1
|
Pu L, Wang J, Chen Y. A similar severe fibrosis pattern in a monozygotic twin pair with the TRIM63 variant manifesting as hypertrophic cardiomyopathy. Eur Heart J 2024:ehae607. [PMID: 39365612 DOI: 10.1093/eurheartj/ehae607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Affiliation(s)
- Lutong Pu
- Department of Cardiology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
| | - Jie Wang
- Department of Cardiology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
- Cardiac Imaging and Target Therapy Lab, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
| | - Yucheng Chen
- Department of Cardiology, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
- Cardiac Imaging and Target Therapy Lab, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
- Center of Rare Diseases, West China Hospital, Sichuan University, No.37, Guo Xue Alley, Chengdu 610041, Sichuan, China
| |
Collapse
|
2
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
3
|
Lopes LR, Ho CY, Elliott PM. Genetics of hypertrophic cardiomyopathy: established and emerging implications for clinical practice. Eur Heart J 2024; 45:2727-2734. [PMID: 38984491 PMCID: PMC11313585 DOI: 10.1093/eurheartj/ehae421] [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: 08/23/2023] [Revised: 12/05/2023] [Accepted: 06/19/2024] [Indexed: 07/11/2024] Open
Abstract
Pathogenic variation in genes encoding proteins of the cardiac sarcomere is responsible for 30%-40% of cases of hypertrophic cardiomyopathy. The main clinical utility of genetic testing is to provide diagnostic confirmation and facilitation of family screening. It also assists in the detection of aetiologies, which require distinct monitoring and treatment approaches. Other clinical applications, including the use of genetic information to inform risk prediction models, have been limited by the challenge of establishing robust genotype-phenotype correlations with actionable consequences, but new data on the interaction between rare and common genetic variation, as well as the emergence of therapies targeting disease-specific pathogenic mechanisms, herald a new era for genetic testing in routine practice.
Collapse
Affiliation(s)
- Luis R Lopes
- Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 5 University St, London WC1E 6JF, UK
| | - Carolyn Y Ho
- Cardiovascular Division, Brigham and Women’s Hospital, Boston, MA, USA
| | - Perry M Elliott
- Barts Heart Centre, St. Bartholomew’s Hospital, West Smithfield, London EC1A 7BE, UK
- Centre for Heart Muscle Disease, Institute of Cardiovascular Science, University College London, 5 University St, London WC1E 6JF, UK
| |
Collapse
|
4
|
García-Hernández S, de la Higuera Romero L, Ochoa JP, McKenna WJ. Emerging Themes in Genetics of Hypertrophic Cardiomyopathy: Current Status and Clinical Application. Can J Cardiol 2024; 40:742-753. [PMID: 38244984 DOI: 10.1016/j.cjca.2024.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024] Open
Abstract
Hypertrophic cardiomyopathy (HCM), defined clinically by the presence of unexplained left ventricular hypertrophy (LVH), with wall thickness ≥ 1.5 cm, is a phenotype in search of a diagnosis, which is most often a genetically determined, cardiac exclusive, or systemic disorder. Familial evaluation and genetic testing are required for definitive diagnosis. The role of genetic findings in predicting development of disease, outcomes, and increasingly to guide management is evolving with access to larger data sets. The specific mutation and sex of the patient are important determinants that ultimately are likely to guide management. The genetic/familial evaluation is influenced by the accuracy of the clinical diagnosis and the extent/expertise of the genetic laboratory. Genetic testing in a patient with unexplained LVH without systemic manifestations will yield a definite/likely pathogenetic mutation in a sarcomere (30%-50%), regulatory/functional (10%-15%) or metabolic/syndromic (< 5%) gene associated with Mendelian inheritance. The importance of oligo- and polygenic determinants, usually in the absence of Mendelian inheritance, is under investigation with important implications, particularly related to familial evaluation and definition of risk of disease development in relatives of probands. The results of genetic testing are increasingly important in management strategies related to the use of the implantable cardioverter defibrillator for prevention of sudden death, use of myosin inhibitors for refractory symptoms in patients with and without outflow tract obstruction, and-on the immediate horizon-gene therapy. This review will focus on genetic and outcome data in sarcomeric HCM, and minor causative genes with robust evidence of their association will also be considered.
Collapse
Affiliation(s)
| | | | - Juan Pablo Ochoa
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Centro Nacional de Investigaciones Cardiovasculades (CNIC), Madrid, Spain; Health in Code S.L., A Coruña, Spain
| | - William J McKenna
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Universidade da Coruña, A Coruña, Spain; Institute of Cardiovascular Science, University College London, London, United Kingdom; Health in Code S.L., A Coruña, Spain.
| |
Collapse
|
5
|
Allouba M, Walsh R, Afify A, Hosny M, Halawa S, Galal A, Fathy M, Theotokis PI, Boraey A, Ellithy A, Buchan R, Govind R, Whiffin N, Anwer S, ElGuindy A, Ware JS, Barton PJR, Yacoub M, Aguib Y. Ethnicity, consanguinity, and genetic architecture of hypertrophic cardiomyopathy. Eur Heart J 2023; 44:5146-5158. [PMID: 37431535 PMCID: PMC10733735 DOI: 10.1093/eurheartj/ehad372] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 03/28/2023] [Accepted: 05/24/2023] [Indexed: 07/12/2023] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is characterized by phenotypic heterogeneity that is partly explained by the diversity of genetic variants contributing to disease. Accurate interpretation of these variants constitutes a major challenge for diagnosis and implementing precision medicine, especially in understudied populations. The aim is to define the genetic architecture of HCM in North African cohorts with high consanguinity using ancestry-matched cases and controls. METHODS AND RESULTS Prospective Egyptian patients (n = 514) and controls (n = 400) underwent clinical phenotyping and genetic testing. Rare variants in 13 validated HCM genes were classified according to standard clinical guidelines and compared with a prospective HCM cohort of majority European ancestry (n = 684). A higher prevalence of homozygous variants was observed in Egyptian patients (4.1% vs. 0.1%, P = 2 × 10-7), with variants in the minor HCM genes MYL2, MYL3, and CSRP3 more likely to present in homozygosity than the major genes, suggesting these variants are less penetrant in heterozygosity. Biallelic variants in the recessive HCM gene TRIM63 were detected in 2.1% of patients (five-fold greater than European patients), highlighting the importance of recessive inheritance in consanguineous populations. Finally, rare variants in Egyptian HCM patients were less likely to be classified as (likely) pathogenic compared with Europeans (40.8% vs. 61.6%, P = 1.6 × 10-5) due to the underrepresentation of Middle Eastern populations in current reference resources. This proportion increased to 53.3% after incorporating methods that leverage new ancestry-matched controls presented here. CONCLUSION Studying consanguineous populations reveals novel insights with relevance to genetic testing and our understanding of the genetic architecture of HCM.
Collapse
Affiliation(s)
- Mona Allouba
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Roddy Walsh
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, The Netherlands
| | - Alaa Afify
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mohammed Hosny
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Sarah Halawa
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Aya Galal
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Mariam Fathy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Pantazis I Theotokis
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| | - Ahmed Boraey
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- Cardiology Department, Kasr Al Aini Medical School, Cairo University, Kasr Al Aini Street, Cairo 11562, Egypt
| | - Amany Ellithy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Rachel Buchan
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
| | - Risha Govind
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Institute of Psychiatry, Psychology and Neuroscience, King's College London, 16 De Crespigny Park, London SE5 8AF, UK
- Present affiliation: National Institute for Health Research (NIHR) Biomedical Research Centre, South London and Maudsley NHS Foundation Trust and King's College London, 16 De Crespigny Park, London SE5 8AF, UK
| | - Nicola Whiffin
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- Present affiliation: Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Dr, Headington, Oxford OX3 7BN, UK
| | - Shehab Anwer
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - Ahmed ElGuindy
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
| | - James S Ware
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Paul J R Barton
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Royal Brompton & Harefield Hospitals, Guy’s and St. Thomas’ NHS Foundation Trust, London, Sydney St, London SW3 6NP, UK
- MRC London Institute of Medical Sciences, Imperial College London, Du Cane Rd, London W12 0NN, UK
| | - Magdi Yacoub
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
- Harefield Heart Science Centre, Hill End Rd, Harefield, Uxbridge UB9 6JH, UK
| | - Yasmine Aguib
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Kasr El Haggar Street, Aswan 81512, Egypt
- National Heart and Lung Institute, Imperial College London, London, Guy Scadding Building, Dovehouse St, London SW3 6LY, UK
| |
Collapse
|
6
|
Ponińska JK, Pelczar-Płachta W, Pollak A, Jończyk-Potoczna K, Truszkowska G, Michałowska I, Szafran E, Bilińska ZT, Bobkowski W, Płoski R. Double Heterozygous Pathogenic Variants in the LOX and PKD1 Genes in a 5-Year-Old Patient with Thoracic Aortic Aneurysm and Polycystic Kidney Disease. Genes (Basel) 2023; 14:1983. [PMID: 38002926 PMCID: PMC10671125 DOI: 10.3390/genes14111983] [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: 09/22/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Familial thoracic aortic aneurysms and dissections may occur as an isolated hereditary trait or as part of connective tissue disorders with Mendelian inheritance, but severe cardiovascular disease in pediatric patients is extremely rare. There is growing knowledge on pathogenic variants causing the disease; however, much of the phenotypic variability and gene-gene interactions remain to be discovered. We present a case report of a 5.5-year-old girl with an aortic aneurysm and concomitant polycystic kidney disease. Whole exome sequencing was performed, followed by family screening by amplicon deep sequencing and diagnostic imaging studies. In the proband, two pathogenic variants were identified: p.Tyr257Ter in the LOX gene inherited from her mother, and p.Thr2977Ile in the PKD1 gene inherited from her father. All adult carriers of either of these variants showed symptoms of aortic disease. We conclude that the coexistence of two independent genetic variants in the proband may be the reason for an early onset of disease.
Collapse
Affiliation(s)
- Joanna Kinga Ponińska
- Department of Medical Biology, National Institute of Cardiology, 04-628 Warszawa, Poland;
| | - Weronika Pelczar-Płachta
- Department of Pediatric Cardiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Agnieszka Pollak
- Department of Medical Genetics, Centre of Biostructure, Medical University of Warsaw, 02-106 Warszawa, Poland
| | | | - Grażyna Truszkowska
- Department of Medical Biology, National Institute of Cardiology, 04-628 Warszawa, Poland;
| | - Ilona Michałowska
- Department of Radiology, National Institute of Cardiology, 04-628 Warszawa, Poland
| | - Emilia Szafran
- Department of Pediatric Cardiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Zofia T. Bilińska
- Unit for Screening Studies in Inherited Cardiovascular Diseases, National Institute of Cardiology, 04-628 Warszawa, Poland;
| | - Waldemar Bobkowski
- Department of Pediatric Cardiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Centre of Biostructure, Medical University of Warsaw, 02-106 Warszawa, Poland
| |
Collapse
|
7
|
Lipov A, Jurgens SJ, Mazzarotto F, Allouba M, Pirruccello JP, Aguib Y, Gennarelli M, Yacoub MH, Ellinor PT, Bezzina CR, Walsh R. Exploring the complex spectrum of dominance and recessiveness in genetic cardiomyopathies. NATURE CARDIOVASCULAR RESEARCH 2023; 2:1078-1094. [PMID: 38666070 PMCID: PMC11041721 DOI: 10.1038/s44161-023-00346-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/07/2023] [Indexed: 04/28/2024]
Abstract
Discrete categorization of Mendelian disease genes into dominant and recessive models often oversimplifies their underlying genetic architecture. Cardiomyopathies (CMs) are genetic diseases with complex etiologies for which an increasing number of recessive associations have recently been proposed. Here, we comprehensively analyze all published evidence pertaining to biallelic variation associated with CM phenotypes to identify high-confidence recessive genes and explore the spectrum of monoallelic and biallelic variant effects in established recessive and dominant disease genes. We classify 18 genes with robust recessive association with CMs, largely characterized by dilated phenotypes, early disease onset and severe outcomes. Several of these genes have monoallelic association with disease outcomes and cardiac traits in the UK Biobank, including LMOD2 and ALPK3 with dilated and hypertrophic CM, respectively. Our data provide insights into the complex spectrum of dominance and recessiveness in genetic heart disease and demonstrate how such approaches enable the discovery of unexplored genetic associations.
Collapse
Affiliation(s)
- Alex Lipov
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| | - Sean J. Jurgens
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Mona Allouba
- National Heart and Lung Institute, Imperial College London, London, UK
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Aswan, Egypt
| | - James P. Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA USA
| | - Yasmine Aguib
- National Heart and Lung Institute, Imperial College London, London, UK
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Aswan, Egypt
| | - Massimo Gennarelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Genetics Unit, Istituto di Ricovero e Cura a Carattere Scientifico, Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Magdi H. Yacoub
- National Heart and Lung Institute, Imperial College London, London, UK
- Aswan Heart Centre, Magdi Yacoub Heart Foundation, Aswan, Egypt
- Harefield Heart Science Centre, Uxbridge, UK
| | - Patrick T. Ellinor
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA USA
| | - Connie R. Bezzina
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
- European Reference Network for Rare and Low Prevalence Complex Diseases of the Heart, Amsterdam, the Netherlands
| | - Roddy Walsh
- Department of Experimental Cardiology, Heart Centre, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Heart Failure & Arrhythmias, Amsterdam, the Netherlands
| |
Collapse
|
8
|
Renaux A, Terwagne C, Cochez M, Tiddi I, Nowé A, Lenaerts T. A knowledge graph approach to predict and interpret disease-causing gene interactions. BMC Bioinformatics 2023; 24:324. [PMID: 37644440 PMCID: PMC10463539 DOI: 10.1186/s12859-023-05451-5] [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: 06/07/2023] [Accepted: 08/22/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND Understanding the impact of gene interactions on disease phenotypes is increasingly recognised as a crucial aspect of genetic disease research. This trend is reflected by the growing amount of clinical research on oligogenic diseases, where disease manifestations are influenced by combinations of variants on a few specific genes. Although statistical machine-learning methods have been developed to identify relevant genetic variant or gene combinations associated with oligogenic diseases, they rely on abstract features and black-box models, posing challenges to interpretability for medical experts and impeding their ability to comprehend and validate predictions. In this work, we present a novel, interpretable predictive approach based on a knowledge graph that not only provides accurate predictions of disease-causing gene interactions but also offers explanations for these results. RESULTS We introduce BOCK, a knowledge graph constructed to explore disease-causing genetic interactions, integrating curated information on oligogenic diseases from clinical cases with relevant biomedical networks and ontologies. Using this graph, we developed a novel predictive framework based on heterogenous paths connecting gene pairs. This method trains an interpretable decision set model that not only accurately predicts pathogenic gene interactions, but also unveils the patterns associated with these diseases. A unique aspect of our approach is its ability to offer, along with each positive prediction, explanations in the form of subgraphs, revealing the specific entities and relationships that led to each pathogenic prediction. CONCLUSION Our method, built with interpretability in mind, leverages heterogenous path information in knowledge graphs to predict pathogenic gene interactions and generate meaningful explanations. This not only broadens our understanding of the molecular mechanisms underlying oligogenic diseases, but also presents a novel application of knowledge graphs in creating more transparent and insightful predictors for genetic research.
Collapse
Affiliation(s)
- Alexandre Renaux
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles - Vrije Universiteit Brussel, Brussels, Belgium
- Machine Learning Group, Université Libre de Bruxelles, Brussels, Belgium
- Artificial Intelligence lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Chloé Terwagne
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles - Vrije Universiteit Brussel, Brussels, Belgium
- Machine Learning Group, Université Libre de Bruxelles, Brussels, Belgium
| | - Michael Cochez
- Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Discovery Lab, Elsevier, Amsterdam, The Netherlands
| | - Ilaria Tiddi
- Computer Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ann Nowé
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles - Vrije Universiteit Brussel, Brussels, Belgium
- Artificial Intelligence lab, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tom Lenaerts
- Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles - Vrije Universiteit Brussel, Brussels, Belgium
- Machine Learning Group, Université Libre de Bruxelles, Brussels, Belgium
- Artificial Intelligence lab, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
9
|
Glavaški M, Velicki L, Vučinić N. Hypertrophic Cardiomyopathy: Genetic Foundations, Outcomes, Interconnections, and Their Modifiers. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1424. [PMID: 37629714 PMCID: PMC10456451 DOI: 10.3390/medicina59081424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most prevalent heritable cardiomyopathy. HCM is considered to be caused by mutations in cardiac sarcomeric protein genes. Recent research suggests that the genetic foundation of HCM is much more complex than originally postulated. The clinical presentations of HCM are very variable. Some mutation carriers remain asymptomatic, while others develop severe HCM, terminal heart failure, or sudden cardiac death. Heterogeneity regarding both genetic mutations and the clinical course of HCM hinders the establishment of universal genotype-phenotype correlations. However, some trends have been identified. The presence of a mutation in some genes encoding sarcomeric proteins is associated with earlier HCM onset, more severe left ventricular hypertrophy, and worse clinical outcomes. There is a diversity in the mechanisms implicated in the pathogenesis of HCM. They may be classified into groups, but they are interrelated. The lack of known supplementary elements that control the progression of HCM indicates that molecular mechanisms that exist between genotype and clinical presentations may be crucial. Secondary molecular changes in pathways implicated in HCM pathogenesis, post-translational protein modifications, and epigenetic factors affect HCM phenotypes. Cardiac loading conditions, exercise, hypertension, diet, alcohol consumption, microbial infection, obstructive sleep apnea, obesity, and environmental factors are non-molecular aspects that change the HCM phenotype. Many mechanisms are implicated in the course of HCM. They are mostly interconnected and contribute to some extent to final outcomes.
Collapse
Affiliation(s)
- Mila Glavaški
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| | - Lazar Velicki
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
- Institute of Cardiovascular Diseases Vojvodina, Put Doktora Goldmana 4, 21204 Sremska Kamenica, Serbia
| | - Nataša Vučinić
- Faculty of Medicine, University of Novi Sad, Hajduk Veljkova 3, 21000 Novi Sad, Serbia; (L.V.)
| |
Collapse
|
10
|
Chumakova OS, Baulina NM. Advanced searching for hypertrophic cardiomyopathy heritability in real practice tomorrow. Front Cardiovasc Med 2023; 10:1236539. [PMID: 37583586 PMCID: PMC10425241 DOI: 10.3389/fcvm.2023.1236539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/17/2023] [Indexed: 08/17/2023] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common inherited cardiac disease associated with morbidity and mortality at any age. As studies in recent decades have shown, the genetic architecture of HCM is quite complex both in the entire population and in each patient. In the rapidly advancing era of gene therapy, we have to provide a detailed molecular diagnosis to our patients to give them the chance for better and more personalized treatment. In addition to emphasizing the importance of genetic testing in routine practice, this review aims to discuss the possibility to go a step further and create an expanded genetic panel that contains not only variants in core genes but also new candidate genes, including those located in deep intron regions, as well as structural variations. It also highlights the benefits of calculating polygenic risk scores based on a combination of rare and common genetic variants for each patient and of using non-genetic HCM markers, such as microRNAs that can enhance stratification of risk for HCM in unselected populations alongside rare genetic variants and clinical factors. While this review is focusing on HCM, the discussed issues are relevant to other cardiomyopathies.
Collapse
Affiliation(s)
- Olga S. Chumakova
- Laboratory of Functional Genomics of Cardiovascular Diseases, National Medical Research Centre of Cardiology Named After E.I. Chazov, Moscow, Russia
| | | |
Collapse
|
11
|
Jiang XY, Guan FF, Ma JX, Dong W, Qi XL, Zhang X, Chen W, Gao S, Gao X, Pan S, Wang JZ, Ma YW, Zhang LF, Lu D. Cardiac-specific Trim44 knockout in rat attenuates isoproterenol-induced cardiac remodeling via inhibition of AKT/mTOR pathway. Dis Model Mech 2023; 16:276033. [PMID: 35855640 PMCID: PMC9441189 DOI: 10.1242/dmm.049444] [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: 01/03/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
When pathological hypertrophy progresses to heart failure (HF), the prognosis is often very poor. Therefore, it is crucial to find new and effective intervention targets. Here, myocardium-specific Trim44 knockout rats were generated using CRISPR-Cas9 technology. Cardiac phenotypic observations revealed that Trim44 knockout affected cardiac morphology at baseline. Rats with Trim44 deficiency exhibited resistance to cardiac pathological changes in response to stimulation via isoproterenol (ISO) treatment, including improvement of cardiac remodeling and dysfunction by morphological and functional observations, reduced myocardial fibrosis and reduced expression of molecular markers of cardiac stress. Furthermore, signal transduction validation associated with growth and hypertrophy development in vivo and in vitro demonstrated that Trim44 deficiency inhibited the activation of signaling pathways involved in myocardial hypertrophy, especially response to pathological stress. In conclusion, the present study indicates that Trim44 knockout attenuates ISO-induced pathological cardiac remodeling through blocking the AKT/mTOR/GSK3β/P70S6K signaling pathway. This is the first study to demonstrate the function and importance of Trim44 in the heart at baseline and under pathological stress. Trim44 could be a novel therapeutic target for prevention of cardiac hypertrophy and HF. Summary: This is the first study to demonstrate the function of Trim44 in the heart at baseline and under pathological stress. Trim44 could be a novel therapeutic target for prevention of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiao-Yu Jiang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Fei-Fei Guan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Jia-Xin Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xiao-Long Qi
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Wei Chen
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Shan Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Xiang Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Shuo Pan
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Ji-Zheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037, China
| | - Yuan-Wu Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Lian-Feng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| | - Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100021, China.,Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China.,National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing 100021, China
| |
Collapse
|
12
|
Li J, Hu Y, Li J, Wang H, Wu H, Zhao C, Tan T, Zhang L, Zhu D, Liu X, Li N, Hu X. Loss of MuRF1 in Duroc pigs promotes skeletal muscle hypertrophy. Transgenic Res 2023; 32:153-167. [PMID: 37071377 DOI: 10.1007/s11248-023-00342-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/03/2023] [Indexed: 04/19/2023]
Abstract
Muscle mass development depends on increased protein synthesis and reduced muscle protein degradation. Muscle ring-finger protein-1 (MuRF1) plays a key role in controlling muscle atrophy. Its E3 ubiquitin ligase activity recognizes and degrades skeletal muscle proteins through the ubiquitin-proteasome system. The loss of Murf1, which encodes MuRF1, in mice leads to the accumulation of skeletal muscle proteins and alleviation of muscle atrophy. However, the function of Murf1 in agricultural animals remains unclear. Herein, we bred F1 generation Murf1+/- and F2 generation Murf1-/- Duroc pigs from F0 Murf1-/- pigs to investigate the effect of Murf1 knockout on skeletal muscle development. We found that the Murf1+/- pigs retained normal levels of muscle growth and reproduction, and their percentage of lean meat increased by 6% compared to that of the wild type (WT) pigs. Furthermore, the meat color, pH, water-holding capacity, and tenderness of the Murf1+/- pigs were similar to those of the WT pigs. The drip loss rate and intramuscular fat decreased slightly in the Murf1+/- pigs. However, the cross-sectional area of the myofibers in the longissimus dorsi increased in the adult Murf1+/- pigs. The skeletal muscle proteins MYBPC3 and actin, which are targeted by MuRF1, accumulated in the Murf1+/- and Murf1-/- pigs. Our findings show that inhibiting muscle protein degradation in MuRF1-deficient Duroc pigs increases the size of their myofibers and their percentage of lean meat without influencing their growth or pork quality. Our study demonstrates that Murf1 is a target gene for promoting skeletal muscle hypertrophy in pig breeding.
Collapse
Affiliation(s)
- Jiaping Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Yiqing Hu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- National Center for Cardiovascular Diseases, Beijing, People's Republic of China
- National Institute of Biological Sciences, Beijing, People's Republic of China
| | - Jiajia Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Haitao Wang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Hanyu Wu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Chengcheng Zhao
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Tan Tan
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- Development Center of Science and Technology, Ministry of Agriculture and Rural Affairs, Beijing, People's Republic of China
| | - Li Zhang
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, 225300, China
| | - Di Zhu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Xu Liu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China.
| | - Xiaoxiang Hu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, People's Republic of China.
| |
Collapse
|
13
|
Ganekal P, Vastrad B, Vastrad C, Kotrashetti S. Identification of biomarkers, pathways, and potential therapeutic targets for heart failure using next-generation sequencing data and bioinformatics analysis. Ther Adv Cardiovasc Dis 2023; 17:17539447231168471. [PMID: 37092838 PMCID: PMC10134165 DOI: 10.1177/17539447231168471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Heart failure (HF) is the most common cardiovascular diseases and the leading cause of cardiovascular diseases related deaths. Increasing molecular targets have been discovered for HF prognosis and therapy. However, there is still an urgent need to identify novel biomarkers. Therefore, we evaluated biomarkers that might aid the diagnosis and treatment of HF. METHODS We searched next-generation sequencing (NGS) dataset (GSE161472) and identified differentially expressed genes (DEGs) by comparing 47 HF samples and 37 normal control samples using limma in R package. Gene ontology (GO) and pathway enrichment analyses of the DEGs were performed using the g: Profiler database. The protein-protein interaction (PPI) network was plotted with Human Integrated Protein-Protein Interaction rEference (HiPPIE) and visualized using Cytoscape. Module analysis of the PPI network was done using PEWCC1. Then, miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed by Cytoscape software. Finally, we performed receiver operating characteristic (ROC) curve analysis to predict the diagnostic effectiveness of the hub genes. RESULTS A total of 930 DEGs, 464 upregulated genes and 466 downregulated genes, were identified in HF. GO and REACTOME pathway enrichment results showed that DEGs mainly enriched in localization, small molecule metabolic process, SARS-CoV infections, and the citric acid tricarboxylic acid (TCA) cycle and respiratory electron transport. After combining the results of the PPI network miRNA-hub gene regulatory network and TF-hub gene regulatory network, 10 hub genes were selected, including heat shock protein 90 alpha family class A member 1 (HSP90AA1), arrestin beta 2 (ARRB2), myosin heavy chain 9 (MYH9), heat shock protein 90 alpha family class B member 1 (HSP90AB1), filamin A (FLNA), epidermal growth factor receptor (EGFR), phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1), cullin 4A (CUL4A), YEATS domain containing 4 (YEATS4), and lysine acetyltransferase 2B (KAT2B). CONCLUSIONS This discovery-driven study might be useful to provide a novel insight into the diagnosis and treatment of HF. However, more experiments are needed in the future to investigate the functional roles of these genes in HF.
Collapse
Affiliation(s)
- Prashanth Ganekal
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, India
| | - Basavaraj Vastrad
- Department of Pharmaceutical Chemistry, K.L.E. College of Pharmacy, Gadag, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, #253, Bharthinagar, Dharwad 580001, India
| | | |
Collapse
|
14
|
Agarwal R, Wakimoto H, Paulo JA, Zhang Q, Reichart D, Toepfer C, Sharma A, Tai AC, Lun M, Gorham J, DePalma SR, Gygi SP, Seidman J, Seidman CE. Pathogenesis of Cardiomyopathy Caused by Variants in ALPK3, an Essential Pseudokinase in the Cardiomyocyte Nucleus and Sarcomere. Circulation 2022; 146:1674-1693. [PMID: 36321451 PMCID: PMC9698156 DOI: 10.1161/circulationaha.122.059688] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND ALPK3 encodes α-kinase 3, a muscle-specific protein of unknown function. ALPK3 loss-of-function variants cause cardiomyopathy with distinctive clinical manifestations in both children and adults, but the molecular functions of ALPK3 remain poorly understood. METHODS We explored the putative kinase activity of ALPK3 and the consequences of damaging variants using isogenic human induced pluripotent stem cell-derived cardiomyocytes, mice, and human patient tissues. RESULTS Multiple sequence alignment of all human α-kinase domains and their orthologs revealed 4 conserved residues that were variant only in ALPK3, demonstrating evolutionary divergence of the ALPK3 α-kinase domain sequence. Phosphoproteomic evaluation of both ALPK3 kinase domain inhibition and overexpression failed to detect significant changes in catalytic activity, establishing ALPK3 as a pseudokinase. Investigations into alternative functions revealed that ALPK3 colocalized with myomesin proteins (MYOM1, MYOM2) at both the nuclear envelope and the sarcomere M-band. ALPK3 loss-of-function variants caused myomesin proteins to mislocalize and also dysregulated several additional M-band proteins involved in sarcomere protein turnover, which ultimately impaired cardiomyocyte structure and function. CONCLUSIONS ALPK3 is an essential cardiac pseudokinase that inserts in the nuclear envelope and the sarcomere M-band. Loss of ALPK3 causes mislocalization of myomesins, critical force-buffering proteins in cardiomyocytes, and also dysregulates M-band proteins necessary for sarcomere protein turnover. We conclude that ALPK3 cardiomyopathy induces ventricular dilatation caused by insufficient myomesin-mediated force buffering and hypertrophy by impairment of sarcomere proteostasis.
Collapse
Affiliation(s)
- Radhika Agarwal
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Hiroko Wakimoto
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Joao A. Paulo
- Department of Cell Biology (J.A.P., S.P.G.), Harvard Medical School, Boston, MA
| | - Qi Zhang
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Daniel Reichart
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Christopher Toepfer
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Radcliffe Department of Medicine (C.T.), University of Oxford, United Kingdom.,Wellcome Centre for Human Genetics (C.T.), University of Oxford, United Kingdom
| | - Arun Sharma
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Board of Governors Regenerative Medicine Institute (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA.,Smidt Heart Institute (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA.,Department of Biomedical Sciences (A.S.), Cedars-Sinai Medical Center, Los Angeles, CA
| | - Angela C. Tai
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Mingyue Lun
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Joshua Gorham
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Steven R. DePalma
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Steven P. Gygi
- Department of Cell Biology (J.A.P., S.P.G.), Harvard Medical School, Boston, MA
| | - J.G. Seidman
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA
| | - Christine E. Seidman
- Department of Genetics (R.A., H.W., Q.Z., D.R., C.T., A.S., A.C.T., M.L., J.G., S.R.D., J.G.S., C.E.S.), Harvard Medical School, Boston, MA.,Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA (C.E.S.).,Howard Hughes Medical Institute, Chevy Chase, MD (C.E.S.)
| |
Collapse
|
15
|
Arbustini E, Behr ER, Carrier L, van Duijn C, Evans P, Favalli V, van der Harst P, Haugaa KH, Jondeau G, Kääb S, Kaski JP, Kavousi M, Loeys B, Pantazis A, Pinto Y, Schunkert H, Di Toro A, Thum T, Urtis M, Waltenberger J, Elliott P. Interpretation and actionability of genetic variants in cardiomyopathies: a position statement from the European Society of Cardiology Council on cardiovascular genomics. Eur Heart J 2022; 43:1901-1916. [PMID: 35089333 DOI: 10.1093/eurheartj/ehab895] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/03/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
This document describes the contribution of clinical criteria to the interpretation of genetic variants using heritable Mendelian cardiomyopathies as an example. The aim is to assist cardiologists in defining the clinical contribution to a genetic diagnosis and the interpretation of molecular genetic reports. The identification of a genetic variant of unknown or uncertain significance is a limitation of genetic testing, but current guidelines for the interpretation of genetic variants include essential contributions from clinical family screening that can establish a de novo assignment of the variant or its segregation with the phenotype in the family. A partnership between clinicians and patients helps to solve major uncertainties and provides reliable and clinically actionable information.
Collapse
Affiliation(s)
- Eloisa Arbustini
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Elijah R Behr
- Cardiology Research Section and Cardiovascular Clinical Academic Group, Institute of Molecular and Clinical Sciences, St George's, University of London and St George's University Hospitals NHS Foundation Trust, London, UK
| | - Lucie Carrier
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Cornelia van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Paul Evans
- Department of Infection, Immunity and Cardiovascular Disease, and INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK
| | | | - Pim van der Harst
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kristina Hermann Haugaa
- ProCardio Center for Innovation, Department of Cardiology, Oslo University Hospital, Rikshospitalet, Postboks 4950 Nydalen, Oslo 0424, Norway
- University of Oslo, Boks 1072 Blindern, Oslo 0316, Norway
| | - Guillaume Jondeau
- CNMR Syndrome de Marfan et apparentés, Member of VASCERN, AP-HP Hopital Bichat, Service de Cardiologie, 46 rue Henri Huchard, Paris 75018, France
- INSERM LVTS U1148, Paris 75018, France
- Université de Paris, Paris, France
| | - Stefan Kääb
- Medizinische Klinik und Poliklinik I, LMU University Hospital Munich, Munich, Germany
- German Center for Cardiovascular Research, Munich Heart Alliance, Munich, Germany
| | - Juan Pablo Kaski
- Institute of Cardiovascular Science, University College London, London, UK
- Centre for Inherited Cardiovascular Diseases, Great Ormond Street Hospital, London, UK
| | - Maryam Kavousi
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bart Loeys
- Cardiogenomics, Center for Medical Genetics, Antwerp University Hospital/University of Antwerp, Antwerp, Belgium
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Antonis Pantazis
- The Royal Brompton and Harefield Hospitals, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Yigal Pinto
- Department of Experimental Cardiology, University of Amsterdam, Amsterdam University Medical Center, Meibergdreef 15, Amsterdam 1105 AZ, The Netherlands
| | - Heribert Schunkert
- Department of Cardiology, Deutsches Herzzentrum München, Technische Universität München, München, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Munich Heart Alliance, Munich, Germany
| | - Alessandro Di Toro
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
- Fraunhofer Institute of Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Mario Urtis
- Transplant Research Area and Centre for Inherited Cardiovascular Diseases, Department of Medical Sciences and Infectious Diseases, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Johannes Waltenberger
- Department of Cardiology and Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
- Cardiovascular Medicine, Hirslanden Klinik Im Park, Seestrasse 220, Zürich 8027, Switzerland
| | - Perry Elliott
- Barts Heart Centre St Bartholomew's Hospital, London, UK
- Institute for Cardiovascular Science, University College London, London, UK
| |
Collapse
|
16
|
Guenette RG, Yang SW, Min J, Pei B, Potts PR. Target and tissue selectivity of PROTAC degraders. Chem Soc Rev 2022; 51:5740-5756. [PMID: 35587208 DOI: 10.1039/d2cs00200k] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Targeted protein degradation (TPD) strategies have revolutionized how scientists tackle challenging protein targets deemed undruggable with traditional small molecule inhibitors. Many promising campaigns to inhibit proteins have failed due to factors surrounding inhibition selectivity and targeting of compounds to specific tissues and cell types. One of the major improvements that PROTAC (proteolysis targeting chimera) and molecular glue technology can exert is highly selective control of target inhibition. Multiple studies have shown that PROTACs can gain selectivity for their protein targets beyond that of their parent ligands via optimization of linker length and stabilization of ternary complexes. Due to the bifunctional nature of PROTACs, the tissue selective nature of E3 ligases can be exploited to uncover novel targeting mechanisms. In this review, we provide critical analysis of the recent progress towards making selective PROTAC molecules and new PROTAC technologies that will continue to push the boundaries of achieving selectivity. These efforts have wide implications in the future of treating disease as they will broaden the possible targets that can be addressed by small molecules, like undruggable proteins or broadly active targets that would benefit from degradation in specific tissue types.
Collapse
Affiliation(s)
| | - Seung Wook Yang
- Induced Proximity Platform, Amgen, Thousand Oaks, CA 91320, USA.
| | - Jaeki Min
- Induced Proximity Platform, Amgen, Thousand Oaks, CA 91320, USA.
| | - Baikang Pei
- Genome Analysis Unit, Amgen, Thousand Oaks, CA 91320, USA
| | | |
Collapse
|
17
|
Sepp R, Hategan L, Csányi B, Borbás J, Tringer A, Pálinkás ED, Nagy V, Takács H, Latinovics D, Nyolczas N, Pálinkás A, Faludi R, Rábai M, Szabó GT, Czuriga D, Balogh L, Halmosi R, Borbély A, Habon T, Hegedűs Z, Nagy I. The Genetic Architecture of Hypertrophic Cardiomyopathy in Hungary: Analysis of 242 Patients with a Panel of 98 Genes. Diagnostics (Basel) 2022; 12:diagnostics12051132. [PMID: 35626289 PMCID: PMC9139509 DOI: 10.3390/diagnostics12051132] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/24/2022] [Accepted: 04/29/2022] [Indexed: 12/03/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a primary disease of the myocardium most commonly caused by mutations in sarcomeric genes. We aimed to perform a nationwide large-scale genetic analysis of a previously unreported, representative HCM cohort in Hungary. A total of 242 consecutive HCM index patients (127 men, 44 ± 11 years) were studied with next generation sequencing using a custom-designed gene-panel comprising 98 cardiomyopathy-related genes. A total of 90 patients (37%) carried pathogenic/likely pathogenic (P/LP) variants. The percentage of patients with P/LP variants in genes with definitive evidence for HCM association was 93%. Most of the patients with P/LP variants had mutations in MYBPC3 (55 pts, 61%) and in MYH7 (21 pts, 23%). Double P/LP variants were present in four patients (1.7%). P/LP variants in other genes could be detected in ≤3% of patients. Of the patients without P/LP variants, 46 patients (19%) carried a variant of unknown significance. Non-HCM P/LP variants were identified in six patients (2.5%), with two in RAF1 (p.Leu633Val, p.Ser257Leu) and one in DES (p.Arg406Trp), FHL1 (p.Glu96Ter), TTN (p.Lys23480fs), and in the mitochondrial genome (m.3243A>G). Frameshift, nonsense, and splice-variants made up 82% of all P/LP MYBPC3 variants. In all the other genes, missense mutations were the dominant form of variants. The MYBPC3 p.Gln1233Ter, the MYBPC3 p.Pro955ArgfsTer95, and the MYBPC3 p.Ser593ProfsTer11 variants were identified in 12, 7, and 13 patients, respectively. These three variants made up 36% of all patients with identified P/LP variants, raising the possibility of a possible founder effect for these mutations. Similar to other HCM populations, the MYBPC3 and the MYH7 genes seemed to be the most frequently affected genes in Hungarian HCM patients. The high prevalence of three MYBPC3 mutations raises the possibility of a founder effect in our HCM cohort.
Collapse
Affiliation(s)
- Róbert Sepp
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
- Correspondence: ; Tel.: +36-30-267-5845; Fax: +36-62-545-820
| | - Lidia Hategan
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Beáta Csányi
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - János Borbás
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Annamária Tringer
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Eszter Dalma Pálinkás
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Viktória Nagy
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Hedvig Takács
- Division of Non-Invasive Cardiology, Department of Internal Medicine, Faculty of Medicine, University of Szeged, Semmelweis u. 8, H-6725 Szeged, Hungary; (L.H.); (B.C.); (J.B.); (A.T.); (E.D.P.); (V.N.); (H.T.)
| | - Dóra Latinovics
- SeqOmics Biotechnology Ltd., Vállalkozók útja 7, H-6782 Mórahalom, Hungary; (D.L.); (I.N.)
| | - Noémi Nyolczas
- Gottsegen National Cardiovascular Center, Haller u. 29, H-1096 Budapest, Hungary;
- Military Hospital-State Health Center, Róbert Károly körút 44, H-1134 Budapest, Hungary
| | - Attila Pálinkás
- Elisabeth Hospital, Dr. Imre József u. 9, H-6800 Hódmezővásárhely, Hungary;
| | - Réka Faludi
- Heart Institute, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary;
| | - Miklós Rábai
- Division of Cardiology, First Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary; (M.R.); (R.H.); (T.H.)
| | - Gábor Tamás Szabó
- Division of Cardiology and Division of Clinical Physiology, Department of Cardiology, University of Debrecen, Móricz Zsigmond körút 22, H-4032 Debrecen, Hungary; (G.T.S.); (D.C.); (L.B.); (A.B.)
| | - Dániel Czuriga
- Division of Cardiology and Division of Clinical Physiology, Department of Cardiology, University of Debrecen, Móricz Zsigmond körút 22, H-4032 Debrecen, Hungary; (G.T.S.); (D.C.); (L.B.); (A.B.)
| | - László Balogh
- Division of Cardiology and Division of Clinical Physiology, Department of Cardiology, University of Debrecen, Móricz Zsigmond körút 22, H-4032 Debrecen, Hungary; (G.T.S.); (D.C.); (L.B.); (A.B.)
| | - Róbert Halmosi
- Division of Cardiology, First Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary; (M.R.); (R.H.); (T.H.)
- Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, H-7624 Pécs, Hungary
| | - Attila Borbély
- Division of Cardiology and Division of Clinical Physiology, Department of Cardiology, University of Debrecen, Móricz Zsigmond körút 22, H-4032 Debrecen, Hungary; (G.T.S.); (D.C.); (L.B.); (A.B.)
| | - Tamás Habon
- Division of Cardiology, First Department of Medicine, Medical School, University of Pécs, Ifjúság útja 13, H-7624 Pécs, Hungary; (M.R.); (R.H.); (T.H.)
| | - Zoltán Hegedűs
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary;
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Szigeti út 12, H-7624 Pécs, Hungary
| | - István Nagy
- SeqOmics Biotechnology Ltd., Vállalkozók útja 7, H-6782 Mórahalom, Hungary; (D.L.); (I.N.)
- Institute of Biochemistry, Biological Research Center, Eötvös Loránd Research Network, Temesvári krt. 62, H-6726 Szeged, Hungary
| |
Collapse
|
18
|
Andreeva S, Chumakova O, Karelkina E, Lebedeva V, Lubimtseva T, Semenov A, Nikitin A, Speshilov G, Kozyreva A, Sokolnikova P, Zhuk S, Fomicheva Y, Moiseeva O, Kostareva A. Case Report: Two New Cases of Autosomal-Recessive Hypertrophic Cardiomyopathy Associated With TRIM63-Compound Heterozygous Variant. Front Genet 2022; 13:743472. [PMID: 35273634 PMCID: PMC8901572 DOI: 10.3389/fgene.2022.743472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 01/14/2022] [Indexed: 12/15/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is one of the most common hereditary diseases, and it is associated with fatal complications. The clinical heterogeneity of HCM requires risk prediction models to identify patients at a high risk of adverse events. Most HCM cases are caused by mutations in genes encoding sarcomere proteins. However, HCM is associated with rare genetic variants with limited data about its clinical course and prognosis, and existing risk prediction models are not validated for such patients' cohorts. TRIM63 is one of the rare genes recently described as a cause of HCM with autosomal-recessive inheritance. Herein, we present two cases of HCM associated with TRIM63-compound heterozygous variants in young male sportsmen. They demonstrated progressively marked hypertrophy, advanced diastolic dysfunction, a significant degree of fibrosis detected by magnetic resonance imaging, and clear indications for implantable cardioverter-defibrillator. One of the cases includes the first description of TRIM63-HCM with extreme hypertrophy. The presented cases are discussed in light of molecular consequences that might underlie cardiac and muscle phenotype in patients with mutations of TRIM63, the master regulator of striated muscle mass.
Collapse
Affiliation(s)
- Sofiya Andreeva
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Olga Chumakova
- Central State Medical Academy of Department of Presidential Affairs, City Clinical Hospital #17, Moscow, Russia
| | - Elena Karelkina
- Institute of Heart and Vessels, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Viktoriya Lebedeva
- Institute of Heart and Vessels, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Tamara Lubimtseva
- Institute of Heart and Vessels, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Andrey Semenov
- Institute of Heart and Vessels, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Alexey Nikitin
- Pulmonology Research Institute, Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Gleb Speshilov
- Laboratory of Genotyping, N. F. Gamaleya National Research Center, Moscow, Russia
| | - Alexandra Kozyreva
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Polina Sokolnikova
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Sergey Zhuk
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Yuliya Fomicheva
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Olga Moiseeva
- Institute of Heart and Vessels, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Anna Kostareva
- Institute of Molecular Biology and Genetics and World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Solna, Sweden
| |
Collapse
|
19
|
Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol 2022; 19:151-167. [PMID: 34526680 DOI: 10.1038/s41569-021-00608-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2021] [Indexed: 01/06/2023]
Abstract
Hypertrophic cardiomyopathy (HCM) was traditionally described as an autosomal dominant Mendelian disease but is now increasingly recognized as having a complex genetic aetiology. Although eight core genes encoding sarcomeric proteins account for >90% of the pathogenic variants in patients with HCM, variants in several additional genes (ACTN2, ALPK3, CSRP3, FHOD3, FLNC, JPH2, KLHL24, PLN and TRIM63), encoding non-sarcomeric proteins with diverse functions, have been shown to be disease-causing in a small number of patients. Genome-wide association studies (GWAS) have identified numerous loci in cardiomyopathy case-control studies and biobank investigations of left ventricular functional traits. Genes associated with Mendelian cardiomyopathy are enriched in the putative causal gene lists at these loci. Intriguingly, many loci are associated with both HCM and dilated cardiomyopathy but with opposite directions of effect on left ventricular traits, highlighting a genetic basis underlying the contrasting pathophysiological effects observed in each condition. This overlap extends to rare Mendelian variants with distinct variant classes in several genes associated with HCM and dilated cardiomyopathy. In this Review, we appraise the complex contribution of the non-sarcomeric, HCM-associated genes to cardiomyopathies across a range of variant classes (from common non-coding variants of individually low effect size to complete gene knockouts), which provides insights into the genetic basis of cardiomyopathies, causal genes at GWAS loci and the application of clinical genetic testing.
Collapse
|
20
|
Moksnes MR, Røsjø H, Richmond A, Lyngbakken MN, Graham SE, Hansen AF, Wolford BN, Gagliano Taliun SA, LeFaive J, Rasheed H, Thomas LF, Zhou W, Aung N, Surakka I, Douville NJ, Campbell A, Porteous DJ, Petersen SE, Munroe PB, Welsh P, Sattar N, Smith GD, Fritsche LG, Nielsen JB, Åsvold BO, Hveem K, Hayward C, Willer CJ, Brumpton BM, Omland T. Genome-wide association study of cardiac troponin I in the general population. Hum Mol Genet 2021; 30:2027-2039. [PMID: 33961016 PMCID: PMC8522636 DOI: 10.1093/hmg/ddab124] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
Circulating cardiac troponin proteins are associated with structural heart disease and predict incident cardiovascular disease in the general population. However, the genetic contribution to cardiac troponin I (cTnI) concentrations and its causal effect on cardiovascular phenotypes are unclear. We combine data from two large population-based studies, the Trøndelag Health Study and the Generation Scotland Scottish Family Health Study, and perform a genome-wide association study of high-sensitivity cTnI concentrations with 48 115 individuals. We further use two-sample Mendelian randomization to investigate the causal effects of circulating cTnI on acute myocardial infarction (AMI) and heart failure (HF). We identified 12 genetic loci (8 novel) associated with cTnI concentrations. Associated protein-altering variants highlighted putative functional genes: CAND2, HABP2, ANO5, APOH, FHOD3, TNFAIP2, KLKB1 and LMAN1. Phenome-wide association tests in 1688 phecodes and 83 continuous traits in UK Biobank showed associations between a genetic risk score for cTnI and cardiac arrhythmias, metabolic and anthropometric measures. Using two-sample Mendelian randomization, we confirmed the non-causal role of cTnI in AMI (5948 cases, 355 246 controls). We found indications for a causal role of cTnI in HF (47 309 cases and 930 014 controls), but this was not supported by secondary analyses using left ventricular mass as outcome (18 257 individuals). Our findings clarify the biology underlying the heritable contribution to circulating cTnI and support cTnI as a non-causal biomarker for AMI in the general population. Using genetically informed methods for causal inference helps inform the role and value of measuring cTnI in the general population.
Collapse
Affiliation(s)
- Marta R Moksnes
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Helge Røsjø
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Anne Richmond
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Magnus N Lyngbakken
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| | - Sarah E Graham
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ailin Falkmo Hansen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Brooke N Wolford
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Sarah A Gagliano Taliun
- Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Montréal Heart Institute, Montréal, QC H1T 1C8, Canada
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonathon LeFaive
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Humaira Rasheed
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Laurent F Thomas
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, 7491 Trondheim. Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, 7491 Trondheim, Norway
| | - Wei Zhou
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Nay Aung
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Ida Surakka
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nicholas J Douville
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Archie Campbell
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - David J Porteous
- Medical Genetics Section, CGEM, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Steffen E Petersen
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
- Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London EC1A 7BE, UK
| | - Patricia B Munroe
- William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Centre, Queen Mary University of London, London E1 4NS, UK
| | - Paul Welsh
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| | - Lars G Fritsche
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jonas B Nielsen
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology Research, Statens Serum Institute, 2300 Copenhagen, Denmark
- Department of Cardiology, Copenhagen University Hospital, 2100 Copenhagen, Denmark
| | - Bjørn Olav Åsvold
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
- Department of Endocrinology, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Kristian Hveem
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Department of Public Health and Nursing, HUNT Research Centre, NTNU - Norwegian University of Science and Technology, 7600 Levanger, Norway
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Cristen J Willer
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ben M Brumpton
- Department of Public Health and Nursing, K.G. Jebsen Center for Genetic Epidemiology, NTNU - Norwegian University of Science and Technology, 7491 Trondheim, Norway
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Clinic of Thoracic and Occupational Medicine, St. Olavs Hospital, Trondheim University Hospital, 7006 Trondheim, Norway
| | - Torbjørn Omland
- Division of Research and Innovation, Akershus University Hospital, 1478 Lørenskog, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
- Division of Medicine, Department of Cardiology, Akershus University Hospital, 1478 Lørenskog, Norway
| |
Collapse
|
21
|
Affiliation(s)
- Roddy Walsh
- Department of Experimental Cardiology, Amsterdam UMC, AMC Heart Center, Amsterdam, The Netherlands
| | - Connie R Bezzina
- Department of Experimental Cardiology, Amsterdam UMC, AMC Heart Center, Amsterdam, The Netherlands
| |
Collapse
|
22
|
Abstract
Hypertrophic cardiomyopathy (HCM) is a genetic disease of the myocardium characterized by a hypertrophic left ventricle with a preserved or increased ejection fraction. Cardiac hypertrophy is often asymmetrical, which is associated with left ventricular outflow tract obstruction. Myocyte hypertrophy, disarray, and myocardial fibrosis constitute the histological features of HCM. HCM is a relatively benign disease but an important cause of sudden cardiac death in the young and heart failure in the elderly. Pathogenic variants (PVs) in genes encoding protein constituents of the sarcomeres are the main causes of HCM. PVs exhibit a gradient of effect sizes, as reflected in their penetrance and variable phenotypic expression of HCM. MYH7 and MYBPC3, encoding β-myosin heavy chain and myosin binding protein C, respectively, are the two most common causal genes and responsible for ≈40% of all HCM cases but a higher percentage of HCM in large families. PVs in genes encoding protein components of the thin filaments are responsible for ≈5% of the HCM cases. Whereas pathogenicity of the genetic variants in large families has been firmly established, ascertainment causality of the PVs in small families and sporadic cases is challenging. In the latter category, PVs are best considered as probabilistic determinants of HCM. Deciphering the genetic basis of HCM has enabled routine genetic testing and has partially elucidated the underpinning mechanism of HCM as increased number of the myosin molecules that are strongly bound to actin. The discoveries have led to the development of mavacamten that targets binding of the myosin molecule to actin filaments and imparts beneficial clinical effects. In the coming years, the yield of the genetic testing is expected to be improved and the so-called missing causal gene be identified. The advances are also expected to enable development of additional specific therapies and editing of the mutations in HCM.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetics, Institute of Molecular Medicine and Department of Medicine, University of Texas Health Sciences Center at Houston
| |
Collapse
|
23
|
Prashanth G, Vastrad B, Tengli A, Vastrad C, Kotturshetti I. Investigation of candidate genes and mechanisms underlying obesity associated type 2 diabetes mellitus using bioinformatics analysis and screening of small drug molecules. BMC Endocr Disord 2021; 21:80. [PMID: 33902539 PMCID: PMC8074411 DOI: 10.1186/s12902-021-00718-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity associated type 2 diabetes mellitus is a metabolic disorder ; however, the etiology of obesity associated type 2 diabetes mellitus remains largely unknown. There is an urgent need to further broaden the understanding of the molecular mechanism associated in obesity associated type 2 diabetes mellitus. METHODS To screen the differentially expressed genes (DEGs) that might play essential roles in obesity associated type 2 diabetes mellitus, the publicly available expression profiling by high throughput sequencing data (GSE143319) was downloaded and screened for DEGs. Then, Gene Ontology (GO) and REACTOME pathway enrichment analysis were performed. The protein - protein interaction network, miRNA - target genes regulatory network and TF-target gene regulatory network were constructed and analyzed for identification of hub and target genes. The hub genes were validated by receiver operating characteristic (ROC) curve analysis and RT- PCR analysis. Finally, a molecular docking study was performed on over expressed proteins to predict the target small drug molecules. RESULTS A total of 820 DEGs were identified between healthy obese and metabolically unhealthy obese, among 409 up regulated and 411 down regulated genes. The GO enrichment analysis results showed that these DEGs were significantly enriched in ion transmembrane transport, intrinsic component of plasma membrane, transferase activity, transferring phosphorus-containing groups, cell adhesion, integral component of plasma membrane and signaling receptor binding, whereas, the REACTOME pathway enrichment analysis results showed that these DEGs were significantly enriched in integration of energy metabolism and extracellular matrix organization. The hub genes CEBPD, TP73, ESR2, TAB1, MAP 3K5, FN1, UBD, RUNX1, PIK3R2 and TNF, which might play an essential role in obesity associated type 2 diabetes mellitus was further screened. CONCLUSIONS The present study could deepen the understanding of the molecular mechanism of obesity associated type 2 diabetes mellitus, which could be useful in developing therapeutic targets for obesity associated type 2 diabetes mellitus.
Collapse
Affiliation(s)
- G Prashanth
- Department of General Medicine, Basaveshwara Medical College, Chitradurga, Karnataka, 577501, India
| | - Basavaraj Vastrad
- Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka, 582103, India
| | - Anandkumar Tengli
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru and JSS Academy of Higher Education & Research, Mysuru, Karnataka, 570015, India
| | - Chanabasayya Vastrad
- Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad, Karnataka, 580001, India.
| | - Iranna Kotturshetti
- Department of Ayurveda, Rajiv Gandhi Education Society`s Ayurvedic Medical College, Ron, Karnataka, 582209, India
| |
Collapse
|
24
|
Otto CM. Heartbeat: taking care of patients with cardiovascular disease during a pandemic. BRITISH HEART JOURNAL 2020; 106:1283-1285. [DOI: 10.1136/heartjnl-2020-317881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
25
|
Affiliation(s)
- Johanna Kuusisto
- Department of Medicine and Clinical Research, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland
| |
Collapse
|