57 results on '"Steven R. DePalma"'
Search Results
2. GATA6 mutations in hiPSCs inform mechanisms for maldevelopment of the heart, pancreas, and diaphragm
- Author
-
Arun Sharma, Lauren K Wasson, Jon AL Willcox, Sarah U Morton, Joshua M Gorham, Daniel M DeLaughter, Meraj Neyazi, Manuel Schmid, Radhika Agarwal, Min Young Jang, Christopher N Toepfer, Tarsha Ward, Yuri Kim, Alexandre C Pereira, Steven R DePalma, Angela Tai, Seongwon Kim, David Conner, Daniel Bernstein, Bruce D Gelb, Wendy K Chung, Elizabeth Goldmuntz, George Porter, Martin Tristani-Firouzi, Deepak Srivastava, Jonathan G Seidman, Christine E Seidman, and Pediatric Cardiac Genomics Consortium
- Subjects
stem cells ,iPSC ,gene mutation ,CRISPR ,heart ,development ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Damaging GATA6 variants cause cardiac outflow tract defects, sometimes with pancreatic and diaphragmic malformations. To define molecular mechanisms for these diverse developmental defects, we studied transcriptional and epigenetic responses to GATA6 loss of function (LoF) and missense variants during cardiomyocyte differentiation of isogenic human induced pluripotent stem cells. We show that GATA6 is a pioneer factor in cardiac development, regulating SMYD1 that activates HAND2, and KDR that with HAND2 orchestrates outflow tract formation. LoF variants perturbed cardiac genes and also endoderm lineage genes that direct PDX1 expression and pancreatic development. Remarkably, an exon 4 GATA6 missense variant, highly associated with extra-cardiac malformations, caused ectopic pioneer activities, profoundly diminishing GATA4, FOXA1/2, and PDX1 expression and increasing normal retinoic acid signaling that promotes diaphragm development. These aberrant epigenetic and transcriptional signatures illuminate the molecular mechanisms for cardiovascular malformations, pancreas and diaphragm dysgenesis that arise in patients with distinct GATA6 variants.
- Published
- 2020
- Full Text
- View/download PDF
3. Pathogenesis of Cardiomyopathy Caused by Variants in ALPK3 , an Essential Pseudokinase in the Cardiomyocyte Nucleus and Sarcomere
- Author
-
Radhika Agarwal, Hiroko Wakimoto, Joao A. Paulo, Qi Zhang, Daniel Reichart, Christopher Toepfer, Arun Sharma, Angela C. Tai, Mingyue Lun, Joshua Gorham, Steven R. DePalma, Steven P. Gygi, J.G. Seidman, and Christine E. Seidman
- Subjects
Physiology (medical) ,Cardiology and Cardiovascular Medicine - Abstract
Background: ALPK3 encodes α-kinase 3, a muscle-specific protein of unknown function. ALPK3 loss-of-function variants cause cardiomyopathy with distinctive clinical manifestations in both children and adults, but the molecular functions of ALPK3 remain poorly understood. Methods: We explored the putative kinase activity of ALPK3 and the consequences of damaging variants using isogenic human induced pluripotent stem cell–derived cardiomyocytes, mice, and human patient tissues. Results: Multiple sequence alignment of all human α-kinase domains and their orthologs revealed 4 conserved residues that were variant only in ALPK3, demonstrating evolutionary divergence of the ALPK3 α-kinase domain sequence. Phosphoproteomic evaluation of both ALPK3 kinase domain inhibition and overexpression failed to detect significant changes in catalytic activity, establishing ALPK3 as a pseudokinase. Investigations into alternative functions revealed that ALPK3 colocalized with myomesin proteins (MYOM1, MYOM2) at both the nuclear envelope and the sarcomere M-band. ALPK3 loss-of-function variants caused myomesin proteins to mislocalize and also dysregulated several additional M-band proteins involved in sarcomere protein turnover, which ultimately impaired cardiomyocyte structure and function. Conclusions: ALPK3 is an essential cardiac pseudokinase that inserts in the nuclear envelope and the sarcomere M-band. Loss of ALPK3 causes mislocalization of myomesins, critical force-buffering proteins in cardiomyocytes, and also dysregulates M-band proteins necessary for sarcomere protein turnover. We conclude that ALPK3 cardiomyopathy induces ventricular dilatation caused by insufficient myomesin-mediated force buffering and hypertrophy by impairment of sarcomere proteostasis.
- Published
- 2022
- Full Text
- View/download PDF
4. Contribution of Previously Unrecognized RNA Splice-Altering Variants to Congenital Heart Disease
- Author
-
Min Young Jang, Parth N. Patel, Alexandre C. Pereira, Jon A.L. Willcox, Alireza Haghighi, Angela C. Tai, Kaoru Ito, Sarah U. Morton, Joshua M. Gorham, David M. McKean, Steven R. DePalma, Daniel Bernstein, Martina Brueckner, Wendy K. Chung, Alessandro Giardini, Elizabeth Goldmuntz, Jonathan R. Kaltman, Richard Kim, Jane W. Newburger, Yufeng Shen, Deepak Srivastava, Martin Tristani-Firouzi, Bruce D. Gelb, George A. Porter, Christine E. Seidman, and Jonathan G. Seidman
- Subjects
General Medicine - Abstract
BACKGROUND: Known genetic causes of congenital heart disease (CHD) explain METHODS: We tested de novo variants from trio studies of 2649 CHD probands and their parents, as well as rare (allele frequency, − 6 ) variants from 4472 CHD probands in the Pediatric Cardiac Genetics Consortium through a combined computational and in vitro approach. RESULTS: We identified 53 de novo and 74 rare variants in CHD cases that alter splicing and thus are loss of function. Of these, 77 variants are in known dominant, recessive, and candidate CHD genes, including KMT2D and RBFOX2 . In 1 case, we confirmed the variant’s predicted impact on RNA splicing in RNA transcripts from the proband’s cardiac tissue. Two probands were found to have 2 loss-of-function variants for recessive CHD genes HECTD1 and DYNC2H1 . In addition, SpliceAI—a predictive algorithm for altered RNA splicing—has a positive predictive value of ≈93% in our cohort. CONCLUSIONS: Through assessment of RNA splicing, we identified a new loss-of-function variant within a CHD gene in 78 probands, of whom 69 (1.5%; n=4472) did not have a previously established genetic explanation for CHD. Identification of splice-altering variants improves diagnostic classification and genetic diagnoses for CHD. REGISTRATION: URL: https://clinicaltrials.gov ; Unique identifier: NCT01196182.
- Published
- 2023
- Full Text
- View/download PDF
5. Filamin C Cardiomyopathy Variants Cause Protein and Lysosome Accumulation
- Author
-
Joshua M. Gorham, Christine E. Seidman, Christopher S. Chen, Jonathan G. Seidman, Subramanian Sundaram, Christopher N. Toepfer, Radhika Agarwal, Jourdan K. Ewoldt, Steven P. Gygi, Joao A. Paulo, Steven R. DePalma, Qi Zhang, and Anant Chopra
- Subjects
Sarcomeres ,Physiology ,Filamins ,Induced Pluripotent Stem Cells ,Autophagy ,Biology ,Filamin ,Myocardial Contraction ,Sarcomere ,Article ,Protein–protein interaction ,Cell biology ,medicine.anatomical_structure ,Lysosome ,medicine ,Humans ,Myocytes, Cardiac ,FLNC ,Sarcomere organization ,Cardiomyopathies ,Lysosomes ,Cardiology and Cardiovascular Medicine ,Haploinsufficiency ,Cells, Cultured - Abstract
Rationale: Dominant heterozygous variants in filamin C ( FLNC ) cause diverse cardiomyopathies, although the underlying molecular mechanisms remain poorly understood. Objective: We aimed to define the molecular mechanisms by which FLNC variants altered human cardiomyocyte gene and protein expression, sarcomere structure, and contractile performance. Methods and Results: Using CRISPR/Cas9, we introduced FLNC variants into human induced pluripotent stem cell–derived cardiomyocytes (hiPSC-CMs). We compared isogenic hiPSC-CMs with normal (wild-type), ablated expression ( FLNC −/− ), or haploinsufficiency ( FLNC +/− ) that causes dilated cardiomyopathy. We also studied a heterozygous in-frame deletion ( FLNC +/Δ7aa ) which did not affect FLNC expression but caused aggregate formation, similar to FLNC variants associated with hypertrophic cardiomyopathy. FLNC −/− hiPSC-CMs demonstrated profound sarcomere misassembly and reduced contractility. Although sarcomere formation and function were unaffected in FLNC +/ − and FLNC +/Δ7aa hiPSC-CMs, these heterozygous variants caused increases in lysosome content, enhancement of autophagic flux, and accumulation of FLNC-binding partners and Z-disc proteins. Conclusions: FLNC expression is required for sarcomere organization and physiological function. Variants that produce misfolded FLNC proteins cause the accumulation of FLNC and FLNC-binding partners which leads to increased lysosome expression and activation of autophagic pathways. Surprisingly, similar pathways were activated in FLNC haploinsufficient hiPSC-CMs, likely initiated by the loss of stoichiometric FLNC protein interactions and impaired turnover of proteins at the Z-disc. These results indicate that both FLNC haploinsufficient variants and variants that produce misfolded FLNC protein cause disease by similar proteotoxic mechanisms and indicate the therapeutic potential for augmenting protein degradative pathways to treat a wide range of FLNC -related cardiomyopathies.
- Published
- 2021
- Full Text
- View/download PDF
6. Damaging variants in FOXI3 cause microtia and craniofacial microsomia
- Author
-
Daniel Quiat, Andrew T. Timberlake, Justin J. Curran, Michael L. Cunningham, Barbara McDonough, Maria A. Artunduaga, Steven R. DePalma, Milagros M. Duenas-Roque, Joshua M. Gorham, Jonas A. Gustafson, Usama Hamdan, Anne V. Hing, Paula Hurtado-Villa, Yamileth Nicolau, Gabriel Osorno, Harry Pachajoa, Gloria L. Porras-Hurtado, Lourdes Quintanilla-Dieck, Luis Serrano, Melissa Tumblin, Ignacio Zarante, Daniela V. Luquetti, Roland D. Eavey, Carrie L. Heike, Jonathan G. Seidman, and Christine E. Seidman
- Subjects
Genetics (clinical) - Abstract
Craniofacial microsomia (CFM) represents a spectrum of craniofacial malformations, ranging from isolated microtia with or without aural atresia to underdevelopment of the mandible, maxilla, orbit, facial soft tissue, and/or facial nerve. The genetic causes of CFM remain largely unknown.We performed genome sequencing and linkage analysis in patients and families with microtia and CFM of unknown genetic etiology. The functional consequences of damaging missense variants were evaluated through expression of wild-type and mutant proteins in vitro.We studied a 5-generation kindred with microtia, identifying a missense variant in FOXI3 (p.Arg236Trp) as the cause of disease (logarithm of the odds = 3.33). We subsequently identified 6 individuals from 3 additional kindreds with microtia-CFM spectrum phenotypes harboring damaging variants in FOXI3, a regulator of ectodermal and neural crest development. Missense variants in the nuclear localization sequence were identified in cases with isolated microtia with aural atresia and found to affect subcellular localization of FOXI3. Loss of function variants were found in patients with microtia and mandibular hypoplasia (CFM), suggesting dosage sensitivity of FOXI3.Damaging variants in FOXI3 are the second most frequent genetic cause of CFM, causing 1% of all cases, including 13% of familial cases in our cohort.
- Published
- 2022
7. An ancient founder mutation located between
- Author
-
Daniel, Quiat, Seong Won, Kim, Qi, Zhang, Sarah U, Morton, Alexandre C, Pereira, Steven R, DePalma, Jon A L, Willcox, Barbara, McDonough, Daniel M, DeLaughter, Joshua M, Gorham, Justin J, Curran, Melissa, Tumblin, Yamileth, Nicolau, Maria A, Artunduaga, Lourdes, Quintanilla-Dieck, Gabriel, Osorno, Luis, Serrano, Usama, Hamdan, Roland D, Eavey, Christine E, Seidman, and J G, Seidman
- Subjects
Mutation ,Humans ,Nerve Tissue Proteins ,Ear, External ,Receptors, Immunologic ,Founder Effect ,American Indian or Alaska Native ,Congenital Microtia - Abstract
Microtia is a congenital malformation that encompasses mild hypoplasia to complete loss of the external ear, or pinna. Although the contribution of genetic variation and environmental factors to microtia remains elusive, Amerindigenous populations have the highest reported incidence. Here, using both transmission disequilibrium tests and association studies in microtia trios (parents and affected child) and microtia cohorts enrolled in Latin America, we map an ∼10-kb microtia locus (odds ratio = 4.7; P = 6.78e-18) to the intergenic region between Roundabout 1 (ROBO1) and Roundabout 2 (ROBO2) (chr3: 78546526 to 78555137). While alleles at the microtia locus significantly increase the risk of microtia, their penetrance is low (1%). We demonstrate that the microtia locus contains a polymorphic complex repeat element that is expanded in affected individuals. The locus is located near a chromatin loop region that regulates ROBO1 and ROBO2 expression in induced pluripotent stem cell–derived neural crest cells. Furthermore, we use single nuclear RNA sequencing to demonstrate ROBO1 and ROBO2 expression in both fibroblasts and chondrocytes of the mature human pinna. Because the microtia allele is enriched in Amerindigenous populations and is shared by some East Asian subjects with craniofacial malformations, we propose that both populations share a mutation that arose in a common ancestor prior to the ancient migration of Eurasian populations into the Americas and that the high incidence of microtia among Amerindigenous populations reflects the population bottleneck that occurred during the migration out of Eurasia.
- Published
- 2022
8. Genomic analyses implicate noncoding de novo variants in congenital heart disease
- Author
-
Daniel Bernstein, Martin Tristani-Firouzi, Jane W. Newburger, Sarah U. Morton, Diane E. Dickel, Lauren K. Wasson, Seong Won Kim, Jonathan G. Seidman, Martina Brueckner, Hongjian Qi, Elizabeth Goldmuntz, George A. Porter, Eric E. Schadt, Olga G. Troyanskaya, Kathryn B. Manheimer, Jian Zhou, Jason Homsy, Michael Parfenov, Steven R. DePalma, Bruce D. Gelb, Andrew Farrell, Alexander Kitaygorodsky, Matt Velinder, Gabor T. Marth, Richard B. Kim, Nihir Patel, Jonathan R. Kaltman, Felix Richter, Deepak Srivastava, Kathleen M. Chen, Yufeng Shen, Joshua M. Gorham, Christine E. Seidman, Alessandro Giardini, and Wendy K. Chung
- Subjects
Proband ,Genetics ,0303 health sciences ,Heart disease ,Genomics ,Odds ratio ,Biology ,medicine.disease ,Genome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Young adult ,Enhancer ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
A genetic etiology is identified for one-third of patients with congenital heart disease (CHD), with 8% of cases attributable to coding de novo variants (DNVs). To assess the contribution of noncoding DNVs to CHD, we compared genome sequences from 749 CHD probands and their parents with those from 1,611 unaffected trios. Neural network prediction of noncoding DNV transcriptional impact identified a burden of DNVs in individuals with CHD (n = 2,238 DNVs) compared to controls (n = 4,177; P = 8.7 × 10-4). Independent analyses of enhancers showed an excess of DNVs in associated genes (27 genes versus 3.7 expected, P = 1 × 10-5). We observed significant overlap between these transcription-based approaches (odds ratio (OR) = 2.5, 95% confidence interval (CI) 1.1-5.0, P = 5.4 × 10-3). CHD DNVs altered transcription levels in 5 of 31 enhancers assayed. Finally, we observed a DNV burden in RNA-binding-protein regulatory sites (OR = 1.13, 95% CI 1.1-1.2, P = 8.8 × 10-5). Our findings demonstrate an enrichment of potentially disruptive regulatory noncoding DNVs in a fraction of CHD at least as high as that observed for damaging coding DNVs.
- Published
- 2020
- Full Text
- View/download PDF
9. Contribution of Noncanonical Splice Variants toTTNTruncating Variant Cardiomyopathy
- Author
-
Barbara McDonough, Alireza Haghighi, Joshua M. Gorham, Christine E. Seidman, Min Young Jang, Diane Fatkin, Neal K. Lakdawala, Parth N Patel, Amy E. Roberts, Kaoru Ito, Lien Lam, Steven R. DePalma, Jon G. Seidman, Renee Johnson, Stuart A. Cook, Jon A. L. Willcox, Paul J.R. Barton, Guys & St Thomas NHS Foundation Trust, Imperial College Healthcare NHS Trust- BRC Funding, and British Heart Foundation
- Subjects
Genetics ,intron ,Cardiomyopathy ,Intron ,heart failure ,General Medicine ,Biology ,medicine.disease ,Exon ,Heart failure ,RNA splicing ,Idiopathic dilated cardiomyopathy ,myocardium ,medicine ,splice ,exon ,Invariant (mathematics) ,cardiomyopathy - Abstract
Background:HeterozygousTTNtruncating variants cause 10% to 20% of idiopathic dilated cardiomyopathy (DCM). Although variants which disrupt canonical splice signals (ie, invariant dinucleotide of the splice donor site, invariant dinucleotide of the splice acceptor site) at exon-intron junctions are readily recognized asTTNtruncating variants, the effects of other nearby sequence variations on splicing and their contribution to disease is uncertain.Methods:Rare variants of unknown significance located in the splice regions of highly expressedTTNexons from 203 DCM cases, 3329 normal subjects, and clinical variant databases were identified. The effects of these variants on splicing were assessed using an in vitro splice assay.Results:Splice-altering variants of unknown significance were enriched in DCM cases over controls and present in 2% of DCM patients (P=0.002). Application of this method to clinical variant databases demonstrated 20% of similar variants of unknown significance inTTNsplice regions affect splicing. Noncanonical splice-altering variants were most frequently located at position +5 of the donor site (P=4.4×107) and position -3 of the acceptor site (P=0.002). SpliceAI, an emerging in silico prediction tool, had a high positive predictive value (86%–95%) but poor sensitivity (15%–50%) for the detection of splice-altering variants. Alternate exons spliced out of mostTTNtranscripts frequently lacked the consensus base at +5 donor and −3 acceptor positions.Conclusions:Noncanonical splice-altering variants inTTNexplain 1-2% of DCM and offer a 10-20% increase in the diagnostic power ofTTNsequencing in this disease. These data suggest rules that may improve efforts to detect splice-altering variants in other genes and may explain the low percent splicing observed for many alternateTTNexons.
- Published
- 2021
- Full Text
- View/download PDF
10. Contribution of Noncanonical Splice Variants to
- Author
-
Parth N, Patel, Kaoru, Ito, Jon A L, Willcox, Alireza, Haghighi, Min Young, Jang, Joshua M, Gorham, Steven R, DePalma, Lien, Lam, Barbara, McDonough, Renee, Johnson, Neal K, Lakdawala, Amy, Roberts, Paul J R, Barton, Stuart A, Cook, Diane, Fatkin, Christine E, Seidman, and J G, Seidman
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Heterozygote ,Adolescent ,RNA Splicing ,Humans ,Connectin ,Female ,Exons ,Middle Aged ,Article - Abstract
BACKGROUND: Heterozygous truncating variants in titin (TTNtv) cause 10–20% of idiopathic dilated cardiomyopathy (DCM). Though variants which disrupt canonical splice signals (i.e. GT, AG) at exon-intron junctions are readily recognized as TTNtv, the effects of other nearby sequence variations on splicing and their contribution to disease is uncertain. METHODS: Rare variants of unknown significance (VUS) located in the splice regions of highly expressed TTN exons from 203 DCM cases, 3329 normal subjects, and clinical variant databases were identified. The effects of these variants on splicing were assessed using an in vitro splice assay. RESULTS: Splice-altering VUS were enriched in DCM cases over controls and present in 2% of DCM patients (p=0.002). Application of this method to clinical variant databases demonstrated 20% of similar VUS in TTN splice regions affect splicing. Non-canonical splice-altering variants were most frequently located at position +5 of the donor site (p=4.4e-7) and position −3 of the acceptor site (p=0.002). SpliceAI, an emerging in-silico prediction tool, had a high positive predictive value (86–95%) but poor sensitivity (15–50%) for the detection of splice-altering variants. Alternate exons spliced out of most TTN transcripts frequently lacked the consensus base at +5 donor and −3 acceptor positions. CONCLUSION: Non-canonical splice-altering variants in TTN explain 1–2% of DCM and offer a 10–20% increase in the diagnostic power of TTN sequencing in this disease. These data suggest rules that may improve efforts to detect splice-altering variants in other genes and may explain the low percent splicing observed for many alternate TTN exons.
- Published
- 2021
11. Genetic and Phenotypic Landscape of Peripartum Cardiomyopathy
- Author
-
Denise Hilfiker-Kleiner, Zolt Arany, Quentin McAfee, Kenneth B. Margulies, Rami Alharethi, Eileen Hsich, Lisa D. Levine, Sorel Goland, Christine E. Seidman, Peter Damm, Jonathan G. Seidman, Sarosh Rana, Daniel Jacoby, Thomas P. Cappola, Chizuko Kamiya, Julie B. Damp, Anne S Ersbøll, Jeff Brandimarto, Steven R. DePalma, Rahul R. Goli, Richard Sheppard, Imac, Uri Elkayam, Ipac Investigators, Valerie Riis, John P. Boehmer, Finn Gustafsson, George A. Macones, Dennis M. McNamara, Jeffrey D. Alexis, Alireza Haghighi, Daniel P. Judge, and Jian Li
- Subjects
Adult ,0303 health sciences ,medicine.medical_specialty ,Peripartum cardiomyopathy ,Obstetrics ,business.industry ,030204 cardiovascular system & hematology ,medicine.disease ,Article ,03 medical and health sciences ,0302 clinical medicine ,Phenotype ,Pregnancy ,Physiology (medical) ,medicine ,Peripartum Period ,Humans ,Female ,Cardiology and Cardiovascular Medicine ,business ,Cardiomyopathies ,030304 developmental biology ,Retrospective Studies - Abstract
Background: Peripartum cardiomyopathy (PPCM) occurs in ≈1:2000 deliveries in the United States and worldwide. The genetic underpinnings of PPCM remain poorly defined. Approximately 10% of women with PPCM harbor truncating variants in TTN (TTNtvs). Whether mutations in other genes can predispose to PPCM is not known. It is also not known if the presence of TTNtvs predicts clinical presentation or outcomes. Nor is it known if the prevalence of TTNtvs differs in women with PPCM and preeclampsia, the strongest risk factor for PPCM. Methods: Women with PPCM were retrospectively identified from several US and international academic centers, and clinical information and DNA samples were acquired. Next-generation sequencing was performed on 67 genes, including TTN , and evaluated for burden of truncating and missense variants. The impact of TTNtvs on the severity of clinical presentation, and on clinical outcomes, was evaluated. Results: Four hundred sixty-nine women met inclusion criteria. Of the women with PPCM, 10.4% bore TTNtvs (odds ratio=9.4 compared with 1.2% in the reference population; Bonferroni-corrected P [ P *]=1.2×10 –46 ). We additionally identified overrepresentation of truncating variants in FLNC (odds ratio=24.8, P *=7.0×10 –8 ), DSP (odds ratio=14.9, P *=1.0×10 –8 ), and BAG3 (odds ratio=53.1, P *=0.02), genes not previously associated with PPCM. This profile is highly similar to that found in nonischemic dilated cardiomyopathy. Women with TTNtvs had lower left ventricular ejection fraction on presentation than did women without TTNtvs (23.5% versus 29%, P =2.5×10 –4 ), but did not differ significantly in timing of presentation after delivery, in prevalence of preeclampsia, or in rates of clinical recovery. Conclusions: This study provides the first extensive genetic and phenotypic landscape of PPCM and demonstrates that predisposition to heart failure is an important risk factor for PPCM. The work reveals a degree of genetic similarity between PPCM and dilated cardiomyopathy, suggesting that gene-specific therapeutic approaches being developed for dilated cardiomyopathy may also apply to PPCM, and that approaches to genetic testing in PPCM should mirror those taken in dilated cardiomyopathy. Last, the clarification of genotype/phenotype associations has important implications for genetic counseling.
- Published
- 2021
12. Discordant clinical features of identical hypertrophic cardiomyopathy twins
- Author
-
Steven R. DePalma, Neal K. Lakdawala, Barbara McDonough, Jodie Ingles, Giuliana G. Repetti, Mark W. Russell, Sharlene M. Day, Daniel Quiat, Eric M. Green, Carolyn Y. Ho, Christopher Semsarian, Christine E. Seidman, Alexandre C. Pereira, Yuri Kim, and Jonathan G. Seidman
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Adolescent ,Heart Ventricles ,Muscle Proteins ,Disease ,030204 cardiovascular system & hematology ,Biology ,Sarcomere ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Genetic variation ,medicine ,Humans ,cardiovascular diseases ,Epigenetics ,Multidisciplinary ,Ejection fraction ,Hypertrophic cardiomyopathy ,Twins, Monozygotic ,Cardiomyopathy, Hypertrophic ,Middle Aged ,Biological Sciences ,medicine.disease ,Phenotype ,030104 developmental biology ,Echocardiography ,Child, Preschool ,cardiovascular system ,Cardiology ,Etiology ,Female ,Follow-Up Studies - Abstract
Hypertrophic cardiomyopathy (HCM) is a disease of heart muscle, which affects ∼1 in 500 individuals and is characterized by increased left ventricular wall thickness. While HCM is caused by pathogenic variants in any one of eight sarcomere protein genes, clinical expression varies considerably, even among patients with the same pathogenic variant. To determine whether background genetic variation or environmental factors drive these differences, we studied disease progression in 11 pairs of monozygotic HCM twins. The twin pairs were followed for 5 to 14 y, and left ventricular wall thickness, left atrial diameter, and left ventricular ejection fraction were collected from echocardiograms at various time points. All nine twin pairs with sarcomere protein gene variants and two with unknown disease etiologies had discordant morphologic features of the heart, demonstrating the influence of nonhereditable factors on clinical expression of HCM. Whole genome sequencing analysis of the six monozygotic twins with discordant HCM phenotypes did not reveal notable somatic genetic variants that might explain their clinical differences. Discordant cardiac morphology of identical twins highlights a significant role for epigenetics and environment in HCM disease progression.
- Published
- 2021
- Full Text
- View/download PDF
13. Mechanisms of Congenital Heart Disease Caused by NAA15 Haploinsufficiency
- Author
-
Bruce D. Gelb, Yuri Kim, Jonathan G. Seidman, Joshua M. Gorham, Kehan Zhang, Sylvia Varland, Christine E. Seidman, Tarsha Ward, Kris Gevaert, Richard P. Lifton, Min Young Jang, Alireza Haghighi, Alexandre C. Pereira, Elizabeth Goldmuntz, Wendy K. Chung, Jason Homsy, Warren Tai, Martina Brueckner, Jon A. L. Willcox, Christopher S. Chen, George A. Porter, Thomas Arnesen, Sarah U. Morton, Benoit G. Bruneau, H. Joseph Yost, Craig C. Benson, Evy Timmerman, Petra Van Damme, Francis Impens, Delphi Van Haver, Steven R. DePalma, and Gabriela Venturini
- Subjects
0301 basic medicine ,Heart Defects, Congenital ,NatA complex ,GENES ,ACETYLATION ,Proteome ,induced pluripotent stem cells ,Physiology ,Induced Pluripotent Stem Cells ,Mutation, Missense ,Haploinsufficiency ,Biology ,Proteomics ,Article ,ribosomes ,CELL-PROLIFERATION ,03 medical and health sciences ,proteomics ,0302 clinical medicine ,MOLECULAR-BASIS ,Medicine and Health Sciences ,Protein biosynthesis ,Humans ,RIBOSOME ,YEAST ,Acetyltransferase complex ,N-Terminal Acetyltransferase E ,Induced pluripotent stem cell ,Child ,Peptide sequence ,Cells, Cultured ,N-Terminal Acetyltransferase A ,NATA ,Acetylation ,congenital heart defects ,proteins ,haploinsufficiency ,Cell biology ,N-TERMINAL ACETYLTRANSFERASES ,030104 developmental biology ,DE-NOVO MUTATIONS ,030220 oncology & carcinogenesis ,Cardiology and Cardiovascular Medicine ,Protein Processing, Post-Translational ,PROTEIN HYPK ,NAA15 - Abstract
Rationale: NAA15 (N-alpha-acetyltransferase 15) is a component of the NatA (N-terminal acetyltransferase complex). The mechanism by which NAA15 haploinsufficiency causes congenital heart disease remains unknown. To better understand molecular processes by which NAA15 haploinsufficiency perturbs cardiac development, we introduced NAA15 variants into human induced pluripotent stem cells (iPSCs) and assessed the consequences of these mutations on RNA and protein expression. Objective: We aim to understand the role of NAA15 haploinsufficiency in cardiac development by investigating proteomic effects on NatA complex activity and identifying proteins dependent upon a full amount of NAA15. Methods and Results: We introduced heterozygous loss of function, compound heterozygous, and missense residues (R276W) in iPSCs using CRISPR/Cas9. Haploinsufficient NAA15 iPSCs differentiate into cardiomyocytes, unlike NAA15 -null iPSCs, presumably due to altered composition of NatA. Mass spectrometry analyses reveal ≈80% of identified iPSC NatA targeted proteins displayed partial or complete N-terminal acetylation. Between null and haploinsufficient NAA15 cells, N-terminal acetylation levels of 32 and 9 NatA-specific targeted proteins were reduced, respectively. Similar acetylation loss in few proteins occurred in NAA15 R276W induced pluripotent stem cells. In addition, steady-state protein levels of 562 proteins were altered in both null and haploinsufficient NAA15 cells; 18 were ribosomal-associated proteins. At least 4 proteins were encoded by genes known to cause autosomal dominant congenital heart disease. Conclusions: These studies define a set of human proteins that requires a full NAA15 complement for normal synthesis and development. A 50% reduction in the amount of NAA15 alters levels of at least 562 proteins and N-terminal acetylation of only 9 proteins. One or more modulated proteins are likely responsible for NAA15-haploinsufficiency mediated congenital heart disease. Additionally, genetically engineered induced pluripotent stem cells provide a platform for evaluating the consequences of amino acid sequence variants of unknown significance on NAA15 function.
- Published
- 2021
14. Author response: GATA6 mutations in hiPSCs inform mechanisms for maldevelopment of the heart, pancreas, and diaphragm
- Author
-
Angela Tai, Elizabeth Goldmuntz, Lauren K. Wasson, Yuri Kim, Manuel Schmid, Deepak Srivastava, Steven R. DePalma, Daniel M. DeLaughter, Jonathan G. Seidman, George A. Porter, Min Young Jang, Arun Sharma, Jon A. L. Willcox, Sarah U. Morton, Alexandre C. Pereira, Radhika Agarwal, Martin Tristani-Firouzi, Seongwon Kim, Joshua M. Gorham, Christine E. Seidman, Tarsha Ward, Christopher N. Toepfer, Meraj Neyazi, Daniel Bernstein, Wendy K. Chung, Bruce D. Gelb, and David A. Conner
- Subjects
GATA6 ,medicine.anatomical_structure ,business.industry ,Maldevelopment ,Medicine ,Anatomy ,business ,Pancreas ,Diaphragm (structural system) - Published
- 2020
- Full Text
- View/download PDF
15. GATA6 mutations in hiPSCs inform mechanisms for maldevelopment of the heart, pancreas, and diaphragm
- Author
-
Min Young Jang, Yuri Kim, Daniel Bernstein, Deepak Srivastava, Jonathan G. Seidman, Elizabeth Goldmuntz, Daniel M. DeLaughter, Manuel Schmid, Christopher N. Toepfer, David A. Conner, Angela Tai, Steven R. DePalma, Alexandre C. Pereira, Bruce D. Gelb, Jon A. L. Willcox, Meraj Neyazi, George A. Porter, Arun Sharma, Radhika Agarwal, Martin Tristani-Firouzi, Tarsha Ward, Lauren K. Wasson, Sarah U. Morton, Seongwon Kim, Joshua M. Gorham, Christine E. Seidman, and Wendy K. Chung
- Subjects
0301 basic medicine ,Gene mutation ,Cardiovascular ,0302 clinical medicine ,GATA6 Transcription Factor ,2.1 Biological and endogenous factors ,gene mutation ,Aetiology ,Biology (General) ,Pediatric ,GATA6 ,iPSC ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,General Neuroscience ,Cell Differentiation ,General Medicine ,Stem Cells and Regenerative Medicine ,Cell biology ,Heart Disease ,medicine.anatomical_structure ,CRISPR ,cardiovascular system ,Medicine ,PDX1 ,Pancreas ,HAND2 ,Cardiac ,Research Article ,Human ,endocrine system ,QH301-705.5 ,Science ,Diaphragm ,Induced Pluripotent Stem Cells ,regenerative medicine ,heart ,Biology ,General Biochemistry, Genetics and Molecular Biology ,developmental biology ,03 medical and health sciences ,Genetic ,stem cells ,Genetics ,medicine ,Humans ,human ,Epigenetics ,development ,Myocytes ,Stem Cell Research - Induced Pluripotent Stem Cell ,General Immunology and Microbiology ,Gene Expression Profiling ,Human Genome ,Pioneer factor ,Stem Cell Research ,Pediatric Cardiac Genomics Consortium ,030104 developmental biology ,Mutation ,biology.protein ,Congenital Structural Anomalies ,Biochemistry and Cell Biology ,Missense ,FOXA1 ,030217 neurology & neurosurgery ,Epigenesis ,Developmental Biology - Abstract
Damaging GATA6 variants cause cardiac outflow tract defects, sometimes with pancreatic and diaphragmic malformations. To define molecular mechanisms for these diverse developmental defects, we studied transcriptional and epigenetic responses to GATA6 loss of function (LoF) and missense variants during cardiomyocyte differentiation of isogenic human induced pluripotent stem cells. We show that GATA6 is a pioneer factor in cardiac development, regulating SMYD1 that activates HAND2, and KDR that with HAND2 orchestrates outflow tract formation. LoF variants perturbed cardiac genes and also endoderm lineage genes that direct PDX1 expression and pancreatic development. Remarkably, an exon 4 GATA6 missense variant, highly associated with extra-cardiac malformations, caused ectopic pioneer activities, profoundly diminishing GATA4, FOXA1/2, and PDX1 expression and increasing normal retinoic acid signaling that promotes diaphragm development. These aberrant epigenetic and transcriptional signatures illuminate the molecular mechanisms for cardiovascular malformations, pancreas and diaphragm dysgenesis that arise in patients with distinct GATA6 variants.
- Published
- 2020
16. Founder mutation in N-terminus of cardiac troponin I causes malignant hypertrophic cardiomyopathy
- Author
-
Samir Arnaout, Georges Nemer, Ossama K. Abou Hassan, Steven R. DePalma, Manal Batrawi, Fadi Bitar, James S. Ware, Antoine Abchee, Mariam Arabi, Jonathan G. Seidman, Athar Khalil, Akl C. Fahed, Christine E. Seidman, Barbara McDonough, Wellcome Trust, and British Heart Foundation
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,Cardiac troponin ,Adolescent ,030204 cardiovascular system & hematology ,Gene mutation ,medicine.disease_cause ,Sudden cardiac death ,TNNI3 ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,Internal medicine ,medicine ,Humans ,Child ,cardiomyopathy, hypertrophic ,risk ,Mutation ,disease ,business.industry ,Myocardium ,Troponin I ,death, sudden, cardiac ,Hypertrophic cardiomyopathy ,Original Articles ,General Medicine ,Middle Aged ,medicine.disease ,Phenotype ,Founder Effect ,Pedigree ,N-terminus ,030104 developmental biology ,Echocardiography ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Cardiology ,cardiovascular system ,Female ,mutation ,business - Abstract
Supplemental Digital Content is available in the text., Background: Cardiac troponin I (TNNI3) gene mutations account for 3% of hypertrophic cardiomyopathy and carriers have a heterogeneous phenotype, with increased risk of sudden cardiac death (SCD). Only one mutation (p.Arg21Cys) has been reported in the N terminus of the protein. In model organisms, it impairs PKA (protein kinase A) phosphorylation, increases calcium sensitivity, and causes diastolic dysfunction. The phenotype of this unique mutation in patients with hypertrophic cardiomyopathy remains unknown. Methods: We sequenced 29 families with hypertrophic cardiomyopathy enriched for pediatric-onset disease and identified 5 families with the TNNI3 p.Arg21Cys mutation. Using cascade screening, we studied the clinical phenotype of 57 individuals from the 5 families with TNNI3 p.Arg21Cys-related cardiomyopathy. We performed survival analysis investigating the age at first SCD in carriers of the mutation. Results: All 5 families with TNNI3 p.Arg21Cys were from South Lebanon. TNNI3 p.Arg21Cys-related cardiomyopathy manifested a malignant phenotype—SCD occurred in 30 (53%) of 57 affected individuals at a median age of 22.5 years. In select carriers without left ventricular hypertrophy on echocardiogram, SCD occurred, myocyte disarray was found on autopsy heart, and tissue Doppler and cardiac magnetic resonance imaging identified subclinical disease features such as diastolic dysfunction and late gadolinium enhancement. Conclusions: The TNNI3 p.Arg21Cys mutation has a founder effect in South Lebanon and causes malignant hypertrophic cardiomyopathy with early SCD even in the absence of hypertrophy. Genetic diagnosis with this mutation may be sufficient for risk stratification for SCD.
- Published
- 2020
17. BET bromodomain proteins regulate transcriptional reprogramming in genetic dilated cardiomyopathy
- Author
-
Jonathan G. Seidman, Hiroko Wakimoto, Steven R. DePalma, Joshua M. Gorham, James E. Bradner, Christine E. Seidman, Da Young Lee, Andrew Antolic, Saptarsi M. Haldar, Jun Qi, David A. Conner, Michael A. Burke, Madeleine E. Lemieux, Jonathan D. Brown, and Zhe Jiao
- Subjects
0301 basic medicine ,Cardiomyopathy, Dilated ,Male ,BRD4 ,Cardiac fibrosis ,Mutant ,Biology ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Gene expression ,medicine ,Animals ,Gene Regulatory Networks ,Epigenetics ,Mice, Knockout ,Gene Expression Profiling ,Calcium-Binding Proteins ,Nuclear Proteins ,General Medicine ,Azepines ,Triazoles ,medicine.disease ,Fibrosis ,Phospholamban ,Bromodomain ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,cardiovascular system ,Research Article ,Transcription Factors - Abstract
The bromodomain and extraterminal (BET) family comprises epigenetic reader proteins that are important regulators of inflammatory and hypertrophic gene expression in the heart. We previously identified the activation of proinflammatory gene networks as a key early driver of dilated cardiomyopathy (DCM) in transgenic mice expressing a mutant form of phospholamban (PLN(R9C)) — a genetic cause of DCM in humans. We hypothesized that BETs coactivate this inflammatory process, representing a critical node in the progression of DCM. To test this hypothesis, we treated PLN(R9C) or age-matched WT mice longitudinally with the small molecule BET bromodomain inhibitor JQ1 or vehicle. BET inhibition abrogated adverse cardiac remodeling, reduced cardiac fibrosis, and prolonged survival in PLN(R9C) mice by inhibiting expression of proinflammatory gene networks at all stages of disease. Specifically, JQ1 had profound effects on proinflammatory gene network expression in cardiac fibroblasts, while having little effect on gene expression in cardiomyocytes. Cardiac fibroblast proliferation was also substantially reduced by JQ1. Mechanistically, we demonstrated that BRD4 serves as a direct and essential regulator of NF-κB–mediated proinflammatory gene expression in cardiac fibroblasts. Suppressing proinflammatory gene expression via BET bromodomain inhibition could be a novel therapeutic strategy for chronic DCM in humans.
- Published
- 2020
18. EM-mosaic detects mosaic point mutations that contribute to congenital heart disease
- Author
-
Jane W. Newburger, Emily Leann Griffin, Jonathan G. Seidman, Martina Brueckner, Bruce D. Gelb, Alexander Hsieh, Steven R. DePalma, Kathryn B. Manheimer, David M. McKean, Joshua M. Gorham, Jon A. L. Willcox, Deepak Srivastava, Elizabeth Goldmuntz, Christine E. Seidman, George A. Porter, Angela C. Tai, Sarah U. Morton, Daniel Bernstein, Hongjian Qi, Richard P. Lifton, Yufeng Shen, Richard W. Kim, Wendy K. Chung, and Martin Tristani-Firouzi
- Subjects
Proband ,Exome sequencing ,Heart disease ,lcsh:Medicine ,Cardiovascular ,Congenital ,Tissue mosaicism ,2.1 Biological and endogenous factors ,Aetiology ,Child ,Exome ,Genetics (clinical) ,Heart Defects ,Genetics ,screening and diagnosis ,Mosaicism ,Detection ,Heart Disease ,Child, Preschool ,Molecular Medicine ,Heart Defects, Congenital ,Adult ,lcsh:QH426-470 ,Adolescent ,Clinical Sciences ,Biology ,Young Adult ,Clinical Research ,medicine ,Humans ,Point Mutation ,Somatic ,Allele ,Preschool ,Molecular Biology ,Genotyping ,Congenital heart disease ,Research ,Point mutation ,lcsh:R ,Human Genome ,Infant ,medicine.disease ,4.1 Discovery and preclinical testing of markers and technologies ,lcsh:Genetics ,Mosaic ,Software - Abstract
Background The contribution of somatic mosaicism, or genetic mutations arising after oocyte fertilization, to congenital heart disease (CHD) is not well understood. Further, the relationship between mosaicism in blood and cardiovascular tissue has not been determined. Methods We developed a new computational method, EM-mosaic (Expectation-Maximization-based detection of mosaicism), to analyze mosaicism in exome sequences derived primarily from blood DNA of 2530 CHD proband-parent trios. To optimize this method, we measured mosaic detection power as a function of sequencing depth. In parallel, we analyzed our cohort using MosaicHunter, a Bayesian genotyping algorithm-based mosaic detection tool, and compared the two methods. The accuracy of these mosaic variant detection algorithms was assessed using an independent resequencing method. We then applied both methods to detect mosaicism in cardiac tissue-derived exome sequences of 66 participants for which matched blood and heart tissue was available. Results EM-mosaic detected 326 mosaic mutations in blood and/or cardiac tissue DNA. Of the 309 detected in blood DNA, 85/97 (88%) tested were independently confirmed, while 7/17 (41%) candidates of 17 detected in cardiac tissue were confirmed. MosaicHunter detected an additional 64 mosaics, of which 23/46 (50%) among 58 candidates from blood and 4/6 (67%) of 6 candidates from cardiac tissue confirmed. Twenty-five mosaic variants altered CHD-risk genes, affecting 1% of our cohort. Of these 25, 22/22 candidates tested were confirmed. Variants predicted as damaging had higher variant allele fraction than benign variants, suggesting a role in CHD. The estimated true frequency of mosaic variants above 10% mosaicism was 0.14/person in blood and 0.21/person in cardiac tissue. Analysis of 66 individuals with matched cardiac tissue available revealed both tissue-specific and shared mosaicism, with shared mosaics generally having higher allele fraction. Conclusions We estimate that ~ 1% of CHD probands have a mosaic variant detectable in blood that could contribute to cardiac malformations, particularly those damaging variants with relatively higher allele fraction. Although blood is a readily available DNA source, cardiac tissues analyzed contributed ~ 5% of somatic mosaic variants identified, indicating the value of tissue mosaicism analyses.
- Published
- 2020
19. BET Bromodomain Proteins Regulate Transcriptional Reprogramming in Genetic Dilated Cardiomyopathy
- Author
-
Da Young Lee, Saptarsi M. Haldar, David A. Conner, Joshua M. Gorham, Andrew Antolic, James E. Bradner, Christine E. Seidman, Zhe Jiao, Jun Qi, Hiroko Wakimoto, Jonathan D. Brown, Steven R. DePalma, Jonathan G. Seidman, and Michael A. Burke
- Subjects
BET inhibitor ,BRD4 ,Cardiac fibrosis ,Gene expression ,cardiovascular system ,medicine ,Gene regulatory network ,Epigenetics ,Biology ,medicine.disease ,Bromodomain ,Phospholamban ,Cell biology - Abstract
The bromodomain and extraterminal (BET) family of epigenetic reader proteins are key regulators of pathologic gene expression in the heart. Using mice carrying a human mutation in phospholamban (PLNR9C) that develop progressive dilated cardiomyopathy (DCM), we previously identified the activation of inflammatory gene networks as a key early driver of DCM. We reasoned that BETs control this inflammatory process, representing a key node in the progression of genetic DCM. Using a chemical genetic strategy, PLNR9C or age-matched wild type mice were treated longitudinally with the BET inhibitor JQ1 or vehicle. JQ1 abrogated DCM, reduced cardiac fibrosis, and prolonged survival in PLNR9C mice by inhibiting inflammatory gene network expression at all disease stages. Cardiac fibroblast proliferation was also substantially reduced by JQ1. Interestingly, JQ1 had profound effects on pathologic gene network expression in cardiac fibroblasts, while having little effect on transcription in cardiomyocytes. Using co-immunoprecipitation, we identified BRD4 as a direct and essential regulator of NFκB-mediated inflammatory gene transcription in cardiac fibroblasts. In this this model of chronic, heritable DCM, BETs activate inflammatory gene networks in cardiac fibroblasts via an NFκB-dependent mechanism, marking them as critical effectors of pathologic gene expression.
- Published
- 2020
- Full Text
- View/download PDF
20. Genome-wide assessment for genetic variants associated with ventricular dysfunction after primary coronary artery bypass graft surgery.
- Author
-
Amanda A Fox, Mias Pretorius, Kuang-Yu Liu, Charles D Collard, Tjorvi E Perry, Stanton K Shernan, Philip L De Jager, David A Hafler, Daniel S Herman, Steven R DePalma, Dan M Roden, Jochen D Muehlschlegel, Brian S Donahue, Dawood Darbar, J G Seidman, Simon C Body, and Christine E Seidman
- Subjects
Medicine ,Science - Abstract
Postoperative ventricular dysfunction (VnD) occurs in 9-20% of coronary artery bypass graft (CABG) surgical patients and is associated with increased postoperative morbidity and mortality. Understanding genetic causes of postoperative VnD should enhance patient risk stratification and improve treatment and prevention strategies. We aimed to determine if genetic variants associate with occurrence of in-hospital VnD after CABG surgery.A genome-wide association study identified single nucleotide polymorphisms (SNPs) associated with postoperative VnD in male subjects of European ancestry undergoing isolated primary CABG surgery with cardiopulmonary bypass. VnD was defined as the need for ≥2 inotropes or mechanical ventricular support after CABG surgery. Validated SNPs were assessed further in two replication CABG cohorts and meta-analysis was performed.Over 100 SNPs were associated with VnD (P2.1) of developing in-hospital VnD after CABG surgery. However, three genetic loci identified by meta-analysis were more modestly associated with development of postoperative VnD. Studies of larger cohorts to assess these loci as well as to define other genetic mechanisms and related biology that link genetic variants to postoperative ventricular dysfunction are warranted.
- Published
- 2011
- Full Text
- View/download PDF
21. Contribution of rare inherited and de novo variants in 2,871 congenital heart disease probands
- Author
-
Cecelia W. Lo, Stephen Sanders, Sarah U. Morton, Irina R. Tikhonoa, Samir Zaidi, Elizabeth Goldmuntz, Hongjian Qi, Richard B. Kim, Jonathan R. Kaltman, Jonathan G. Seidman, Xue Zeng, Jason Homsy, George A. Porter, W. Scott Watkins, Deepak Srivastava, Weni Chang, Martin Tristani-Firouzi, Seema Mital, James R. Knight, Qiongshi Lu, Steven R. DePalma, John E. Deanfield, Christopher Castaldi, J. William Gaynor, Yufeng Shen, Bruce D. Gelb, Mark W. Russell, Richard P. Lifton, Alessandro Giardini, Kaya Bilguvar, Wendy K. Chung, Jane W. Newburger, H. Joseph Yost, Sheng Chih Jin, Mark Yandell, Martina Brueckner, Shrikant Mane, Robert D. Bjornson, Wei Chien Hung, Amy E. Roberts, Junhui Zhang, Christine E. Seidman, Michael C. Sierant, Hongyu Zhao, and Shozeb Haider
- Subjects
Heart Defects, Congenital ,Risk ,Adult ,Male ,0301 basic medicine ,Proband ,Heterozygote ,Heart disease ,Gene Expression ,Genome-wide association study ,Biology ,Medical and Health Sciences ,Article ,Growth Differentiation Factor 1 ,Congenital ,03 medical and health sciences ,Genotype ,Genetics ,medicine ,Humans ,Genetic Predisposition to Disease ,Exome ,cardiovascular diseases ,Autistic Disorder ,Child ,Exome sequencing ,Heart Defects ,Tetralogy of Fallot ,Myosin Heavy Chains ,Homozygote ,Case-control study ,High-Throughput Nucleotide Sequencing ,Biological Sciences ,Vascular Endothelial Growth Factor Receptor-3 ,medicine.disease ,Pedigree ,3. Good health ,Editorial ,030104 developmental biology ,Case-Control Studies ,Mutation ,Female ,Cardiac Myosins ,Genome-Wide Association Study ,Developmental Biology - Abstract
Congenital heart disease (CHD) is the leading cause of mortality from birth defects. Here, exome sequencing of a single cohort of 2,871 CHD probands, including 2,645 parent-offspring trios, implicated rare inherited mutations in 1.8%, including a recessive founder mutation in GDF1 accounting for ∼5% of severe CHD in Ashkenazim, recessive genotypes in MYH6 accounting for ∼11% of Shone complex, and dominant FLT4 mutations accounting for 2.3% of Tetralogy of Fallot. De novo mutations (DNMs) accounted for 8% of cases, including ∼3% of isolated CHD patients and ∼28% with both neurodevelopmental and extra-cardiac congenital anomalies. Seven genes surpassed thresholds for genome-wide significance, and 12 genes not previously implicated in CHD had >70% probability of being disease related. DNMs in ∼440 genes were inferred to contribute to CHD. Striking overlap between genes with damaging DNMs in probands with CHD and autism was also found.
- Published
- 2017
- Full Text
- View/download PDF
22. Abstract 785: Modeling Congenital Heart Disease-Associated Variants in GATA6 Using CRISPR/Cas9 and Human Induced Pluripotent Stem Cells
- Author
-
Seongwon Kim, Joshua M. Gorham, Christopher N. Toepfer, David A. Conner, Jon A. L. Willcox, Lauren K. Wasson, Christine E. Seidman, Megan Jang, Daniel M. DeLaughter, Tarsha Ward, Steven R. DePalma, Alexandre C. Pereira, Angela Tai, Manuel Schmid, Sarah U. Morton, Meraj Neyazi, Arun Sharma, Yuri Kim, Benoit G. Bruneau, Jon G. Seidman, and Radhika Agarwal
- Subjects
Genetics ,endocrine system ,GATA6 ,Heart disease ,Physiology ,Biology ,Gene mutation ,medicine.disease ,medicine ,CRISPR ,Identification (biology) ,Human Induced Pluripotent Stem Cells ,Cardiology and Cardiovascular Medicine ,Stem cell biology ,Exome sequencing - Abstract
The discovery of damaging gene mutations in congenital heart disease (CHD) patients enables identification of regulators of cardiac development. Exome sequencing identified de novo heterozygous loss-of-function (LoF) and missense variants in GATA6 among CHD probands, most with outflow tract malformations. Other subjects with GATA6 LoF mutations developed pancreatic agenesis. To elucidate the molecular basis for the predominance of this heart defect, we modeled GATA6 mutations in cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). GATA6 variants were introduced into isogenic hiPSCs using CRISPR/Cas9 genome editing. Genome-wide molecular profiles including chromatin accessibility (ATAC-Seq) and gene expression (single cell and bulk RNA-Seq) were evaluated during hiPSC-CM differentiation. Analyses of GATA6 mutant hiPSC-CMs showed deficits in hiPSC-CM differentiation, chromatin accessibility and transcriptional profiles. Heterozygous GATA6 LoF hiPSCs made hiPSC-CMs but exhibited reduced expression of second heart field genes. Homozygous GATA6 LoF hiPSCs failed to differentiate and adopted fibroblast expression profiles. hiPSCs carrying a homozygous GATA6 missense variant, R456G, which altered a DNA-binding domain residue, showed enhanced capacity to differentiate into neuroepithelial-like cells. Chromatin-accessibility studies confirmed that GATA6 normally binds to genes in the promoter region and other genes at distal enhancers. Human GATA6 haploinsufficiency disrupts developmental transcriptional responses driving cardiac morphogenesis. The HAND2 -dependent genetic program, operant during outflow tract development, is particularly sensitive to GATA6 dosage. The mixed differentiation patterns observed in mutation-carrying hiPSCs likely contributes to vascular phenotypes observed in CHD patients. GATA6 haploinsufficiency preferentially alters binding of distal enhancers to promoters in genes where GATA6 normally binds the enhancer rather than the promoter. We speculate that pathogenicity of GATA6 haploinsufficiency is mediated by weaker binding of GATA6 to distal enhancers than to promoter elements, altering expression of these genes in GATA6 haploinsufficient patients.
- Published
- 2019
- Full Text
- View/download PDF
23. Abstract 104: Identification and Characterization of a Titin Enhancer using CRISPR/Cas9 Genome Editing and hiPSC-Derived Cardiomyocytes
- Author
-
Seong Won Kim, Yuri Kim, Arun Sharma, Jonathan G. Seidman, Joshua M. Gorham, Lauren K. Wasson, Steven R. DePalma, Christine E. Seidman, Jon A. L. Willcox, Daniel M. DeLaughter, Manuel Schmid, Christopher N. Toepfer, Radhika Agarwal, Angela Tai, and Meraj Neyazi
- Subjects
biology ,Physiology ,Cardiomyopathy ,Dilated cardiomyopathy ,Computational biology ,medicine.disease ,Genome editing ,Heart failure ,medicine ,biology.protein ,CRISPR ,Titin ,Identification (biology) ,Cardiology and Cardiovascular Medicine ,Enhancer - Abstract
Dilated cardiomyopathy (DCM) is a leading cause for heart failure and is associated with a rate of mortality of 20% within 5 years of diagnosis. The most common genetic causes for DCM are mutations of the sarcomere protein titin (encoded by TTN ), which occurs in 10-20% of DCM cases. Dominant DCM mutations truncate titin (TTNtv) and result in haploinsufficiency. Thus, strategies to increase the expression of the wild type TTN allele could attenuate damaging effects of TTNtv. Utilizing bioinformatic tools, we identified a putative enhancer for TTN in its intron 1. We deleted a 658 bp region from intron 1 which encompasses the region of interest in human induced pluripotent stem cells (hiPSCs) using CRISPR/Cas9 genome editing to validate its function. Utilizing RNA sequencing and qPCR of RNA harvested from hiPSC-derived cardiomyocytes (hiPSC-CMs), we demonstrated that a homozygous deletion in this region leads to a drop in TTN expression compared to the wild type (WT) control (0.344-fold change, p < 0.001). To further characterize this region, we subdivided it into three parts which we called E1 (296 bp), E2 (206 bp), and E3 (139 bp). E1 includes a highly conserved region and a region of open chromatin as identified by the Assay for Transposase-Accessible Chromatin Sequencing (ATAC-Seq) performed on hiPSC-CMs. A homozygous E1 deletion resulted in a decreased TTN expression of 0.63-fold compared to the WT control (p < 0.001) when performing RNA sequencing on hiPSC-CMs. Both homozygous E2 and E3 deletions resulted in an increased TTN expression (1.56-fold change, p < 0.001; 1.19 fold change, p < 0.001). Utilizing a published sarcomere tracking platform, SarcTrack, to investigate hiPSC-CM physiology, we saw a decreased contractility of 6.6% in hiPSC-CMs carrying a homozygous E1 deletion compared to 10.1% in the WT control (p < 0.001). Cells carrying homozygous E2 or E3 deletions were hypercontractile (13.8%, p < 0.001; 13.7%, p < 0.001). Given our results, we hypothesize that TTN expression depends on the E1 region. If confirmed, we expect that increasing the activity of this enhancer using small molecules may provide a novel therapeutic target for DCM caused by TTNtv.
- Published
- 2019
- Full Text
- View/download PDF
24. Abstract 202: The R21C Mutation in Troponin I Has a Founder Effect in South Lebanon and Causes Malignant Hypertrophic Cardiomyopathy
- Author
-
Samir Arnaout, Mariam Arabi, Athar Khalil, Jonathan G. Seidman, Akl C. Fahed, Steven R. DePalma, Christine E. Seidman, Manal Batrawi, James S. Ware, Fadi Bitar, Georges Nemer, Antoine Abche, and Barbara McDonough
- Subjects
medicine.medical_specialty ,Mutation ,Physiology ,business.industry ,Cardiomyopathy ,Hypertrophic cardiomyopathy ,macromolecular substances ,medicine.disease ,medicine.disease_cause ,Sarcomere ,Sudden cardiac death ,Wide phenotypic variability ,Internal medicine ,Troponin I ,cardiovascular system ,medicine ,Cardiology ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,business ,Founder effect - Abstract
Hypertrophic Cardiomyopathy (HCM) occurs in 1 of every 500 people and has a wide phenotypic variability. In the majority of cases, HCM is caused by known mutations in genes that code for sarcomere proteins. Although gene testing is widely available for HCM, knowing the phenotype caused by different gene mutations remains a challenging task. We recruited 28 families with HCM, of which 19 (67.8%) have at least one patient with pediatric onset. Index patients from 20 families received targeted sequencing for a panel of genes including TNNI3 , and 7 families received Sanger sequencing for the TNNI3 . We identified a missense mutation p.R21C in TNNI3 segregating with HCM in four families from South Lebanon. Through cascade screening, we identified 30 patients from the four families; twenty of them (67%) had a clinical diagnosis of HCM with a median age of 37 years, while 9 (30%), with a median age 21 years, had no evidence of HCM on echocardiography. An additional 27 members of the families had evidence of HCM, including 22 with SCD in the setting of no past medical history, and their carrier status for p.R21C was implied from the pedigrees. Survival analysis for 57 HCM patients with the mutation revealed a markedly decreased age at first adverse event as compared to 47 HCM patients with the MYBPC3 p.R502W mutation. Founder mutations in HCM that cause a severe phenotype are uncommon. The p.R21C mutation in TNNI3 is the first HCM mutation described in the Lebanese population and has a founder effect in South Lebanon. Early and more frequent screening with different imaging modalities as well as tailored management might be warranted for carriers of this mutation.
- Published
- 2019
- Full Text
- View/download PDF
25. THSD1 (Thrombospondin Type 1 Domain Containing Protein 1) Mutation in the Pathogenesis of Intracranial Aneurysm and Subarachnoid Hemorrhage
- Author
-
Ming-Sum Lee, Morgan L. Hennessy, Barbara McDonough, Calum A. MacRae, Christine E. Seidman, Georgene W. Hergenroeder, Xiaoqian Fang, Sarah M. Colosimo, Susan M. Dymecki, Steven R. DePalma, Dong H. Kim, John P. Hagan, Stephen V. Nalbach, Krista J. Qualmann, Kyla J. Patek, Teresa Santiago-Sim, Jonathan G. Seidman, Steven C. Greenway, and Dianna M. Milewicz
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Subarachnoid hemorrhage ,Aneurysm, Ruptured ,Pathogenesis ,Mice ,03 medical and health sciences ,Aneurysm ,Animal Disease Models ,medicine ,Animals ,Humans ,Exome ,Genetic Predisposition to Disease ,Thrombospondins ,Zebrafish ,Advanced and Specialized Nursing ,Thrombospondin ,business.industry ,Intracranial Aneurysm ,Subarachnoid Hemorrhage ,Zebrafish Proteins ,medicine.disease ,Pedigree ,Disease Models, Animal ,030104 developmental biology ,Codon, Nonsense ,Mutation (genetic algorithm) ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background and Purpose— A ruptured intracranial aneurysm (IA) is the leading cause of a subarachnoid hemorrhage. This study seeks to define a specific gene whose mutation leads to disease. Methods— More than 500 IA probands and 100 affected families were enrolled and clinically characterized. Whole exome sequencing was performed on a large family, revealing a segregating THSD1 (thrombospondin type 1 domain containing protein 1) mutation. THSD1 was sequenced in other probands and controls. Thsd1 loss-of-function studies in zebrafish and mice were used for in vivo analyses and functional studies performed using an in vitro endothelial cell model. Results— A nonsense mutation in THSD1 was identified that segregated with the 9 affected (3 suffered subarachnoid hemorrhage and 6 had unruptured IA) and was absent in 13 unaffected family members (LOD score 4.69). Targeted THSD1 sequencing identified mutations in 8 of 507 unrelated IA probands, including 3 who had suffered subarachnoid hemorrhage (1.6% [95% confidence interval, 0.8%–3.1%]). These THSD1 mutations/rare variants were highly enriched in our IA patient cohort relative to 89 040 chromosomes in Exome Aggregation Consortium (ExAC) database ( P THSD1 loss impaired endothelial cell focal adhesion to the basement membrane. These adhesion defects could be rescued by expression of wild-type THSD1 but not THSD1 mutants identified in IA patients. Conclusions— This report identifies THSD1 mutations in familial and sporadic IA patients and shows that THSD1 loss results in cerebral bleeding in 2 animal models. This finding provides new insight into IA and subarachnoid hemorrhage pathogenesis and provides new understanding of THSD1 function, which includes endothelial cell to extracellular matrix adhesion.
- Published
- 2016
- Full Text
- View/download PDF
26. Rare genetic variation at transcription factor binding sites modulates local DNA methylation profiles
- Author
-
Daniel Bernstein, Steven R. DePalma, George A. Porter, Alejandro Martin-Trujillo, Bruce D. Gelb, Sarah U. Morton, Nihir Patel, Bharati Jadhav, Elizabeth Goldmuntz, David M. McKean, Felix Richter, Paras Garg, Martin Tristani-Firouzi, Jonathan G. Seidman, Christine E. Seidman, Alessandro Giardini, Jane W. Newburger, Wendy K. Chung, Andrew J. Sharp, Dorota Gruber, and Richard B. Kim
- Subjects
Male ,Cancer Research ,QH426-470 ,Biochemistry ,Epigenesis, Genetic ,Cohort Studies ,Database and Informatics Methods ,0302 clinical medicine ,Child ,Genetics (clinical) ,Genetics ,0303 health sciences ,DNA methylation ,Chemical Reactions ,Genomics ,Methylation ,Middle Aged ,Chromatin ,Nucleic acids ,Chemistry ,CpG site ,Child, Preschool ,Physical Sciences ,Chromatin Immunoprecipitation Sequencing ,Epigenetics ,Female ,DNA modification ,Sequence Analysis ,Chromatin modification ,Research Article ,Chromosome biology ,Adult ,Heart Defects, Congenital ,Cell biology ,Adolescent ,Bioinformatics ,Biology ,Research and Analysis Methods ,Polymorphism, Single Nucleotide ,DNA sequencing ,Young Adult ,03 medical and health sciences ,Sequence Motif Analysis ,Gene Disruption ,Humans ,Molecular Biology ,Gene ,Ecology, Evolution, Behavior and Systematics ,030304 developmental biology ,Evolutionary Biology ,Binding Sites ,Biology and life sciences ,Population Biology ,Whole Genome Sequencing ,Genome, Human ,Infant, Newborn ,Infant ,DNA ,DNA binding site ,Genetic Polymorphism ,CpG Islands ,Gene expression ,Population Genetics ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Although DNA methylation is the best characterized epigenetic mark, the mechanism by which it is targeted to specific regions in the genome remains unclear. Recent studies have revealed that local DNA methylation profiles might be dictated by cis-regulatory DNA sequences that mainly operate via DNA-binding factors. Consistent with this finding, we have recently shown that disruption of CTCF-binding sites by rare single nucleotide variants (SNVs) can underlie cis-linked DNA methylation changes in patients with congenital anomalies. These data raise the hypothesis that rare genetic variation at transcription factor binding sites (TFBSs) might contribute to local DNA methylation patterning. In this work, by combining blood genome-wide DNA methylation profiles, whole genome sequencing-derived SNVs from 247 unrelated individuals along with 133 predicted TFBS motifs derived from ENCODE ChIP-Seq data, we observed an association between the disruption of binding sites for multiple TFs by rare SNVs and extreme DNA methylation values at both local and, to a lesser extent, distant CpGs. While the majority of these changes affected only single CpGs, 24% were associated with multiple outlier CpGs within ±1kb of the disrupted TFBS. Interestingly, disruption of functionally constrained sites within TF motifs lead to larger DNA methylation changes at nearby CpG sites. Altogether, these findings suggest that rare SNVs at TFBS negatively influence TF-DNA binding, which can lead to an altered local DNA methylation profile. Furthermore, subsequent integration of DNA methylation and RNA-Seq profiles from cardiac tissues enabled us to observe an association between rare SNV-directed DNA methylation and outlier expression of nearby genes. In conclusion, our findings not only provide insights into the effect of rare genetic variation at TFBS on shaping local DNA methylation and its consequences on genome regulation, but also provide a rationale to incorporate DNA methylation data to interpret the functional role of rare variants., Author summary One of the major challenges for human genetics in the post-genomic era is to interpret the functional relevance of genetic variation. Quantitative trait locus (QTL) analyses have associated an important fraction of genetic variants with a wide range of molecular phenotypes including gene expression (eQTL) and DNA methylation (meQTL), providing insights into the mechanisms by which genetic variation can contribute to health and disease. Although QTL mapping represents an excellent approach to identify biologically relevant functional variants, these studies have been mainly focused on common variants and do not include low-frequency and rare variants. Here, we observed that rare regulatory variants, i.e, single nucleotide variants (SNVs) that disrupt transcription factor binding sites (TFBSs), are associated with changes in DNA methylation at both local and, to a lesser extent, broader locations, most likely, by altering the DNA-binding affinity of transcription factors (TFs). Interestingly, we have also shown that this change in DNA methylation can alter expression levels of nearby genes. Overall, these data suggest a role of rare regulatory SNVs in shaping DNA methylation, and suggest that the incorporation of DNA methylation data may help to interpret the functional consequences of human genetic variation.
- Published
- 2020
- Full Text
- View/download PDF
27. SEQUENCE VARIANTS IN TITIN CAUSING SPLICING DEFECTS AND CARDIOMYOPATHY: INSIGHTS FOR GENE BASED DIAGNOSIS AND NORMAL PHYSIOLOGY
- Author
-
Parth Patel, Joshua M. Gorham, Christine E. Seidman, Barbara McDonough, Alireza Haghighi, Diane Fatkin, Arun Sharma, Jon A. L. Willcox, Jonathan G. Seidman, Steven R. DePalma, Kaoru Ito, Lien Lam, and Renee Johnson
- Subjects
Genetics ,biology ,business.industry ,Cardiomyopathy ,Dilated cardiomyopathy ,musculoskeletal system ,medicine.disease ,RNA splicing ,cardiovascular system ,biology.protein ,medicine ,Titin ,Cardiology and Cardiovascular Medicine ,business ,Gene ,Sequence (medicine) - Abstract
Heterozygous truncating variants in titin (TTNtv) are the major genetic cause of dilated cardiomyopathy (DCM). Though variants which disrupt essential splicing dinucleotides (GT/AG) are readily recognized as TTNtv, the effects of other nearby sequence variations on splicing is uncertain. We
- Published
- 2020
- Full Text
- View/download PDF
28. Abstract 231: Identification of a Titin Enhancer using hiPSC-Derived Cardiomyocytes and CRISPR/Cas9 Genome Editing
- Author
-
Arun Sharma, Jonathan G. Seidman, Seong Won Kim, Steven R. DePalma, Manuel Schmid, Meraj Neyazi, Lauren K. Wasson, Joshua M. Gorham, and Christine E. Seidman
- Subjects
biology ,Physiology ,Cardiomyopathy ,Dilated cardiomyopathy ,Computational biology ,medicine.disease ,Genome editing ,Heart failure ,medicine ,biology.protein ,CRISPR ,Identification (biology) ,Titin ,Cardiology and Cardiovascular Medicine ,Enhancer - Abstract
Dilated cardiomyopathy (DCM), a disorder that occurs in 1:250 individuals, is associated with rates of mortality of 20% within 5 years of diagnosis and is a leading cause for heart failure and cardiac transplantation. Mutations in the massive sarcomere protein titin (encoded by TTN ) are the most common genetic cause of DCM, occurring in 10-20% of cases. As dominant DCM mutations truncate titin (TTNtv) and result in haploinsufficiency, we predict that strategies to increase the expression of the wild type (WT) TTN allele might attenuate the damaging effects of TTNtv. We used bioinformatic analyses to identify a putative TTN enhancer within intron 1. To confirm its function, we deleted 658 bp from intron 1 that encompasses the putative TTN enhancer in human induced pluripotent stem cells (hiPSCs) using CRISPR/Cas9 genome editing. We used qPCR and RNA sequencing of RNA harvested from hiPSC-derived cardiomyocytes (hiPSC-CMs) and demonstrated that a homozygous deletion in this region leads to decreased TTN gene expression compared to the WT control (0.344 fold change, p < 0.001), and also to decreased expression of other sarcomeric genes such as TNNT2 (0.074 fold change, p < 0.001), MYH6 (0.18 fold change, p < 0.001), MYH7 (0.008 fold change, p < 0.001), and ACTN2 (0.118 fold change, p < 0.001). The expression of transcription factors (TF) that have binding sites in this region is also affected, such as MEIS2 (0.4 fold change, p < 0.001) and KLF6 (0.33 fold change, p < 0.001), both of which are known to be involved in cardiogenesis. These TF may act as a link between the deletion in TTN Intron 1 and a decreased transcription of other cardiac-relevant genes. We also utilized Assay for Transposase-Accessible Chromatin Sequencing (ATAC-Seq) on WT hiPSC-CMs to identify open regions of chromatin that are accessible for TF binding. This provided additional evidence that this 658 bp region in TTN intron 1 has enhancer activity. Ongoing studies are aiming to refine the TTN Intron 1 enhancer by further studies using CRISPR/Cas9, CRISPR droplet sequencing (CROP-Seq), and luciferase-based enhancer activity assays. If confirmed, we expect that increasing the activity of this 658 bp region with small molecules may provide a novel therapeutic target for DCM caused by TTNtv.
- Published
- 2018
- Full Text
- View/download PDF
29. 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
30. De novo mutations in congenital heart disease with neurodevelopmental and other congenital anomalies
- Author
-
Josh Gorham, David M. McKean, Stephen Sanders, Elizabeth Goldmuntz, Francesc López-Giráldez, Angela Romano-Adesman, Kaya Bilguvar, James S. Ware, Jonathan G. Seidman, Mark J. Daly, Amy E. Roberts, Konrad J. Karczewski, J. William Gaynor, Richard P. Lifton, Christine E. Seidman, Mark W. Russell, Hongjian Qi, Steven R. DePalma, Jonathan R. Kaltman, Michael Ronemus, Badri N. Vardarajan, Alessandro Giardini, Ivan Iossifov, Roger E. Breitbart, Jason Homsy, Shrikant Mane, Richard B. Kim, Wendy K. Chung, George A. Porter, Samir Zaidi, Hiroko Wakimoto, Jane W. Newburger, Bruce D. Gelb, Kaitlin E. Samocha, Lijiang Ma, Martina Brueckner, Yufeng Shen, Seema Mital, John E. Deanfield, Sheng Chih Jin, and Irina Tikhonova
- Subjects
Genetics ,0303 health sciences ,Mutation ,Multidisciplinary ,Heart disease ,030204 cardiovascular system & hematology ,Biology ,Bioinformatics ,medicine.disease_cause ,medicine.disease ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,RNA splicing ,medicine ,Transcriptional regulation ,Chromatin modification ,cardiovascular diseases ,Gene ,De novo mutations ,Exome sequencing ,030304 developmental biology - Abstract
Congenital heart disease (CHD) patients have an increased prevalence of extracardiac congenital anomalies (CAs) and risk of neurodevelopmental disabilities (NDDs). Exome sequencing of 1213 CHD parent-offspring trios identified an excess of protein-damaging de novo mutations, especially in genes highly expressed in the developing heart and brain. These mutations accounted for 20% of patients with CHD, NDD, and CA but only 2% of patients with isolated CHD. Mutations altered genes involved in morphogenesis, chromatin modification, and transcriptional regulation, including multiple mutations in RBFOX2, a regulator of mRNA splicing. Genes mutated in other cohorts examined for NDD were enriched in CHD cases, particularly those with coexisting NDD. These findings reveal shared genetic contributions to CHD, NDD, and CA and provide opportunities for improved prognostic assessment and early therapeutic intervention in CHD patients.
- Published
- 2015
- Full Text
- View/download PDF
31. Using Next-generation RNA Sequencing to Examine Ischemic Changes Induced by Cold Blood Cardioplegia on the Human Left Ventricular Myocardium Transcriptome
- Author
-
David M. McKean, Steven R. DePalma, Joshua M. Gorham, Christine E. Seidman, Mahyar Heydarpour, Erica Mazaika, Stanton K. Shernan, Danos C. Christodoulou, Sary F. Aranki, Simon C. Body, Jon G. Seidman, Tjorvi E. Perry, Amanda A. Fox, Grace M. Lee, and Jochen D. Muehlschlegel
- Subjects
Male ,medicine.medical_specialty ,Myocardial ischemia ,Heart Ventricles ,Myocardial Ischemia ,Ischemia ,Article ,law.invention ,Transcriptome ,Downregulation and upregulation ,law ,Internal medicine ,Cardiopulmonary bypass ,Humans ,Medicine ,Blood cardioplegia ,Aged ,Aged, 80 and over ,Sequence Analysis, RNA ,business.industry ,Myocardium ,RNA ,medicine.disease ,Cold Temperature ,Anesthesiology and Pain Medicine ,Heart Arrest, Induced ,Left ventricular myocardium ,Cardiology ,Female ,business - Abstract
Background: The exact mechanisms that underlie the pathological processes of myocardial ischemia in humans are unclear. Cardiopulmonary bypass with cardioplegic arrest allows the authors to examine the whole transcriptional profile of human left ventricular myocardium at baseline and after exposure to cold cardioplegia-induced ischemia as a human ischemia model. Methods: The authors obtained biopsies from 45 patients undergoing aortic valve replacement surgery at baseline and after an average of 79 min of cold cardioplegic arrest. Samples were RNA sequenced and analyzed with the Partek® Genomics Suite (Partek Inc., St. Louis, MO) for differential expression. Ingenuity Pathway Analysis (Ingenuity Systems, Redwood City, CA) and Biobase ExPlain (Biobase GmbH, Wolfenbuettel, Germany) systems were used for functional and pathway analyses. Results: Of the 4,098 genes with a mean expression value greater than 5, 90% were down-regulated and 9.1% were up-regulated. Of those, 1,241 were significantly differentially expressed. Gene ontology analysis revealed significant down-regulation in immune inflammatory response and complement activation categories and highly consistent was the down-regulation of intelectin 1, proteoglycan, and secretory leukocyte peptidase inhibitor. Up-regulated genes of interest were FBJ murine osteosarcoma viral oncogene homolog and the hemoglobin genes hemoglobin α1 (HBA1) and hemoglobin β. In addition, analysis of transcription factor–binding sites revealed interesting targets in factors regulating reactive oxygen species production, apoptosis, immunity, cytokine production, and inflammatory response. Conclusions: The authors have shown that the human left ventricle exhibits significant changes in gene expression in response to cold cardioplegia-induced ischemia during cardiopulmonary bypass, which provides great insight into the pathophysiology of ventricular ischemia, and thus, may help guide efforts to reduce myocardial damage during surgery.
- Published
- 2015
- Full Text
- View/download PDF
32. Increased Frequency of De Novo Copy Number Variants in Congenital Heart Disease by Integrative Analysis of Single Nucleotide Polymorphism Array and Exome Sequence Data
- Author
-
Jason Homsy, Elizabeth Goldmuntz, Stephen Sanders, Ryan Golhar, Kaoru Ito, Badri N. Vardarajan, Richard P. Lifton, Peter White, Wendy K. Chung, Michael Ronemus, Matthew W. State, Laura Rodriguez-Murillo, Steven R. DePalma, Jeremy Leipzig, Dorothy Warburton, Hakon Hakonarson, Menachem Fromer, Ivan Iossifov, Alexander G. Bick, Boris Yamrom, Jonathan R. Kaltman, Erica Mazaika, Christine E. Seidman, Martina Brueckner, Jonathan G. Seidman, Michael J. Italia, A. Jeremy Willsey, Bruce D. Gelb, Yufeng Shen, and Joseph T. Glessner
- Subjects
DNA copy number variations ,single nucleotide ,Physiology ,Molecular Sequence Data ,Clinical Sciences ,Single-nucleotide polymorphism ,Genomics ,Cardiorespiratory Medicine and Haematology ,Biology ,Cardiovascular ,polymorphism ,law.invention ,Cohort Studies ,Pathogenesis ,Congenital ,Gene Frequency ,law ,genomics ,Genetics ,Humans ,2.1 Biological and endogenous factors ,Gene Regulatory Networks ,Exome ,Copy-number variation ,Aetiology ,Exome sequencing ,Polymerase chain reaction ,Heart Defects ,Pediatric ,Human Genome ,Odds ratio ,Heart Disease ,Cardiovascular System & Hematology ,Case-Control Studies ,Congenital Structural Anomalies ,microarray analysis ,Cardiology and Cardiovascular Medicine - Abstract
Rationale : Congenital heart disease (CHD) is among the most common birth defects. Most cases are of unknown pathogenesis. Objective : To determine the contribution of de novo copy number variants (CNVs) in the pathogenesis of sporadic CHD. Methods and Results : We studied 538 CHD trios using genome-wide dense single nucleotide polymorphism arrays and whole exome sequencing. Results were experimentally validated using digital droplet polymerase chain reaction. We compared validated CNVs in CHD cases with CNVs in 1301 healthy control trios. The 2 complementary high-resolution technologies identified 63 validated de novo CNVs in 51 CHD cases. A significant increase in CNV burden was observed when comparing CHD trios with healthy trios, using either single nucleotide polymorphism array ( P =7×10 −5 ; odds ratio, 4.6) or whole exome sequencing data ( P =6×10 −4 ; odds ratio, 3.5) and remained after removing 16% of de novo CNV loci previously reported as pathogenic ( P =0.02; odds ratio, 2.7). We observed recurrent de novo CNVs on 15q11.2 encompassing CYFIP1, NIPA1 , and NIPA2 and single de novo CNVs encompassing DUSP1, JUN, JUP, MED15, MED9, PTPRE SREBF1, TOP2A , and ZEB2 , genes that interact with established CHD proteins NKX2-5 and GATA4 . Integrating de novo variants in whole exome sequencing and CNV data suggests that ETS1 is the pathogenic gene altered by 11q24.2-q25 deletions in Jacobsen syndrome and that CTBP2 is the pathogenic gene in 10q subtelomeric deletions. Conclusions : We demonstrate a significantly increased frequency of rare de novo CNVs in CHD patients compared with healthy controls and suggest several novel genetic loci for CHD.
- Published
- 2014
- Full Text
- View/download PDF
33. Increased Burden of Cardiovascular Disease in Carriers of APOL1 Genetic Variants
- Author
-
Steven R. DePalma, Kaoru Ito, Christopher Newton-Cheh, Jason Flannick, Jonathan G. Seidman, Namrata Gupta, David J. Friedman, Sekar Kathiresan, James G. Wilson, Alexander G. Bick, Martin R. Pollak, Giulio Genovese, Christine E. Seidman, Stacey Gabriel, Michael Parfenov, David Altshuler, Ervin R. Fox, Joel N. Hirschhorn, and Herman A. Taylor
- Subjects
Genetics ,education.field_of_study ,medicine.medical_specialty ,biology ,Physiology ,Apolipoprotein L1 ,business.industry ,Population ,Disease ,medicine.disease ,Internal medicine ,Genotype ,Genetic variation ,Epidemiology ,biology.protein ,Medicine ,Allele ,Cardiology and Cardiovascular Medicine ,education ,business ,Kidney disease - Abstract
Rationale: Two distinct alleles in the gene encoding apolipoprotein L1 ( APOL1 ), a major component of high-density lipoprotein, confer protection against Trypanosoma brucei rhodesiense infection and also increase risk for chronic kidney disease. Approximately 14% of Americans with African ancestry carry 2 APOL1 risk alleles, accounting for the high chronic kidney disease burden in this population. Objective: We tested whether APOL1 risk alleles significantly increase risk for atherosclerotic cardiovascular disease (CVD) in African Americans. Methods and Results: We sequenced APOL1 in 1959 randomly selected African American participants in the Jackson Heart Study (JHS) and evaluated associations between APOL1 genotypes and renal and cardiovascular phenotypes. Previously identified association between APOL1 genotypes and chronic kidney disease was confirmed ( P =2.4×10 −6 ). Among JHS participants with 2 APOL1 risk alleles, we observed increased risk for CVD (50/763 events among participants without versus 37/280 events among participants with 2 risk alleles; odds ratio, 2.17; P =9.4×10 −4 ). We replicated this novel association of APOL1 genotype with CVD in Women’s Health Initiative (WHI) participants (66/292 events among participants without versus 37/101 events among participants with 2 risk alleles; odds ratio, 1.98; P =8.37×10 −3 ; JHS and WHI combined, P =8.5×10 −5 ; odds ratio, 2.12). The increased risk for CVD conferred by APOL1 alleles was robust to correction for both traditional CVD risk factors and chronic kidney disease. Conclusions: APOL1 variants contribute to atherosclerotic CVD risk, indicating a genetic component to cardiovascular health disparities in individuals of African ancestry. The considerable population of African Americans with 2 APOL1 risk alleles may benefit from intensive interventions to reduce CVD.
- Published
- 2014
- Full Text
- View/download PDF
34. <scp>HOXA</scp>2Haploinsufficiency in Dominant Bilateral Microtia and Hearing Loss
- Author
-
Lourdes Quintanilla-Dieck, Christine E. Seidman, Maria A. Artunduaga, Roland D. Eavey, Gabriel Osorno, Steven R. DePalma, Jonathan G. Seidman, Patricia Jarrin, Lucas M. Viana, Cecilia C. Helwig, Barbara McDonough, and Kerry K. Brown
- Subjects
Male ,Nonsynonymous substitution ,Hearing loss ,media_common.quotation_subject ,Nonsense ,Haploinsufficiency ,Biology ,Article ,Congenital Abnormalities ,Genetics ,medicine ,Humans ,Exome ,Ear, External ,Hearing Loss ,Genetics (clinical) ,Exome sequencing ,Congenital Microtia ,Genes, Dominant ,media_common ,Homeodomain Proteins ,Microtia ,High-Throughput Nucleotide Sequencing ,Autosomal dominant trait ,Ear ,medicine.disease ,Pedigree ,Phenotype ,Female ,medicine.symptom - Abstract
Microtia is a rare, congenital malformation of the external ear that in some cases has a genetic etiology. We ascertained a three-generation family with bilateral microtia and hearing loss segregating as an autosomal dominant trait. Exome sequencing of affected family members detected only seven shared, rare, heterozygous, nonsynonymous variants, including one protein truncating variant, a HOXA2 nonsense change (c.703C>T, p.Q235*). The HOXA2 variant was segregated with microtia and hearing loss in the family and was not seen in 6,500 individuals sequenced by the NHLBI Exome Sequencing Project or in 218 control individuals sequenced in this study. HOXA2 has been shown to be critical for outer and middle ear development through mouse models and has previously been associated with autosomal recessive bilateral microtia. Our data extend these conclusions and define HOXA2 haploinsufficiency as the first genetic cause for autosomal-dominant nonsyndromic microtia.
- Published
- 2013
- Full Text
- View/download PDF
35. Burden of Rare Sarcomere Gene Variants in the Framingham and Jackson Heart Study Cohorts
- Author
-
Sekar Kathiresan, James G. Wilson, Jonathan G. Seidman, Heidi L. Rehm, Ramachandran S. Vasan, Jayashri Aragam, Michael Parfenov, Joel N. Hirschhorn, Susan Cheng, Ervin R. Fox, Birgit Funke, Christine E. Seidman, Steven R. DePalma, Daniel S. Herman, Jason Flannick, Christopher J. O'Donnell, Stacey Gabriel, Kaoru Ito, Christopher Newton-Cheh, Emelia J. Benjamin, Namrata Gupta, Alexander G. Bick, David Altshuler, and Herman A. Taylor
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Sarcomeres ,medicine.medical_specialty ,Population ,Cardiomyopathy ,030204 cardiovascular system & hematology ,Biology ,Sarcomere ,Cohort Studies ,03 medical and health sciences ,0302 clinical medicine ,Framingham Heart Study ,Risk Factors ,Report ,Internal medicine ,Genetics ,medicine ,Humans ,Genetics(clinical) ,education ,Genetics (clinical) ,Aged ,030304 developmental biology ,Aged, 80 and over ,0303 health sciences ,education.field_of_study ,Framingham Risk Score ,Models, Cardiovascular ,Hypertrophic cardiomyopathy ,Genetic Variation ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,3. Good health ,Cardiovascular Diseases ,Cohort ,Cardiology ,Female ,Cohort study - Abstract
Rare sarcomere protein variants cause dominant hypertrophic and dilated cardiomyopathies. To evaluate whether allelic variants in eight sarcomere genes are associated with cardiac morphology and function in the community, we sequenced 3,600 individuals from the Framingham Heart Study (FHS) and Jackson Heart Study (JHS) cohorts. Out of the total, 11.2% of individuals had one or more rare nonsynonymous sarcomere variants. The prevalence of likely pathogenic sarcomere variants was 0.6%, twice the previous estimates; however, only four of the 22 individuals had clinical manifestations of hypertrophic cardiomyopathy. Rare sarcomere variants were associated with an increased risk for adverse cardiovascular events (hazard ratio: 2.3) in the FHS cohort, suggesting that cardiovascular risk assessment in the general population can benefit from rare variant analysis.
- Published
- 2012
- Full Text
- View/download PDF
36. Epigenomic Control of Cardiac Fibrosis by Bet Bromodomain Proteins in Dilated Cardiomyopathy
- Author
-
Jonathan G. Seidman, Jonathan D. Brown, Zhe Jiao, Jun Qi, Steven R. DePalma, Michael A. Burke, Joshua M. Gorham, James E. Bradner, Christine E. Seidman, Hiroko Wakimoto, Saptarsi M. Haldar, and David A. Conner
- Subjects
0301 basic medicine ,Regulation of gene expression ,030102 biochemistry & molecular biology ,Cardiac fibrosis ,business.industry ,Dilated cardiomyopathy ,musculoskeletal system ,medicine.disease ,complex mixtures ,Bromodomain ,Phospholamban ,BET inhibitor ,03 medical and health sciences ,Fibrosis ,Gene expression ,cardiovascular system ,Cancer research ,medicine ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,business - Abstract
Pathologic gene expression is a hallmark of DCM. Mice carrying a human DCM mutation in phospholamban (PLNR9C) develop fibrosis, DCM, HF and premature death. RNAseq showed fibrotic gene expression to be a key early driver of DCM in PLNR9C mice. Recently, bromodomain and extraterminal (BET) epigenetic reader proteins have been identified as key regulators of pathologic gene expression in the heart. Using a chemical genetic strategy, we studied the role of BET proteins on the temporal regulation of gene expression in DCM. PLNR9C and age-matched wild type (WT) mice were treated longitudinally with the BET inhibitor JQ1 or vehicle. Vehicle-treated PLNR9C mice developed progressive DCM with reduced LV function, chamber dilation and LV hypertrophy; this was blunted with JQ1 (fractional shortening 15±3% vs. 27±3%, p
- Published
- 2018
- Full Text
- View/download PDF
37. Locus for Familial Migrainous Vertigo Disease Maps to Chromosome 5q35
- Author
-
Fayez Bahmad, Jonathan G. Seidman, Christine E. Seidman, Saumil N. Merchant, Steven R. DePalma, Roberta Lemos Bezerra, and Carlos Augusto Costa Pires de Oliveira
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pediatrics ,Migraine Disorders ,Locus (genetics) ,Disease ,Article ,Genetic linkage ,Internal medicine ,Vertigo ,otorhinolaryngologic diseases ,medicine ,Humans ,Chromosomes, Human, 4-5 ,Genotyping ,Aged ,Aged, 80 and over ,biology ,business.industry ,Chromosome Mapping ,Autosomal dominant trait ,General Medicine ,Middle Aged ,medicine.disease ,biology.organism_classification ,Endocrinology ,Otorhinolaryngology ,Migraine ,Disease Progression ,Female ,Headaches ,medicine.symptom ,business - Abstract
Objectives: Migrainous vertigo (episodic vertigo associated with migraine) is sometimes inherited as an autosomal dominant trait. However, neither disease genes nor loci that might be responsible have been reported. We sought to map the genetic locus for familial migrainous vertigo in a 4-generation family and to define the progression of disease in this family. Methods: We studied 23 members in a family in whom migrainous vertigo was inherited as an autosomal dominant trait. Clinical information obtained included case histories and results of otolaryngological, neurologic, audiometric, and imaging evaluations. Genome-wide linkage analysis was performed with Affymetrix Genechip Human Mapping 10K microarrays. Genotyping of family members' DNA with microsatellite markers was used to further assess candidate loci identified from the whole-genome scan. Results: Of 23 family members, 10 suffered from migrainous vertigo beginning after 35 years of age. Migraine headaches usually preceded the onset of vertigo by 15 to 20 years. Longitudinal audiometric studies over 12 years showed stable, high-frequency sensorineural hearing loss consistent with presbycusis. Low-frequency or fluctuating hearing loss was not observed. The results of vestibular testing and imaging studies were unremarkable. Genetic analysis defined a 12.0 MB interval on chromosome 5q35 between loci rs244895 and D5S2073 that contained the disease gene (logarithm of odds score, 4.21). Conclusions: We report the first locus for familial migrainous vertigo, which mapped to 5q35.
- Published
- 2009
- Full Text
- View/download PDF
38. NKX2-5 Mutations in an Inbred Consanguineous Population: Genetic and Phenotypic Diversity
- Author
-
Mariam Arabi, Fadi Bitar, Jonathan G. Seidman, Akl C. Fahed, Ossama K. Abou Hassan, Manal Batrawi, Georges Nemer, Steven R. DePalma, Christine E. Seidman, and Marwan M. Refaat
- Subjects
Adult ,Heart Defects, Congenital ,Male ,Adolescent ,Genotype ,Genetic Linkage ,Population ,Penetrance ,Kaplan-Meier Estimate ,Disease ,Consanguinity ,Biology ,medicine.disease_cause ,Article ,Cohort Studies ,Electrocardiography ,Young Adult ,Genetic linkage ,Genetic variation ,Prevalence ,medicine ,Humans ,Lebanon ,Child ,education ,Genetic Association Studies ,Homeodomain Proteins ,Genetics ,Mutation ,education.field_of_study ,Multidisciplinary ,Genetic Variation ,Middle Aged ,respiratory system ,Pedigree ,Phenotype ,Child, Preschool ,Homeobox Protein Nkx-2.5 ,Female ,Transcription Factors - Abstract
NKX2-5 mutations are associated with different forms of congenital heart disease. Despite the knowledge gained from molecular and animal studies, genotype-phenotype correlations in humans are limited by the lack of large cohorts and the incomplete assessment of family members. We hypothesized that studying the role of NKX2-5 in inbred populations with homogeneous genetic backgrounds and high consanguinity rates such as Lebanon could help closing this gap. We sequenced NKX2-5 in 188 index CHD cases (25 with ASD). Five variants (three segregated in families) were detected in eleven families including the previously documented p.R25C variant, which was found in seven patients from different families and in one healthy individual. In 3/5 familial dominant ASD cases, we identified an NKX2-5 mutation. In addition to the heterogeneity of NKX2-5 mutations, a diversity of phenotypes occurred within the families with predominant ASD and AV block. We did in fact identify a large prevalence of Sudden Cardiac Death (SCD) in families with truncating mutations and two patients with coronary sinus disease. NKX2-5 is thus responsible for dominant familial ASD even in consanguineous populations and a wide genetic and phenotypic diversity is characteristic of NKX2-5 mutations in the Lebanese population.
- Published
- 2015
- Full Text
- View/download PDF
39. A novel COL1A1 mutation in infantile cortical hyperostosis (Caffey disease) expands the spectrum of collagen-related disorders
- Author
-
Richard Couper, Harald Jüppner, Stefan Mundlos, Jonathan G. Seidman, Hilal Abu-Zahra, Robert C. Gensure, William G. Cole, David Sillence, Steven R. DePalma, Catherine Barclay, Outi Mäkitie, Murat Bastepe, Leena Ala Kokko, and Catherine K Y Chan
- Subjects
0303 health sciences ,Pathology ,medicine.medical_specialty ,Infantile cortical hyperostosis ,Haplotype ,Chromosome ,Locus (genetics) ,General Medicine ,Biology ,medicine.disease ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Genetic linkage ,medicine ,Missense mutation ,030217 neurology & neurosurgery ,Type I collagen ,030304 developmental biology - Abstract
Infantile cortical hyperostosis (Caffey disease) is characterized by spontaneous episodes of subperiosteal new bone formation along 1 or more bones commencing within the first 5 months of life. A genome-wide screen for genetic linkage in a large family with an autosomal dominant form of Caffey disease (ADC) revealed a locus on chromosome 17q21 (LOD score, 6.78). Affected individuals and obligate carriers were heterozygous for a missense mutation (3040C→T) in exon 41 of the gene encoding the α1(I) chain of type I collagen (COL1A1), altering residue 836 (R836C) in the triple-helical domain of this chain. The same mutation was identified in affected members of 2 unrelated, smaller families with ADC, but not in 2 prenatal cases and not in more than 300 chromosomes from healthy individuals. Fibroblast cultures from an affected individual produced abnormal disulfide-bonded dimeric α1(I) chains. Dermal collagen fibrils of the same individual were larger, more variable in shape and size, and less densely packed than those in control samples. Individuals bearing the mutation, whether they had experienced an episode of cortical hyperostosis or not, had joint hyperlaxity, hyperextensible skin, and inguinal hernias resembling symptoms of a mild form of Ehlers-Danlos syndrome type III. These findings extend the spectrum of COL1A1-related diseases to include a hyperostotic disorder.
- Published
- 2005
- Full Text
- View/download PDF
40. Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: expanded panels offer limited additional sensitivity
- Author
-
Jun Shen, Jonathan G. Seidman, Melissa A. Kelly, Larry Babb, Stephanie Cox, Heidi L. Rehm, Matthew S. Lebo, Eugene Clark, Heather M. McLaughlin, Ahmed Alfares, Gregory McDermott, Christine E. Seidman, Samantha Baxter, Steven R. DePalma, Birgit Funke, and Carolyn Y. Ho
- Subjects
Proband ,Oncology ,Adult ,Male ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Adolescent ,Cardiomyopathy ,Sensitivity and Specificity ,Young Adult ,Internal medicine ,medicine ,Clinical genetic ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Child ,Genetics (clinical) ,Aged ,Oligonucleotide Array Sequence Analysis ,Aged, 80 and over ,business.industry ,Hypertrophic cardiomyopathy ,Genetic Variation ,High-Throughput Nucleotide Sequencing ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,Child, Preschool ,Cardiology ,Costs and Cost Analysis ,Female ,business - Abstract
Hypertrophic cardiomyopathy (HCM) is caused primarily by pathogenic variants in genes encoding sarcomere proteins. We report genetic testing results for HCM in 2,912 unrelated individuals with nonsyndromic presentations from a broad referral population over 10 years.Genetic testing was performed by Sanger sequencing for 10 genes from 2004 to 2007, by HCM CardioChip for 11 genes from 2007 to 2011 and by next-generation sequencing for 18, 46, or 51 genes from 2011 onward.The detection rate is ~32% among unselected probands, with inconclusive results in an additional 15%. Detection rates were not significantly different between adult and pediatric probands but were higher in females compared with males. An expanded gene panel encompassing more than 50 genes identified only a very small number of additional pathogenic variants beyond those identifiable in our original panels, which examined 11 genes. Familial genetic testing in at-risk family members eliminated the need for longitudinal cardiac evaluations in 691 individuals. Based on the projected costs derived from Medicare fee schedules for the recommended clinical evaluations of HCM family members by the American College of Cardiology Foundation/American Heart Association, our data indicate that genetic testing resulted in a minimum cost savings of about $0.7 million.Clinical HCM genetic testing provides a definitive molecular diagnosis for many patients and provides cost savings to families. Expanded gene panels have not substantively increased the clinical sensitivity of HCM testing, suggesting major additional causes of HCM still remain to be identified.
- Published
- 2014
41. Nationwide study on hypertrophic cardiomyopathy in Iceland: evidence of a MYBPC3 founder mutation
- Author
-
Barry J. Maron, Ragnar Danielsen, Christine E. Seidman, Daniel F. Gudbjartsson, Erica Mazaika, Michael A. Burke, Hilma Holm, Berglind Adalsteinsdottir, Polakit Teekakirikul, Barbara McDonough, Jonathan G. Seidman, Steven R. DePalma, Kari Stefansson, and Gunnar Gunnarsson
- Subjects
Adult ,Male ,Adolescent ,Population ,Iceland ,Young Adult ,Physiology (medical) ,Prevalence ,Medicine ,Humans ,Genetic Predisposition to Disease ,Age of Onset ,education ,Child ,Gene ,Founder mutation ,Aged ,Genetics ,Aged, 80 and over ,education.field_of_study ,business.industry ,Haplotype ,Hypertrophic cardiomyopathy ,Cardiomyopathy, Hypertrophic ,Middle Aged ,medicine.disease ,language.human_language ,Founder Effect ,Phenotype ,Haplotypes ,Child, Preschool ,Mutation (genetic algorithm) ,language ,MYH7 ,Female ,Cardiology and Cardiovascular Medicine ,Icelandic ,business ,Carrier Proteins - Abstract
Background— The geographic isolation and homogeneous population of Iceland are ideally suited to ascertain clinical and genetic characteristics of hypertrophic cardiomyopathy (HCM) at the population level. Methods and Results— Medical records and cardiac imaging studies obtained between 1997 and 2010 were reviewed to identify Icelandic patients with HCM. Surviving patients were recruited for clinical and genetic studies. A previously identified Icelandic mutation, MYBPC3 c.927-2A>G, was genotyped, and mutation-negative samples were sequenced for HCM genes and other hypertrophic genes. Record review identified 180 patients with HCM. Genetic analyses of 151 patients defined pathogenic mutations in 101 (67%), including MYBPC3 c.927-2A>G (88 patients, 58%), 4 other MYBPC3 or MYH7 mutations (5 patients, 3.3%), and 2 GLA mutations (8 patients, 5.3%). Haplotype and genetic genealogical data defined MYBPC3 c.927-2A>G as a founder mutation, introduced into the Icelandic population in the 15th century, with a current population prevalence of 0.36%. MYBPC3 c.927-2A>G mutation carriers exhibited phenotypic diversity but were younger at diagnosis (42 versus 49 years; P =0.001) and sustained more adverse events (15% versus 2%; P =0.02) than mutation-negative patients. All-cause mortality for patients with HCM was similar to that of an age-matched Icelandic population (hazard ratio, 0.98; P =0.9). HCM-related mortality (0.78%/y) occurred at a mean age of 68 compared with 81 years for non–HCM-related mortality ( P =0.02). Conclusions— A founder MYBPC3 mutation that arose >550 years ago is the predominant cause of HCM in Iceland. The MYBPC3 c.927-2A>G mutation is associated with low adverse event rates but earlier cardiovascular mortality, illustrating the impact of genotype on outcomes in HCM.
- Published
- 2014
42. A Nonsense Mutation in MSX1 Causes Witkop Syndrome
- Author
-
Marianna Bei, Dolrudee Jumlongras, Ute Felbor, Jonathan G. Seidman, Jean M. Stimson, Richard L. Maas, Christine E. Seidman, WenFang Wang, Bjorn R. Olsen, and Steven R. DePalma
- Subjects
Male ,Ectodermal dysplasia ,Genetic Linkage ,DNA Mutational Analysis ,Mice ,0302 clinical medicine ,Genetics(clinical) ,In Situ Hybridization ,Genetics (clinical) ,Genes, Dominant ,Mice, Knockout ,Genetics ,0303 health sciences ,integumentary system ,Chromosome Mapping ,Syndrome ,Articles ,Pedigree ,Phenotype ,medicine.anatomical_structure ,Codon, Nonsense ,Nail (anatomy) ,Female ,Adult ,Heterozygote ,Nonsense mutation ,Nails, Malformed ,Biology ,03 medical and health sciences ,Genetic linkage ,medicine ,Animals ,Humans ,Tooth and nail syndrome ,Amino Acid Sequence ,RNA, Messenger ,Anodontia ,030304 developmental biology ,Homeodomain Proteins ,MSX1 Transcription Factor ,Polymorphism, Genetic ,Base Sequence ,Heterozygote advantage ,030206 dentistry ,medicine.disease ,Protein Structure, Tertiary ,stomatognathic diseases ,Nail disease ,PAX9 ,Transcription Factors - Abstract
Witkop syndrome, also known as tooth and nail syndrome (TNS), is a rare autosomal dominant disorder. Affected individuals have nail dysplasia and several congenitally missing teeth. To identify the gene responsible for TNS, we used candidate-gene linkage analysis in a three-generation family affected by the disorder. We found linkage between TNS and polymorphic markers surrounding the MSX1 locus. Direct sequencing and restriction-enzyme analysis revealed that a heterozygous stop mutation in the homeodomain of MSX1 cosegregated with the phenotype. In addition, histological analysis of Msx1-knockout mice, combined with a finding of Msx1 expression in mesenchyme of developing nail beds, revealed that not only was tooth development disrupted in these mice, but nail development was affected as well. Nail plates in Msx1-null mice were defective and were thinner than those of their wild-type littermates. The resemblance between the tooth and nail phenotype in the human family and that of Msx1-knockout mice strongly supports the conclusions that a nonsense mutation in MSX1 causes TNS and that Msx1 is critical for both tooth and nail development.
- Published
- 2001
- Full Text
- View/download PDF
43. Mutations in Sarcomere Protein Genes as a Cause of Dilated Cardiomyopathy
- Author
-
Mitsuhiro Kamisago, Sapna D. Sharma, Steven R. DePalma, Scott Solomon, Pankaj Sharma, Barbara McDonough, Leslie Smoot, Mary P. Mullen, Paul K. Woolf, E. Douglas Wigle, J.G. Seidman, John Jarcho, Lawrence R. Shapiro, and Christine E. Seidman
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Sarcomeres ,medicine.medical_specialty ,Adolescent ,TNNT2 ,Molecular Sequence Data ,Mutation, Missense ,Cardiomyopathy ,medicine.disease_cause ,Troponin T ,Internal medicine ,Idiopathic dilated cardiomyopathy ,Troponin I ,medicine ,Humans ,Amino Acid Sequence ,Child ,Aged ,Genes, Dominant ,Ultrasonography ,Chromosomes, Human, Pair 14 ,Genetics ,Mutation ,Nonmuscle Myosin Type IIB ,Myosin Heavy Chains ,biology ,Myocardium ,Chromosome Mapping ,Infant ,Dilated cardiomyopathy ,General Medicine ,Myocardial Disorder ,Middle Aged ,medicine.disease ,Myocardial Contraction ,Troponin ,Pedigree ,Child, Preschool ,biology.protein ,Cardiology ,Female - Abstract
The molecular basis of idiopathic dilated cardiomyopathy, a primary myocardial disorder that results in reduced contractile function, is largely unknown. Some cases of familial dilated cardiomyopathy are caused by mutations in cardiac cytoskeletal proteins; this finding implicates defects in contractile-force transmission as one mechanism underlying this disorder. To elucidate this important cause of heart failure, we investigated other genetic causes of dilated cardiomyopathy.Clinical evaluations were performed in 21 kindreds with familial dilated cardiomyopathy. A genome-wide linkage study prompted a search of the genes encoding beta-myosin heavy chain, troponin T, troponin I, and alpha-tropomyosin for disease-causing mutations.A genetic locus for mutations associated with dilated cardiomyopathy was identified at chromosome 14q11.2-13 (maximal lod score, 5.11; theta=0), where the gene for cardiac beta-myosin heavy chain is encoded. Analyses of this and other genes for sarcomere proteins identified disease-causing dominant mutations in four kindreds. Cardiac beta-myosin heavy-chain missense mutations (Ser532Pro and Phe764Leu) and a deletion in cardiac troponin T (deltaLys210) caused early-onset ventricular dilatation (average age at diagnosis, 24 years) and diminished contractile function and frequently resulted in heart failure. Affected persons had neither antecedent cardiac hypertrophy (average maximal left-ventricular-wall thickness, 8.5 mm) nor histopathological findings characteristic of hypertrophy.Mutations in sarcomere protein genes account for approximately 10 percent of cases of familial dilated cardiomyopathy and are particularly prevalent in families with early-onset ventricular dilatation and dysfunction. Because distinct mutations in sarcomere proteins cause either dilated or hypertrophic cardiomyopathy, the effects of mutant sarcomere proteins on muscle mechanics must trigger two different series of events that remodel the heart.
- Published
- 2000
- Full Text
- View/download PDF
44. PEDIATRIC CARDIOMYOPATHY MUTATIONS IN A HIGHLY CONSANGUINEOUS POPULATION
- Author
-
Christine E. Seidman, Şükrü Candan, Steven R. DePalma, Akl C. Fahed, Jonathan G. Seidman, Kazum Öztarhan, Hatip Aydin, Helen Bornaun, Betül Erer, Ahmet Ekmekçi, Barbara McDonough, and Alireza Haghighi
- Subjects
education.field_of_study ,Pediatrics ,medicine.medical_specialty ,Pediatric cardiomyopathy ,business.industry ,Population ,Cardiomyopathy ,medicine ,Disease ,Cardiology and Cardiovascular Medicine ,education ,medicine.disease ,business - Abstract
Pediatric-onset cardiomyopathy is more common in inbred and highly consanguineous populations that are enriched for familial and recessive disease. The genetic causes of pediatric cardiomyopathy remain poorly understood. Nearly one quarter of marriages in Turkey are consanguineous. Studying this
- Published
- 2016
- Full Text
- View/download PDF
45. Spectrum of somatic mitochondrial mutations in five cancers
- Author
-
Alexei Protopopov, Jonathan G. Seidman, Jianhua Zhang, Raju Kucherlapati, Tatianna Larman, Angela Hadjipanayis, Stacey Gabriel, Christine E. Seidman, Lynda Chin, and Steven R. DePalma
- Subjects
Nonsynonymous substitution ,Mitochondrial DNA ,Somatic cell ,Mutation, Missense ,Biology ,Adenocarcinoma ,medicine.disease_cause ,DNA, Mitochondrial ,medicine ,Missense mutation ,Humans ,Gene ,Genetics ,Ovarian Neoplasms ,Multidisciplinary ,Genomics ,Biological Sciences ,medicine.disease ,Cystadenocarcinoma, Serous ,Gene Expression Regulation, Neoplastic ,Leukemia ,Leukemia, Myeloid, Acute ,Genes, Mitochondrial ,Colonic Neoplasms ,Female ,Carcinogenesis ,Colorectal Neoplasms ,Gene Deletion - Abstract
Somatic mtDNA mutations have been reported in some human tumors, but their spectrum in different malignancies and their role in cancer development remain incompletely understood. Here, we describe the breadth of somatic and inherited mutations across the mitochondrial genome by sequence analyses of paired tumor and normal tissue samples from 226 individuals with five types of cancer using whole-genome data generated by The Cancer Genome Atlas Research Network. The frequencies of deleterious tumor-specific somatic mutations found in mtDNA varied across tumor types, ranging from 13% of glioblastomas to 63% of rectal adenocarcinomas. Compared with inherited mtDNA variants, somatic mtDNA mutations were enriched for nonsynonymous vs. synonymous changes (93 vs. 15; P < 2.2E−16) and were predicted to functionally impact the encoded protein. Somatic missense mutations in tumors were distributed uniformly among the mitochondrial protein genes, but 65% of somatic truncating mutations occurred in NADH dehydrogenase 5. Analysis of staging data in colon and rectal cancers revealed that the frequency of damaging mitochondrial mutations is the same in stages I and IV tumors. In summary, these data suggest that damaging somatic mtDNA mutations occur frequently (13–63%) in these five tumor types and likely confer a selective advantage in oncogenesis.
- Published
- 2012
46. Truncations of titin causing dilated cardiomyopathy
- Author
-
Elizabeth Sparks, Lauren Conner, Debbie Lin Teodorescu, Daniel S. Herman, Steven R. DePalma, Polakit Teekakirikul, Sharon L. Graw, Charles E. Murry, Dudley J. Pennell, Lien Lam, Nicholas R. Banner, Paul J.R. Barton, Allison L. Cirino, Michael J. Ackerman, Andrea Di Lenarda, Matthew R.G. Taylor, Barbara McDonough, Carolyn Y. Ho, Richard N. Mitchell, Marco Merlo, Danos C. Christodoulou, Stuart A. Cook, Neal K. Lakdawala, Christine E. Seidman, Libin Wang, Jonathan G. Seidman, Gianfranco Sinagra, Luisa Mestroni, J. Martijn Bos, Herman, D, Lam, L, Taylor, Mr, Wang, L, Teekakirikul, P, Christodoulou, D, Conner, L, Depalma, Sr, Mcdonough, B, Sparks, E, Teodorescu, Dl, Cirino, Al, Banner, Nr, Pennell, Dj, Graw, S, Merlo, M, Di Lenarda, A, Sinagra, Gianfranco, Bos, Jm, Ackerman, Mj, Mitchell, Rn, Murry, Ce, Lakdawala, Nk, Ho, Cy, Barton, Pj, Cook, Sa, Mestroni, L, Seidman, Jg, and Seidman, C. E.
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,Genotyping Techniques ,Cardiomyopathy ,Dilated cardiomyopathy ,Muscle Proteins ,medicine.disease_cause ,Frameshift mutation ,symbols.namesake ,medicine ,Humans ,Connectin ,titin ,Gene ,Sequence Deletion ,Sanger sequencing ,Genetics ,Mutation ,biology ,business.industry ,Myocardium ,Hypertrophic cardiomyopathy ,Sequence Analysis, DNA ,General Medicine ,Middle Aged ,medicine.disease ,mutations ,hypertrophic cardiomyopathy ,Molecular biology ,symbols ,biology.protein ,Female ,Titin ,mutation ,business ,Protein Kinases - Abstract
Dilated cardiomyopathy and hypertrophic cardiomyopathy arise from mutations in many genes. TTN, the gene encoding the sarcomere protein titin, has been insufficiently analyzed for cardiomyopathy mutations because of its enormous size.We analyzed TTN in 312 subjects with dilated cardiomyopathy, 231 subjects with hypertrophic cardiomyopathy, and 249 controls by using next-generation or dideoxy sequencing. We evaluated deleterious variants for cosegregation in families and assessed clinical characteristics.We identified 72 unique mutations (25 nonsense, 23 frameshift, 23 splicing, and 1 large tandem insertion) that altered full-length titin. Among subjects studied by means of next-generation sequencing, the frequency of TTN mutations was significantly higher among subjects with dilated cardiomyopathy (54 of 203 [27%]) than among subjects with hypertrophic cardiomyopathy (3 of 231 [1%], P=3×10(-16)) or controls (7 of 249 [3%], P=9×10(-14)). TTN mutations cosegregated with dilated cardiomyopathy in families (combined lod score, 11.1) with high (95%) observed penetrance after the age of 40 years. Mutations associated with dilated cardiomyopathy were overrepresented in the titin A-band but were absent from the Z-disk and M-band regions of titin (P≤0.01 for all comparisons). Overall, the rates of cardiac outcomes were similar in subjects with and those without TTN mutations, but adverse events occurred earlier in male mutation carriers than in female carriers (P=4×10(-5)).TTN truncating mutations are a common cause of dilated cardiomyopathy, occurring in approximately 25% of familial cases of idiopathic dilated cardiomyopathy and in 18% of sporadic cases. Incorporation of sequencing approaches that detect TTN truncations into genetic testing for dilated cardiomyopathy should substantially increase test sensitivity, thereby allowing earlier diagnosis and therapeutic intervention for many patients with dilated cardiomyopathy. Defining the functional effects of TTN truncating mutations should improve our understanding of the pathophysiology of dilated cardiomyopathy. (Funded by the Howard Hughes Medical Institute and others.).
- Published
- 2012
47. Quantification of Gene Transcripts with Deep Sequencing Analysis of Gene Expression (DSAGE) Using 1 to 2 µg Total RNA
- Author
-
Daniel S. Herman, Danos C. Christodoulou, Masataka Kawana, Hiroko Wakimoto, Joshua M. Gorham, and Steven R. DePalma
- Subjects
Genetics ,Massive parallel sequencing ,Gene Expression Profiling ,High-Throughput Nucleotide Sequencing ,RNA-Seq ,Biology ,Article ,DNA sequencing ,Deep sequencing ,Massively parallel signature sequencing ,Gene expression profiling ,Complementary DNA ,RNA ,Reference genome - Abstract
Deep sequencing analysis of gene expression (DSAGE) measures global gene transcript levels by massively parallel sequencing of cDNA tags, using 1-2μg total RNA. 21-bp cDNA tags are generated by NlaIII digestion of the cDNA, followed by MmeI cleavage offset from the NlaIII site. cDNA tags are then queried by massively parallel sequencing and aligned to a reference genome and transcriptome, or any available gene sequences, using Bowtie, an ultra high-throughput short-read aligner, and Tophat, a fast splice-junction mapper. Analysis of 10-20-million tags, acquired using one lane of an Illumina Genome Analyzer II, provides sufficient depth to quantify gene expression and detect rare transcripts. Typically, we observe the expression of 15,000 genes in the cardiac left ventricle, including gene transcripts expressed as low as one copy per cell. These expression profiles are highly reproducible (r>0.99 between technical replicates), enabling sensitive detection of differences between experimental conditions as well as assessment of relative transcript abundance between different genes. The significance of these differences is assessed, while accounting for multiple comparisons, using a false discovery rate approach (Audic and Claverie, 1997). Thus, DSAGE can be used to quantify gene expression profiles and assess differential expression with high sensitivity requiring small amounts of biological material.
- Published
- 2011
- Full Text
- View/download PDF
48. Genomewide Linkage in a Large Caucasian Family Maps a New Locus for Intracranial Aneurysms to Chromosome 13q
- Author
-
Steven R. DePalma, Jonathan G. Seidman, Dong H. Kim, Christine E. Seidman, Teresa Santiago-Sim, Barbara McDonough, and Kevin L. Ju
- Subjects
Adult ,Male ,Pathology ,medicine.medical_specialty ,Genetic Linkage ,Locus (genetics) ,Disease ,Polymorphism, Single Nucleotide ,White People ,Genetic linkage ,medicine ,Humans ,Genetic Predisposition to Disease ,Gene ,Aged ,Advanced and Specialized Nursing ,Genetics ,Chromosomes, Human, Pair 13 ,business.industry ,Family aggregation ,Chromosome ,Intracranial Aneurysm ,Middle Aged ,Pedigree ,Genetic marker ,Susceptibility locus ,Female ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background and Purpose— Familial aggregation of intracranial aneurysms (IAs) indicates a genetic role in the pathogenesis of this disease. Despite a number of reported susceptibility loci, no disease-causing gene variants have been identified. In this study, we used a parametric genomewide linkage approach to search for new IA susceptibility loci in a large Caucasian family. Methods— The affection status of family members with clinical signs of IA was confirmed with medical records or through radiological or surgical examinations. All other relatives were screened using MR angiography. Genomewide linkage analysis was performed on 35 subjects using approximately 250 000 single nucleotide polymorphic markers. Results— Ten individuals had an IA. Linkage analysis using a dominant model showed significant linkage to a 7-cM region in 13q14.12–21.1 with a maximum logarithm of odds score of 4.56. Conclusion— A new IA susceptibility locus on 13q was identified, adding to the number of IA loci already reported. Given that no coding variants have been reported to date, it is possible that alternative genetic variants such as regulatory elements or copy number variation are important in IA pathogenesis. We are proceeding with attempts to identify such variants in our locus.
- Published
- 2009
- Full Text
- View/download PDF
49. De novo copy number variants identify new genes and loci in isolated sporadic tetralogy of Fallot
- Author
-
Michael E. Weinblatt, Maria A. Artunduaga, Steven C. Greenway, Steven R. DePalma, Philip L. De Jager, Robert M. Plenge, Jonathan G. Seidman, Roger E. Breitbart, Sonia M Mesquita, Maria de Lourdes Quintanilla-Dieck, Joshua M. Korn, Joshua M. Gorham, Roland D. Eavey, Jessie H. Conta, Christine E. Seidman, Steven A. McCarroll, Alexandre C. Pereira, Stacey Gabriel, David A. Hafler, Jennifer C Lin, Nancy A. Shadick, Samuel J Israel, David Altshuler, and Emel Ergul
- Subjects
JAG1 ,congenital, hereditary, and neonatal diseases and abnormalities ,endocrine system diseases ,Heart malformation ,Heart Ventricles ,Gene Dosage ,Locus (genetics) ,Biology ,Gene dosage ,Article ,Genetic variation ,mental disorders ,Genetics ,medicine ,Humans ,Copy-number variation ,Tetralogy of Fallot ,Chromosome ,Genetic Variation ,medicine.disease ,Phenotype ,Gene Expression Regulation ,Chromosomes, Human, Pair 1 ,Child, Preschool ,Chromosomes, Human, Pair 3 - Abstract
Tetralogy of Fallot (TOF), the most common severe congenital heart malformation, occurs sporadically, without other anomaly, and from unknown cause in 70% of cases. Through a genome-wide survey of 114 subjects with TOF and their unaffected parents, we identified 11 de novo copy number variants (CNVs) that were absent or extremely rare (
- Published
- 2009
50. THE R21C MUTATION IN TROPONIN I HAS A FOUNDER EFFECT IN SOUTH LEBANON AND CAUSES MALIGNANT HYPERTROPHIC CARDIOMYOPATHY
- Author
-
Christine E. Seidman, Georges Nemer, Barbara McDonough, Mariam T. Arabi, Akl C. Fahed, Steven R. DePalma, Antoine B. Abchee, Fadi F. Bitar, Jonathan G. Seidman, Samir Arnaout, Manal Batrawi, and James S. Ware
- Subjects
Genetics ,medicine.medical_specialty ,business.industry ,Hypertrophic cardiomyopathy ,medicine.disease ,Phenotype ,Internal medicine ,Troponin I ,Mutation (genetic algorithm) ,cardiovascular system ,Cardiology ,Medicine ,cardiovascular diseases ,Cardiology and Cardiovascular Medicine ,business ,Gene ,Founder effect - Abstract
Although gene testing is widely available for Hypertrophic Cardiomyopathy (HCM), determining the clinical implications of a mutation is limited by an understanding of its phenotype. Inbred populations with founder mutations provide an opportunity to study genotype-phenotype correlations. We
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.