1
|
Zuela-Sopilniak N, Morival J, Lammerding J. Multi-level transcriptomic analysis of LMNA -related dilated cardiomyopathy identifies disease-driving processes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598511. [PMID: 38915720 PMCID: PMC11195185 DOI: 10.1101/2024.06.11.598511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
LMNA- related dilated cardiomyopathy ( LMNA -DCM) is one of the most severe forms of DCM. The incomplete understanding of the molecular disease mechanisms results in lacking treatment options, leading to high mortality amongst patients. Here, using an inducible, cardiomyocyte-specific lamin A/C depletion mouse model, we conducted a comprehensive transcriptomic study, combining both bulk and single nucleus RNA sequencing, and spanning LMNA -DCM disease progression, to identify potential disease drivers. Our refined analysis pipeline identified 496 genes already misregulated early in disease. The expression of these genes was largely driven by disease specific cardiomyocyte sub-populations and involved biological processes mediating cellular response to DNA damage, cytosolic pattern recognition, and innate immunity. Indeed, DNA damage in LMNA -DCM hearts was significantly increased early in disease and correlated with reduced cardiomyocyte lamin A levels. Activation of cytosolic pattern recognition in cardiomyocytes was independent of cGAS, which is rarely expressed in cardiomyocytes, but likely occurred downstream of other pattern recognition sensors such as IFI16. Altered gene expression in cardiac fibroblasts and immune cell infiltration further contributed to tissue-wide changes in gene expression. Our transcriptomic analysis further predicted significant alterations in cell-cell communication between cardiomyocytes, fibroblasts, and immune cells, mediated through early changes in the extracellular matrix (ECM) in the LMNA -DCM hearts. Taken together, our work suggests a model in which nuclear damage in cardiomyocytes leads to activation of DNA damage responses, cytosolic pattern recognition pathway, and other signaling pathways that activate inflammation, immune cell recruitment, and transcriptional changes in cardiac fibroblasts, which collectively drive LMNA -DCM pathogenesis.
Collapse
|
2
|
RBM20-Related Cardiomyopathy: Current Understanding and Future Options. J Clin Med 2021; 10:jcm10184101. [PMID: 34575212 PMCID: PMC8468976 DOI: 10.3390/jcm10184101] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/13/2022] Open
Abstract
Splice regulators play an essential role in the transcriptomic diversity of all eukaryotic cell types and organ systems. Recent evidence suggests a contribution of splice-regulatory networks in many diseases, such as cardiomyopathies. Adaptive splice regulators, such as RNA-binding motif protein 20 (RBM20) determine the physiological mRNA landscape formation, and rare variants in the RBM20 gene explain up to 6% of genetic dilated cardiomyopathy (DCM) cases. With ample knowledge from RBM20-deficient mice, rats, swine and induced pluripotent stem cells (iPSCs), the downstream targets and quantitative effects on splicing are now well-defined and the prerequisites for corrective therapeutic approaches are set. This review article highlights some of the recent advances in the field, ranging from aspects of granule formation to 3D genome architectures underlying RBM20-related cardiomyopathy. Promising therapeutic strategies are presented and put into context with the pathophysiological characteristics of RBM20-related diseases.
Collapse
|
3
|
Sayed N, Liu C, Ameen M, Himmati F, Zhang JZ, Khanamiri S, Moonen JR, Wnorowski A, Cheng L, Rhee JW, Gaddam S, Wang KC, Sallam K, Boyd JH, Woo YJ, Rabinovitch M, Wu JC. Clinical trial in a dish using iPSCs shows lovastatin improves endothelial dysfunction and cellular cross-talk in LMNA cardiomyopathy. Sci Transl Med 2021; 12:12/554/eaax9276. [PMID: 32727917 DOI: 10.1126/scitranslmed.aax9276] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 02/13/2020] [Accepted: 07/09/2020] [Indexed: 12/15/2022]
Abstract
Mutations in LMNA, the gene that encodes lamin A and C, causes LMNA-related dilated cardiomyopathy (DCM) or cardiolaminopathy. LMNA is expressed in endothelial cells (ECs); however, little is known about the EC-specific phenotype of LMNA-related DCM. Here, we studied a family affected by DCM due to a frameshift variant in LMNA Human induced pluripotent stem cell (iPSC)-derived ECs were generated from patients with LMNA-related DCM and phenotypically characterized. Patients with LMNA-related DCM exhibited clinical endothelial dysfunction, and their iPSC-ECs showed decreased functionality as seen by impaired angiogenesis and nitric oxide (NO) production. Moreover, genome-edited isogenic iPSC lines recapitulated the EC disease phenotype in which LMNA-corrected iPSC-ECs showed restoration of EC function. Simultaneous profiling of chromatin accessibility and gene expression dynamics by combining assay for transposase-accessible chromatin using sequencing (ATAC-seq) and RNA sequencing (RNA-seq) as well as loss-of-function studies identified Krüppel-like factor 2 (KLF2) as a potential transcription factor responsible for the EC dysfunction. Gain-of-function studies showed that treatment of LMNA iPSC-ECs with KLF2 agonists, including lovastatin, rescued the EC dysfunction. Patients with LMNA-related DCM treated with lovastatin showed improvements in clinical endothelial dysfunction as indicated by increased reactive hyperemia index. Furthermore, iPSC-derived cardiomyocytes (iPSC-CMs) from patients exhibiting the DCM phenotype showed improvement in CM function when cocultured with iPSC-ECs and lovastatin. These results suggest that impaired cross-talk between ECs and CMs can contribute to the pathogenesis of LMNA-related DCM, and statin may be an effective therapy for vascular dysfunction in patients with cardiolaminopathy.
Collapse
Affiliation(s)
- Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mohamed Ameen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Farhan Himmati
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joe Z Zhang
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Saereh Khanamiri
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jan-Renier Moonen
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexa Wnorowski
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Bioengineering, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Linling Cheng
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sadhana Gaddam
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin C Wang
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karim Sallam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jack H Boyd
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Y Joseph Woo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Cardiothoracic Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Marlene Rabinovitch
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA.,Vera Moulton Wall Center for Pulmonary Vascular Disease, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Yeung C, Enriquez A, Suarez-Fuster L, Baranchuk A. Atrial fibrillation in patients with inherited cardiomyopathies. Europace 2020; 21:22-32. [PMID: 29684120 DOI: 10.1093/europace/euy064] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/13/2018] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) often complicates the course of inherited cardiomyopathies and, in some cases, may be the presenting feature. Each inherited cardiomyopathy has its own peculiar pathogenetic characteristics that can contribute to the development and maintenance of AF. Atrial fibrillation may occur as a consequence of disease-specific defects, non-specific cardiac chamber changes secondary to the primary illness, or a combination thereof. The presence of AF can denote a turning point in the progression of the disease, promoting clinical deterioration and increasing morbidity and mortality. Furthermore, the management of AF can be particularly challenging in patients with inherited cardiomyopathies. In this article, we review the current information on the prevalence, pathophysiology, risk factors, and treatment of AF in three different inherited cardiomyopathies: hypertrophic cardiomyopathy, arrhythmogenic right ventricular dysplasia/cardiomyopathy, familial dilated cardiomyopathy, and left ventricular non-compaction cardiomyopathy.
Collapse
Affiliation(s)
- Cynthia Yeung
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | - Andres Enriquez
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| | | | - Adrian Baranchuk
- Kingston General Hospital, Queen's University, Kingston, ON, Canada
| |
Collapse
|
5
|
Grassi S, Campuzano O, Coll M, Brión M, Arena V, Iglesias A, Carracedo Á, Brugada R, Oliva A. Genetic variants of uncertain significance: How to match scientific rigour and standard of proof in sudden cardiac death? Leg Med (Tokyo) 2020; 45:101712. [PMID: 32361481 DOI: 10.1016/j.legalmed.2020.101712] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 02/17/2020] [Accepted: 04/21/2020] [Indexed: 02/08/2023]
Abstract
In many SCD cases, in particular in pediatric age, autopsy can be completely negative and then a post-mortem genetic testing (molecular autopsy) is indicated. In NGS era finding new/rare variants is extremely frequent and, when only variants of unknown significance are found, molecular autopsy fails to find a cause of death. We describe the emblematic case of the sudden death of a 7-year-old girl. We performed a full-body micro-CT analysis, an accurate autopsy, a serum tryptase test and toxicological tests. Since the only macroscopic abnormality we found was a myocardial bridging (length: 1,1 cm, thickness: 0,5 cm) of the left anterior descending coronary artery, a molecular autopsy has been performed. NGS analysis on victim DNA detected rare variants in DPP6, MYH7, SCN2B and NOTCH1 and segregation analysis was then achieved. On the basis of ACMG/AMP (clinical) guidelines, all the found variants were classified as of unknown significance. In other words, both the macroscopic and genetic anomalies we found were of uncertain significance and then the autopsy failed to find the cause of the death. Our case raises three main discussion points: (a) economical, ethical and legal limitations of genetic investigation; (b) risk that genetic testing does not succeed in finding a certain cause of the death; (c) absence of specific guidelines to face the problem of VUS in forensic cases.
Collapse
Affiliation(s)
- Simone Grassi
- Institute of Public Health, Section of Legal Medicine, Catholic University, Rome, Italy
| | - Oscar Campuzano
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Centro Investigación Biomédica Red Enfermedades Cardiovasculares, Madrid, Spain; Department of Biochemistry and Molecular Genetics, Hospital Clinic, IDIBAPS, Barcelona, Spain
| | - Mònica Coll
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - María Brión
- Genetics of Cardiovascular and Ophthalmological Diseases, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain; Genomic Medicine, University of Santiago de Compostela, IDIS, CIBERER, Santiago de Compostela, Spain
| | - Vincenzo Arena
- Institute of Anatomical Pathology, Catholic University, Rome, Italy
| | - Anna Iglesias
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain
| | - Ángel Carracedo
- Genomic Medicine, University of Santiago de Compostela, IDIS, CIBERER, Santiago de Compostela, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Center, University of Girona-IDIBGI, Girona, Spain; Medical Science Department, School of Medicine, University of Girona, Girona, Spain; Centro Investigación Biomédica Red Enfermedades Cardiovasculares, Madrid, Spain; Cardiology Service, Hospital Josep Trueta, Girona, Spain
| | - Antonio Oliva
- Institute of Public Health, Section of Legal Medicine, Catholic University, Rome, Italy.
| |
Collapse
|
6
|
Ameri P, Schiattarella GG, Crotti L, Torchio M, Bertero E, Rodolico D, Forte M, Di Mauro V, Paolillo R, Chimenti C, Torella D, Catalucci D, Sciarretta S, Basso C, Indolfi C, Perrino C. Novel Basic Science Insights to Improve the Management of Heart Failure: Review of the Working Group on Cellular and Molecular Biology of the Heart of the Italian Society of Cardiology. Int J Mol Sci 2020; 21:E1192. [PMID: 32054029 PMCID: PMC7072832 DOI: 10.3390/ijms21041192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022] Open
Abstract
Despite important advances in diagnosis and treatment, heart failure (HF) remains a syndrome with substantial morbidity and dismal prognosis. Although implementation and optimization of existing technologies and drugs may lead to better management of HF, new or alternative strategies are desirable. In this regard, basic science is expected to give fundamental inputs, by expanding the knowledge of the pathways underlying HF development and progression, identifying approaches that may improve HF detection and prognostic stratification, and finding novel treatments. Here, we discuss recent basic science insights that encompass major areas of translational research in HF and have high potential clinical impact.
Collapse
Affiliation(s)
- Pietro Ameri
- IRCCS Ospedale Policlinico San Martino—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16132 Genova, Italy;
| | | | - Lia Crotti
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, 20149 Milan, Italy;
- Department of Medicine and Surgery, University of Milano-Bicocca, 20126 Milan, Italy
| | - Margherita Torchio
- Istituto Auxologico Italiano, IRCCS, Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin, and Laboratory of Cardiovascular Genetics, 20095 Milan, Italy;
| | - Edoardo Bertero
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany;
| | - Daniele Rodolico
- Agostino Gemelli Medical School, Catholic University of the Sacred Heart, 00168 Rome, Italy;
| | - Maurizio Forte
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzili, Italy; (M.F.); (S.S.)
| | - Vittoria Di Mauro
- National Research Council (CNR) Institute of Genetics & Biomedical Research, Milan Unit, 20138 Milan, Italy; (V.D.M.); (D.C.)
- Humanitas Clinical and Research Hospital, 20090 Rozzano (MI), Italy
| | - Roberta Paolillo
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| | - Cristina Chimenti
- Department of Cardiovascular, Respiratory, Nephrologic, and Geriatric Sciences, Sapienza University of Rome, 00100 Rome, Italy;
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Daniele Catalucci
- National Research Council (CNR) Institute of Genetics & Biomedical Research, Milan Unit, 20138 Milan, Italy; (V.D.M.); (D.C.)
- Humanitas Clinical and Research Hospital, 20090 Rozzano (MI), Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology, IRCCS Neuromed, 86077 Pozzili, Italy; (M.F.); (S.S.)
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padua, 35121 Padua, Italy;
| | - Ciro Indolfi
- Division of Cardiology, Department of Medical and Surgical Sciences & Center of Cardiovascular Research, Magna Graecia University, 88100 Catanzaro, Italy;
- URT-CNR, Magna Graecia University, 88100 Catanzaro, Italy
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| |
Collapse
|
7
|
Modeling of LMNA-Related Dilated Cardiomyopathy Using Human Induced Pluripotent Stem Cells. Cells 2019; 8:cells8060594. [PMID: 31208058 PMCID: PMC6627421 DOI: 10.3390/cells8060594] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/31/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the leading causes of heart failure and heart transplantation. A portion of familial DCM is due to mutations in the LMNA gene encoding the nuclear lamina proteins lamin A and C and without adequate treatment these patients have a poor prognosis. To get better insights into pathobiology behind this disease, we focused on modeling LMNA-related DCM using human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM). Primary skin fibroblasts from DCM patients carrying the most prevalent Finnish founder mutation (p.S143P) in LMNA were reprogrammed into hiPSCs and further differentiated into cardiomyocytes (CMs). The cellular structure, functionality as well as gene and protein expression were assessed in detail. While mutant hiPSC-CMs presented virtually normal sarcomere structure under normoxia, dramatic sarcomere damage and an increased sensitivity to cellular stress was observed after hypoxia. A detailed electrophysiological evaluation revealed bradyarrhythmia and increased occurrence of arrhythmias in mutant hiPSC-CMs on β-adrenergic stimulation. Mutant hiPSC-CMs also showed increased sensitivity to hypoxia on microelectrode array and altered Ca2+ dynamics. Taken together, p.S143P hiPSC-CM model mimics hallmarks of LMNA-related DCM and provides a useful tool to study the underlying cellular mechanisms of accelerated cardiac degeneration in this disease.
Collapse
|
8
|
Abstract
PURPOSE OF REVIEW To describe recent advancements in cardiovascular genetics made possible by leveraging next-generation sequencing (NGS), and to provide a framework for practical applications of genetic testing for hypertrophic (HCM), dilated (DCM), and arrhythmogenic right ventricular cardiomyopathies (ARVC). RECENT FINDINGS The availability of NGS has made possible extensive reference databases. These, combined with recent initiatives to compile previously siloed commercial and research cardiomyopathy data sets, provide a more powerful and precise approach to cardiovascular genetic medicine. HCM, DCM and ARVC are cardiomyopathies usually inherited in an autosomal dominant pattern. Over 1000 pathogenic mutations have been identified: HCM in genes encoding proteins of the sarcomere, and ARVC in genes encoding proteins of the desosome. DCM shows considerably more diverse ontology, suggesting more complex pathophysiology. In addition to allelic and locus heterogeneity, reduced penetrance and variable expressivity among affected individuals can make the clinical diagnosis of 'familial cardiomyopathy' less apparent. SUMMARY Current evidence supports the use of genetic testing in clinical practice to improve risk stratification for clinically affected patients and their at-risk relatives for hypertrophic, arrhythmogenic, and dilated cardiomyopathies. Understanding how to implement genetic testing and to evaluate at-risk family members, provide clinical implications of results as well as discuss limitations of genetic testing is essential to improving personalized care.
Collapse
|
9
|
Lin XF, Luo JW, Liu G, Zhu YB, Jin Z, Lin X. Genetic mutation of familial dilated cardiomyopathy based on next‑generation semiconductor sequencing. Mol Med Rep 2018; 18:4271-4280. [PMID: 30221713 PMCID: PMC6172371 DOI: 10.3892/mmr.2018.9455] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 08/02/2018] [Indexed: 01/10/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a complex myocardial disease of multifactorial etiologies, including enlarged cardiac chambers and contractile dysfunction. It has been suggested that the inheritance of DCM‑associated mutations predominates its onset. Therefore, the present study investigated the pathogenesis of DCM via pedigree analysis and genetic diagnosis by massive whole‑exome screening, and targeted exon capture. To study the familial gene‑phenotype association, the exon and splice sites of 325 hereditary disease‑associated genes in the proband with familial dilated cardiomyopathy (FDC), including 61 cardiac disease‑associated genes, such as the lamins A/C (LMNA), were analyzed by ultra‑high multiplex polymerase chain reaction and the Ion AmpliSeq™ Inherited Disease Panel. The present study also conducted Sanger DNA Sequencing for family members with global minor allele frequencies <1% to verify potential pathogenic mutation sites. A total of three rare missense mutations were detected, including heterozygous c.244G>A in LMNA, c.546C>G in potassium voltage‑gated channel subfamily KQT (KCNQ4) and c.1276G>A in EYA transcriptional coactivator and phosphatase 1 (EYA1), indicating a glutamic acid to lysine substitution at amino acid 82 (p.E82K) in LMNA, a p.F182L in KCNQ4 (a mutation associated with pathogenic deafness) and p.G426S in EYA1 (associated with Branchiootorenal syndrome 1 and Branchiootic syndrome 1 pathogenesis). In the present study, a carrier with slight hearing impairment was detected in the family analyzed; however, no patients with deafness or branchiootorenal syndrome were observed. LMNA p.E82K revealed SIFT and PolyPhen‑2 scores of 0 and 1, respectively. In the second generation, 3 patients with DCM underwent permanent pacemaker implantation due to sick sinus syndrome, atrioventricular block and unstable cardiac electrophysiology. The present study suggested that LMNA p.E82K may contribute to the pathogenesis of FDC and concomitant atrioventricular block. At present, only three families with DCM resulting from similar mutations have been reported. The present study demonstrated the strong pathogenic effects of LMNA p.E82K on DCM.
Collapse
Affiliation(s)
- Xin-Fu Lin
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Jie-Wei Luo
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Gui Liu
- Department of Traditional Chinese Medicine, Fujian Provincial Hospital, Fuzhou, Fujian 350001, P.R. China
| | - Yao-Bin Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian 350005, P.R. China
| | - Zhao Jin
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| | - Xing Lin
- Provincial Clinical Medical College, Fujian Medical University, Fuzhou, Fujian 350001, P.R. China
| |
Collapse
|
10
|
Bhide S, Trujillo AS, O'Connor MT, Young GH, Cryderman DE, Chandran S, Nikravesh M, Wallrath LL, Melkani GC. Increasing autophagy and blocking Nrf2 suppress laminopathy-induced age-dependent cardiac dysfunction and shortened lifespan. Aging Cell 2018; 17:e12747. [PMID: 29575479 PMCID: PMC5946079 DOI: 10.1111/acel.12747] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2018] [Indexed: 12/16/2022] Open
Abstract
Mutations in the human LMNA gene cause a collection of diseases known as laminopathies. These include myocardial diseases that exhibit age-dependent penetrance of dysrhythmias and heart failure. The LMNA gene encodes A-type lamins, intermediate filaments that support nuclear structure and organize the genome. Mechanisms by which mutant lamins cause age-dependent heart defects are not well understood. To address this issue, we modeled human disease-causing mutations in the Drosophila melanogaster Lamin C gene and expressed mutant Lamin C exclusively in the heart. This resulted in progressive cardiac dysfunction, loss of adipose tissue homeostasis, and a shortened adult lifespan. Within cardiac cells, mutant Lamin C aggregated in the cytoplasm, the CncC(Nrf2)/Keap1 redox sensing pathway was activated, mitochondria exhibited abnormal morphology, and the autophagy cargo receptor Ref2(P)/p62 was upregulated. Genetic analyses demonstrated that simultaneous over-expression of the autophagy kinase Atg1 gene and an RNAi against CncC eliminated the cytoplasmic protein aggregates, restored cardiac function, and lengthened lifespan. These data suggest that simultaneously increasing rates of autophagy and blocking the Nrf2/Keap1 pathway are a potential therapeutic strategy for cardiac laminopathies.
Collapse
Affiliation(s)
- Shruti Bhide
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Adriana S. Trujillo
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Maureen T. O'Connor
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Grant H. Young
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Diane E. Cryderman
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Sahaana Chandran
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Mastaneh Nikravesh
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| | - Lori L. Wallrath
- Department of Biochemistry; Carver College of Medicine; University of Iowa; Iowa City IA USA
| | - Girish C. Melkani
- Department of Biology, Molecular Biology and Heart Institutes; San Diego State University; San Diego CA USA
| |
Collapse
|
11
|
Peretto G, Sala S, Benedetti S, Di Resta C, Gigli L, Ferrari M, Della Bella P. Updated clinical overview on cardiac laminopathies: an electrical and mechanical disease. Nucleus 2018; 9:380-391. [PMID: 29929425 PMCID: PMC7000139 DOI: 10.1080/19491034.2018.1489195] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cardiac laminopathies, associated with mutations in the LMNA gene, encompass a wide spectrum of clinical manifestations, involving electrical and mechanical alterations of cardiomyocytes. Thus, dilated cardiomyopathy, bradyarrhythmias and atrial or ventricular tachyarrhythmias may occur in a number of combined phenotypes. Nowadays, some attempt has been made to identify clinical predictors for the most life-threatening complications of LMNA-associated heart disease, i.e. sudden cardiac death and end-stage heart failure. The goal of this manuscript is to combine the most recent evidences in an updated review to show the state-of-the-art of such a complex disease group. This is supposed to be the starting point to collect more data and design new ad hoc studies to identify clinically useful predictors to stratify risk in mutation carriers, including probands and their asymptomatic relatives.
Collapse
Affiliation(s)
- G. Peretto
- Department of Cardiac Electrophysyology and Arrhythmology, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - S. Sala
- Department of Cardiac Electrophysyology and Arrhythmology, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - S. Benedetti
- Laboratory of Clinical Molecular Biology and Cytogenetics, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - C. Di Resta
- Genomic Unit for the diagnosis of human pathologies, Division of Genetics and Cellular Biology, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - L. Gigli
- Department of Cardiac Electrophysyology and Arrhythmology, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - M. Ferrari
- Laboratory of Clinical Molecular Biology and Cytogenetics, IRCCS San Raffaele Hospital and University, Milan, Italy
- Genomic Unit for the diagnosis of human pathologies, Division of Genetics and Cellular Biology, IRCCS San Raffaele Hospital and University, Milan, Italy
| | - P. Della Bella
- Department of Cardiac Electrophysyology and Arrhythmology, IRCCS San Raffaele Hospital and University, Milan, Italy
| |
Collapse
|
12
|
Stroud MJ, Fang X, Veevers J, Chen J. Generation and Analysis of Striated Muscle Selective LINC Complex Protein Mutant Mice. Methods Mol Biol 2018; 1840:251-281. [PMID: 30141050 PMCID: PMC6887482 DOI: 10.1007/978-1-4939-8691-0_18] [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] [Indexed: 03/23/2023]
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex mediates intracellular cross talk between the nucleus and the cytoplasm. In striated muscle, the LINC complex provides structural support to the myocyte nucleus and plays an essential role in regulating gene expression and mechanotransduction. A wide range of cardiac and skeletal myopathies have been linked to mutations in LINC complex proteins. Studies utilizing tissue-specific knockout and mutant mouse models have revealed important insights into the roles of the LINC complex in striated muscle. In this chapter, we describe several feasible approaches for generating striated muscle-specific gene knockout and mutant mouse models to study LINC complex protein function in cardiac and skeletal muscle. The experimental procedures used for phenotyping and analysis of LINC complex knockout mice are also described.
Collapse
Affiliation(s)
- Matthew J Stroud
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Cardiovascular Division, King's College London, British Heart Foundation Centre of Excellence, London, UK
| | - Xi Fang
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jennifer Veevers
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ju Chen
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Li X, Zhang P. Genetic determinants of myocardial dysfunction. J Med Genet 2016; 54:1-10. [DOI: 10.1136/jmedgenet-2016-104308] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 10/26/2016] [Accepted: 10/27/2016] [Indexed: 12/30/2022]
|
14
|
Skeletal Muscle Laminopathies: A Review of Clinical and Molecular Features. Cells 2016; 5:cells5030033. [PMID: 27529282 PMCID: PMC5040975 DOI: 10.3390/cells5030033] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/01/2016] [Accepted: 06/08/2016] [Indexed: 01/12/2023] Open
Abstract
LMNA-related disorders are caused by mutations in the LMNA gene, which encodes for the nuclear envelope proteins, lamin A and C, via alternative splicing. Laminopathies are associated with a wide range of disease phenotypes, including neuromuscular, cardiac, metabolic disorders and premature aging syndromes. The most frequent diseases associated with mutations in the LMNA gene are characterized by skeletal and cardiac muscle involvement. This review will focus on genetics and clinical features of laminopathies affecting primarily skeletal muscle. Although only symptomatic treatment is available for these patients, many achievements have been made in clarifying the pathogenesis and improving the management of these diseases.
Collapse
|
15
|
Furusawa T, Rochman M, Taher L, Dimitriadis EK, Nagashima K, Anderson S, Bustin M. Chromatin decompaction by the nucleosomal binding protein HMGN5 impairs nuclear sturdiness. Nat Commun 2015; 6:6138. [PMID: 25609380 PMCID: PMC4304400 DOI: 10.1038/ncomms7138] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/15/2014] [Indexed: 12/19/2022] Open
Abstract
In most metazoan nuclei, heterochromatin is located at the nuclear periphery in contact with the nuclear lamina, which provides mechanical stability to the nucleus. We show that in cultured cells, chromatin de-compaction by the nucleosome binding protein HMGN5 decreases the sturdiness, elasticity, and rigidity of the nucleus. Mice overexpressing HMGN5, either globally or only in the heart, are normal at birth but develop hypertrophic heart with large cardiomyoctyes, deformed nuclei and disrupted lamina, and die of cardiac malfunction. Chromatin de-compaction is seen in cardiomyocytes of newborn mice but misshaped nuclei with disrupted lamina are seen only in adult cardiomyocytes, suggesting that loss of heterochromatin diminishes the ability of the nucleus to withstand the mechanical forces of the contracting heart. Thus, heterochromatin enhances the ability of the nuclear lamina to maintain the sturdiness and shape of the eukaryotic nucleus; a structural role for chromatin that is distinct from its genetic functions.
Collapse
Affiliation(s)
- Takashi Furusawa
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mark Rochman
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Leila Taher
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, University of Rostock, Rostock 18057, Germany
| | - Emilios K Dimitriadis
- Biomedical Engineering and Physical Science Shared Resource Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kunio Nagashima
- Electron microscopy laboratory, Leidos Biomedical Research Inc., SAIC-Frederick Inc.,Frederick National Laboratory for Cancer Research, Frederick, Maryland 21701, USA
| | - Stasia Anderson
- Animal Core Facility, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Michael Bustin
- Protein Section, Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
16
|
Stroud MJ, Banerjee I, Veevers J, Chen J. Linker of nucleoskeleton and cytoskeleton complex proteins in cardiac structure, function, and disease. Circ Res 2014; 114:538-48. [PMID: 24481844 DOI: 10.1161/circresaha.114.301236] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The linker of nucleoskeleton and cytoskeleton (LINC) complex, composed of proteins within the inner and the outer nuclear membranes, connects the nuclear lamina to the cytoskeleton. The importance of this complex has been highlighted by the discovery of mutations in genes encoding LINC complex proteins, which cause skeletal or cardiac myopathies. Herein, this review summarizes structure, function, and interactions of major components of the LINC complex, highlights how mutations in these proteins may lead to cardiac disease, and outlines future challenges in the field.
Collapse
Affiliation(s)
- Matthew J Stroud
- From the Department of Cardiology, University of California San Diego School of Medicine, La Jolla, CA
| | | | | | | |
Collapse
|
17
|
Abstract
Nuclear envelope
spectrin-repeat
proteins (Nesprins), are a novel family of
nuclear and cytoskeletal proteins with rapidly expanding roles as intracellular scaffolds
and linkers. Originally described as proteins that localise to the nuclear envelope (NE)
and establish nuclear-cytoskeletal connections, nesprins have now been found to comprise a
diverse spectrum of tissue specific isoforms that localise to multiple sub-cellular
compartments. Here, we describe how nesprins are necessary in maintaining cellular
architecture by acting as essential scaffolds and linkers at both the NE and other
sub-cellular domains. More importantly, we speculate how nesprin mutations may disrupt
tissue specific nesprin scaffolds and explain the tissue specific nature of many
nesprin-associated diseases, including laminopathies.
Collapse
|
18
|
Carboni N, Mateddu A, Marrosu G, Cocco E, Marrosu MG. Genetic and clinical characteristics of skeletal and cardiac muscle in patients with lamin A/C gene mutations. Muscle Nerve 2013; 48:161-70. [PMID: 23450819 DOI: 10.1002/mus.23827] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2013] [Indexed: 12/12/2022]
Abstract
Alterations of the lamin A/C (LMNA) gene are associated with different clinical entities, including disorders that affect skeletal and cardiac muscle, peripheral nerves, metabolism, bones, and disorders that cause premature aging. In this article we review the clinical and genetic characteristics of cardiac and skeletal muscle diseases related to alterations in the LMNA gene. There is no single explanation of how LMNA gene alterations may cause these disorders; however, important goals have been achieved in understanding the pathogenic effects of LMNA gene mutations on cardiac and skeletal muscle.
Collapse
Affiliation(s)
- Nicola Carboni
- Department of Public Health, Clinical and Molecular Medicine, Multiple Sclerosis Centre, Via Is Guadazzonis 2, 09100 Cagliari, University of Cagliari, Italy.
| | | | | | | | | |
Collapse
|
19
|
Yang J, Zhao Y, Hao P, Meng X, Dong M, Wang Y, Zhang Y, Zhang C. Impact of angiotensin I converting enzyme insertion/deletion polymorphisms on dilated cardiomyopathy and hypertrophic cardiomyopathy risk. PLoS One 2013; 8:e63309. [PMID: 23691019 PMCID: PMC3653933 DOI: 10.1371/journal.pone.0063309] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 04/02/2013] [Indexed: 01/13/2023] Open
Abstract
Background Genetic factors in the pathogenesis of cardiomyopathies have received a lot attention during the past two decades. Angiotensin I converting enzyme (ACE) insertion/deletion (I/D) polymorphisms were found to be associated with cardiomyopathies. However, the previous results were inconsistent. The current meta-analysis aims to examine the association of ACE I/D polymorphisms and dilated cardiomyopathy (DCM) or hypertrophic cardiomyopathy (HCM). Methods Eight studies on DCM (1387 controls and 977 patients) and eight studies on HCM (1055 controls and 827 patients) were included in this meta-analysis. Results The overall data showed no significant association between ACE I/D polymorphism and DCM risk. Further subgroup analysis by ethnicity also did not find a significantly increased risk for D allele carriers among East Asians and Europeans. However, the overall analysis suggested that the D allele carriers might be associated with increased risk of HCM (DD/ID vs. II: OR = 1.69, 95% CI 1.04–2.74, P = 0.03). Conclusion In summary, the meta-analysis indicated that certain ACE I/D polymorphism might be associated with HCM but not DCM susceptibility. Given the limited sample sizes, further large multicenter case-control investigation is needed.
Collapse
Affiliation(s)
- Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yunhan Zhao
- Shandong University School of Medicine, Jinan, Shandong, P.R. China
| | - Panpan Hao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Xiao Meng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Mei Dong
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Ying Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
| | - Yun Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
- * E-mail: (CZ); (YZ)
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, Shandong, P.R. China
- * E-mail: (CZ); (YZ)
| |
Collapse
|
20
|
Kloos W, Katus HA, Meder B. Genetic cardiomyopathies. Lessons learned from humans, mice, and zebrafish. Herz 2013; 37:612-7. [PMID: 22767018 DOI: 10.1007/s00059-012-3651-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dilated cardiomyopathy (DCM) is a multifactorial disease of the heart muscle and a leading cause of congestive heart failure. Human genetic studies and the establishment of suitable animal models such as mice and zebrafish have already revealed parts of its genetic etiology. With the next generation of genomic sequencing technologies (NGS) on the rise, the comprehensive genetic dissection of DCM patients will reveal clinically relevant information, novel causes, and modifiers of this complex disorder. The recent exploration of the epigenome as another mechanism of cardiac gene regulation will further elucidate unexplained variations observed in the correlation between the patient's genotype and phenotype. Some of these intriguing advances being made in basic genetic research will soon find their way into clinical practice for more individualized treatment of cardiomyopathy patients.
Collapse
Affiliation(s)
- W Kloos
- Abteilung Innere Medizin III, Kardiologie, Angiologie und Pulmologie, Universitätsklinik Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Gemany
| | | | | |
Collapse
|
21
|
Roncarati R, Viviani Anselmi C, Krawitz P, Lattanzi G, von Kodolitsch Y, Perrot A, di Pasquale E, Papa L, Portararo P, Columbaro M, Forni A, Faggian G, Condorelli G, Robinson PN. Doubly heterozygous LMNA and TTN mutations revealed by exome sequencing in a severe form of dilated cardiomyopathy. Eur J Hum Genet 2013; 21:1105-11. [PMID: 23463027 DOI: 10.1038/ejhg.2013.16] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 12/20/2012] [Accepted: 01/17/2013] [Indexed: 01/18/2023] Open
Abstract
Familial dilated cardiomyopathy (DCM) is a heterogeneous disease; although 30 disease genes have been discovered, they explain only no more than half of all cases; in addition, the causes of intra-familial variability in DCM have remained largely unknown. In this study, we exploited the use of whole-exome sequencing (WES) to investigate the causes of clinical variability in an extended family with 14 affected subjects, four of whom showed particular severe manifestations of cardiomyopathy requiring heart transplantation in early adulthood. This analysis, followed by confirmative conventional sequencing, identified the mutation p.K219T in the lamin A/C gene in all 14 affected patients. An additional variant in the gene for titin, p.L4855F, was identified in the severely affected patients. The age for heart transplantation was substantially less for LMNA:p.K219T/TTN:p.L4855F double heterozygotes than that for LMNA:p.K219T single heterozygotes. Myocardial specimens of doubly heterozygote individuals showed increased nuclear length, sarcomeric disorganization, and myonuclear clustering compared with samples from single heterozygotes. In conclusion, our results show that WES can be used for the identification of causal and modifier variants in families with variable manifestations of DCM. In addition, they not only indicate that LMNA and TTN mutational status may be useful in this family for risk stratification in individuals at risk for DCM but also suggest titin as a modifier for DCM.
Collapse
Affiliation(s)
- Roberta Roncarati
- 1] Biomedical and Genetic Research Institute (IRGB), Milan Unit, National Research Council of Italy, Milan, Italy [2] Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Jadhav KB, Karpe KK, Maramattom BV. An Indian family with an Emery-Dreifuss myopathy and familial dilated cardiomyopathy due to a novel LMNA mutation. Ann Indian Acad Neurol 2013; 15:344-6. [PMID: 23349612 PMCID: PMC3548385 DOI: 10.4103/0972-2327.104355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 01/11/2012] [Accepted: 06/23/2012] [Indexed: 11/15/2022] Open
Abstract
Emery-Dreifuss myopathy can be associated with a cardiomyopathy and cardiac dysrhythmias. The inheritance pattern of Emery-Dreifuss muscular dystrophy (EDMD) is X linked, whereas EDMD2 is autosomal dominant. EDMD2 is caused by lamin A/C gene (LMNA) mutations that produce alterations in the lamin proteins that are integral to nuclear and cell integrity. A 53-year-old man was brought to us with a right internal carotid artery dissection. Detailed work-up of the patient and family members revealed some unusual features, and genetic sequencing of the LMNA gene was undertaken. A novel mutation was identified in two of the samples sent for analysis. We present the first Indian family of EDMD2 with familial dilated cardiomyopathy and cardiac dysrhythmias in whom LMNA gene sequencing was performed. A novel mutation was identified and additional unusual clinical features were described.
Collapse
Affiliation(s)
- Khushal B Jadhav
- Department of Neurology, Lourdes Heart Institute and Neuro Center, Kochi, Kerala, India
| | | | | |
Collapse
|
23
|
Sims-Williams HP, Nye HJ, Walker PR. Dilated cardiomyopathy and skeletal myopathy: presenting features of a laminopathy. BMJ Case Rep 2013; 2013:bcr-2012-007574. [PMID: 23329710 DOI: 10.1136/bcr-2012-007574] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Mutations in the lamin A/C (LMNA) gene cause significant disruption to skeletal and myocardial muscle, as well as nervous tissue. We describe a case illustrating varied manifestations of a LMNA mutation and the implications for diagnosis and management. We turn to several family studies that describe considerable phenotypic variation arising from LMNA mutations. The discussion focuses on educating the reader in recognition of potential presentations of LMNA mutations.
Collapse
Affiliation(s)
- Hugh P Sims-Williams
- Department of Neurology, University Hospital of North Staffordshire, Stoke on Trent, UK.
| | | | | |
Collapse
|
24
|
McNally EM, Golbus JR, Puckelwartz MJ. Genetic mutations and mechanisms in dilated cardiomyopathy. J Clin Invest 2013; 123:19-26. [PMID: 23281406 DOI: 10.1172/jci62862] [Citation(s) in RCA: 323] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Genetic mutations account for a significant percentage of cardiomyopathies, which are a leading cause of congestive heart failure. In hypertrophic cardiomyopathy (HCM), cardiac output is limited by the thickened myocardium through impaired filling and outflow. Mutations in the genes encoding the thick filament components myosin heavy chain and myosin binding protein C (MYH7 and MYBPC3) together explain 75% of inherited HCMs, leading to the observation that HCM is a disease of the sarcomere. Many mutations are "private" or rare variants, often unique to families. In contrast, dilated cardiomyopathy (DCM) is far more genetically heterogeneous, with mutations in genes encoding cytoskeletal, nucleoskeletal, mitochondrial, and calcium-handling proteins. DCM is characterized by enlarged ventricular dimensions and impaired systolic and diastolic function. Private mutations account for most DCMs, with few hotspots or recurring mutations. More than 50 single genes are linked to inherited DCM, including many genes that also link to HCM. Relatively few clinical clues guide the diagnosis of inherited DCM, but emerging evidence supports the use of genetic testing to identify those patients at risk for faster disease progression, congestive heart failure, and arrhythmia.
Collapse
Affiliation(s)
- Elizabeth M McNally
- Department of Human Genetics, University of Chicago, Chicago, Illinois 60637, USA.
| | | | | |
Collapse
|
25
|
Chen SC, Kennedy BK, Lampe PD. Phosphorylation of connexin43 on S279/282 may contribute to laminopathy-associated conduction defects. Exp Cell Res 2012; 319:888-96. [PMID: 23261543 DOI: 10.1016/j.yexcr.2012.12.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 12/11/2012] [Accepted: 12/13/2012] [Indexed: 01/07/2023]
Abstract
An understanding of the molecular mechanism behind the arrhythmic phenotype associated with laminopathies has yet to emerge. A-type lamins have been shown to interact and sequester activated phospho-ERK1/2(pERK1/2) at the nucleus. The gap junction protein connexin43 (Cx43) can be phosphorylated by pERK1/2 on S279/282 (pS279/282), inhibiting intercellular communication. We hypothesized that without A-type lamins, pS279/282 Cx43 will increase due to inappropriate phosphorylation by pERK1/2, resulting in decreased gap junction function. We observed a 1.6-fold increase in pS279/282 Cx43 levels in Lmna(-/-) mouse embryonic fibroblasts (MEFs) compared to Lmna(+/+), and 1.8-fold more pERK1/2 co-precipitated from Lmna(-/-) MEFs with Cx43 antibodies. We found a 3-fold increase in the fraction of non-nuclear pERK1/2 and a concomitant 2-fold increase in the fraction of pS279/282 Cx43 in Lmna(-/-) MEFs by immunofluorescence. In an assay of gap junctional function, Lmna(-/-) MEFs transferred dye to 60% fewer partners compared to Lmna(+/+) controls. These results are mirrored in 5-6 week-old Lmna(-/-) mice compared to their Lmna(+/+) littermates as we detect increased pS279/282 Cx43 in gap junctions by immunofluorescence and 1.7-fold increased levels by immunoblot. We conclude that increased pS279/282 Cx43 in the Lmna(-/-) background results in decreased cell communication and may contribute to the arrhythmic pathology in vivo.
Collapse
Affiliation(s)
- Steven C Chen
- Fred Hutchinson Cancer Research Center (FHCRC), Public Health Sciences Division, 1100 Fairview Ave. N., Seattle, WA 98109, USA
| | | | | |
Collapse
|
26
|
van Rijsingen IAW, Bakker A, Azim D, Hermans-van Ast JF, van der Kooi AJ, van Tintelen JP, van den Berg MP, Christiaans I, Lekanne Dit Deprez RH, Wilde AAM, Zwinderman AH, Meijers JCM, Grootemaat AE, Nieuwland R, Pinto YM, Pinto-Sietsma SJ. Lamin A/C mutation is independently associated with an increased risk of arterial and venous thromboembolic complications. Int J Cardiol 2012; 168:472-7. [PMID: 23073275 DOI: 10.1016/j.ijcard.2012.09.118] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 09/10/2012] [Accepted: 09/22/2012] [Indexed: 11/15/2022]
Abstract
BACKGROUND Lamin A/C (LMNA) mutation carriers suffer from a variety of clinical phenotypes, including dilated cardiomyopathy (DCM). Although it has been suggested that carriers are at risk for thromboembolic complications, it is unknown whether this risk is higher than can be expected from the underlying cardiac abnormalities. The purpose of this study was to determine whether a LMNA mutation is associated with an increased risk of thromboembolic complications. METHODS We compared a cohort of 76 LMNA mutation carriers with a cohort of 224 idiopathic DCM patients without a LMNA mutation, with respect to the prevalence of arterial and venous thromboembolic complications. Furthermore, we carried out a case-control study to explore whether a prothrombotic phenotype was present in LMNA mutation carriers without DCM or atrial tachyarrhythmias (n=14) and compared this with mutation negative relatives (n=13). RESULTS The prevalence of thromboembolic complications was higher in the cohort of LMNA mutation carriers than in DCM patients (22 vs 11%; p<0.05), after respectively mean follow-up of 42 ± 12 and 49 ± 12 years. After adjustment for possible confounders, including atrial tachyarrhythmias and left ventricular ejection fraction, LMNA mutation carriership was independently associated with an increased risk of thromboembolic complications (HR 4.8, 95% CI: 2.2-10.6). The results of the case-control study suggested a prothrombotic phenotype in LMNA mutation carriers, as reflected by an altered platelet function and increased thrombin generation. CONCLUSIONS LMNA mutation is independently associated with an increased risk of arterial and venous thromboembolic complications. Laboratory research in LMNA mutation carriers without severe cardiac abnormalities suggests a prothrombotic phenotype.
Collapse
|
27
|
Affiliation(s)
- Srijita Sen-Chowdhry
- Inherited Cardiovascular Disease Group, University College London, The Heart Hospital, 16-18 Westmoreland Street, London, UK
| | | |
Collapse
|
28
|
Carboni N, Sardu C, Cocco E, Marrosu G, Manzi RC, Nissardi V, Isola F, Mateddu A, Solla E, Maioli MA, Oppo V, Piras R, Coghe G, Lai C, Marrosu MG. Cardiac involvement in patients with lamin A/C gene mutations: A cohort observation. Muscle Nerve 2012; 46:187-92. [DOI: 10.1002/mus.23294] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
29
|
Meder B, Katus H. Klinik und Genetik der hypertrophen und dilatativen Kardiomyopathie. Internist (Berl) 2012; 53:408-14, 417-8. [DOI: 10.1007/s00108-011-2988-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
|
31
|
Ho JCY, Zhou T, Lai WH, Huang Y, Chan YC, Li X, Wong NLY, Li Y, Au KW, Guo D, Xu J, Siu CW, Pei D, Tse HF, Esteban MA. Generation of induced pluripotent stem cell lines from 3 distinct laminopathies bearing heterogeneous mutations in lamin A/C. Aging (Albany NY) 2011; 3:380-90. [PMID: 21483033 PMCID: PMC3117453 DOI: 10.18632/aging.100277] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The term laminopathies defines a group of genetic disorders caused by defects in the nuclear envelope, mostly the lamins. Lamins are the main constituents of the nuclear lamina, a filamentous meshwork associated with the inner nuclear membrane that provides mechanical stability and plays important roles in processes such as transcription, DNA replication and chromatin organization. More than 300 mutations in lamin A/C have been associated with diverse clinical phenotypes, understanding the molecular basis of these diseases may provide a rationale for treating them. Here we describe the generation of induced pluripotent stem cells (iPSCs) from a patient with inherited dilated cardiomiopathy and 2 patients with distinct accelerated forms of aging, atypical Werner syndrome and Hutchinson Gilford progeria, all of which are caused by mutations in lamin A/C. These cell lines were pluripotent and displayed normal nuclear membrane morphology compared to donor fibroblasts. Their differentiated progeny reproduced the disease phenotype, reinforcing the idea that they represent excellent tools for understanding the role of lamin A/C in normal physiology and the clinical diversity associated with these diseases.
Collapse
Affiliation(s)
- Jenny C Y Ho
- Cardiology Division, Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Pokfulam, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Holmström M, Kivistö S, Heliö T, Jurkko R, Kaartinen M, Antila M, Reissell E, Kuusisto J, Kärkkäinen S, Peuhkurinen K, Koikkalainen J, Lötjönen J, Lauerma K. Late gadolinium enhanced cardiovascular magnetic resonance of lamin A/C gene mutation related dilated cardiomyopathy. J Cardiovasc Magn Reson 2011; 13:30. [PMID: 21689390 PMCID: PMC3135551 DOI: 10.1186/1532-429x-13-30] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 06/20/2011] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The purpose of this study was to identify early features of lamin A/C gene mutation related dilated cardiomyopathy (DCM) with cardiovascular magnetic resonance (CMR). We characterise myocardial and functional findings in carriers of lamin A/C mutation to facilitate the recognition of these patients using this method. We also investigated the connection between myocardial fibrosis and conduction abnormalities. METHODS Seventeen lamin A/C mutation carriers underwent CMR. Late gadolinium enhancement (LGE) and cine images were performed to evaluate myocardial fibrosis, regional wall motion, longitudinal myocardial function, global function and volumetry of both ventricles. The location, pattern and extent of enhancement in the left ventricle (LV) myocardium were visually estimated. RESULTS Patients had LV myocardial fibrosis in 88% of cases. Segmental wall motion abnormalities correlated strongly with the degree of enhancement. Myocardial enhancement was associated with conduction abnormalities. Sixty-nine percent of our asymptomatic or mildly symptomatic patients showed mild ventricular dilatation, systolic failure or both in global ventricular analysis. Decreased longitudinal systolic LV function was observed in 53% of patients. CONCLUSIONS Cardiac conduction abnormalities, mildly dilated LV and depressed systolic dysfunction are common in DCM caused by a lamin A/C gene mutation. However, other cardiac diseases may produce similar symptoms. CMR is an accurate tool to determine the typical cardiac involvement in lamin A/C cardiomyopathy and may help to initiate early treatment in this malignant familiar form of DCM.
Collapse
MESH Headings
- Adolescent
- Adult
- Cardiomyopathy, Dilated/diagnosis
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/physiopathology
- Chi-Square Distribution
- Contrast Media
- Electrocardiography
- Female
- Fibrosis
- Finland
- Genetic Predisposition to Disease
- Humans
- Hypertrophy, Left Ventricular/diagnosis
- Hypertrophy, Left Ventricular/genetics
- Lamin Type A/genetics
- Magnetic Resonance Imaging, Cine
- Male
- Meglumine
- Middle Aged
- Mutation
- Myocardium/pathology
- Organometallic Compounds
- Phenotype
- Predictive Value of Tests
- Severity of Illness Index
- Ventricular Dysfunction, Left/diagnosis
- Ventricular Dysfunction, Left/genetics
- Ventricular Function, Left
- Ventricular Function, Right
Collapse
Affiliation(s)
- Miia Holmström
- Department of Radiology, University of Helsinki and HUS Radiology (Medical Imaging Center) P.O. Box 340, FI-00029 HUS, Finland
| | - Sari Kivistö
- Department of Radiology, University of Helsinki and HUS Radiology (Medical Imaging Center) P.O. Box 340, FI-00029 HUS, Finland
| | - Tiina Heliö
- Department of Cardiology, Helsinki University Central Hospital, P.O. Box 340, FI-00029 HUS, Finland
| | - Raija Jurkko
- Department of Cardiology, Helsinki University Central Hospital, P.O. Box 340, FI-00029 HUS, Finland
| | - Maija Kaartinen
- Department of Cardiology, Helsinki University Central Hospital, P.O. Box 340, FI-00029 HUS, Finland
| | - Margareta Antila
- Department of Radiology, University of Helsinki and HUS Radiology (Medical Imaging Center) P.O. Box 340, FI-00029 HUS, Finland
| | - Eeva Reissell
- Boehringer Ingelheim Finland Ky Tammasaarenkatu 5, FI-00180 Helsinki, Finland
| | - Johanna Kuusisto
- Heart Center, Kuopio University Hospital, P.O. Box 1777, FI-70211 Kuopio, Finland
| | - Satu Kärkkäinen
- Heart Center, Kuopio University Hospital, P.O. Box 1777, FI-70211 Kuopio, Finland
| | - Keijo Peuhkurinen
- Heart Center, Kuopio University Hospital, P.O. Box 1777, FI-70211 Kuopio, Finland
| | - Juha Koikkalainen
- VTT Technical Research Centre of Finland, P.O. Box 1300, FI-33101 Tampere, Finland
| | - Jyrki Lötjönen
- VTT Technical Research Centre of Finland, P.O. Box 1300, FI-33101 Tampere, Finland
| | - Kirsi Lauerma
- Department of Radiology, University of Helsinki and HUS Radiology (Medical Imaging Center) P.O. Box 340, FI-00029 HUS, Finland
| |
Collapse
|
33
|
Affiliation(s)
- Hugh Watkins
- Department of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom.
| | | | | |
Collapse
|
34
|
Sysi-Aho M, Koikkalainen J, Seppänen-Laakso T, Kaartinen M, Kuusisto J, Peuhkurinen K, Kärkkäinen S, Antila M, Lauerma K, Reissell E, Jurkko R, Lötjönen J, Heliö T, Orešič M. Serum lipidomics meets cardiac magnetic resonance imaging: profiling of subjects at risk of dilated cardiomyopathy. PLoS One 2011; 6:e15744. [PMID: 21283746 PMCID: PMC3024392 DOI: 10.1371/journal.pone.0015744] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 11/27/2010] [Indexed: 11/19/2022] Open
Abstract
Dilated cardiomyopathy (DCM), characterized by left ventricular dilatation and systolic dysfunction, constitutes a significant cause for heart failure, sudden cardiac death or need for heart transplantation. Lamin A/C gene (LMNA) on chromosome 1p12 is the most significant disease gene causing DCM and has been reported to cause 7-9% of DCM leading to cardiac transplantation. We have previously performed cardiac magnetic resonance imaging (MRI) to LMNA carriers to describe the early phenotype. Clinically, early recognition of subjects at risk of developing DCM would be important but is often difficult. Thus we have earlier used the MRI findings of these LMNA carriers for creating a model by which LMNA carriers could be identified from the controls at an asymptomatic stage. Some LMNA mutations may cause lipodystrophy. To characterize possible effects of LMNA mutations on lipid profile, we set out to apply global serum lipidomics using Ultra Performance Liquid Chromatography coupled to mass spectrometry in the same LMNA carriers, DCM patients without LMNA mutation and controls. All DCM patients, with or without LMNA mutation, differed from controls in regard to distinct serum lipidomic profile dominated by diminished odd-chain triglycerides and lipid ratios related to desaturation. Furthermore, we introduce a novel approach to identify associations between the molecular lipids from serum and the MR images from the LMNA carriers. The association analysis using dependency network and regression approaches also helped us to obtain novel insights into how the affected lipids might relate to cardiac shape and volume changes. Our study provides a framework for linking serum derived molecular markers not only with clinical endpoints, but also with the more subtle intermediate phenotypes, as derived from medical imaging, of potential pathophysiological relevance.
Collapse
Affiliation(s)
- Marko Sysi-Aho
- VTT Technical Research Centre of Finland, Espoo, Finland
| | | | | | | | | | | | | | | | - Kirsi Lauerma
- Helsinki Medical Imaging Center, Helsinki University Central Hospital, Helsinki, Finland
| | - Eeva Reissell
- Helsinki University Central Hospital, Helsinki, Finland
| | - Raija Jurkko
- Helsinki University Central Hospital, Helsinki, Finland
| | - Jyrki Lötjönen
- VTT Technical Research Centre of Finland, Tampere, Finland
| | - Tiina Heliö
- Helsinki University Central Hospital, Helsinki, Finland
| | - Matej Orešič
- VTT Technical Research Centre of Finland, Espoo, Finland
- * E-mail:
| |
Collapse
|
35
|
Raman SV, Basso C, Tandri H, Taylor MRG. Imaging phenotype vs genotype in nonhypertrophic heritable cardiomyopathies: dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy. Circ Cardiovasc Imaging 2010; 3:753-65. [PMID: 21081743 DOI: 10.1161/circimaging.110.957563] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Subha V Raman
- Ohio State University College of Medicine, Columbus, Ohio, USA.
| | | | | | | |
Collapse
|
36
|
Abstract
Sumoylation is a posttranslational modification process in which SUMO proteins are covalently and reversibly conjugated to their targets via enzymatic cascade reactions. Since the discovery of SUMO-1 in 1996, the SUMO pathway has garnered increased attention due to its role in a number of important biological activities such as cell cycle progression, epigenetic modulation, signal transduction, and DNA replication/repair, as well as its potential implication in human pathogenesis such as in cancer development and metastasis, neurodegenerative disorders and craniofacial defects. The role of the SUMO pathway in regulating cardiogenic gene activity, development and/or disorders is just emerging. Our review is based on recent advances that highlight the regulation of cardiac gene activity in cardiac development and disease by the SUMO conjugation pathway.
Collapse
Affiliation(s)
- Jun Wang
- Center for Stem Cell Engineering, Department of Basic Research Laboratories, Texas Heart Institute, Houston, TX 77030
| | - Robert J Schwartz
- Center for Stem Cell Engineering, Department of Basic Research Laboratories, Texas Heart Institute, Houston, TX 77030
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| |
Collapse
|
37
|
Proteomic Profiling of the Dystrophin-Deficient MDX Heart Reveals Drastically Altered Levels of Key Metabolic and Contractile Proteins. J Biomed Biotechnol 2010; 2010:648501. [PMID: 20508850 PMCID: PMC2874991 DOI: 10.1155/2010/648501] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Accepted: 02/25/2010] [Indexed: 12/13/2022] Open
Abstract
Although Duchenne muscular dystrophy is primarily classified as a neuromuscular disease, cardiac complications play an important role in the course of this X-linked inherited disorder. The pathobiochemical steps causing a progressive decline in the dystrophic heart are not well understood. We therefore carried out a fluorescence difference in-gel electrophoretic analysis of 9-month-old dystrophin-deficient versus age-matched normal heart, using the established MDX mouse model of muscular dystrophy-related cardiomyopathy. Out of 2,509 detectable protein spots, 79 2D-spots showed a drastic differential expression pattern, with the concentration of 3 proteins being increased, including nucleoside diphosphate kinase and lamin-A/C, and of 26 protein species being decreased, including ATP synthase, fatty acid binding-protein, isocitrate dehydrogenase, NADH dehydrogenase, porin, peroxiredoxin, adenylate kinase, tropomyosin, actin, and myosin light chains. Hence, the lack of cardiac dystrophin appears to trigger a generally perturbed protein expression pattern in the MDX heart, affecting especially energy metabolism and contractile proteins.
Collapse
|
38
|
Verstraeten VLRM, Lammerding J. Another broken heart: loss of lamina-associated polypeptide 2alpha causes systolic dysfunction. Circ Res 2010; 106:234-7. [PMID: 20133908 DOI: 10.1161/circresaha.109.212001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
39
|
Jensen BC. Skin deep: what can the study of dermal fibroblasts teach us about dilated cardiomyopathy? J Mol Cell Cardiol 2009; 48:576-8. [PMID: 20005233 DOI: 10.1016/j.yjmcc.2009.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Accepted: 11/26/2009] [Indexed: 11/17/2022]
|