1
|
Gerritse M, van Ham WB, Denning C, van Veen TAB, Maas RGC. Characteristics and pharmacological responsiveness in hiPSC models of inherited cardiomyopathy. Pharmacol Ther 2025; 272:108845. [PMID: 40250811 DOI: 10.1016/j.pharmthera.2025.108845] [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: 11/11/2024] [Revised: 02/17/2025] [Accepted: 03/24/2025] [Indexed: 04/20/2025]
Abstract
Inherited cardiomyopathies are a major cause of heart failure in all age groups, often with an onset in adolescence or early adult life. More than a thousand variants in approximately one hundred genes are associated with cardiomyopathies. Interestingly, many genetic cardiomyopathies display overlapping phenotypical defects in patients, despite the diversity of the initial pathogenic variants. Understanding how the underlying pathophysiology of genetic cardiomyopathies leads to these phenotypes will improve insights into a patient's disease course, and creates the opportunity for conceiving treatment strategies. Moreover, therapeutic strategies can be used to treat multiple cardiomyopathies based on shared phenotypes. Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) offer reliable, high-throughput models for studying molecular and cellular characteristics of hereditary cardiomyopathies. hiPSC-CMs are produced relatively easily, either by directly originating them from patients, or by introducing patient-specific genetic variants in healthy lines. This review evaluates 90 studies on 24 cardiomyopathy-associated genes and systematically summarises the morphological and functional phenotypes observed in hiPSC-CMs. Additionally, treatment strategies applied in cardiomyopathic hiPSC-CMs are compiled and scored for effectiveness. Multiple overlapping phenotypic defects were identified in cardiomyocytes with different variants, whereas certain characteristics were only associated with specific genetic variants. Based on these findings, common mechanisms, therapeutic prospects, and considerations for future research are discussed with the aim to improve clinical translation from hiPSC-CMs to patients.
Collapse
Affiliation(s)
- Merel Gerritse
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Willem B van Ham
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Chris Denning
- Department of Stem Cell Biology, Biodiscovery Institute, University of Nottingham, University Park, Nottingham, NG7 2RD, UK.
| | - Toon A B van Veen
- Department of Medical Physiology, Division Heart & Lungs, University Medical Center Utrecht, 3584 CM Utrecht, the Netherlands.
| | - Renee G C Maas
- Utrecht Regenerative Medicine Center, Circulatory Health Research Center, University Utrecht, 3584 CS Utrecht, the Netherlands; Department of Cardiology, Experimental Cardiology Laboratory, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
2
|
Mukhopadhyay S, Dixit P, Khanom N, Sanghera G, McGurk KA. The Genetic Factors Influencing Cardiomyopathies and Heart Failure across the Allele Frequency Spectrum. J Cardiovasc Transl Res 2024; 17:1119-1139. [PMID: 38771459 PMCID: PMC11519107 DOI: 10.1007/s12265-024-10520-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/03/2024] [Indexed: 05/22/2024]
Abstract
Heart failure (HF) remains a major cause of mortality and morbidity worldwide. Understanding the genetic basis of HF allows for the development of disease-modifying therapies, more appropriate risk stratification, and personalised management of patients. The advent of next-generation sequencing has enabled genome-wide association studies; moving beyond rare variants identified in a Mendelian fashion and detecting common DNA variants associated with disease. We summarise the latest GWAS and rare variant data on mixed and refined HF aetiologies, and cardiomyopathies. We describe the recent understanding of the functional impact of titin variants and highlight FHOD3 as a novel cardiomyopathy-associated gene. We describe future directions of research in this field and how genetic data can be leveraged to improve the care of patients with HF.
Collapse
Affiliation(s)
- Srinjay Mukhopadhyay
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
- School of Medicine, Cardiff University, Wales, UK
| | - Prithvi Dixit
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Najiyah Khanom
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Gianluca Sanghera
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK
| | - Kathryn A McGurk
- National Heart and Lung Institute, Imperial College London, LMS Building, Hammersmith Campus, London, UK.
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK.
| |
Collapse
|
3
|
Tseng WC, Chiu SN, Juang JMJ, Chen WP, Lee NC, Wu MH. Genophenotypic correlates and long-term outcome prognosticators of left ventricular non-compaction in children. J Formos Med Assoc 2024:S0929-6646(24)00452-2. [PMID: 39332976 DOI: 10.1016/j.jfma.2024.09.032] [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: 06/21/2024] [Revised: 08/05/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024] Open
Abstract
BACKGROUND To investigate the outcomes, clinical prognosticators, and genetic profiles of pediatric left ventricular non-compaction (LVNC). METHODS All subjects were <18 years old, diagnosed with LVNC between January 2008 and December 2020. Whole-exome sequencing was undertaken. The primary endpoint was composite outcome, including death, heart transplant, and left ventricular assist device implantation. RESULTS Thirty-three patients were enrolled, males predominating (57.6%). Median age at diagnosis was 0.33 (0.1-7.2) years. Family history was documented in four (12.1%). Five (15.2%) had sustained arrhythmias. Mean follow-up period was 9.5 years, and 5- and 10-year event-free survival were 84.8% and 66.9%, respectively. Seven died of heart failure, four received heart transplants, and one required left ventricular assist device placement. Log of baseline NT-proBNP (adjusted odds ratio [aOR] = 4.4, p = 0.012) and lack of improvement in NT-proBNP (aOR = 41.2, p = 0.033) impacted the primary outcome most significantly. Eighteen out of 25 genetic testing (72%) revealed chromosomal anomalies, or pathogenic or likely pathogenic variants. Three genetic variants (PLEKHM2 p.G419R, RYR2 p.V2571A, and SCN5A p.M1676I) were significantly associated with the primary outcome (p = 1.52 × 10-6). CONCLUSIONS Pediatric LVNC is a rare disorder with variable genetic underpinnings. Baseline NT-proBNP values and lack of improvement in NT-proBNP levels were important predictors of poor long-term outcomes. Pathogenic genetic variants or chromosomal anomalies are not unusual.
Collapse
Affiliation(s)
- Wei-Chieh Tseng
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shuenn-Nan Chiu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Jyh-Ming Jimmy Juang
- Department of Heart failure Center and Cardiovascular Center, National Taiwan University Hospital, Taipei, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Pin Chen
- Graduate Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ni-Chung Lee
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Hwan Wu
- Department of Pediatrics, National Taiwan University Hospital, Taipei, Taiwan; Department of Pediatrics, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
4
|
Dababneh S, Hamledari H, Maaref Y, Jayousi F, Hosseini DB, Khan A, Jannati S, Jabbari K, Arslanova A, Butt M, Roston TM, Sanatani S, Tibbits GF. Advances in Hypertrophic Cardiomyopathy Disease Modelling Using hiPSC-Derived Cardiomyocytes. Can J Cardiol 2024; 40:766-776. [PMID: 37952715 DOI: 10.1016/j.cjca.2023.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/21/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023] Open
Abstract
The advent of human induced pluripotent stem cells (hiPSCs) and their capacity to be differentiated into beating human cardiomyocytes (CMs) in vitro has revolutionized human disease modelling, genotype-phenotype predictions, and therapeutic testing. Hypertrophic cardiomyopathy (HCM) is a common inherited cardiomyopathy and the leading known cause of sudden cardiac arrest in young adults and athletes. On a molecular level, HCM is often driven by single pathogenic genetic variants, usually in sarcomeric proteins, that can alter the mechanical, electrical, signalling, and transcriptional properties of the cell. A deeper knowledge of these alterations is critical to better understanding HCM manifestation, progression, and treatment. Leveraging hiPSC-CMs to investigate the molecular mechanisms driving HCM presents a unique opportunity to dissect the consequences of genetic variants in a sophisticated and controlled manner. In this review, we summarize the molecular underpinnings of HCM and the role of hiPSC-CM studies in advancing our understanding, and we highlight the advances in hiPSC-CM-based modelling of HCM, including maturation, contractility, multiomics, and genome editing, with the notable exception of electrophysiology, which has been previously covered.
Collapse
Affiliation(s)
- Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Homa Hamledari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Yasaman Maaref
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Farah Jayousi
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Dina B Hosseini
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aasim Khan
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Shayan Jannati
- Faculty of Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kosar Jabbari
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Thomas M Roston
- Division of Cardiology and Centre for Cardiovascular Innovation, University of British Columbia, Vancouver, British Columbia, Canada
| | - Shubhayan Sanatani
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Glen F Tibbits
- Cellular and Regenerative Medicine Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada.
| |
Collapse
|
5
|
Grebur K, Mester B, Fekete BA, Kiss AR, Gregor Z, Horváth M, Farkas-Sütő K, Csonka K, Bödör C, Merkely B, Vágó H, Szűcs A. Genetic, clinical and imaging implications of a noncompaction phenotype population with preserved ejection fraction. Front Cardiovasc Med 2024; 11:1337378. [PMID: 38380180 PMCID: PMC10876896 DOI: 10.3389/fcvm.2024.1337378] [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: 11/12/2023] [Accepted: 01/22/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction The genotype of symptomatic left ventricular noncompaction phenotype (LVNC) subjects with preserved left ventricular ejection fraction (LVEF) and its effect on clinical presentation are less well studied. We aimed to characterize the genetic, cardiac magnetic resonance (CMR) and clinical background, and genotype-phenotype relationship in LVNC with preserved LVEF. Methods We included 54 symptomatic LVNC individuals (LVEF: 65 ± 5%) whose samples were analyzed with a 174-gene next-generation sequencing panel and 54 control (C) subjects. The results were evaluated using the criteria of the American College of Medical Genetics and Genomics. Medical data suggesting a higher risk of cardiovascular complications were considered "red flags". Results Of the LVNC population, 24% carried pathogenic or likely pathogenic (P) mutations; 56% carried variants of uncertain significance (VUS); and 20% were free from cardiomyopathy-related mutations. Regarding the CMR parameters, the LVNC and C groups differed significantly, while the three genetic subgroups were comparable. We found a significant relationship between red flags and genotype; furthermore, the number of red flags in a single subject differed significantly among the genetic subgroups (p = 0.002) and correlated with the genotype (r = 0.457, p = 0.01). In 6 out of 7 LVNC subjects diagnosed in childhood, P or VUS mutations were found. Discussion The large number of P mutations and the association between red flags and genotype underline the importance of genetic-assisted risk stratification in symptomatic LVNC with preserved LVEF.
Collapse
Affiliation(s)
- Kinga Grebur
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Balázs Mester
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Bálint András Fekete
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Anna Réka Kiss
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Zsófia Gregor
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Márton Horváth
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Katalin Csonka
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Csaba Bödör
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Hajnalka Vágó
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Andrea Szűcs
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
6
|
Ni M, Li Y, Wei J, Song Z, Wang H, Yao J, Chen YX, Belke D, Estillore JP, Wang R, Vallmitjana A, Benitez R, Hove-Madsen L, Feng W, Chen J, Roston TM, Sanatani S, Lehman A, Chen SRW. Increased Ca 2+ Transient Underlies RyR2-Related Left Ventricular Noncompaction. Circ Res 2023; 133:177-192. [PMID: 37325910 DOI: 10.1161/circresaha.123.322504] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/07/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND A loss-of-function cardiac ryanodine receptor (RyR2) mutation, I4855M+/-, has recently been linked to a new cardiac disorder termed RyR2 Ca2+ release deficiency syndrome (CRDS) as well as left ventricular noncompaction (LVNC). The mechanism by which RyR2 loss-of-function causes CRDS has been extensively studied, but the mechanism underlying RyR2 loss-of-function-associated LVNC is unknown. Here, we determined the impact of a CRDS-LVNC-associated RyR2-I4855M+/- loss-of-function mutation on cardiac structure and function. METHODS We generated a mouse model expressing the CRDS-LVNC-associated RyR2-I4855M+/- mutation. Histological analysis, echocardiography, ECG recording, and intact heart Ca2+ imaging were performed to characterize the structural and functional consequences of the RyR2-I4855M+/- mutation. RESULTS As in humans, RyR2-I4855M+/- mice displayed LVNC characterized by cardiac hypertrabeculation and noncompaction. RyR2-I4855M+/- mice were highly susceptible to electrical stimulation-induced ventricular arrhythmias but protected from stress-induced ventricular arrhythmias. Unexpectedly, the RyR2-I4855M+/- mutation increased the peak Ca2+ transient but did not alter the L-type Ca2+ current, suggesting an increase in Ca2+-induced Ca2+ release gain. The RyR2-I4855M+/- mutation abolished sarcoplasmic reticulum store overload-induced Ca2+ release or Ca2+ leak, elevated sarcoplasmic reticulum Ca2+ load, prolonged Ca2+ transient decay, and elevated end-diastolic Ca2+ level upon rapid pacing. Immunoblotting revealed increased level of phosphorylated CaMKII (Ca2+-calmodulin dependent protein kinases II) but unchanged levels of CaMKII, calcineurin, and other Ca2+ handling proteins in the RyR2-I4855M+/- mutant compared with wild type. CONCLUSIONS The RyR2-I4855M+/- mutant mice represent the first RyR2-associated LVNC animal model that recapitulates the CRDS-LVNC overlapping phenotype in humans. The RyR2-I4855M+/- mutation increases the peak Ca2+ transient by increasing the Ca2+-induced Ca2+ release gain and the end-diastolic Ca2+ level by prolonging Ca2+ transient decay. Our data suggest that the increased peak-systolic and end-diastolic Ca2+ levels may underlie RyR2-associated LVNC.
Collapse
Affiliation(s)
- Mingke Ni
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yanhui Li
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinhong Wei
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
- School of Medicine, Northwest University, Xi 'an, China (J.W.)
| | - Zhenpeng Song
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Hui Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Jinjing Yao
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Yong-Xiang Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Darrell Belke
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - John Paul Estillore
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Ruiwu Wang
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| | - Alexander Vallmitjana
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
| | - Raul Benitez
- Department of Automatic Control, Universitat Politècnica de Catalunya, Barcelona, Spain (A.V., R.B.)
- Institut de Recerca Sant Joan de Déu (IRSJD), Barcelona, Spain (R.B.)
| | - Leif Hove-Madsen
- Biomedical Research Institute Barcelona IIBB-CSIC, IIB Sant Pau and CIBERCV, Hospital de Sant Pau, Barcelona, Spain (L.H.-M.)
| | - Wei Feng
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Ju Chen
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla (W.F., J.C.)
| | - Thomas M Roston
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Shubhayan Sanatani
- Division of Pediatric Cardiology, Department of Pediatrics (T.M.R., S.S.), University of British Columbia, Vancouver, Canada
| | - Anna Lehman
- Department of Medical Genetics (A.L.), University of British Columbia, Vancouver, Canada
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, Libin Cardiovascular Institute, University of Calgary, Alberta, Canada (M.N., Y.L., J.W., Z.S., H.W., J.Y., Y.-X.C., D.B., J.P.E., R.W., S.R.W.C.)
| |
Collapse
|
7
|
Lehman SJ, Meller A, Solieva SO, Lotthammer JM, Greenberg L, Langer SJ, Greenberg MJ, Tardiff JC, Bowman GR, Leinwand L. Divergent Molecular Phenotypes in Point Mutations at the Same Residue in Beta-Myosin Heavy Chain Lead to Distinct Cardiomyopathies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547580. [PMID: 37461648 PMCID: PMC10349964 DOI: 10.1101/2023.07.03.547580] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
In genetic cardiomyopathies, a frequently described phenomenon is how similar mutations in one protein can lead to discrete clinical phenotypes. One example is illustrated by two mutations in beta myosin heavy chain (β-MHC) that are linked to hypertrophic cardiomyopathy (HCM) (Ile467Val, I467V) and left ventricular non-compaction (LVNC) (Ile467Thr, I467T). To investigate how these missense mutations lead to independent diseases, we studied the molecular effects of each mutation using recombinant human β-MHC Subfragment 1 (S1) in in vitro assays. Both HCM-I467V and LVNC-I467T S1 mutations exhibited similar mechanochemical function, including unchanged ATPase and enhanced actin velocity but had opposing effects on the super-relaxed (SRX) state of myosin. HCM-I467V S1 showed a small reduction in the SRX state, shifting myosin to a more actin-available state that may lead to the "gain-of-function" phenotype commonly described in HCM. In contrast, LVNC-I467T significantly increased the population of myosin in the ultra-slow SRX state. Interestingly, molecular dynamics simulations reveal that I467T allosterically disrupts interactions between ADP and the nucleotide-binding pocket, which may result in an increased ADP release rate. This predicted change in ADP release rate may define the enhanced actin velocity measured in LVNC-I467T, but also describe the uncoupled mechanochemical function for this mutation where the enhanced ADP release rate may be sufficient to offset the increased SRX population of myosin. These contrasting molecular effects may lead to contractile dysregulation that initiates LVNC-associated signaling pathways that progress the phenotype. Together, analysis of these mutations provides evidence that phenotypic complexity originates at the molecular level and is critical to understanding disease progression and developing therapies.
Collapse
Affiliation(s)
- Sarah J Lehman
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Artur Meller
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
- Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO, USA
| | - Shahlo O Solieva
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Jeffrey M Lotthammer
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Lina Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Stephen J Langer
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| | - Michael J Greenberg
- Washington University in St. Louis, Department of Biochemistry and Molecular Biophysics, St. Louis, MO, USA
| | - Jil C Tardiff
- University of Arizona, Department of Biomedical Engineering, Tucson, AZ, USA
| | - Gregory R Bowman
- University of Pennsylvania, Department of Biochemistry and Biophysics, Philadelphia, PA, USA
| | - Leslie Leinwand
- University of Colorado, Molecular, Cellular, and Developmental Biology, Boulder, CO, USA
| |
Collapse
|
8
|
Different Phenotypes of Sarcomeric MyBPC3-Cardiomyopathy in the Same Family: Hypertrophic, Left Ventricular Noncompaction and Restrictive Phenotypes (in Association with Sarcoidosis). Genes (Basel) 2022; 13:genes13081344. [PMID: 36011256 PMCID: PMC9407345 DOI: 10.3390/genes13081344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/04/2022] [Accepted: 07/26/2022] [Indexed: 01/27/2023] Open
Abstract
The same variants in sarcomeric genes can lead to different cardiomyopathies within the same family. This gave rise to the concept of a continuum of sarcomeric cardiomyopathies. However, the manifestations and evolution of these cardiomyopathies in pathogenic variant carriers, including members of the same family, remains poorly understood. We present a case of familial sarcomeric cardiomyopathy caused by heterozygous truncating pathogenic variant p.Q1233* in cardiac myosin-binding protein C (MyBPC3) gene. The proband was first diagnosed with restrictive cardiomyopathy combined with left ventricular noncompaction (LVNC) and sarcoidosis at the age of 64. The predominantly restrictive phenotype of cardiomyopathy is considered to be a result of interaction between LVNC and sarcoid myocarditis. His 39-year-old son and 35-year-old daughter have identical non-obstructive asymmetric hypertrophic cardiomyopathy. The risk of sudden cardiac death in the son is high due to myocardial fibrosis, ischemia and nonsustained VT. We assume that both phenotypes in the family may have originally been different or there may have been a gradual transformation of the hypertrophic phenotype into LVNC. Myocarditis is regarded as an important epigenomic modifier of sarcomeric cardiomyopathy. In the proband and his son, cardioverter-defibrillators were implanted, and the proband experienced appropriate shocks due to ventricular tachycardia/fibrillation. The proband was also treated with corticosteroids. His death at the age of 69 years occurred due to acute gastric hemorrhage accompanied by progressive heart failure. This report confirms the concept of the phenotypic continuum of sarcomeric cardiomyopathies and describes possible phenotypic patterns and their transformation over time.
Collapse
|
9
|
Genetic Profile of Left Ventricular Noncompaction Cardiomyopathy in Children-A Single Reference Center Experience. Genes (Basel) 2022; 13:genes13081334. [PMID: 35893073 PMCID: PMC9332142 DOI: 10.3390/genes13081334] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/14/2022] [Accepted: 07/24/2022] [Indexed: 02/06/2023] Open
Abstract
Background: Left ventricular noncompaction cardiomyopathy (LVNC) is a rare cardiac disorder characterised by the presence of a two-layer myocardium with prominent ventricular trabeculation, intertrabecular deep depressions and an increased risk of heart failure, atrial and ventricular arrhythmias and systemic thromboembolic events in affected patients. The heterogeneous molecular aetiology solved in 10%–50% of patients more frequently involves sarcomeric, cytoskeletal or ion channel protein dysfunction—mainly related to causative MYH7, TTN or MYBPC3 variants. The aim of the study was to determine the molecular spectrum of isolated LVNC in a group of children examined in a single paediatric reference centre. Methods: Thirty-one paediatric patients prospectively diagnosed with LVNC by echocardiography and cardiovascular magnetic resonance examination were recruited into the study group. The molecular analysis included next-generation sequencing (gene panel or whole exome) and classic Sanger sequencing. All selected variants with high priority were co-segregated in the available parents. Results: We identified 16 distinct variants in 11 genes in 16 patients (52%), including 10 novel alterations. The most frequent defects in our cohort were found in the genes HCN4 (n = 4), MYH7 (n = 2) and PRDM16 (n = 2). Other likely disease-causing variants were detected in ACTC1, ACTN2, HCCS, LAMA4, MYH6, RBM20, TAFFAZIN and TTN. Patients with established molecular defects more often presented with arrhythmia, thromboembolic events and death, whereas the predominant symptoms in patients with no identified molecular defects were heart failure and the presence of late gadolinium enhancement. Conclusion: This study expands the genetic and clinical spectrum of childhood LVNC. Although the molecular aetiology of LVNC varies widely, the comprehensive testing of a wide panel of cardiomyopathy-related genes helped to identify underlying molecular defects in more than half of the children in the study group. The molecular spectrum in our cohort correlated with the occurrence of arrhythmia, death and a family history of cardiomyopathy. We confirmed that genetic testing is an integral part of the work-up and management LVNC in children.
Collapse
|
10
|
Abstract
Purpose of Review The advent of induced pluripotent stem cells (iPSC) has paved the way for new in vitro models of human cardiomyopathy. Herein, we will review existing models of disease as well as strengths and limitations of the system. Recent Findings Preclinical studies have now demonstrated that iPSCs generated from patients with both acquired or heritable genetic diseases retain properties of the disease in vitro and can be used as a model to study novel therapeutics. iPSCs can be differentiated in vitro into the cardiomyocyte lineage into cells resembling adult ventricular myocytes that retain properties of cardiovascular disease from their respective donor. iPSC pluripotency allows for them to be frozen, stored, and continually used to generate iPSC-derived myocytes for future experiments without need for invasive procedures or repeat myocyte isolations to obtain animal or human cardiac tissues. Summary While not without their limitations, iPSC models offer new ways for studying patient-specific cardiomyopathies. iPSCs offer a high-throughput avenue for drug development, modeling of disease pathophysiology in vitro, and enabling experimental repair strategies without need for invasive procedures to obtain cardiac tissues.
Collapse
|
11
|
Lin Y, Huang J, Zhu Z, Zhang Z, Xian J, Yang Z, Qin T, Chen L, Huang J, Huang Y, Wu Q, Hu Z, Lin X, Xu G. Overlap phenotypes of the left ventricular noncompaction and hypertrophic cardiomyopathy with complex arrhythmias and heart failure induced by the novel truncated DSC2 mutation. Orphanet J Rare Dis 2021; 16:496. [PMID: 34819141 PMCID: PMC8611834 DOI: 10.1186/s13023-021-02112-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/06/2021] [Indexed: 12/18/2022] Open
Abstract
Background The left ventricular noncompaction cardiomyopathy (LVNC) is a rare subtype of cardiomyopathy associated with a high risk of heart failure (HF), thromboembolism, arrhythmia, and sudden cardiac death. Methods The proband with overlap phenotypes of LVNC and hypertrophic cardiomyopathy (HCM) complicates atrial fibrillation (AF), ventricular tachycardia (VT), and HF due to the diffuse myocardial lesion, which were diagnosed by electrocardiogram, echocardiogram and cardiac magnetic resonance imaging. Peripheral blood was collected from the proband and his relatives. DNA was extracted from the peripheral blood of proband for high-throughput target capture sequencing. The Sanger sequence verified the variants. The protein was extracted from the skin of the proband and healthy volunteer. The expression difference of desmocollin2 was detected by Western blot. Results The novel heterozygous truncated mutation (p.K47Rfs*2) of the DSC2 gene encoding an important component of desmosomes was detected by targeted capture sequencing. The western blots showed that the expressing level of functional desmocollin2 protein (~ 94kd) was lower in the proband than that in the healthy volunteer, indicating that DSC2 p.K47Rfs*2 obviously reduced the functional desmocollin2 protein expression in the proband. Conclusion The heterozygous DSC2 p.K47Rfs*2 remarkably and abnormally reduced the functional desmocollin2 expression, which may potentially induce the overlap phenotypes of LVNC and HCM, complicating AF, VT, and HF.
Collapse
Affiliation(s)
- Yubi Lin
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Jiana Huang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.,Reproductive Center, The Six Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510000, China
| | - Zhiling Zhu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zuoquan Zhang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Jianzhong Xian
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zhe Yang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Tingfeng Qin
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Linxi Chen
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Jingmin Huang
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China
| | - Yin Huang
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Qiaoyun Wu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China
| | - Zhenyu Hu
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore
| | - Xiufang Lin
- The Center of Cardiovascular Diseases, The Department of Cardiology, Radiology and Ultrasonography, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, 519000, China.
| | - Geyang Xu
- Department of Physiology, The School of Medicine of Jinan University, Guangzhou, 510000, China.
| |
Collapse
|
12
|
Jiang X, Chen Y, Liu X, Ye L, Yu M, Shen Z, Lei W, Hu S. Uncovering Inherited Cardiomyopathy With Human Induced Pluripotent Stem Cells. Front Cell Dev Biol 2021; 9:672039. [PMID: 34079803 PMCID: PMC8166268 DOI: 10.3389/fcell.2021.672039] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/06/2021] [Indexed: 11/13/2022] Open
Abstract
In the past decades, researchers discovered the contribution of genetic defects to the pathogenesis of primary cardiomyopathy and tried to explain the pathogenesis of these diseases by establishing a variety of disease models. Although human heart tissues and primary cardiomyocytes have advantages in modeling human heart diseases, they are difficult to obtain and culture in vitro. Defects developed in genetically modified animal models are notably different from human diseases at the molecular level. The advent of human induced pluripotent stem cells (hiPSCs) provides an unprecedented opportunity to further investigate the pathogenic mechanisms of inherited cardiomyopathies in vitro using patient-specific hiPSC-derived cardiomyocytes. In this review, we will make a summary of recent advances in in vitro inherited cardiomyopathy modeling using hiPSCs.
Collapse
Affiliation(s)
- Xue Jiang
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Xiaofeng Liu
- The Affiliated Haian Hospital of Nantong University, Nantong, China
| | - Lingqun Ye
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Miao Yu
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Wei Lei
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| | - Shijun Hu
- Department of Cardiovascular Surgery of The First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, China
| |
Collapse
|
13
|
Micheu MM, Rosca AM. Patient-specific induced pluripotent stem cells as "disease-in-a-dish" models for inherited cardiomyopathies and channelopathies - 15 years of research. World J Stem Cells 2021; 13:281-303. [PMID: 33959219 PMCID: PMC8080539 DOI: 10.4252/wjsc.v13.i4.281] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 02/06/2023] Open
Abstract
Among inherited cardiac conditions, a special place is kept by cardiomyopathies (CMPs) and channelopathies (CNPs), which pose a substantial healthcare burden due to the complexity of the therapeutic management and cause early mortality. Like other inherited cardiac conditions, genetic CMPs and CNPs exhibit incomplete penetrance and variable expressivity even within carriers of the same pathogenic deoxyribonucleic acid variant, challenging our understanding of the underlying pathogenic mechanisms. Until recently, the lack of accurate physiological preclinical models hindered the investigation of fundamental cellular and molecular mechanisms. The advent of induced pluripotent stem cell (iPSC) technology, along with advances in gene editing, offered unprecedented opportunities to explore hereditary CMPs and CNPs. Hallmark features of iPSCs include the ability to differentiate into unlimited numbers of cells from any of the three germ layers, genetic identity with the subject from whom they were derived, and ease of gene editing, all of which were used to generate "disease-in-a-dish" models of monogenic cardiac conditions. Functionally, iPSC-derived cardiomyocytes that faithfully recapitulate the patient-specific phenotype, allowed the study of disease mechanisms in an individual-/allele-specific manner, as well as the customization of therapeutic regimen. This review provides a synopsis of the most important iPSC-based models of CMPs and CNPs and the potential use for modeling disease mechanisms, personalized therapy and deoxyribonucleic acid variant functional annotation.
Collapse
Affiliation(s)
- Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Bucharest 014452, Romania.
| | - Ana-Maria Rosca
- Cell and Tissue Engineering Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest 050568, Romania
| |
Collapse
|
14
|
Wang Y, Jiang T, Xu J, Gu Y, Zhou Y, Lin Y, Wu Y, Li W, Wang C, Shen B, Mo X, Wang X, Zhou B, Ding C, Hu Z. Mutations in RNA Methyltransferase Gene NSUN5 Confer High Risk of Outflow Tract Malformation. Front Cell Dev Biol 2021; 9:623394. [PMID: 33968922 PMCID: PMC8097101 DOI: 10.3389/fcell.2021.623394] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/01/2021] [Indexed: 11/13/2022] Open
Abstract
NSUN5, encoding a cytosine-5 RNA methyltransferase and located in the 7q11.23 locus, is a candidate gene for tetralogy of Fallot (TOF). Deletion of the 7q11.23 locus in humans is linked to cardiac outflow tract (OFT) disorders including TOF. We identified four potential pathogenic mutations in the coding region of NSUN5 and which were enriched in TOF patients by an association study of 132 TOF patients and 2,000 in-house controls (P = 1.44 × 10-5). We then generated a Nsun5 null (Nsun5 -/-) mouse model to validate the human findings by defining the functions of Nsun5 in OFT morphogenesis. The OFT did not develop properly in the Nsun5 deletion embryonic heart. We found a misalignment of the aorta and septum defects caused by the delayed fusion of the membraneous ventricular spetum as an OFT development delay. This caused OFT development delay in 27 of 64 (42.2%) Nsun5 -/- mice. Moreover, we also found OFT development delay in 8 of 51 (15.7%) Nsun5 +/- mice. Further functional experiments showed that the loss of Nsun5 function impaired the 5-methylcytosine (m5C) modification and translation efficiency of essential cardiac genes. Nsun5 is required for normal OFT morphogenesis and it regulates the m5C modification of essential cardiac genes. Our findings suggest the involvement of NSUN5 in the pathogenesis of TOF.
Collapse
Affiliation(s)
- Yifeng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Tao Jiang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Jiani Xu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yayun Gu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yan Zhou
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yuan Lin
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Yifei Wu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Wei Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Cheng Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| | - Bin Shen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Xuming Mo
- Department of Cardiothoracic Surgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaowei Wang
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bin Zhou
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, United States
| | - Chenyue Ding
- Center of Reproduction and Genetics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhibin Hu
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, China.,Department of Epidemiology and Biostatistics, Center for Global Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Alimohamed MZ, Johansson LF, Posafalvi A, Boven LG, van Dijk KK, Walters L, Vos YJ, Westers H, Hoedemaekers YM, Sinke RJ, Sijmons RH, Sikkema-Raddatz B, Jongbloed JDH, van der Zwaag PA. Diagnostic yield of targeted next generation sequencing in 2002 Dutch cardiomyopathy patients. Int J Cardiol 2021; 332:99-104. [PMID: 33662488 DOI: 10.1016/j.ijcard.2021.02.069] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/13/2021] [Accepted: 02/17/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Next-generation sequencing (NGS) is increasingly used for clinical evaluation of cardiomyopathy patients as it allows for simultaneous screening of multiple cardiomyopathy-associated genes. Adding copy number variant (CNV) analysis of NGS data is not routine yet and may contribute to the diagnostic yield. OBJECTIVES Determine the diagnostic yield of our targeted NGS gene panel in routine clinical diagnostics of Dutch cardiomyopathy patients and explore the impact of exon CNVs on diagnostic yield. METHODS Patients (N = 2002) referred for clinical genetic analysis underwent diagnostic testing of 55-61 genes associated with cardiomyopathies. Samples were analyzed and evaluated for single nucleotide variants (SNVs), indels and CNVs. CNVs identified in the NGS data and suspected of being pathogenic based on type, size and location were confirmed by additional molecular tests. RESULTS A (likely) pathogenic (L)P variant was detected in 22.7% of patients, including 3 with CNVs and 25 where a variant was identified in a gene currently not associated with the patient's cardiomyopathy subtype. Only 15 out of 2002 patients (0.8%) were found to carry two (L)P variants. CONCLUSION The yield of routine clinical diagnostics of cardiomyopathies was relatively low when compared to literature. This is likely due to the fact that our study reports the outcome of patients in daily routine diagnostics, therefore also including patients not fully fulfilling (subtype specific) cardiomyopathy criteria. This may also explain why (L)P variants were identified in genes not associated with the reported subtype. The added value of CNV analysis was shown to be limited but not negligible.
Collapse
Affiliation(s)
- Mohamed Z Alimohamed
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Lennart F Johansson
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands; University of Groningen, University Medical Center Groningen, Genomics Coordination Center, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Anna Posafalvi
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Ludolf G Boven
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Krista K van Dijk
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Lisa Walters
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Yvonne J Vos
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Helga Westers
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Yvonne M Hoedemaekers
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Richard J Sinke
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Rolf H Sijmons
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Birgit Sikkema-Raddatz
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| | - Jan D H Jongbloed
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Paul A van der Zwaag
- University of Groningen, University Medical Center Groningen, Department of Genetics, Hanzeplein 1, 9713 GZ Groningen, the Netherlands
| |
Collapse
|
16
|
Risi C, Schäfer LU, Belknap B, Pepper I, White HD, Schröder GF, Galkin VE. High-Resolution Cryo-EM Structure of the Cardiac Actomyosin Complex. Structure 2021; 29:50-60.e4. [PMID: 33065066 PMCID: PMC7796959 DOI: 10.1016/j.str.2020.09.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/29/2020] [Accepted: 09/25/2020] [Indexed: 12/31/2022]
Abstract
Heart contraction depends on a complicated array of interactions between sarcomeric proteins required to convert chemical energy into mechanical force. Cyclic interactions between actin and myosin molecules, controlled by troponin and tropomyosin, generate the sliding force between the actin-based thin and myosin-based thick filaments. Alterations in this sophisticated system due to missense mutations can lead to cardiovascular diseases. Numerous structural studies proposed pathological mechanisms of missense mutations at the myosin-myosin, actin-tropomyosin, and tropomyosin-troponin interfaces. However, despite the central role of actomyosin interactions a detailed structural description of the cardiac actomyosin interface remained unknown. Here, we report a cryo-EM structure of a cardiac actomyosin complex at 3.8 Å resolution. The structure reveals the molecular basis of cardiac diseases caused by missense mutations in myosin and actin proteins.
Collapse
Affiliation(s)
- Cristina Risi
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Luisa U Schäfer
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Betty Belknap
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Ian Pepper
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Howard D White
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA
| | - Gunnar F Schröder
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich, 52425 Jülich, Germany; Physics Department, Heinrich-Heine Universität Düsseldorf, 40225 Düsseldorf, Germany
| | - Vitold E Galkin
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA 23507, USA.
| |
Collapse
|
17
|
A comprehensive guide to genetic variants and post-translational modifications of cardiac troponin C. J Muscle Res Cell Motil 2020; 42:323-342. [PMID: 33179204 DOI: 10.1007/s10974-020-09592-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/24/2020] [Indexed: 02/07/2023]
Abstract
Familial cardiomyopathy is an inherited disease that affects the structure and function of heart muscle and has an extreme range of phenotypes. Among the millions of affected individuals, patients with hypertrophic (HCM), dilated (DCM), or left ventricular non-compaction (LVNC) cardiomyopathy can experience morphologic changes of the heart which lead to sudden death in the most detrimental cases. TNNC1, the gene that codes for cardiac troponin C (cTnC), is a sarcomere gene associated with cardiomyopathies in which probands exhibit young age of presentation and high death, transplant or ventricular fibrillation events relative to TNNT2 and TNNI3 probands. Using GnomAD, ClinVar, UniProt and PhosphoSitePlus databases and published literature, an extensive list to date of identified genetic variants in TNNC1 and post-translational modifications (PTMs) in cTnC was compiled. Additionally, a recent cryo-EM structure of the cardiac thin filament regulatory unit was used to localize each functionally studied amino acid variant and each PTM (acetylation, glycation, s-nitrosylation, phosphorylation) in the structure of cTnC. TNNC1 has a large number of variants (> 100) relative to other genes of the same transcript size. Surprisingly, the mapped variant amino acids and PTMs are distributed throughout the cTnC structure. While many cardiomyopathy-associated variants are localized in α-helical regions of cTnC, this was not statistically significant χ2 (p = 0.72). Exploring the variants in TNNC1 and PTMs of cTnC in the contexts of cardiomyopathy association, physiological modulation and potential non-canonical roles provides insights into the normal function of cTnC along with the many facets of TNNC1 as a cardiomyopathic gene.
Collapse
|
18
|
Eintracht J, Toms M, Moosajee M. The Use of Induced Pluripotent Stem Cells as a Model for Developmental Eye Disorders. Front Cell Neurosci 2020; 14:265. [PMID: 32973457 PMCID: PMC7468397 DOI: 10.3389/fncel.2020.00265] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Approximately one-third of childhood blindness is attributed to developmental eye disorders, of which 80% have a genetic cause. Eye morphogenesis is tightly regulated by a highly conserved network of transcription factors when disrupted by genetic mutations can result in severe ocular malformation. Human-induced pluripotent stem cells (hiPSCs) are an attractive tool to study early eye development as they are more physiologically relevant than animal models, can be patient-specific and their use does not elicit the ethical concerns associated with human embryonic stem cells. The generation of self-organizing hiPSC-derived optic cups is a major advancement to understanding mechanisms of ocular development and disease. Their development in vitro has been found to mirror that of the human eye and these early organoids have been used to effectively model microphthalmia caused by a VSX2 variant. hiPSC-derived optic cups, retina, and cornea organoids are powerful tools for future modeling of disease phenotypes and will enable a greater understanding of the pathophysiology of many other developmental eye disorders. These models will also provide an effective platform for identifying molecular therapeutic targets and for future clinical applications.
Collapse
Affiliation(s)
| | - Maria Toms
- UCL Institute of Ophthalmology, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom
| | - Mariya Moosajee
- UCL Institute of Ophthalmology, London, United Kingdom.,The Francis Crick Institute, London, United Kingdom.,Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom.,Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
19
|
Finsterer J, Stöllberger C. Left Ventricular Noncompaction Syndrome: Genetic Insights and Therapeutic Perspectives. Curr Cardiol Rep 2020; 22:84. [DOI: 10.1007/s11886-020-01339-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
20
|
Pioner JM, Fornaro A, Coppini R, Ceschia N, Sacconi L, Donati MA, Favilli S, Poggesi C, Olivotto I, Ferrantini C. Advances in Stem Cell Modeling of Dystrophin-Associated Disease: Implications for the Wider World of Dilated Cardiomyopathy. Front Physiol 2020; 11:368. [PMID: 32477154 PMCID: PMC7235370 DOI: 10.3389/fphys.2020.00368] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Familial dilated cardiomyopathy (DCM) is mostly caused by mutations in genes encoding cytoskeletal and sarcomeric proteins. In the pediatric population, DCM is the predominant type of primitive myocardial disease. A severe form of DCM is associated with mutations in the DMD gene encoding dystrophin, which are the cause of Duchenne Muscular Dystrophy (DMD). DMD-associated cardiomyopathy is still poorly understood and orphan of a specific therapy. In the last 5 years, a rise of interest in disease models using human induced pluripotent stem cells (hiPSCs) has led to more than 50 original studies on DCM models. In this review paper, we provide a comprehensive overview on the advances in DMD cardiomyopathy disease modeling and highlight the most remarkable findings obtained from cardiomyocytes differentiated from hiPSCs of DMD patients. We will also describe how hiPSCs based studies have contributed to the identification of specific myocardial disease mechanisms that may be relevant in the pathogenesis of DCM, representing novel potential therapeutic targets.
Collapse
Affiliation(s)
- Josè Manuel Pioner
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | | | - Raffaele Coppini
- Department of NeuroFarBa, Università degli Studi di Firenze, Florence, Italy
| | - Nicole Ceschia
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Leonardo Sacconi
- LENS, Università degli Studi di Firenze and National Institute of Optics (INO-CNR), Florence, Italy
| | | | - Silvia Favilli
- Pediatric Cardiology, Meyer Children's Hospital, Florence, Italy
| | - Corrado Poggesi
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| | - Iacopo Olivotto
- Cardiomyopathy Unit, Careggi University Hospital, Florence, Italy
| | - Cecilia Ferrantini
- Division of Physiology, Department of Experimental and Clinical Medicine, Università degli Studi di Firenze, Florence, Italy
| |
Collapse
|
21
|
Li C, Liu F, Liu S, Pan H, Du H, Huang J, Xie Y, Li Y, Zhao R, Wei Y. Elevated myocardial SORBS2 and the underlying implications in left ventricular noncompaction cardiomyopathy. EBioMedicine 2020; 53:102695. [PMID: 32143182 PMCID: PMC7058526 DOI: 10.1016/j.ebiom.2020.102695] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 01/19/2023] Open
Abstract
Background Left ventricular noncompaction cardiomyopathy (LVNC) is a hereditary heart disease characterized by an excessive trabecular meshwork of deep intertrabecular recesses within the ventricular myocardium. The guidelines for management of LVNC patients aim to improve quality of life by preventing cardiac heart failure. However, the mechanism underlying LVNC-associated heart failure remains poorly understood. Methods Using protein mass spectrometry analysis, we established that Sorbin And SH3 Domain Containing 2 (SORBS2) is up-regulated in LVNC hearts without changes to structure proteins. We conducted in vivo experiments wherein the heart tissues of wild-type mice were injected with an AAV9 vector to overexpress SORBS2, followed by analysis using echocardiography, T-tubule analysis and Ca2+ imaging to identify functional and morphological changes. In addition, we analyzed the function and structure of SORBS2 overexpressing human embryonic stem cell (hESC) derived cardiomyocytes (hESC-CM) via immunoblotting, immunohistochemistry, immunofluorescence, and confocal Ca2+ imaging. Findings LVNC myocardial tissues feature strongly elevated expression of SORBS2, microtubule densification and redistribution of Junctophilin 2 (JP2). SORBS2 interacts with β-tubulin, promoting its polymerization in 293T cells and hESC-derived CMs. In vivo, cardiac dysfunction, β-tubulin densification, JP2 translocation, T-tubule disorganization and Ca2+ handling dysfunction were observed in mice overexpressing SORBS2. Interpretation We identified a novel mechanism through which SORBS2 interacts with β-tubulin and promotes microtubule densification, eventually effecting JP2 distribution and T-tubule, potentially contributing to heart failure in LVNC disease. Fund This work was supported by a CAMS Initiative for Innovative Medicine grant (CAMS-I2M, 2016-I2M-1-015 to Y.J.Wei)
Collapse
Affiliation(s)
- Chunyan Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Fan Liu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shenghua Liu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Haizhou Pan
- Children's Heart Center, the Second Affiliated Hospital and Yuying Children's Hospital, Institute of Cardiovascular Development and Translational Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haiwei Du
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Jian Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yuanyuan Xie
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yanfen Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Ranxu Zhao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China
| | - Yingjie Wei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Beijing 100037, China.
| |
Collapse
|
22
|
Poetsch MS, Guan K. iPSCs for modeling of sarcomeric cardiomyopathies. RECENT ADVANCES IN IPSC DISEASE MODELING, VOLUME 1 2020:237-273. [DOI: 10.1016/b978-0-12-822227-0.00012-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
23
|
Ichida F. Left ventricular noncompaction - Risk stratification and genetic consideration. J Cardiol 2019; 75:1-9. [PMID: 31629663 DOI: 10.1016/j.jjcc.2019.09.011] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 09/18/2019] [Indexed: 11/30/2022]
Abstract
Left ventricular noncompaction (LVNC) is a cardiomyopathy characterized by two layered structures composed of prominent trabecular meshwork and deep intertrabecular recesses. LVNC was thought to be rare; however, heightened awareness has resulted in an increased detection of the morphological features of LVNC in routine clinical practice especially in the adult population. Although LVNC was classified as an independent primary cardiomyopathy of genetic origin by the American Heart Association in 2006, its definition, diagnostic criteria and clinical implications are still being debated. Clinical manifestations are highly variable, even in the same family, ranging from no symptoms to disabling congestive heart failure, life-threatening arrhythmias, systemic thromboemboli, and sudden cardiac death. Among phenotypic subtypes of LVNC, children with isolated LVNC with normal cardiac function had the best outcomes: children with LVNC and dilated cardiomyopathy had the worst outcomes. Myocardial dysfunction or ventricular arrhythmias are predictors of mortality in adults with LVNC. LVNC, like other forms of inherited cardiomyopathy, is genetically heterogeneous and can be inherited as an autosomal dominant or X-linked recessive disorder. It has been linked to mutations in many genes, including ZASP, TAZ/G4.5, and those encoding sarcomeric, Z-disc, cytoskeleton proteins, and mitochondria. Disturbance of the NOTCH signaling pathway has been reported to be part of genetic pathway for LVNC as well. Although there are an increasing number of reports, genotype-phenotype correlations have been challenging and investigations are ongoing. Patients with mutations are more likely to have major adverse cardiovascular events, further, LV systolic dysfunction in mutation carriers makes them at high risk for cardiac events. Treatments focus on improvement in cardiac function and reduction of mechanical stress in patients with systolic dysfunction and on treatment of arrhythmia and implantation of an automatic implantable cardioverter-defibrillator for prevention of sudden death. Given that 20-40% of cases may be familial, family screening is recommended.
Collapse
Affiliation(s)
- Fukiko Ichida
- Department of Pediatrics, International University of Health and Welfare, Sanno Hospital, 8-10-16, Akasaka, Minato-ku, Tokyo 107-0052, Japan.
| |
Collapse
|