Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Circ Genom Precis Med ; 16(5): 452-461, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37767697

RESUMEN

BACKGROUND: Many cardiovascular disorders propel the development of advanced heart failure that necessitates cardiac transplantation. When treatable causes are excluded, studies to define causes are often abandoned, resulting in a diagnosis of end-stage idiopathic cardiomyopathy. We studied whether DNA sequence analyses could identify unrecognized causes of end-stage nonischemic cardiomyopathy requiring heart transplantation and whether the prevalence of genetic causes differed from ambulatory cardiomyopathy cases. METHODS: We performed whole exome and genome sequencing of 122 explanted hearts from 101 adult and 21 pediatric patients with idiopathic cardiomyopathy from a single center. Data were analyzed for pathogenic/likely pathogenic variants in nuclear and mitochondrial genomes and assessed for nonhuman microbial sequences. The frequency of damaging genetic variants was compared among cardiomyopathy cohorts with different clinical severity. RESULTS: Fifty-four samples (44.3%) had pathogenic/likely pathogenic cardiomyopathy gene variants. The frequency of pathogenic variants was similar in pediatric (42.9%) and adult (43.6%) samples, but the distribution of mutated genes differed (P=8.30×10-4). The prevalence of causal genetic variants was significantly higher in end-stage than in previously reported ambulatory adult dilated cardiomyopathy cases (P<0.001). Among remaining samples with unexplained causes, no damaging mitochondrial variants were identified, but 28 samples contained parvovirus genome sequences, including 2 samples with 6- to 9-fold higher levels than the overall mean levels in other samples. CONCLUSIONS: Pathogenic variants and viral myocarditis were identified in 45.9% of patients with unexplained end-stage cardiomyopathy. Damaging gene variants are significantly more frequent among transplant compared with patients with ambulatory cardiomyopathy. Genetic analyses can help define cause of end-stage cardiomyopathy to guide management and risk stratification of patients and family members.


Asunto(s)
Cardiomiopatías , Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Trasplante de Corazón , Adulto , Humanos , Niño , Cardiomiopatías/genética , Cardiomiopatías/patología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/cirugía , Cardiomiopatía Dilatada/diagnóstico , Insuficiencia Cardíaca/diagnóstico
2.
Genet Med ; 25(1): 143-150, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36260083

RESUMEN

PURPOSE: 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. METHODS: 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. RESULTS: 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. CONCLUSION: 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.


Asunto(s)
Microtia Congénita , Síndrome de Goldenhar , Micrognatismo , Humanos , Síndrome de Goldenhar/genética , Microtia Congénita/genética , Oído/anomalías , Cara
3.
Science ; 377(6606): eabo1984, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35926050

RESUMEN

Pathogenic variants in genes that cause dilated cardiomyopathy (DCM) and arrhythmogenic cardiomyopathy (ACM) convey high risks for the development of heart failure through unknown mechanisms. Using single-nucleus RNA sequencing, we characterized the transcriptome of 880,000 nuclei from 18 control and 61 failing, nonischemic human hearts with pathogenic variants in DCM and ACM genes or idiopathic disease. We performed genotype-stratified analyses of the ventricular cell lineages and transcriptional states. The resultant DCM and ACM ventricular cell atlas demonstrated distinct right and left ventricular responses, highlighting genotype-associated pathways, intercellular interactions, and differential gene expression at single-cell resolution. Together, these data illuminate both shared and distinct cellular and molecular architectures of human heart failure and suggest candidate therapeutic targets.


Asunto(s)
Displasia Ventricular Derecha Arritmogénica , Cardiomiopatía Dilatada , Insuficiencia Cardíaca , Análisis de la Célula Individual , Transcriptoma , Displasia Ventricular Derecha Arritmogénica/genética , Atlas como Asunto , Cardiomiopatía Dilatada/genética , Núcleo Celular/genética , Insuficiencia Cardíaca/genética , Ventrículos Cardíacos , Humanos , RNA-Seq
4.
Proc Natl Acad Sci U S A ; 119(21): e2203928119, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35584116

RESUMEN

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 cell­derived 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.


Asunto(s)
Indio Americano o Nativo de Alaska , Microtia Congénita , Microtia Congénita/genética , Oído Externo , Efecto Fundador , Humanos , Mutación , Proteínas del Tejido Nervioso/genética , Receptores Inmunológicos/genética , Indio Americano o Nativo de Alaska/genética , Proteínas Roundabout
5.
Circ Genom Precis Med ; 14(5): e003389, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34461741

RESUMEN

BACKGROUND: Heterozygous TTN truncating 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 as TTN truncating 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 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 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 in TTN splice 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 most TTN transcripts frequently lacked the consensus base at +5 donor and -3 acceptor positions. CONCLUSIONS: Noncanonical 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.


Asunto(s)
Cardiomiopatía Dilatada/genética , Conectina/genética , Exones , Heterocigoto , Empalme del ARN , Adolescente , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad
6.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33658374

RESUMEN

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.


Asunto(s)
Cardiomiopatía Hipertrófica , Ecocardiografía , Epigénesis Genética , Ventrículos Cardíacos , Proteínas Musculares , Gemelos Monocigóticos , Adolescente , Adulto , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/fisiopatología , Preescolar , Femenino , Estudios de Seguimiento , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Proteínas Musculares/genética , Proteínas Musculares/metabolismo
8.
Circ Genom Precis Med ; 13(5): 444-452, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32885985

RESUMEN

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.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Troponina I/genética , Adolescente , Adulto , Cardiomiopatía Hipertrófica/diagnóstico , Niño , Muerte Súbita Cardíaca/etiología , Ecocardiografía , Femenino , Efecto Fundador , Humanos , Masculino , Persona de Mediana Edad , Miocardio/patología , Linaje , Fenotipo , Dominios Proteicos/genética , Troponina I/química , Adulto Joven
9.
Nature ; 588(7838): 466-472, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32971526

RESUMEN

Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.


Asunto(s)
Miocardio/citología , Análisis de la Célula Individual , Transcriptoma , Adipocitos/clasificación , Adipocitos/metabolismo , Adulto , Enzima Convertidora de Angiotensina 2/análisis , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Células Epiteliales/clasificación , Células Epiteliales/metabolismo , Epitelio , Femenino , Fibroblastos/clasificación , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Atrios Cardíacos/anatomía & histología , Atrios Cardíacos/citología , Atrios Cardíacos/inervación , Ventrículos Cardíacos/anatomía & histología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/inervación , Homeostasis/inmunología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Miocitos Cardíacos/clasificación , Miocitos Cardíacos/metabolismo , Neuronas/clasificación , Neuronas/metabolismo , Pericitos/clasificación , Pericitos/metabolismo , Receptores de Coronavirus/análisis , Receptores de Coronavirus/genética , Receptores de Coronavirus/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Células del Estroma/clasificación , Células del Estroma/metabolismo
10.
Sci Transl Med ; 11(476)2019 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-30674652

RESUMEN

The mechanisms by which truncating mutations in MYBPC3 (encoding cardiac myosin-binding protein C; cMyBPC) or myosin missense mutations cause hypercontractility and poor relaxation in hypertrophic cardiomyopathy (HCM) are incompletely understood. Using genetic and biochemical approaches, we explored how depletion of cMyBPC altered sarcomere function. We demonstrated that stepwise loss of cMyBPC resulted in reciprocal augmentation of myosin contractility. Direct attenuation of myosin function, via a damaging missense variant (F764L) that causes dilated cardiomyopathy (DCM), normalized the increased contractility from cMyBPC depletion. Depletion of cMyBPC also altered dynamic myosin conformations during relaxation, enhancing the myosin state that enables ATP hydrolysis and thin filament interactions while reducing the super relaxed conformation associated with energy conservation. MYK-461, a pharmacologic inhibitor of myosin ATPase, rescued relaxation deficits and restored normal contractility in mouse and human cardiomyocytes with MYBPC3 mutations. These data define dosage-dependent effects of cMyBPC on myosin that occur across the cardiac cycle as the pathophysiologic mechanisms by which MYBPC3 truncations cause HCM. Therapeutic strategies to attenuate cMyBPC activity may rescue depressed cardiac contractility in patients with DCM, whereas inhibiting myosin by MYK-461 should benefit the substantial proportion of patients with HCM with MYBPC3 mutations.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Proteínas Portadoras/genética , Mutación/genética , Miosinas/metabolismo , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/metabolismo , Animales , Cardiomiopatía Hipertrófica/fisiopatología , Modelos Animales de Enfermedad , Haploinsuficiencia , Humanos , Ratones , Contracción Miocárdica , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Fenotipo , ortoaminobenzoatos/metabolismo
11.
Genet Med ; 21(1): 133-143, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-29892087

RESUMEN

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 <0.0001). For some of these genes, DCM-associated variants appeared to be clustered in key protein functional domains. Multiple rare variants were present in many family probands, however, there was generally only one "driver" pathogenic variant that cosegregated with disease. CONCLUSION: Rare variants in cardiomyopathy genes can be effectively stratified by combining variant-level and gene-level information. Prioritization of genes based on their a priori likelihood of disease causation is a key factor in identifying clinically actionable variants in cardiac genetic testing.


Asunto(s)
Cardiomiopatía Dilatada/genética , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedades Raras/genética , Cardiomiopatía Dilatada/diagnóstico , Cardiomiopatía Dilatada/patología , Femenino , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Linaje , Enfermedades Raras/diagnóstico , Enfermedades Raras/patología
12.
Circ Cardiovasc Genet ; 10(5)2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29030401

RESUMEN

BACKGROUND: As DNA sequencing costs decline, genetic testing options have expanded. Whole exome sequencing and whole genome sequencing (WGS) are entering clinical use, posing questions about their incremental value compared with disease-specific multigene panels that have been the cornerstone of genetic testing. METHODS AND RESULTS: Forty-one patients with hypertrophic cardiomyopathy who had undergone targeted hypertrophic cardiomyopathy genetic testing (either multigene panel or familial variant test) were recruited into the MedSeq Project, a clinical trial of WGS. Results from panel genetic testing and WGS were compared. In 20 of 41 participants, panel genetic testing identified variants classified as pathogenic, likely pathogenic, or uncertain significance. WGS identified 19 of these 20 variants, but the variant detection algorithm missed a pathogenic 18 bp duplication in myosin binding protein C (MYBPC3) because of low coverage. In 3 individuals, WGS identified variants in genes implicated in cardiomyopathy but not included in prior panel testing: a pathogenic protein tyrosine phosphatase, non-receptor type 11 (PTPN11) variant and variants of uncertain significance in integrin-linked kinase (ILK) and filamin-C (FLNC). WGS also identified 84 secondary findings (mean=2 per person, range=0-6), which mostly defined carrier status for recessive conditions. CONCLUSIONS: WGS detected nearly all variants identified on panel testing, provided 1 new diagnostic finding, and allowed interrogation of posited disease genes. Several variants of uncertain clinical use and numerous secondary genetic findings were also identified. Whereas panel testing and WGS provided similar diagnostic yield, WGS offers the advantage of reanalysis over time to incorporate advances in knowledge, but requires expertise in genomic interpretation to appropriately incorporate WGS into clinical care. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov. Unique identifier: NCT01736566.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Pruebas Genéticas/métodos , Secuenciación Completa del Genoma/métodos , Anciano , Algoritmos , Proteínas Portadoras/genética , Femenino , Filaminas/genética , Duplicación de Gen , Variación Genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Población Blanca/genética
13.
Proc Natl Acad Sci U S A ; 114(29): 7689-7694, 2017 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-28679633

RESUMEN

Genetic variants that cause haploinsufficiency account for many autosomal dominant (AD) disorders. Gene-based diagnosis classifies variants that alter canonical splice signals as pathogenic, but due to imperfect understanding of RNA splice signals other variants that may create or eliminate splice sites are often clinically classified as variants of unknown significance (VUS). To improve recognition of pathogenic splice-altering variants in AD disorders, we used computational tools to prioritize VUS and developed a cell-based minigene splicing assay to confirm aberrant splicing. Using this two-step procedure we evaluated all rare variants in two AD cardiomyopathy genes, lamin A/C (LMNA) and myosin binding protein C (MYBPC3). We demonstrate that 13 LMNA and 35 MYBPC3 variants identified in cardiomyopathy patients alter RNA splicing, representing a 50% increase in the numbers of established damaging splice variants in these genes. Over half of these variants are annotated as VUS by clinical diagnostic laboratories. Familial analyses of one variant, a synonymous LMNA VUS, demonstrated segregation with cardiomyopathy affection status and altered cardiac LMNA splicing. Application of this strategy should improve diagnostic accuracy and variant classification in other haploinsufficient AD disorders.


Asunto(s)
Proteínas Portadoras/genética , Lamina Tipo A/genética , Mutación , Empalme del ARN , Adulto , Anciano , Alelos , Cardiomiopatías/genética , Biología Computacional , Femenino , Variación Genética , Genotipo , Células HEK293 , Haploinsuficiencia , Cardiopatías/genética , Trasplante de Corazón , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Marcapaso Artificial , Linaje , Sitios de Empalme de ARN , Análisis de Secuencia de ADN , Adulto Joven
14.
JACC Basic Transl Sci ; 2(2): 122-131, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28596995

RESUMEN

The degradation and release of cardiac myosin binding protein-C (cMyBP-C) upon cardiac damage may stimulate an inflammatory response and autoantibody (AAb) production. We determined whether the presence of cMyBP-C-AAbs associated with adverse cardiac function in CVD patients. Importantly, cMyBP-C-AAbs were significantly detected in ACS patient sera upon arrival to the emergency department, particularly in STEMI patients. Patients positive for cMyBP-C-AAbs had a reduced LVEF and elevated levels of clinical biomarkers of MI. We conclude that cMyBP-C-AAbs may serve as early predictive indicators of deteriorating cardiac function and patient outcome in ACS patients prior to the infarction.

15.
Stroke ; 47(12): 3005-3013, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27895300

RESUMEN

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<0.0001). In zebrafish and mice, Thsd1 loss-of-function caused cerebral bleeding (which localized to the subarachnoid space in mice) and increased mortality. Mechanistically, 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.


Asunto(s)
Aneurisma Roto/genética , Aneurisma Intracraneal/genética , Hemorragia Subaracnoidea/genética , Trombospondinas/genética , Animales , Codón sin Sentido , Modelos Animales de Enfermedad , Exoma , Predisposición Genética a la Enfermedad , Humanos , Ratones , Linaje , Pez Cebra , Proteínas de Pez Cebra
16.
Circulation ; 130(14): 1158-67, 2014 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-25078086

RESUMEN

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.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/mortalidad , Proteínas Portadoras/genética , Efecto Fundador , Predisposición Genética a la Enfermedad/epidemiología , Predisposición Genética a la Enfermedad/genética , Adolescente , Adulto , Edad de Inicio , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Haplotipos , Humanos , Islandia/epidemiología , Masculino , Persona de Mediana Edad , Fenotipo , Prevalencia , Adulto Joven
17.
Ann Neurol ; 75(5): 793-798, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24771548

RESUMEN

We report a 5-generation family with phenotypically diverse neurodegenerative disease including relentlessly progressive choreoathetoid movements, dysarthria, dysphagia, spastic paralysis, and behavioral dementia in descendants of a 67-year-old woman with amyotrophic lateral sclerosis. Disease onset varied with gender, occurring in male children and adult women. Exome sequence analyses revealed a novel mutation (c.1490C>T, p.P497L) in the ubiquilin-2 gene (UBQLN2) with X-linked inheritance in all studied affected individuals. As ubiquilin-2-positive inclusions were identified in brain, we suggest that mutant peptide predisposes to protein misfolding and accumulation. Our findings expand the spectrum of neurodegenerative phenotypes caused by UBQLN2 mutations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Heterogeneidad Genética , Trastornos Heredodegenerativos del Sistema Nervioso/diagnóstico , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Mutación/genética , Ubiquitinas/genética , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Adulto , Anciano , Proteínas Relacionadas con la Autofagia , Preescolar , Femenino , Genes Dominantes , Humanos , Masculino , Linaje , Pliegue de Proteína , Adulto Joven
18.
J Clin Invest ; 124(3): 1364-70, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24509080

RESUMEN

The transcriptome is subject to multiple changes during pathogenesis, including the use of alternate 5' start-sites that can affect transcription levels and output. Current RNA sequencing techniques can assess mRNA levels, but do not robustly detect changes in 5' start-site use. Here, we developed a transcriptome sequencing strategy that detects genome-wide changes in start-site usage (5'RNA-Seq) and applied this methodology to identify regulatory events that occur in hypertrophic cardiomyopathy (HCM). Compared with transcripts from WT mice, 92 genes had altered start-site usage in a mouse model of HCM, including four-and-a-half LIM domains protein 1 (Fhl1). HCM-induced altered transcriptional regulation of Fhl1 resulted in robust myocyte expression of a distinct protein isoform, a response that was conserved in humans with genetic or acquired cardiomyopathies. Genetic ablation of Fhl1 in HCM mice was deleterious, which suggests that Fhl1 transcriptional changes provide salutary effects on stressed myocytes in this disease. Because Fhl1 is a chromosome X-encoded gene, stress-induced changes in its transcription may contribute to gender differences in the clinical severity of HCM. Our findings indicate that 5'RNA-Seq has the potential to identify genome-wide changes in 5' start-site usage that are associated with pathogenic phenotypes.


Asunto(s)
Cardiomiopatía Dilatada/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas con Dominio LIM/genética , Proteínas Musculares/genética , Región de Flanqueo 5' , Animales , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Codón Iniciador , Femenino , Humanos , Masculino , Ratones , Ratones de la Cepa 129 , Mutación Missense , Miocardio/metabolismo , Miocardio/patología , Miocitos Cardíacos/metabolismo , Cadenas Pesadas de Miosina/genética , Análisis de Secuencia de ARN , Transcriptoma
19.
Hum Mutat ; 34(10): 1347-51, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23775976

RESUMEN

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.


Asunto(s)
Anomalías Congénitas/genética , Oído/anomalías , Genes Dominantes , Haploinsuficiencia , Pérdida Auditiva/genética , Proteínas de Homeodominio/genética , Microtia Congénita , Oído Externo/anomalías , Exoma , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Linaje , Fenotipo
20.
Circ Cardiovasc Genet ; 5(5): 503-10, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22949430

RESUMEN

BACKGROUND: Sarcomere mutations cause both dilated cardiomyopathy (DCM) and hypertrophic cardiomyopathy (HCM); however, the steps leading from mutation to disease are not well described. By studying mutation carriers before a clinical diagnosis develops, we characterize the early manifestations of sarcomere mutations in DCM and investigate how these manifestations differ from sarcomere mutations associated with HCM. METHODS AND RESULTS: Sixty-two genotyped individuals in families with sarcomeric DCM underwent clinical evaluation including strain echocardiography. The group included 12 subclinical DCM mutation carriers with normal cardiac dimensions and left ventricular ejection fraction (LVEF ≥55%), 21 overt DCM subjects, and 29 related mutation (-) normal controls. Results were compared with a previously characterized cohort of 60 subclinical HCM subjects (sarcomere mutation carriers without left ventricular hypertrophy). Systolic myocardial tissue velocity, longitudinal, circumferential, and radial strain, and longitudinal and radial strain rate were reduced by 10%-23% in subclinical DCM mutation carriers compared with controls (P<0.001 for all comparisons), after adjusting for age and family relations. No significant differences in diastolic parameters were identified comparing the subclinical and control cohorts. The opposite pattern of contractile abnormalities with reduced diastolic but preserved systolic function was seen in subclinical HCM. CONCLUSIONS: Subtle abnormalities in systolic function are present in subclinical DCM mutation carriers, despite normal left ventricular size and ejection fraction. In contrast, impaired relaxation and preserved systolic function appear to be the predominant early manifestations of sarcomere mutations that lead to HCM. These findings support the theory that the mutation's intrinsic impact on sarcomere function influences whether a dilated or hypertrophic phenotype develops.


Asunto(s)
Cardiomiopatía Dilatada/genética , Contracción Miocárdica/fisiología , Sarcómeros/genética , Adolescente , Adulto , Anciano , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/fisiopatología , Niño , Preescolar , Estudios de Cohortes , Ecocardiografía , Femenino , Genotipo , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Sarcómeros/fisiología , Función Ventricular Izquierda/fisiología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...