1
|
Hauptmeijer RWL, Lippert L, Ten Cate FEAU, Fejzic Z, Leenders E, Wolf CM, Draaisma JMT. Differentiating primary sarcomeric hypertrophic cardiomyopathy from Noonan syndrome: can the electrocardiogram be of use? Cardiol Young 2024; 34:597-603. [PMID: 37649442 DOI: 10.1017/s1047951123003177] [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: 09/01/2023]
Abstract
Noonan syndrome is a multi-system genetic disorder and patients may suffer from hypertrophic cardiomyopathy. Previous studies have identified electrocardiographic features that may support a diagnosis of Noonan syndrome. In this two-centre retrospective study, we analysed typical Noonan syndrome-related electrocardiographic features in 30 patients with Noonan syndrome with hypertrophic cardiomyopathy and compared these with the electrocardiographic features in 15 children with sarcomeric hypertrophic cardiomyopathy. Typical Noonan syndrome-related electrocardiographic features are a negative aVF, small left precordial R-waves, large right precordial S-waves, and abnormal Q-wave. We also analysed electrocardiographic features of hypertrophic cardiomyopathy: ST-segment abnormalities and T-wave abnormalities. A negative aVF was seen in 83% of patients with Noonan syndrome-related hypertrophic cardiomyopathy in contrast to 27% of patients with primary sarcomeric hypertrophic cardiomyopathy (p < 0.001). An extreme QRS axis in the north-west was seen only in patients with Noonan syndrome-related hypertrophic cardiomyopathy. This QRS axis deviation is likely to be determined by the Noonan syndrome-related hypertrophic cardiomyopathy and not by the type of hypertrophic cardiomyopathy. There were no differences between the two groups in the frequency of large right precordial S-waves and small R-waves in the left precordial leads V5 and V6. However, an abnormal R/S ratio was more often seen in patients with Noonan syndrome-related hypertrophic cardiomyopathy (p < 0.001). Pathologic Q-waves were seen statistically more frequently in patients with sarcomeric hypertrophic cardiomyopathy (p = 0.009). The occurrence of ST-segment and T-wave pathology did not statistically differ between the two groups. Electrography can be of use in differentiating sarcomeric hypertrophic cardiomyopathy from Noonan syndrome-related hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Robert W L Hauptmeijer
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lea Lippert
- Department of Congenital Heart Disease and Pediatric Cardiology, Technical University of Munich, School of Medicine & Health, Munich, Germany
| | - Floris E A Udink Ten Cate
- Department of Pediatric Cardiology, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zina Fejzic
- Department of Pediatric Cardiology, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erika Leenders
- Department of Human genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cordula M Wolf
- Department of Congenital Heart Disease and Pediatric Cardiology, Technical University of Munich, School of Medicine & Health, Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Jos M T Draaisma
- Department of Pediatrics, Radboud Institute for Health Sciences, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
2
|
Yang W, Zhu Y, Tang F, Jian Z, Xiao Y. Cardiac proteomic profiling suggests that hypertrophic and dilated cardiomyopathy share a common pathogenetic pathway of the calcium signalling pathway. Eur J Clin Invest 2023; 53:e14051. [PMID: 37381592 DOI: 10.1111/eci.14051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 06/04/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
OBJECTIVE Hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM) are classified as different diseases but have many similar pathogenic genes and clinical symptoms. Previous research has focused on mutated genes. This study was conducted to identify key molecular mechanisms and explore effective therapeutic targets. METHODS Myocardial tissue was harvested from patients with HCM (n = 3) or DCM (n = 4) during surgery. Hearts donated by healthy traffic accident victims were treated as controls (n = 4). Total proteins were extracted for liquid chromatography-tandem mass spectrometry. Differentially expressed proteins (DEPs) were annotated via GO and KEGG analyses. Selected distinguishing protein abundance was confirmed by western blotting. RESULTS Compared with the control group, there were 121 and 76 DEPs in the HCM and DCM groups, respectively. GO terms for these two comparisons are associated with contraction-related components and actin binding. Additionally, the most significantly upregulated and downregulated proteins were periostin and tropomyosin alpha-3 chain in both comparisons. Moreover, when comparing the HCM and DCM groups, we found 60 significant DEPs, and the GO and KEGG terms are related to the calcium signalling pathway. Expression of the calcium regulation-related protein peptidyl-prolyl cis-trans isomerase (FKBP1A) was significantly upregulated in multiple samples. CONCLUSION HCM and DCM have many mutual pathogenetic pathways. Calcium ion-related processes are among the most significant factors affecting disease development. For HCM and DCM, research on regulating linchpin protein expression or interfering with key calcium-related pathways may be more beneficial than genetic research.
Collapse
Affiliation(s)
- Wenjuan Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yu Zhu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
- Department of Cardiovascular Surgery, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Fuqin Tang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Zhao Jian
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Yingbin Xiao
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| |
Collapse
|
3
|
Crean AM, Small GR, Saleem Z, Maharajh G, Ruel M, Chow BJW. Application of Cardiovascular Computed Tomography to the Assessment of Patients With Hypertrophic Cardiomyopathy. Am J Cardiol 2023; 205:481-492. [PMID: 37683571 DOI: 10.1016/j.amjcard.2023.06.096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 06/24/2023] [Accepted: 06/29/2023] [Indexed: 09/10/2023]
Abstract
Hypertrophic cardiomyopathy is a common inherited cardiac condition in which regional myocardial thickening and scarring can lead to a range of symptoms including breathlessness, dizziness, chest pain, and collapse with loss of consciousness. It is vital to be able to understand the mechanisms behind these epiphenomena and to be able to distinguish, for example, between syncope because of arrhythmia versus syncope because of mechanical outflow tract obstruction. Therefore, we require a technique that can characterize anatomy, physiology, and myocardial substrate. Traditionally, this role has been the preserve of cardiac magnetic resonance (CMR) imaging. This review makes the case for cardiac computed tomography (CT) as an alternative imaging method. We review the use of functional CT to identify the components of outflow tract obstruction (and obstruction at other levels, which may be simultaneous), and as an aid to interventional and surgical planning. We demonstrate the added value of multiplanar isotropic reformats in this condition, particularly in cases where the diagnosis may be more challenging or where complications (such as early apical aneurysm) may be difficult to recognize with 2-dimensional techniques. In conclusion, our aim is to convince readers that cardiac CT is a highly valuable and versatile tool, which deserves wider usage and greater recognition in those caring for patients with hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Andrew M Crean
- Division of Cardiology, Ottawa Heart Institute, Ottawa, Ontario, Canada.
| | - Gary R Small
- Division of Cardiology, Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Zain Saleem
- Division of Cardiology, University of Ottawa, Ottawa, Ontario, Canada
| | - Gyaandeo Maharajh
- Division of Cardiovascular Surgery, Children's Hospital of Eastern Ontario, Ottawa, Canada
| | - Marc Ruel
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Benjamin J W Chow
- Division of Cardiology, Ottawa Heart Institute, Ottawa, Ontario, Canada
| |
Collapse
|
4
|
Tsatsopoulou A, Protonotarios I, Xylouri Z, Papagiannis I, Anastasakis A, Germanakis I, Patrianakos A, Nyktari E, Gavras C, Papadopoulos G, Meditskou S, Lazarou E, Miliou A, Lazaros G. Cardiomyopathies in children: An overview. Hellenic J Cardiol 2023; 72:43-56. [PMID: 36870438 DOI: 10.1016/j.hjc.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Paediatric cardiomyopathies form a heterogeneous group of disorders characterized by structural and electrical abnormalities of the heart muscle, commonly due to a gene variant of the myocardial cell structure. Mostly inherited as a dominant or occasionally recessive trait, they might be part of a syndromic disorder of underlying metabolic or neuromuscular defects or combine early developing extracardiac abnormalities (i.e., Naxos disease). The annual incidence of 1 per 100,000 children appears higher during the first two years of life. Dilated and hypertrophic cardiomyopathy phenotypes share an incidence of 60% and 25%, respectively. Arrhythmogenic right ventricular cardiomyopathy (ARVC), restrictive cardiomyopathy, and left ventricular noncompaction are less commonly diagnosed. Adverse events such as severe heart failure, heart transplantation, or death usually appear early after the initial presentation. In ARVC patients, high-intensity aerobic exercise has been associated with worse clinical outcomes and increased penetrance in at-risk genotype-positive relatives. Acute myocarditis in children has an incidence of 1.4-2.1 cases/per 100,000 children per year, with a 6-14% mortality rate during the acute phase. A genetic defect is considered responsible for the progression to dilated cardiomyopathy phenotype. Similarly, a dilated or arrhythmogenic cardiomyopathy phenotype might emerge with an episode of acute myocarditis in childhood or adolescence. This review provides an overview of childhood cardiomyopathies focusing on clinical presentation, outcome, and pathology.
Collapse
Affiliation(s)
- Adalena Tsatsopoulou
- General Paediatrics and Clinical Research, Private Clinic, Naxos, Greece; Unit of Inherited Cardiac Conditions and Sports Cardiology, 1st Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece; Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Centre, Athens, Greece; Laboratory of Histology and Embryology, Department of Medicine, School of Life Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Protonotarios
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, Hampshire, SO16 6YD, UK
| | - Zafeirenia Xylouri
- University Hospital Southampton NHS Foundation Trust, Tremona Road, Southampton, Hampshire, SO16 6YD, UK
| | - Ioannis Papagiannis
- Department of Paediatric Cardiology and Adult Congenital Heart Disease, Onassis Cardiac Surgery Centre, Athens, Greece
| | - Aris Anastasakis
- Unit of Inherited and Rare Cardiovascular Diseases, Onassis Cardiac Surgery Centre, Athens, Greece
| | - Ioannis Germanakis
- Department of Paediatrics, University Hospital Heraklion, School of Medicine, University of Crete, Heraklion, Greece
| | | | | | | | | | - Soultana Meditskou
- Laboratory of Histology and Embryology, Department of Medicine, School of Life Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Emilia Lazarou
- Unit of Inherited Cardiac Conditions and Sports Cardiology, 1st Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Miliou
- Unit of Inherited Cardiac Conditions and Sports Cardiology, 1st Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece
| | - George Lazaros
- Unit of Inherited Cardiac Conditions and Sports Cardiology, 1st Department of Cardiology, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
5
|
Nakhaei-Rad S, Haghighi F, Bazgir F, Dahlmann J, Busley AV, Buchholzer M, Kleemann K, Schänzer A, Borchardt A, Hahn A, Kötter S, Schanze D, Anand R, Funk F, Kronenbitter AV, Scheller J, Piekorz RP, Reichert AS, Volleth M, Wolf MJ, Cirstea IC, Gelb BD, Tartaglia M, Schmitt JP, Krüger M, Kutschka I, Cyganek L, Zenker M, Kensah G, Ahmadian MR. Molecular and cellular evidence for the impact of a hypertrophic cardiomyopathy-associated RAF1 variant on the structure and function of contractile machinery in bioartificial cardiac tissues. Commun Biol 2023; 6:657. [PMID: 37344639 PMCID: PMC10284840 DOI: 10.1038/s42003-023-05013-8] [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: 06/08/2022] [Accepted: 06/02/2023] [Indexed: 06/23/2023] Open
Abstract
Noonan syndrome (NS), the most common among RASopathies, is caused by germline variants in genes encoding components of the RAS-MAPK pathway. Distinct variants, including the recurrent Ser257Leu substitution in RAF1, are associated with severe hypertrophic cardiomyopathy (HCM). Here, we investigated the elusive mechanistic link between NS-associated RAF1S257L and HCM using three-dimensional cardiac bodies and bioartificial cardiac tissues generated from patient-derived induced pluripotent stem cells (iPSCs) harboring the pathogenic RAF1 c.770 C > T missense change. We characterize the molecular, structural, and functional consequences of aberrant RAF1-associated signaling on the cardiac models. Ultrastructural assessment of the sarcomere revealed a shortening of the I-bands along the Z disc area in both iPSC-derived RAF1S257L cardiomyocytes and myocardial tissue biopsies. The aforementioned changes correlated with the isoform shift of titin from a longer (N2BA) to a shorter isoform (N2B) that also affected the active force generation and contractile tensions. The genotype-phenotype correlation was confirmed using cardiomyocyte progeny of an isogenic gene-corrected RAF1S257L-iPSC line and was mainly reversed by MEK inhibition. Collectively, our findings uncovered a direct link between a RASopathy gene variant and the abnormal sarcomere structure resulting in a cardiac dysfunction that remarkably recapitulates the human disease.
Collapse
Affiliation(s)
- Saeideh Nakhaei-Rad
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Stem Cell Biology and Regenerative Medicine Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fereshteh Haghighi
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Julia Dahlmann
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Alexandra Viktoria Busley
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Marcel Buchholzer
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Karolin Kleemann
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, Giessen, Germany
| | - Andrea Borchardt
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Sebastian Kötter
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Denny Schanze
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Ruchika Anand
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Florian Funk
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Annette Vera Kronenbitter
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Roland P Piekorz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Andreas S Reichert
- Institute of Biochemistry and Molecular Biology I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Marianne Volleth
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany
| | - Matthew J Wolf
- Department of Medicine and Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, University of Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and Departments of Pediatrics and Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, 00146, Rome, Italy
| | - Joachim P Schmitt
- Institute of Pharmacology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Martina Krüger
- Institute of Cardiovascular Physiology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Ingo Kutschka
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Lukas Cyganek
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells", University of Göttingen, Göttingen, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto von Guericke-University, Magdeburg, Germany.
| | - George Kensah
- Clinic for Cardiothoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| | - Mohammad R Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
6
|
Chauhan PK, Sowdhamini R. Transcriptome data analysis of primary cardiomyopathies reveals perturbations in arachidonic acid metabolism. Front Cardiovasc Med 2023; 10:1110119. [PMID: 37288265 PMCID: PMC10242083 DOI: 10.3389/fcvm.2023.1110119] [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: 11/28/2022] [Accepted: 05/09/2023] [Indexed: 06/09/2023] Open
Abstract
Introduction Cardiomyopathies are complex heart diseases with significant prevalence around the world. Among these, primary forms are the major contributors to heart failure and sudden cardiac death. As a high-energy demanding engine, the heart utilizes fatty acids, glucose, amino acid, lactate and ketone bodies for energy to meet its requirement. However, continuous myocardial stress and cardiomyopathies drive towards metabolic impairment that advances heart failure (HF) pathogenesis. So far, metabolic profile correlation across different cardiomyopathies remains poorly understood. Methods In this study, we systematically explore metabolic differences amongst primary cardiomyopathies. By assessing the metabolic gene expression of all primary cardiomyopathies, we highlight the significantly shared and distinct metabolic pathways that may represent specialized adaptations to unique cellular demands. We utilized publicly available RNA-seq datasets to profile global changes in the above diseases (|log2FC| ≥ 0.28 and BH adjusted p-val 0.1) and performed gene set analysis (GSA) using the PAGE statistics on KEGG pathways. Results Our analysis demonstrates that genes in arachidonic acid metabolism (AA) are significantly perturbed across cardiomyopathies. In particular, the arachidonic acid metabolism gene PLA2G2A interacts with fibroblast marker genes and can potentially influence fibrosis during cardiomyopathy. Conclusion The profound significance of AA metabolism within the cardiovascular system renders it a key player in modulating the phenotypes of cardiomyopathies.
Collapse
Affiliation(s)
- Pankaj Kumar Chauhan
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore, India
| | - Ramanathan Sowdhamini
- National Centre for Biological Sciences (Tata Institute of Fundamental Research), Bangalore, India
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
- Institute of Bioinformatics and Applied Biotechnology, Bangalore, India
| |
Collapse
|
7
|
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: 2.0] [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
|
8
|
Lau C, Gul U, Liu B, Captur G, Hothi SS. Cardiovascular Magnetic Resonance Imaging in Familial Dilated Cardiomyopathy. Medicina (B Aires) 2023; 59:medicina59030439. [PMID: 36984439 PMCID: PMC10057087 DOI: 10.3390/medicina59030439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common cause of non-ischaemic heart failure, conferring high morbidity and mortality, including sudden cardiac death due to systolic dysfunction or arrhythmic sudden death. Within the DCM cohort exists a group of patients with familial disease. In this article we review the pathophysiology and cardiac imaging findings of familial DCM, with specific attention to known disease subtypes. The role of advanced cardiac imaging cardiovascular magnetic resonance is still accumulating, and there remains much to be elucidated. We discuss its potential clinical roles as currently known, with respect to diagnostic utility and risk stratification. Advances in such risk stratification may help target pharmacological and device therapies to those at highest risk.
Collapse
Affiliation(s)
- Clement Lau
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
| | - Uzma Gul
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
| | - Boyang Liu
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Gabriella Captur
- MRC Unit for Lifelong Health and Ageing at UCL, University College London, London WC1E 6BT, UK
- Institute of Cardiovascular Science, University College London, London WC1E 6BT, UK
- Centre for Inherited Heart Muscle Conditions, Cardiology Department, The Royal Free Hospital, London NW3 2QG, UK
| | - Sandeep S. Hothi
- New Cross Hospital, Royal Wolverhampton NHS Trust, Wolverhampton WV10 0QP, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
- Correspondence:
| |
Collapse
|
9
|
de Winter JM, Bouman K, Strom J, Methawasin M, Jongbloed JDH, van der Roest W, Wijngaarden JV, Timmermans J, Nijveldt R, van den Heuvel F, Kamsteeg EJ, van Engelen BG, Galli R, Bogaards SJP, Boon RA, van der Pijl RJ, Granzier H, Koeleman B, Amin AS, van der Velden J, van Tintelen JP, van den Berg MP, van Spaendonck-Zwarts KY, Voermans NC, Ottenheijm CAC. KBTBD13 is a novel cardiomyopathy gene. Hum Mutat 2022; 43:1860-1865. [PMID: 36335629 PMCID: PMC10100581 DOI: 10.1002/humu.24499] [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: 02/28/2022] [Revised: 08/22/2022] [Accepted: 09/12/2022] [Indexed: 11/08/2022]
Abstract
KBTBD13 variants cause nemaline myopathy type 6 (NEM6). The majority of NEM6 patients harbors the Dutch founder variant, c.1222C>T, p.Arg408Cys (KBTBD13 p.R408C). Although KBTBD13 is expressed in cardiac muscle, cardiac involvement in NEM6 is unknown. Here, we constructed pedigrees of three families with the KBTBD13 p.R408C variant. In 65 evaluated patients, 12% presented with left ventricle dilatation, 29% with left ventricular ejection fraction< 50%, 8% with atrial fibrillation, 9% with ventricular tachycardia, and 20% with repolarization abnormalities. Five patients received an implantable cardioverter defibrillator, three cases of sudden cardiac death were reported. Linkage analysis confirmed cosegregation of the KBTBD13 p.R408C variant with the cardiac phenotype. Mouse studies revealed that (1) mice harboring the Kbtbd13 p.R408C variant display mild diastolic dysfunction; (2) Kbtbd13-deficient mice have systolic dysfunction. Hence, (1) KBTBD13 is associated with cardiac dysfunction and cardiomyopathy; (2) KBTBD13 should be added to the cardiomyopathy gene panel; (3) NEM6 patients should be referred to the cardiologist.
Collapse
Affiliation(s)
| | - Karlijn Bouman
- Department of Neurology, Radboudumc, Nijmegen, The Netherlands
| | - Joshua Strom
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Mei Methawasin
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Jan D H Jongbloed
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | - Wilma van der Roest
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands
| | | | | | - Robin Nijveldt
- Department of Cardiology, Radboudumc, Nijmegen, The Netherlands
| | | | | | | | - Ricardo Galli
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Reinier A Boon
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Robbert J van der Pijl
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| | - Bobby Koeleman
- Department of Human Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Ahmad S Amin
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - J Peter van Tintelen
- Department of Human Genetics, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Cardiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Karin Y van Spaendonck-Zwarts
- Department of Genetics, University Medical Center Groningen, Groningen, The Netherlands.,Department of Human Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | | | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
10
|
Atrial fibrillation in hypertrophic cardiomyopathy-a contemporary mini review. Hellenic J Cardiol 2022; 67:66-72. [DOI: 10.1016/j.hjc.2022.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 11/21/2022] Open
|
11
|
Migliore L, Galvagni F, Pierantozzi E, Sorrentino V, Rossi D. Allele-specific silencing by RNAi of R92Q and R173W mutations in cardiac troponin T. Exp Biol Med (Maywood) 2022; 247:805-814. [PMID: 35067102 PMCID: PMC9160939 DOI: 10.1177/15353702211072453] [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: 08/25/2021] [Accepted: 12/17/2021] [Indexed: 08/30/2024] Open
Abstract
Autosomal dominant mutations in sarcomere proteins such as the cardiac troponin T (TNNT2) are the main genetic causes of human hypertrophic cardiomyopathy and dilated cardiomyopathy. Allele-specific silencing by RNA interference (ASP-RNAi) holds promise as a therapeutic strategy for downregulating a single mutant allele with minimal suppression of the corresponding wild-type allele. Here, we propose ASP-RNAi as a possible strategy to specifically knockdown mutant alleles coding for R92Q and R173W mutant TNNT2 proteins, identified in hypertrophic and dilated cardiomyopathy, respectively. Different siRNAs were designed and validated by luciferase reporter assay and following analysis in HEK293T cells expressing either the wild-type or mutant TNNT2 alleles. This study is the first exploration of ASP-RNAi on TNNT2-R173W and TNNT2-R92Q mutations in vitro and gives a base for further application of allele silencing as a therapeutic treatment for TNNT2-mutation-associated cardiomyopathies.
Collapse
Affiliation(s)
- Loredana Migliore
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Federico Galvagni
- Department of Biotechnology,
Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Enrico Pierantozzi
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Vincenzo Sorrentino
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Daniela Rossi
- Department of Molecular and
Developmental Medicine, University of Siena, 53100 Siena, Italy
| |
Collapse
|
12
|
Wang X, Wang P, Pei H. Left Atrial Appendage Occlusion in Hypertrophic Cardiomyopathy Patients Should Be Taken Into Account. J Am Coll Cardiol 2021; 78:e157. [PMID: 34736570 DOI: 10.1016/j.jacc.2021.08.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/17/2022]
|
13
|
Bogatyreva FM, Kaplunova VY, Kozhevnikova MV, Shakaryants GA, Yatsenko DA, Emelianov АV, Lishuta AS, Khabarova NV, Privalova EV, Belenkov YN. Assesment of Markers of Endothelial Dysfunction and Myocardial Stress in Patients with Hypertrophic Cardyomyopathy. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2021. [DOI: 10.20996/1819-6446-2021-06-06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Aim. To study the level of serum biomarkers of endothelial dysfunction and myocardial stress in patients with various types of hypertrophic cardiomyopathy (HCM).Material and Methods. 48 patients with hypertrophic cardiomyopathy (27 men and 21 women) were examined, the mean age was 54±13 years. The patients were divided into two groups according to the course options: group 1 - symptomatic stable course (n=14); group 2 - progressive course of symptomatic HCM (n=34). In accordance with the assigned tasks, all patients underwent determination of biological markers of neurohumoral systems in blood serum using immunofluorescence assay analysis: N-terminal fragment of brain natriuretic peptide (NT-proBNP), von Willebrand factor (vWF), endothelin-1, E-selectin with subsequent evaluation and correlation of results with clinical and instrumental characteristics of patients. When performing echocardiography following parameters were assessed: dimensions of the heart chambers, the thickness of the interventricular septum, the thickness of the posterior wall of the left ventricle (LV), LV mass, LV mass index, the ratio between LV filling in diastole (peak E) and atrial systole (peak A)-(E/A), relative myocardial thickness index, left atrial volume index.Results. Both groups showed increased levels of NT-proBNP and endothelin-1. E-selectin and vonWillebrand factor remained within the normal range. There was no statistically significant intergroup difference. There was a correlation between the level of the index of the relative myocardial thickness and the level of NT-proBNP (r=0.30; p=0.04). A correlation was found between the level of the vWF marker and the left atrial volume index (r=0.32; p=0.04). When assessing the association of indicators of intracardiac hemodynamics with other markers (E-selectin and endothelin-1, no statistically significant relationships were found.Conclusion. In the course of the research, it was found that the high activity of endothelin-1 and NT-proBNP reflects endothelial dysfunction and myocardial stress in patients with HCM, especially in patients with a progressive variant of HCM. However, we did not find any changes in the levels of E-selectin and von Willebrand factor, as well as their intergroup differences. These results require additional studies to assess endothelial dysfunction in patients with HCM.
Collapse
Affiliation(s)
- F. M. Bogatyreva
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - V. Yu. Kaplunova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - M. V. Kozhevnikova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - G. A. Shakaryants
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - D. A. Yatsenko
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - А. V. Emelianov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - A. S. Lishuta
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - N. V. Khabarova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - E. V. Privalova
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | - Yu. N. Belenkov
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
14
|
Abreu SM, Trigo C, Pinto FF. Cardiomyopathy. Like mother, like daughter? A new phenotype for a MYH7 mutation. PROGRESS IN PEDIATRIC CARDIOLOGY 2021. [DOI: 10.1016/j.ppedcard.2021.101348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
15
|
De Gaspari M, Basso C, Perazzolo Marra M, Elia S, Bueno Marinas M, Angelini A, Thiene G, Rizzo S. Small Vessel Disease: Another Component of the Hypertrophic Cardiomyopathy Phenotype Not Necessarily Associated with Fibrosis. J Clin Med 2021; 10:jcm10040575. [PMID: 33557065 PMCID: PMC7913811 DOI: 10.3390/jcm10040575] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 02/04/2023] Open
Abstract
Background: Hypertrophic cardiomyopathy (HCM) is characterized by myocardial disarray, small vessel disease (SVD), and fibrosis. The relationship between SVD and replacement-type fibrosis is still unclear. Methods: Histopathologic assessment of replacement-type fibrosis and SVD in HCM patients with either end-stage heart failure (HF) or sudden cardiac death (SCD). Chronic ischemic heart disease (IHD) patients served as controls. Results: Forty HCM hearts, 10 HF and 30 SCD, were studied. Replacement-type fibrosis was detected in all HF and in 57% of SCD cases. In SCD, replacement-type fibrosis was associated with older age, greater septal thickness, SVD prevalence, and score (all p < 0.05). Prevalence of SVD did not show significant differences among SCD, HF, and IHD (73%, 100% and 95%, respectively), while SVD score was higher in HF than IHD and SCD (2.4, 1.95, and 1.18, respectively) and in areas with replacement-type fibrosis vs. those without in HF (3.4 vs. 1.4) and SCD (1.4 vs. 0.8) (all p < 0.05). Conclusions: SVD is a frequent feature in HCM independent of the clinical presentation. A higher SVD score is observed in HCM-HF and in areas with replacement-type fibrosis. Although SVD is part of the HCM phenotype, further remodeling of the microcirculation might occur secondarily to fibrosis.
Collapse
|
16
|
Towards an Enhanced Tool for Quantifying the Degree of LV Hyper-Trabeculation. J Clin Med 2021; 10:jcm10030503. [PMID: 33535420 PMCID: PMC7867055 DOI: 10.3390/jcm10030503] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/22/2021] [Accepted: 01/28/2021] [Indexed: 11/17/2022] Open
Abstract
Left ventricular non-compaction (LVNC) is defined by an increase of trabeculations in left ventricular (LV) endomyocardium. Although LVNC can be in isolation, an increase in hypertrabeculation often accompanies genetic cardiomyopathies. Current methods for quantification of LV trabeculae have limitations. Several improvements are proposed and implemented to enhance a software tool to quantify the trabeculae degree in the LV myocardium in an accurate and automatic way for a population of patients with genetic cardiomyopathies (QLVTHCI). The software tool is developed and evaluated for a population of 59 patients (470 end-diastole cardiac magnetic resonance images). This tool produces volumes of the compact sector and the trabecular area, the proportion between these volumes, and the left ventricular and trabeculated masses. Substantial enhancements are obtained over the manual process performed by cardiologists, so saving important diagnosis time. The parallelization of the detection of the external layer is proposed to ensure real-time processing of a patient, obtaining speed-ups from 7.5 to 1500 with regard to QLVTHCI and the manual process used traditionally by cardiologists. Comparing the method proposed with the fractal proposal to differentiate LVNC and non-LVNC patients among 27 subjects with previously diagnosed cardiomyopathies, QLVTHCI presents a full diagnostic accuracy, while the fractal criteria achieve 78%. Moreover, QLTVHCI can be installed and integrated in hospitals on request, whereas the high cost of the license of the fractal method per year of this tool has prevented reproducibility by other medical centers.
Collapse
|
17
|
Scolari FL, Faganello LS, Garbin HI, Piva E Mattos B, Biolo A. A systematic review of microRNAs in patients with hypertrophic cardiomyopathy. Int J Cardiol 2020; 327:146-154. [PMID: 33212095 DOI: 10.1016/j.ijcard.2020.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/20/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Several microRNAs (miRNA) have been associated with hypertrophic cardiomyopathy (HCM), but studies differ regarding methods employed. In an attempt to understand their role in the disease, we performed a systematic review of studies assessing miRNAs and their association with HCM. METHODS The literature search was based on The Medical Subject Headings (MeSH) terms "Hypertrophic Cardiomyopathy" and "MicroRNA" combined with other synonyms on Embase, Medline and LILACS databases in April 2020. The selected studies and data extraction were independently evaluated. Only human reports with a clear definition of HCM diagnosis were included. RESULTS The search found 68 studies, 13 fulfilled the selection criteria, with a total of 329 patients. Eighty-seven miRNA were differentially expressed in HCM patients, being mir-21, mir-29a and mir-133 the most reported. The miRNA were mainly up-regulated, where mir-29a was up-regulated in 6 studies, followed by mir-133 in 4 and mir-21 in 3. The other miRNAs were mainly up-regulated. Blood samples were evaluated in the majority of patients (86%), but a greater number of miRNAs (79%) were assessed in myocardium. Six studies evaluating the phenotype correlation demonstrated that several miRNAs, mainly mir-1-3p, mir-19b, mir-21, mir-29a, mir-155, and mir-221, were related to either hypertrophy or fibrosis. Mir-29a showed a more consistent phenotypic correlation. CONCLUSION Eighty-seven miRNAs were differentially expressed in HCM patients, the majority in up-regulation. Mir-21, mir-29a and mir-133 were the most reported. Correlation with left ventricular hypertrophy and fibrosis was evaluated in six studies for several miRNAs, nevertheless, mir-29a showed more consistent findings and seems to be a promising biomarker.
Collapse
Affiliation(s)
- Fernando Luís Scolari
- Division of Cardiology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Faculty of Medicine, Universidade Federal do Rio Grande do Sul, 2350 Ramiro Barcelos St, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Lucas Simonetto Faganello
- Department of Cardiac Electrophysiology, McGill University Health Centre, 1001 Decarie Boulevard, Montreal, Quebec H4A 3J1, Canada
| | - Henrique Iahnke Garbin
- Faculty of Medicine, Universidade Federal do Rio Grande do Sul, 2350 Ramiro Barcelos St, Porto Alegre, Rio Grande do Sul, Brazil
| | - Beatriz Piva E Mattos
- Division of Cardiology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Faculty of Medicine, Universidade Federal do Rio Grande do Sul, 2350 Ramiro Barcelos St, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Andreia Biolo
- Division of Cardiology, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Faculty of Medicine, Universidade Federal do Rio Grande do Sul, 2350 Ramiro Barcelos St, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
18
|
Cramer GE, Gommans DHF, Dieker HJ, Michels M, Verheugt F, de Boer MJ, Bakker J, Fouraux MA, Timmermans J, Kofflard M, Brouwer M. Exercise and myocardial injury in hypertrophic cardiomyopathy. Heart 2020; 106:1169-1175. [PMID: 32001622 PMCID: PMC7398456 DOI: 10.1136/heartjnl-2019-315818] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/09/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Objective Troponin and high signal intensity on T2-weighted (HighT2) cardiovascular magnetic resonance imaging (CMRi) are both markers of myocardial injury in hypertrophic cardiomyopathy (HCM). The interplay between exercise and disease development remains uncertain in HCM. We sought to assess the occurrence of postexercise troponin rises and its determinants. Methods Multicentre project on patients with HCM and mutation carriers without hypertrophy (controls). Participants performed a symptom limited bicycle test with hs-cTnT assessment pre-exercise and 6 hours postexercise. Pre-exercise CMRi was performed in patients with HCM to assess measures of hypertrophy and myocardial injury. Depending on baseline troponin (< or >13 ng/L), a rise was defined as a >50% or >20% increase, respectively. Results Troponin rises occurred in 18% (23/127) of patients with HCM and 4% (2/53) in mutation carriers (p=0.01). Comparing patients with HCM with and without a postexercise troponin rise, maximum heart rates (157±19 vs 143±23, p=0.004) and maximal wall thickness (20 mm vs 17 mm, p=0.023) were higher in the former, as was the presence of late gadolinium enhancement (85% vs 57%, p=0.02). HighT2 was seen in 65% (13/20) and 19% (15/79), respectively (p<0.001). HighT2 was the only independent predictor of troponin rise (adjusted odds ratio 7.9; 95% CI 2.7 to 23.3; p<0.001). Conclusions Postexercise troponin rises were seen in about 20% of patients with HCM, almost five times more frequent than in mutation carriers. HighT2 on CMRi may identify a group of particularly vulnerable patients, supporting the concept that HighT2 reflects an active disease state, prone to additional injury after a short episode of high oxygen demand.
Collapse
Affiliation(s)
| | | | | | - Michelle Michels
- Cardiology, Thoraxcenter, Erasmus MC, Rotterdam, Zuid-Holland, The Netherlands
| | - Freek Verheugt
- Cardiology, Radboudumc, Nijmegen, Gelderland, The Netherlands
| | | | - Jeannette Bakker
- Radiology, Albert Schweitzer Ziekenhuis, Dordrecht, Zuid-Holland, The Netherlands
| | - Michael A Fouraux
- Clinical Chemistry, Albert Schweitzer Ziekenhuis, Dordrecht, Zuid-Holland, The Netherlands
| | | | - Marcel Kofflard
- Cardiology, Albert Schweitzer Hospital, Dordrecht, Zuid-Holland, The Netherlands
| | - Marc Brouwer
- Cardiology, Radboudumc, Nijmegen, Gelderland, The Netherlands
| |
Collapse
|
19
|
Yeh JK, Liu WH, Wang CY, Lu JJ, Chen CH, Wu-Chou YH, Chang PY, Chang SC, Yang CH, Tsai ML, Ho MY, Hsieh IC, Wen MS. Targeted Next Generation Sequencing for Genetic Mutations of Dilated Cardiomyopathy. ACTA CARDIOLOGICA SINICA 2019; 35:571-584. [PMID: 31879508 PMCID: PMC6859096 DOI: 10.6515/acs.201911_35(6).20190402a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 04/02/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND Approximately one-third of cases of dilated cardiomyopathy (DCM) are caused by genetic mutations. With new sequencing technologies, numerous variants have been associated with this inherited cardiomyopathy, however the prevalence and genotype-phenotype correlations in different ethnic cohorts remain unclear. This study aimed to investigate the variants in Chinese DCM patients and correlate them with clinical presentations and prognosis. METHODS AND RESULTS From September 2013 to December 2016, 70 index patients underwent DNA sequencing for 12 common disease-causing genes with next generation sequencing. Using a bioinformatics filtering process, 12 rare truncating variants (7 nonsense variants, 4 frameshift variants, and 1 splice site variant) and 29 rare missense variants were identified. Of these, 3 patients were double heterozygotes and 10 patients were compound heterozygotes. Overall, 47.1% (33/70) of the index patients had the seputatively pathogenic variants. The majority (33/41, 80.4%) of these variants were located in titin (TTN). More than 80% of the TTN variants (27/33, 81.8%) were distributed in the A band region of the sarcomere. Patients carrying these variants did not have a different phenotype in disease severity, clinical outcome and reversibility of ventricular function compared with non-carriers. CONCLUSIONS Several new rare variants were identified in a Chinese population in this study, indicating that there are ethnic differences in genetic mutations in DCM patients. TTN remains the major disease-causing gene. Our results could be a reference for future genetic tests in Chinese populations. No specific genotype-phenotype correlations were found, however a prospective large cohort study may be needed to confirm our findings.
Collapse
Affiliation(s)
| | - Wei-Hsiu Liu
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital
| | - Chao-Yung Wang
- Department of Cardiology
- College of Medicine, Chang Gung University, Taoyuan
| | - Jang-Jih Lu
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital
- College of Medicine, Chang Gung University, Taoyuan
| | | | - Yah-Huei Wu-Chou
- Department of Medical Research, Linkou Chang Gung Memorial Hospital and Graduate of Institute of Clinical Medical Science, Chang Gung University, Taoyuan, Taiwan
| | - Pi-Yueh Chang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital
| | - Shih-Cheng Chang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital
| | | | | | | | - I-Chang Hsieh
- Department of Cardiology
- College of Medicine, Chang Gung University, Taoyuan
| | - Ming-Shien Wen
- Department of Cardiology
- College of Medicine, Chang Gung University, Taoyuan
| |
Collapse
|
20
|
Kaltenecker E, Schleihauf J, Meierhofer C, Shehu N, Mkrtchyan N, Hager A, Kühn A, Cleuziou J, Klingel K, Seidel H, Zenker M, Ewert P, Hessling G, Wolf CM. Long-term outcomes of childhood onset Noonan compared to sarcomere hypertrophic cardiomyopathy. Cardiovasc Diagn Ther 2019; 9:S299-S309. [PMID: 31737538 DOI: 10.21037/cdt.2019.05.01] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background To compare outcome and cardiac pathology between patients with Noonan syndrome (N-HCM) and sarcomere protein-associated (S-HCM) childhood onset hypertrophic cardiomyopathy (HCM). Methods Clinical data were recorded from medical charts. Primary endpoint was survival. Secondary endpoints were survival without hospitalization, without intervention or without arrhythmic events. Functional clinical status and results from genetic testing, imaging, electrocardiographic (ECG) studies, cardiopulmonary exercise testing (CPET) and histopathology were compared between groups. Results Childhood HCM was diagnosed in 29 N-HCM and 34 S-HCM patients. Follow-up time was greater than 10 years in more than half of all patients. Mortality was below 7% and not different between groups. Children with N-HCM presented at a younger age and there was less time of survival without hospitalization for heart failure or intervention in N-HCM compared to S-HCM patients. Clinical functional status improved over time in N-HCM patients. On long-term follow-up, left ventricular posterior wall thickness indexed to body surface area decreased in N-HCM and increased in S-HCM patients. There was a trend to lower risk for severe arrhythmic events in N-HCM patients and only S-HCM individuals received an implantable cardioverter-defibrillator. There were no differences between groups in ventricular function, ECG and CPET parameters. Myocardial fibrosis as assessed by histopathology of myocardial specimens and cardiovascular magnetic resonance with late gadolinium enhancement or T1 mapping was present in both groups. Conclusions When compared to S-HCM patients, children with N-HCM have increased morbidity during early disease course, but favorable long-term outcome with low mortality, stagnation of myocardial hypertrophy, and low risk for malignant arrhythmias.
Collapse
Affiliation(s)
- Emanuel Kaltenecker
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Julia Schleihauf
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Christian Meierhofer
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Nerejda Shehu
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Naira Mkrtchyan
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Alfred Hager
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Andreas Kühn
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Julie Cleuziou
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University of Munich, Munich, Germany.,(INSURE) Institute for Translational Cardiac Surgery, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Heide Seidel
- Institute of Human Genetics, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Martin Zenker
- Institute of Human Genetics, University Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Peter Ewert
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Gabriele Hessling
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Cordula M Wolf
- Department of Congenital Heart Disease and Pediatric Cardiology, German Heart Center Munich, Technical University of Munich, Munich, Germany
| |
Collapse
|
21
|
Rizzo S, Carturan E, De Gaspari M, Pilichou K, Thiene G, Basso C. Update on cardiomyopathies and sudden cardiac death. Forensic Sci Res 2019; 4:202-210. [PMID: 31489386 PMCID: PMC6713087 DOI: 10.1080/20961790.2019.1631957] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Sudden cardiac death (SCD) remains a leading mode of death in western countries. Since SCD can be the first and last clinical presentation of the underlying disease, autopsy could be the only medical examination available for early diagnosis and it should be performed according to the guidelines of the Association for European Cardiovascular Pathology. Although the vast majority of SCD are due to coronary artery disease, non-ischemic causes of SCD do exist and are prevalent in young people with structural (i.e. arrhythmogenic, hypertrophic and inflammatory cardiomyopathy) and non-structural (ion channel diseases) cardiomyopathies, accounting for up to one half of cases. A standardized autopsy protocol, in combination with blood sampling to ensure feasibility of postmortem molecular testing if needed, is mandatory. The pathologist is called to provide the correct diagnosis and to advice the relatives on the need of a cascade clinical and genetic screening in the presence of a heredo-familial disease.
Collapse
Affiliation(s)
- Stefania Rizzo
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| | - Elisa Carturan
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| | - Monica De Gaspari
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| | - Kalliopi Pilichou
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| | - Gaetano Thiene
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| | - Cristina Basso
- Cardiovascular Pathology, Department of Cardio-Thoracic-Vascular Sciences & Public Health and Azienda Ospedaliera, University of Padua Medical School, Padua, Italy
| |
Collapse
|
22
|
van der Velden J, Tocchetti CG, Varricchi G, Bianco A, Sequeira V, Hilfiker-Kleiner D, Hamdani N, Leite-Moreira AF, Mayr M, Falcão-Pires I, Thum T, Dawson DK, Balligand JL, Heymans S. Metabolic changes in hypertrophic cardiomyopathies: scientific update from the Working Group of Myocardial Function of the European Society of Cardiology. Cardiovasc Res 2019; 114:1273-1280. [PMID: 29912308 PMCID: PMC6054261 DOI: 10.1093/cvr/cvy147] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022] Open
Abstract
Disturbed metabolism as a consequence of obesity and diabetes may cause cardiac diseases (recently highlighted in the cardiovascular research spotlight issue on metabolic cardiomyopathies).1 In turn, the metabolism of the heart may also be disturbed in genetic and acquired forms of hypertrophic cardiac disease. Herein, we provide an overview of recent insights on metabolic changes in genetic hypertrophic cardiomyopathy and discuss several therapies, which may be explored to target disturbed metabolism and prevent onset of cardiac hypertrophy. This article is part of the Mini Review Series from the Varenna 2017 meeting of the Working Group of Myocardial Function of the European Society of Cardiology.
Collapse
Affiliation(s)
- Jolanda van der Velden
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.,Netherlands Heart Institute, Utrecht, The Netherlands
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy
| | - Anna Bianco
- Department of Translational Medical Sciences, Federico II University, Naples, NA, Italy.,Department of Cardiology, Maastricht University Medical Center & CARIM, Maastricht University, Maastricht, The Netherlands
| | - Vasco Sequeira
- Amsterdam UMC, Vrije Universiteit Amsterdam, Physiology, Amsterdam Cardiovascular Sciences, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Denise Hilfiker-Kleiner
- Molecular Cardiology, Department of Cardiology and Angiology, Medical School Hannover, Germany
| | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University Bochum, Bochum, Germany
| | - Adelino F Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research Centre, University of Porto, Porto, Portugal
| | - Manuel Mayr
- The James Black Centre & King's British Heart Foundation Centre, King's College, University of London, London, UK
| | - Ines Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, Cardiovascular Research Centre, University of Porto, Porto, Portugal
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,National Heart and Lung Institute, Imperial College London, London, UK.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Dana K Dawson
- School of Medicine & Dentistry, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics, Institut de Recherche Experimentale et Clinique (IREC), and Clinique Universitaire Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Stephane Heymans
- Netherlands Heart Institute, Utrecht, The Netherlands.,Department of Cardiology, Maastricht University Medical Center & CARIM, Maastricht University, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Leuven University, Leuven, Belgium
| |
Collapse
|
23
|
Wijnker PJ, Sequeira V, Kuster DW, van der Velden J. Hypertrophic Cardiomyopathy: A Vicious Cycle Triggered by Sarcomere Mutations and Secondary Disease Hits. Antioxid Redox Signal 2019; 31:318-358. [PMID: 29490477 PMCID: PMC6602117 DOI: 10.1089/ars.2017.7236] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 02/23/2018] [Accepted: 02/25/2018] [Indexed: 02/06/2023]
Abstract
Significance: Hypertrophic cardiomyopathy (HCM) is a cardiac genetic disease characterized by left ventricular hypertrophy, diastolic dysfunction, and myocardial disarray. Disease onset occurs between 20 and 50 years of age, thus affecting patients in the prime of their life. HCM is caused by mutations in sarcomere proteins, the contractile building blocks of the heart. Despite increased knowledge of causal mutations, the exact path from genetic defect leading to cardiomyopathy is complex and involves additional disease hits. Recent Advances: Laboratory-based studies indicate that HCM development not only depends on the primary sarcomere impairment caused by the mutation but also on secondary disease-related alterations in the heart. Here we propose a vicious mutation-induced disease cycle, in which a mutation-induced energy depletion alters cellular metabolism with increased mitochondrial work, which triggers secondary disease modifiers that will worsen disease and ultimately lead to end-stage HCM. Critical Issues: Evidence shows excessive cellular reactive oxygen species (ROS) in HCM patients and HCM animal models. Oxidative stress markers are increased in the heart (oxidized proteins, DNA, and lipids) and serum of HCM patients. In addition, increased mitochondrial ROS production and changes in endogenous antioxidants are reported in HCM. Mutant sarcomeric protein may drive excessive levels of cardiac ROS via changes in cardiac efficiency and metabolism, mitochondrial activation and/or dysfunction, impaired protein quality control, and microvascular dysfunction. Future Directions: Interventions restoring metabolism, mitochondrial function, and improved ROS balance may be promising therapeutic approaches. We discuss the effects of current HCM pharmacological therapies and potential future therapies to prevent and reverse HCM. Antioxid. Redox Signal. 31, 318-358.
Collapse
Affiliation(s)
- Paul J.M. Wijnker
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Vasco Sequeira
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Diederik W.D. Kuster
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| |
Collapse
|
24
|
Sequeira V, Bertero E, Maack C. Energetic drain driving hypertrophic cardiomyopathy. FEBS Lett 2019; 593:1616-1626. [PMID: 31209876 DOI: 10.1002/1873-3468.13496] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common form of hereditary cardiomyopathy and is mainly caused by mutations of genes encoding cardiac sarcomeric proteins. HCM is characterized by hypertrophy of the left ventricle, frequently involving the septum, that is not explained solely by loading conditions. HCM has a heterogeneous clinical profile, but diastolic dysfunction and ventricular arrhythmias represent two dominant features of the disease. Preclinical evidence indicates that the enhanced Calcium (Ca2+ ) sensitivity of the myofilaments plays a key role in the pathophysiology of HCM. Notably, this is not always a direct consequence of sarcomeric mutations, but can also result from secondary mutation-driven alterations. Here, we review experimental and clinical evidence indicating that increased myofilament Ca2+ sensitivity lies upstream of numerous cellular derangements which potentially contribute to the progression of HCM toward heart failure and sudden cardiac death.
Collapse
Affiliation(s)
- Vasco Sequeira
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| | - Edoardo Bertero
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany
| |
Collapse
|
25
|
Sarantis P, Gaitanaki C, Beis D. Ventricular remodeling of single-chambered myh6 -/- adult zebrafish hearts occurs via a hyperplastic response and is accompanied by elastin deposition in the atrium. Cell Tissue Res 2019; 378:279-288. [PMID: 31129720 DOI: 10.1007/s00441-019-03044-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 05/07/2019] [Indexed: 12/19/2022]
Abstract
Zebrafish (Danio rerio) is widely used as an animal model to understand the pathophysiology of cardiovascular diseases. Here, we present the adult cardiac phenotype of weak atrium, myh6-/-, which carry mutations in the zebrafish atrial myosin heavy chain. Homozygous mutants survive to adulthood and are fertile despite their initial weak atrial beat. In adult mutants, the atrium remains hypoplastic and shows elastin deposition while mutant ventricles exhibit increased size. In mammals, hypertrophy is the most common mechanism resulting in cardiomegaly. Using immunohistochemistry and confocal microscopy to measure cardiomyocyte cell size, density and proliferation, we show that the enlargement of the myh6-/- ventricle is predominantly due to hyperplasia. However, we identified similar transcriptional profiles to the mammalian hypertrophy response via RT-PCR of the hyperplastic ventricles. Furthermore, we show activation of the ER-stress pathway by western blot analysis. In conclusion, we can assume, based on our model, that molecular signaling pathways associated with hypertrophy in mammals, in combination with ER-stress activation, result in hyperplasia in zebrafish. In addition, to our knowledge, this is the first time to report elastin deposition in the atrium.
Collapse
Affiliation(s)
- Panagiotis Sarantis
- Zebrafish Disease Models lab, Center for Clinical Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece
- Department of Animal & Human Physiology, School of Biology, National and Kapodistrian University of Athens, University Campus, 157 84, Athens, Greece
| | - Catherine Gaitanaki
- Department of Animal & Human Physiology, School of Biology, National and Kapodistrian University of Athens, University Campus, 157 84, Athens, Greece
| | - Dimitris Beis
- Zebrafish Disease Models lab, Center for Clinical Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, 11527, Athens, Greece.
| |
Collapse
|
26
|
The co-segregation of the MYL2 R58Q mutation in Chinese hypertrophic cardiomyopathy family and its pathological effect on cardiomyopathy disarray. Mol Genet Genomics 2019; 294:1241-1249. [PMID: 31104103 DOI: 10.1007/s00438-019-01578-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/08/2019] [Indexed: 01/19/2023]
Abstract
Hypertrophic cardiomyopathy (HCM), a major cause of sudden death in youth, is largely affected by genetic factors. The R58Q mutation in the MYL2 gene was identified in some HCM patients and was considered as a deleterious HCM mutation. However, the passing of R58Q between generations along with HCM symptoms was observed only in small families with only two or three members; thus, whether R58Q is as deleterious as previously claimed remains questionable. Here, we reported a large four-generation Chinese family, and found that R58Q existed in all six members with HCM and two healthy juveniles who had not yet developed HCM yet, and presumably in three deceased members who suffered from sudden death. In addition, we also found that compared with other mutations, R58Q had a more severe effect on the cellular level. Therefore, we confirmed that R58Q could be passed from generation to generation along with HCM symptoms and that it was indeed a deleterious mutation for HCM. However, further study is needed to identify additional factors that may determine the various symptoms shown in different family members within the same family.
Collapse
|
27
|
Olivotto I, Camici PG, Merlini PA, Rapezzi C, Patten M, Climent V, Sinagra G, Tomberli B, Marin F, Ehlermann P, Maier LS, Fornaro A, Jacobshagen C, Ganau A, Moretti L, Hernandez Madrid A, Coppini R, Reggiardo G, Poggesi C, Fattirolli F, Belardinelli L, Gensini G, Mugelli A. Efficacy of Ranolazine in Patients With Symptomatic Hypertrophic Cardiomyopathy: The RESTYLE-HCM Randomized, Double-Blind, Placebo-Controlled Study. Circ Heart Fail 2019; 11:e004124. [PMID: 29321131 DOI: 10.1161/circheartfailure.117.004124] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/18/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The late sodium current inhibitor ranolazine reverses the main electrophysiological and mechanical abnormalities of human hypertrophic cardiomyopathy (HCM) cardiomyocytes in vitro, suggesting potential clinical benefit. We aimed to assess the effect of ranolazine on functional capacity, symptomatic status, diastolic function, and arrhythmias in HCM. METHODS AND RESULTS In this multicenter, double-blind, phase 2 study, 80 adult patients with nonobstructive HCM (age 53±14 years, 34 women) were randomly assigned to placebo (n=40) or ranolazine 1000 mg bid (n=40) for 5 months. The primary end point was change in peak VO2 compared with baseline using cardiopulmonary exercise test. Echocardiographic lateral and septal E/E' ratio, prohormone brain natriuretic peptide levels, 24-hour Holter arrhythmic profile, and quality of life were assessed. Ranolazine was safe and well tolerated. Overall, there was no significant difference in VO2 peak change at 5 months in the ranolazine versus placebo group (delta 0.15±3.96 versus -0.02±4.25 mL/kg per minute; P=0.832). Ranolazine treatment was associated with a reduction in 24-hour burden of premature ventricular complexes compared with placebo (>50% reduction versus baseline in 61% versus 31%, respectively; P=0.042). However, changes in prohormone brain natriuretic peptide levels did not differ in the ranolazine compared with the placebo group (geometric mean median [interquartile range], -3 pg/mL [-107, 142 pg/mL] versus 78 pg/mL [-71, 242 pg/mL]; P=0.251). Furthermore, E/E' ratio and quality of life scores showed no significant difference. CONCLUSIONS In patients with nonobstructive HCM, ranolazine showed no overall effect on exercise performance, plasma prohormone brain natriuretic peptide levels, diastolic function, or quality of life. The drug showed an excellent safety profile and was associated with reduced premature ventricular complex burden. Late sodium current inhibition does not seem to improve functional capacity in HCM. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrialsregister.eu. Unique identifier: 2011-004507-20.
Collapse
Affiliation(s)
- Iacopo Olivotto
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Paolo G Camici
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Piera Angelica Merlini
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Claudio Rapezzi
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Monica Patten
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Vicent Climent
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Gianfranco Sinagra
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Benedetta Tomberli
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.).
| | - Francisco Marin
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Philipp Ehlermann
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Lars S Maier
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Alessandra Fornaro
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Claudius Jacobshagen
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Antonello Ganau
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Luciano Moretti
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Antonio Hernandez Madrid
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Raffaele Coppini
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Giorgio Reggiardo
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Corrado Poggesi
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Francesco Fattirolli
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Luiz Belardinelli
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Gianfranco Gensini
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| | - Alessandro Mugelli
- >From the Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy (I.O., B.T., A.F.); Vita Salute University and Scientific Institute San Raffaele, Milan, Italy (P.G.C.); Dipartimento Cardiotoracovascolare, Ospedale Niguarda, Milan, Italy (P.A.M.); Ospedale S. Orsola Malpighi, Bologna, Italy (C.R.); Clinic of General and Interventional Cardiology, University Heart Center, Hamburg, Germany (M.P.); Cardiology Department, Hospital General Universitario de Alicante, ISABIAL - FISABIO, Alicante, Spain (V.C.); Cardiovascular Department, Ospedale di Cattinara, Trieste, Italy (G.S.); Hospital Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain (F.M.); University Hospital, Heidelberg, Germany (P.E.); University Hospital Regensburg, Germany (L.S.M.); Herz zentrum Georg-August-Universitaet, Göttingen, Germany (C.J.); Department of Clinical and Experimental Medicine, Cardiology, Sassari Hospital, Sassari, Italy (A.G.); Ospedale Mazzoni, Ascoli Piceno, Italy (L.M.); Hospital Ramòn y Cajal, Alcalá University, Madrid, Spain (A.H.M.); Department Neurofarba, University of Florence, Italy (R.C., A.M.); Medi Service, Genoa, Italy (G.R.); Department of Experimental and Clinical Medicine, University of Florence, Italy (C.P., F.F.); Gilead Sciences, Foster City, CA (L.B.); and CESMAV, Florence, Italy (G.G.)
| |
Collapse
|
28
|
Lorén CE, Dahl CP, Do L, Almaas VM, Geiran OR, Mörner S, Hellman U. Low Molecular Mass Myocardial Hyaluronan in Human Hypertrophic Cardiomyopathy. Cells 2019; 8:cells8020097. [PMID: 30699940 PMCID: PMC6406527 DOI: 10.3390/cells8020097] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/11/2019] [Accepted: 01/23/2019] [Indexed: 12/23/2022] Open
Abstract
During the development of hypertrophic cardiomyopathy, the heart returns to fetal energy metabolism where cells utilize more glucose instead of fatty acids as a source of energy. Metabolism of glucose can increase synthesis of the extracellular glycosaminoglycan hyaluronan, which has been shown to be involved in the development of cardiac hypertrophy and fibrosis. The aim of this study was to investigate hyaluronan metabolism in cardiac tissue from patients with hypertrophic cardiomyopathy in relation to cardiac growth. NMR and qRT-PCR analysis of human cardiac tissue from hypertrophic cardiomyopathy patients and healthy control hearts showed dysregulated glucose and hyaluronan metabolism in the patients. Gas phase electrophoresis revealed a higher amount of low molecular mass hyaluronan and larger cardiomyocytes in cardiac tissue from patients with hypertrophic cardiomyopathy. Histochemistry showed high concentrations of hyaluronan around individual cardiomyocytes in hearts from hypertrophic cardiomyopathy patients. Experimentally, we could also observe accumulation of low molecular mass hyaluronan in cardiac hypertrophy in a rat model. In conclusion, the development of hypertrophic cardiomyopathy with increased glucose metabolism affected both hyaluronan molecular mass and amount. The process of regulating cardiomyocyte size seems to involve fragmentation of hyaluronan.
Collapse
Affiliation(s)
- Christina E Lorén
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Christen P Dahl
- Department of Cardiology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
- Department of Clinical Medicine, UiT, the Arctic University of Norway, 9019 Tromsø, Norway.
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0372 Oslo, Norway.
| | - Lan Do
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Vibeke M Almaas
- Department of Cardiology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
| | - Odd R Geiran
- Faculty of Medicine, University of Oslo, 0318 Oslo, Norway.
- Department of Thoracic and Cardiovascular Surgery, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway.
| | - Stellan Mörner
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| | - Urban Hellman
- Cardiology, Heart Centre, Department of Public Health and Clinical Medicine, Umeå University, 901 85 Umeå, Sweden.
| |
Collapse
|
29
|
Hypertrophic cardiomyopathy-linked variants of cardiac myosin-binding protein C3 display altered molecular properties and actin interaction. Biochem J 2018; 475:3933-3948. [PMID: 30446606 DOI: 10.1042/bcj20180685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 12/31/2022]
Abstract
The most common inherited cardiac disorder, hypertrophic cardiomyopathy (HCM), is characterized by thickening of heart muscle, for which genetic mutations in cardiac myosin-binding protein C3 (c-MYBPC3) gene, is the leading cause. Notably, patients with HCM display a heterogeneous clinical presentation, onset and prognosis. Thus, delineating the molecular mechanisms that explain how disparate c-MYBPC3 variants lead to HCM is essential for correlating the impact of specific genotypes on clinical severity. Herein, five c-MYBPC3 missense variants clinically associated with HCM were investigated; namely V1 (R177H), V2 (A216T), V3 (E258K), V4 (E441K) and double mutation V5 (V3 + V4), all located within the C1 and C2 domains of MyBP-C, a region known to interact with sarcomeric protein, actin. Injection of the variant complementary RNAs in zebrafish embryos was observed to recapitulate phenotypic aspects of HCM in patients. Interestingly, V3- and V5-cRNA injection produced the most severe zebrafish cardiac phenotype, exhibiting increased diastolic/systolic myocardial thickness and significantly reduced heart rate compared with control zebrafish. Molecular analysis of recombinant C0-C2 protein fragments revealed that c-MYBPC3 variants alter the C0-C2 domain secondary structure, thermodynamic stability and importantly, result in a reduced binding affinity to cardiac actin. V5 (double mutant), displayed the greatest protein instability with concomitant loss of actin-binding function. Our study provides specific mechanistic insight into how c-MYBPC3 pathogenic variants alter both functional and structural characteristics of C0-C2 domains leading to impaired actin interaction and reduced contractility, which may provide a basis for elucidating the disease mechanism in HCM patients with c- MYBPC3 mutations.
Collapse
|
30
|
Garg L, Gupta M, Sabzwari SRA, Agrawal S, Agarwal M, Nazir T, Gordon J, Bozorgnia B, Martinez MW. Atrial fibrillation in hypertrophic cardiomyopathy: prevalence, clinical impact, and management. Heart Fail Rev 2018; 24:189-197. [DOI: 10.1007/s10741-018-9752-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
31
|
Point mutations in the tri-helix bundle of the M-domain of cardiac myosin binding protein-C influence systolic duration and delay cardiac relaxation. J Mol Cell Cardiol 2018; 119:116-124. [PMID: 29729251 DOI: 10.1016/j.yjmcc.2018.05.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 04/06/2018] [Accepted: 05/01/2018] [Indexed: 11/23/2022]
Abstract
Cardiac myosin binding protein-C (cMyBP-C) is an essential regulatory protein required for proper systolic contraction and diastolic relaxation. We previously showed that N'-terminal domains of cMyBP-C stimulate contraction by binding to actin and activating the thin filament in vitro. In principle, thin filament activating effects of cMyBP-C could influence contraction and relaxation rates, or augment force amplitude in vivo. cMyBP-C binding to actin could also contribute to an internal load that slows muscle shortening velocity as previously hypothesized. However, the functional significance of cMyBP-C binding to actin has not yet been established in vivo. We previously identified an actin binding site in the regulatory M-domain of cMyBP-C and described two missense mutations that either increased (L348P) or decreased (E330K) binding affinity of recombinant cMyBP-C N'-terminal domains for actin in vitro. Here we created transgenic mice with either the L348P or E330K mutations to determine the functional significance of cMyBP-C binding to actin in vivo. Results showed that enhanced binding of cMyBP-C to actin in L348P-Tg mice prolonged the time to end-systole and slowed relaxation rates. Reduced interactions between cMyBP-C and actin in E330K-Tg mice had the opposite effect and significantly shortened the duration of ejection. Neither mouse model displayed overt systolic dysfunction, but L348P-Tg mice showed diastolic dysfunction presumably resulting from delayed relaxation. We conclude that cMyBP-C binding to actin contributes to sustained thin filament activation at the end of systole and during isovolumetric relaxation. These results provide the first functional evidence that cMyBP-C interactions with actin influence cardiac function in vivo.
Collapse
|
32
|
Ferrantini C, Pioner JM, Mazzoni L, Gentile F, Tosi B, Rossi A, Belardinelli L, Tesi C, Palandri C, Matucci R, Cerbai E, Olivotto I, Poggesi C, Mugelli A, Coppini R. Late sodium current inhibitors to treat exercise-induced obstruction in hypertrophic cardiomyopathy: an in vitro study in human myocardium. Br J Pharmacol 2018; 175:2635-2652. [PMID: 29579779 PMCID: PMC6003658 DOI: 10.1111/bph.14223] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022] Open
Abstract
Background and Purpose In 30–40% of hypertrophic cardiomyopathy (HCM) patients, symptomatic left ventricular (LV) outflow gradients develop only during exercise due to catecholamine‐induced LV hypercontractility (inducible obstruction). Negative inotropic pharmacological options are limited to β‐blockers or disopyramide, with low efficacy and tolerability. We assessed the potential of late sodium current (INaL)‐inhibitors to treat inducible obstruction in HCM. Experimental Approach The electrophysiological and mechanical responses to β‐adrenoceptor stimulation were studied in human myocardium from HCM and control patients. Effects of INaL‐inhibitors (ranolazine and GS‐967) in HCM samples were investigated under conditions simulating rest and exercise. Key Results In cardiomyocytes and trabeculae from 18 surgical septal samples of patients with obstruction, the selective INaL‐inhibitor GS‐967 (0.5 μM) hastened twitch kinetics, decreased diastolic [Ca2+] and shortened action potentials, matching the effects of ranolazine (10μM). Mechanical responses to isoprenaline (inotropic and lusitropic) were comparable in HCM and control myocardium. However, isoprenaline prolonged action potentials in HCM myocardium, while it shortened them in controls. Unlike disopyramide, neither GS‐967 nor ranolazine reduced force at rest. However, in the presence of isoprenaline, they reduced Ca2+‐transient amplitude and twitch tension, while the acceleration of relaxation was maintained. INaL‐inhibitors were more effective than disopyramide in reducing contractility during exercise. Finally, INaL‐inhibitors abolished arrhythmias induced by isoprenaline. Conclusions and Implications Ranolazine and GS‐967 reduced septal myocardium tension during simulated exercise in vitro and therefore have the potential to ameliorate symptoms caused by inducible obstruction in HCM patients, with some advantages over disopyramide and β‐blockers.
Collapse
Affiliation(s)
- Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.,Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Josè Manuel Pioner
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luca Mazzoni
- Department NeuroFarBa, University of Florence, Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Benedetta Tosi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessandra Rossi
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Palandri
- Department NeuroFarBa, University of Florence, Florence, Italy
| | - Rosanna Matucci
- Department NeuroFarBa, University of Florence, Florence, Italy
| | | | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | |
Collapse
|
33
|
Olivotto I, Finocchiaro G, Maurizi N, Crotti L. Common presentation of rare cardiac diseases: Arrhythmias. Int J Cardiol 2018; 257:351-357. [PMID: 29506731 DOI: 10.1016/j.ijcard.2018.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 12/13/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
Ventricular or supraventricular ectopic beats or atrial fibrillation may be the first presentation of uncommon cardiac disease, both acquired and genetically determined. In some patients, these manifestations can be the first sign of the underlying cardiac disorder. In others, however, they are also important as prognostic indicators, reflecting electrical instability and risk. Most cardiology clinics are busy environments where the implementation of complex diagnostic algorithms is not feasible. However, it is equally impossible to reach a final diagnosis, among the thousands of rare diseases that involve the heart, moving from a first line clinical and instrumental examination. Cardiac and extra-cardiac red flags, an accurate family and clinical history and ECG interpretation may be of help in identifying a rare disease. Advanced imaging and laboratory testing at experienced referral centers is then necessary to reach a final diagnosis, but the first step in the right direction, based on these simple elements, is the most important. We here review arrhythmic presentations of rare or relatively rare diseases, and suggest a simple "rule out-rule in" approach to help direct clinical suspicion and minimize risk of neglect.
Collapse
Affiliation(s)
- Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy.
| | - Gherardo Finocchiaro
- Cardiology Clinical and Academic Group, St George's University of London, United Kingdom
| | - Niccolò Maurizi
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Lia Crotti
- IRCCS Istituto Auxologico Italiano, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics, Milan, Italy; Department of Cardiovascular, Neural and Metabolic Sciences, San Luca Hospital, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medicine and Surgery, University of Milan Bicocca, Italy
| |
Collapse
|
34
|
Boban M, Pesa V, Beck N, Manola S, Zulj M, Rotim A, Vcev A. Supplementary Diagnostic Landmarks of Left Ventricular Non-Compaction on Magnetic Resonance Imaging. Yonsei Med J 2018; 59:63-71. [PMID: 29214778 PMCID: PMC5725366 DOI: 10.3349/ymj.2018.59.1.63] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/09/2017] [Accepted: 09/12/2017] [Indexed: 01/11/2023] Open
Abstract
PURPOSE Diagnostic criteria for left ventricular non-compaction (LVNC) are still a matter of dispute. The aim of our present study was to test the diagnostic value of two novel diagnostic cardiac magnetic resonance (CMR) parameters: proof of non-compact (NC) myocardium blood flow using T2 sequences and changes in geometry of the left ventricle. MATERIALS AND METHODS The study included cases with LVNC and controls, from a data base formed in a period of 3.5 years (n=1890 exams), in which CMR protocol included T2 sequences. Measurement of perpendicular maximal and minimal end diastolic dimensions in the region with NC myocardium from short axis plane was recorded, and calculated as a ratio (MaxMinEDDR), while flow through trabecula was proven by intracavital T2-weighted hyperintensity (ICT2HI). LVNC diagnosis met the following three criteria: thickening of compact (C) layer, NC:C>2.3:1 and NC>20%LV. RESULTS The study included 200 patients; 71 with LVNC (35.5%; i.e., 3.76% of CMRs) and 129 (64.5%) controls. MaxMinEDDR in patients with LVNC was significantly different from that in controls (1.17±0.08 vs. 1.06±0.04, respectively; p<0.001). MaxMinEDDR >1.10 had sensitivity of 91.6% [95% confidence intervals (CI) 82.5-96.8], specificity of 85.3% (95% CI 78.0-90.0), and area under curve (AUC) 0.919 (95% CI 0.872-0.953; p<0.001) for LVNC. Existence of ICT2HI had sensitivity of 100.0% (95% CI 94.9-100.0), specificity of 91.5% (95% CI 85.3-95.7), and AUC 0.957 (95% CI 0.919-0.981; p<0.001) for LVNC. CONCLUSION Two additional diagnostic parameters for LVNC were identified in this study. ICT2HI and geometric eccentricity of the ventricle both had relatively high sensitivity and specificity for diagnosing LVNC.
Collapse
Affiliation(s)
- Marko Boban
- Department of Cardiology, University Hospital "Thalassotherapia Opatija," Medical Faculty University of Rijeka, Opatija, Croatia
- Department of Internal Medicine, Medical Faculty "J.J. Strossmayer" University of Osijek, Osijek, Croatia
- Department of Radiology, University Hospital "Thalassotherapia Opatija," Opatija, Croatia.
| | - Vladimir Pesa
- Department of Cardiology, University Hospital "Thalassotherapia Opatija," Medical Faculty University of Rijeka, Opatija, Croatia
- Department of Radiology, University Hospital "Thalassotherapia Opatija," Opatija, Croatia
| | - Natko Beck
- Department of Cardiology, University Hospital "Thalassotherapia Opatija," Medical Faculty University of Rijeka, Opatija, Croatia
- Department of Radiology, University Hospital "Thalassotherapia Opatija," Opatija, Croatia
| | - Sime Manola
- Department of Cardiology-Arrhythmology and Electrophysiology, University Hospital "Sestre Milosrdnice," Zagreb, Croatia
| | - Marinko Zulj
- Department of Internal Medicine, Medical Faculty "J.J. Strossmayer" University of Osijek, Osijek, Croatia
| | - Ante Rotim
- Department of Cardiology, University Hospital "Thalassotherapia Opatija," Medical Faculty University of Rijeka, Opatija, Croatia
| | - Aleksandar Vcev
- Department of Internal Medicine, Medical Faculty "J.J. Strossmayer" University of Osijek, Osijek, Croatia
| |
Collapse
|
35
|
Gal D, Sipido KR, Vandevelde W. Editorial highlights from Cardiovascular Research. Cardiovasc Res 2017; 113:e64-e68. [PMID: 29186440 DOI: 10.1093/cvr/cvx210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Diane Gal
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| | - Karin R Sipido
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| | - Wouter Vandevelde
- Division of Experimental Cardiology, Department of Cardiovascular Sciences, Campus Gasthuisberg, KU Leuven, Belgium
| |
Collapse
|
36
|
Sabater-Molina M, Pérez-Sánchez I, Hernández del Rincón J, Gimeno J. Genetics of hypertrophic cardiomyopathy: A review of current state. Clin Genet 2017; 93:3-14. [DOI: 10.1111/cge.13027] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022]
Affiliation(s)
- M. Sabater-Molina
- Inherited Cardiac Disease Unit; University Hospital Virgen Arrixaca; Murcia Spain
- Internal Medicine Department, University of Murcia; Murcia Spain
| | - I. Pérez-Sánchez
- Inherited Cardiac Disease Unit; University Hospital Virgen Arrixaca; Murcia Spain
| | - J.P. Hernández del Rincón
- Internal Medicine Department, University of Murcia; Murcia Spain
- Pathology Department; Institute of Legal Medicine; Murcia Spain
| | - J.R. Gimeno
- Inherited Cardiac Disease Unit; University Hospital Virgen Arrixaca; Murcia Spain
- Internal Medicine Department, University of Murcia; Murcia Spain
| |
Collapse
|
37
|
Kawai M, Johnston JR, Karam T, Wang L, Singh RK, Pinto JR. Myosin Rod Hypophosphorylation and CB Kinetics in Papillary Muscles from a TnC-A8V KI Mouse Model. Biophys J 2017; 112:1726-1736. [PMID: 28445763 DOI: 10.1016/j.bpj.2017.02.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 02/17/2017] [Accepted: 02/22/2017] [Indexed: 12/11/2022] Open
Abstract
The cardiac troponin C (TnC)-A8V mutation is associated with hypertrophic and restrictive cardiomyopathy (HCM and RCM) in human and mice. The residue affected lies in the N-helix, a region known to affect Ca2+-binding affinity to the N-terminal domain. Here we report on the functional effects of this mutation in skinned papillary muscle fibers from homozygous knock-in TnC-A8V mice. Muscle fibers from left ventricle were activated at 25°C under the ionic conditions of working cardiomyocytes. The pCa-tension relationship showed a 3× increase in Ca2+-sensitivity and a decrease (0.8×) in cooperativity (nH) in mutant fibers. The elementary steps of the cross-bridge (CB) cycle were investigated by sinusoidal analysis. The ATP study revealed that there is no significant change in the affinity of ATP (K1) for the myosin head. In TnC-A8V mutant fibers, the CB detachment rate (k2) and its equilibrium constant (K2) increased (1.5×). The phosphate study revealed that rate constant of the force-generation step (k4) decreased (0.5×), reversal step (k-4) increased (2×), and the phosphate-release step (1/K5) increased (2×). Pro-Q Diamond staining of the skinned fibers samples revealed no significant changes in total phosphorylation of multiple sarcomeric proteins. Further investigation using liquid chromatography-tandem mass spectrometry revealed hypophosphorylation of the rod domain of myosin heavy chain in TnC-A8V mutant fibers compared to wild-type. Immunoblotting confirmed the results observed in the mass spectrometry analysis. The results suggest perturbed CB kinetics-possibly caused by changes in the α-myosin heavy chain phosphorylation profile-as a novel mechanism, to our knowledge, by which a mutation in TnC can have rippling effects in the myofilament and contribute to the pathogenesis of HCM/RCM.
Collapse
Affiliation(s)
- Masataka Kawai
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa.
| | - Jamie R Johnston
- Department of Biomedical Sciences, College of Medicine, The Florida State University, Tallahassee, Florida
| | - Tarek Karam
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa
| | - Li Wang
- Departments of Anatomy and Cell Biology, and Internal Medicine, College of Medicine, University of Iowa, Iowa City, Iowa; School of Nursing, Soochow University, Suzhou, Jiangsu, China
| | - Rakesh K Singh
- Translational Science Laboratory, College of Medicine, The Florida State University, Tallahassee, Florida
| | - Jose R Pinto
- Department of Biomedical Sciences, College of Medicine, The Florida State University, Tallahassee, Florida
| |
Collapse
|
38
|
Ferrantini C, Coppini R, Pioner JM, Gentile F, Tosi B, Mazzoni L, Scellini B, Piroddi N, Laurino A, Santini L, Spinelli V, Sacconi L, De Tombe P, Moore R, Tardiff J, Mugelli A, Olivotto I, Cerbai E, Tesi C, Poggesi C. Pathogenesis of Hypertrophic Cardiomyopathy is Mutation Rather Than Disease Specific: A Comparison of the Cardiac Troponin T E163R and R92Q Mouse Models. J Am Heart Assoc 2017; 6:JAHA.116.005407. [PMID: 28735292 PMCID: PMC5586279 DOI: 10.1161/jaha.116.005407] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background In cardiomyocytes from patients with hypertrophic cardiomyopathy, mechanical dysfunction and arrhythmogenicity are caused by mutation‐driven changes in myofilament function combined with excitation‐contraction (E‐C) coupling abnormalities related to adverse remodeling. Whether myofilament or E‐C coupling alterations are more relevant in disease development is unknown. Here, we aim to investigate whether the relative roles of myofilament dysfunction and E‐C coupling remodeling in determining the hypertrophic cardiomyopathy phenotype are mutation specific. Methods and Results Two hypertrophic cardiomyopathy mouse models carrying the R92Q and the E163R TNNT2 mutations were investigated. Echocardiography showed left ventricular hypertrophy, enhanced contractility, and diastolic dysfunction in both models; however, these phenotypes were more pronounced in the R92Q mice. Both E163R and R92Q trabeculae showed prolonged twitch relaxation and increased occurrence of premature beats. In E163R ventricular myofibrils or skinned trabeculae, relaxation following Ca2+ removal was prolonged; resting tension and resting ATPase were higher; and isometric ATPase at maximal Ca2+ activation, the energy cost of tension generation, and myofilament Ca2+ sensitivity were increased compared with that in wild‐type mice. No sarcomeric changes were observed in R92Q versus wild‐type mice, except for a large increase in myofilament Ca2+ sensitivity. In R92Q myocardium, we found a blunted response to inotropic interventions, slower decay of Ca2+ transients, reduced SERCA function, and increased Ca2+/calmodulin kinase II activity. Contrarily, secondary alterations of E‐C coupling and signaling were minimal in E163R myocardium. Conclusions In E163R models, mutation‐driven myofilament abnormalities directly cause myocardial dysfunction. In R92Q, diastolic dysfunction and arrhythmogenicity are mediated by profound cardiomyocyte signaling and E‐C coupling changes. Similar hypertrophic cardiomyopathy phenotypes can be generated through different pathways, implying different strategies for a precision medicine approach to treatment.
Collapse
MESH Headings
- Animals
- Calcium Signaling
- Calcium-Calmodulin-Dependent Protein Kinases/metabolism
- Cardiomyopathy, Hypertrophic/diagnostic imaging
- Cardiomyopathy, Hypertrophic/genetics
- Cardiomyopathy, Hypertrophic/metabolism
- Cardiomyopathy, Hypertrophic/physiopathology
- Disease Models, Animal
- Excitation Contraction Coupling
- Fibrosis
- Genetic Markers
- Genetic Predisposition to Disease
- Hypertrophy, Left Ventricular/diagnostic imaging
- Hypertrophy, Left Ventricular/genetics
- Hypertrophy, Left Ventricular/metabolism
- Hypertrophy, Left Ventricular/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Mutation
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myofibrils/metabolism
- Myofibrils/pathology
- Phenotype
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
- Troponin T/genetics
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/genetics
- Ventricular Dysfunction, Left/metabolism
- Ventricular Dysfunction, Left/physiopathology
- Ventricular Function, Left
- Ventricular Remodeling
Collapse
Affiliation(s)
- Cecilia Ferrantini
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | - Josè Manuel Pioner
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Francesca Gentile
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Benedetta Tosi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Luca Mazzoni
- Department of NeuroFarBa, University of Florence, Italy
| | - Beatrice Scellini
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Nicoletta Piroddi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | | | | | - Leonardo Sacconi
- LENS, University of Florence & National Institute of Optics (INO-CNR), Florence, Italy
| | - Pieter De Tombe
- Loyola University Medical Center Department of Physiology, Chicago, IL
| | | | | | - Alessandro Mugelli
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | | | | | - Chiara Tesi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Corrado Poggesi
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| |
Collapse
|
39
|
Zile MA, Trayanova NA. Myofilament protein dynamics modulate EAD formation in human hypertrophic cardiomyopathy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2017. [PMID: 28648627 DOI: 10.1016/j.pbiomolbio.2017.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Patients with hypertrophic cardiomyopathy (HCM), a disease associated with sarcomeric protein mutations, often suffer from sudden cardiac death (SCD) resulting from arrhythmia. In order to advance SCD prevention strategies, our understanding of how sarcomeric mutations in HCM patients contribute to enhanced arrhythmogenesis needs to be improved. Early afterdepolarizations (EADs) are an important mechanism underlying arrhythmias associated with HCM-SCD. Although the ionic mechanisms underlying EADs have been studied in general, whether myofilament protein dynamics mechanisms also underlie EADs remains unknown. Thus, our goals were to investigate if myofilament protein dynamics mechanisms underlie EADs and to uncover how those mechanisms are affected by pacing rate, sarcomere length (SL), and different levels of HCM-induced myofilament remodeling. To achieve this, a mechanistically-based bidirectionally coupled human electrophysiology-force myocyte model under the conditions of HCM was constructed. HCM ionic remodeling included a reduced repolarization reserve, while HCM myofilament modeling involved altered thin filament activation. We found that the mechanoelectric feedback (MEF) on calcium dynamics in the bidirectionally coupled model, via Troponin C buffering of cytoplasmic Ca2+, was the myofilament mechanism underlying EADs. Incorporating MEF diminished the degree of repolarization reserve reduction necessary for EADs to emerge and increased the frequency of EAD occurrence, especially at faster pacing rates. Longer SLs and enhanced thin filament activation diminished the effects of MEF on EADs. Together these findings demonstrate that myofilament protein dynamics mechanisms play an important role in EAD formation.
Collapse
Affiliation(s)
- Melanie A Zile
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, 3400 N Charles St, 208 Hackerman Hall, Baltimore, MD 21218, USA.
| | - Natalia A Trayanova
- Institute for Computational Medicine and Department of Biomedical Engineering at Johns Hopkins University, 3400 N Charles St, 208 Hackerman Hall, Baltimore, MD 21218, USA.
| |
Collapse
|
40
|
Michels M, Olivotto I, Asselbergs FW, van der Velden J. Life-long tailoring of management for patients with hypertrophic cardiomyopathy : Awareness and decision-making in changing scenarios. Neth Heart J 2017; 25:186-199. [PMID: 28005231 PMCID: PMC5313451 DOI: 10.1007/s12471-016-0943-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common genetic heart disease, characterised by complex pathophysiology and extensive genetic and clinical heterogeneity. In most patients, HCM is caused by mutations in cardiac sarcomere protein genes and inherited as an autosomal dominant trait. The clinical phenotype ranges from severe presentations at a young age to lack of left ventricular hypertrophy in genotype-positive individuals. No preventative treatment is available as the sequence and causality of the pathomechanisms that initiate and exacerbate HCM are unknown. Sudden cardiac death and end-stage heart failure are devastating expressions of this disease. Contemporary management including surgical myectomy and implantable cardiac defibrillators has shown significant impact on long-term prognosis. However, timely recognition of specific scenarios - including transition to the end-stage phase - may be challenging due to limited awareness of the progression patterns of HCM. This in turn may lead to missed therapeutic opportunities. To illustrate these difficulties, we describe two HCM patients who progressed from the typical hyperdynamic stage of asymmetric septal thickening to end-stage heart failure with severely reduced ejection fraction. We highlight the different stages of this complex inherited cardiomyopathy based on the clinical staging proposed by Olivotto and colleagues. In this way, we aim to provide a practical guide for clinicians and hope to increase awareness for this common form of cardiac disease.
Collapse
Affiliation(s)
- M Michels
- Thoraxcenter, Department of Cardiology, Erasmus MC, Rotterdam, The Netherlands
| | - I Olivotto
- Careggi University Hospital, Florence, Italy
| | - F W Asselbergs
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - J van der Velden
- VU University Medical Center, Amsterdam, The Netherlands.
- Netherlands Heart Institute, Utrecht, The Netherlands.
| |
Collapse
|
41
|
Gal D, Sipido KR, Vandevelde W. 'A picture is worth a thousand words': image highlights from Cardiovascular Research. Cardiovasc Res 2016; 112:622-625. [PMID: 27979810 DOI: 10.1093/cvr/cvw226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Diane Gal
- Department of Cardiovascular Sciences, Experimental Cardiology, KU Leuven, University of Leuven, Campus Gasthuisberg O/N1 704, Herestraat 49, B-3000 Leuven, Belgium
| | - Karin R Sipido
- Department of Cardiovascular Sciences, Experimental Cardiology, KU Leuven, University of Leuven, Campus Gasthuisberg O/N1 704, Herestraat 49, B-3000 Leuven, Belgium
| | - Wouter Vandevelde
- Department of Cardiovascular Sciences, Experimental Cardiology, KU Leuven, University of Leuven, Campus Gasthuisberg O/N1 704, Herestraat 49, B-3000 Leuven, Belgium
| |
Collapse
|
42
|
Bottillo I, Giordano C, Cerbelli B, D'Angelantonio D, Lipari M, Polidori T, Majore S, Bertini E, D'Amico A, Giannarelli D, De Bernardo C, Masuelli L, Musumeci F, Avella A, Re F, Zachara E, d'Amati G, Grammatico P. A novel LAMP2 mutation associated with severe cardiac hypertrophy and microvascular remodeling in a female with Danon disease: a case report and literature review. Cardiovasc Pathol 2016; 25:423-31. [PMID: 27497751 DOI: 10.1016/j.carpath.2016.07.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 07/19/2016] [Accepted: 07/20/2016] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Danon disease (DD) is a rare disorder characterized by cardiomyopathy, intellectual disability, and proximal myopathy. It is caused by mutations in the LAMP2 gene on X chromosome. Female patients most often present with late-onset cardiomyopathy and slow disease progression, but early-onset cases with unfavorable prognosis have been reported. CASE REPORT We describe the clinical, pathological, and molecular features of a novel LAMP2 c.453delT mutation in a female patient with severe hypertrophic cardiomyopathy, Wolff Parkinson White (WPW) syndrome and rapid progression to heart failure, requiring heart transplant. Immunohistochemical analysis of LAMP2 in the explanted heart revealed a mosaic pattern of distribution, with discrete clusters of either stained or unstained cardiac myocytes, the latter being more frequent in the septum. These findings paralleled X chromosome inactivation within the myocardium. Interestingly, multiple foci of microscarring were found on histology in the Left Ventricle (LV) free wall and septum, in a close spatial relationship with remodeling and severe stenosis of intramural coronary arterioles. CONCLUSIONS Our findings suggest that several features may contribute to the early and severe cardiac phenotype in female DD patients. The type of mutation may account for the early disease onset, while both the inhomogeneous distribution of LAMP2 loss and the presence of microvascular remodeling may be determinant in the rapid progression to heart failure.
Collapse
Affiliation(s)
- Irene Bottillo
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy.
| | - Carla Giordano
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Bruna Cerbelli
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Daniela D'Angelantonio
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Martina Lipari
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Taisia Polidori
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Silvia Majore
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Enrico Bertini
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Adele D'Amico
- Unit for Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Diana Giannarelli
- Biostatistic Unit, Regina Elena National Cancer Institute, Rome, Italy
| | - Carmelilia De Bernardo
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| | - Laura Masuelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Francesco Musumeci
- Department of Cardiac Surgery, San Camillo-Forlanini Hospital, Rome, Italy
| | - Andrea Avella
- Cardiomyopathies Unit, Division of Cardiology and Cardiac Arrhythmias, San Camillo-Forlanini Hospital, Rome, Italy
| | - Federica Re
- Cardiomyopathies Unit, Division of Cardiology and Cardiac Arrhythmias, San Camillo-Forlanini Hospital, Rome, Italy
| | - Elisabetta Zachara
- Cardiomyopathies Unit, Division of Cardiology and Cardiac Arrhythmias, San Camillo-Forlanini Hospital, Rome, Italy
| | - Giulia d'Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | - Paola Grammatico
- Medical Genetics Laboratory, Department of Molecular Medicine, Sapienza University, San Camillo-Forlanini Hospital, Rome, Italy
| |
Collapse
|
43
|
Matsa E, Ahrens JH, Wu JC. Human Induced Pluripotent Stem Cells as a Platform for Personalized and Precision Cardiovascular Medicine. Physiol Rev 2016; 96:1093-126. [PMID: 27335446 PMCID: PMC6345246 DOI: 10.1152/physrev.00036.2015] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have revolutionized the field of human disease modeling, with an enormous potential to serve as paradigm shifting platforms for preclinical trials, personalized clinical diagnosis, and drug treatment. In this review, we describe how hiPSCs could transition cardiac healthcare away from simple disease diagnosis to prediction and prevention, bridging the gap between basic and clinical research to bring the best science to every patient.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - John H Ahrens
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Department of Medicine, Division of Cardiology, and Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
44
|
Cardiovascular magnetic resonance in rheumatology: Current status and recommendations for use. Int J Cardiol 2016; 217:135-48. [PMID: 27179903 DOI: 10.1016/j.ijcard.2016.04.158] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/25/2016] [Indexed: 01/14/2023]
Abstract
Targeted therapies in connective tissue diseases (CTDs) have led to improvements of disease-associated outcomes, but life expectancy remains lower compared to general population due to emerging co-morbidities, particularly due to excess cardiovascular risk. Cardiovascular magnetic resonance (CMR) is a noninvasive imaging technique which can provide detailed information about multiple cardiovascular pathologies without using ionizing radiation. CMR is considered the reference standard for quantitative evaluation of left and right ventricular volumes, mass and function, cardiac tissue characterization and assessment of thoracic vessels; it may also be used for the quantitative assessment of myocardial blood flow with high spatial resolution and for the evaluation of the proximal coronary arteries. These applications are of particular interest in CTDs, because of the potential of serious and variable involvement of the cardiovascular system during their course. The International Consensus Group on CMR in Rheumatology was formed in January 2012 aiming to achieve consensus among CMR and rheumatology experts in developing initial recommendations on the current state-of-the-art use of CMR in CTDs. The present report outlines the recommendations of the participating CMR and rheumatology experts with regards to: (a) indications for use of CMR in rheumatoid arthritis, the spondyloarthropathies, systemic lupus erythematosus, vasculitis of small, medium and large vessels, myositis, sarcoidosis (SRC), and scleroderma (SSc); (b) CMR protocols, terminology for reporting CMR and diagnostic CMR criteria for assessment and quantification of cardiovascular involvement in CTDs; and (c) a research agenda for the further development of this evolving field.
Collapse
|
45
|
San Román I, Navarro M, Martínez F, Albert L, Polo L, Guardiola J, García-Molina E, Muñoz-Esparza C, López-Ayala JM, Sabater-Molina M, Gimeno JR. Unclassifiable arrhythmic cardiomyopathy associated with Emery-Dreifuss caused by a mutation in FHL1. Clin Genet 2016; 90:171-6. [DOI: 10.1111/cge.12760] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 01/14/2023]
Affiliation(s)
- I. San Román
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - M. Navarro
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - F. Martínez
- Neurology Department; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - L. Albert
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - L. Polo
- Pathology Department; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - J. Guardiola
- Pneumology Department; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - E. García-Molina
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - C. Muñoz-Esparza
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - J. M. López-Ayala
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
| | - M. Sabater-Molina
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
- Internal Medicine Department, University of Murcia; Murcia Spain
| | - J. R. Gimeno
- Inherited Cardiac Disease Unit; University Hospital Virgen de la Arrixaca; Murcia Spain
- Internal Medicine Department, University of Murcia; Murcia Spain
| |
Collapse
|
46
|
Comprehensive maternal serum proteomics identifies the cytoskeletal proteins as non-invasive biomarkers in prenatal diagnosis of congenital heart defects. Sci Rep 2016; 6:19248. [PMID: 26750556 PMCID: PMC4707500 DOI: 10.1038/srep19248] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/09/2015] [Indexed: 12/27/2022] Open
Abstract
Congenital heart defects (CHDs) are the most common group of major birth defects. Presently there are no clinically used biomarkers for prenatally detecting CHDs. Here, we performed a comprehensive maternal serum proteomics assessment, combined with immunoassays, for the discovery of non-invasive biomarkers for prenatal diagnosis of CHDs. A total of 370 women were included in this study. An isobaric tagging for relative and absolute quantification (iTRAQ) proteomic approach was used first to compare protein profiles in pooled serum collected from women who had CHD-possessing or normal fetuses, and 47 proteins displayed significant differential expressions. Targeted verifications were performed on 11 proteins using multiple reaction monitoring mass spectrometry (MRM-MS), and the resultant candidate biomarkers were then further validated using ELISA analysis. Finally, we identified a biomarker panel composed of 4 cytoskeletal proteins capable of differentiating CHD-pregnancies from normal ones [with an area under the receiver operating characteristic curve (AUC) of 0.938, P < 0.0001]. The discovery of cytoskeletal protein changes in maternal serum not only could help us in prenatal diagnosis of CHDs, but also may shed new light on CHD embryogenesis studies.
Collapse
|
47
|
Maron BJ, Fox PR. Hypertrophic cardiomyopathy in man and cats. J Vet Cardiol 2015; 17 Suppl 1:S6-9. [DOI: 10.1016/j.jvc.2015.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 03/15/2015] [Accepted: 03/17/2015] [Indexed: 12/19/2022]
|
48
|
Pasqualucci D, Fornaro A, Castelli G, Rossi A, Arretini A, Chiriatti C, Targetti M, Girolami F, Corda M, Orrù P, Matta G, Stefàno P, Cecchi F, Porcu M, Olivotto I. Clinical Spectrum, Therapeutic Options, and Outcome of Advanced Heart Failure in Hypertrophic Cardiomyopathy. Circ Heart Fail 2015; 8:1014-21. [PMID: 26446673 DOI: 10.1161/circheartfailure.114.001843] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 09/17/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND The clinical course of patients with hypertrophic cardiomyopathy and advanced heart failure (HF) subtended by progressive left ventricular dysfunction has received limited attention. Our aim was to assess the outcome of HF and impact of treatment options including the implantable cardioverter-defibrillator and heart transplantation (HT) in patients with hypertrophic cardiomyopathy evaluated at 2 Italian referral centers >3 decades. METHODS AND RESULTS All-cause mortality and a combined end point including death, HT, or appropriate implantable cardioverter-defibrillator shock were assessed in 71 consecutive patients with HF not related to outflow obstruction (7% of the entire hypertrophic cardiomyopathy cohort) followed up for 6.1±6.9 years after development of New York Heart Association class III to IV symptoms. At enrollment, left ventricular ejection fraction was <50% in 55 patients and >50% in 16; all had restrictive left ventricular filling. During follow-up, 35 patients died (49%%; 5-year rate, 49%) and 53 met the combined end point (75%; 5-year rate, 62%). Most events occurred in the 3 years after HF onset (17% per year compared with only 3% per year subsequently). Appropriate implantable cardioverter-defibrillator shocks occurred in 11 of 34 implanted patients. Of 37 patients evaluated for HT, 14 were transplanted, 10 listed, and 13 excluded; 2 early post-HT deaths occurred in patients with elevated pulmonary vascular resistance. Eleven of the 14 HT patients were alive at 10±8 years. CONCLUSIONS In hypertrophic cardiomyopathy, advanced HF not associated with outflow obstruction portends a severely unfavorable prognosis, particularly in the first 3 years after onset of symptoms, despite frequently preserved systolic function in about one quarter of the patients. Outcome of HT is favorable but requires early consideration, as the window of opportunity may be short.
Collapse
Affiliation(s)
- Daniele Pasqualucci
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.).
| | - Alessandra Fornaro
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Gabriele Castelli
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Alessandra Rossi
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Anna Arretini
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Chiara Chiriatti
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Mattia Targetti
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Francesca Girolami
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Marco Corda
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Pierpaolo Orrù
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Gildo Matta
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Pierluigi Stefàno
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Franco Cecchi
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Maurizio Porcu
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| | - Iacopo Olivotto
- From the Departments of Cardiology (D.P., M.C., P.O., M.P.) and Radiology (G.M.), Brotzu Hospital, Cagliari, Italy; Referral Center for Cardiomyopathies (A.F., G.C., A.R., A.A., C.C., M.T., I.O.), Genetic Diagnostics Unit (F.G.), and Cardiac Surgery (P.S.), Careggi University Hospital, Florence, Italy; and Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy (F.C.)
| |
Collapse
|
49
|
Imaging in Deciphering Histological Substrates in Hypertrophic Cardiomyopathy. CURRENT CARDIOVASCULAR IMAGING REPORTS 2015. [DOI: 10.1007/s12410-015-9355-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
50
|
Cardiac myosin-binding protein C (MYBPC3) in cardiac pathophysiology. Gene 2015; 573:188-97. [PMID: 26358504 DOI: 10.1016/j.gene.2015.09.008] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 08/21/2015] [Accepted: 09/01/2015] [Indexed: 12/27/2022]
Abstract
More than 350 individual MYPBC3 mutations have been identified in patients with inherited hypertrophic cardiomyopathy (HCM), thus representing 40–50% of all HCM mutations, making it the most frequently mutated gene in HCM. HCM is considered a disease of the sarcomere and is characterized by left ventricular hypertrophy, myocyte disarray and diastolic dysfunction. MYBPC3 encodes for the thick filament associated protein cardiac myosin-binding protein C (cMyBP-C), a signaling node in cardiac myocytes that contributes to the maintenance of sarcomeric structure and regulation of contraction and relaxation. This review aims to provide a succinct overview of how mutations in MYBPC3 are considered to affect the physiological function of cMyBP-C, thus causing the deleterious consequences observed inHCM patients. Importantly, recent advances to causally treat HCM by repairing MYBPC3 mutations by gene therapy are discussed here, providing a promising alternative to heart transplantation for patients with a fatal form of neonatal cardiomyopathy due to bi-allelic truncating MYBPC3 mutations.
Collapse
|