49 results on '"Francesco Mazzarotto"'
Search Results
2. Calcium handling maturation and adaptation to increased substrate stiffness in human iPSC-derived cardiomyocytes: The impact of full-length dystrophin deficiency
- Author
-
Josè Manuel Pioner, Lorenzo Santini, Chiara Palandri, Marianna Langione, Bruno Grandinetti, Silvia Querceto, Daniele Martella, Costanza Mazzantini, Beatrice Scellini, Lucrezia Giammarino, Flavia Lupi, Francesco Mazzarotto, Aoife Gowran, Davide Rovina, Rosaria Santoro, Giulio Pompilio, Chiara Tesi, Camilla Parmeggiani, Michael Regnier, Elisabetta Cerbai, David L. Mack, Corrado Poggesi, Cecilia Ferrantini, and Raffaele Coppini
- Subjects
human iPSC derived cardiomyocytes ,dystrophin (DMD) ,substrate stiffness ,calcium handing ,duchenne muscular dystrophy (DMD) ,Physiology ,QP1-981 - Abstract
Cardiomyocytes differentiated from human induced Pluripotent Stem Cells (hiPSC- CMs) are a unique source for modelling inherited cardiomyopathies. In particular, the possibility of observing maturation processes in a simple culture dish opens novel perspectives in the study of early-disease defects caused by genetic mutations before the onset of clinical manifestations. For instance, calcium handling abnormalities are considered as a leading cause of cardiomyocyte dysfunction in several genetic-based dilated cardiomyopathies, including rare types such as Duchenne Muscular Dystrophy (DMD)-associated cardiomyopathy. To better define the maturation of calcium handling we simultaneously measured action potential and calcium transients (Ca-Ts) using fluorescent indicators at specific time points. We combined micropatterned substrates with long-term cultures to improve maturation of hiPSC-CMs (60, 75 or 90 days post-differentiation). Control-(hiPSC)-CMs displayed increased maturation over time (90 vs 60 days), with longer action potential duration (APD), increased Ca-T amplitude, faster Ca-T rise (time to peak) and Ca-T decay (RT50). The progressively increased contribution of the SR to Ca release (estimated by post-rest potentiation or Caffeine-induced Ca-Ts) appeared as the main determinant of the progressive rise of Ca-T amplitude during maturation. As an example of severe cardiomyopathy with early onset, we compared hiPSC-CMs generated from a DMD patient (DMD-ΔExon50) and a CRISPR-Cas9 genome edited cell line isogenic to the healthy control with deletion of a G base at position 263 of the DMD gene (c.263delG-CMs). In DMD-hiPSC-CMs, changes of Ca-Ts during maturation were less pronounced: indeed, DMD cells at 90 days showed reduced Ca-T amplitude and faster Ca-T rise and RT50, as compared with control hiPSC-CMs. Caffeine-Ca-T was reduced in amplitude and had a slower time course, suggesting lower SR calcium content and NCX function in DMD vs control cells. Nonetheless, the inotropic and lusitropic responses to forskolin were preserved. CRISPR-induced c.263delG-CM line recapitulated the same developmental calcium handling alterations observed in DMD-CMs. We then tested the effects of micropatterned substrates with higher stiffness. In control hiPSC-CMs, higher stiffness leads to higher amplitude of Ca-T with faster decay kinetics. In hiPSC-CMs lacking full-length dystrophin, however, stiffer substrates did not modify Ca-Ts but only led to higher SR Ca content. These findings highlighted the inability of dystrophin-deficient cardiomyocytes to adjust their calcium homeostasis in response to increases of extracellular matrix stiffness, which suggests a mechanism occurring during the physiological and pathological development (i.e. fibrosis).
- Published
- 2022
- Full Text
- View/download PDF
3. Disease Progression of Hypertrophic Cardiomyopathy: Modeling Using Machine Learning
- Author
-
Matej Pičulin, Tim Smole, Bojan Žunkovič, Enja Kokalj, Marko Robnik-Šikonja, Matjaž Kukar, Dimitrios I Fotiadis, Vasileios C Pezoulas, Nikolaos S Tachos, Fausto Barlocco, Francesco Mazzarotto, Dejana Popović, Lars S Maier, Lazar Velicki, Iacopo Olivotto, Guy A MacGowan, Djordje G Jakovljević, Nenad Filipović, and Zoran Bosnić
- Subjects
Computer applications to medicine. Medical informatics ,R858-859.7 - Abstract
BackgroundCardiovascular disorders in general are responsible for 30% of deaths worldwide. Among them, hypertrophic cardiomyopathy (HCM) is a genetic cardiac disease that is present in about 1 of 500 young adults and can cause sudden cardiac death (SCD). ObjectiveAlthough the current state-of-the-art methods model the risk of SCD for patients, to the best of our knowledge, no methods are available for modeling the patient's clinical status up to 10 years ahead. In this paper, we propose a novel machine learning (ML)-based tool for predicting disease progression for patients diagnosed with HCM in terms of adverse remodeling of the heart during a 10-year period. MethodsThe method consisted of 6 predictive regression models that independently predict future values of 6 clinical characteristics: left atrial size, left atrial volume, left ventricular ejection fraction, New York Heart Association functional classification, left ventricular internal diastolic diameter, and left ventricular internal systolic diameter. We supplemented each prediction with the explanation that is generated using the Shapley additive explanation method. ResultsThe final experiments showed that predictive error is lower on 5 of the 6 constructed models in comparison to experts (on average, by 0.34) or a consortium of experts (on average, by 0.22). The experiments revealed that semisupervised learning and the artificial data from virtual patients help improve predictive accuracies. The best-performing random forest model improved R2 from 0.3 to 0.6. ConclusionsBy engaging medical experts to provide interpretation and validation of the results, we determined the models' favorable performance compared to the performance of experts for 5 of 6 targets.
- Published
- 2022
- Full Text
- View/download PDF
4. Quantitative approaches to variant classification increase the yield and precision of genetic testing in Mendelian diseases: the case of hypertrophic cardiomyopathy
- Author
-
Roddy Walsh, Francesco Mazzarotto, Nicola Whiffin, Rachel Buchan, William Midwinter, Alicja Wilk, Nicholas Li, Leanne Felkin, Nathan Ingold, Risha Govind, Mian Ahmad, Erica Mazaika, Mona Allouba, Xiaolei Zhang, Antonio de Marvao, Sharlene M. Day, Euan Ashley, Steven D. Colan, Michelle Michels, Alexandre C. Pereira, Daniel Jacoby, Carolyn Y. Ho, Kate L. Thomson, Hugh Watkins, Paul J. R. Barton, Iacopo Olivotto, Stuart A. Cook, and James S. Ware
- Subjects
Variant interpretation ,Mendelian genetics ,Hypertrophic cardiomyopathy ,ACMG/AMP guidelines ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background International guidelines for variant interpretation in Mendelian disease set stringent criteria to report a variant as (likely) pathogenic, prioritising control of false-positive rate over test sensitivity and diagnostic yield. Genetic testing is also more likely informative in individuals with well-characterised variants from extensively studied European-ancestry populations. Inherited cardiomyopathies are relatively common Mendelian diseases that allow empirical calibration and assessment of this framework. Methods We compared rare variants in large hypertrophic cardiomyopathy (HCM) cohorts (up to 6179 cases) to reference populations to identify variant classes with high prior likelihoods of pathogenicity, as defined by etiological fraction (EF). We analysed the distribution of variants using a bespoke unsupervised clustering algorithm to identify gene regions in which variants are significantly clustered in cases. Results Analysis of variant distribution identified regions in which variants are significantly enriched in cases and variant location was a better discriminator of pathogenicity than generic computational functional prediction algorithms. Non-truncating variant classes with an EF ≥ 0.95 were identified in five established HCM genes. Applying this approach leads to an estimated 14–20% increase in cases with actionable HCM variants, i.e. variants classified as pathogenic/likely pathogenic that might be used for predictive testing in probands’ relatives. Conclusions When found in a patient confirmed to have disease, novel variants in some genes and regions are empirically shown to have a sufficiently high probability of pathogenicity to support a “likely pathogenic” classification, even without additional segregation or functional data. This could increase the yield of high confidence actionable variants, consistent with the framework and recommendations of current guidelines. The techniques outlined offer a consistent and unbiased approach to variant interpretation for Mendelian disease genetic testing. We propose adaptations to ACMG/AMP guidelines to incorporate such evidence in a quantitative and transparent manner.
- Published
- 2019
- Full Text
- View/download PDF
5. Contemporary Insights Into the Genetics of Hypertrophic Cardiomyopathy: Toward a New Era in Clinical Testing?
- Author
-
Francesco Mazzarotto, Iacopo Olivotto, Beatrice Boschi, Francesca Girolami, Corrado Poggesi, Paul J. R. Barton, and Roddy Walsh
- Subjects
genetic association ,genetic testing ,hypertrophic cardiomyopathy ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Abstract Genetic testing for hypertrophic cardiomyopathy (HCM) is an established clinical technique, supported by 30 years of research into its genetic etiology. Although pathogenic variants are often detected in patients and used to identify at‐risk relatives, the effectiveness of genetic testing has been hampered by ambiguous genetic associations (yielding uncertain and potentially false‐positive results), difficulties in classifying variants, and uncertainty about genotype‐negative patients. Recent case‐control studies on rare variation, improved data sharing, and meta‐analysis of case cohorts contributed to new insights into the genetic basis of HCM. In particular, although research into new genes and mechanisms remains essential, reassessment of Mendelian genetic associations in HCM argues that current clinical genetic testing should be limited to a small number of validated disease genes that yield informative and interpretable results. Accurate and consistent variant interpretation has benefited from new standardized variant interpretation guidelines and innovative approaches to improve classification. Most cases lacking a pathogenic variant are now believed to indicate non‐Mendelian HCM, with more benign prognosis and minimal risk to relatives. Here, we discuss recent advances in the genetics of HCM and their application to clinical genetic testing together with practical issues regarding implementation. Although this review focuses on HCM, many of the issues discussed are also relevant to other inherited cardiac diseases.
- Published
- 2020
- Full Text
- View/download PDF
6. A machine learning-based risk stratification model for ventricular tachycardia and heart failure in hypertrophic cardiomyopathy.
- Author
-
Tim Smole, Bojan Zunkovic, Matej Piculin, Enja Kokalj, Marko Robnik-Sikonja, Matjaz Kukar, Dimitrios I. Fotiadis, Vasileios C. Pezoulas, Nikolaos S. Tachos, Fausto Barlocco, Francesco Mazzarotto, Dejana Popovic, Lars Maier, Lazar U. Velicki, Guy A. MacGowan, Iacopo Olivotto, Nenad Filipovic, Djordje G. Jakovljevic, and Zoran Bosnic
- Published
- 2021
- Full Text
- View/download PDF
7. Slower calcium handling balances faster cross-bridge cycling in human MYBPC3 HCM
- Author
-
Josè Manuel Pioner, Giulia Vitale, Sonette Steczina, Marianna Langione, Francesca Margara, Lorenzo Santini, Francesco Giardini, Erica Lazzeri, Nicoletta Piroddi, Beatrice Scellini, Chiara Palandri, Maike Schuldt, Valentina Spinelli, Francesca Girolami, Francesco Mazzarotto, Jolanda van der Velden, Elisabetta Cerbai, Chiara Tesi, Iacopo Olivotto, Alfonso Bueno-Orovio, Leonardo Sacconi, Raffaele Coppini, Cecilia Ferrantini, Michael Regnier, and Corrado Poggesi
- Subjects
myosin binding protein-C ,founder effect ,Physiology ,hypertrophic cardiomyopathy ,personalized medicine ,sarcomere energetics ,Cardiology and Cardiovascular Medicine - Abstract
Background: The pathogenesis of MYBPC3 -associated hypertrophic cardiomyopathy (HCM) is still unresolved. In our HCM patient cohort, a large and well-characterized population carrying the MYBPC3 :c772G>A variant (p.Glu258Lys, E258K) provides the unique opportunity to study the basic mechanisms of MYBPC3 -HCM with a comprehensive translational approach. Methods: We collected clinical and genetic data from 93 HCM patients carrying the MYBPC3 :c772G>A variant. Functional perturbations were investigated using different biophysical techniques in left ventricular samples from 4 patients who underwent myectomy for refractory outflow obstruction, compared with samples from non-failing non-hypertrophic surgical patients and healthy donors. Human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes and engineered heart tissues (EHTs) were also investigated. Results: Haplotype analysis revealed MYBPC3 :c772G>A as a founder mutation in Tuscany. In ventricular myocardium, the mutation leads to reduced cMyBP-C (cardiac myosin binding protein-C) expression, supporting haploinsufficiency as the main primary disease mechanism. Mechanical studies in single myofibrils and permeabilized muscle strips highlighted faster cross-bridge cycling, and higher energy cost of tension generation. A novel approach based on tissue clearing and advanced optical microscopy supported the idea that the sarcomere energetics dysfunction is intrinsically related with the reduction in cMyBP-C. Studies in single cardiomyocytes (native and hiPSC-derived), intact trabeculae and hiPSC-EHTs revealed prolonged action potentials, slower Ca 2+ transients and preserved twitch duration, suggesting that the slower excitation-contraction coupling counterbalanced the faster sarcomere kinetics. This conclusion was strengthened by in silico simulations. Conclusions: HCM-related MYBPC3 :c772G>A mutation invariably impairs sarcomere energetics and cross-bridge cycling. Compensatory electrophysiological changes (eg, reduced potassium channel expression) appear to preserve twitch contraction parameters, but may expose patients to greater arrhythmic propensity and disease progression. Therapeutic approaches correcting the primary sarcomeric defects may prevent secondary cardiomyocyte remodeling.
- Published
- 2023
- Full Text
- View/download PDF
8. 1152 INVESTIGATION ON THE HIGH INCIDENCE OF THE ATTRV-CAUSING TRANSTHYRETIN VARIANT VAL142ILE IN CENTRAL ITALY
- Author
-
Francesco Cappelli, Alessia Argiro´, Mattia Zampieri, Irene Giotti, Beatrice Boschi, Sabrina Frusconi, Joel Buxbaum, Massimo Gennarelli, Renato Polimanti, Iacopo Olivotto, Federico Perfetto, and Francesco Mazzarotto
- Subjects
Cardiology and Cardiovascular Medicine - Abstract
The p.Val142Ile variant in transthyretin (encoded by the TTR gene) is the most common genetic cause of transthyretin-related amyloidosis. This allele is particularly prevalent in communities of African descent compared with populations of different ancestries, where its frequency is two orders of magnitude lower. For this reason, p.Val142Ile has always been considered an “African” variant, with limited studies performed on individuals of European descent. However, recent reports of higher-than-expected prevalence in European-ancestry populations question the African specificity of this allele. Here we show that the high recurrence of p.Val142Ile in central Italy is due to a founder effect and not to recent admixture from African populations, highlighting how this may be the case in other communities. This suggests a probable underestimate of the global prevalence of p.Val142Ile, and further emphasizes the importance of routine inclusion of TTR in gene panels used for clinical genetic testing in hypertrophic cardiomyopathy (independently of the patient's geographical origin), that transthyretin-related amyloidosis can mimic. Figure.Principal Component Analysis (PCA) of the 16 probands together with the 25044 individuals of the 1000 Genomes Project (Phase 3) having been genotyped both within the 1000 Genomes Project (NA10851_1KG) and in-house (NA10851_internal). The proband clustering within the smear of native Americans is A228.
- Published
- 2022
- Full Text
- View/download PDF
9. Investigation on the high recurrence of the ATTRv-causing transthyretin variant Val142Ile in central Italy
- Author
-
Francesco Mazzarotto, Alessia Argirò, Mattia Zampieri, Chiara Magri, Irene Giotti, Beatrice Boschi, Sabrina Frusconi, Massimo Gennarelli, Joel Buxbaum, Renato Polimanti, Iacopo Olivotto, Federico Perfetto, and Francesco Cappelli
- Subjects
Genetics ,Genetics (clinical) - Abstract
The p.Val142Ile variant in transthyretin (encoded by the TTR gene) is the most common genetic cause of transthyretin-related amyloidosis. This allele is particularly prevalent in communities ofAfrican descent compared with populations of different ancestries, where its frequency is two orders of magnitude lower. For this reason, p.Val142Ile has always been considered an "African" variant, with limited studies performed on individuals of European descent. However, recent reports of higher-than-expected prevalence in European-ancestry populations question the African specificity of this allele. Here we show that the high recurrence of p.Val142Ile in central Italy is due to a founder effect and not to recent admixture from African populations, highlighting how this may be the case in other communities. This suggests a probable underestimate of the global prevalence of p.Val142Ile, and further emphasizes the importance of routine inclusion of TTR in gene panels used for clinical genetic testing in hypertrophic cardiomyopathy (independently of the patient's geographical origin), that transthyretin-related amyloidosis can mimic.
- Published
- 2022
10. Systematic large-scale assessment of the genetic architecture of left ventricular noncompaction reveals diverse etiologies
- Author
-
Antonio de Marvao, Francesca Girolami, Antonis Pantazis, Francesco Mazzarotto, A. John Baksi, James S. Ware, Kathryn A. McGurk, Iacopo Olivotto, Megan H. Hawley, Angharad M. Roberts, Sanjay K Prasad, Roddy Walsh, Elisabetta Cerbai, Paul J.R. Barton, Beatrice Boschi, Ben Statton, Soha Romeih, Leander Beekman, Elisabeth M. Lodder, Declan P. O'Regan, Matteo Beltrami, Connie R. Bezzina, Magdi H. Yacoub, Birgit Funke, Mona Allouba, Yasmine Aguib, Stuart A. Cook, Fondation Leducq, British Heart Foundation, Wellcome Trust, Guys & St Thomas NHS Foundation Trust, Department of Health, Imperial College Healthcare NHS Trust- BRC Funding, Mason Medical Research Foundation, The Academy of Medical Sciences, Cardiology, ACS - Amsterdam Cardiovascular Sciences, Human Genetics, and ACS - Heart failure & arrhythmias
- Subjects
Cardiomyopathy, Dilated ,Heart Defects, Congenital ,0301 basic medicine ,Population ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Biology ,DIAGNOSIS ,Article ,CLASSIFICATION ,DISEASE ,Congenital ,03 medical and health sciences ,0302 clinical medicine ,Dilated ,medicine ,Humans ,Genetic Testing ,cardiovascular diseases ,education ,Genetics (clinical) ,Heart Defects ,CARDIOLOGY ,Genetic testing ,Genetics & Heredity ,Genetics ,0604 Genetics ,education.field_of_study ,Science & Technology ,CARDIOMYOPATHY ,medicine.diagnostic_test ,MUTATIONS ,STATEMENT ,Hypertrophic cardiomyopathy ,1103 Clinical Sciences ,Dilated cardiomyopathy ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Genetic architecture ,030104 developmental biology ,Hypertrophic ,cardiovascular system ,Left ventricular noncompaction ,MYH7 ,Cardiomyopathies ,Life Sciences & Biomedicine - Abstract
Purpose: To characterize the genetic architecture of left ventricular noncompaction (LVNC) and investigate the extent to which it may represent a distinct pathology or a secondary phenotype associated with other cardiac diseases. Methods: We performed rare variant association analysis with 840 LVNC cases and 125,748 gnomAD population controls, and compared results to similar analyses on dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM). Results: We observed substantial genetic overlap indicating that LVNC often represents a phenotypic variation of DCM or HCM. In contrast, truncating variants in MYH7, ACTN2, and PRDM16 were uniquely associated with LVNC and may reflect a distinct LVNC etiology. In particular, MYH7 truncating variants (MYH7tv), generally considered nonpathogenic for cardiomyopathies, were 20-fold enriched in LVNC cases over controls. MYH7tv heterozygotes identified in the UK Biobank and healthy volunteer cohorts also displayed significantly greater noncompaction compared with matched controls. RYR2 exon deletions and HCN4 transmembrane variants were also enriched in LVNC, supporting prior reports of association with arrhythmogenic LVNC phenotypes. Conclusion: LVNC is characterized by substantial genetic overlap with DCM/HCM but is also associated with distinct noncompaction and arrhythmia etiologies. These results will enable enhanced application of LVNC genetic testing and help to distinguish pathological from physiological noncompaction.
- Published
- 2021
- Full Text
- View/download PDF
11. Spatial and Functional Distribution of MYBPC3 Pathogenic Variants and Clinical Outcomes in Patients With Hypertrophic Cardiomyopathy
- Author
-
Francesco Mazzarotto, Carolyn Y. Ho, Helen Woolcock, Adam S. Helms, Michelle Michels, Euan A. Ashley, Steven D. Colan, Sharlene M. Day, Iacopo Olivotto, Samuel G. Wittekind, Jodie Ingles, Alexandre C. Pereira, James S. Ware, Amelia A. Glazier, Christopher Semsarian, Daniel Jacoby, Jaime Yob, Sara Saberi, Neal K. Lakdawala, Juliani Rodriguez, Neha Hafeez, Samat Kabani, Andrea D. Thompson, Wellcome Trust, British Heart Foundation, and Cardiology
- Subjects
0301 basic medicine ,Genetics ,Familial Hypertrophic Cardiomyopathy ,actins ,genotype ,Hypertrophic cardiomyopathy ,myosin ,General Medicine ,030204 cardiovascular system & hematology ,Biology ,hypertrophic cardiomyopathy ,medicine.disease ,3. Good health ,03 medical and health sciences ,MYBPC3 ,030104 developmental biology ,0302 clinical medicine ,Myosin-binding protein C ,Genotype ,Myosin ,medicine ,Distribution (pharmacology) ,In patient ,sarcomere ,Actin - Abstract
Background: Pathogenic variants in MYBPC3 , encoding cardiac MyBP-C (myosin binding protein C), are the most common cause of familial hypertrophic cardiomyopathy. A large number of unique MYBPC3 variants and relatively small genotyped hypertrophic cardiomyopathy cohorts have precluded detailed genotype-phenotype correlations. Methods: Patients with hypertrophic cardiomyopathy and MYBPC3 variants were identified from the Sarcomeric Human Cardiomyopathy Registry. Variant types and locations were analyzed, morphological severity was assessed, and time-event analysis was performed (composite clinical outcome of sudden death, class III/IV heart failure, left ventricular assist device/transplant, atrial fibrillation). For selected missense variants falling in enriched domains, myofilament localization and degradation rates were measured in vitro. Results: Among 4756 genotyped patients with hypertrophic cardiomyopathy in Sarcomeric Human Cardiomyopathy Registry, 1316 patients were identified with adjudicated pathogenic truncating (N=234 unique variants, 1047 patients) or nontruncating (N=22 unique variants, 191 patients) variants in MYBPC3 . Truncating variants were evenly dispersed throughout the gene, and hypertrophy severity and outcomes were not associated with variant location (grouped by 5′–3′ quartiles or by founder variant subgroup). Nontruncating pathogenic variants clustered in the C3, C6, and C10 domains (18 of 22, 82%, P Conclusions: Truncating variants account for 91% of MYBPC3 pathogenic variants and cause similar clinical severity and outcomes regardless of location, consistent with locus-independent loss-of-function. Nontruncating MYBPC3 pathogenic variants are regionally clustered, and a subset also cause loss of function through failure of myofilament incorporation and rapid degradation. Cardiac morphology and clinical outcomes are similar in patients with truncating versus nontruncating variants.
- Published
- 2020
- Full Text
- View/download PDF
12. Phenotypic Expression and Outcomes in Individuals With Rare Genetic Variants of Hypertrophic Cardiomyopathy
- Author
-
Carlo Biffi, Marina Quinlan, Antonio de Marvao, Declan P. O'Regan, Francesco Mazzarotto, Antonis Pantazis, Daniel Rueckert, Marjola Thanaj, James S. Ware, Paul J.R. Barton, Kathryn A. McGurk, Brian P Halliday, Pawel Tokarczuk, Nicola Whiffin, Roddy Walsh, Mikyung Jang, Timothy J W Dawes, Jinming Duan, Catherine Francis, A. John Baksi, Carolyn Y. Ho, Upasana Tayal, Sanjay K Prasad, Ben Statton, Sean L. Zheng, Stuart A. Cook, Pantazis I. Theotokis, Xiao Xu, Nicoló Savioli, Alaine Berry, Wenjia Bai, Rachel Buchan, Xiaolei Zhang, Cardiology, The Academy of Medical Sciences, Imperial College Healthcare NHS Trust- BRC Funding, British Heart Foundation, Wellcome Trust, National Heart & Lung Institute Foundation, Engineering & Physical Science Research Council (EPSRC), and Mason Medical Research Foundation
- Subjects
Male ,Sarcomeres ,Cardiac & Cardiovascular Systems ,cardiovascular magnetic resonance ,deep learning ,genetics ,hypertrophic cardiomyopathy ,penetrance ,Aged ,Cardiomyopathy, Hypertrophic ,Cohort Studies ,Deep Learning ,Female ,Heart Ventricles ,Humans ,Magnetic Resonance Imaging ,Middle Aged ,Penetrance ,Phenotype ,Cardiomyopathy ,MEDICAL GENETICS ,AMERICAN-COLLEGE ,DIAGNOSIS ,GUIDELINES ,RECOMMENDATIONS ,DISEASE ,1117 Public Health and Health Services ,Medicine ,Clinical significance ,cardiovascular diseases ,CLINICAL EXOME ,Gene ,1102 Cardiorespiratory Medicine and Haematology ,Genetics ,RISK ,Science & Technology ,business.industry ,Genetic variants ,Hypertrophic cardiomyopathy ,medicine.disease ,ddc ,Cardiovascular System & Hematology ,Hypertrophic ,Cardiovascular System & Cardiology ,Cardiology and Cardiovascular Medicine ,business ,BURDEN ,Life Sciences & Biomedicine - Abstract
Background: Hypertrophic cardiomyopathy (HCM) is caused by rare variants in sarcomere-encoding genes, but little is known about the clinical significance of these variants in the general population. Objectives: The goal of this study was to compare lifetime outcomes and cardiovascular phenotypes according to the presence of rare variants in sarcomere-encoding genes among middle-aged adults. Methods: This study analyzed whole exome sequencing and cardiac magnetic resonance imaging in UK Biobank participants stratified according to sarcomere-encoding variant status. Results: The prevalence of rare variants (allele frequency
- Published
- 2021
13. Disease Progression of Hypertrophic Cardiomyopathy: Modeling Using Machine Learning (Preprint)
- Author
-
Matej Pičulin, Tim Smole, Bojan Žunkovič, Enja Kokalj, Marko Robnik-Šikonja, Matjaž Kukar, Dimitrios I Fotiadis, Vasileios C Pezoulas, Nikolaos S Tachos, Fausto Barlocco, Francesco Mazzarotto, Dejana Popović, Lars S Maier, Lazar Velicki, Iacopo Olivotto, Guy A MacGowan, Djordje G Jakovljević, Nenad Filipović, and Zoran Bosnić
- Abstract
BACKGROUND Cardiovascular disorders in general are responsible for 30% of deaths worldwide. Among them, hypertrophic cardiomyopathy (HCM) is a genetic cardiac disease that is present in about 1 of 500 young adults and can cause sudden cardiac death (SCD). OBJECTIVE Although the current state-of-the-art methods model the risk of SCD for patients, to the best of our knowledge, no methods are available for modeling the patient's clinical status up to 10 years ahead. In this paper, we propose a novel machine learning (ML)-based tool for predicting disease progression for patients diagnosed with HCM in terms of adverse remodeling of the heart during a 10-year period. METHODS The method consisted of 6 predictive regression models that independently predict future values of 6 clinical characteristics: left atrial size, left atrial volume, left ventricular ejection fraction, New York Heart Association functional classification, left ventricular internal diastolic diameter, and left ventricular internal systolic diameter. We supplemented each prediction with the explanation that is generated using the Shapley additive explanation method. RESULTS The final experiments showed that predictive error is lower on 5 of the 6 constructed models in comparison to experts (on average, by 0.34) or a consortium of experts (on average, by 0.22). The experiments revealed that semisupervised learning and the artificial data from virtual patients help improve predictive accuracies. The best-performing random forest model improved R2 from 0.3 to 0.6. CONCLUSIONS By engaging medical experts to provide interpretation and validation of the results, we determined the models' favorable performance compared to the performance of experts for 5 of 6 targets.
- Published
- 2021
- Full Text
- View/download PDF
14. Evidence-Based Assessment of Genes in Dilated Cardiomyopathy
- Author
-
Francesco Mazzarotto, Matthew S. Edwards, Jodie Ingles, Christopher Semsarian, Stacey Peters, James S. Ware, Ronald H. Lekanne Deprez, Petros Syrris, Rudy Celeghin, Judy Fan, Argelia Medeiros Domingo, Jessica Wang, Ray E. Hershberger, Lucas Bronicki, Kalliopi Pilichou, Laiken Peterson, Daniel P. Judge, Najim Lahrouchi, Rebecca L. Miller, Ana Morales, Tomohiko Ai, Brittney Murray, Renee Johnson, Elizabeth Jordan, J. Peter van Tintelen, Courtney Thaxton, Alexandros Protonotarios, Emily Brown, Cynthia A. James, Babken Asatryan, Palak Shah, R. Thomas Lumbers, Roddy Walsh, Olga Jarinova, Wellcome Trust, British Heart Foundation, National Heart & Lung Institute Foundation, Cardiology, ACS - Heart failure & arrhythmias, Human Genetics, ACS - Pulmonary hypertension & thrombosis, and ARD - Amsterdam Reproduction and Development
- Subjects
Cardiomyopathy, Dilated ,medicine.medical_specialty ,Cardiomyopathy ,Clinical Sciences ,610 Medicine & health ,030204 cardiovascular system & hematology ,Cardiorespiratory Medicine and Haematology ,Cardiovascular ,Right ventricular cardiomyopathy ,1117 Public Health and Health Services ,03 medical and health sciences ,0302 clinical medicine ,Rare Diseases ,Physiology (medical) ,Internal medicine ,Dilated ,Genetics ,Medicine ,Humans ,genetics ,Genetic Predisposition to Disease ,cardiovascular diseases ,Genetic Testing ,1102 Cardiorespiratory Medicine and Haematology ,Expert Testimony ,030304 developmental biology ,0303 health sciences ,Evidence-Based Medicine ,business.industry ,Hypertrophic cardiomyopathy ,Dilated cardiomyopathy ,1103 Clinical Sciences ,medicine.disease ,Heart Disease ,Cardiovascular System & Hematology ,Cardiology ,Public Health and Health Services ,cardiovascular system ,cardiomyopathy ,Cardiology and Cardiovascular Medicine ,business ,Evidence based assessment ,Biotechnology - Abstract
Background: Each of the cardiomyopathies, classically categorized as hypertrophic cardiomyopathy, dilated cardiomyopathy (DCM), and arrhythmogenic right ventricular cardiomyopathy, has a signature genetic theme. Hypertrophic cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy are largely understood as genetic diseases of sarcomere or desmosome proteins, respectively. In contrast, >250 genes spanning >10 gene ontologies have been implicated in DCM, representing a complex and diverse genetic architecture. To clarify this, a systematic curation of evidence to establish the relationship of genes with DCM was conducted. Methods: An international panel with clinical and scientific expertise in DCM genetics evaluated evidence supporting monogenic relationships of genes with idiopathic DCM. The panel used the Clinical Genome Resource semiquantitative gene-disease clinical validity classification framework with modifications for DCM genetics to classify genes into categories on the basis of the strength of currently available evidence. Representation of DCM genes on clinically available genetic testing panels was evaluated. Results: Fifty-one genes with human genetic evidence were curated. Twelve genes (23%) from 8 gene ontologies were classified as having definitive ( BAG3 , DES , FLNC , LMNA , MYH7 , PLN , RBM20 , SCN5A , TNNC1 , TNNT2 , TTN ) or strong ( DSP ) evidence. Seven genes (14%; ACTC1 , ACTN2 , JPH2 , NEXN , TNNI3 , TPM1 , VCL ) including 2 additional ontologies were classified as moderate evidence; these genes are likely to emerge as strong or definitive with additional evidence. Of these 19 genes, 6 were similarly classified for hypertrophic cardiomyopathy and 3 for arrhythmogenic right ventricular cardiomyopathy. Of the remaining 32 genes (63%), 25 (49%) had limited evidence, 4 (8%) were disputed, 2 (4%) had no disease relationship, and 1 (2%) was supported by animal model data only. Of the 16 evaluated clinical genetic testing panels, most definitive genes were included, but panels also included numerous genes with minimal human evidence. Conclusions: In the curation of 51 genes, 19 had high evidence (12 definitive/strong, 7 moderate). It is notable that these 19 genes explain only a minority of cases, leaving the remainder of DCM genetic architecture incompletely addressed. Clinical genetic testing panels include most high-evidence genes; however, genes lacking robust evidence are also commonly included. We recommend that high-evidence DCM genes be used for clinical practice and that caution be exercised in the interpretation of variants in variable-evidence DCM genes.
- Published
- 2021
- Full Text
- View/download PDF
15. Shared genetic pathways contribute to risk of hypertrophic and dilated cardiomyopathies with opposite directions of effect
- Author
-
Antonio de Marvao, Roddy Walsh, Jean-Claude Tardif, R. Thomas Lumbers, Eric Villard, Rafik Tadros, Peter Lichtner, Catherine Francis, Julie Amyot, Michelle Michels, Hugh Watkins, Julia Cadrin-Tourigny, Najim Lahrouchi, Rudolf A. de Boer, Patrick Garceau, Karin J. H. Verweij, Paul M. Matthews, Paul Elliott, S. Matthijs Boekholdt, Folkert W. Asselbergs, Declan P. O'Regan, Benjamin Meder, Joost A. Offerhaus, Nicola Whiffin, Jacco C. Karper, Jason D. Roberts, Marie-Pierre Dubé, Hideaki Suzuki, James S. Ware, Yigal M. Pinto, Thomas Meitinger, Guillaume Lettre, Hannah G. van Velzen, Arthur A.M. Wilde, Marjon van Slegtenhorst, Francesco Mazzarotto, Wouter P. te Rijdt, Paul J.R. Barton, Sanjay K Prasad, A. John Baksi, Michael W.T. Tanck, Mario Talajic, Roy Huurman, J. Peter van Tintelen, Connie R. Bezzina, Antonis Pantazis, Robert A. Hegele, Jentien M Vermeulen, Rachel Buchan, Imke Christiaans, Jan H. Veldink, Edgar T. Hoorntje, Elham Kayvanpour, Pascale Richard, Geneviève Giraldeau, Flavie Ader, Andrew Thain, Philippe L. L’Allier, Xiao Xu, Leander Beekman, David McCarty, Alexa M.C. Vermeer, Geraldine Sloane, Wenjia Bai, Andrew R. Harper, Jolanda van der Velden, Stuart A. Cook, Ken Kelu Bisabu, Philippe Charron, Deborah Schneider-Luftman, Human Genetics, ACS - Heart failure & arrhythmias, Cardiology, Adult Psychiatry, APH - Mental Health, ANS - Complex Trait Genetics, ANS - Compulsivity, Impulsivity & Attention, ANS - Mood, Anxiety, Psychosis, Stress & Sleep, ACS - Amsterdam Cardiovascular Sciences, Epidemiology and Data Science, APH - Methodology, ACS - Atherosclerosis & ischemic syndromes, Human genetics, Physiology, Cardiovascular Centre (CVC), Clinical Genetics, Wellcome Trust, Department of Health, British Heart Foundation, Engineering & Physical Science Research Council (EPSRC), UK DRI Ltd, The Academy of Medical Sciences, and Imperial College Healthcare NHS Trust- BRC Funding
- Subjects
Linkage disequilibrium ,Cardiomyopathy, Dilated/genetics ,Left ,Cardiomyopathy ,Genome-wide association study ,Kaplan-Meier Estimate ,VARIANTS ,Ventricular Function, Left ,Linkage Disequilibrium ,0302 clinical medicine ,Gene Frequency ,Dilated ,Ventricular Function ,11 Medical and Health Sciences ,Cardiomyopathy, Hypertrophic/genetics ,Genetics & Heredity ,0303 health sciences ,HERITABILITY ,Single Nucleotide ,MENDELIAN RANDOMIZATION ,Cardiology ,cardiovascular system ,HEART ,Life Sciences & Biomedicine ,Ventricular Function, Left/genetics ,Cardiomyopathy, Dilated ,medicine.medical_specialty ,Heart Ventricles ,Quantitative Trait Loci ,Biology ,Quantitative trait locus ,Sudden death ,Polymorphism, Single Nucleotide ,Article ,Heart Ventricles/physiopathology ,03 medical and health sciences ,Internal medicine ,Mendelian randomization ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,cardiovascular diseases ,GENOME-WIDE ASSOCIATION ,Polymorphism ,030304 developmental biology ,Genetic association ,Science & Technology ,Hypertrophic/genetics ,Left/genetics ,Case-control study ,CONTRACTILITY ,06 Biological Sciences ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Hypertrophic ,Dilated/genetics ,Case-Control Studies ,Genome-Wide Association Study ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The heart muscle diseases hypertrophic (HCM) and dilated (DCM) cardiomyopathies are leading causes of sudden death and heart failure in young, otherwise healthy, individuals. We conducted genome-wide association studies and multi-trait analyses in HCM (1,733 cases), DCM (5,521 cases) and nine left ventricular (LV) traits (19,260 UK Biobank participants with structurally normal hearts). We identified 16 loci associated with HCM, 13 with DCM and 23 with LV traits. We show strong genetic correlations between LV traits and cardiomyopathies, with opposing effects in HCM and DCM. Two-sample Mendelian randomization supports a causal association linking increased LV contractility with HCM risk. A polygenic risk score explains a significant portion of phenotypic variability in carriers of HCM-causing rare variants. Our findings thus provide evidence that polygenic risk score may account for variability in Mendelian diseases. More broadly, we provide insights into how genetic pathways may lead to distinct disorders through opposing genetic effects.
- Published
- 2021
- Full Text
- View/download PDF
16. Outcomes and phenotypic expression of rare variants in hypertrophic cardiomyopathy genes amongst UK Biobank participants
- Author
-
de Marvao A, Daniel Rueckert, Alaine Berry, Nicoló Savioli, Carlo Biffi, Declan P. O'Regan, Marina Quinlan, Roddy Walsh, Carolyn Y. Ho, S Prasad, Jang M, Francesco Mazzarotto, Nicola Whiffin, Upasana Tayal, Pantazis I. Theotokis, Pawel Tokarczuk, Kathryn A. McGurk, S A Cook, Timothy J W Dawes, James S. Ware, Brian P Halliday, Antonis Pantazis, Benjamin Statton, Marjola Thanaj, Jinming Duan, Catherine Francis, Sean L. Zheng, Paul J.R. Barton, Xiaolei Zhang, Wenjia Bai, Baksi Aj, Xiao Xu, and Rachel Buchan
- Subjects
medicine.medical_specialty ,education.field_of_study ,medicine.diagnostic_test ,business.industry ,Population ,Cardiomyopathy ,Hypertrophic cardiomyopathy ,medicine.disease ,Gastroenterology ,Penetrance ,Cardiac magnetic resonance imaging ,Internal medicine ,Heart failure ,medicine ,Clinical significance ,education ,business ,Allele frequency - Abstract
BackgroundHypertrophic cardiomyopathy (HCM) is caused by rare variants in sarcomere-encoding genes, but little is known about the clinical significance of these variants in the general population.MethodsWe compared outcomes and cardiovascular phenotypes in UK Biobank participants with whole exome sequencing stratified by sarcomere-encoding variant status.ResultsThe prevalence of rare variants (allele frequency ConclusionsIn the general population, SARC-P/LP variants have low aggregate penetrance for overt HCM but are associated with an increased risk of adverse cardiovascular outcomes and a sub-clinical cardiomyopathic phenotype. In contrast, rare sarcomeric variants that do not meet criteria to be classified as P/LP appear to have minimal clinical impact.
- Published
- 2021
- Full Text
- View/download PDF
17. Arrhythmogenic potential of myocardial disarray in hypertrophic cardiomyopathy: genetic basis, functional consequences and relation to sudden cardiac death
- Author
-
Francesco Mazzarotto, Michael Papadakis, Leonardo Sacconi, Nabeel Sheikh, Gherardo Finocchiaro, Iacopo Olivotto, Antonis Pantazis, Ornella Leone, Cecilia Ferrantini, Mary N. Sheppard, Joe Westaby, and Sanjay Sharma
- Subjects
medicine.medical_specialty ,Heart Diseases ,Cardiomyopathy ,Disease ,030204 cardiovascular system & hematology ,Sudden death ,030218 nuclear medicine & medical imaging ,Sudden cardiac death ,Hypertrophic cardiomyopathy ,Myocardial disarray ,Death, Sudden, Cardiac ,Humans ,Myocardium ,Myocytes, Cardiac ,Cardiomyopathy, Hypertrophic ,03 medical and health sciences ,0302 clinical medicine ,Molecular level ,Physiology (medical) ,Internal medicine ,medicine ,cardiovascular diseases ,Pathological ,Myocytes ,business.industry ,Diagnostic marker ,medicine.disease ,Sudden ,Death ,Hypertrophic ,cardiovascular system ,Cardiology ,Cardiology and Cardiovascular Medicine ,business ,Cardiac - Abstract
Myocardial disarray is defined as disorganized cardiomyocyte spatial distribution, with loss of physiological fibre alignment and orientation. Since the first pathological descriptions of hypertrophic cardiomyopathy (HCM), disarray appeared as a typical feature of this condition and sparked vivid debate regarding its specificity to the disease and clinical significance as a diagnostic marker and a risk factor for sudden death. Although much of the controversy surrounding its diagnostic value in HCM persists, it is increasingly recognized that myocardial disarray may be found in physiological contexts and in cardiac conditions different from HCM, raising the possibility that central focus should be placed on its quantity and distribution, rather than a mere presence. While further studies are needed to establish what amount of disarray should be considered as a hallmark of the disease, novel experimental approaches and emerging imaging techniques for the first time allow ex vivo and in vivo characterization of the myocardium to a molecular level. Such advances hold the promise of filling major gaps in our understanding of the functional consequences of myocardial disarray in HCM and specifically on arrhythmogenic propensity and as a risk factor for sudden death. Ultimately, these studies will clarify whether disarray represents a major determinant of the HCM clinical profile, and a potential therapeutic target, as opposed to an intriguing but largely innocent bystander.
- Published
- 2021
18. Evaluation of gene validity for CPVT and short QT syndrome in sudden arrhythmic death
- Author
-
Simona Amenta, Amy C. Sturm, Ahmad S. Amin, Francesco Mazzarotto, Valentina Trevisan, Eline A. Nannenberg, Arnon Adler, Roddy Walsh, Emanuela Abiusi, Harriet Feilotter, Melanie Care, Arthur A.M. Wilde, Marco V Perez, Hennie Bikker, Wojciech Zareba, James S. Ware, Ray E. Hershberger, Michael H. Gollob, John Garcia, Valeria Novelli, Cardiology, ACS - Heart failure & arrhythmias, Human Genetics, ACS - Pulmonary hypertension & thrombosis, ARD - Amsterdam Reproduction and Development, ACS - Atherosclerosis & ischemic syndromes, ACS - Amsterdam Cardiovascular Sciences, and Human genetics
- Subjects
Cardiac & Cardiovascular Systems ,Genetic testing ,VARIANTS ,030204 cardiovascular system & hematology ,GUIDELINES ,0302 clinical medicine ,Catecholaminergic polymorphic ventricular tachycardia ,Mendelian genetics ,Short QT syndrome ,1102 Cardiorespiratory Medicine and Haematology ,Genetics ,0303 health sciences ,education.field_of_study ,medicine.diagnostic_test ,ASSOCIATION ,3. Good health ,KCNQ1 MUTATION ,KCNQ1 Potassium Channel ,symbols ,Cardiology and Cardiovascular Medicine ,Life Sciences & Biomedicine ,Settore BIO/18 - GENETICA ,Population ,Sudden death ,03 medical and health sciences ,symbols.namesake ,Calmodulin ,Channelopathy ,ANK2 ,medicine ,Humans ,education ,030304 developmental biology ,CARNITINE DEFICIENCY ,Science & Technology ,POLYMORPHIC VENTRICULAR-TACHYCARDIA ,business.industry ,Arrhythmias, Cardiac ,Ryanodine Receptor Calcium Release Channel ,1103 Clinical Sciences ,medicine.disease ,Death, Sudden, Cardiac ,Cardiovascular System & Hematology ,Tachycardia, Ventricular ,Cardiovascular System & Cardiology ,Mendelian inheritance ,business - Abstract
Aims Catecholaminergic polymorphic ventricular tachycardia (CPVT) and short QT syndrome (SQTS) are inherited arrhythmogenic disorders that can cause sudden death. Numerous genes have been reported to cause these conditions, but evidence supporting these gene–disease relationships varies considerably. To ensure appropriate utilization of genetic information for CPVT and SQTS patients, we applied an evidence-based reappraisal of previously reported genes. Methods and results Three teams independently curated all published evidence for 11 CPVT and 9 SQTS implicated genes using the ClinGen gene curation framework. The results were reviewed by a Channelopathy Expert Panel who provided the final classifications. Seven genes had definitive to moderate evidence for disease causation in CPVT, with either autosomal dominant (RYR2, CALM1, CALM2, CALM3) or autosomal recessive (CASQ2, TRDN, TECRL) inheritance. Three of the four disputed genes for CPVT (KCNJ2, PKP2, SCN5A) were deemed by the Expert Panel to be reported for phenotypes that were not representative of CPVT, while reported variants in a fourth gene (ANK2) were too common in the population to be disease-causing. For SQTS, only one gene (KCNH2) was classified as definitive, with three others (KCNQ1, KCNJ2, SLC4A3) having strong to moderate evidence. The majority of genetic evidence for SQTS genes was derived from very few variants (five in KCNJ2, two in KCNH2, one in KCNQ1/SLC4A3). Conclusions Seven CPVT and four SQTS genes have valid evidence for disease causation and should be included in genetic testing panels. Additional genes associated with conditions that may mimic clinical features of CPVT/SQTS have potential utility for differential diagnosis.
- Published
- 2021
- Full Text
- View/download PDF
19. An Evidence-based Assessment of Genes in Dilated Cardiomyopathy
- Author
-
Petros Syrris, Judy Fan, Ana Morales, Olga Jarinova, Cynthia A. James, Tomohiko Ai, Argelia Medeiros Domingo, Emily Brown, R. Thomas Lumbers, Brittney Murray, Ronald H. Lekanne Deprez, Ray E. Hershberger, Lucas Bronicki, Laiken Peterson, Christopher Semsarian, Courtney Thaxton, Alexandros Protonotarios, Kalliopi Pilichou, Roddy Walsh, Stacey Peters, Rudy Celeghin, Daniel P. Judge, Rebecca C. Miller, Jessica Wang, Francesco Mazzarotto, Matthew S. Edwards, Jodie Ingles, Babken Asatryan, Palak Shah, James S. Ware, J. Peter van Tintelen, Elizabeth Jordan, Renee Johnson, and Najim Lahrouchi
- Subjects
medicine.diagnostic_test ,TNNT2 ,TPM1 ,Computational biology ,Biology ,Genetic architecture ,TNNI3 ,LMNA ,cardiovascular system ,medicine ,MYH7 ,cardiovascular diseases ,FLNC ,Genetic testing - Abstract
Background: The cardiomyopathies are classically categorized as hypertrophic (HCM), dilated (DCM), and arrhythmogenic right ventricular (ARVC), and each have a signature genetic theme. HCM and ARVC are largely understood as genetic diseases of sarcomere or desmosome proteins, respectively. In contrast, >250 genes spanning more than 10 gene ontologies have been implicated in DCM, representing a complex and diverse genetic architecture. To clarify this, a systematic curation of evidence to establish the relationship of genes with DCM was conducted. Methods: An international Panel with clinical and scientific expertise in DCM genetics was assembled to evaluate evidence supporting monogenic relationships of genes with idiopathic DCM. The Panel utilized the ClinGen semi-quantitative gene-disease clinical validity classification framework. Results: Fifty-one genes with human genetic evidence were curated. Twelve genes (23%) from eight gene ontologies were classified as having definitive (BAG3, DES, FLNC, LMNA, MYH7, PLN, RBM20, SCN5A, TNNC1, TNNT2, TTN) or strong (DSP) evidence. Seven genes (14%) (ACTC1, ACTN2, JPH2, NEXN, TNNI3, TPM1, VCL) including two additional ontologies were classified as moderate evidence; these genes are likely to emerge as strong or definitive with additional evidence. Of the 19 genes classified as definitive, strong or moderate, six were similarly classified for HCM and three for ARVC. Of the remaining 32 genes (63%), 25 (49%) had limited evidence, 4 (8%) were disputed, 2 (4%) had no disease relationship, and 1 (2%) was supported by animal model data only. Of 16 commercially available genetic testing panels evaluated, most definitive genes were included, but panels also included numerous genes with minimal human evidence. Conclusions: In a systematic curation of published evidence for genes considered relevant for monogenic DCM, 12 were classified as definitive or strong and seven as moderate evidence spanning 10 gene ontologies. Notably, these 19 genes only explain a minority of DCM cases, leaving the remainder of DCM genetic architecture incompletely addressed. While clinical genetic testing panels include most high evidence genes, genes lacking robust evidence are also commonly included. Until the genetic architecture of DCM is more fully defined, care should be taken in the interpretation of variable evidence DCM genes in clinical practice.
- Published
- 2020
- Full Text
- View/download PDF
20. Computational prediction of protein subdomain stability in MYBPC3 enables clinical risk stratification in hypertrophic cardiomyopathy and enhances variant interpretation
- Author
-
Carolyn Y. Ho, Sara Saberi, Francesco Mazzarotto, Samuel G. Wittekind, Euan A. Ashley, Sharlene M. Day, James S. Ware, Alexandre C. Pereira, Anamika Kannan, Michelle Michels, Iacopo Olivotto, Jaime Yob, Adam S. Helms, Andrea D. Thompson, Neal K. Lakdawala, Christopher Semsarian, Daniel Jacoby, Steven D. Colan, Jodie Ingles, Wellcome Trust, British Heart Foundation, and Cardiology
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Cardiomyopathy ,Mutation, Missense ,030105 genetics & heredity ,Risk Assessment ,Article ,03 medical and health sciences ,SDG 3 - Good Health and Well-being ,Internal medicine ,medicine ,Missense mutation ,Humans ,Stroke ,Uncertain significance ,Genetics (clinical) ,Loss function ,Genetics & Heredity ,0604 Genetics ,Science & Technology ,business.industry ,Hazard ratio ,Hypertrophic cardiomyopathy ,1103 Clinical Sciences ,Atrial fibrillation ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Carrier Proteins ,Mutation ,Cardiomyopathies ,030104 developmental biology ,Hypertrophic ,Heart failure ,Cardiology ,Missense ,business ,Life Sciences & Biomedicine ,Clinical risk factor - Abstract
PurposeVariants inMYBPC3causing loss-of-function are the most common cause of HCM. However, a substantial number of patients carry missense variants of uncertain significance (VUS) inMYBPC3.We hypothesize that a structural-based algorithm, STRUM, which estimates the effect of missense variants on protein folding, will improve clinical risk stratification of patients with HCM and aMYBPC3VUS.MethodsAmong 7,963 patients in the multi-center Sarcomeric Human Cardiomyopathy Registry, 120 unique missense VUSs inMYBPC3were identified. Variants were evaluated for their effect on subdomain folding and a stratified time-to-event analysis for an overall composite endpoint (first occurrence of ventricular arrhythmia, heart failure, all-cause mortality, atrial fibrillation, and stroke) was performed for patients with HCM and aMYBPC3missense VUS.ResultsWe demonstrated that patients carrying aMYBPC3VUS predicted to cause subdomain misfolding (STRUM +, ΔΔG ≤-1.2 kcal/mol) exhibited a higher rate of adverse events compared to those with a STRUM-VUS (Hazard Ratio=2.29, P=0.0282).In silicosaturation mutagenesis ofMYBPC3identified 4,943/23,427 (21%) missense variants that were predicted to cause subdomain misfolding.ConclusionsSTRUM enables clinical risk stratification of patients with HCM and aMYBPC3VUS and has the capacity to improve prognostic predictions and clinical decision making.
- Published
- 2020
21. Enhancing rare variant interpretation in inherited arrhythmias through quantitative analysis of 5182 cases from long QT syndrome and Brugada syndrome consortia cohorts and gnomAD population controls
- Author
-
Rafik Tadros, Najim Lahrouchi, Connie R. Bezzina, Roddy Walsh, C Krijger, J S Ware, Nicola Whiffin, Doris Škorić-Milosavljević, Charlotte Glinge, and Francesco Mazzarotto
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Pediatrics ,medicine.medical_specialty ,education.field_of_study ,business.industry ,Long QT syndrome ,Interpretation (philosophy) ,Population ,medicine.disease ,medicine ,Cardiology and Cardiovascular Medicine ,business ,education ,Brugada syndrome - Abstract
Background/Introduction Guidelines for variant interpretation in Mendelian disease set stringent criteria to report a variant as (likely) pathogenic, prioritising control of false positive rate over test sensitivity and diagnostic yield, and require customisation for the specific genetic characteristics of gene-disease dyads. Inherited arrhythmias like long QT syndrome (LQTS) and Brugada syndrome (BrS) are genetically heterogeneous, with missense variants constituting the preponderance of disease-causing variants. Evidence from family segregation or functional assays to confirm pathogenicity are often unavailable or impractical in clinical genetic testing, leading to high rates of variants of uncertain significance and false negative test results. Methods We compared rare variant frequencies from 1847 LQTS (KCNQ1, KCNH2, SCN5A) and 3335 BrS (SCN5A) cases from the International LQTS/BrS Genetics Consortia to population-specific gnomAD data. We propose disease-specific criteria for ACMG/AMP evidence classes – rarity (PM2/BS1 rules) and enrichment of individual (PS4) and domain-specific (PM1) variants in cases over controls. Results Rare SCN5A variant prevalence differed between BrS cases with spontaneous (28.7%) versus induced (15.8%) type 1 Brugada ECG (p=1.3x10–13) and between European (20.8%) and Japanese (8.9%) patients (p=8.8x10–18). Transmembrane regions and specific N-terminus (KCNH2) and C-terminus (KCNQ1/KCNH2) domains were characterised by high enrichment of case variants and >95% probability of pathogenicity. Applying the customised rules, 17.5% of European BrS cases and 73.7% of European LQTS cases had variants classified as (likely) pathogenic, compared to estimated diagnostic yields (case excess over gnomAD) of 19.3%/82.6%. Conclusions Large case/control datasets enable quantitative implementation of ACMG/AMP guidelines and increased sensitivity for inherited arrhythmia genetic testing. Classification of Brugada/LQTS variants Funding Acknowledgement Type of funding source: Foundation. Main funding source(s): Dutch Heart Foundation, Netherlands Organisation for Scientific Research
- Published
- 2020
- Full Text
- View/download PDF
22. The genetic architecture of left ventricular non-compaction reveals both substantial overlap with other cardiomyopathies and a distinct aetiology in a subset of cases
- Author
-
Francesca Girolami, Francesco Mazzarotto, Megan H. Hawley, Roddy Walsh, Elisabetta Cerbai, Stuart A. Cook, Leander Beekman, Beatrice Boschi, Matteo Beltrami, Connie R. Bezzina, Birgit Funke, Angharad M. Roberts, James S. Ware, Iacopo Olivotto, and Paul J.R. Barton
- Subjects
business.industry ,Evolutionary biology ,Etiology ,Medicine ,Cardiology and Cardiovascular Medicine ,business ,Genetic architecture - Abstract
Background Left ventricular non-compaction (LVNC) is a condition characterised by trabeculations in the myocardial wall and is the subject of considerable conjecture as to whether it represents a distinct pathology or a secondary phenotype associated with other cardiac diseases, particularly cardiomyopathies. Purpose To investigate the genetic architecture of LVNC by identifying genes and variant classes robustly associated with disease and comparing these to other genetically characterised cardiomyopathies. Methods We performed rare variant association analysis using six different LVNC cohorts comprising 840 cases together with 125,748 gnomAD population controls and compared results to similar analyses with dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) cases. Results We observed substantial overlap in genes and variant classes enriched in LVNC and DCM/HCM, indicating that in many cases LVNC belongs to a spectrum of more established cardiomyopathies, with non-compaction representing a phenotypic variation in patients with DCM- or HCM-causing variants. In contrast, five variant classes were uniquely enriched in LVNC cases, of which truncating variants in MYH7, ACTN2 and PRDM16 may represent a distinct LVNC aetiology. MYH7 truncating variants are generally considered as non-pathogenic but were detected in 2% of LVNC cases compared to 0.1% of controls, including a cluster of variants around a single splice region. Additionally, structural variants (exon deletions) in RYR2 and missense variants in the transmembrane region of HCN4 were enriched in LVNC cases, confirming prior reports regarding the association of these variant classes with combined LVNC and arrhythmia phenotypes. Conclusions We demonstrated that genetic association analysis can clarify the relationship between LVNC and established cardiomyopathies, highlighted substantial overlap with DCM/HCM but also identified variant classes associated with distinct LVNC and with joint LVNC/arrhythmia phenotypes. These results underline the complex genetic landscape of LVNC and inform how genetic testing in LVNC cases should be pursued and interpreted. Cardiomyopathy rare variant frequencies Funding Acknowledgement Type of funding source: None
- Published
- 2020
- Full Text
- View/download PDF
23. Spatial and Functional Distribution of
- Author
-
Adam S, Helms, Andrea D, Thompson, Amelia A, Glazier, Neha, Hafeez, Samat, Kabani, Juliani, Rodriguez, Jaime M, Yob, Helen, Woolcock, Francesco, Mazzarotto, Neal K, Lakdawala, Samuel G, Wittekind, Alexandre C, Pereira, Daniel L, Jacoby, Steven D, Colan, Euan A, Ashley, Sara, Saberi, James S, Ware, Jodie, Ingles, Christopher, Semsarian, Michelle, Michels, Iacopo, Olivotto, Carolyn Y, Ho, and Sharlene M, Day
- Subjects
Adult ,Male ,Adolescent ,Genotype ,myosin ,Severity of Illness Index ,Young Adult ,Myofibrils ,Humans ,Registries ,Child ,Spatial Analysis ,Polymorphism, Genetic ,actins ,Original Articles ,Cardiomyopathy, Hypertrophic ,Middle Aged ,hypertrophic cardiomyopathy ,Phenotype ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Female ,sarcomere ,Carrier Proteins - Abstract
Supplemental Digital Content is available in the text., Background: Pathogenic variants in MYBPC3, encoding cardiac MyBP-C (myosin binding protein C), are the most common cause of familial hypertrophic cardiomyopathy. A large number of unique MYBPC3 variants and relatively small genotyped hypertrophic cardiomyopathy cohorts have precluded detailed genotype-phenotype correlations. Methods: Patients with hypertrophic cardiomyopathy and MYBPC3 variants were identified from the Sarcomeric Human Cardiomyopathy Registry. Variant types and locations were analyzed, morphological severity was assessed, and time-event analysis was performed (composite clinical outcome of sudden death, class III/IV heart failure, left ventricular assist device/transplant, atrial fibrillation). For selected missense variants falling in enriched domains, myofilament localization and degradation rates were measured in vitro. Results: Among 4756 genotyped patients with hypertrophic cardiomyopathy in Sarcomeric Human Cardiomyopathy Registry, 1316 patients were identified with adjudicated pathogenic truncating (N=234 unique variants, 1047 patients) or nontruncating (N=22 unique variants, 191 patients) variants in MYBPC3. Truncating variants were evenly dispersed throughout the gene, and hypertrophy severity and outcomes were not associated with variant location (grouped by 5′–3′ quartiles or by founder variant subgroup). Nontruncating pathogenic variants clustered in the C3, C6, and C10 domains (18 of 22, 82%, P
- Published
- 2020
24. Enhancing rare variant interpretation in inherited arrhythmias through quantitative analysis of consortium disease cohorts and population controls
- Author
-
Charles Antzelevitch, Ahmad S. Amin, Bo Gregers Winkel, Dan M. Roden, Stefan Kääb, Jonathan R. Skinner, Seiko Ohno, Julien Barc, Birgit Stallmeyer, Carla Giustetto, Connie R. Bezzina, Francesco Mazzarotto, Tomas Robyns, Carlo Napolitano, Stellan Mörner, Doris Škorić-Milosavljević, Sven Dittmann, Kenichiro Yamagata, Sonia Van Dooren, Anniek Corveleyn, Carlo de Asmundis, Ramon Brugada, K Usuda, Yuji Tanaka, Sven Zumhagen, Tadashi Nakajima, Johan Saenen, Elijah R. Behr, Hector Barajas-Martinez, Stéphane Bézieau, Masao Yoshinaga, Georgia Sarquella-Brugada, Paul G.A. Volders, Juan R. Gimeno, Lia Crotti, Charlotte Glinge, Andrea Mazzanti, Ingrid P.C. Krapels, Nicola Whiffin, Sebastian Clauss, Yoshiaki Kaneko, James S. Ware, Minoru Horie, Keiko Shimamoto, Isabelle Denjoy, Pieter G. Postema, Christian Krijger, Takeshi Aiba, Masahiko Kurabayashi, Pyotr G. Platonov, Regina Sebastiano, Cristina Gil Ortuño, Annika Rydberg, Roddy Walsh, Michael J. Ackerman, Hideki Itoh, M. Benjamin Shoemaker, Can Hasdemir, Pascale Guicheney, J. Martijn Bos, Frederic Sacher, Takeru Makiyama, Julieta Lazarte, Maarten P. van den Berg, Dominique Babuty, David J. Tester, Silvia Castelletti, Jacques Mansourati, Antoine Leenhardt, Paul A. van der Zwaag, Sanjay Sharma, Elena Arbelo, Candan Celen, Pier D. Lambiase, Maria Christina Kotta, Johannes Steinfurt, Jean-Baptiste Gourraud, Pedro Brugada, Wataru Shimizu, Josep Brugada, Jørgen K. Kanters, Eline A. Nannenberg, Silvia G. Priori, Mary N. Sheppard, Richard Redon, Morten S. Olesen, Jeroen Breckpot, Britt M. Beckmann, Naomasa Makita, Martin Borggrefe, Rafik Tadros, Jean-Jacques Schott, Jacob Tfelt-Hansen, Steven A. Lubitz, Hatice Şahin, Najim Lahrouchi, Michael Papadakis, Daisuke Hazeki, Kenshi Hayashi, Oscar Campuzano, Katja E. Odening, Federica Dagradi, Eric Schulze-Bahr, Boris Rudic, Hiroki Kimoto, Vincent Probst, Jason D. Roberts, Raphaël P. Martins, Bart Loeys, Daniela F. Giachino, F. Kyndt, Kimie Ohkubo, Taisuke Ishikawa, Catarina Lundin, Lut Van Laer, Patrick T. Ellinor, Maria Sabater Molina, Peter J. Schwartz, Annika Winbo, Wellcome Trust, Rosetrees Trust, British Heart Foundation, Walsh, R, Lahrouchi, N, Tadros, R, Kyndt, F, Glinge, C, Postema, P, Amin, A, Nannenberg, E, Ware, J, Whiffin, N, Mazzarotto, F, Skoric-Milosavljevic, D, Krijger, C, Arbelo, E, Babuty, D, Barajas-Martinez, H, Beckmann, B, Bezieau, S, Bos, J, Breckpot, J, Campuzano, O, Castelletti, S, Celen, C, Clauss, S, Corveleyn, A, Crotti, L, Dagradi, F, de Asmundis, C, Denjoy, I, Dittmann, S, Ellinor, P, Ortuno, C, Giustetto, C, Gourraud, J, Hazeki, D, Horie, M, Ishikawa, T, Itoh, H, Kaneko, Y, Kanters, J, Kimoto, H, Kotta, M, Krapels, I, Kurabayashi, M, Lazarte, J, Leenhardt, A, Loeys, B, Lundin, C, Makiyama, T, Mansourati, J, Martins, R, Mazzanti, A, Morner, S, Napolitano, C, Ohkubo, K, Papadakis, M, Rudic, B, Molina, M, Sacher, F, Sahin, H, Sarquella-Brugada, G, Sebastiano, R, Sharma, S, Sheppard, M, Shimamoto, K, Shoemaker, M, Stallmeyer, B, Steinfurt, J, Tanaka, Y, Tester, D, Usuda, K, van der Zwaag, P, Van Dooren, S, Van Laer, L, Winbo, A, Winkel, B, Yamagata, K, Zumhagen, S, Volders, P, Lubitz, S, Antzelevitch, C, Platonov, P, Odening, K, Roden, D, Roberts, J, Skinner, J, Tfelt-Hansen, J, van den Berg, M, Olesen, M, Lambiase, P, Borggrefe, M, Hayashi, K, Rydberg, A, Nakajima, T, Yoshinaga, M, Saenen, J, Kaab, S, Brugada, P, Robyns, T, Giachino, D, Ackerman, M, Brugada, R, Brugada, J, Gimeno, J, Hasdemir, C, Guicheney, P, Priori, S, Schulze-Bahr, E, Makita, N, Schwartz, P, Shimizu, W, Aiba, T, Schott, J, Redon, R, Ohno, S, Probst, V, Arnaout, A, Amelot, M, Anselme, F, Billon, O, Defaye, P, Dupuis, J, Jesel, L, Laurent, G, Maury, P, Pasquie, J, Wiart, F, Behr, E, Barc, J, Bezzina, C, unité de recherche de l'institut du thorax UMR1087 UMR6291 (ITX), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN), Centre Hospitalier Régional Universitaire de Tours (CHRU TOURS), CIC - CHU Bichat, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Régional Universitaire de Brest (CHRU Brest), CHU Pontchaillou [Rennes], Unité de Recherche sur les Maladies Cardiovasculaires, du Métabolisme et de la Nutrition = Institute of cardiometabolism and nutrition (ICAN), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU), Ege Üniversitesi, Cardiovascular Centre (CVC), Nantes Referral Ctr Inherited Car, Cardiology, Graduate School, ACS - Heart failure & arrhythmias, Human Genetics, ACS - Amsterdam Cardiovascular Sciences, ACS - Atherosclerosis & ischemic syndromes, MUMC+: DA KG Polikliniek (9), RS: Carim - H02 Cardiomyopathy, Cardiologie, MUMC+: MA Med Staf Spec Cardiologie (9), RS: Carim - H04 Arrhythmogenesis and cardiogenetics, Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional Universitaire de Tours (CHRU Tours), Clinical sciences, Heartrhythmmanagement, Medical Genetics, Reproduction and Genetics, and Cardio-vascular diseases
- Subjects
0301 basic medicine ,[SDV]Life Sciences [q-bio] ,Nantes Referral Center for inherited cardiac arrhythmia ,Disease ,Arrhythmias ,030105 genetics & heredity ,ACMG/AMP guidelines ,Brugada ,LQTS ,variant interpretation ,Medicine ,Genetics (clinical) ,Brugada Syndrome ,Brugada syndrome ,Genetics ,Genetics & Heredity ,education.field_of_study ,medicine.diagnostic_test ,Molecular pathology ,3. Good health ,Long QT Syndrome ,Medical genetics ,Population Control ,Cardiology and Cardiovascular Medicine ,Cardiac ,Medical Genetics ,Life Sciences & Biomedicine ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Long QT syndrome ,Population ,610 Medicine & health ,BIO/18 - GENETICA ,Article ,03 medical and health sciences ,Humans ,Genetic Testing ,cardiovascular diseases ,education ,Medicinsk genetik ,Genetic testing ,0604 Genetics ,Science & Technology ,business.industry ,Genetic heterogeneity ,MUTATIONS ,ACMG/AMP guideline ,Arrhythmias, Cardiac ,1103 Clinical Sciences ,MED/11 - MALATTIE DELL'APPARATO CARDIOVASCOLARE ,medicine.disease ,Mutation ,030104 developmental biology ,Human medicine ,business - Abstract
Purpose: Stringent variant interpretation guidelines can lead to high rates of variants of uncertain significance (VUS) for genetically heterogeneous disease like long QT syndrome (LQTS) and Brugada syndrome (BrS). Quantitative and disease-specific customization of American College of Medical Genetics and Genomics/Association for Molecular Pathology (ACMG/AMP) guidelines can address this false negative rate. Methods: We compared rare variant frequencies from 1847 LQTS (KCNQ1/KCNH2/SCN5A) and 3335 BrS (SCN5A) cases from the International LQTS/BrS Genetics Consortia to population-specific gnomAD data and developed disease-specific criteria for ACMG/AMP evidence classes-rarity (PM2/BS1 rules) and case enrichment of individual (PS4) and domain-specific (PM1) variants. Results: Rare SCN5A variant prevalence differed between European (20.8%) and Japanese (8.9%) BrS patients (p = 5.7 x 10(-18)) and diagnosis with spontaneous (28.7%) versus induced (15.8%) Brugada type 1 electrocardiogram (ECG) (p = 1.3 x 10(-13)). Ion channel transmembrane regions and specific N-terminus (KCNH2) and C-terminus (KCNQ1/KCNH2) domains were characterized by high enrichment of case variants and >95% probability of pathogenicity. Applying the customized rules, 17.4% of European BrS and 74.8% of European LQTS cases had (likely) pathogenic variants, compared with estimated diagnostic yields (case excess over gnomAD) of 19.2%/82.1%, reducing VUS prevalence to close to background rare variant frequency. Conclusion: Large case-control data sets enable quantitative implementation of ACMG/AMP guidelines and increased sensitivity for inherited arrhythmia genetic testing., Amsterdam Cardiovascular Sciences fellowship; Dutch Heart Foundation (CVON Predict-2/Concor-genes); Netherlands Organization for Scientific ResearchNetherlands Organization for Scientific Research (NWO) [VICI 016.150.610]; Fondation LeducqLeducq Foundation, R.W. is supported by an Amsterdam Cardiovascular Sciences fellowship. C.R.B. is supported by the Dutch Heart Foundation (CVON Predict-2/Concor-genes), Netherlands Organization for Scientific Research (VICI 016.150.610), and Fondation Leducq. See Supplement for all Acknowledgements.
- Published
- 2020
- Full Text
- View/download PDF
25. Contemporary Insights Into the Genetics of Hypertrophic Cardiomyopathy: Toward a New Era in Clinical Testing?
- Author
-
Francesca Girolami, Paul J.R. Barton, Corrado Poggesi, Francesco Mazzarotto, Roddy Walsh, Beatrice Boschi, Iacopo Olivotto, Imperial College Healthcare NHS Trust- BRC Funding, and Cardiology
- Subjects
genetic association ,Cardiomyopathy ,Diagnostic Testing ,030204 cardiovascular system & hematology ,Risk Assessment ,genetic testing ,Genetic, Association Studies ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Predictive Value of Tests ,Risk Factors ,Genetic etiology ,Contemporary Review ,Clinical genetic ,Humans ,Medicine ,Genetic Predisposition to Disease ,In patient ,1102 Cardiorespiratory Medicine and Haematology ,030304 developmental biology ,Genetic association ,Genetic testing ,Disease gene ,Genetics ,0303 health sciences ,medicine.diagnostic_test ,business.industry ,Hypertrophic cardiomyopathy ,Genetic Variation ,Reproducibility of Results ,Hypertrophy ,Cardiomyopathy, Hypertrophic ,hypertrophic cardiomyopathy ,Prognosis ,medicine.disease ,3. Good health ,Phenotype ,Mendelian inheritance ,symbols ,Cardiology and Cardiovascular Medicine ,business - Abstract
Genetic testing for hypertrophic cardiomyopathy ( HCM ) is an established clinical technique, supported by 30 years of research into its genetic etiology. Although pathogenic variants are often detected in patients and used to identify at‐risk relatives, the effectiveness of genetic testing has been hampered by ambiguous genetic associations (yielding uncertain and potentially false‐positive results), difficulties in classifying variants, and uncertainty about genotype‐negative patients. Recent case‐control studies on rare variation, improved data sharing, and meta‐analysis of case cohorts contributed to new insights into the genetic basis of HCM . In particular, although research into new genes and mechanisms remains essential, reassessment of Mendelian genetic associations in HCM argues that current clinical genetic testing should be limited to a small number of validated disease genes that yield informative and interpretable results. Accurate and consistent variant interpretation has benefited from new standardized variant interpretation guidelines and innovative approaches to improve classification. Most cases lacking a pathogenic variant are now believed to indicate non‐Mendelian HCM , with more benign prognosis and minimal risk to relatives. Here, we discuss recent advances in the genetics of HCM and their application to clinical genetic testing together with practical issues regarding implementation. Although this review focuses on HCM , many of the issues discussed are also relevant to other inherited cardiac diseases.
- Published
- 2020
- Full Text
- View/download PDF
26. Disease-specific variant pathogenicity prediction significantly improves variant interpretation in inherited cardiac conditions
- Author
-
Carolyn Y. Ho, Declan P. O'Regan, Michelle Michels, Magdi H. Yacoub, William Midwinter, Roddy Walsh, Mian Ahmad, Francesco Mazzarotto, Antonio de Marvao, Euan A. Ashley, Risha Govind, Alexandre C. Pereira, John L. Jefferies, Jodie Ingles, Alicja Wilk, Nicola Whiffin, Xiaolei Zhang, Leonardo Bottolo, Nicholas Li, James S. Ware, Angharad M. Roberts, Christopher Semsarian, Iacopo Olivotto, Paul J.R. Barton, Pantazis I. Theotokis, Daniel Jacoby, Mona Allouba, Yasmine Aguib, Stuart A. Cook, Steven D. Colan, Sharlene M. Day, Chee Jian Pua, Erica Mazaika, Rachel Buchan, Gunnar Gunnarsson, Ware, James S. [0000-0002-6110-5880], Apollo - University of Cambridge Repository, Cardiology, Wellcome Trust, Ware, James S [0000-0002-6110-5880], Royal Brompton & Harefield NHS Foundation Trust, Imperial College Healthcare NHS Trust- BRC Funding, and British Heart Foundation
- Subjects
pathogenicity prediction ,Cardiomyopathy ,Disease ,030204 cardiovascular system & hematology ,Bioinformatics ,Genome ,0302 clinical medicine ,Missense mutation ,MUTATION ,Uncertain significance ,Genetics (clinical) ,Brugada syndrome ,Genetics & Heredity ,Disease gene ,0303 health sciences ,Virulence ,Hypertrophic cardiomyopathy ,article ,Middle Aged ,GENOTYPE ,3. Good health ,Area Under Curve ,symbols ,cardiomyopathy ,long QT syndrome ,missense variant interpretation ,Algorithms ,Humans ,Cardiomyopathies ,Mutation, Missense ,Life Sciences & Biomedicine ,Disease specific ,Long QT syndrome ,Computational biology ,Biology ,03 medical and health sciences ,symbols.namesake ,medicine ,030304 developmental biology ,0604 Genetics ,Science & Technology ,business.industry ,1103 Clinical Sciences ,medicine.disease ,Pathogenicity ,Confidence interval ,Mendelian inheritance ,Missense ,business - Abstract
BackgroundAccurate discrimination of benign and pathogenic rare variation remains a priority for clinical genome interpretation. State-of-the-art machine learning tools are useful for genome-wide variant prioritisation but remain imprecise. Since the relationship between molecular consequence and likelihood of pathogenicity varies between genes with distinct molecular mechanisms, we hypothesised that a disease-specific classifier may outperform existing genome-wide tools.MethodsWe present a novel disease-specific variant classification tool, CardioBoost, that estimates the probability of pathogenicity for rare missense variants in inherited cardiomyopathies and arrhythmias, trained with variants of known clinical effect. To benchmark against state-of-the-art genome-wide pathogenicity classification tools, we assessed classification of hold-out test variants using both overall performance metrics, and metrics of high-confidence (>90%) classifications relevant to variant interpretation. We further evaluated the prioritisation of variants associated with disease and patient clinical outcomes, providing validations that are robust to potential mis-classification in gold-standard reference datasets.ResultsCardioBoost has higher discriminating power than published genome-wide variant classification tools in distinguishing between pathogenic and benign variants based on overall classification performance measures with the highest area under the Precision-Recall Curve as 91% for cardiomyopathies and as 96% for inherited arrhythmias. When assessed at high-confidence (>90%) classification thresholds, prediction accuracy is improved by at least 120% over existing tools for both cardiomyopathies and arrhythmias, with significantly improved sensitivity and specificity. Finally, CardioBoost improves prioritisation of variants significantly associated with disease, and stratifies survival of patients with cardiomyopathies, confirming biologically relevant variant classification.ConclusionsWe demonstrate that a disease-specific variant pathogenicity prediction tool outperforms state-of-the-art genome-wide tools for the classification of rare missense variants of uncertain significance for inherited cardiac conditions. To facilitate evaluation of CardioBoost, we provide pre-computed pathogenicity scores for all possible rare missense variants in genes associated with cardiomyopathies and arrhythmias (https://www.cardiodb.org/cardioboost/). Our results also highlight the need to develop and evaluate variant classification tools focused on specific diseases and clinical application contexts. Our proposed model for assessing variants in known disease genes, and the use of application-specific evaluations, is broadly applicable to improve variant interpretation across a wide range of Mendelian diseases.
- Published
- 2020
- Full Text
- View/download PDF
27. Advantages and Perils of Clinical Whole-Exome and Whole-Genome Sequencing in Cardiomyopathy
- Author
-
Roddy Walsh, Francesco Mazzarotto, Iacopo Olivotto, and Cardiology
- Subjects
0301 basic medicine ,Cardiac & Cardiovascular Systems ,MEDICAL GENETICS ,Disease ,030204 cardiovascular system & hematology ,VARIANTS ,FAMILIAL HYPERTROPHIC CARDIOMYOPATHY ,DISEASE ,0302 clinical medicine ,Pharmacology (medical) ,Pharmacology & Pharmacy ,Exome ,Exome sequencing ,medicine.diagnostic_test ,High-Throughput Nucleotide Sequencing ,General Medicine ,Cardiomyopathy ,Clinical genetic testing ,Whole-exome sequencing ,Whole-genome sequencing ,3. Good health ,Phenotype ,Molecular Diagnostic Techniques ,Medical genetics ,MYOSIN ,1115 Pharmacology and Pharmaceutical Sciences ,Cardiology and Cardiovascular Medicine ,Cardiomyopathies ,Life Sciences & Biomedicine ,medicine.medical_specialty ,Context (language use) ,Computational biology ,AMERICAN-COLLEGE ,03 medical and health sciences ,Predictive Value of Tests ,Exome Sequencing ,medicine ,Animals ,Humans ,Genetic Predisposition to Disease ,Genetic testing ,Pharmacology ,Whole genome sequencing ,Science & Technology ,Whole Genome Sequencing ,business.industry ,MUTATIONS ,Genetic Variation ,Reproducibility of Results ,Genetic architecture ,HUMAN HEART ,HIGH-RISK ,030104 developmental biology ,Cardiovascular System & Hematology ,Cardiovascular System & Cardiology ,business ,SUDDEN CARDIAC DEATH - Abstract
As the price of next-generation sequencing keeps decreasing, cost is becoming a less important discriminator for diagnostic laboratories in choosing the preferred type of approach to genetic testing. Genome-wide sequencing strategies will plausibly become the standard first-tier tools for genetic testing, with the potential for deeper understanding of the genetic architecture of cardiomyopathies and discovery of the underlying aetiology in the many patients in whom the genetic cause remains elusive. Routine usage of extended sequencing assays will also enable “genetic-first diagnostics”, particularly for those patients affected with syndromic conditions of unclear genetic origin, often resulting in costly and distressing diagnostic odysseys before reaching a diagnosis. However, access to genome-wide data for all patients will need to be managed with rigour and caution by (cardiovascular) genetic professionals to avoid erroneous variant pathogenicity assertions and over-reporting uncertain findings, both damaging scenarios to patients and their family members. Researchers will also be required to adopt robust methods to demonstrate novel genetic associations with disease, given the high “narrative potential” of such large datasets and the dangers of generating further false positive associations (that have previously blighted the field of cardiac genetics). Here, we discuss advantages and dangers associated with the routine adoption of whole-exome (and whole-genome) sequencing in diagnostic facilities and in the research setting in the context of cardiomyopathies but relevant to several other conditions.
- Published
- 2020
28. The genetic architecture of left ventricular non-compaction reveals both substantial overlap with other cardiomyopathies and a distinct aetiology in a subset of cases
- Author
-
Mona Allouba, Yasmine Aguib, Stuart A. Cook, Francesco Mazzarotto, Sanjay K Prasad, Antonis Pantazis, Soha Romeih, Roddy Walsh, Ben Statton, Magdi H. Yacoub, Leander Beekman, Antonio de Marvao, Kathryn A. McGurk, Elisabetta Cerbai, Connie R. Bezzina, Beatrice Boschi, Paul J.R. Barton, Elisabeth M. Lodder, James S. Ware, Megan H. Hawley, Angharad M. Roberts, Declan P. O'Regan, Birgit Funke, Iacopo Olivotto, Francesca Girolami, A. John Baksi, and Matteo Beltrami
- Subjects
Genetics ,0303 health sciences ,education.field_of_study ,medicine.diagnostic_test ,Population ,Hypertrophic cardiomyopathy ,Dilated cardiomyopathy ,Disease ,030204 cardiovascular system & hematology ,Biology ,medicine.disease ,Genetic architecture ,03 medical and health sciences ,0302 clinical medicine ,medicine ,cardiovascular system ,Missense mutation ,MYH7 ,cardiovascular diseases ,education ,030304 developmental biology ,Genetic testing - Abstract
BackgroundLeft ventricular non-compaction (LVNC) is a condition characterised by trabeculations in the myocardial wall and is the subject of considerable conjecture as to whether it represents a distinct pathology or a secondary phenotype associated with other cardiac diseases, particularly cardiomyopathies. We sought to investigate the genetic architecture of LVNC by identifying genes and variant classes robustly associated with disease and comparing these to other genetically characterised cardiomyopathies.MethodsWe performed rare variant association analysis using six different LVNC cohorts comprising 840 cases together with 125,748 gnomAD population controls and compared results to similar analyses with dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM) cases.ResultsWe observed substantial overlap in genes and variant classes enriched in LVNC and DCM/HCM, indicating that in many cases LVNC belongs to a spectrum of more established cardiomyopathies, with non-compaction representing a phenotypic variation in patients with DCM- or HCM-causing variants. In contrast, five variant classes were uniquely enriched in LVNC cases, of which truncating variants inMYH7, ACTN2andPRDM16may represent a distinct LVNC aetiology.MYH7truncating variants are generally considered as non-pathogenic but were detected in 2% of LVNC cases compared to 0.1% of controls, including a cluster of variants around a single splice region. Individuals withMYH7truncating variants identified in the UK Biobank and cohorts of healthy volunteers also displayed significantly greater non-compaction compared to matched controls, with 50% meeting the diagnostic criteria for LVNC. Additionally, structural variants (exon deletions) inRYR2and missense variants in the transmembrane region ofHCN4were enriched in LVNC cases, confirming prior reports regarding the association of these variant classes with combined LVNC and arrhythmia phenotypes.ConclusionsWe demonstrated that genetic association analysis can clarify the relationship between LVNC and established cardiomyopathies, highlighted substantial overlap with DCM/HCM but also identified variant classes associated with distinct LVNC and with joint LVNC/arrhythmia phenotypes. These results underline the complex genetic landscape of LVNC and inform how genetic testing in LVNC cases should be pursued and interpreted.
- Published
- 2020
- Full Text
- View/download PDF
29. Temporal Trend of Age at Diagnosis in Hypertrophic Cardiomyopathy: An Analysis of the International Sarcomeric Human Cardiomyopathy Registry
- Author
-
Marco Canepa, Carlo Fumagalli, Daniel Jacoby, Justin Vincent-Tompkins, Sharlene M. Day, Francesco Mazzarotto, Euan A. Ashley, Michelle Michels, Carolyn Y. Ho, Giacomo Tini, Iacopo Olivotto, James S. Ware, and Cardiology
- Subjects
Adult ,Male ,medicine.medical_specialty ,Internationality ,phenotype ,genotype ,prevalence ,Cardiomyopathy ,Age at diagnosis ,heart failure ,hypertrophic ,030204 cardiovascular system & hematology ,Diagnostic tools ,03 medical and health sciences ,Sex Factors ,0302 clinical medicine ,Internal medicine ,Epidemiology ,medicine ,Humans ,Genetic Testing ,Registries ,030212 general & internal medicine ,cardiomyopathy ,cardiomyopathy, hypertrophic ,Aged ,Retrospective Studies ,Aged, 80 and over ,business.industry ,Age Factors ,Hypertrophic cardiomyopathy ,Original Articles ,Middle Aged ,medicine.disease ,United States ,3. Good health ,Heart failure ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background: Over the last 50 years, the epidemiology of hypertrophic cardiomyopathy (HCM) has changed because of increased awareness and availability of advanced diagnostic tools. We aim to describe the temporal trends in age, sex, and clinical characteristics at HCM diagnosis over >4 decades. Methods: We retrospectively analyzed records from the ongoing multinational Sarcomeric Human Cardiomyopathy Registry. Overall, 7286 patients with HCM diagnosed at an age ≥18 years between 1961 and 2019 were included in the analysis and divided into 3 eras of diagnosis (2010). Results: Age at diagnosis increased markedly over time (40±14 versus 47±15 versus 51±16 years, P P P P 30 mm Hg: 31.9% versus 39.3% versus 39.0%, P =0.001). Consistent with decreasing phenotypic severity, yield of pathogenic/likely pathogenic variants at genetic testing decreased over time (57.7% versus 45.6% versus 38.4%, P Conclusions: Evolving HCM populations include progressively greater representation of older patients with sporadic disease, mild phenotypes, and genotype-negative status. Such trend suggests a prominent role of imaging over genetic testing in promoting HCM diagnoses and urges efforts to understand genotype-negative disease eluding the classic monogenic paradigm.
- Published
- 2020
30. Comparison of long-term outcome in anthracycline-related versus idiopathic dilated cardiomyopathy: a single centre experience
- Author
-
Cecilia Ferrantini, Francesco Mazzarotto, Alessandra Fornaro, Niccolò Marchionni, Benedetta Tomberli, Ciaccheri M, Iacopo Olivotto, Luigi Rigacci, Massimo Milli, Raffaele Coppini, Gabriele Castelli, Francesca Girolami, and Marco Chiostri
- Subjects
Heart transplantation ,medicine.medical_specialty ,Ejection fraction ,business.industry ,medicine.medical_treatment ,Hazard ratio ,Cardiomyopathy ,030204 cardiovascular system & hematology ,medicine.disease ,Confidence interval ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Heart failure ,Internal medicine ,Cohort ,Idiopathic dilated cardiomyopathy ,medicine ,Cardiology ,Cardiology and Cardiovascular Medicine ,business - Abstract
AIMS Cardiac dysfunction is a severe complication of anthracycline-containing anticancer therapy. The outcome of anthracycline-induced cardiomyopathy (AICM) compared with other non-ischaemic causes of heart failure (HF), such as idiopathic dilated cardiomyopathy (IDCM), is unresolved. The aim of this study was to compare the survival of AICM patients with an IDCM cohort followed at our centre from 1990 to 2016. METHODS AND RESULTS We included 67 patients (67% female, 50 ± 15 years) with AICM, defined as onset of otherwise unexplained left ventricular ejection fraction (LVEF) ≤50% following anthracycline therapy, and 488 IDCM patients (28% female, 55 ± 12 years). Patients were followed with constantly optimized HF therapy, for 7.6 ± 5.5 and 8.1 ± 5.5 years, respectively. In both cohorts, 25% of patients reached the combined endpoint of death/heart transplantation. Overall survival rates at 5 and 10 years were similar (AICM: 86% and 61%, IDCM: 88% and 75%; P = 0.61), and so was cardiovascular survival (AICM: 91% and 76%, IDCM: 91% and 80%; P = 0.373), also after 1:1 propensity matching (P = 0.27) and adjusting for age, LVEF and left ventricular size. A trend toward higher all-cause mortality was present in AICM patients [hazard ratio (HR) 1.67, 95% confidence interval (CI) 0.95-2.92, P = 0.076]. No differences were observed between AICM and IDCM with regard to pharmacological HF therapy, but AICM patients were less likely to receive devices (13% vs. 41.8% in IDCM, P
- Published
- 2017
- Full Text
- View/download PDF
31. P2731Genetic ancestry analysis of the Italian founder population carrying the cardiac amyloidosis-causing variant Val122Ile in the transthyretin gene
- Author
-
Raffaele Martone, C. Di Mario, Renato Polimanti, Simone Bartolini, Giulia Taborchi, Joel N. Buxbaum, Elisabetta Pelo, Francesco Mazzarotto, I Olivotto, Sofia Morini, Federico Perfetto, Elisa Contini, Sabrina Frusconi, and Francesco Cappelli
- Subjects
Proband ,Genetics ,Amyloid ,biology ,business.industry ,Amyloidosis ,Single-nucleotide polymorphism ,medicine.disease ,Transthyretin ,Cardiac amyloidosis ,medicine ,biology.protein ,Cardiology and Cardiovascular Medicine ,business ,Allele frequency ,Founder effect - Abstract
Background Transthyretin amyloidosis is a life-threatening disorder caused by the deposition of TTR amyloid in various tissues and organs. The most common worldwide pathogenic variant with almost exclusive cardiac involvement is Val122Ile (rs76992529), with an allele frequency of 3.5% in the U.S. African-American population, but rare in Caucasians. Unexpectedly, we identified 23 Caucasian individuals with Val122Ile in our amyloidosis referral center (9 affected patients, 14 carriers), belonging to 9 unrelated families. Purpose To determine the ancestral origin of the Tuscan founder population of TTR Val122Ile carriers. Methods A total of 24 individuals were included in the analysis (our 23 probands and relatives from Val122Ile families and the Caucasian reference sample NA10851 (CEU – Utah resident with European ancestry). All samples were genotyped using the EUROFORGEN Global AIM-SNP array1, inclusive of 127 highly informative SNPs to infer genetic ancestry. We have performed a principal component analysis (PCA) of the 9 unrelated probands and NA10851, compared with the Phase 3 of the 1000 Genomes Project data, comprising 2504 unrelated individuals from >20 distinct populations.(Figure 1). Results As shown in Figure 1, all our samples but one (from Argentina) cluster very close to the super-cluster of European populations, and distant from the populations of African ancestry. The proband from Argentina and the Caucasian reference sample NA10851 cluster close to Mexicans and Peruvians, and the super-cluster of European populations, respectively, confirming the robustness of the analysis. Conclusion Based on this result, we can confidently conclude that our samples from Tuscan families in which the TTR Val122Ile variant segregates are of ancestral European origin, with no mixed African ancestry, implying that the same variant originated in Africans and Europeans independently and not as result of genetic admixture. These findings suggest the presence of a mutational hot spots in TTR, with potential impact on the epidemiology of amyloidosis worldwide. Acknowledgement/Funding The present study was supported by an Investigator-Initiated Research to Azienda Ospedaliero Universitaria Careggi from Pfizer Srl.
- Published
- 2019
- Full Text
- View/download PDF
32. 4258Re-evaluating the genetic contribution of monogenic dilated cardiomyopathy
- Author
-
S Prasad, Francesco Mazzarotto, B Funke, Nicola Whiffin, Rachel Buchan, I Olivotto, Alicja Wilk, William Midwinter, Roddy Walsh, Paul J.R. Barton, Declan P. O'Regan, Sarah Cook, J S Ware, H Watkins, and Upasana Tayal
- Subjects
medicine.medical_specialty ,business.industry ,Internal medicine ,Cardiology ,medicine ,Dilated cardiomyopathy ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business - Abstract
Background Dilated cardiomyopathy (DCM) is genetically heterogeneous, with >100 purported disease genes tested in clinical laboratories. However, many genes were originally identified based on candidate-gene studies that did not adequately account for background population variation. Here we define the frequency of rare variation in 2538 DCM patients across protein-coding regions of 56 commonly tested genes and compare this to both 912 confirmed healthy controls and a reference population of 60,706 individuals. Purpose To identify clinically interpretable genes robustly associated with dominant monogenic DCM. Methods We used the TruSight Cardio sequencing panel to evaluate the burden of rare variants in 56 putative DCM genes in 1040 DCM patients and 912 healthy volunteers processed with identical sequencing and bioinformatics pipelines. We further aggregated data from 1498 DCM patients sequenced in diagnostic laboratories and the ExAC database for replication and meta-analysis. Results Specific variant classes in TTN, DSP, MYH7 and LMNA were associated with DCM in all comparisons. Variants in BAG3, TNNT2, TPM1, NEXN and VCL were significantly enriched specific patient subsets, with the last 3 genes likely contributing primarily to early-onset forms of DCM. Overall, rare variants in these 9 genes potentially explained 19–26% of cases. Whilst the absence of a significant excess in other genes cannot preclude a role in disease, such genes have limited diagnostic value since novel variants will be uninterpretable and therefore non-actionable, and their diagnostic yield is minimal. Conclusion In the largest sequenced DCM cohort yet described, we observe robust disease association only with a limited number of genes, highlighting their importance in DCM and translating into high interpretability in diagnostic testing. The other genes evaluated have limited value in diagnostic testing in DCM. This data will contribute to community gene curation efforts, and will reduce erroneous and inconclusive findings in diagnostic testing. Acknowledgement/Funding Wellcome Trust (107469/Z/15/Z), BHF (SP/10/10/28431), MRC (MR/M003191/1), Fondation Leducq (11-CVD01), Italian Ministry of Health (RF-2013-02356787)
- Published
- 2019
- Full Text
- View/download PDF
33. P1243Comparison of long-term clinical course and outcome of MYBPC3 - versus MYH7 - related hypertrophic cardiomyopathy
- Author
-
Elisa Fedele, Matteo Beltrami, Corrado Poggesi, Francesca Cecchi, Francesco Mazzarotto, Carlo Fumagalli, Raffaele Coppini, Niccolò Maurizi, Silvia Passantino, Katia Baldini, Mattia Targetti, A Arretini, Cecilia Ferrantini, Alessia Tomberli, and I Olivotto
- Subjects
Pediatrics ,medicine.medical_specialty ,business.industry ,Hypertrophic cardiomyopathy ,medicine ,Clinical course ,MYH7 ,Cardiology and Cardiovascular Medicine ,medicine.disease ,business ,Outcome (game theory) ,Term (time) - Abstract
Introduction The presence of sarcomere mutations is a powerful predictor of heart failure-related outcomes in Hypertrophic Cardiomyopathy (HCM). However, whether the prevalence of left ventricular (LV) dysfunction differs in patients with mutations in the two most prevalent HCM-associated genes (i.e. MYBPC3 and MYH7) is unclear. Purpose To ascertain lifetime trends in prevalence of LV dysfunction in HCM associated with pathogenic or likely-pathogenic MYBPC3 versus MYH7 mutations. Methods Clinical and instrumental records of 402 HCM patients with MYBPC3 (N=251) or MYH7 (N=151) mutations were retrospectively reviewed. Presence of systolic dysfunction (ejection fraction [EF] Results Patients were diagnosed at a mean age of 39±17 years and 63% were men. At first evaluation MYBPC3-HCM patients were less frequently obstructive (15% vs 26% in MYH7; p=0.005) and had lower LVEF (61±11% vs 64±9%; p=0.01). Prevalence of diastolic dysfunction increased with age and was lowest in MYBPC3 patients No significant differences were observed in terms of NYHA class change, atrial fibrillation, stroke, heart failure, appropriate ICD intervention or cardiovascular death. However, prevalence of NSVT was higher for MYBPC3 (39% vs 14% in MYH7, p In vitro cross-sectional evaluation of myocardial samples taken during septal myectomy at different ages showed a decline in contraction-relaxation properties after age 40 in MYPBC3 carriers, but preserved function in MYH7 patients (Figure). Kinetic of myosin cross-bridges Conclusions In HCM patients, mutations in the MYBPC3 gene and early diagnosis are associated with slowly progressing systolic impairment leading to overt dysfunction in 15% compared to 5% in MYH7-HCM. However, outcome was similar in the two subsets. These differences in lifetime myocardial performance between the two most common HCM-associated genes suggest diverse pathways of disease progression, potentially amenable to requiring different molecular approaches.
- Published
- 2019
- Full Text
- View/download PDF
34. 121 Re-evaluating the genetic contribution of monogenic dilated cardiomyopathy
- Author
-
Francesco Mazzarotto, Paz Tayal, Rachel Buchan, William Midwinter, Alicja Wilk, Nicola Whiffin, Risha Govind, Erica Mazaika, Antonio De Marvao, Leanne Felkin, Timothy Dawes, Mian Ahmad, Elizabeth Edwards, Alexander Ing, Kate Thomson, Laura Chan, David Sim, John Baksi, Antonis Pantazis, Angharad Roberts, Hugh Watkins, Birgit Funke, Declan O’Regan, Iacopo Olivotto, Paul Barton, Sanjay Prasad, Stuart Cook, James Ware, and Roddy Walsh
- Published
- 2019
- Full Text
- View/download PDF
35. Sex-related differences in exercise performance and outcome of patients with hypertrophic cardiomyopathy
- Author
-
A Arretini, Francesco Mazzarotto, Franco Cecchi, Silvia Passantino, Andrea Oddo, Francesco Pieri, Flavio Ribichini, Carlo Fumagalli, Luca Ghiselli, Martin S. Maron, Mattia Targetti, Mariantonietta Cicoira, Niccolò Maurizi, Alberto Marchi, Iacopo Olivotto, Ethan J. Rowin, Francesca Girolami, Niccolò Marchionni, and Fabio Mori
- Subjects
Male ,medicine.medical_specialty ,Functional impairment ,Epidemiology ,exercise performance ,Outcome (game theory) ,Internal medicine ,Exercise performance ,Stress Echocardiography ,Humans ,Medicine ,sex ,Retrospective Studies ,exercise echocardiography ,business.industry ,Hypertrophic cardiomyopathy ,Sex related ,Cardiomyopathy, Hypertrophic ,medicine.disease ,Exercise echocardiography ,Defibrillators, Implantable ,Death, Sudden, Cardiac ,outcome ,Echocardiography ,Cardiology ,Female ,Cardiology and Cardiovascular Medicine ,business - Abstract
Aims Exercise performance is known to predict outcome in hypertrophic cardiomyopathy (HCM), but whether sex-related differences exist is unresolved. We explored whether functional impairment, assessed by exercise echocardiography, has comparable predictive accuracy in females and males with HCM. Methods We retrospectively evaluated 292 HCM patients (46 ± 16 years, 72% males), consecutively referred for exercise echocardiography; 242 were followed for 5.9 ± 4.2 years. Results Peak exercise capacity was 6.5 ± 1.6 metabolic equivalents (METs). Sixty patients (21%) showed impaired exercise capacity (≤5 METs). Exercise performance was reduced in females, compared with males (5.6 ± 1.6 vs 6.9 ± 1.5 METs, p 50 years of age. At multivariable analysis, female sex was independently associated with impaired exercise capacity (odds ratio: 4.67; 95% confidence interval (CI): 1.83–11.90; p = 0.001). During follow-up, 24 patients (10%) met the primary endpoint (a combination of cardiac death, heart failure requiring hospitalization, sustained ventricular tachycardia, appropriate implantable cardioverter defibrillator discharge, resuscitated sudden cardiac death and cardioembolic stroke). Event-free survival was reduced in females (p = 0.035 vs males). Peak METs were inversely related to outcome in males (hazard ratio (HR) per unit increase: 0.57; 95% CI: 0.39–0.84; p = 0.004) but not in females (HR: 1.22; 95% CI: 0.66–2.24; p = 0.53). Conclusions Female patients with HCM showed significant age-related impairment in functional capacity compared with males, particularly evident in post-menopausal age groups. While women were at greater risk of HCM-related complications and death, impaired exercise capacity predicted adverse outcome only in men. These findings suggest the need for sex-specific management strategies in HCM.
- Published
- 2019
36. Quantitative approaches to variant classification increase the yield and precision of genetic testing in Mendelian diseases: the case of hypertrophic cardiomyopathy
- Author
-
Steven D. Colan, Rachel Buchan, Daniel Jacoby, Alexandre C. Pereira, Hugh Watkins, Leanne E. Felkin, Michelle Michels, Carolyn Y. Ho, Mian Ahmad, Sharlene M. Day, Mona Allouba, Roddy Walsh, Iacopo Olivotto, Antonio de Marvao, Francesco Mazzarotto, Risha Govind, Stuart A. Cook, Alicja Wilk, Xiaolei Zhang, James S. Ware, Erica Mazaika, Nathan Ingold, Paul J.R. Barton, Nicola Whiffin, Nicholas Li, William Midwinter, Euan A. Ashley, K Thomson, British Heart Foundation, Fondation Leducq, Wellcome Trust, Department of Health, Royal Brompton & Harefield NHS Foundation Trust, Imperial College Healthcare NHS Trust- BRC Funding, and Cardiology
- Subjects
lcsh:QH426-470 ,Cardiomyopathy ,lcsh:Medicine ,ESC ,Computational biology ,Disease ,030204 cardiovascular system & hematology ,Biology ,Mendelian disease ,GUIDELINES ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,medicine ,Humans ,ACMG/AMP guidelines ,Genetic Testing ,Likely pathogenic ,Variant interpretation ,030304 developmental biology ,Genetic testing ,Genetics & Heredity ,0303 health sciences ,0604 Genetics ,Science & Technology ,medicine.diagnostic_test ,VENTRICULAR-ARRHYTHMIAS ,Research ,lcsh:R ,Hypertrophic cardiomyopathy ,1103 Clinical Sciences ,Cardiomyopathy, Hypertrophic ,Functional prediction ,medicine.disease ,3. Good health ,lcsh:Genetics ,Hypertrophic ,ACMG ,Mendelian genetics ,Practice Guidelines as Topic ,Mutation ,Mendelian inheritance ,symbols ,AMP guidelines ,False positive rate ,Life Sciences & Biomedicine ,PATHOGENICITY - Abstract
Background International guidelines for variant interpretation in Mendelian disease set stringent criteria to report a variant as (likely) pathogenic, prioritising control of false-positive rate over test sensitivity and diagnostic yield. Genetic testing is also more likely informative in individuals with well-characterised variants from extensively studied European-ancestry populations. Inherited cardiomyopathies are relatively common Mendelian diseases that allow empirical calibration and assessment of this framework. Methods We compared rare variants in large hypertrophic cardiomyopathy (HCM) cohorts (up to 6179 cases) to reference populations to identify variant classes with high prior likelihoods of pathogenicity, as defined by etiological fraction (EF). We analysed the distribution of variants using a bespoke unsupervised clustering algorithm to identify gene regions in which variants are significantly clustered in cases. Results Analysis of variant distribution identified regions in which variants are significantly enriched in cases and variant location was a better discriminator of pathogenicity than generic computational functional prediction algorithms. Non-truncating variant classes with an EF ≥ 0.95 were identified in five established HCM genes. Applying this approach leads to an estimated 14–20% increase in cases with actionable HCM variants, i.e. variants classified as pathogenic/likely pathogenic that might be used for predictive testing in probands’ relatives. Conclusions When found in a patient confirmed to have disease, novel variants in some genes and regions are empirically shown to have a sufficiently high probability of pathogenicity to support a “likely pathogenic” classification, even without additional segregation or functional data. This could increase the yield of high confidence actionable variants, consistent with the framework and recommendations of current guidelines. The techniques outlined offer a consistent and unbiased approach to variant interpretation for Mendelian disease genetic testing. We propose adaptations to ACMG/AMP guidelines to incorporate such evidence in a quantitative and transparent manner. Electronic supplementary material The online version of this article (10.1186/s13073-019-0616-z) contains supplementary material, which is available to authorized users.
- Published
- 2019
37. Defining the diagnostic effectiveness of genes for inclusion in panels: the experience of two decades of genetic testing for hypertrophic cardiomyopathy at a single center
- Author
-
Francesco Mazzarotto, Franco Cecchi, Elisa Contini, Roddy Walsh, Stuart A. Cook, Raffaele Coppini, Katia Baldini, Ilaria Tanini, Alessia Tomberli, Fausto Barlocco, Iacopo Olivotto, Francesca Girolami, Sara Bardi, Corrado Poggesi, Elisabetta Cerbai, Beatrice Boschi, Francesca Torricelli, Elisabetta Pelo, and British Heart Foundation
- Subjects
Male ,0301 basic medicine ,Proband ,Routine testing ,VARIANTS ,030204 cardiovascular system & hematology ,GUIDELINES ,DISEASE ,Cohort Studies ,0302 clinical medicine ,Medicine ,Genetics (clinical) ,Genetics & Heredity ,Sanger sequencing ,medicine.diagnostic_test ,Hypertrophic cardiomyopathy ,High-Throughput Nucleotide Sequencing ,Middle Aged ,Mendelian HCM genetics ,Patient management ,NGS ,cardiovascular system ,symbols ,Female ,Life Sciences & Biomedicine ,Adult ,Sarcomeres ,Diagnostic effectiveness ,HCM mimics ,ESC ,macromolecular substances ,Computational biology ,Article ,Young Adult ,03 medical and health sciences ,symbols.namesake ,Humans ,Genetic Testing ,cardiovascular diseases ,Gene ,Aged ,Genetic testing ,0604 Genetics ,Science & Technology ,MUTATIONS ,business.industry ,Reproducibility of Results ,1103 Clinical Sciences ,Cardiomyopathy, Hypertrophic ,medicine.disease ,030104 developmental biology ,Mendelian inheritance ,business - Abstract
Purpose Genetic testing in hypertrophic cardiomyopathy (HCM) has long relied on Sanger sequencing of sarcomeric genes. The advent of next-generation sequencing (NGS) has catalyzed routine testing of additional genes of dubious HCM-causing potential. We used 19 years of genetic testing results to define a reliable set of genes implicated in Mendelian HCM and assess the value of expanded NGS panels. Methods We dissected genetic testing results from 1,198 single-center HCM probands and devised a widely applicable score to identify which genes yield effective results in the diagnostic setting. Results Compared with early panels targeting only fully validated sarcomeric HCM genes, expanded NGS panels allow the prompt recognition of probands with HCM-mimicking diseases. Scoring by “diagnostic effectiveness” highlighted that PLN should also be routinely screened besides historically validated genes for HCM and its mimics. Conclusion The additive value of expanded panels in HCM genetic testing lies in the systematic screening of genes associated with HCM mimics, requiring different patient management. Only variants in a limited set of genes are highly actionable and interpretable in the clinic, suggesting that larger panels offer limited additional sensitivity. A score estimating the relative effectiveness of a given gene’s inclusion in diagnostic panels is proposed.
- Published
- 2019
38. A gene-centric strategy for identifying disease-causing rare variants in dilated cardiomyopathy
- Author
-
Anne Keogh, Christopher S. Hayward, Claire Horvat, Paul J.R. Barton, Barbara McDonough, Alireza Haghighi, Jonathan G. Seidman, Leanne E. Felkin, Diane Fatkin, Francesco Mazzarotto, Peter S. Macdonald, Michael Parfenov, Christine E. Seidman, Steven R. DePalma, Daniel S. Herman, Angharad M. Roberts, Eleni Giannoulatou, Jacob E Munro, Amy E. Roberts, Stuart A. Cook, Renee Johnson, Lien Lam, British Heart Foundation, Fondation Leducq, and Royal Brompton & Harefield NHS Foundation Trust
- Subjects
0301 basic medicine ,Proband ,Cardiomyopathy, Dilated ,Male ,Dilated cardiomyopathy ,Cardiomyopathy ,Mutation, Missense ,Disease ,TITIN ,030204 cardiovascular system & hematology ,Biology ,Article ,03 medical and health sciences ,pathogenic variant ,0302 clinical medicine ,Rare Diseases ,medicine ,SCN5A MUTATION ,Missense mutation ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Gene ,Genetics (clinical) ,Genetic testing ,SERVER ,Genetics ,Genetics & Heredity ,0604 Genetics ,Science & Technology ,medicine.diagnostic_test ,BAG3 ,High-Throughput Nucleotide Sequencing ,1103 Clinical Sciences ,ASSOCIATION ,Middle Aged ,medicine.disease ,SODIUM-CHANNEL NA(V)1.5 ,Human genetics ,Pedigree ,pathogenic variant Genetic testing ,030104 developmental biology ,Next-generation sequencing ,HEART-FAILURE ,Female ,Life Sciences & Biomedicine - Abstract
© 2018, American College of Medical Genetics and Genomics. Purpose: We evaluated strategies for identifying disease-causing variants in genetic testing for dilated cardiomyopathy (DCM). Methods: Cardiomyopathy gene panel testing was performed in 532 DCM patients and 527 healthy control subjects. Rare variants in 41 genes were stratified using variant-level and gene-level characteristics. Results: A majority of DCM cases and controls carried rare protein-altering cardiomyopathy gene variants. Variant-level characteristics alone had limited discriminative value. Differentiation between groups was substantially improved by addition of gene-level information that incorporated ranking of genes based on literature evidence for disease association. The odds of DCM were increased to nearly 9-fold for truncating variants or high-impact missense variants in the subset of 14 genes that had the strongest biological links to DCM (P
- Published
- 2018
39. Comparison of long-term outcome in anthracycline-related versus idiopathic dilated cardiomyopathy: a single centre experience
- Author
-
Alessandra, Fornaro, Iacopo, Olivotto, Luigi, Rigacci, Mauro, Ciaccheri, Benedetta, Tomberli, Cecilia, Ferrantini, Raffaele, Coppini, Francesca, Girolami, Francesco, Mazzarotto, Marco, Chiostri, Massimo, Milli, Niccolò, Marchionni, and Gabriele, Castelli
- Subjects
Cardiomyopathy, Dilated ,Male ,Heart Ventricles ,Incidence ,Stroke Volume ,Middle Aged ,Severity of Illness Index ,Ventricular Function, Left ,Survival Rate ,Italy ,Echocardiography ,Risk Factors ,Disease Progression ,Humans ,Anthracyclines ,Female ,Follow-Up Studies ,Forecasting ,Retrospective Studies - Abstract
Cardiac dysfunction is a severe complication of anthracycline-containing anticancer therapy. The outcome of anthracycline-induced cardiomyopathy (AICM) compared with other non-ischaemic causes of heart failure (HF), such as idiopathic dilated cardiomyopathy (IDCM), is unresolved. The aim of this study was to compare the survival of AICM patients with an IDCM cohort followed at our centre from 1990 to 2016.We included 67 patients (67% female, 50 ± 15 years) with AICM, defined as onset of otherwise unexplained left ventricular ejection fraction (LVEF) ≤50% following anthracycline therapy, and 488 IDCM patients (28% female, 55 ± 12 years). Patients were followed with constantly optimized HF therapy, for 7.6 ± 5.5 and 8.1 ± 5.5 years, respectively. In both cohorts, 25% of patients reached the combined endpoint of death/heart transplantation. Overall survival rates at 5 and 10 years were similar (AICM: 86% and 61%, IDCM: 88% and 75%; P = 0.61), and so was cardiovascular survival (AICM: 91% and 76%, IDCM: 91% and 80%; P = 0.373), also after 1:1 propensity matching (P = 0.27) and adjusting for age, LVEF and left ventricular size. A trend toward higher all-cause mortality was present in AICM patients [hazard ratio (HR) 1.67, 95% confidence interval (CI) 0.95-2.92, P = 0.076]. No differences were observed between AICM and IDCM with regard to pharmacological HF therapy, but AICM patients were less likely to receive devices (13% vs. 41.8% in IDCM, P 0.001).Cardiovascular mortality in patients with AICM did not differ from that of a matched IDCM cohort, despite cancer-related morbidity and less prevalent use of devices. These data suggest that patients with AICM should be treated with appropriate guideline-directed medical therapies similar to other non-ischaemic dilated cardiomyopathies.
- Published
- 2017
40. Reassessment of Mendelian gene pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference samples
- Author
-
Karen McGuire, Edward Blair, K Thomson, James S. Ware, Hugh Watkins, Daniel G. MacArthur, Roddy Walsh, Stuart A. Cook, Jessica Woodley, Francesco Mazzarotto, Jenny C. Taylor, Eric Vallabh Minikel, Anneke Seller, Martin Farrall, and Birgit Funke
- Subjects
0303 health sciences ,Cardiomyopathy ,Computational biology ,030204 cardiovascular system & hematology ,Biology ,Pathogenicity ,medicine.disease ,Mendelian disease ,03 medical and health sciences ,symbols.namesake ,0302 clinical medicine ,Variation (linguistics) ,Data sequences ,Genetic variation ,Mendelian inheritance ,symbols ,medicine ,Gene ,030304 developmental biology - Abstract
The accurate interpretation of variation in Mendelian disease genes has lagged behind data generation as sequencing has become increasingly accessible. Ongoing large sequencing efforts present huge interpretive challenges, but also provide an invaluable opportunity to characterize the spectrum and importance of rare variation. Here we analyze sequence data from 7,855 clinical cardiomyopathy cases and 60,706 ExAC reference samples to better understand genetic variation in a representative autosomal dominant disorder. We show that in some genes previously reported as important causes of a given cardiomyopathy, rare variation is not clinically informative and there is a high likelihood of false positive interpretation. By contrast, in other genes, we find that diagnostic laboratories may be overly conservative when assessing variant pathogenicity. We outline improved interpretation approaches for specific genes and variant classes and propose that these will increase the clinical utility of testing across a range of Mendelian diseases.
- Published
- 2017
41. Two decades of genetic testing in hypertrophic cardiomyopathy in a single center: The additive value of extended next-generation sequencing panels lies in the early diagnosis of metabolic mimics
- Author
-
Raffaele Coppini, Fausto Barlocco, Francesco Mazzarotto, Franco Cecchi, Francesca Girolami, Katia Baldini, Roddy Walsh, Iacopo Olivotto, Alessia Tomberli, Beatrice Boschi, and Elisabetta Pelo
- Subjects
Pharmacology ,medicine.diagnostic_test ,Physiology ,business.industry ,Hypertrophic cardiomyopathy ,Computational biology ,030204 cardiovascular system & hematology ,medicine.disease ,Single Center ,DNA sequencing ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Molecular Medicine ,030212 general & internal medicine ,business ,Value (mathematics) ,Genetic testing - Published
- 2018
- Full Text
- View/download PDF
42. Integrated allelic, transcriptional, and phenomic dissection of the cardiac effects of titin truncations in health and disease
- Author
-
Anne Keogh, Sanjay K Prasad, Jason Flannick, Diane Fatkin, Magdi H. Yacoub, Antonio de Marvao, David Altshuler, Francesco Mazzarotto, Peter S. Macdonald, Daniel S. Herman, Jonathan G. Seidman, Sebastian Schafer, Michael Gotthardt, Leanne E. Felkin, Rachel Buchan, Ankur Gulati, Christopher J. O'Donnell, Stuart A. Cook, Michael H. Radke, Jacqueline A. L. MacArthur, Christine E. Seidman, Dudley J. Pennell, James G. Wilson, Christopher S. Hayward, John Baksi, Sungsam Gong, S Wilkinson, James S. Ware, Timothy J W Dawes, Fiona Cunningham, Nicholas R. Banner, Norbert Hubner, Shibu John, Paul J.R. Barton, Stacey Gabriel, Roddy Walsh, Matthias Heinig, Declan P. O'Regan, Tan Ru San, Angharad M. Roberts, Alexander G. Bick, Emma J. Birks, British Heart Foundation, Heart Research UK, and Commission of the European Communities
- Subjects
Transcription, Genetic ,Cardiomyopathy ,FAMILIAL DILATED CARDIOMYOPATHY ,Disease ,Research & Experimental Medicine ,VARIANTS ,Transcriptome ,Cohort Studies ,Protein Isoforms ,Connectin ,10. No inequality ,POPULATION ,11 Medical and Health Sciences ,Genetics ,education.field_of_study ,biology ,Dilated cardiomyopathy ,Heart ,General Medicine ,Exons ,Middle Aged ,Penetrance ,Healthy Volunteers ,3. Good health ,PROGNOSTIC VALUE ,Medicine, Research & Experimental ,CARDIOVASCULAR MAGNETIC-RESONANCE ,HEART-FAILURE ,Titin ,Life Sciences & Biomedicine ,Adult ,Cardiomyopathy, Dilated ,GENES ,Adolescent ,Population ,Immunoglobulins ,Computational biology ,SEQUENCE ,Young Adult ,medicine ,Humans ,Allele ,education ,Alleles ,Aged ,Heart Failure ,Science & Technology ,MUTATIONS ,Genetic Variation ,Cell Biology ,06 Biological Sciences ,medicine.disease ,Mutation ,RISK-FACTORS ,biology.protein - Abstract
The recent discovery of heterozygous human mutations that truncate full-length titin (TTN, an abundant structural, sensory, and signaling filament in muscle) as a common cause of end-stage dilated cardiomyopathy (DCM) promises new prospects for improving heart failure management. However, realization of this opportunity has been hindered by the burden of TTN-truncating variants (TTNtv) in the general population and uncertainty about their consequences in health or disease. To elucidate the effects of TTNtv, we coupled TTN gene sequencing with cardiac phenotyping in 5267 individuals across the spectrum of cardiac physiology and integrated these data with RNA and protein analyses of human heart tissues. We report diversity of TTN isoform expression in the heart, define the relative inclusion of TTN exons in different isoforms (using the TTN transcript annotations available at http://cardiodb.org/titin), and demonstrate that these data, coupled with the position of the TTNtv, provide a robust strategy to discriminate pathogenic from benign TTNtv. We show that TTNtv is the most common genetic cause of DCM in ambulant patients in the community, identify clinically important manifestations of TTNtv-positive DCM, and define the penetrance and outcomes of TTNtv in the general population. By integrating genetic, transcriptome, and protein analyses, we provide evidence for a length-dependent mechanism of disease. These data inform diagnostic criteria and management strategies for TTNtv-positive DCM patients and for TTNtv that are identified as incidental findings.
- Published
- 2015
43. ZBTB17 (MIZ1) Is Important for the Cardiac Stress Response and a Novel Candidate Gene for Cardiomyopathy and Heart Failure
- Author
-
Byambajav Buyandelger, Martin Eilers, Rachel Buchan, Ju Chen, Albrecht Schmidt, Francesco Pesce, Norbert Hubner, Wilhelm Haverkamp, Angharad M. Roberts, Keat-Eng Ng, Peter Nürnberg, Francesco Mazzarotto, Michael D. Schneider, Rivka L. Isaacson, Enrico Petretto, Hendrik Milting, James S. Ware, Niels Teucher, Gerd Hasenfuss, Paul J.R. Barton, Enrique Lara-Pezzi, Conny Pfeiffer, Leanne E. Felkin, Thomas Brand, Martin Teichmann, Ian R. Gould, Heming Wei, Mohammad R. Toliat, Catherine Mansfield, Sylvia Gunkel, Thomas Braun, Sara J. McSweeney, Vera Regitz-Zagrosek, Jesús M. Gómez-Salinero, Declan P. O'Regan, Sawa Kostin, Andreas Perrot, Ewelina M. Krysztofinska, Marina López-Olañeta, Josee Vouffo, Onjee Choi, Sebastian Schafer, Ralph Knöll, Dudley J. Pennell, Sanjay Prasad, Gudrun Knöll, and Kristina M. Ilieva
- Subjects
Candidate gene ,Pathology ,medicine.medical_specialty ,Ubiquitin-Protein Ligases ,Kruppel-Like Transcription Factors ,Cardiomyopathy ,Muscle Proteins ,Locus (genetics) ,030204 cardiovascular system & hematology ,Biology ,Tissue Culture Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Stress, Physiological ,Genetics ,medicine ,Animals ,Humans ,Myocyte ,genetics, animal models ,Gene ,Transcription factor ,Genetics (clinical) ,030304 developmental biology ,Heart Failure ,0303 health sciences ,Cardiac myocyte ,Nuclear Proteins ,Heart ,LIM Domain Proteins ,medicine.disease ,Protein Inhibitors of Activated STAT ,Rats ,3. Good health ,Cell biology ,DNA-Binding Proteins ,Heart failure ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,cardiomyopathy - Abstract
Background— Mutations in sarcomeric and cytoskeletal proteins are a major cause of hereditary cardiomyopathies, but our knowledge remains incomplete as to how the genetic defects execute their effects. Methods and Results— We used cysteine and glycine-rich protein 3, a known cardiomyopathy gene, in a yeast 2-hybrid screen and identified zinc-finger and BTB domain-containing protein 17 ( ZBTB17 ) as a novel interacting partner. ZBTB17 is a transcription factor that contains the peak association signal (rs10927875) at the replicated 1p36 cardiomyopathy locus. ZBTB17 expression protected cardiac myocytes from apoptosis in vitro and in a mouse model with cardiac myocyte–specific deletion of Zbtb17 , which develops cardiomyopathy and fibrosis after biomechanical stress. ZBTB17 also regulated cardiac myocyte hypertrophy in vitro and in vivo in a calcineurin-dependent manner. Conclusions— We revealed new functions for ZBTB17 in the heart, a transcription factor that may play a role as a novel cardiomyopathy gene.
- Published
- 2015
44. 142 Effects of Truncating Variants in Titin on Cardiac Phenotype and Left Ventricular Remodelling in Dilated Cardiomyopathy
- Author
-
Rachel Buchan, Nicola Whiffin, Francesco Mazzarotto, Upasana Tayal, Stuart A. Cook, Simon Newsome, Roddy Walsh, James S. Ware, and Sanjay K Prasad
- Subjects
medicine.medical_specialty ,Ejection fraction ,biology ,business.industry ,Dilated cardiomyopathy ,Stroke volume ,medicine.disease ,Surgery ,Internal medicine ,cardiovascular system ,medicine ,Cardiology ,Left ventricular Stroke volume ,biology.protein ,End-diastolic volume ,Titin ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,Cardiac phenotype ,business ,End-systolic volume - Abstract
Background The clinical course of dilated cardiomyopathy (DCM) is variable: while 20% of patients die within 5 years of diagnosis, up to 15% recover fully. DNA variants that truncate the sarcomeric protein titin (TTNtv) are found in up to 20% of DCM. We sought to characterise the phenotype of TTNtv DCM and evaluate the effect of TTNtv on left ventricular remodelling in DCM. Methods 661 prospectively recruited DCM patients underwent targeted sequencing of TTN and cardiac MRI (CMR) scanning to evaluate left and right ventricular volumes, function, wall thickness and mass (Siemens scanners, 1.5T). TTNtv in constitutive exons were confirmed by Sanger sequencing or visual inspection of reads on IGV. Quantitative phenotypes were compared using t-tests or Mann-Whitney U tests as appropriate. A subgroup of 122 patients underwent follow up CMR scanning. Linear regression analysis was used to determine the effects of TTNtv on interval change in left ventricular ejection fraction (LVEF), left ventricular volumes and mass. Results Targeted sequencing of 661 patients with DCM (mean age 57.3 years, 68% male) identified 62 patients (9.4%) with confirmed truncating variants in TTN (A band, n = 48; I band, n = 11; M band, n = 3). There was no difference in age at diagnosis between patients with and without TTNtv (54.1 yrs vs 57.7 yrs, p = 0.05). Patients with TTNtv had lower maximum and mean left ventricular wall thickness and lower indexed left ventricular stroke volume and mass (Table 1). There was no difference in baseline left or right ventricular ejection fraction between patients with and without TTNtv. 122 DCM patients (mean age 54.3 years, 66% male) underwent an additional CMR with a median follow-up interval of 2.6 years (IQR 1.4–4.6 years). Amongst these, 21 patients (16.9%) had TTNtv in constitutive exons (A band, n = 17; I band, n = 3; M band, n = 1). 67% of patients with TTNtv (14/21) showed an improvement in LVEF >5% compared to 50% of patients without TTNtv (51/101). The mean interval improvement in LVEF between baseline and follow up studies was 6.2% in patients with TTNtv compared to 4.6% in those without (p = 0.55). In regression analysis, the presence of a truncating variant in TTN was not predictive of the interval change in LVEF, indexed left ventricular end diastolic volume, end systolic volume, stroke volume and mass (Table 2). Conclusion These data show that TTNtv DCM is phenotypically characterised by thinner left ventricular walls, lower left ventricular mass and indexed stroke volume in the absence of overt differences in ejection fraction or age at diagnosis. Notably, these data show that there is no evidence that DCM patients with TTNtv have a different pattern of left ventricular remodelling compared to patients without TTNtv. This implies that the presence of a titin truncating mutation in a patient with DCM does not preclude the possibility of functional recovery.
- Published
- 2016
- Full Text
- View/download PDF
45. 143 Clinical and Genetic Characteristics of Familial Dilated Cardiomyopathy in a Large UK Prospective Cohort: Abstract 143 Table 1
- Author
-
Francesco Mazzarotto, Upasana Tayal, Simon Newsome, Nicola Whiffin, James S. Ware, Rachel Buchan, Stuart A. Cook, Roddy Walsh, and Sanjay K Prasad
- Subjects
Proband ,medicine.medical_specialty ,Pathology ,Ejection fraction ,medicine.diagnostic_test ,business.industry ,Cardiomyopathy ,030204 cardiovascular system & hematology ,musculoskeletal system ,medicine.disease ,complex mixtures ,LMNA ,03 medical and health sciences ,Exact test ,0302 clinical medicine ,Internal medicine ,Idiopathic dilated cardiomyopathy ,cardiovascular system ,medicine ,cardiovascular diseases ,030212 general & internal medicine ,Cardiology and Cardiovascular Medicine ,business ,Prospective cohort study ,Genetic testing - Abstract
Background Up to fifty percent of idiopathic dilated cardiomyopathy (DCM) has a familial basis. Variants can occur in over 40 genes, though truncating variants in the sarcomeric gene titin account for the largest proportion (~20%). At least half of familial DCM cases are genetically orphan. We sought to study whether familial DCM was associated with distinct clinical characteristics, independently of the underlying genetic variant. Methods 595 prospectively recruited DCM patients underwent detailed phenotyping with cardiac MRI (Siemens scanners, 1.5T) and were sequenced using a customised panel of ~100 cardiomyopathy genes on Illumina and 5500xl platforms. Variants were identified and annotated using a customised bioinformatics pipeline. Clinical information including family pedigree data, ECG, and arrhythmia status at diagnosis (presence of confirmed ventricular or atrial arrhythmias) was collected on all patients. Familial DCM was defined as DCM occurring in 2 or more 1st or 2nd degree family members. Chi squared or Fisher’s exact test was used to compare across categorical variables and t-tests or Mann-Whitney U tests across continuous variables as appropriate. Results Overall, 16% of patients (95 out of 595) had familial DCM. Thirty individuals came from 13 families, the remaining were unrelated probands. Patients with familial DCM had an earlier age of disease onset (49.8 years vs 58.8 years, p Patients with familial DCM had less conduction disease at baseline (11% vs 36%, p Patients with familial DCM had a milder intermediate phenotype of DCM (left ventricular ejection fraction 45.2% vs 38.2%, p There were 44 potentially disease-causing variants in DCM genes in the familial DCM cohort (Table 1). Genetic testing had a yield of 44% in familial (n = 42), and 22% in non-familial DCM (n = 117). Five patients carried 2 variants. Truncating variants in titin were the most common variant (n = 17) and were over twice as common in patients with familial DCM compared to those without (18% vs 6.8%, p Conclusions Disease causing variants in TTN and LMNA were more commonly associated with familial DCM, but 56% of patients with familial DCM remain genetically unexplained. This highlights the need for further novel DCM disease gene discovery. Our data show that familial DCM is characterised by a younger age of disease onset and less severe ventricular dysfunction as compared to non-familial DCM.
- Published
- 2016
- Full Text
- View/download PDF
46. FineSplice, enhanced splice junction detection and quantification: a novel pipeline based on the assessment of diverse RNA-Seq alignment solutions
- Author
-
Francesco Mazzarotto, Carlos Torroja-Fungairiño, Paul J.R. Barton, Enrique Lara-Pezzi, Stuart A. Cook, Alberto Gatto, Fátima Sánchez-Cabo, Unión Europea. Comisión Europea, European Research Council, Ministerio de Ciencia e Innovación (España), Comunidad de Madrid (España), Ministerio de Economía y Competitividad (España), Fundación ProCNIC, National Health Service (Reino Unido), and British Heart Foundation
- Subjects
Biochemistry & Molecular Biology ,Pipeline (computing) ,05 Environmental Sciences ,CODE ,Biology ,ULTRAFAST ,Bioinformatics ,computer.software_genre ,DISEASE ,Bottleneck ,DIFFERENTIAL EXPRESSION ,Genetics ,False positive paradox ,HUMAN GENOME ,splice ,Science & Technology ,LANDSCAPE ,Sequence Analysis, RNA ,Alternative splicing ,TRANSCRIPTOMES ,06 Biological Sciences ,GENE ,Alternative Splicing ,Filter (video) ,RNA splicing ,Methods Online ,Anomaly detection ,08 Information and Computing Sciences ,Data mining ,RNA Splice Sites ,MESSENGER-RNA ,Life Sciences & Biomedicine ,computer ,Sequence Alignment ,Algorithms ,Software ,Developmental Biology - Abstract
Alternative splicing is the main mechanism governing protein diversity. The recent developments in RNA-Seq technology have enabled the study of the global impact and regulation of this biological process. However, the lack of standardized protocols constitutes a major bottleneck in the analysis of alternative splicing. This is particularly important for the identification of exon-exon junctions, which is a critical step in any analysis workflow. Here we performed a systematic benchmarking of alignment tools to dissect the impact of design and method on the mapping, detection and quantification of splice junctions from multi-exon reads. Accordingly, we devised a novel pipeline based on TopHat2 combined with a splice junction detection algorithm, which we have named FineSplice. FineSplice allows effective elimination of spurious junction hits arising from artefactual alignments, achieving up to 99% precision in both real and simulated data sets and yielding superior F1 scores under most tested conditions. The proposed strategy conjugates an efficient mapping solution with a semi-supervised anomaly detection scheme to filter out false positives and allows reliable estimation of expressed junctions from the alignment output. Ultimately this provides more accurate information to identify meaningful splicing patterns. FineSplice is freely available at https://sourceforge.net/p/finesplice/. European Union’s FP7 [ERG-239158 to E.L.-P., CardioNeT-ITN-289600 to E.L.-P., S.A.C. and P.J.R.B.]; Spanish Ministry of Science and Innovation [BFU2009-10016, SAF2012-31451 to E.L.-P.]; Regional Government of Madrid [2010-BMD-2321 ‘Fibroteam’ to E.L.-P.]; Spanish Ministry of Economy and Competitiveness and by the Pro-CNIC Foundation (towards CNIC); National Institute for Health Research Cardiovascular BRU at the Royal Brompton & Harefield NHS Foundation Trust and Imperial College London (to S.A.C. and P.J.B.). Funding for open access charge: European Union’s FP7 [ERG-239158, CardioNeT-ITN289600 to E.L.-P.] Sí
- Published
- 2014
47. 163 Integrated allelic, transcriptional, and phenotypic dissection of the cardiac effects of titin variation in health and disease
- Author
-
Angharad Roberts, James Ware, Daniel Herman, Sebastian Schafer, Francesco Mazzarotto, John Baksi, Rachel Buchan, Roddy Walsh, Shibu John, Samuel Wilkinson, Leanne Felkin, Alexander Bick, Michael Radke, Michael Gotthardt, Paul Barton, Norbert Hubner, J Seidman, Christine Seidman, and Stuart Cook
- Subjects
Cardiology and Cardiovascular Medicine - Published
- 2015
- Full Text
- View/download PDF
48. 76 Comprehensive Assessment of Rare Genetic Variation in Dilated Cardiomyopathy Genes in Patients and Controls: Abstract 76 Table 1
- Author
-
James S. Ware, Francesco Mazzarotto, Stuart A. Cook, Paul J.R. Barton, Upasana Tayal, Roddy Walsh, and Rachel Buchan
- Subjects
Pathology ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Cardiomyopathy ,Dilated cardiomyopathy ,Single-nucleotide polymorphism ,musculoskeletal system ,medicine.disease ,Gastroenterology ,Internal medicine ,Heart failure ,Genetic variation ,cardiovascular system ,medicine ,cardiovascular diseases ,MYH6 ,Cardiology and Cardiovascular Medicine ,business ,Exome sequencing ,Genetic testing - Abstract
Introduction Dilated cardiomyopathy (DCM) is a genetically heterogenous condition with mutations reported in at least 50 associated genes. 1 Our consortium has demonstrated the presence of truncating mutations in the giant sarcomeric gene titin (TTN) in up to 27% of DCM 2 making this the commonest genetic cause of DCM. Here we conducted a comprehensive analysis of the frequency of rare coding variants in cardiomyopathy genes in DCM patients and ethnically-matched healthy controls. Methods We sequenced 64 cardiomyopathy genes in 651 individuals, comprising 234 patients referred for prospective evaluation of DCM with cardiac MRI (54 ± 14 yrs at scan, 70.5% males), 98 end-stage DCM (39 ± 16 yrs on date of transplant/left-ventricular assist device implantation, 83.7% males), and 319 prospectively recruited adult healthy volunteers (HVOL, 42 ± 14 yrs at scan, 46.1% males), who underwent detailed phenotyping with cardiac MRI. Next-generation targeted exon sequencing was performed on the SOLiD 5500xl platform. Variant calling was performed using Lifescope v2.5.1. Data were mapped to the Hg19 (GRCh37) human genome reference. Caucasian ethnicity was confirmed using Principal Component Analysis. Burden testing for potentially disease-causing variation was performed on each gene. Results DCM patients were enriched for novel protein-altering variants in 28 genes (Table 1: top 7 genes), but only 5 achieved nominal significance. Truncating variants in TTN (TTNtv) remained significant after multiple testing correction (12% DCM, 1.9% controls, p = 8.1 × 10–6). Rare variation in SCN5A and MYH6, previously reported to play a key role in DCM, were not enriched in DCM (SCN5A – 2.1% DCM, 2.8% controls, p = 0.80; MYH6 –0.9% DCM, 2.2% controls, p = 0.95) and nor were non-synonymous SNPs in TTN (TTNns, 25.6% DCM, 25.7% controls, p = 0.55). Importantly, an additive effect of variation in multiple DCM genes on DCM risk was identified using logistic regression models (p = 5.7 × 10 x -4), demonstrating a multi-genic basis for DCM in some cases. Conclusions To the best of our knowledge, this is the first comprehensive study of the genetic architecture of DCM compared to an ethnically-matched healthy control cohort. The majority of genes reported to contain rare variants in DCM have similar burdens of variation in controls. Some variants may contribute to disease, but excluding TTNtv the majority of such variants are uninterpretable in the absence of functional or segregation data. References Mestroni L, Taylor MR. Genetics and genetic testing of dilated cardiomyopathy: a new perspective. Discov Med. 2013;15(80):43-9 Herman DS, Lam L, Taylor MR, Wang L, Teekakirikul P, Christodoulou D, et al . Truncations of titin causing dilated cardiomyopathy. N Engl J Med. 2012;366(7):619-28
- Published
- 2015
- Full Text
- View/download PDF
49. Titin-truncating variants affect heart function in disease cohorts and the general population
- Author
-
Francesco Mazzarotto, Laura Lihua Chan, Calvin W. L. Chin, Ester Khin, Carlo Biffi, Paul J.R. Barton, Upasana Tayal, Sebastiaan van Heesch, Sebastian Schafer, Roddy Walsh, Miao Kui, Lorna R. Fiedler, Nicole Tee, Norbert Hubner, Daniel Rueckert, Antonio de Marvao, Stuart A. Cook, James S. Ware, Valentin Schneider, David Sim, Teresa Totman, Franziska Kreuchwig, Declan P. O'Regan, Nicole S. J. Ko, Timothy J W Dawes, Chee Jian Pua, Benjamin Ng, Owen J. L. Rackham, Sanjay K Prasad, Allison Faber, Dominique P.V. de Kleijn, Jonathan G. Seidman, Eleonora Adami, Wolfgang A. Linke, Jean-Paul Kovalik, Vera Regitz-Zagrosek, Christine E. Seidman, British Heart Foundation, Imperial College Healthcare NHS Trust- BRC Funding, Commission of the European Communities, Fondation Leducq, National Institute for Health Research, Engineering & Physical Science Research Council (EPSRC), Wellcome Trust, Department of Health, and Medical Research Council
- Subjects
0301 basic medicine ,Cardiomyopathy, Dilated ,Male ,Population ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Bioinformatics ,Article ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Genetics ,Journal Article ,Animals ,Humans ,Connectin ,Comparative Study ,Allele ,education ,Cardiac imaging ,education.field_of_study ,biology ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Dilated cardiomyopathy ,Heart ,11 Medical And Health Sciences ,06 Biological Sciences ,medicine.disease ,Stop codon ,Cardiovascular physiology ,Rats ,030104 developmental biology ,Case-Control Studies ,biology.protein ,cardiovascular system ,Titin ,Developmental Biology - Abstract
Titin-truncating variants (TTNtv) commonly cause dilated cardiomyopathy (DCM). TTNtv are also encountered in 1/41% of the general population, where they may be silent, perhaps reflecting allelic factors. To better understand TTNtv, we integrated TTN allelic series, cardiac imaging and genomic data in humans and studied rat models with disparate TTNtv. In patients with DCM, TTNtv throughout titin were significantly associated with DCM. Ribosomal profiling in rat showed the translational footprint of premature stop codons in Ttn, TTNtv-position-independent nonsense-mediated degradation of the mutant allele and a signature of perturbed cardiac metabolism. Heart physiology in rats with TTNtv was unremarkable at baseline but became impaired during cardiac stress. In healthy humans, machine-learning-based analysis of high-resolution cardiac imaging showed TTNtv to be associated with eccentric cardiac remodeling. These data show that TTNtv have molecular and physiological effects on the heart across species, with a continuum of expressivity in health and disease.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.