1
|
Holtzhausen C, Heil L, Klingel K, Fox H, Gummert J, Gärtner A, Schmidt A, Krüger M, Kirfel G, van der Ven PFM, Milting H, Clemen CS, Schröder R, Fürst DO, Tiesmeier J. Sudden cardiac death, arrhythmogenic cardiomyopathy and intercalated disc pathology due to reduced filamin C protein levels: a matter of life and death. Hum Mol Genet 2025; 34:726-738. [PMID: 39895064 DOI: 10.1093/hmg/ddaf014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
Mutations in the human FLNC gene encoding filamin C (FLNc) cause a broad spectrum of sporadic and familial cardiomyopathies and myopathies. We report on the genetic, clinical, morphological and biochemical findings in a German family harboring an FLNC variant that leads to severe cardiac disease comprising sudden cardiac death and arrhythmogenic cardiomyopathy. Genetic analysis identified a novel heterozygous FLNC variant in exon 16 (NM_001458.4:c.2495_2498delAGTA, het; p.K832TfsX45) in i) the index patient suffering from dilated cardiomyopathy necessitating heart transplantation, ii) a son, who died from sudden cardiac death, iii) a second son, who survived an episode of sudden cardiac arrest and iv) a third son affected by isolated skeletal muscle myopathy. FLNc protein levels were markedly reduced in cardiac tissue obtained from the index patient, implying that the p.K832TfsX45 FLNc variant most probably caused nonsense-mediated decay of the corresponding mRNA. Morphological analysis of the diseased cardiac tissue revealed extensive fibrotic remodeling, and marked degenerative changes of the contractile apparatus of cardiomyocytes and severe structural alterations of intercalated discs. Connexin-43 signal intensity at intercalated discs was diminished and FLNc labelling of myofibrils was attenuated or even absent. Proteome analyses demonstrated complex alterations of extracellular matrix and intercalated disc proteins. Our findings demonstrate that this novel, truncating FLNC mutation likely leads to haploinsufficiency, thereby causing a deleterious sequence of degenerative changes of cardiac tissue with extensive fibrotic remodeling and intercalated disc pathology as the structural basis for FLNC-related cardiomyopathy with life-threatening cardiac arrhythmias.
Collapse
MESH Headings
- Female
- Humans
- Male
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/pathology
- Arrhythmogenic Right Ventricular Dysplasia/genetics
- Arrhythmogenic Right Ventricular Dysplasia/pathology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/pathology
- Connexin 43/metabolism
- Connexin 43/genetics
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/pathology
- Filamins/genetics
- Filamins/metabolism
- Mutation
- Myocardium/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Pedigree
Collapse
Affiliation(s)
- Christian Holtzhausen
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Lorena Heil
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076 Tübingen, Germany
| | - Henrik Fox
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Jan Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Andreas Schmidt
- Center for Molecular Medicine (CMMC), Medical Faculty, and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50937 Cologne, Germany
| | - Marcus Krüger
- Center for Molecular Medicine (CMMC), Medical Faculty, and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50937 Cologne, Germany
| | - Gregor Kirfel
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Höhe, 51147 Cologne, Germany
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Jens Tiesmeier
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
- Institute for Anesthesiology, Intensive Care- and Emergency Medicine, MLK-Hospital, Voedestr. 79, Luebbecke, Campus OWL, Ruhr-University Bochum, 32312 Lübbecke, Germany
| |
Collapse
|
2
|
Wang X, Lang Z, Yan Z, Xu J, Zhang J, Jiao L, Zhang H. Dilated cardiomyopathy: from genes and molecules to potential treatments. Mol Cell Biochem 2025:10.1007/s11010-025-05269-0. [PMID: 40155570 DOI: 10.1007/s11010-025-05269-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Dilated cardiomyopathy is a myocardial condition marked by the enlargement of the heart's ventricular chambers and the gradual decline in systolic function, frequently resulting in congestive heart failure. Dilated cardiomyopathy has obvious familial characteristics, and mutations in related pathogenic genes can account for about 50% of patients with dilated cardiomyopathy. The most common genes related to dilated cardiomyopathy include TTN, LMNA, MYH7, etc. With more and more research on these genes, it will undoubtedly provide more potential targets and therapeutic pathways for the treatment of dilated cardiomyopathy. In addition, myocardial inflammation, myocardial metabolism abnormalities and cardiomyocyte apoptosis all have an important impact on the pathogenesis of dilated cardiomyopathy. Approximately half of sudden deaths among children and adolescents, along with the majority of patients undergoing heart transplantation, stem from cardiomyopathy. Therefore, precise and prompt clinical diagnosis holds paramount importance. Currently, diagnosis primarily hinges on the patient's medical background and imaging tests, with the significance of genetic testing steadily gaining prominence. The primary treatment for dilated cardiomyopathy remains heart transplantation. However, the scarcity of donors and the risk of severe immune rejection underscore the pressing need for novel therapies. Presently, research is actively exploring preclinical treatments like stem cell therapy as potential solutions.
Collapse
Affiliation(s)
- Xiumei Wang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zekun Lang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Zeyi Yan
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Jinyuan Zhang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Lianhang Jiao
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China
| | - Haijun Zhang
- Department of Anesthesiology and Operating Theater, The First Hospital of Lanzhou University, Lanzhou, 730000, The People's Republic of China.
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, Gansu, The People's Republic of China.
| |
Collapse
|
3
|
Kim JH, Baggish AL, Levine BD, Ackerman MJ, Day SM, Dineen EH, Guseh JS, La Gerche A, Lampert R, Martinez MW, Papadakis M, Phelan DM, Shafer KM. Clinical Considerations for Competitive Sports Participation for Athletes With Cardiovascular Abnormalities: A Scientific Statement From the American Heart Association and American College of Cardiology. Circulation 2025; 151:e716-e761. [PMID: 39973614 DOI: 10.1161/cir.0000000000001297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
COLLABORATORS Larry A. Allen, MD, MHS, FAHA, FACC; Mats Börjesson, MD, PhD, FACC; Alan C. Braverman, MD, FACC; Julie A. Brothers, MD; Silvia Castelletti, MD, MSc, FESC; Eugene H. Chung, MD, MPH, FHRS, FAHA, FACC; Timothy W. Churchill, MD, FACC; Guido Claessen, MD, PhD; Flavio D'Ascenzi, MD, PhD; Douglas Darden, MD; Peter N. Dean, MD, FACC; Neal W. Dickert, MD, PhD, FACC; Jonathan A. Drezner, MD; Katherine E. Economy, MD, MPH; Thijs M.H. Eijsvogels, PhD; Michael S. Emery, MD, MS, FACC; Susan P. Etheridge, MD, FHRS, FAHA, FACC; Sabiha Gati, BSc (Hons), MBBS, PhD, MRCP, FESC; Belinda Gray, BSc (Med), MBBS, PhD; Martin Halle, MD; Kimberly G. Harmon, MD; Jeffrey J. Hsu, MD, PhD, FAHA, FACC; Richard J. Kovacs, MD, FAHA, MACC; Sheela Krishnan, MD, FACC; Mark S. Link, MD, FHRS, FAHA, FACC; Martin Maron, MD; Silvana Molossi, MD, PhD, FACC; Antonio Pelliccia, MD; Jack C. Salerno, MD, FACC, FHRS; Ankit B. Shah, MD, MPH, FACC; Sanjay Sharma, BSc (Hons), MBChB, MRCP (UK), MD; Tamanna K. Singh, MD, FACC; Katie M. Stewart, NP, MS; Paul D. Thompson, MD, FAHA, FACC; Meagan M. Wasfy, MD, MPH, FACC; Matthias Wilhelm, MD. This American Heart Association/American College of Cardiology scientific statement on clinical considerations for competitive sports participation for athletes with cardiovascular abnormalities or diseases is organized into 11 distinct sections focused on sports-specific topics or disease processes that are relevant when considering the potential risks of adverse cardiovascular events, including sudden cardiac arrest, during competitive sports participation. Task forces comprising international experts in sports cardiology and the respective topics covered were assigned to each section and prepared specific clinical considerations tables for practitioners to reference. Comprehensive literature review and an emphasis on shared decision-making were integral in the writing of all clinical considerations presented.
Collapse
|
4
|
Kim JH, Baggish AL, Levine BD, Ackerman MJ, Day SM, Dineen EH, Guseh Ii JS, La Gerche A, Lampert R, Martinez MW, Papadakis M, Phelan DM, Shafer KM, Allen LA, Börjesson M, Braverman AC, Brothers JA, Castelletti S, Chung EH, Churchill TW, Claessen G, D'Ascenzi F, Darden D, Dean PN, Dickert NW, Drezner JA, Economy KE, Eijsvogels TMH, Emery MS, Etheridge SP, Gati S, Gray B, Halle M, Harmon KG, Hsu JJ, Kovacs RJ, Krishnan S, Link MS, Maron M, Molossi S, Pelliccia A, Salerno JC, Shah AB, Sharma S, Singh TK, Stewart KM, Thompson PD, Wasfy MM, Wilhelm M. Clinical Considerations for Competitive Sports Participation for Athletes With Cardiovascular Abnormalities: A Scientific Statement From the American Heart Association and American College of Cardiology. J Am Coll Cardiol 2025; 85:1059-1108. [PMID: 39976316 DOI: 10.1016/j.jacc.2024.12.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
This American Heart Association/American College of Cardiology scientific statement on clinical considerations for competitive sports participation for athletes with cardiovascular abnormalities or diseases is organized into 11 distinct sections focused on sports-specific topics or disease processes that are relevant when considering the potential risks of adverse cardiovascular events, including sudden cardiac arrest, during competitive sports participation. Task forces comprising international experts in sports cardiology and the respective topics covered were assigned to each section and prepared specific clinical considerations tables for practitioners to reference. Comprehensive literature review and an emphasis on shared decision-making were integral in the writing of all clinical considerations presented.
Collapse
|
5
|
Horgan S, Kotwal H, Malan A, Sekhri N, Lopes LR. Reassessment and reclassification of variants of unknown significance in patients with cardiomyopathy in a specialist department. J Med Genet 2025; 62:185-190. [PMID: 39694818 DOI: 10.1136/jmg-2024-110208] [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: 06/28/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The utility of diagnostic genetic testing in cardiomyopathy has grown significantly, due to the discovery of novel genes and greater awareness among healthcare professionals. However, a substantial proportion of cases (around 50%) yield no causative genetic variants or have variants of unknown significance (VUS), limiting their use in clinical management and familial screening. The increase in data quantity and quality in reference databases, coupled with variant interpretation guidelines, allows for periodic reanalysis of VUS, potentially reducing diagnostic gaps. METHODS This study presents a review of VUS results identified in hypertrophic cardiomyopathy (HCM), dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy (ACM) probands over a 5-year period, using American College of Medical Genetics and Genomics criteria. A total of 248 VUS from 233 reports were reviewed, with the majority of patients with a diagnosis of HCM (n=112), followed by DCM (n=99) and ACM (n=22). RESULTS Four (1.6%) VUS showed sufficient evidence to upgrade to likely pathogenic/pathogenic status, while 8 (3.2%) were downgraded to benign. The majority 236 (95.2%) remained VUS after reanalysis, of which 12 (4.7%) had potential to reclassification to benign or likely pathogenic/pathogenic depending on further data. CONCLUSION The study emphasises the importance of periodic re-evaluation of VUS results for clinical management of probands as well as cascade testing. We show feasibility of conducting reclassification analysis in a referral centre, but highlight the need for ongoing collaboration between clinical and laboratory experts. Our work supports the current recommendation of reclassification every 3-5 years to keep pace with evolving evidence.
Collapse
Affiliation(s)
| | | | | | - Neha Sekhri
- ICVD, St Bartholomew's Hospital, London, UK
- Queen Mary University of London, London, UK
| | - Luis R Lopes
- ICVD, St Bartholomew's Hospital, London, UK
- University College London Institute of Cardiovascular Science, London, UK
| |
Collapse
|
6
|
Ilchuk LA, Kochegarova KK, Baikova IP, Safonova PD, Bruter AV, Kubekina MV, Okulova YD, Minkovskaya TE, Kuznetsova NA, Dolmatova DM, Ryabinina AY, Mozhaev AA, Belousov VV, Ershov BP, Timashev PS, Filatov MA, Silaeva YY. Mutations in Filamin C Associated with Both Alleles Do Not Affect the Functioning of Mice Cardiac Muscles. Int J Mol Sci 2025; 26:1409. [PMID: 40003875 PMCID: PMC11855563 DOI: 10.3390/ijms26041409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Filamin C (FLNC) is a structural protein of muscle fibers. Mutations in the FLNC gene are known to cause myopathies and cardiomyopathies in humans. Here we report the generation by a CRISPR/Cas9 editing system injected into zygote pronuclei of two mouse strains carrying filamin C mutations-one of them (AGA) has a deletion of three nucleotides at position c.7418_7420, causing E>>D substitution and N deletion at positions 2472 and 2473, respectively. The other strain carries a deletion of GA nucleotides at position c.7419_7420, leading to a frameshift and a premature stop codon. Homozygous animals (FlncAGA/AGA and FlncGA/GA) were embryonically lethal. We determined that FlncGA/GA embryos died prior to the E12.5 stage and illustrated delayed development after the E9.5 stage. We performed histological analysis of heart tissue and skeletal muscles of heterozygous strains carrying mutations in different combinations (FlncGA/wt, FlncAGA/wt, and FlncGA/AGA). By performing physiological tests (grip strength and endurance tests), we have shown that heterozygous animals of both strains (FlncGA/wt, FlncAGA/wt) are functionally indistinguishable from wild-type animals. Interestingly, compound heterozygous mice (FlncGA/AGA) are viable, develop normally, reach puberty and it was verified by ECG and Eco-CG that their cardiac muscle is functionally normal. Intriguingly, FlncGA/AGA mice demonstrated better results in the grip strength physiological test in comparison to WT animals. We also propose a structural model that explains the complementary interaction of two mutant variants of filamin C.
Collapse
Affiliation(s)
- Leonid A. Ilchuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Ksenia K. Kochegarova
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; (K.K.K.); (B.P.E.); (P.S.T.)
| | - Iuliia P. Baikova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
| | - Polina D. Safonova
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Alexandra V. Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Marina V. Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Yulia D. Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Tatiana E. Minkovskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
| | - Nadezhda A. Kuznetsova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Daria M. Dolmatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Anna Yu. Ryabinina
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia;
- Laboratory of Molecular Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.M.); (V.V.B.)
| | - Andrey A. Mozhaev
- Laboratory of Molecular Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.M.); (V.V.B.)
- Group of Genome Editing Techniques, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
| | - Vsevolod V. Belousov
- Laboratory of Molecular Technologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (A.A.M.); (V.V.B.)
- Group of Genome Editing Techniques, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 117513 Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, 117513 Moscow, Russia
| | - Boris P. Ershov
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; (K.K.K.); (B.P.E.); (P.S.T.)
| | - Peter S. Timashev
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; (K.K.K.); (B.P.E.); (P.S.T.)
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Maxim A. Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia; (K.K.K.); (B.P.E.); (P.S.T.)
| | - Yulia Yu. Silaeva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia; (L.A.I.); (I.P.B.); (A.V.B.); (M.V.K.); (Y.D.O.); (T.E.M.); (N.A.K.); (D.M.D.); (Y.Y.S.)
- Core Facility Center, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia;
| |
Collapse
|
7
|
Hiruma T, Inoue S, Dai Z, Nomura S, Kubo T, Sugiura K, Suzuki A, Kashimura T, Matsushima S, Yamada T, Tobita T, Katoh M, Ko T, Ito M, Ishida J, Amiya E, Hatano M, Takeda N, Takimoto E, Akazawa H, Morita H, Yamaguchi J, Inomata T, Tsutsui H, Kitaoka H, Aburatani H, Takeda N, Komuro I. Association of Multiple Nonhypertrophic Cardiomyopathy-Related Genetic Variants and Outcomes in Patients With Hypertrophic Cardiomyopathy. JACC. HEART FAILURE 2024; 12:2041-2052. [PMID: 39340495 DOI: 10.1016/j.jchf.2024.08.005] [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: 07/24/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND Approximately 10% of hypertrophic cardiomyopathy (HCM) patients have left ventricular systolic dysfunction (end-stage HCM) leading to severe heart-failure; however, risk stratification to identify patients at risk of progressing to end-stage HCM remains insufficient. OBJECTIVES In this study, the authors sought to elucidate whether the coexistence of other cardiovascular disease (CVD)-related variants is associated with progression to end-stage HCM in patients with HCM harboring pathogenic or likely pathogenic (P/LP) sarcomeric variants. METHODS The authors performed genetic analysis of 83 CVD-related genes in HCM patients from a Japanese multicenter cohort. P/LP variants in 8 major sarcomeric genes (MYBPC3, MYH7, TNNT2, TNNI3, TPM1, MYL2, MYL3, and ACTC1) definitive for HCM were defined as "sarcomeric variants." In addition, P/LP variants associated with other CVDs, such as dilated cardiomyopathy and arrhythmogenic cardiomyopathy, were referred to as "other CVD-related variants." RESULTS Among 394 HCM patients, 139 carried P/LP sarcomeric variants: 11 (7.9%) carried other CVD-related variants, 6 (4.3%) multiple sarcomeric variants, and 122 (87.8%) single sarcomeric variants. In a multivariable Cox regression analysis, presence of multiple sarcomeric variants (adjusted HR [aHR]: 3.35 [95% CI: 1.25-8.95]; P = 0.016) and coexistence of other CVD-related variants (aHR: 2.80 [95% CI: 1.16-6.78]; P = 0.022) were independently associated with progression to end-stage HCM. Coexisting other CVD-related variants were also associated with heart failure events (aHR: 2.75 [95% CI: 1.27-5.94]; P = 0.010). CONCLUSIONS Approximately 8% of sarcomeric HCM patients carried other CVD-related variants, which were associated with progression to end-stage HCM and heart failure events. Comprehensive surveillance of CVD-related variants within sarcomeric HCM patients contributes to risk stratification and understanding of mechanisms underlying end-stage HCM.
Collapse
Affiliation(s)
- Takashi Hiruma
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Inoue
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Zhehao Dai
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan.
| | - Toru Kubo
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University, Kochi, Japan
| | - Kenta Sugiura
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University, Kochi, Japan
| | - Atsushi Suzuki
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takeshi Kashimura
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanobu Yamada
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashige Tobita
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Manami Katoh
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Toshiyuki Ko
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Masamichi Ito
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Ishida
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eisuke Amiya
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaru Hatano
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norifumi Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Eiki Takimoto
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Junichi Yamaguchi
- Department of Cardiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Takayuki Inomata
- Department of Cardiovascular Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan; School of Medicine and Graduate School, International University of Health and Welfare, Okawa, Japan
| | - Hiroaki Kitaoka
- Department of Cardiology and Geriatrics, Kochi Medical School, Kochi University, Kochi, Japan
| | - Hiroyuki Aburatani
- Genome Science Laboratory, Research Center for Advanced Science and Technologies, The University of Tokyo, Tokyo, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Frontier Cardiovascular Science, The University of Tokyo Graduate School of Medicine, Tokyo, Japan; International University of Health and Welfare, Tokyo, Japan.
| |
Collapse
|
8
|
Dong J, Zhang W, Chen Q, Zha L. Identification of a Missense Mutation in the FLNC Gene from a Chinese Family with Restrictive Cardiomyopathy. J Multidiscip Healthc 2024; 17:5363-5373. [PMID: 39582878 PMCID: PMC11585995 DOI: 10.2147/jmdh.s494831] [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: 10/07/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
Objective Restrictive cardiomyopathy (RCM) is a heterogenous cardiomyopathy with various causes, and genetic variants take an important part of the pathogenesis. Whole-exome sequencing (WES) is effective to discover genes that cause genetic diseases. By using WES, we attempted to identify the genetic cause of an RCM family and clarify the clinical diagnosis of the patient and then provide a personalized treatment plan. Materials and Methods Blood samples were obtained from the proband and his healthy parents. WES and Sanger sequencing were performed to identify the possible pathogenic gene. Co-segregation analysis was conducted for candidate variants, and the allele frequency was checked in databases including Ensembl, Exome Aggregation Consortium (ExAC) and Human Gene Mutation Database (HGMD). Furthermore, the potential effect of variant was predicted using various-free software such as SIFT, Polyphen-2 and Mutation Taster and the conservation was tested using multiple sequence alignments by ClustalX. Results The proband was a 20 years old boy with severe heart failure symptoms including dyspnea, massive ascites, edema of both lower limbs and chest congestion. Echocardiography showed significant biatrial enlargement, normal left ventricular wall thickness and preserved systolic function of both ventricles. A missense mutation in FLNC (c.6451G>A, p.G2151S), encoded filamin-C was detected in proband by WES and Sanger sequencing, while it was not be found in his parents, we supposed that the FLNC mutation (c.6451G>A, p.G2151S) may be a de-novo mutation. Through multiple functional predictions, we found that it is a deleterious mutation and the mutation in filamin-C could alter its structure and normal function, contributing to RCM. Conclusion Here, an FLNC missense mutation (c.6451G>A, p.G2151S) known to be pathogenic in hypertrophic cardiomyopathy, was found to be associated with RCM, indicating the genetic overlap among cardiomyopathies. This study provides insights into Phenotype-Genotype Correlations of RCM in patients with FLNC mutations.
Collapse
Affiliation(s)
- Jiangtao Dong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Wenjuan Zhang
- Department of Geriatrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Qianwen Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Department of Pediatric Cardiology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430070, People’s Republic of China
| | - Lingfeng Zha
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
9
|
Dong R, Zhou X, Zhang H, Shi B, Liu G, Liu Y. Novel FLNC variants in pediatric cardiomyopathy: an insight into disease mechanisms. Hum Genomics 2024; 18:118. [PMID: 39472949 PMCID: PMC11520881 DOI: 10.1186/s40246-024-00683-9] [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: 09/06/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND FLNC gene variants have predominantly been reported in adult populations with cardiomyopathies, and early-onset cases are less common. The genotype-phenotype relationship indicates that dilated cardiomyopathy (DCM) is often associated with FLNC truncating variants. METHODS We conducted a comprehensive genetic analysis using next generation sequencing (NGS) to identify FLNC variants in patients with cardiovascular conditions. Detailed phenotypic and variant analyses were performed to characterize the clinical features and genetic alterations. Minigene assays and structural modeling were used to investigate the pathogenicity caused by the identified variants. RESULTS In a cohort of 58 patients, novel heterozygous FLNC variants, c.3962A > T (p.Glu1321Val) and c.7543C > T (p.Leu2515Phe), were identified in patients presenting with dilated and mixed restrictive/hypertrophic cardiomyopathies, respectively. The c.3962A > T variant disrupted normal splicing, as demonstrated through the splicing prediction tool and minigene studies, further emphasizing its pathogenic potential. CONCLUSION For missense variants of FLNC in patients with DCM, the splicing effect of the variant should be carefully checked. Early detection and intervention are crucial given the high risk of sudden cardiac death and severe cardiac complications.
Collapse
Affiliation(s)
- Rui Dong
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, China
| | - Xin Zhou
- Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, China
- Cardiovascular department, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China
| | - Haiyan Zhang
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China
- Shandong Provincial Clinical Research Center for Children's Health and Disease, Jinan, China
| | - Bingyi Shi
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China.
- Cardiovascular department, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China.
| | - Guohua Liu
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China.
- Department of Pediatrics, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital, Jinan), Jinan, China.
| | - Yi Liu
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China.
- Cardiovascular department, Children's Hospital Affiliated to Shandong University (Jinan Children's Hospital), Jinan, China.
| |
Collapse
|
10
|
Newman NA, Burke MA. Dilated Cardiomyopathy: A Genetic Journey from Past to Future. Int J Mol Sci 2024; 25:11460. [PMID: 39519012 PMCID: PMC11546582 DOI: 10.3390/ijms252111460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by reduced systolic function and cardiac dilation. Cases without an identified secondary cause are classified as idiopathic dilated cardiomyopathy (IDC). Over the last 35 years, many cases of IDC have increasingly been recognized to be genetic in etiology with a core set of definitively causal genes in up to 40% of cases. While over 200 genes have been associated with DCM, the evidence supporting pathogenicity for most remains limited. Further, rapid advances in sequencing and bioinformatics have recently revealed a complex genetic spectrum ranging from monogenic to polygenic in DCM. These advances have also led to the discovery of causal and modifier genetic variants in secondary forms of DCM (e.g., alcohol-induced cardiomyopathy). Current guidelines recommend genetic counseling and screening, as well as endorsing a handful of genotype-specific therapies (e.g., device placement in LMNA cardiomyopathy). The future of genetics in DCM will likely involve polygenic risk scores, direct-to-consumer testing, and pharmacogenetics, requiring providers to have a thorough understanding of this rapidly developing field. Herein we outline three decades of genetics in DCM, summarize recent advances, and project possible future avenues for the field.
Collapse
Affiliation(s)
- Noah A. Newman
- Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Michael A. Burke
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
11
|
Goliusova DV, Sharikova MY, Lavrenteva KA, Lebedeva OS, Muranova LK, Gusev NB, Bogomazova AN, Lagarkova MA. Role of Filamin C in Muscle Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1546-1557. [PMID: 39418514 DOI: 10.1134/s0006297924090025] [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: 06/13/2024] [Revised: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Filamin C (FLNC) is a member of a high-molecular weight protein family, which bind actin filaments in the cytoskeleton of various cells. In human genome FLNC is encoded by the FLNC gene located on chromosome 7 and is expressed predominantly in striated skeletal and cardiac muscle cells. Filamin C is involved in organization and stabilization of thin actin filaments three-dimensional network in sarcomeres, and is supposed to play a role of mechanosensor transferring mechanical signals to different protein targets. Under mechanical stress FLNC can undergo unfolding that increases the risk of its aggregation. FLNC molecules with an impaired native structure could be eliminated by the BAG3-mediated chaperone-assisted selective autophagy. Mutations in the FLNC gene could be accompanied by the changes in FLNC interaction with its protein partners and could lead to formation of aggregates, which overload the autophagy and proteasome protein degradation systems, thus facilitating development of various pathological processes. Molecular mechanisms of the FLNC-associated congenital disorders, called filaminopathies, remain poorly understood. This review is devoted to analysis of the structure and mechanisms of filamin C function in muscle and heart cells in normal state and in the FLNC-associated pathologies. The presented data summarize the results of research at the molecular, cellular, and tissue levels and allow us to outline promising ways for further investigation of pathogenetic mechanisms in filaminopathies.
Collapse
Affiliation(s)
- Daria V Goliusova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia.
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita Y Sharikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Kristina A Lavrenteva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga S Lebedeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Lidia K Muranova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nikolai B Gusev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexandra N Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| |
Collapse
|
12
|
García-Hernández S, de la Higuera Romero L, Ochoa JP, McKenna WJ. Emerging Themes in Genetics of Hypertrophic Cardiomyopathy: Current Status and Clinical Application. Can J Cardiol 2024; 40:742-753. [PMID: 38244984 DOI: 10.1016/j.cjca.2024.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM), defined clinically by the presence of unexplained left ventricular hypertrophy (LVH), with wall thickness ≥ 1.5 cm, is a phenotype in search of a diagnosis, which is most often a genetically determined, cardiac exclusive, or systemic disorder. Familial evaluation and genetic testing are required for definitive diagnosis. The role of genetic findings in predicting development of disease, outcomes, and increasingly to guide management is evolving with access to larger data sets. The specific mutation and sex of the patient are important determinants that ultimately are likely to guide management. The genetic/familial evaluation is influenced by the accuracy of the clinical diagnosis and the extent/expertise of the genetic laboratory. Genetic testing in a patient with unexplained LVH without systemic manifestations will yield a definite/likely pathogenetic mutation in a sarcomere (30%-50%), regulatory/functional (10%-15%) or metabolic/syndromic (< 5%) gene associated with Mendelian inheritance. The importance of oligo- and polygenic determinants, usually in the absence of Mendelian inheritance, is under investigation with important implications, particularly related to familial evaluation and definition of risk of disease development in relatives of probands. The results of genetic testing are increasingly important in management strategies related to the use of the implantable cardioverter defibrillator for prevention of sudden death, use of myosin inhibitors for refractory symptoms in patients with and without outflow tract obstruction, and-on the immediate horizon-gene therapy. This review will focus on genetic and outcome data in sarcomeric HCM, and minor causative genes with robust evidence of their association will also be considered.
Collapse
Affiliation(s)
| | | | - Juan Pablo Ochoa
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Centro Nacional de Investigaciones Cardiovasculades (CNIC), Madrid, Spain; Health in Code S.L., A Coruña, Spain
| | - William J McKenna
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Institute of Cardiovascular Science, University College London, London, United Kingdom; Health in Code S.L., A Coruña, Spain.
| |
Collapse
|
13
|
Lazzarino M, Zanetti M, Chen SN, Gao S, Peña B, Lam CK, Wu JC, Taylor MRG, Mestroni L, Sbaizero O. Defective Biomechanics and Pharmacological Rescue of Human Cardiomyocytes with Filamin C Truncations. Int J Mol Sci 2024; 25:2942. [PMID: 38474188 PMCID: PMC10932268 DOI: 10.3390/ijms25052942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Actin-binding filamin C (FLNC) is expressed in cardiomyocytes, where it localizes to Z-discs, sarcolemma, and intercalated discs. Although FLNC truncation variants (FLNCtv) are an established cause of arrhythmias and heart failure, changes in biomechanical properties of cardiomyocytes are mostly unknown. Thus, we investigated the mechanical properties of human-induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) carrying FLNCtv. CRISPR/Cas9 genome-edited homozygous FLNCKO-/- hiPSC-CMs and heterozygous knock-out FLNCKO+/- hiPSC-CMs were analyzed and compared to wild-type FLNC (FLNCWT) hiPSC-CMs. Atomic force microscopy (AFM) was used to perform micro-indentation to evaluate passive and dynamic mechanical properties. A qualitative analysis of the beating traces showed gene dosage-dependent-manner "irregular" peak profiles in FLNCKO+/- and FLNCKO-/- hiPSC-CMs. Two Young's moduli were calculated: E1, reflecting the compression of the plasma membrane and actin cortex, and E2, including the whole cell with a cytoskeleton and nucleus. Both E1 and E2 showed decreased stiffness in mutant FLNCKO+/- and FLNCKO-/- iPSC-CMs compared to that in FLNCWT. The cell adhesion force and work of adhesion were assessed using the retraction curve of the SCFS. Mutant FLNC iPSC-CMs showed gene dosage-dependent decreases in the work of adhesion and adhesion forces from the heterozygous FLNCKO+/- to the FLNCKO-/- model compared to FLNCWT, suggesting damaged cytoskeleton and membrane structures. Finally, we investigated the effect of crenolanib on the mechanical properties of hiPSC-CMs. Crenolanib is an inhibitor of the Platelet-Derived Growth Factor Receptor α (PDGFRA) pathway which is upregulated in FLNCtv hiPSC-CMs. Crenolanib was able to partially rescue the stiffness of FLNCKO-/- hiPSC-CMs compared to control, supporting its potential therapeutic role.
Collapse
Affiliation(s)
- Marco Lazzarino
- CNR-IOM, Area Science Park, 34149 Trieste, Italy; (M.L.); (M.Z.)
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Michele Zanetti
- CNR-IOM, Area Science Park, 34149 Trieste, Italy; (M.L.); (M.Z.)
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Suet Nee Chen
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Shanshan Gao
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Brisa Peña
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
- Bioengineering Department, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; (C.K.L.); (J.C.W.)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; (C.K.L.); (J.C.W.)
| | - Matthew R. G. Taylor
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Luisa Mestroni
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Orfeo Sbaizero
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
- Engineering and Architecture Department, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
14
|
Gao S, He L, Lam CK, Taylor MRG, Mestroni L, Lombardi R, Chen SN. Filamin C Deficiency Impairs Sarcomere Stability and Activates Focal Adhesion Kinase through PDGFRA Signaling in Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Cells 2024; 13:278. [PMID: 38334670 PMCID: PMC10854597 DOI: 10.3390/cells13030278] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/10/2024] Open
Abstract
Truncating mutations in filamin C (FLNC) are associated with dilated cardiomyopathy and arrhythmogenic cardiomyopathy. FLNC is an actin-binding protein and is known to interact with transmembrane and structural proteins; hence, the ablation of FLNC in cardiomyocytes is expected to dysregulate cell adhesion, cytoskeletal organization, sarcomere structural integrity, and likely nuclear function. Our previous study showed that the transcriptional profiles of FLNC homozygous deletions in human pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are highly comparable to the transcriptome profiles of hiPSC-CMs from patients with FLNC truncating mutations. Therefore, in this study, we used CRISPR-Cas-engineered hiPSC-derived FLNC knockout cardiac myocytes as a model of FLNC cardiomyopathy to determine pathogenic mechanisms and to examine structural changes caused by FLNC deficiency. RNA sequencing data indicated the significant upregulation of focal adhesion signaling and the dysregulation of thin filament genes in FLNC-knockout (FLNCKO) hiPSC-CMs compared to isogenic hiPSC-CMs. Furthermore, our findings suggest that the complete loss of FLNC in cardiomyocytes led to cytoskeletal defects and the activation of focal adhesion kinase. Pharmacological inhibition of PDGFRA signaling using crenolanib (an FDA-approved drug) reduced focal adhesion kinase activation and partially normalized the focal adhesion signaling pathway. The findings from this study suggest the opportunity in repurposing FDA-approved drug as a therapeutic strategy to treat FLNC cardiomyopathy.
Collapse
Affiliation(s)
- Shanshan Gao
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
| | - Lingaonan He
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
| | - Chi Keung Lam
- Department of Biological Sciences, University of Delaware, Newark, NE 19716, USA;
| | - Matthew R. G. Taylor
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
| | - Luisa Mestroni
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
| | - Raffaella Lombardi
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
- Department of Advanced Biomedical Sciences, “Federico II” University of Naples, 80138 Naples, Italy
| | - Suet Nee Chen
- University of Colorado Cardiovascular Institute, University of Colorado-Anschutz Medical and Boulder Campuses, Aurora, CO 80045, USA; (S.G.); (L.H.); (M.R.G.T.); (L.M.); (R.L.)
| |
Collapse
|
15
|
Onnée M, Bénézit A, Bastu S, Nadaj-Pakleza A, Lannes B, Ader F, Thèze C, Cintas P, Cances C, Carlier RY, Metay C, Cossée M, Malfatti E. The FLNC Ala1186Val Variant Linked to Cytoplasmic Body Myopathy and Cardiomyopathy Causes Protein Instability. Biomedicines 2024; 12:322. [PMID: 38397924 PMCID: PMC10887408 DOI: 10.3390/biomedicines12020322] [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: 12/29/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Filamin C-related disorders include myopathies and cardiomyopathies linked to variants in the FLNC gene. Filamin C belongs to a family of actin-binding proteins involved in sarcomere stability. This study investigates the pathogenic impact of the FLNC c.3557C > T (p.Ala1186Val) pathogenic variant associated with an early-onset cytoplasmic body myopathy and cardiomyopathy in three unrelated patients. We performed clinical imaging and myopathologic and genetic characterization of three patients with an early-onset myopathy and cardiomyopathy. Bioinformatics analysis, variant interpretation, and protein structure analysis were performed to validate and assess the effects of the filamin C variant. All patients presented with a homogeneous clinical phenotype marked by a severe contractural myopathy, leading to loss of gait. There was prominent respiratory involvement and restrictive or hypertrophic cardiomyopathies. The Ala1186Val variant is located in the interstrand loop involved in intradomain stabilization and/or interdomain interactions with neighbor Ig-like domains. 3D modeling highlights local structural changes involving nearby residues and probably impacts the protein stability, causing protein aggregation in the form of cytoplasmic bodies. Myopathologic studies have disclosed the prominent aggregation and upregulation of the aggrephagy-associated proteins LC3B and p62. As a whole, the Ala1186Val variant in the FLNC gene provokes a severe myopathy with contractures, respiratory involvement, and cardiomyopathy due to protein aggregation in patients' muscles.
Collapse
Affiliation(s)
- Marion Onnée
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
| | - Audrey Bénézit
- Neurologie et Réanimation Pédiatrique, Assistance Publique–Hôpitaux de Paris, Université Paris Saclay, Département Médico-Universitaire Santé de l’Enfant et de l’Adolescent, Hôpital Raymond Poincaré, 92380 Garches, France;
| | - Sultan Bastu
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
| | - Aleksandra Nadaj-Pakleza
- Centre de Référence des Maladies Neuromusculaires Nord Est Ile-de-France, Service de Neurologie, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France;
- European Reference Network, EURO-NMD, Neuromuscular Centre at Hautepierre Hospital, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Béatrice Lannes
- Département de Pathologie, Hôpitaux Universitaires de Strasbourg, 67091 Strasbourg, France;
| | - Flavie Ader
- Assistance Publique–Hôpitaux de Paris, Sorbonne Université, Département Médico-Universitaire BioGem, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, 75013 Paris, France;
- Institut National de la Santé et de la Recherche Médicale UMRS1166, Université Paris Cité, 75006 Paris, France
| | - Corinne Thèze
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, Université de Montpellier, 34095 Montpellier, France;
| | - Pascal Cintas
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Département de Neurologie, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (P.C.); (M.C.)
| | - Claude Cances
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Unité de Neurologie Pédiatrique, Hôpital des Enfants, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Robert-Yves Carlier
- Assistance Publique–Hôpitaux de Paris, Groupe Hospitalier Universitaire Paris Saclay, Département Médico-Universitaire Smart Imaging, Service d’Imagerie Médicale, Institut National de la Santé et de la Recherche Médicale UMR1179, Hôpital Raymond Poincaré, 92380 Garches, France;
| | - Corinne Metay
- Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Centre de Génétique Moléculaire et Chromosomique, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Groupe Hospitalier La Pitié-Salpêtrière, 75013 Paris, France;
| | - Mireille Cossée
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Département de Neurologie, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (P.C.); (M.C.)
- PhyMedExp, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, 34295 Montpellier, France
| | - Edoardo Malfatti
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
- Assistance Publique–Hôpitaux de Paris, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Hôpital Henri Mondor, 94000 Créteil, France
| |
Collapse
|
16
|
Allouba M, Walsh R, Afify A, Hosny M, Halawa S, Galal A, Fathy M, Theotokis PI, Boraey A, Ellithy A, Buchan R, Govind R, Whiffin N, Anwer S, ElGuindy A, Ware JS, Barton PJR, Yacoub M, Aguib Y. Ethnicity, consanguinity, and genetic architecture of hypertrophic cardiomyopathy. Eur Heart J 2023; 44:5146-5158. [PMID: 37431535 PMCID: PMC10733735 DOI: 10.1093/eurheartj/ehad372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/28/2023] [Accepted: 05/24/2023] [Indexed: 07/12/2023] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity that is partly explained by the diversity of genetic variants contributing to disease. Accurate interpretation of these variants constitutes a major challenge for diagnosis and implementing precision medicine, especially in understudied populations. The aim is to define the genetic architecture of HCM in North African cohorts with high consanguinity using ancestry-matched cases and controls. METHODS AND RESULTS Prospective Egyptian patients (n = 514) and controls (n = 400) underwent clinical phenotyping and genetic testing. Rare variants in 13 validated HCM genes were classified according to standard clinical guidelines and compared with a prospective HCM cohort of majority European ancestry (n = 684). A higher prevalence of homozygous variants was observed in Egyptian patients (4.1% vs. 0.1%, P = 2 × 10-7), with variants in the minor HCM genes MYL2, MYL3, and CSRP3 more likely to present in homozygosity than the major genes, suggesting these variants are less penetrant in heterozygosity. Biallelic variants in the recessive HCM gene TRIM63 were detected in 2.1% of patients (five-fold greater than European patients), highlighting the importance of recessive inheritance in consanguineous populations. Finally, rare variants in Egyptian HCM patients were less likely to be classified as (likely) pathogenic compared with Europeans (40.8% vs. 61.6%, P = 1.6 × 10-5) due to the underrepresentation of Middle Eastern populations in current reference resources. This proportion increased to 53.3% after incorporating methods that leverage new ancestry-matched controls presented here. CONCLUSION Studying consanguineous populations reveals novel insights with relevance to genetic testing and our understanding of the genetic architecture of HCM.
Collapse
Affiliation(s)
- Mona Allouba
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Roddy Walsh
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Alaa Afify
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mohammed Hosny
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Sarah Halawa
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Aya Galal
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mariam Fathy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Pantazis I Theotokis
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Ahmed Boraey
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Amany Ellithy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Rachel Buchan
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
| | - Risha Govind
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
- Present affiliation: National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Nicola Whiffin
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, UK
| | - Shehab Anwer
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Ahmed ElGuindy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Paul J R Barton
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Magdi Yacoub
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Harefield Heart Science Centre, Hill End Rd, Harefield, Uxbridge UB9 6JH, UK
| | - Yasmine Aguib
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| |
Collapse
|
17
|
Hespe S, Isbister JC, Duflou J, Puranik R, Bagnall RD, Semsarian C, Gray B, Ingles J. A case series of patients with filamin-C truncating variants attending a specialized cardiac genetic clinic. Eur Heart J Case Rep 2023; 7:ytad572. [PMID: 38116480 PMCID: PMC10729912 DOI: 10.1093/ehjcr/ytad572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/25/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023]
Abstract
Background FLNC encodes for filamin-C, a protein expressed in Z-discs of cardiac and skeletal muscle, involved in intracellular signalling and mechanical stabilization. Variants can cause diverse phenotypes with skeletal (myofibrillar or distal myopathy) and/or cardiac (hypertrophic, restrictive, and arrhythmogenic cardiomyopathies) manifestations. Truncating variants have recently been implicated in arrhythmogenic cardiomyopathy (ACM) without skeletal disease. Case summary Retrospective review of medical records, including cardiac investigations, was performed for families attending a specialized clinic with a FLNC truncating variant (FLNCtv). Variants were classified according to accepted variant interpretation criteria. Of seven families identified, six had primary cardiac phenotypes with one nonsense and five frameshift variants (nonsense-mediated decay competent) identified. One family had no cardiac phenotype, with a pathogenic variant (p.Arg2467Alafs*62) identified as secondary genetic finding. Of the six with cardiac phenotypes, proband age at diagnosis ranged 27-35 years (four females). Five families experienced sudden cardiac death (SCD) of a young relative (age range: 30-43 years), and one patient listed for cardiac transplant. Left ventricular (LV) ejection fraction ranged from 13 to 46%, with LV fibrosis (late gadolinium enhancement) on cardiac imaging or on postmortem histology seen in three families. Two families had one genotype-positive/phenotype-negative relative. Discussion The FLNCtv causes a left-sided ACM phenotype with a high risk of severe cardiac outcomes including end-stage heart failure and SCD. Incomplete penetrance is observed with implications for reporting secondary genetic findings.
Collapse
Affiliation(s)
- Sophie Hespe
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and University of New South Wales, 384 Victoria Street, Darlinghurst, 2010 NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
| | - Julia C Isbister
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, 2050 NSW, Australia
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, 2050 NSW, Australia
| | - Johan Duflou
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
| | - Raj Puranik
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, 2050 NSW, Australia
| | - Richard D Bagnall
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, 2050 NSW, Australia
| | - Christopher Semsarian
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, 2050 NSW, Australia
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, The University of Sydney, Sydney, 2050 NSW, Australia
| | - Belinda Gray
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, 2050 NSW, Australia
| | - Jodie Ingles
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, and University of New South Wales, 384 Victoria Street, Darlinghurst, 2010 NSW, Australia
- Faculty of Medicine and Health, The University of Sydney, Camperdown, 2050 NSW, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, 2050 NSW, Australia
| |
Collapse
|
18
|
Jacobs J, Van Aelst L, Breckpot J, Corveleyn A, Kuiperi C, Dupont M, Heggermont W, De Vadder K, Willems R, Van Cleemput J, Bogaert JG, Robyns T. Tools to differentiate between Filamin C and Titin truncating variant carriers: value of MRI. Eur J Hum Genet 2023; 31:1323-1332. [PMID: 37032351 PMCID: PMC10620392 DOI: 10.1038/s41431-023-01357-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/01/2023] [Accepted: 03/22/2023] [Indexed: 04/11/2023] Open
Abstract
Whereas truncating variants of the giant protein Titin (TTNtv) are the main cause of familial dilated cardiomyopathy (DCM), recently Filamin C truncating variants (FLNCtv) were identified as a cause of arrhythmogenic cardiomyopathy (ACM). Our aim was to characterize and compare clinical and MRI features of TTNtv and FLNCtv in the Belgian population. In index patients referred for genetic testing of ACM/DCM, FLNCtv and TTNtv were found in 17 (3.6%) and 33 (12.3%) subjects, respectively. Further family cascade screening yielded 24 and 19 additional truncating variant carriers in FLNC and TTN, respectively. The main phenotype was ACM in FLNCtv carriers whereas TTNtv carriers showed either an ACM or DCM phenotype. Non-sustained Ventricular Tachycardia was frequent in both populations. MRI data, available in 28/40 FLNCtv and 32/52 TTNtv patients, showed lower Left Ventricular (LV) ejection fraction and lower LV strain in TTNtv patients (p < 0.01). Conversely, both the frequency (68% vs 22%) and extent of non-ischemic myocardial late gadolinium enhancement (LGE) was significantly higher in FLNCtv patients (p < 0.01). Hereby, ring-like LGE was found in 16/19 (84%) FLNCtv versus 1/7 (14%) of TTNtv patients (p < 0.01). In conclusion, a large number of FLNCtv and TTNtv patients present with an ACM phenotype but can be separated by cardiac MRI. Whereas FLNCtv patients often have extensive myocardial fibrosis, typically following a ring-like pattern, LV dysfunction without or limited replacement fibrosis is the common TTNtv phenotype.
Collapse
Affiliation(s)
- Johanna Jacobs
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium.
- Department of Cardiovascular Diseases, University Hospitals Leuven, 3000, Leuven, Belgium.
| | - Lucas Van Aelst
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, 3000, Leuven, Belgium
| | | | | | - Cuno Kuiperi
- Center for Human Genetics, UZ Leuven, 3000, Leuven, Belgium
| | - Matthias Dupont
- Department of Cardiology, Ziekenhuis Oost-Limburg (ZOL), 3600, Genk, Belgium
| | - Ward Heggermont
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
- Department of Cardiology, Onze-Lieve-Vrouwziekenhuis Aalst, 9300, Aalst, Belgium
| | | | - Rik Willems
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Johan Van Cleemput
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, 3000, Leuven, Belgium
| | - Jan G Bogaert
- Department of Radiology, UZ Leuven, 3000, Leuven, Belgium
- Department of Imaging and Pathology, KU Leuven, 3000, Leuven, Belgium
| | - Tomas Robyns
- Department of Cardiovascular Sciences, KU Leuven, 3000, Leuven, Belgium
- Department of Cardiovascular Diseases, University Hospitals Leuven, 3000, Leuven, Belgium
| |
Collapse
|
19
|
Wang Y, Jia H, Song J. Accurate Classification of Non-ischemic Cardiomyopathy. Curr Cardiol Rep 2023; 25:1299-1317. [PMID: 37721634 PMCID: PMC10651539 DOI: 10.1007/s11886-023-01944-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE OF REVIEW This article aims to review the accurate classification of non-ischemic cardiomyopathy, including the methods, basis, subtype characteristics, and prognosis, especially the similarities and differences between different classifications. RECENT FINDINGS Non-ischemic cardiomyopathy refers to a myocardial disease that excludes coronary artery disease or ischemic injury and has a variety of etiologies and high incidence. Recent studies suggest that traditional classification methods based on primary/mixed/acquired or genetic/non-genetic cannot meet the precise needs of contemporary clinical management. This article systematically describes the history of classifications of cardiomyopathy and presents etiological and genetic differences between cardiomyopathies. The accurate classification is described from the perspective of morphology, function, and genomics in hypertrophic cardiomyopathy, dilated cardiomyopathy, restrictive cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, left ventricular noncompaction, and partially acquired cardiomyopathy. The different clinical characteristics and treatment needs of these cardiomyopathies are elaborated. Some single-gene mutant cardiomyopathies have unique phenotypes, and some cardiomyopathies have mixed phenotypes. These special classifications require personalized precision treatment, which is worthy of independent research. This article describes recent advances in the accurate classification of non-ischemic cardiomyopathy from clinical phenotypes and causative genes, discusses the advantages and usage scenarios of each classification, compares the differences in prognosis and patient management needs of different subtypes, and summarizes common methods and new exploration directions for accurate classification.
Collapse
Affiliation(s)
- Yifan Wang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.
| |
Collapse
|
20
|
Wong J, Peters S, Marwick TH. Phenotyping heart failure by genetics and associated conditions. Eur Heart J Cardiovasc Imaging 2023; 24:1293-1301. [PMID: 37279791 DOI: 10.1093/ehjci/jead125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 05/26/2023] [Indexed: 06/08/2023] Open
Abstract
Heart failure is a highly heterogeneous disease, and genetic testing may allow phenotypic distinctions that are incremental to those obtainable from imaging. Advances in genetic testing have allowed for the identification of deleterious variants in patients with specific heart failure phenotypes (dilated cardiomyopathy, arrhythmogenic right ventricular cardiomyopathy, and hypertrophic cardiomyopathy), and many of these have specific treatment implications. The diagnostic yield of genetic testing in heart failure is modest, and many rare variants are associated with incomplete penetrance and variable expressivity. Environmental factors and co-morbidities have a large role in the heterogeneity of the heart failure phenotype. Future endeavours should concentrate on the cumulative impact of genetic polymorphisms in the development of heart failure.
Collapse
Affiliation(s)
- Joshua Wong
- Baker Heart and Diabetes Institute and Department of Cardiometabolic Health, University of Melbourne, PO Box 6492, Melbourne, VIC 3004, Australia
| | - Stacey Peters
- Baker Heart and Diabetes Institute and Department of Cardiometabolic Health, University of Melbourne, PO Box 6492, Melbourne, VIC 3004, Australia
| | - Thomas H Marwick
- Baker Heart and Diabetes Institute and Department of Cardiometabolic Health, University of Melbourne, PO Box 6492, Melbourne, VIC 3004, Australia
| |
Collapse
|
21
|
Zhou X, Fang X, Ithychanda SS, Wu T, Gu Y, Chen C, Wang L, Bogomolovas J, Qin J, Chen J. Interaction of Filamin C With Actin Is Essential for Cardiac Development and Function. Circ Res 2023; 133:400-411. [PMID: 37492967 PMCID: PMC10529502 DOI: 10.1161/circresaha.123.322750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 07/17/2023] [Indexed: 07/27/2023]
Abstract
BACKGROUND FLNC (filamin C), a member of the filamin family predominantly expressed in striated muscles, plays a crucial role in bridging the cytoskeleton and ECM (extracellular matrix) in cardiomyocytes, thereby maintaining heart integrity and function. Although genetic variants within the N-terminal ABD (actin-binding domain) of FLNC have been identified in patients with cardiomyopathy, the precise contribution of the actin-binding capability to FLNC's function in mammalian hearts remains poorly understood. METHODS We conducted in silico analysis of the 3-dimensional structure of mouse FLNC to identify key amino acid residues within the ABD that are essential for FLNC's actin-binding capacity. Subsequently, we performed coimmunoprecipitation and immunofluorescent assays to validate the in silico findings and assess the impact of these mutations on the interactions with other binding partners and the subcellular localization of FLNC. Additionally, we generated and analyzed knock-in mouse models in which the FLNC-actin interaction was completely disrupted by these mutations. RESULTS Our findings revealed that F93A/L98E mutations completely disrupted FLNC-actin interaction while preserving FLNC's ability to interact with other binding partners ITGB1 (β1 integrin) and γ-SAG (γ-sarcoglycan), as well as maintaining FLNC subcellular localization. Loss of FLNC-actin interaction in embryonic cardiomyocytes resulted in embryonic lethality and cardiac developmental defects, including ventricular wall malformation and reduced cardiomyocyte proliferation. Moreover, disruption of FLNC-actin interaction in adult cardiomyocytes led to severe dilated cardiomyopathy, enhanced lethality and dysregulation of key cytoskeleton components. CONCLUSIONS Our data strongly support the crucial role of FLNC as a bridge between actin filaments and ECM through its interactions with actin, ITGB1, γ-SAG, and other associated proteins in cardiomyocytes. Disruption of FLN-actin interaction may result in detachment of actin filaments from the extracellular matrix, ultimately impairing normal cardiac development and function. These findings also provide insights into mechanisms underlying cardiomyopathy associated with genetic variants in FLNC ABD and other regions.
Collapse
Affiliation(s)
- Xiaohai Zhou
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Xi Fang
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Sujay Subbayya Ithychanda
- Department of Cardiovascular and Metabolic Sciences (S.S.I., J.Q.), Lerner Research Institute, Cleveland Clinic, OH
| | - Tongbin Wu
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Yusu Gu
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Chao Chen
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Li Wang
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Julius Bogomolovas
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| | - Jun Qin
- Department of Cardiovascular and Metabolic Sciences (S.S.I., J.Q.), Lerner Research Institute, Cleveland Clinic, OH
| | - Ju Chen
- Department of Medicine (X.Z., X.F., T.W., Y.G., C.C., L.W., J.B., J.C.), University of California San Diego, La Jolla
| |
Collapse
|
22
|
Bermúdez-Jiménez FJ, Carriel V, Santos-Mateo JJ, Fernández A, García-Hernández S, Ramos KA, Piqueras-Flores J, Cabrera-Romero E, Barriales-Villa R, de la Higuera Romero L, Alcalá López JE, Gimeno Blanes JR, Sánchez-Porras D, Campos F, Alaminos M, Oyonarte-Ramírez JM, Álvarez M, Tercedor L, Brodehl A, Jiménez-Jáimez J. ROD2 domain filamin C missense mutations exhibit a distinctive cardiac phenotype with restrictive/hypertrophic cardiomyopathy and saw-tooth myocardium. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2023; 76:301-311. [PMID: 35952944 DOI: 10.1016/j.rec.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/28/2022] [Indexed: 04/29/2023]
Abstract
INTRODUCTION AND OBJECTIVES Missense mutations in the filamin C (FLNC) gene have been reported as cause of inherited cardiomyopathy. Knowledge of the pathogenicity and genotype-phenotype correlation remains scarce. Our aim was to describe a distinctive cardiac phenotype related to rare missense FLNC variants in the ROD2 domain. METHODS We recruited 21 unrelated families genetically evaluated because of hypertrophic cardiomyopathy (HCM)/restrictive cardiomyopathy (RCM) phenotype carrying rare missense variants in the ROD2 domain of FLNC (FLNC-mRod2). Carriers underwent advanced cardiac imaging and genetic cascade screening. Myocardial tissue from 3 explanted hearts of a missense FLNC carrier was histologically analyzed and compared with an FLNC-truncating variant heart sample and a healthy control. Plasmids independently containing 3 FLNC missense variants were transfected and analyzed using confocal microscopy. RESULTS Eleven families (52%) with 20 assessed individuals (37 [23.7-52.7]) years showed 15 cases with a cardiac phenotype consisting of an overlap of HCM-RCM and left ventricular hypertrabeculation (saw-tooth appearance). During a median follow-up of 6.49 years, they presented with advanced heart failure: 16 (80%) diastolic dysfunction, 3 heart transplants, 3 heart failure deaths) and absence of cardiac conduction disturbances or skeletal myopathy. A total of 6 families had moderate genotype-phenotype segregation, and the remaining were de novo variants. Differential extracellular matrix remodeling and FLNC distribution among cardiomyocytes were confirmed on histology. HT1080 and H9c2 cells did not reveal cytoplasmic aggregation of mutant FLNC. CONCLUSIONS FLNC-mRod2 variants show a high prevalence of an overlapped phenotype comprising RCM, HCM and deep hypertrabeculation with saw-tooth appearance and distinctive cardiac histopathological remodeling.
Collapse
Affiliation(s)
- Francisco José Bermúdez-Jiménez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Víctor Carriel
- Departamento de Histología, Grupo de Ingeniería Tisular, Universidad de Granada, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Juan José Santos-Mateo
- Servicio de Cardiología, Hospital Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca Murcia (IMIB), Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-Guard Heart), Amsterdam, Netherlands
| | - Adrián Fernández
- Servicio de Cardiología, Hospital Universitario Fundación Favaloro, Buenos Aires, Argentina
| | - Soledad García-Hernández
- Health in Code SL, Cardiología y Departamento Científico, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Karina Analía Ramos
- Servicio de Cardiología, Hospital Centenario, Facultad de Ciencias Médicas, Universidad de Rosario, Argentina
| | - Jesús Piqueras-Flores
- Servicio de Cardiología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Eva Cabrera-Romero
- Servicio de Cardiología, Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Roberto Barriales-Villa
- Complexo Hospitalario Universitario A Coruña, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Luis de la Higuera Romero
- Health in Code SL, Cardiología y Departamento Científico, Instituto de Investigación Biomédica de A Coruña (INIBIC), A Coruña, Spain
| | - Juan Emilio Alcalá López
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Juan Ramón Gimeno Blanes
- Servicio de Cardiología, Hospital Universitario Virgen de la Arrixaca, Instituto Murciano de Investigación Biosanitaria Virgen de la Arrixaca Murcia (IMIB), Murcia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain; European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart (ERN-Guard Heart), Amsterdam, Netherlands
| | - David Sánchez-Porras
- Departamento de Histología, Grupo de Ingeniería Tisular, Universidad de Granada, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Fernando Campos
- Departamento de Histología, Grupo de Ingeniería Tisular, Universidad de Granada, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Miguel Alaminos
- Departamento de Histología, Grupo de Ingeniería Tisular, Universidad de Granada, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - José Manuel Oyonarte-Ramírez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Miguel Álvarez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Luis Tercedor
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain
| | - Andreas Brodehl
- Erich and Hanna Klessmann Institute for Cardiovascular Research & Development (EHKI), Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Juan Jiménez-Jáimez
- Servicio de Cardiología, Hospital Universitario Virgen de las Nieves, Instituto de Investigación Biosanitaria ibsGRANADA, Granada, Spain.
| |
Collapse
|
23
|
Lawley CM, Kaski JP. Clinical and Genetic Screening for Hypertrophic Cardiomyopathy in Paediatric Relatives: Changing Paradigms in Clinical Practice. J Clin Med 2023; 12:2788. [PMID: 37109125 PMCID: PMC10146293 DOI: 10.3390/jcm12082788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/02/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is an important cause of morbidity and mortality in children. While the aetiology is heterogeneous, most cases are caused by variants in the genes encoding components of the cardiac sarcomere, which are inherited as an autosomal dominant trait. In recent years, there has been a paradigm shift in the role of clinical screening and predictive genetic testing in children with a first-degree relative with HCM, with the recognition that phenotypic expression can, and often does, manifest in young children and that familial disease in the paediatric age group may not be benign. The care of the child and family affected by HCM relies on a multidisciplinary team, with a key role for genomics. This review article summarises current evidence in clinical and genetic screening for hypertrophic cardiomyopathy in paediatric relatives and highlights aspects that remain to be resolved.
Collapse
Affiliation(s)
- Claire M. Lawley
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London WC1N 3JH, UK
- The University of Sydney Children’s Hospital Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Juan Pablo Kaski
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London WC1N 3JH, UK
- Centre for Paediatric Inherited and Rare Cardiovascular Disease, University College London Institute of Cardiovascular Science, London WC1E 6DD, UK
| |
Collapse
|
24
|
Wang BZ, Nash TR, Zhang X, Rao J, Abriola L, Kim Y, Zakharov S, Kim M, Luo LJ, Morsink M, Liu B, Lock RI, Fleischer S, Tamargo MA, Bohnen M, Welch CL, Chung WK, Marx SO, Surovtseva YV, Vunjak-Novakovic G, Fine BM. Engineered cardiac tissue model of restrictive cardiomyopathy for drug discovery. Cell Rep Med 2023; 4:100976. [PMID: 36921598 PMCID: PMC10040415 DOI: 10.1016/j.xcrm.2023.100976] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/19/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023]
Abstract
Restrictive cardiomyopathy (RCM) is defined as increased myocardial stiffness and impaired diastolic relaxation leading to elevated ventricular filling pressures. Human variants in filamin C (FLNC) are linked to a variety of cardiomyopathies, and in this study, we investigate an in-frame deletion (c.7416_7418delGAA, p.Glu2472_Asn2473delinAsp) in a patient with RCM. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with this variant display impaired relaxation and reduced calcium kinetics in 2D culture when compared with a CRISPR-Cas9-corrected isogenic control line. Similarly, mutant engineered cardiac tissues (ECTs) demonstrate increased passive tension and impaired relaxation velocity compared with isogenic controls. High-throughput small-molecule screening identifies phosphodiesterase 3 (PDE3) inhibition by trequinsin as a potential therapy to improve cardiomyocyte relaxation in this genotype. Together, these data demonstrate an engineered cardiac tissue model of RCM and establish the translational potential of this precision medicine approach to identify therapeutics targeting myocardial relaxation.
Collapse
Affiliation(s)
- Bryan Z Wang
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Trevor R Nash
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Xiaokan Zhang
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Jenny Rao
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Laura Abriola
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Youngbin Kim
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sergey Zakharov
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Michael Kim
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Lori J Luo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Margaretha Morsink
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bohao Liu
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Roberta I Lock
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Sharon Fleischer
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Manuel A Tamargo
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Michael Bohnen
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Carrie L Welch
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Wendy K Chung
- Department of Pediatrics, Columbia University, New York, NY 10032, USA
| | - Steven O Marx
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Yulia V Surovtseva
- Yale Center for Molecular Discovery, Yale University, New Haven, CT 06520, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA; Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA; College of Dental Medicine, Columbia University, New York, NY 10032, USA
| | - Barry M Fine
- Department of Medicine, Division of Cardiology, Columbia University Medical Center, New York, NY 10032, USA.
| |
Collapse
|
25
|
Khurshid S, Lazarte J, Pirruccello JP, Weng LC, Choi SH, Hall AW, Wang X, Friedman SF, Nauffal V, Biddinger KJ, Aragam KG, Batra P, Ho JE, Philippakis AA, Ellinor PT, Lubitz SA. Clinical and genetic associations of deep learning-derived cardiac magnetic resonance-based left ventricular mass. Nat Commun 2023; 14:1558. [PMID: 36944631 PMCID: PMC10030590 DOI: 10.1038/s41467-023-37173-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/04/2023] [Indexed: 03/23/2023] Open
Abstract
Left ventricular mass is a risk marker for cardiovascular events, and may indicate an underlying cardiomyopathy. Cardiac magnetic resonance is the gold-standard for left ventricular mass estimation, but is challenging to obtain at scale. Here, we use deep learning to enable genome-wide association study of cardiac magnetic resonance-derived left ventricular mass indexed to body surface area within 43,230 UK Biobank participants. We identify 12 genome-wide associations (1 known at TTN and 11 novel for left ventricular mass), implicating genes previously associated with cardiac contractility and cardiomyopathy. Cardiac magnetic resonance-derived indexed left ventricular mass is associated with incident dilated and hypertrophic cardiomyopathies, and implantable cardioverter-defibrillator implant. An indexed left ventricular mass polygenic risk score ≥90th percentile is also associated with incident implantable cardioverter-defibrillator implant in separate UK Biobank (hazard ratio 1.22, 95% CI 1.05-1.44) and Mass General Brigham (hazard ratio 1.75, 95% CI 1.12-2.74) samples. Here, we perform a genome-wide association study of cardiac magnetic resonance-derived indexed left ventricular mass to identify 11 novel variants and demonstrate that cardiac magnetic resonance-derived and genetically predicted indexed left ventricular mass are associated with incident cardiomyopathy.
Collapse
Affiliation(s)
- Shaan Khurshid
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Julieta Lazarte
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - James P Pirruccello
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Amelia W Hall
- Gene Regulation Observatory, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Wang
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel F Friedman
- Data Sciences Platform, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Victor Nauffal
- Division of Cardiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Kiran J Biddinger
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Krishna G Aragam
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Boston, MA, USA
| | - Puneet Batra
- Data Sciences Platform, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jennifer E Ho
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- CardioVascular Institute and Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Anthony A Philippakis
- Data Sciences Platform, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Steven A Lubitz
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
26
|
Gaudreault N, Ruel LJ, Henry C, Schleit J, Lagüe P, Champagne J, Sénéchal M, Sarrazin JF, Philippon F, Bossé Y, Steinberg C. Novel filamin C (FLNC) variant causes a severe form of familial mixed hypertrophic-restrictive cardiomyopathy. Am J Med Genet A 2023; 191:1508-1517. [PMID: 36864778 DOI: 10.1002/ajmg.a.63169] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/12/2022] [Accepted: 02/10/2023] [Indexed: 03/04/2023]
Abstract
Variants of filamin C (FLNC) have been identified as rare genetic substrate for hypertrophic cardiomyopathy (HCM). Data on the clinical course of FLNC-related HCM are conflicting with some studies suggesting mild phenotypes whereas other studies have reported more severe outcomes. In this study, we present a novel FLNC variant (Ile1937Asn) that was identified in a large family of French-Canadian descent with excellent segregation data. FLNC-Ile1937Asn is a novel missense variant characterized by full penetrance and poor clinical outcomes. End stage heart failure requiring transplantation occurred in 43% and sudden cardiac death in 29% of affected family members. Other particular features of FLNC-Ile1937Asn include an early disease onset (mean age of 19 years) and the development of a marked atrial myopathy (severe biatrial dilatation with remodeling and multiple complex atrial arrhythmias) that was present in all gene carriers. The FLNC-Ile1937Asn variant is a novel, pathogenic mutation resulting in a severe form of HCM with full disease penetrance. The variant is associated with a high proportion of end-stage heart failure, heart transplantation, and disease-related mortality. Close follow-up and appropriate risk stratification of affected individuals at specialized heart centers is recommended.
Collapse
Affiliation(s)
- Nathalie Gaudreault
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - Louis-Jacques Ruel
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - Cyndi Henry
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | | | - Patrick Lagüe
- PROTEO, The Quebec Network for Research on Protein Function, Engineering, and Applications, Quebec, Canada.,The Institute of integrative biology and systems (IBIS), Laval University, Quebec, Canada
| | - Jean Champagne
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Multidisciplinary Department of Cardiology and Cardiac Surgery, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - Mario Sénéchal
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Multidisciplinary Department of Cardiology and Cardiac Surgery, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - Jean-François Sarrazin
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Multidisciplinary Department of Cardiology and Cardiac Surgery, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - François Philippon
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Multidisciplinary Department of Cardiology and Cardiac Surgery, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| | - Yohan Bossé
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Department of Molecular Medicine, Laval University, Quebec, Canada
| | - Christian Steinberg
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada.,Multidisciplinary Department of Cardiology and Cardiac Surgery, Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec, Canada
| |
Collapse
|
27
|
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
|
28
|
Expression of LIM domain-binding 3 (LDB3), a striated muscle Z-band alternatively spliced PDZ-motif protein in the nervous system. Sci Rep 2023; 13:270. [PMID: 36609526 PMCID: PMC9822979 DOI: 10.1038/s41598-023-27531-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023] Open
Abstract
LIM domain-binding 3 (LDB3) is a member of the Enigma family of PDZ-LIM proteins. LDB3 has been reported as a striated muscle-specific Z-band alternatively spliced protein that plays an important role in mechanosensory actin cytoskeleton remodeling. This study shows that LDB3 is broadly expressed in the central and peripheral nervous system of human and mouse. LDB3 is predominantly expressed in the adult stages compared to early development and at a significantly higher level in the spinal cord than in the brain. As in skeletal muscle and heart, LDB3 is extensively alternatively spliced in the neurons. Three novel splice isoforms were identified suggesting splicing-dependent regulation of LDB3 expression in the nervous system. Expression of LDB3 in the motor cortex, cerebellum, spinal motor neuron, peripheral nerve, and neuromuscular junction in addition to skeletal muscle indicates important roles for this PDZ-LIM family protein in motor planning and execution. Moreover, expression in the hippocampal neurons suggests roles for LDB3 in learning and memory. LDB3 interactors filamin C and myotilin are also expressed in the spinal motor neuron, nerve, and neuromuscular junction, thereby providing the basis for neurogenic manifestations in myopathies associated with mutations in these so-called muscle proteins.
Collapse
|
29
|
Ripoll-Vera T. Cardiac filaminopathies: lights and shadows in the phenotype associated with the FLNC gene. REVISTA ESPANOLA DE CARDIOLOGIA (ENGLISH ED.) 2022; 76:284-286. [PMID: 36539187 DOI: 10.1016/j.rec.2022.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 12/23/2022]
Affiliation(s)
- Tomás Ripoll-Vera
- Unidad de Cardiopatías Familiares, Hospital Universitario Son Llàtzer, Instituto de Investigación Sanitaria de Baleares (IdISBa), Palma de Mallorca, Islas Baleares, Spain; Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Spain.
| |
Collapse
|
30
|
Spracklen TF, Keavney B, Laing N, Ntusi N, Shaboodien G. Modern genomic techniques in the identification of genetic causes of cardiomyopathy. Heart 2022; 108:1843-1850. [PMID: 35140110 DOI: 10.1136/heartjnl-2021-320424] [Citation(s) in RCA: 6] [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: 10/06/2021] [Accepted: 01/18/2022] [Indexed: 11/04/2022] Open
Abstract
Over the past three decades numerous disease-causing genes have been linked to the pathogenesis of heritable cardiomyopathies, but many causal genes are yet to be identified. Next-generation sequencing (NGS) platforms have revolutionised clinical testing capacity in familial cardiomyopathy. In this review, we summarise how NGS technologies have advanced our understanding of genetic non-syndromic cardiomyopathy over the last decade. First, 26 putative new disease-causing genes have been identified to date, mostly from whole-exome sequencing, and some of which (FLNC, MTO1, HCN4) have had a considerable clinical impact and are now included in routine diagnostic gene panels. Second, we consider challenges in variant interpretation and the importance of large-scale NGS population control cohorts for this purpose. Third, an emerging role of common variation in some forms of genetic cardiomyopathy is being elucidated through recent studies which have illustrated an additive effect of numerous polymorphic loci on cardiac parameters; this may explain phenotypic variability and low rates of genetic diagnosis from sequencing studies. Finally, we discuss the clinical utility of genetic testing in cardiomyopathy in Western settings, where NGS panel testing of core disease genes is currently recommended with possible implications for patient management. Given the findings of recent studies, whole-exome or whole-genome sequencing should be considered in patients of non-European ancestry with clearly familial disease, or severe paediatric disease, when no result is obtained on panel sequencing. The clinical utility of polygenic risk assessment needs to be investigated further in patients with unexplained dilated cardiomyopathy and hypertrophic cardiomyopathy in whom a pathogenic variant is not identified.
Collapse
Affiliation(s)
- Timothy F Spracklen
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Cape Town, Cape Town, South Africa
| | - Bernard Keavney
- Division of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | - Nakita Laing
- Division of Human Genetics, University of Cape Town, Cape Town, South Africa
| | - Ntobeko Ntusi
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
- Department of Medicine, University of Cape Town, Cape Universities Body Imaging Centre, Cape Town, South Africa
| | - Gasnat Shaboodien
- Cape Heart Institute, University of Cape Town Department of Medicine, Cape Town, South Africa
| |
Collapse
|
31
|
Calcagno DM, Taghdiri N, Ninh VK, Mesfin JM, Toomu A, Sehgal R, Lee J, Liang Y, Duran JM, Adler E, Christman KL, Zhang K, Sheikh F, Fu Z, King KR. Single-cell and spatial transcriptomics of the infarcted heart define the dynamic onset of the border zone in response to mechanical destabilization. NATURE CARDIOVASCULAR RESEARCH 2022; 1:1039-1055. [PMID: 39086770 PMCID: PMC11290420 DOI: 10.1038/s44161-022-00160-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/03/2022] [Indexed: 08/02/2024]
Abstract
The border zone (BZ) of the infarcted heart is a geographically complex and biologically enigmatic interface separating poorly perfused infarct zones (IZs) from remote zones (RZs). The cellular and molecular mechanisms of myocardial BZs are not well understood because microdissection inevitably combines them with uncontrolled amounts of RZs and IZs. Here, we use single-cell/nucleus RNA sequencing, spatial transcriptomics and multiplexed RNA fluorescence in situ hybridization to redefine the BZ based on cardiomyocyte transcriptomes. BZ1 (Nppa + Xirp2 -) forms a hundreds-of-micrometer-thick layer of morphologically intact cells adjacent to RZs that are detectable within an hour of injury. Meanwhile, BZ2 (Nppa + Xirp2 +) forms a near-single-cell-thick layer of morphologically distorted cardiomyocytes at the IZ edge that colocalize with matricellular protein-expressing myofibroblasts and express predominantly mechanotransduction genes. Surprisingly, mechanical injury alone is sufficient to induce BZ genes. We propose a 'loss of neighbor' hypothesis to explain how ischemic cell death mechanically destabilizes the BZ to induce its transcriptional response.
Collapse
Affiliation(s)
- D. M. Calcagno
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally: D.M. Calcagno, N. Taghdiri
| | - N. Taghdiri
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally: D.M. Calcagno, N. Taghdiri
| | - V. K. Ninh
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - J. M. Mesfin
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - A. Toomu
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - R. Sehgal
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - J. Lee
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - Y. Liang
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - J. M. Duran
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - E. Adler
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - K. L. Christman
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, USA
| | - K. Zhang
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - F. Sheikh
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Z. Fu
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
| | - K. R. King
- Department of Bioengineering, Jacobs School of Engineering, University of California San Diego, La Jolla, CA, USA
- Division of Cardiology and Cardiovascular Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
32
|
Ripoll-Vera T. Filaminopatías cardiacas: luces y sombras en el fenotipo asociado con el gen FLNC. Rev Esp Cardiol 2022. [DOI: 10.1016/j.recesp.2022.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
33
|
Dong M, Liu J, Liu C, Wang H, Sun W, Liu B. CRISPR/CAS9: A promising approach for the research and treatment of cardiovascular diseases. Pharmacol Res 2022; 185:106480. [PMID: 36191879 DOI: 10.1016/j.phrs.2022.106480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 10/31/2022]
Abstract
The development of gene-editing technology has been one of the biggest advances in biomedicine over the past two decades. Not only can it be used as a research tool to build a variety of disease models for the exploration of disease pathogenesis at the genetic level, it can also be used for prevention and treatment. This is done by intervening with the expression of target genes and carrying out precise molecular targeted therapy for diseases. The simple and flexible clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene-editing technology overcomes the limitations of zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs). For this reason, it has rapidly become a preferred method for gene editing. As a new gene intervention method, CRISPR/Cas9 has been widely used in the clinical treatment of tumours and rare diseases; however, its application in the field of cardiovascular diseases is currently limited. This article reviews the application of the CRISPR/Cas9 editing technology in cardiovascular disease research and treatment, and discusses the limitations and prospects of this technology.
Collapse
Affiliation(s)
- Mengying Dong
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Jiangen Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Caixia Liu
- Department of Neurology, The Liaoning Province People's Hospital, 33 Wenyi Road, ShenYang, China, 110016
| | - He Wang
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041
| | - Wei Sun
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, 218 Ziqiang Road, Changchun, China, 130041.
| |
Collapse
|
34
|
Muravyev A, Vershinina T, Tesner P, Sjoberg G, Fomicheva Y, Čajbiková NN, Kozyreva A, Zhuk S, Mamaeva E, Tarnovskaya S, Jornholt J, Sokolnikova P, Pervunina T, Vasichkina E, Sejersen T, Kostareva A. Rare clinical phenotype of filaminopathy presenting as restrictive cardiomyopathy and myopathy in childhood. Orphanet J Rare Dis 2022; 17:358. [PMID: 36104822 PMCID: PMC9476594 DOI: 10.1186/s13023-022-02477-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background FLNC is one of the few genes associated with all types of cardiomyopathies, but it also underlies neuromuscular phenotype. The combination of concomitant neuromuscular and cardiac involvement is not often observed in filaminopathies and the impact of this on the disease prognosis has hitherto not been analyzed. Results Here we provide a detailed clinical, genetic, and structural prediction analysis of distinct FLNC-associated phenotypes based on twelve pediatric cases. They include early-onset restrictive cardiomyopathy (RCM) in association with congenital myopathy. In all patients the initial diagnosis was established during the first year of life and in five out of twelve (41.7%) patients the first symptoms were observed at birth. RCM was present in all patients, often in combination with septal defects. No ventricular arrhythmias were noted in any of the patients presented here. Myopathy was confirmed by neurological examination, electromyography, and morphological studies. Arthrogryposes was diagnosed in six patients and remained clinically meaningful with increasing age in three of them. One patient underwent successful heart transplantation at the age of 18 years and two patients are currently included in the waiting list for heart transplantation. Two died due to congestive heart failure. One patient had ICD instally as primary prevention of SCD. In ten out of twelve patients the disease was associated with missense variants and only in two cases loss of function variants were detected. In half of the described cases, an amino acid substitution A1186V, altering the structure of IgFLNc10, was found. Conclusions The present description of twelve cases of early-onset restrictive cardiomyopathy with congenital myopathy and FLNC mutation, underlines a distinct unique phenotype that can be suggested as a separate clinical form of filaminopathies. Amino acid substitution A1186V, which was observed in half of the cases, defines a mutational hotspot for the reported combination of myopathy and cardiomyopathy. Several independent molecular mechanisms of FLNC mutations linked to filamin structure and function can explain the broad spectrum of FLNC-associated phenotypes. Early disease presentation and unfavorable prognosis of heart failure demanding heart transplantation make awareness of this clinical form of filaminopathy of great clinical importance. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02477-5.
Collapse
|
35
|
Zech ATL, Prondzynski M, Singh SR, Pietsch N, Orthey E, Alizoti E, Busch J, Madsen A, Behrens CS, Meyer-Jens M, Mearini G, Lemoine MD, Krämer E, Mosqueira D, Virdi S, Indenbirken D, Depke M, Salazar MG, Völker U, Braren I, Pu WT, Eschenhagen T, Hammer E, Schlossarek S, Carrier L. ACTN2 Mutant Causes Proteopathy in Human iPSC-Derived Cardiomyocytes. Cells 2022; 11:cells11172745. [PMID: 36078153 PMCID: PMC9454684 DOI: 10.3390/cells11172745] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/24/2022] [Accepted: 08/29/2022] [Indexed: 11/28/2022] Open
Abstract
Genetic variants in α-actinin-2 (ACTN2) are associated with several forms of (cardio)myopathy. We previously reported a heterozygous missense (c.740C>T) ACTN2 gene variant, associated with hypertrophic cardiomyopathy, and characterized by an electro-mechanical phenotype in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Here, we created with CRISPR/Cas9 genetic tools two heterozygous functional knock-out hiPSC lines with a second wild-type (ACTN2wt) and missense ACTN2 (ACTN2mut) allele, respectively. We evaluated their impact on cardiomyocyte structure and function, using a combination of different technologies, including immunofluorescence and live cell imaging, RNA-seq, and mass spectrometry. This study showed that ACTN2mut presents a higher percentage of multinucleation, protein aggregation, hypertrophy, myofibrillar disarray, and activation of both the ubiquitin-proteasome system and the autophagy-lysosomal pathway as compared to ACTN2wt in 2D-cultured hiPSC-CMs. Furthermore, the expression of ACTN2mut was associated with a marked reduction of sarcomere-associated protein levels in 2D-cultured hiPSC-CMs and force impairment in engineered heart tissues. In conclusion, our study highlights the activation of proteolytic systems in ACTN2mut hiPSC-CMs likely to cope with ACTN2 aggregation and therefore directs towards proteopathy as an additional cellular pathology caused by this ACTN2 variant, which may contribute to human ACTN2-associated cardiomyopathies.
Collapse
Affiliation(s)
- Antonia T. L. Zech
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Maksymilian Prondzynski
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sonia R. Singh
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Niels Pietsch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Ellen Orthey
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Erda Alizoti
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Josefine Busch
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Alexandra Madsen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Charlotta S. Behrens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Moritz Meyer-Jens
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Giulia Mearini
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Marc D. Lemoine
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Department of Cardiology, University Heart and Vascular Center, 20246 Hamburg, Germany
| | - Elisabeth Krämer
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Diogo Mosqueira
- Biodiscovery Institute, University of Nottingham, Nottingham NG7 2RD, UK
| | - Sanamjeet Virdi
- Heinrich-Pette-Institute, Leibniz Institute of Virology, 20246 Hamburg, Germany
| | - Daniela Indenbirken
- Heinrich-Pette-Institute, Leibniz Institute of Virology, 20246 Hamburg, Germany
| | - Maren Depke
- Department for Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Manuela Gesell Salazar
- Department for Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Uwe Völker
- Department for Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Ingke Braren
- Vector Facility, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - William T. Pu
- Department of Cardiology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Elke Hammer
- Department for Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Greifswald, 17475 Greifswald, Germany
| | - Saskia Schlossarek
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
- Correspondence: ; Tel.: +49-40-7410-57208
| |
Collapse
|
36
|
Wilde AAM, Semsarian C, Márquez MF, Shamloo AS, Ackerman MJ, Ashley EA, Sternick EB, Barajas-Martinez H, Behr ER, Bezzina CR, Breckpot J, Charron P, Chockalingam P, Crotti L, Gollob MH, Lubitz S, Makita N, Ohno S, Ortiz-Genga M, Sacilotto L, Schulze-Bahr E, Shimizu W, Sotoodehnia N, Tadros R, Ware JS, Winlaw DS, Kaufman ES. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases. Europace 2022; 24:1307-1367. [PMID: 35373836 PMCID: PMC9435643 DOI: 10.1093/europace/euac030] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Arthur A M Wilde
- Heart Centre, Department of Cardiology, Amsterdam Universitair Medische
Centra, Amsterdam, location AMC, The Netherlands
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute,
University of Sydney, Sydney, Australia
| | - Manlio F Márquez
- Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de
México, Mexico
- Member of the Latin American Heart Rhythm Society (LAHRS)
| | | | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine,
and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm
Services and Pediatric Cardiology; Windland Smith Rice Genetic Heart Rhythm Clinic and
Windland Smith Rice Sudden Death Genomics Laboratory, Mayo
Clinic, Rochester, MN, USA
| | - Euan A Ashley
- Department of Cardiovascular Medicine, Stanford University,
Stanford, California, USA
| | - Eduardo Back Sternick
- Arrhythmia and Electrophysiology Unit, Biocor Institute,
Minas Gerais, Brazil; and
Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Héctor Barajas-Martinez
- Cardiovascular Research, Lankenau Institute of Medical
Research, Wynnewood, PA, USA; and Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Elijah R Behr
- Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical
Sciences, St. George’s, University of London; St. George’s University Hospitals NHS
Foundation Trust, London, UK; Mayo Clinic Healthcare, London
| | - Connie R Bezzina
- Amsterdam UMC Heart Center, Department of Experimental
Cardiology, Amsterdam, The
Netherlands
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven,
Leuven, Belgium
| | - Philippe Charron
- Sorbonne Université, APHP, Centre de Référence des Maladies Cardiaques
Héréditaires, ICAN, Inserm UMR1166, Hôpital
Pitié-Salpêtrière, Paris, France
| | | | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin,
Istituto Auxologico Italiano, IRCCS, Milan, Italy
- Cardiomyopathy Unit and Cardiac Rehabilitation Unit, San Luca Hospital,
Istituto Auxologico Italiano, IRCCS, Milan,
Italy
- Department of Medicine and Surgery, University of
Milano-Bicocca, Milan, Italy
| | - Michael H Gollob
- Inherited Arrhythmia and Cardiomyopathy Program, Division of Cardiology,
University of Toronto, Toronto, ON, Canada
| | - Steven Lubitz
- Cardiac Arrhythmia Service, Massachusetts General Hospital and Harvard
Medical School, Boston, MA, USA
| | - Naomasa Makita
- National Cerebral and Cardiovascular Center, Research
Institute, Suita, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular
Center, Suita, Japan
| | - Martín Ortiz-Genga
- Clinical Department, Health in Code, A
Coruña, Spain; and Member of the Latin
American Heart Rhythm Society (LAHRS)
| | - Luciana Sacilotto
- Arrhythmia Unit, Instituto do Coracao, Hospital das Clinicas HCFMUSP,
Faculdade de Medicina, Universidade de Sao Paulo, Sao
Paulo, Brazil; and Member of the Latin
American Heart Rhythm Society (LAHRS)
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases, University Hospital
Münster, Münster, Germany
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Nippon
Medical School, Bunkyo-ku, Tokyo, Japan
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of
Medicine, University of Washington, Seattle, WA,
USA
| | - Rafik Tadros
- Cardiovascular Genetics Center, Department of Medicine, Montreal Heart
Institute, Université de Montréal, Montreal,
Canada
| | - James S Ware
- National Heart and Lung Institute and MRC London Institute of Medical
Sciences, Imperial College London, London,
UK
- Royal Brompton & Harefield Hospitals, Guy’s
and St. Thomas’ NHS Foundation Trust, London, UK
| | - David S Winlaw
- Cincinnati Children's Hospital Medical Centre, University of
Cincinnati, Cincinnati, OH, USA
| | - Elizabeth S Kaufman
- Metrohealth Medical Center, Case Western Reserve University,
Cleveland, OH, USA
| |
Collapse
|
37
|
Wilde AAM, Semsarian C, Márquez MF, Sepehri Shamloo A, Ackerman MJ, Ashley EA, Sternick Eduardo B, Barajas‐Martinez H, Behr ER, Bezzina CR, Breckpot J, Charron P, Chockalingam P, Crotti L, Gollob MH, Lubitz S, Makita N, Ohno S, Ortiz‐Genga M, Sacilotto L, Schulze‐Bahr E, Shimizu W, Sotoodehnia N, Tadros R, Ware JS, Winlaw DS, Kaufman ES, Aiba T, Bollmann A, Choi J, Dalal A, Darrieux F, Giudicessi J, Guerchicoff M, Hong K, Krahn AD, Mac Intyre C, Mackall JA, Mont L, Napolitano C, Ochoa Juan P, Peichl P, Pereira AC, Schwartz PJ, Skinner J, Stellbrink C, Tfelt‐Hansen J, Deneke T. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the state of genetic testing for cardiac diseases. J Arrhythm 2022; 38:491-553. [PMID: 35936045 PMCID: PMC9347209 DOI: 10.1002/joa3.12717] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Arthur A. M. Wilde
- Heart Centre, Department of Cardiology, Amsterdam Universitair Medische CentraAmsterdamThe Netherlands
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary InstituteUniversity of SydneySydneyAustralia
| | - Manlio F. Márquez
- Instituto Nacional de Cardiología Ignacio ChávezCiudad de MéxicoMexico
| | | | - Michael J. Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm Services and Pediatric Cardiology; Windland Smith Rice Genetic Heart Rhythm Clinic and Windland Smith Rice Sudden Death Genomics Laboratory, Mayo ClinicRochesterMNUSA
| | - Euan A. Ashley
- Department of Cardiovascular MedicineStanford UniversityStanfordCAUSA
| | | | | | - Elijah R. Behr
- Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical Sciences, St. George’sUniversity of London; St. George’s University Hospitals NHS Foundation TrustLondonUKMayo Clinic HealthcareLondon
| | - Connie R. Bezzina
- Amsterdam UMC Heart Center, Department of Experimental CardiologyAmsterdamThe Netherlands
| | - Jeroen Breckpot
- Center for Human GeneticsUniversity Hospitals LeuvenLeuvenBelgium
| | | | | | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCSMilanItaly
- Cardiomyopathy Unit and Cardiac Rehabilitation Unit, San Luca Hospital, Istituto Auxologico Italiano, IRCCSMilanItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMilanItaly
| | - Michael H. Gollob
- Inherited Arrhythmia and Cardiomyopathy Program, Division of CardiologyUniversity of TorontoTorontoONCanada
| | - Steven Lubitz
- Cardiac Arrhythmia ServiceMassachusetts General Hospital and Harvard Medical SchoolBostonMAUSA
| | - Naomasa Makita
- National Cerebral and Cardiovascular CenterResearch InstituteSuitaJapan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular CenterSuitaJapan
| | | | - Luciana Sacilotto
- Arrhythmia Unit, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao PauloBrazil
| | - Eric Schulze‐Bahr
- Institute for Genetics of Heart DiseasesUniversity Hospital MünsterMünsterGermany
| | - Wataru Shimizu
- Department of Cardiovascular MedicineGraduate School of MedicineTokyoJapan
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of MedicineUniversity of WashingtonSeattleWAUSA
| | - Rafik Tadros
- Cardiovascular Genetics Center, Department of Medicine, Montreal Heart InstituteUniversité de MontréalMontrealCanada
| | - James S. Ware
- National Heart and Lung Institute and MRC London Institute of Medical SciencesImperial College LondonLondonUK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation TrustLondonUK
| | - David S. Winlaw
- Cincinnati Children's Hospital Medical CentreUniversity of CincinnatiCincinnatiOHUSA
| | | | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center, SuitaOsakaJapan
| | - Andreas Bollmann
- Department of ElectrophysiologyHeart Center Leipzig at University of LeipzigLeipzigGermany
- Leipzig Heart InstituteLeipzigGermany
| | - Jong‐Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Anam HospitalKorea University College of MedicineSeoulRepublic of Korea
| | - Aarti Dalal
- Department of Pediatrics, Division of CardiologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Francisco Darrieux
- Arrhythmia Unit, Instituto do Coração, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São PauloSão PauloBrazil
| | - John Giudicessi
- Department of Cardiovascular Medicine (Divisions of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo ClinicRochesterMNUSA
| | - Mariana Guerchicoff
- Division of Pediatric Arrhythmia and Electrophysiology, Italian Hospital of Buenos AiresBuenos AiresArgentina
| | - Kui Hong
- Department of Cardiovascular MedicineThe Second Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Andrew D. Krahn
- Division of CardiologyUniversity of British ColumbiaVancouverCanada
| | - Ciorsti Mac Intyre
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo ClinicRochesterMNUSA
| | - Judith A. Mackall
- Center for Cardiac Electrophysiology and Pacing, University Hospitals Cleveland Medical CenterCase Western Reserve University School of MedicineClevelandOHUSA
| | - Lluís Mont
- Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS). Barcelona, Spain; Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), MadridSpain
| | - Carlo Napolitano
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCSPaviaItaly
- Department of Molecular MedicineUniversity of PaviaPaviaItaly
| | - Pablo Ochoa Juan
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), MadridSpain
- Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de HierroMadridSpain
- Centro de Investigacion Biomedica en Red en Enfermedades Cariovasculares (CIBERCV), MadridSpain
| | - Petr Peichl
- Department of CardiologyInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart InstituteUniversity of São Paulo Medical SchoolSão PauloBrazil
- Hipercol Brasil ProgramSão PauloBrazil
| | - Peter J. Schwartz
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCSMilanItaly
| | - Jon Skinner
- Sydney Childrens Hospital NetworkUniversity of SydneySydneyAustralia
| | - Christoph Stellbrink
- Department of Cardiology and Intensive Care MedicineUniversity Hospital Campus Klinikum BielefeldBielefeldGermany
| | - Jacob Tfelt‐Hansen
- The Department of Cardiology, the Heart Centre, Copenhagen University Hospital, Rigshopitalet, Copenhagen, Denmark; Section of genetics, Department of Forensic Medicine, Faculty of Medical SciencesUniversity of CopenhagenDenmark
| | - Thomas Deneke
- Heart Center Bad NeustadtBad Neustadt a.d. SaaleGermany
| |
Collapse
|
38
|
Velardo D, D'Angelo MG, Citterio A, Panzeri E, Napoli L, Cinnante C, Moggio M, Comi GP, Ronchi D, Bassi MT. Case Reports: Novel Missense Variants in the Filamin C Actin Binding Domain Cause Variable Phenotypes. Front Neurol 2022; 13:930039. [PMID: 35903116 PMCID: PMC9315448 DOI: 10.3389/fneur.2022.930039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/22/2022] [Indexed: 11/24/2022] Open
Abstract
Filamin C is a large dimeric actin-binding protein, most prevalent in skeletal and cardiac muscle Z-discs, where it participates in sarcomere mechanical stabilization and intracellular signaling, interacting with numerous binding partners. Dominant heterozygous mutations of Filamin C gene cause several forms of myopathy and structural or arrhythmogenic cardiomyopathy. In this report we describe clinical and molecular findings of two Italian patients, in whom we identified two novel missense variants located within the Filamin C actin binding domain. Muscle imaging, histological and ultrastructural findings are also reported. Our results underline the extreme inter- and intrafamilial variability of clinical manifestations, hence the need to extend the investigation also to asymptomatic relatives, and the relevance of a broad diagnostic approach involving muscle electron microscopy, skeletal muscle magnetic resonance imaging and next generation sequencing techniques.
Collapse
Affiliation(s)
- Daniele Velardo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
- *Correspondence: Daniele Velardo
| | - Maria Grazia D'Angelo
- NeuroMuscular Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Andrea Citterio
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Elena Panzeri
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Laura Napoli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Claudia Cinnante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuroradiology Unit, Milan, Italy
- Department of Radiology, Istituto Auxologico Italiano, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Milan, Italy
| | - Maurizio Moggio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Giacomo Pietro Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Neuroscience Section, University of Milan, Milan, Italy
| | - Dario Ronchi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Neuroscience Section, University of Milan, Milan, Italy
| | - Maria Teresa Bassi
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| |
Collapse
|
39
|
Wilde AAM, Semsarian C, Márquez MF, Sepehri Shamloo A, Ackerman MJ, Ashley EA, Sternick EB, Barajas-Martinez H, Behr ER, Bezzina CR, Breckpot J, Charron P, Chockalingam P, Crotti L, Gollob MH, Lubitz S, Makita N, Ohno S, Ortiz-Genga M, Sacilotto L, Schulze-Bahr E, Shimizu W, Sotoodehnia N, Tadros R, Ware JS, Winlaw DS, Kaufman ES, Aiba T, Bollmann A, Choi JI, Dalal A, Darrieux F, Giudicessi J, Guerchicoff M, Hong K, Krahn AD, MacIntyre C, Mackall JA, Mont L, Napolitano C, Ochoa JP, Peichl P, Pereira AC, Schwartz PJ, Skinner J, Stellbrink C, Tfelt-Hansen J, Deneke T. European Heart Rhythm Association (EHRA)/Heart Rhythm Society (HRS)/Asia Pacific Heart Rhythm Society (APHRS)/Latin American Heart Rhythm Society (LAHRS) Expert Consensus Statement on the State of Genetic Testing for Cardiac Diseases. Heart Rhythm 2022; 19:e1-e60. [PMID: 35390533 DOI: 10.1016/j.hrthm.2022.03.1225] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 03/25/2022] [Indexed: 12/12/2022]
Affiliation(s)
- Arthur A M Wilde
- Heart Centre, Department of Cardiology, Amsterdam Universitair Medische Centra, Amsterdam, location AMC, The Netherlands.
| | - Christopher Semsarian
- Agnes Ginges Centre for Molecular Cardiology at Centenary Institute, University of Sydney, Sydney, Australia.
| | - Manlio F Márquez
- Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico; and Member of the Latin American Heart Rhythm Society (LAHRS).
| | | | - Michael J Ackerman
- Departments of Cardiovascular Medicine, Pediatric and Adolescent Medicine, and Molecular Pharmacology & Experimental Therapeutics; Divisions of Heart Rhythm Services and Pediatric Cardiology; Windland Smith Rice Genetic Heart Rhythm Clinic and Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN, USA
| | - Euan A Ashley
- Department of Cardiovascular Medicine, Stanford University, Stanford, CA, USA
| | - Eduardo Back Sternick
- Arrhythmia and Electrophysiology Unit, Biocor Institute, Minas Gerais, Brazil; and Member of the Latin American Heart Rhythm Society (LAHRS)
| | | | - Elijah R Behr
- Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical Sciences, St. George's, University of London; St. George's University Hospitals NHS Foundation Trust, London, UK; Mayo Clinic Healthcare, London
| | - Connie R Bezzina
- Amsterdam UMC Heart Center, Department of Experimental Cardiology, Amsterdam, The Netherlands
| | - Jeroen Breckpot
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Charron
- Sorbonne Université, APHP, Centre de Référence des Maladies Cardiaques Héréditaires, ICAN, Inserm UMR1166, Hôpital Pitié-Salpêtrière, Paris, France
| | | | - Lia Crotti
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy; Cardiomyopathy Unit and Cardiac Rehabilitation Unit, San Luca Hospital, Istituto Auxologico Italiano, IRCCS, Milan, Italy; Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Michael H Gollob
- Inherited Arrhythmia and Cardiomyopathy Program, Division of Cardiology, University of Toronto, Toronto, ON, Canada
| | - Steven Lubitz
- Cardiac Arrhythmia Service, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Naomasa Makita
- National Cerebral and Cardiovascular Center, Research Institute, Suita, Japan
| | - Seiko Ohno
- Department of Bioscience and Genetics, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Martín Ortiz-Genga
- Clinical Department, Health in Code, A Coruña, Spain; and Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Luciana Sacilotto
- Arrhythmia Unit, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil; and Member of the Latin American Heart Rhythm Society (LAHRS)
| | - Eric Schulze-Bahr
- Institute for Genetics of Heart Diseases, University Hospital Münster, Münster, Germany
| | - Wataru Shimizu
- Department of Cardiovascular Medicine, Graduate School of Medicine, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Rafik Tadros
- Cardiovascular Genetics Center, Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Canada
| | - James S Ware
- National Heart and Lung Institute and MRC London Institute of Medical Sciences, Imperial College London, London, UK; Royal Brompton & Harefield Hospitals, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - David S Winlaw
- Cincinnati Children's Hospital Medical Centre, University of Cincinnati, Cincinnati, OH, USA
| | - Elizabeth S Kaufman
- Metrohealth Medical Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Takeshi Aiba
- Department of Clinical Laboratory Medicine and Genetics, National Cerebral and Cardiovascular Center, Suita, Osaka, Japan
| | - Andreas Bollmann
- Department of Electrophysiology, Heart Center Leipzig at University of Leipzig, Leipzig, Germany; Leipzig Heart Institute, Leipzig Heart Digital, Leipzig, Germany
| | - Jong-Il Choi
- Division of Cardiology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Aarti Dalal
- Department of Pediatrics, Division of Cardiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Francisco Darrieux
- Arrhythmia Unit, Instituto do Coração, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - John Giudicessi
- Department of Cardiovascular Medicine (Divisions of Heart Rhythm Services and Circulatory Failure and the Windland Smith Rice Genetic Heart Rhythm Clinic), Mayo Clinic, Rochester, MN, USA
| | - Mariana Guerchicoff
- Division of Pediatric Arrhythmia and Electrophysiology, Italian Hospital of Buenos Aires, Buenos Aires, Argentina
| | - Kui Hong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Andrew D Krahn
- Division of Cardiology, University of British Columbia, Vancouver, Canada
| | - Ciorsti MacIntyre
- Department of Cardiovascular Medicine, Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Mayo Clinic, Rochester, MN, USA
| | - Judith A Mackall
- Center for Cardiac Electrophysiology and Pacing, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Lluís Mont
- Institut d'Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigacion Biomedica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Carlo Napolitano
- Molecular Cardiology, Istituti Clinici Scientifici Maugeri, IRCCS, Pavia, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Juan Pablo Ochoa
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Heart Failure and Inherited Cardiac Diseases Unit, Department of Cardiology, Hospital Universitario Puerta de Hierro, Madrid, Spain; Centro de Investigacion Biomedica en Red en Enfermedades Cariovasculares (CIBERCV), Madrid, Spain
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo 05403-000, Brazil; Hipercol Brasil Program, São Paulo, Brazil
| | - Peter J Schwartz
- Center for Cardiac Arrhythmias of Genetic Origin, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Jon Skinner
- Sydney Childrens Hospital Network, University of Sydney, Sydney, Australia
| | - Christoph Stellbrink
- Department of Cardiology and Intensive Care Medicine, University Hospital Campus Klinikum Bielefeld, Bielefeld, Germany
| | - Jacob Tfelt-Hansen
- The Department of Cardiology, the Heart Centre, Copenhagen University Hospital, Rigshopitalet, Copenhagen, Denmark; Section of Genetics, Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Denmark
| | - Thomas Deneke
- Heart Center Bad Neustadt, Bad Neustadt a.d. Saale, Germany
| |
Collapse
|
40
|
Huang C, Zheng Y, Zhang W, Chen Z, Huang Z, Fang Y. Case Report: A Chinese Family of Hypertrophic Cardiomyopathy Caused by a Novel Splicing Mutation in the FLNC Gene. Front Genet 2022; 13:894791. [PMID: 35795207 PMCID: PMC9251305 DOI: 10.3389/fgene.2022.894791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a type of primary cardiomyopathy with genetic etiology, and it carries a high risk of diastolic dysfunction, heart failure, and malignant arrhythmias. We reported the first familial HCM in China, caused by a novel FLNC splicing mutation. We performed duo exome sequencing (ES) to examine the genome of the proband and his mother. For 10 days, a 15-year-old boy was presented to our hospital due to non–exercise-associated chest tightness and asthma. He was diagnosed with HCM [end-diastolic interventricular septal thickness was about 18 mm by transthoracic echocardiography (TTE)]. His mother and sister performed TTE to screen familial cardiomyopathy, which revealed hypertrophic cardiomyopathy only in the proband’s mother. In ES of the mother–son duo, we identified a novel heterozygous mutation of the FLNC gene (chr7:128492808, NM_001127487, c.5905+2T>C, rs1808874360) as the candidate cause of autosomal dominant HCM. Sanger sequencing confirmed this novel mutation in the proband and his mother but absent in the proband’s sister. The potential impact of the novel mutation was predicted by MutationTaster, dbscSNV_ADA_SCORE, dbscSNV_RF_SCORE, CADD_phred, PhyloP20way_mammalian, PhyloP100way_vertebrate, SiPhy_29way_logOdds, and GERP++_RS software. After the administration of furosemide, spironolactone, and metoprolol, the proband’s heart function was improved, and symptoms were alleviated. We presented the first familial HCM caused by a novel FLNC splicing mutation via exome sequencing in China. Therefore, it is necessary that familial screening for patients with HCM should be performed for the early detection of HCM intervention in malignant cardiac events in advance and block genes.
Collapse
Affiliation(s)
- Chunhui Huang
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Yonghong Zheng
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Wei Zhang
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Zhigang Chen
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Zhixin Huang
- Department of Ultrasound Medicine, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Yuan Fang
- Department of Geriatric Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Yuan Fang,
| |
Collapse
|
41
|
Qian C, Xiufu W, Jianxun T, Zihao C, Wenjie S, Jingfeng T, Kahlert UD, Renfei D. A Novel Extracellular Matrix Gene-Based Prognostic Model to Predict Overall Survive in Patients With Glioblastoma. Front Genet 2022; 13:851427. [PMID: 35783254 PMCID: PMC9247148 DOI: 10.3389/fgene.2022.851427] [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: 01/09/2022] [Accepted: 03/30/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Glioblastoma (GBM), one of the most prevalent brain tumor types, is correlated with an extremely poor prognosis. The extracellular matrix (ECM) genes could activate many crucial pathways that facilitate tumor development. This study aims to provide online models to predict GBM survival by ECM genes. Methods: The associations of ECM genes with the prognosis of GBM were analyzed, and the significant prognosis-related genes were used to develop the ECM index in the CGGA dataset. Furthermore, the ECM index was then validated on three datasets, namely, GSE16011, TCGA-GBM, and GSE83300. The prognosis difference, differentially expressed genes, and potential drugs were obtained. Multiple machine learning methods were selected to construct the model to predict the survival status of GBM patients at 6, 12, 18, 24, 30, and 36 months after diagnosis. Results: Five ECM gene signatures (AEBP1, F3, FLNC, IGFBP2, and LDHA) were recognized to be associated with the prognosis. GBM patients were divided into high- and low-ECM index groups with significantly different overall survival rates in four datasets. High-ECM index patients exhibited a worse prognosis than low-ECM index patients. Four small molecules (podophyllotoxin, lasalocid, MG-262, and nystatin) that might reduce GBM development were predicted by the Cmap dataset. In the independent dataset (GSE83300), the maximum values of prediction accuracy at 6, 12, 18, 24, 30, and 36 months were 0.878, 0.769, 0.748, 0.720, 0.705, and 0.868, respectively. These machine learning models were provided on a publicly accessible, open-source website (https://ospg.shinyapps.io/OSPG/). Conclusion: In summary, our findings indicated that ECM genes were prognostic indicators for patient survival. This study provided an online server for the prediction of survival curves of GBM patients.
Collapse
Affiliation(s)
- Chen Qian
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wu Xiufu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Tang Jianxun
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Chen Zihao
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Shi Wenjie
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
- Molecular and Experimental Surgery, Medical Faculty University Hospital Magdeburg, University Clinic for General-, Visceral-, Vascular- and Trans-Plantation Surgery, Otto-von Guericke University, Magdeburg, Germany
| | - Tang Jingfeng
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Ulf D. Kahlert
- Molecular and Experimental Surgery, Medical Faculty University Hospital Magdeburg, University Clinic for General-, Visceral-, Vascular- and Trans-Plantation Surgery, Otto-von Guericke University, Magdeburg, Germany
| | - Du Renfei
- Clinic of Neurosurgery, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
- Chifeng Municipal Hospital, Inner Mongolia, Chifeng, China
| |
Collapse
|
42
|
Lukas Laws J, Lancaster MC, Ben Shoemaker M, Stevenson WG, Hung RR, Wells Q, Marshall Brinkley D, Hughes S, Anderson K, Roden D, Stevenson LW. Arrhythmias as Presentation of Genetic Cardiomyopathy. Circ Res 2022; 130:1698-1722. [PMID: 35617362 PMCID: PMC9205615 DOI: 10.1161/circresaha.122.319835] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
There is increasing evidence regarding the prevalence of genetic cardiomyopathies, for which arrhythmias may be the first presentation. Ventricular and atrial arrhythmias presenting in the absence of known myocardial disease are often labelled as idiopathic, or lone. While ventricular arrhythmias are well-recognized as presentation for arrhythmogenic cardiomyopathy in the right ventricle, the scope of arrhythmogenic cardiomyopathy has broadened to include those with dominant left ventricular involvement, usually with a phenotype of dilated cardiomyopathy. In addition, careful evaluation for genetic cardiomyopathy is also warranted for patients presenting with frequent premature ventricular contractions, conduction system disease, and early onset atrial fibrillation, in which most detected genes are in the cardiomyopathy panels. Sudden death can occur early in the course of these genetic cardiomyopathies, for which risk is not adequately tracked by left ventricular ejection fraction. Only a few of the cardiomyopathy genotypes implicated in early sudden death are recognized in current indications for implantable cardioverter defibrillators which otherwise rely upon a left ventricular ejection fraction ≤0.35 in dilated cardiomyopathy. The genetic diagnoses impact other aspects of clinical management such as exercise prescription and pharmacological therapy of arrhythmias, and new therapies are coming into clinical investigation for specific genetic cardiomyopathies. The expansion of available genetic information and implications raises new challenges for genetic counseling, particularly with the family member who has no evidence of a cardiomyopathy phenotype and may face a potentially negative impact of a genetic diagnosis. Discussions of risk for both probands and relatives need to be tailored to their numeric literacy during shared decision-making. For patients presenting with arrhythmias or cardiomyopathy, extension of genetic testing and its implications will enable cascade screening, intervention to change the trajectory for specific genotype-phenotype profiles, and enable further development and evaluation of emerging targeted therapies.
Collapse
Affiliation(s)
- J Lukas Laws
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Megan C Lancaster
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - M Ben Shoemaker
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - William G Stevenson
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Rebecca R Hung
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Quinn Wells
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - D Marshall Brinkley
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Sean Hughes
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Katherine Anderson
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Dan Roden
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| | - Lynne W Stevenson
- Division of Cardiovascular Medicine, Vanderbilt Heart and Vascular Institute, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
43
|
Genetic Insights into Primary Restrictive Cardiomyopathy. J Clin Med 2022; 11:jcm11082094. [PMID: 35456187 PMCID: PMC9027761 DOI: 10.3390/jcm11082094] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Restrictive cardiomyopathy is a rare cardiac disease causing severe diastolic dysfunction, ventricular stiffness and dilated atria. In consequence, it induces heart failure often with preserved ejection fraction and is associated with a high mortality. Since it is a poor clinical prognosis, patients with restrictive cardiomyopathy frequently require heart transplantation. Genetic as well as non-genetic factors contribute to restrictive cardiomyopathy and a significant portion of cases are of unknown etiology. However, the genetic forms of restrictive cardiomyopathy and the involved molecular pathomechanisms are only partially understood. In this review, we summarize the current knowledge about primary genetic restrictive cardiomyopathy and describe its genetic landscape, which might be of interest for geneticists as well as for cardiologists.
Collapse
|
44
|
Chintanaphol M, Orgil BO, Alberson NR, Towbin JA, Purevjav E. Restrictive cardiomyopathy: from genetics and clinical overview to animal modeling. Rev Cardiovasc Med 2022; 23:108. [PMID: 35345275 DOI: 10.31083/j.rcm2303108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 10/08/2021] [Accepted: 10/25/2021] [Indexed: 11/06/2022] Open
Abstract
Restrictive cardiomyopathy (RCM), a potentially devastating heart muscle disorder, is characterized by diastolic dysfunction due to abnormal muscle relaxation and myocardial stiffness resulting in restrictive filling of the ventricles. Diastolic dysfunction is often accompanied by left atrial or bi-atrial enlargement and normal ventricular size and systolic function. RCM is the rarest form of cardiomyopathy, accounting for 2-5% of pediatric cardiomyopathy cases, however, survival rates have been reported to be 82%, 80%, and 68% at 1-, 2-, and 5-years after diagnosis, respectively. RCM can be idiopathic, familial, or secondary to a systemic disorder, such as amyloidosis, sarcoidosis, and hereditary hemochromatosis. Approximately 30% of cases are familial RCM, and the genes that have been linked to RCM are cTnT, cTnI, MyBP-C, MYH7, MYL2, MYL3, DES, MYPN, TTN, BAG3, DCBLD2, LNMA, and FLNC. Increased Ca2+ sensitivity, sarcomere disruption, and protein aggregates are some of the few mechanisms of pathogenesis that have been revealed by studies utilizing cell lines and animal models. Additional exploration into the pathogenesis of RCM is necessary to create novel therapeutic strategies to reverse restrictive cardiomyopathic phenotypes.
Collapse
Affiliation(s)
- Michelle Chintanaphol
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Buyan-Ochir Orgil
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Neely R Alberson
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Jeffrey A Towbin
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Enkhsaikhan Purevjav
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| |
Collapse
|
45
|
Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol 2022; 19:151-167. [PMID: 34526680 DOI: 10.1038/s41569-021-00608-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2021] [Indexed: 01/06/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) was traditionally described as an autosomal dominant Mendelian disease but is now increasingly recognized as having a complex genetic aetiology. Although eight core genes encoding sarcomeric proteins account for >90% of the pathogenic variants in patients with HCM, variants in several additional genes (ACTN2, ALPK3, CSRP3, FHOD3, FLNC, JPH2, KLHL24, PLN and TRIM63), encoding non-sarcomeric proteins with diverse functions, have been shown to be disease-causing in a small number of patients. Genome-wide association studies (GWAS) have identified numerous loci in cardiomyopathy case-control studies and biobank investigations of left ventricular functional traits. Genes associated with Mendelian cardiomyopathy are enriched in the putative causal gene lists at these loci. Intriguingly, many loci are associated with both HCM and dilated cardiomyopathy but with opposite directions of effect on left ventricular traits, highlighting a genetic basis underlying the contrasting pathophysiological effects observed in each condition. This overlap extends to rare Mendelian variants with distinct variant classes in several genes associated with HCM and dilated cardiomyopathy. In this Review, we appraise the complex contribution of the non-sarcomeric, HCM-associated genes to cardiomyopathies across a range of variant classes (from common non-coding variants of individually low effect size to complete gene knockouts), which provides insights into the genetic basis of cardiomyopathies, causal genes at GWAS loci and the application of clinical genetic testing.
Collapse
|
46
|
Suay-Corredera C, Alegre-Cebollada J. The mechanics of the heart: zooming in on hypertrophic cardiomyopathy and cMyBP-C. FEBS Lett 2022; 596:703-746. [PMID: 35224729 DOI: 10.1002/1873-3468.14301] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 11/10/2022]
Abstract
Hypertrophic cardiomyopathy (HCM), a disease characterized by cardiac muscle hypertrophy and hypercontractility, is the most frequently inherited disorder of the heart. HCM is mainly caused by variants in genes encoding proteins of the sarcomere, the basic contractile unit of cardiomyocytes. The most frequently mutated among them is MYBPC3, which encodes cardiac myosin-binding protein C (cMyBP-C), a key regulator of sarcomere contraction. In this review, we summarize clinical and genetic aspects of HCM and provide updated information on the function of the healthy and HCM sarcomere, as well as on emerging therapeutic options targeting sarcomere mechanical activity. Building on what is known about cMyBP-C activity, we examine different pathogenicity drivers by which MYBPC3 variants can cause disease, focussing on protein haploinsufficiency as a common pathomechanism also in nontruncating variants. Finally, we discuss recent evidence correlating altered cMyBP-C mechanical properties with HCM development.
Collapse
|
47
|
Chen SN, Lam CK, Wan YW, Gao S, Malak OA, Zhao SR, Lombardi R, Ambardekar AV, Bristow MR, Cleveland J, Gigli M, Sinagra G, Graw S, Taylor MR, Wu JC, Mestroni L. Activation of PDGFRA signaling contributes to filamin C-related arrhythmogenic cardiomyopathy. SCIENCE ADVANCES 2022; 8:eabk0052. [PMID: 35196083 PMCID: PMC8865769 DOI: 10.1126/sciadv.abk0052] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/25/2021] [Indexed: 05/07/2023]
Abstract
FLNC truncating mutations (FLNCtv) are prevalent causes of inherited dilated cardiomyopathy (DCM), with a high risk of developing arrhythmogenic cardiomyopathy. We investigated the molecular mechanisms of mutant FLNC in the pathogenesis of arrhythmogenic DCM (a-DCM) using patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs). We demonstrated that iPSC-CMs from two patients with different FLNCtv mutations displayed arrhythmias and impaired contraction. FLNC ablation induced a similar phenotype, suggesting that FLNCtv are loss-of-function mutations. Coimmunoprecipitation and proteomic analysis identified β-catenin (CTNNB1) as a downstream target. FLNC deficiency induced nuclear translocation of CTNNB1 and subsequently activated the platelet-derived growth factor receptor alpha (PDGFRA) pathway, which were also observed in human hearts with a-DCM and FLNCtv. Treatment with the PDGFRA inhibitor, crenolanib, improved contractile function of patient iPSC-CMs. Collectively, our findings suggest that PDGFRA signaling is implicated in the pathogenesis, and inhibition of this pathway is a potential therapeutic strategy in FLNC-related cardiomyopathies.
Collapse
Affiliation(s)
- Suet Nee Chen
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, USA
| | - Ying-Wooi Wan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Shanshan Gao
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Olfat A. Malak
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Shane Rui Zhao
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Raffaella Lombardi
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
- Department of Advanced Biomedical Sciences University of Naples “Federico II”, Naples, Italy
| | - Amrut V. Ambardekar
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Michael R. Bristow
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Joseph Cleveland
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Marta Gigli
- Cardiovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular Department, Azienda Sanitaria-Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Sharon Graw
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Matthew R.G. Taylor
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Luisa Mestroni
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Aurora, CO, USA
| |
Collapse
|
48
|
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
|
49
|
Abstract
PURPOSE OF REVIEW Hypertrophic cardiomyopathy (HCM) is one of the leading causes of sudden cardiac death (SCD) in younger people and athletes. It is crucial to identify the risk factors for SCD in individuals with HCM. This review, based on recent systematic literature studies, will focus on the risk factors for SCD in patients with HCM. RECENT FINDINGS An increasing number of studies have further explored the risk factors for SCD in patients with HCM, and new risk markers have emerged accordingly. In addition, more accurate SCD risk estimation and stratification methods have been proposed and continuously improved. SUMMARY The identification of independent risk factors for HCM-related SCD would likely contribute to risk stratification. However, it is difficult to predict SCD with absolute certainty, as the annual incidence of SCD in adult patients with HCM is approximately 1%. The review discusses the established risk factors, such as a family history of SCD, unexplained syncope and some new risk factors. Taken together, the findings of this review demonstrate that there is a need for further research on individual risk factors and that SCD risk stratification in HCM patients remains a clinical challenge.
Collapse
Affiliation(s)
- Ying Hong
- Department of Cardiology, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| | - Wilber W. Su
- Department of Cardiology, Banner-University Medical Center, Phoenix, Arizona, USA
| | - Xiaoping Li
- Department of Cardiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan, China
| |
Collapse
|
50
|
Matsumura T, Inoue K, Toyooka K, Inoue M, Iida A, Saito Y, Nishikawa T, Moriuchi K, Beck G, Nishino I, Fujimura H. Clinical trajectory of a patient with filaminopathy who developed arrhythmogenic cardiomyopathy, myofibrillar myopathy, and multiorgan tumors. Neuromuscul Disord 2021; 31:1282-1286. [PMID: 34857437 DOI: 10.1016/j.nmd.2021.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/05/2021] [Accepted: 10/05/2021] [Indexed: 11/30/2022]
Abstract
We report a case of a patient presenting with arrhythmogenic cardiomyopathy, myofibrillar myopathy, and multiorgan tumors. A 41-year-old woman with a history of hypertrophic cardiomyopathy, diagnosed at 6 years of age, developed scoliosis after puberty. Following spinal surgery to address the scoliosis, she developed recurrent severe arrhythmia and heart failure. She developed hypoventilation at age 29 years. Proximal dominant weakness and mild elevation of serum creatine kinase indicated possible myopathy. Myofibrillar myopathy was diagnosed by muscle biopsy at age 30 year. Acute abdomen was repeatedly reported from age 33 years, eventually leading to a diagnosis of gastric polyp and erosive ulcer. A urinary bladder tumor was found at age 35 years, and breast cancer was diagnosed at age 40 years. Whole exome sequencing detected a heterozygous missense mutation in Filamin C. Recent evidences suggest that filamins are associated with tumors, and this case further highlights the clinical spectrum of filaminopathy.
Collapse
Affiliation(s)
- Tsuyoshi Matsumura
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, Toneyama 5-1-1, Toyonaka, Osaka 560-8552, Japan.
| | - Kimiko Inoue
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, Toneyama 5-1-1, Toyonaka, Osaka 560-8552, Japan
| | - Keiko Toyooka
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, Toneyama 5-1-1, Toyonaka, Osaka 560-8552, Japan
| | - Michio Inoue
- Department of Neuromuscular Research, National Center of Neurology, Psychiatry, Ogawahigashi 4-1-1, Kodaira, Tokyo 187-8551, Japan
| | - Aritoshi Iida
- Department of Neuromuscular Research, National Center of Neurology, Psychiatry, Ogawahigashi 4-1-1, Kodaira, Tokyo 187-8551, Japan
| | - Yoshihiko Saito
- Department of Neuromuscular Research, National Center of Neurology, Psychiatry, Ogawahigashi 4-1-1, Kodaira, Tokyo 187-8551, Japan
| | - Tatsuya Nishikawa
- Department of Onco-Cardiology, Osaka International Cancer Institute, Otemae 3-1-69, Chuo-ku, Osaka City, Osaka, 541-8567, Japan
| | - Kenji Moriuchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, Kishibe-Shinmachi 6-1, Suita, Osaka 564-8565, Japan
| | - Goichi Beck
- Department of Neurology, Osaka University Graduate School of Medicine, Yamadaoka 2-2, Suita, Osaka 565-0871, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Center of Neurology, Psychiatry, Ogawahigashi 4-1-1, Kodaira, Tokyo 187-8551, Japan
| | - Harutoshi Fujimura
- Department of Neurology, National Hospital Organization Osaka Toneyama Medical Center, Toneyama 5-1-1, Toyonaka, Osaka 560-8552, Japan
| |
Collapse
|