analysis of this region in conjunction with BMP10 identified rs9790621 as associated with ENPEP transcription in LA, while rs7687878 was associated with BMP10 expression in RA. In RA, BMP10 and ENPEP were strongly correlated in noncarriers, which was attenuated in risk-allele carriers, where BMP10 and PITX2 expression were strongly correlated. CONCLUSIONS These results significantly expand knowledge of the intricate, tissue-specific transcriptional landscape in human atria, including DE transcripts and side-specific isoform expression. Furthermore, they suggest the existence of blocks of transcription disequilibrium influenced by genetics.
Collapse
Affiliation(s)
- Aaron Isaacs
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Maastricht Center for Systems Biology (A.I.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stef Zeemering
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Joris Winters
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Montserrat Batlle
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
| | - Elham Bidar
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bas Boukens
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Barbara Casadei
- NIHR Oxford Biomedical Research Center, John Radcliffe Hospital, University of Oxford, United Kingdom (B.C.)
| | - Winnie Chua
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
| | - Harry J G M Crijns
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Cardiology (H.J.G.M.C.), Maastricht University Medical Centre, the Netherlands
| | - Larissa Fabritz
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- University Center of Cardiovascular Sciences, University Medical Center Hamburg Eppendorf, Germany (L.F.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Eduard Guasch
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Stephane N Hatem
- INSERM UMRS 1166, Institute of Cardiometabolism and Nutrition (ICAN), Sorbonne University, Paris, France (S.N.H.)
| | - Ben Hermans
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Stefan Kääb
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Michal Kawczynski
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Bart Maesen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Jos Maessen
- Departments of Cardiothoracic Surgery (E.B., M.K., B.M., J.M.), Maastricht University Medical Centre, the Netherlands
| | - Lluis Mont
- Institute of Biomedical Research August Pi Sunyer, Barcelona, Spain (M.B., E.G., L.M.)
- CIBERCV, Madrid, Spain (M.B., E.G., L.M.)
- Clinic Barcelona, Universitat de Barcelona, Spain (E.G., L.M.)
| | - Moritz F Sinner
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
- University Heart and Vascular Center, University Hospital Hamburg Eppendorf, Germany (S.K., M.F.S.)
- Department of Cardiology, University Hospital of Munich, Germany (S.K., M.F.S.)
| | - Reza Wakili
- Department of Medicine and Cardiology, Goethe University, Frankfurt, Germany (R.W.)
- German Center for Cardiovascular Research DZHK, Rhine-Main (R.W.)
| | - Sander Verheule
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Paulus Kirchhof
- Institute of Cardiovascular Sciences, Birmingham, United Kingdom (W.C., L.F., P.K.)
- Department of Cardiology, University Heart and Vascular Center Hamburg, Germany (P.K.)
- German Center for Cardiovascular Research (DZHK), Hamburg/Kiel/Lübeck, Germany (L.F., S.K., M.F.S., P.K.)
| | - Ulrich Schotten
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Physiology (A.I., S.Z., J.W., B.B., B.H., M.K., S.V., U.S.), Maastricht University, the Netherlands
| | - Monika Stoll
- CARIM School for Cardiovascular Diseases (A.I., S.Z., J.W., B.B., H.J.G.M.C., B.H., M.K., S.V., U.S., M.S.), Maastricht University, the Netherlands
- Department of Biochemistry (M.S.), Maastricht University, the Netherlands
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, Germany (M.S.)
| |
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
|
Zemorshidi F, Töpf A, Claeys KG, McFarlane A, Patton A, Nafissi S, Straub V. Novel OBSCN variants associated with a risk to exercise-intolerance and rhabdomyolysis. Neuromuscul Disord 2024; 34:83-88. [PMID: 38159459 DOI: 10.1016/j.nmd.2023.10.013] [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: 03/26/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024]
Abstract
Obscurin, encoded by the OBSCN gene, is a muscle protein consisting of three main splice isoforms, obscurin-A, obscurin-B, and obscurin kinase-only protein (also known as KIAA1639 or Obsc-kin). Obscurin is located at the M-band and Z-disks and interacts with titin and myomesin. It plays an important role in the stability and maintenance of the A- and M-bands and the subsarcolemmal organization of the microtubule network. Furthermore, obscurin is involved in Ca2+ regulation and sarcoplasmic reticulum function and is connected to several other muscle proteins. OBSCN gene variants have been reported to be relatively common in inherited cardiomyopathies. Here we reported two young patients with a history of cramps, myalgia, exercise intolerance, rhabdomyolysis, and myoglobinuria without any evidence of concomitant cardiomyopathy in association with novel OBSCN variants (c.24822C>A and c.2653+1G>C). Obscurin-deficient muscle fibers seem to have increased susceptibility to damage triggered by exercise that may lead to rhabdomyolysis. More studies are needed to clarify the diverse clinical phenotypes and the pathophysiology of OBSCN gene variants.
Collapse
Affiliation(s)
- Fariba Zemorshidi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Iran; Department of Neurology, Shariati Hospital,Tehran University of Medical Sciences, Tehran, Iran; Department of Neurology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ana Töpf
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Kristl G Claeys
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Adam McFarlane
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Annabel Patton
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Shahriar Nafissi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Iran; Department of Neurology, Shariati Hospital,Tehran University of Medical Sciences, Tehran, Iran.
| | - Volker Straub
- John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
7
|
Mauriello GE, Moncure GE, Nowzari RA, Miller CJ, Wright NT. The N-terminus of obscurin is flexible in solution. Proteins 2023; 91:485-496. [PMID: 36306263 DOI: 10.1002/prot.26442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/06/2022]
Abstract
The N-terminal half of the giant cytoskeletal protein obscurin is comprised of more than 50 Ig-like domains, arranged in tandem. Domains 18-51 are connected to each other through short 5-residue linkers, and this arrangement has been previously shown to form a semi-flexible rod in solution. Domains 1-18 generally have slightly longer ~7 residue interdomain linkers, and the multidomain structure and motion conferred by this kind of linker is understudied. Here, we use NMR, SAXS, and MD to show that these longer linkers are associated with significantly more domain/domain flexibility, with the resulting multidomain structure being moderately compact. Further examination of the relationship between interdomain flexibility and linker length shows there is a 5 residue "sweet spot" linker length that results in dual-domain systems being extended, and conversely that both longer or shorter linkers result in a less extended structure. This detailed knowledge of the obscurin N terminus structure and flexibility allowed for mathematical modeling of domains 1-18, which suggests that this region likely forms tangles if left alone in solution. Given how infrequently protein tangles occur in nature, and given the pathological outcomes that occur when tangles do arise, our data suggest that obscurin is likely either significantly scaffolded or else externally extended in the cell.
Collapse
Affiliation(s)
- Gianna E Mauriello
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, USA
| | - Grace E Moncure
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, USA
| | - Roujon A Nowzari
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, USA
| | - Callie J Miller
- Department of Engineering, James Madison University, Harrisonburg, Virginia, USA
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, USA
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
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
|
11
|
Exploring the Potential of Symmetric Exon Deletion to Treat Non-Ischemic Dilated Cardiomyopathy by Removing Frameshift Mutations in TTN. Genes (Basel) 2022; 13:genes13061093. [PMID: 35741855 PMCID: PMC9222585 DOI: 10.3390/genes13061093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
Non-ischemic dilated cardiomyopathy (DCM) is one of the most frequent pathologies requiring cardiac transplants. Even though the etiology of this disease is complex, frameshift mutations in the giant sarcomeric protein Titin could explain up to 25% of the familial and 18% of the sporadic cases of DCM. Many studies have shown the potential of genome editing using CRISPR/Cas9 to correct truncating mutations in sarcomeric proteins and have established the grounds for myoediting. However, these therapies are still in an immature state, with only few studies showing an efficient treatment of cardiac diseases. This publication hypothesizes that the Titin (TTN)-specific gene structure allows the application of myoediting approaches in a broad range of locations to reframe TTNtvvariants and to treat DCM patients. Additionally, to pave the way for the generation of efficient myoediting approaches for DCM, we screened and selected promising target locations in TTN. We conceptually explored the deletion of symmetric exons as a therapeutic approach to restore TTN’s reading frame in cases of frameshift mutations. We identified a set of 94 potential candidate exons of TTN that we consider particularly suitable for this therapeutic deletion. With this study, we aim to contribute to the development of new therapies to efficiently treat titinopathies and other diseases caused by mutations in genes encoding proteins with modular structures, e.g., Obscurin.
Collapse
|
12
|
The Oxidative Balance Orchestrates the Main Keystones of the Functional Activity of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7714542. [PMID: 35047109 PMCID: PMC8763515 DOI: 10.1155/2022/7714542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/03/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
This review is aimed at providing an overview of the key hallmarks of cardiomyocytes in physiological and pathological conditions. The main feature of cardiac tissue is the force generation through contraction. This process requires a conspicuous energy demand and therefore an active metabolism. The cardiac tissue is rich of mitochondria, the powerhouses in cells. These organelles, producing ATP, are also the main sources of ROS whose altered handling can cause their accumulation and therefore triggers detrimental effects on mitochondria themselves and other cell components thus leading to apoptosis and cardiac diseases. This review highlights the metabolic aspects of cardiomyocytes and wanders through the main systems of these cells: (a) the unique structural organization (such as different protein complexes represented by contractile, regulatory, and structural proteins); (b) the homeostasis of intracellular Ca2+ that represents a crucial ion for cardiac functions and E-C coupling; and (c) the balance of Zn2+, an ion with a crucial impact on the cardiovascular system. Although each system seems to be independent and finely controlled, the contractile proteins, intracellular Ca2+ homeostasis, and intracellular Zn2+ signals are strongly linked to each other by the intracellular ROS management in a fascinating way to form a "functional tetrad" which ensures the proper functioning of the myocardium. Nevertheless, if ROS balance is not properly handled, one or more of these components could be altered resulting in deleterious effects leading to an unbalance of this "tetrad" and promoting cardiovascular diseases. In conclusion, this "functional tetrad" is proposed as a complex network that communicates continuously in the cardiomyocytes and can drive the switch from physiological to pathological conditions in the heart.
Collapse
|
13
|
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
|
14
|
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
|
15
|
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
|
16
|
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
|
17
|
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disease of the myocardium characterized by a hypertrophic left ventricle with a preserved or increased ejection fraction. Cardiac hypertrophy is often asymmetrical, which is associated with left ventricular outflow tract obstruction. Myocyte hypertrophy, disarray, and myocardial fibrosis constitute the histological features of HCM. HCM is a relatively benign disease but an important cause of sudden cardiac death in the young and heart failure in the elderly. Pathogenic variants (PVs) in genes encoding protein constituents of the sarcomeres are the main causes of HCM. PVs exhibit a gradient of effect sizes, as reflected in their penetrance and variable phenotypic expression of HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin binding protein C, respectively, are the two most common causal genes and responsible for ≈40% of all HCM cases but a higher percentage of HCM in large families. PVs in genes encoding protein components of the thin filaments are responsible for ≈5% of the HCM cases. Whereas pathogenicity of the genetic variants in large families has been firmly established, ascertainment causality of the PVs in small families and sporadic cases is challenging. In the latter category, PVs are best considered as probabilistic determinants of HCM. Deciphering the genetic basis of HCM has enabled routine genetic testing and has partially elucidated the underpinning mechanism of HCM as increased number of the myosin molecules that are strongly bound to actin. The discoveries have led to the development of mavacamten that targets binding of the myosin molecule to actin filaments and imparts beneficial clinical effects. In the coming years, the yield of the genetic testing is expected to be improved and the so-called missing causal gene be identified. The advances are also expected to enable development of additional specific therapies and editing of the mutations in HCM.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston
| |
Collapse
|
18
|
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
|
19
|
Grogan A, Coleman A, Joca H, Granzier H, Russel MW, Ward CW, Kontrogianni-Konstantopoulos A. Deletion of obscurin immunoglobulin domains Ig58/59 leads to age-dependent cardiac remodeling and arrhythmia. Basic Res Cardiol 2020; 115:60. [PMID: 32910221 PMCID: PMC9302192 DOI: 10.1007/s00395-020-00818-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/06/2020] [Indexed: 12/23/2022]
Abstract
Obscurin comprises a family of giant modular proteins that play key structural and regulatory roles in striated muscles. Immunoglobulin domains 58/59 (Ig58/59) of obscurin mediate binding to essential modulators of muscle structure and function, including canonical titin, a smaller splice variant of titin, termed novex-3, and phospholamban (PLN). Importantly, missense mutations localized within the obscurin-Ig58/59 region that affect binding to titins and/or PLN have been linked to the development of myopathy in humans. To elucidate the pathophysiological role of this region, we generated a constitutive deletion mouse model, Obscn-ΔIg58/59, that expresses obscurin lacking Ig58/59, and determined the consequences of this manipulation on cardiac morphology and function under conditions of acute stress and through the physiological process of aging. Our studies show that young Obscn-ΔIg58/59 mice are susceptible to acute β-adrenergic stress. Moreover, sedentary Obscn-ΔIg58/59 mice develop left ventricular hypertrophy that progresses to dilation, contractile impairment, atrial enlargement, and arrhythmia as a function of aging with males being more affected than females. Experiments in ventricular cardiomyocytes revealed altered Ca2+ cycling associated with changes in the expression and/or phosphorylation levels of major Ca2+ cycling proteins, including PLN, SERCA2, and RyR2. Taken together, our work demonstrates that obscurin-Ig58/59 is an essential regulatory module in the heart and its deletion leads to age- and sex-dependent cardiac remodeling, ventricular dilation, and arrhythmia due to deregulated Ca2+ cycling.
Collapse
MESH Headings
- Action Potentials
- Age Factors
- Animals
- Arrhythmias, Cardiac/enzymology
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Calcium Signaling
- Calcium-Binding Proteins/metabolism
- Female
- Gene Deletion
- Heart Rate
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/pathology
- Hypertrophy, Left Ventricular/physiopathology
- Immunoglobulin Domains
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphorylation
- Protein Serine-Threonine Kinases/deficiency
- Protein Serine-Threonine Kinases/genetics
- Rho Guanine Nucleotide Exchange Factors/deficiency
- Rho Guanine Nucleotide Exchange Factors/genetics
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Sedentary Behavior
- Sex Factors
- Ventricular Dysfunction, Left/enzymology
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/pathology
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Andrew Coleman
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Humberto Joca
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Henk Granzier
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ, 85724, USA
| | - Mark W Russel
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Christopher W Ward
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | | |
Collapse
|
20
|
Hu LYR, Kontrogianni-Konstantopoulos A. Proteomic Analysis of Myocardia Containing the Obscurin R4344Q Mutation Linked to Hypertrophic Cardiomyopathy. Front Physiol 2020; 11:478. [PMID: 32528308 PMCID: PMC7247546 DOI: 10.3389/fphys.2020.00478] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 04/20/2020] [Indexed: 12/25/2022] Open
Abstract
Obscurin is a giant cytoskeletal protein with structural and regulatory roles encoded by the OBSCN gene. Recently, mutations in OBSCN were associated with the development of different forms of cardiomyopathies, including hypertrophic cardiomyopathy (HCM). We previously reported that homozygous mice carrying the HCM-linked R4344Q obscurin mutation develop arrhythmia by 1-year of age under sedentary conditions characterized by increased heart rate, frequent incidents of premature ventricular contractions, and episodes of spontaneous ventricular tachycardia. In an effort to delineate the molecular mechanisms that contribute to the observed arrhythmic phenotype, we subjected protein lysates prepared from left ventricles of 1-year old R4344Q and wild-type mice to comparative proteomics analysis using tandem mass spectrometry; raw data are available via ProteomeXchange with identifier PXD017314. We found that the expression levels of proteins involved in cardiac function and disease, cytoskeletal organization, electropotential regulation, molecular transport and metabolism were significantly altered. Moreover, phospho-proteomic evaluation revealed changes in the phosphorylation profile of Ca2+ cycling proteins, including sAnk1.5, a major binding partner of obscurin localized in the sarcoplasmic reticulum; notably, this is the first report indicating that sAnk1 undergoes phosphorylation. Taken together, our findings implicate obscurin in diverse cellular processes within the myocardium, which is consistent with its multiple binding partners, localization in different subcellular compartments, and disease association.
Collapse
Affiliation(s)
- Li-Yen R Hu
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD, United States
| | | |
Collapse
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
Forleo C, D’Erchia AM, Sorrentino S, Manzari C, Chiara M, Iacoviello M, Guaricci AI, De Santis D, Musci RL, La Spada A, Marangelli V, Pesole G, Favale S. Targeted next-generation sequencing detects novel gene-phenotype associations and expands the mutational spectrum in cardiomyopathies. PLoS One 2017; 12:e0181842. [PMID: 28750076 PMCID: PMC5531468 DOI: 10.1371/journal.pone.0181842] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 07/08/2017] [Indexed: 12/19/2022] Open
Abstract
Cardiomyopathies are a heterogeneous group of primary diseases of the myocardium, including hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM), and arrhythmogenic right ventricular cardiomyopathy (ARVC), with higher morbidity and mortality. These diseases are genetically diverse and associated with rare mutations in a large number of genes, many of which overlap among the phenotypes. To better investigate the genetic overlap between these three phenotypes and to identify new genotype-phenotype correlations, we designed a custom gene panel consisting of 115 genes known to be associated with cardiomyopathic phenotypes and channelopathies. A cohort of 38 unrelated patients, 16 affected by DCM, 14 by HCM and 8 by ARVC, was recruited for the study on the basis of more severe phenotypes and family history of cardiomyopathy and/or sudden death. We detected a total of 142 rare variants in 40 genes, and all patients were found to be carriers of at least one rare variant. Twenty-eight of the 142 rare variants were also predicted as potentially pathogenic variants and found in 26 patients. In 23 out of 38 patients, we found at least one novel potential gene-phenotype association. In particular, we detected three variants in OBSCN gene in ARVC patients, four variants in ANK2 gene and two variants in DLG1, TRPM4, and AKAP9 genes in DCM patients, two variants in PSEN2 gene and four variants in AKAP9 gene in HCM patients. Overall, our results confirmed that cardiomyopathic patients could carry multiple rare gene variants; in addition, our investigation of the genetic overlap among cardiomyopathies revealed new gene-phenotype associations. Furthermore, as our study confirms, data obtained using targeted next-generation sequencing could provide a remarkable contribution to the molecular diagnosis of cardiomyopathies, early identification of patients at risk for arrhythmia development, and better clinical management of cardiomyopathic patients.
Collapse
Affiliation(s)
- Cinzia Forleo
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Anna Maria D’Erchia
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| | - Sandro Sorrentino
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Caterina Manzari
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| | - Matteo Chiara
- Department of Biosciences, University of Milano, Milano, Italy
| | - Massimo Iacoviello
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Andrea Igoren Guaricci
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Delia De Santis
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Rita Leonarda Musci
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Antonino La Spada
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Vito Marangelli
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Bari, Italy
| | - Stefano Favale
- Cardiology Unit, Department of Emergency and Organ Transplantation, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
32
|
Abstract
Cardiac and skeletal striated muscles are intricately designed machines responsible for muscle contraction. Coordination of the basic contractile unit, the sarcomere, and the complex cytoskeletal networks are critical for contractile activity. The sarcomere is comprised of precisely organized individual filament systems that include thin (actin), thick (myosin), titin, and nebulin. Connecting the sarcomere to other organelles (e.g., mitochondria and nucleus) and serving as the scaffold to maintain cellular integrity are the intermediate filaments. The costamere, on the other hand, tethers the sarcomere to the cell membrane. Unique structures like the intercalated disc in cardiac muscle and the myotendinous junction in skeletal muscle help synchronize and transmit force. Intense investigation has been done on many of the proteins that make up these cytoskeletal assemblies. Yet the details of their function and how they interconnect have just started to be elucidated. A vast number of human myopathies are contributed to mutations in muscle proteins; thus understanding their basic function provides a mechanistic understanding of muscle disorders. In this review, we highlight the components of striated muscle with respect to their interactions, signaling pathways, functions, and connections to disease. © 2017 American Physiological Society. Compr Physiol 7:891-944, 2017.
Collapse
Affiliation(s)
- Christine A Henderson
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Christopher G Gomez
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Stefanie M Novak
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, Arizona, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
33
|
Hu LYR, Ackermann MA, Hecker PA, Prosser BL, King B, O’Connell KA, Grogan A, Meyer LC, Berndsen CE, Wright NT, Jonathan Lederer W, Kontrogianni-Konstantopoulos A. Deregulated Ca 2+ cycling underlies the development of arrhythmia and heart disease due to mutant obscurin. SCIENCE ADVANCES 2017; 3:e1603081. [PMID: 28630914 PMCID: PMC5462502 DOI: 10.1126/sciadv.1603081] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/17/2017] [Indexed: 05/05/2023]
Abstract
Obscurins are cytoskeletal proteins with structural and regulatory roles encoded by OBSCN. Mutations in OBSCN are associated with the development of hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Specifically, the R4344Q mutation present in immunoglobulin domain 58 (Ig58) was the first to be linked with the development of HCM. To assess the effects of R4344Q in vivo, we generated the respective knock-in mouse model. Mutant obscurins are expressed and incorporated normally into sarcomeres. The expression patterns of sarcomeric and Ca2+-cycling proteins are unaltered in sedentary 1-year-old knock-in myocardia, with the exception of sarco/endoplasmic reticulum Ca2+ adenosine triphosphatase 2 (SERCA2) and pentameric phospholamban whose levels are significantly increased and decreased, respectively. Isolated cardiomyocytes from 1-year-old knock-in hearts exhibit increased Ca2+-transients and Ca2+-load in the sarcoplasmic reticulum and faster contractility kinetics. Moreover, sedentary 1-year-old knock-in animals develop tachycardia accompanied by premature ventricular contractions, whereas 2-month-old knock-in animals subjected to pressure overload develop a DCM-like phenotype. Structural analysis revealed that the R4344Q mutation alters the distribution of electrostatic charges over the Ig58 surface, thus interfering with its binding capabilities. Consistent with this, wild-type Ig58 interacts with phospholamban modestly, and this interaction is markedly enhanced in the presence of R4344Q. Together, our studies demonstrate that under sedentary conditions, the R4344Q mutation results in Ca2+ deregulation and spontaneous arrhythmia, whereas in the presence of chronic, pathological stress, it leads to cardiac remodeling and dilation. We postulate that enhanced binding between mutant obscurins and phospholamban leads to SERCA2 disinhibition, which may underlie the observed pathological alterations.
Collapse
Affiliation(s)
- Li-Yen R. Hu
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Maegen A. Ackermann
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Peter A. Hecker
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Benjamin L. Prosser
- Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Physiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Brendan King
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Kelly A. O’Connell
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Alyssa Grogan
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Baltimore, MD 21201, USA
| | - Logan C. Meyer
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Christopher E. Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - Nathan T. Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, VA 22807, USA
| | - W. Jonathan Lederer
- Department of Physiology, Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | |
Collapse
|
34
|
Abstract
The inherited cardiomyopathies, hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) and left ventricular non-compaction (LVNC), have been frequently associated with mutations in sarcomeric proteins. In recent years, advances in DNA sequencing technology has allowed the study of the giant proteins of the sarcomere, such as titin and nebulin. Obscurin has been somewhat neglected in these studies, largely because its functional role is far from clear, although there was an isolated report in 2007 of obscurin mutations associated with HCM. Recently, whole exome sequencing methodology (WES) has been used to address mutations in OBSCN, the gene for obscurin, and OBSCN variants were found to be relatively common in inherited cardiomyopathies. In different studies, 5 OBSCN unique variants have been found in a group of 30 end-stage failing hearts, 6 OBSCN unique variants in 74 HCM cases and 3 OBSCN unique variants in 10 LVNC patients. As yet, the number of known potentially disease-causing OBSCN variants is quite small. The reason for this is that mutations in the OBSCN gene have not been recognised as potentially disease-causing until recently, and were not included in large-scale genetic surveys. OBSCN mutations may be causative of HCM, DCM and LVNC and other cardiomyopathies, or they may work in concert with other variants in the same or other genes to initiate the pathology. Currently, the function of obscurin is not well understood, but we anticipate that many more OBSCN variants linked to cardiomyopathy will be found when the large cohorts of patient sequences available are tested. It is to be hoped that the establishment of the importance of obscurin in pathology will stimulate a thorough investigation of obscurin function.
Collapse
|
35
|
Serie DJ, Crook JE, Necela BM, Axenfeld BC, Dockter TJ, Colon-Otero G, Perez EA, Thompson EA, Norton N. Breast Cancer Clinical Trial of Chemotherapy and Trastuzumab: Potential Tool to Identify Cardiac Modifying Variants of Dilated Cardiomyopathy. J Cardiovasc Dev Dis 2017; 4:jcdd4020006. [PMID: 29367538 PMCID: PMC5715703 DOI: 10.3390/jcdd4020006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin and the ERBB2 targeted therapy, trastuzumab, are routinely used in the treatment of HER2+ breast cancer. In mouse models, doxorubicin is known to cause cardiomyopathy and conditional cardiac knock out of Erbb2 results in dilated cardiomyopathy and increased sensitivity to doxorubicin-induced cell death. In humans, these drugs also result in cardiac phenotypes, but severity and reversibility is highly variable. We examined the association of decline in left ventricular ejection fraction (LVEF) at 15,204 single nucleotide polymorphisms (SNPs) spanning 72 cardiomyopathy genes, in 800 breast cancer patients who received doxorubicin and trastuzumab. For 7033 common SNPs (minor allele frequency (MAF) > 0.01) we performed single marker linear regression. For all SNPs, we performed gene-based testing with SNP-set (Sequence) Kernel Association Tests: SKAT, SKAT-O and SKAT-common/rare under rare variant non-burden; rare variant optimized burden and non-burden tests; and a combination of rare and common variants respectively. Single marker analyses identified seven missense variants in OBSCN (p = 0.0045-0.0009, MAF = 0.18-0.50) and two in TTN (both p = 0.04, MAF = 0.22). Gene-based rare variant analyses, SKAT and SKAT-O, performed very similarly (ILK, TCAP, DSC2, VCL, FXN, DSP and KCNQ1, p = 0.042-0.006). Gene-based tests of rare/common variants were significant at the nominal 5% level for OBSCN as well as TCAP, DSC2, VCL, NEXN, KCNJ2 and DMD (p = 0.044-0.008). Our results suggest that rare and common variants in OBSCN, as well as in other genes, could have modifying effects in cardiomyopathy.
Collapse
Affiliation(s)
- Daniel J Serie
- Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Julia E Crook
- Health Sciences Research, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Brian M Necela
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Bianca C Axenfeld
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Travis J Dockter
- Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA.
| | | | - Edith A Perez
- Hematology/Oncology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| | - Nadine Norton
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
36
|
Manring HR, Carter OA, Ackermann MA. Obscure functions: the location-function relationship of obscurins. Biophys Rev 2017; 9:245-258. [PMID: 28510116 DOI: 10.1007/s12551-017-0254-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 03/05/2017] [Indexed: 12/18/2022] Open
Abstract
The obscurin family of polypeptides is essential for normal striated muscle function and contributes to the pathogenesis of fatal diseases, including cardiomyopathies and cancers. The single mammalian obscurin gene, OBSCN, gives rise to giant (∼800 kDa) and smaller (∼40-500 kDa) proteins that are composed of tandem adhesion and signaling motifs. Mammalian obscurin proteins are expressed in a variety of cell types, including striated muscles, and localize to distinct subcellular compartments where they contribute to diverse cellular processes. Obscurin homologs in Caenorhabditis elegans and Drosophila possess a similar domain architecture and are also expressed in striated muscles. The long sought after question, "what does obscurin do?" is complex and cannot be addressed without taking into consideration the subcellular distribution of these proteins and local isoform concentration. Herein, we present an overview of the functions of obscurins and begin to define the intricate relationship between their subcellular distributions and functions in striated muscles.
Collapse
Affiliation(s)
- Heather R Manring
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Olivia A Carter
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA
| | - Maegen A Ackermann
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA. .,Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
37
|
Abstract
Striated cardiac and skeletal muscles play very different roles in the body, but they are similar at the molecular level. In particular, contraction, regardless of the type of muscle, is a precise and complex process involving the integral protein myofilaments and their associated regulatory components. The smallest functional unit of muscle contraction is the sarcomere. Within the sarcomere can be found a sophisticated ensemble of proteins associated with the thick filaments (myosin, myosin binding protein-C, titin, and obscurin) and thin myofilaments (actin, troponin, tropomyosin, nebulin, and nebulette). These parallel thick and thin filaments slide across one another, pulling the two ends of the sarcomere together to regulate contraction. More specifically, the regulation of both timing and force of contraction is accomplished through an intricate network of intra- and interfilament interactions belonging to each myofilament. This review introduces the sarcomere proteins involved in striated muscle contraction and places greater emphasis on the more recently identified and less well-characterized myofilaments: cardiac myosin binding protein-C, titin, nebulin, and obscurin. © 2017 American Physiological Society. Compr Physiol 7:675-692, 2017.
Collapse
Affiliation(s)
- Brian Leei Lin
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, Illinois, USA
| | - Taejeong Song
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, Illinois, USA.,Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Sakthivel Sadayappan
- Department of Cell and Molecular Physiology, Health Sciences Division, Loyola University Chicago, Maywood, Illinois, USA.,Department of Internal Medicine, Heart, Lung and Vascular Institute, Division of Cardiovascular Health and Sciences, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
38
|
Pernigo S, Fukuzawa A, Beedle AEM, Holt M, Round A, Pandini A, Garcia-Manyes S, Gautel M, Steiner RA. Binding of Myomesin to Obscurin-Like-1 at the Muscle M-Band Provides a Strategy for Isoform-Specific Mechanical Protection. Structure 2016; 25:107-120. [PMID: 27989621 PMCID: PMC5222588 DOI: 10.1016/j.str.2016.11.015] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/16/2016] [Accepted: 11/18/2016] [Indexed: 12/03/2022]
Abstract
The sarcomeric cytoskeleton is a network of modular proteins that integrate mechanical and signaling roles. Obscurin, or its homolog obscurin-like-1, bridges the giant ruler titin and the myosin crosslinker myomesin at the M-band. Yet, the molecular mechanisms underlying the physical obscurin(-like-1):myomesin connection, important for mechanical integrity of the M-band, remained elusive. Here, using a combination of structural, cellular, and single-molecule force spectroscopy techniques, we decode the architectural and functional determinants defining the obscurin(-like-1):myomesin complex. The crystal structure reveals a trans-complementation mechanism whereby an incomplete immunoglobulin-like domain assimilates an isoform-specific myomesin interdomain sequence. Crucially, this unconventional architecture provides mechanical stability up to forces of ∼135 pN. A cellular competition assay in neonatal rat cardiomyocytes validates the complex and provides the rationale for the isoform specificity of the interaction. Altogether, our results reveal a novel binding strategy in sarcomere assembly, which might have implications on muscle nanomechanics and overall M-band organization. The structure of the human obscurin-like-1:myomesin complex has been determined A myomesin sequence complements an immunoglobulin fold of obscurin-like-1 This binding mechanism provides mechanical stability up to forces of ∼135 pN Possible implications on muscle nanomechanics and M-band organization are discussed
Collapse
Affiliation(s)
- Stefano Pernigo
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK
| | - Atsushi Fukuzawa
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK
| | - Amy E M Beedle
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Physics, King's College London, London WC2R 2LS, UK
| | - Mark Holt
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK
| | - Adam Round
- European Molecular Biology Laboratory, Grenoble Outstation, 38042 Grenoble, France; School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, UK
| | - Alessandro Pandini
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Computer Science and Synthetic Biology Theme, Brunel University London, London UB8 3PH, UK
| | - Sergi Garcia-Manyes
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Department of Physics, King's College London, London WC2R 2LS, UK.
| | - Mathias Gautel
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK; Cardiovascular Division, King's College London BHF Centre of Research Excellence, London SE1 1UL, UK.
| | - Roberto A Steiner
- Randall Division of Cell and Molecular Biophysics, King's College London, London SE1 1UL, UK.
| |
Collapse
|
39
|
Rowland TJ, Graw SL, Sweet ME, Gigli M, Taylor MR, Mestroni L. Obscurin Variants in Patients With Left Ventricular Noncompaction. J Am Coll Cardiol 2016; 68:2237-2238. [PMID: 27855815 PMCID: PMC5896764 DOI: 10.1016/j.jacc.2016.08.052] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 08/04/2016] [Accepted: 08/09/2016] [Indexed: 10/20/2022]
Affiliation(s)
| | | | | | | | - Matthew R.G. Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, 12700 East 19th Avenue, F442, Aurora, Colorado 80045,
| | | |
Collapse
|
40
|
Manrai AK, Funke BH, Rehm HL, Olesen MS, Maron BA, Szolovits P, Margulies DM, Loscalzo J, Kohane IS. Genetic Misdiagnoses and the Potential for Health Disparities. N Engl J Med 2016; 375:655-65. [PMID: 27532831 PMCID: PMC5292722 DOI: 10.1056/nejmsa1507092] [Citation(s) in RCA: 544] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND For more than a decade, risk stratification for hypertrophic cardiomyopathy has been enhanced by targeted genetic testing. Using sequencing results, clinicians routinely assess the risk of hypertrophic cardiomyopathy in a patient's relatives and diagnose the condition in patients who have ambiguous clinical presentations. However, the benefits of genetic testing come with the risk that variants may be misclassified. METHODS Using publicly accessible exome data, we identified variants that have previously been considered causal in hypertrophic cardiomyopathy and that are overrepresented in the general population. We studied these variants in diverse populations and reevaluated their initial ascertainments in the medical literature. We reviewed patient records at a leading genetic-testing laboratory for occurrences of these variants during the near-decade-long history of the laboratory. RESULTS Multiple patients, all of whom were of African or unspecified ancestry, received positive reports, with variants misclassified as pathogenic on the basis of the understanding at the time of testing. Subsequently, all reported variants were recategorized as benign. The mutations that were most common in the general population were significantly more common among black Americans than among white Americans (P<0.001). Simulations showed that the inclusion of even small numbers of black Americans in control cohorts probably would have prevented these misclassifications. We identified methodologic shortcomings that contributed to these errors in the medical literature. CONCLUSIONS The misclassification of benign variants as pathogenic that we found in our study shows the need for sequencing the genomes of diverse populations, both in asymptomatic controls and the tested patient population. These results expand on current guidelines, which recommend the use of ancestry-matched controls to interpret variants. As additional populations of different ancestry backgrounds are sequenced, we expect variant reclassifications to increase, particularly for ancestry groups that have historically been less well studied. (Funded by the National Institutes of Health.).
Collapse
Affiliation(s)
- Arjun K Manrai
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Birgit H Funke
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Heidi L Rehm
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Morten S Olesen
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Bradley A Maron
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Peter Szolovits
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - David M Margulies
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Joseph Loscalzo
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| | - Isaac S Kohane
- From the Departments of Biomedical Informatics (A.K.M., D.M.M., I.S.K.), Pathology (B.H.F.), and Medicine (B.A.M., J.L.), Harvard Medical School, the Departments of Pathology, Massachusetts General Hospital (B.H.F.), and the Department of Pathology (H.L.R.), Division of Cardiovascular Medicine (B.A.M.), and Department of Medicine (B.A.M., J.L.), Brigham and Women's Hospital, Boston, and the Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology (MIT) (A.K.M., I.S.K.), the Laboratory for Molecular Medicine, Partners HealthCare Personalized Medicine (B.H.F., H.L.R.), and the Computer Science and Artificial Intelligence Laboratory, MIT (P.S.), Cambridge - all in Massachusetts; and the Laboratory of Molecular Cardiology, Department of Cardiology, the Heart Center, University Hospital of Copenhagen, Rigshospitalet, and the Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen (M.S.O.) - both in Copenhagen
| |
Collapse
|
41
|
In-Utero Low-Dose Irradiation Leads to Persistent Alterations in the Mouse Heart Proteome. PLoS One 2016; 11:e0156952. [PMID: 27276052 PMCID: PMC4898684 DOI: 10.1371/journal.pone.0156952] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 05/23/2016] [Indexed: 02/07/2023] Open
Abstract
Prenatal exposure to stress such as increased level of reactive oxygen species or antiviral therapy are known factors leading to adult heart defects. The risks following a radiation exposure during fetal period are unknown, as are the mechanisms of any potential cardiac damage. The aim of this study was to gather evidence for possible damage by investigating long-term changes in the mouse heart proteome after prenatal exposure to low and moderate radiation doses. Pregnant C57Bl/6J mice received on embryonic day 11 (E11) a single total body dose of ionizing radiation that ranged from 0.02 Gy to 1.0 Gy. The offspring were sacrificed at the age of 6 months or 2 years. Quantitative proteomic analysis of heart tissue was performed using Isotope Coded Protein Label technology and tandem mass spectrometry. The proteomics data were analyzed by bioinformatics and key changes were validated by immunoblotting. Persistent changes were observed in the expression of proteins representing mitochondrial respiratory complexes, redox and heat shock response, and the cytoskeleton, even at the low dose of 0.1 Gy. The level of total and active form of the kinase MAP4K4 that is essential for the embryonic development of mouse heart was persistently decreased at the radiation dose of 1.0 Gy. This study provides the first insight into the molecular mechanisms of cardiac impairment induced by ionizing radiation exposure during the prenatal period.
Collapse
|
42
|
Bang ML. Animal Models of Congenital Cardiomyopathies Associated With Mutations in Z-Line Proteins. J Cell Physiol 2016; 232:38-52. [PMID: 27171814 DOI: 10.1002/jcp.25424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/10/2016] [Indexed: 01/15/2023]
Abstract
The cardiac Z-line at the boundary between sarcomeres is a multiprotein complex connecting the contractile apparatus with the cytoskeleton and the extracellular matrix. The Z-line is important for efficient force generation and transmission as well as the maintenance of structural stability and integrity. Furthermore, it is a nodal point for intracellular signaling, in particular mechanosensing and mechanotransduction. Mutations in various genes encoding Z-line proteins have been associated with different cardiomyopathies, including dilated cardiomyopathy, hypertrophic cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, restrictive cardiomyopathy, and left ventricular noncompaction, and mutations even within the same gene can cause widely different pathologies. Animal models have contributed to a great advancement in the understanding of the physiological function of Z-line proteins and the pathways leading from mutations in Z-line proteins to cardiomyopathy, although genotype-phenotype prediction remains a great challenge. This review presents an overview of the currently available animal models for Z-line and Z-line associated proteins involved in human cardiomyopathies with special emphasis on knock-in and transgenic mouse models recapitulating the clinical phenotypes of human cardiomyopathy patients carrying mutations in Z-line proteins. Pros and cons of mouse models will be discussed and a future outlook will be given. J. Cell. Physiol. 232: 38-52, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research, UOS Milan, National Research Council and Humanitas Clinical and Research Center, Rozzano, Milan, Italy.
| |
Collapse
|
43
|
Kawano H, Kawamura K, Ishijima M, Hayashi T, Abe K, Kawai K, Maemura K. Myocardial fragmentation associated with disruption of the Z-band in hypertrophic cardiomyopathy in Noonan syndrome. Cardiovasc Pathol 2016; 25:329-332. [PMID: 27216919 DOI: 10.1016/j.carpath.2016.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 11/25/2022] Open
Abstract
A 13-year-old female with Noonan syndrome had been diagnosed with hypertrophic cardiomyopathy, and she died of heart failure at the age of 25 years. Light microscopic and electron microscopic examination of her biopsied myocardium and autopsy heart showed myocardial fragmentation associated with Z-band disruption as well as myocardial hypertrophy and disarray with interstitial fibrosis. Myocardial fragmentation associated with Z-band disruption may be related to the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Hiroaki Kawano
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan.
| | - Koichi Kawamura
- Faculty of Science and Engineering, Waseda University, Tokyo, Japan
| | - Mitsuaki Ishijima
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Tomayoshi Hayashi
- Department of Pathology, Shimabara Prefectural Hospital, Shimabara, Japan
| | - Kuniko Abe
- Department of Pathology, Nagasaki University Hospital, Nagasaki, Japan
| | - Kioko Kawai
- Nagasaki Diagnostic Pathology Clinic, Nagasaki, Japan
| | - Koji Maemura
- Department of Cardiovascular Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| |
Collapse
|
44
|
Investigation of Pathogenic Genes in Chinese sporadic Hypertrophic Cardiomyopathy Patients by Whole Exome Sequencing. Sci Rep 2015; 5:16609. [PMID: 26573135 PMCID: PMC4647833 DOI: 10.1038/srep16609] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/16/2015] [Indexed: 11/08/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a cardiovascular disease with high heterogeneity. Limited knowledge concerning the genetic background of nearly 40% HCM cases indicates there is a clear need for further investigation to explore the genetic pathogenesis of the disease. In this study, we undertook a whole exome sequencing (WES) approach to identify novel candidate genes and mutations associated with HCM. The cohort consisted of 74 unrelated patients with sporadic HCM (sHCM) previously determined to be negative for mutations in eight sarcomere genes. The results showed that 7 of 74 patients (9.5%) had damaging mutations in 43 known HCM disease genes. Furthermore, after analysis combining the Transmission and De novo Association (TADA) program and the ToppGene program, 10 putative genes gained priority. A thorough review of public databases and related literature revealed that there is strong supporting evidence for most of the genes playing roles in various aspects of heart development. Findings from recent studies suggest that the putative and known disease genes converge on three functional pathways: sarcomere function, calcium signaling and metabolism pathway. This study illustrates the benefit of WES, in combination with rare variant analysis tools, in providing valuable insight into the genetic etiology of a heterogeneous sporadic disease.
Collapse
|
45
|
Marston S, Montgiraud C, Munster AB, Copeland O, Choi O, dos Remedios C, Messer AE, Ehler E, Knöll R. OBSCN Mutations Associated with Dilated Cardiomyopathy and Haploinsufficiency. PLoS One 2015; 10:e0138568. [PMID: 26406308 PMCID: PMC4583186 DOI: 10.1371/journal.pone.0138568] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/01/2015] [Indexed: 11/18/2022] Open
Abstract
Background Studies of the functional consequences of DCM-causing mutations have been limited to a few cases where patients with known mutations had heart transplants. To increase the number of potential tissue samples for direct investigation we performed whole exon sequencing of explanted heart muscle samples from 30 patients that had a diagnosis of familial dilated cardiomyopathy and screened for potentially disease-causing mutations in 58 HCM or DCM-related genes. Results We identified 5 potentially disease-causing OBSCN mutations in 4 samples; one sample had two OBSCN mutations and one mutation was judged to be not disease-related. Also identified were 6 truncating mutations in TTN, 3 mutations in MYH7, 2 in DSP and one each in TNNC1, TNNI3, MYOM1, VCL, GLA, PLB, TCAP, PKP2 and LAMA4. The mean level of obscurin mRNA was significantly greater and more variable in healthy donor samples than the DCM samples but did not correlate with OBSCN mutations. A single obscurin protein band was observed in human heart myofibrils with apparent mass 960 ± 60 kDa. The three samples with OBSCN mutations had significantly lower levels of obscurin immunoreactive material than DCM samples without OBSCN mutations (45±7, 48±3, and 72±6% of control level).Obscurin levels in DCM controls, donor heart and myectomy samples were the same. Conclusions OBSCN mutations may result in the development of a DCM phenotype via haploinsufficiency. Mutations in the obscurin gene should be considered as a significant causal factor of DCM, alone or in concert with other mutations.
Collapse
Affiliation(s)
- Steven Marston
- NHLI, Imperial College London, London, United Kingdom
- * E-mail:
| | | | | | | | - Onjee Choi
- NHLI, Imperial College London, London, United Kingdom
| | | | | | - Elisabeth Ehler
- Randall Division, King’s College London, London, United Kingdom
| | - Ralph Knöll
- NHLI, Imperial College London, London, United Kingdom
| |
Collapse
|
46
|
Kimura A. Molecular genetics and pathogenesis of cardiomyopathy. J Hum Genet 2015; 61:41-50. [PMID: 26178429 DOI: 10.1038/jhg.2015.83] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyopathy is defined as a disease of functional impairment in the cardiac muscle and its etiology includes both extrinsic and intrinsic factors. Cardiomyopathy caused by the intrinsic factors is called as primary cardiomyopathy of which two major clinical phenotypes are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Genetic approaches have revealed the disease genes for hereditary primary cardiomyopathy and functional studies have demonstrated that characteristic functional alterations induced by the disease-associated mutations are closely related to the clinical types, such that increased and decreased Ca(2+) sensitivities of muscle contraction are associated with HCM and DCM, respectively. In addition, recent studies have suggested that mutations in the Z-disc components found in HCM and DCM may result in increased and decreased stiffness of sarcomere, respectively. Moreover, functional analysis of mutations in the other components of cardiac muscle have suggested that the altered response to metabolic stresses is associated with cardiomyopathy, further indicating the heterogeneity in the etiology and pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
47
|
The sarcomeric M-region: a molecular command center for diverse cellular processes. BIOMED RESEARCH INTERNATIONAL 2015; 2015:714197. [PMID: 25961035 PMCID: PMC4413555 DOI: 10.1155/2015/714197] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/08/2015] [Indexed: 02/07/2023]
Abstract
The sarcomeric M-region anchors thick filaments and withstands the mechanical stress of contractions by deformation, thus enabling distribution of physiological forces along the length of thick filaments. While the role of the M-region in supporting myofibrillar structure and contractility is well established, its role in mediating additional cellular processes has only recently started to emerge. As such, M-region is the hub of key protein players contributing to cytoskeletal remodeling, signal transduction, mechanosensing, metabolism, and proteasomal degradation. Mutations in genes encoding M-region related proteins lead to development of severe and lethal cardiac and skeletal myopathies affecting mankind. Herein, we describe the main cellular processes taking place at the M-region, other than thick filament assembly, and discuss human myopathies associated with mutant or truncated M-region proteins.
Collapse
|
48
|
Pernigo S, Fukuzawa A, Pandini A, Holt M, Kleinjung J, Gautel M, Steiner RA. The Crystal Structure of the Human Titin:Obscurin Complex Reveals a Conserved yet Specific Muscle M-Band Zipper Module. J Mol Biol 2015; 427:718-736. [DOI: 10.1016/j.jmb.2014.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 11/15/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
|
49
|
Abstract
The giant sarcomeric protein titin is a key determinant of myocardial passive stiffness and stress-sensitive signaling. Titin stiffness is modulated by isoform variation, phosphorylation by protein kinases, and, possibly, oxidative stress through disulfide bond formation. Titin has also emerged as an important human disease gene. Early studies in patients with dilated cardiomyopathy (DCM) revealed shifts toward more compliant isoforms, an adaptation that offsets increases in passive stiffness based on the extracellular matrix. Similar shifts are observed in heart failure with preserved ejection fraction. In contrast, hypophosphorylation of PKA/G sites contributes to a net increase in cardiomyocyte resting tension in heart failure with preserved ejection fraction. More recently, titin mutations have been recognized as the most common etiology of inherited DCM. In addition, some DCM-causing mutations affect RBM20, a titin splice factor. Titin mutations are a rare cause of hypertrophic cardiomyopathy and also underlie some cases of arrhythmogenic right ventricular dysplasia. Finally, mutations of genes encoding proteins that interact with and/or bind to titin are responsible for both DCM and hypertrophic cardiomyopathy. Targeting titin as a therapeutic strategy is in its infancy, but it could potentially involve manipulation of isoforms, posttranslational modifications, and upregulation of normal protein in patients with disease-causing mutations.
Collapse
|
50
|
Obscurins: Goliaths and Davids take over non-muscle tissues. PLoS One 2014; 9:e88162. [PMID: 24516603 PMCID: PMC3916441 DOI: 10.1371/journal.pone.0088162] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/03/2014] [Indexed: 12/11/2022] Open
Abstract
Obscurins comprise a family of proteins originally identified in striated muscles, where they play essential roles in myofibrillogenesis, cytoskeletal organization, and Ca2+ homeostasis. They are encoded by the single OBSCN gene, and are composed of tandem adhesion domains and signaling motifs. To date, two giant obscurin isoforms have been described in detail that differ only at the extreme COOH-terminus; while obscurin-A (∼720 kDa) contains a non-modular COOH-terminus that harbors binding sites for the adaptor proteins ankyrins, obscurin-B (∼870 kDa) contains two COOH-terminal serine-threonine kinase domains preceded by adhesion motifs. Besides the two known giant obscurins, a thorough search of transcript databases suggests that complex alternative splicing of the obscurin transcript results in the generation of additional giant as well as small isoforms with molecular masses ranging between ∼50–970 kDa. These novel isoforms share common domains with the characterized isoforms, but also contain unique regions. Using a panel of highly specific antibodies directed against epitopes spanning the entire length of giant obscurins, we employed western blotting and immunohistochemistry to perform a systematic and comprehensive characterization of the expression profile of obscurins in muscle and non-muscle tissues. Our studies demonstrate for the first time that obscurins are not restricted to striated muscles, but are abundantly expressed in several tissues and organs including brain, skin, kidney, liver, spleen, and lung. While some obscurin isoforms are ubiquitously expressed, others are preferentially present in specific tissues and organs. Moreover, obscurins are present in select structures and cell types where they assume nuclear, cytosolic, and membrane distributions. Given the ubiquitous expression of some obscurins, along with the preferential expression of others, it becomes apparent that obscurins may play common and unique roles, respectively, in the regulation and maintenance of cell homeostasis in various tissues and organs throughout the body.
Collapse
|