Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros

Banco de datos
Tipo del documento
Asunto de la revista
Intervalo de año de publicación
1.
Pediatr Res ; 89(3): 628-635, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32375165

RESUMEN

BACKGROUND: Information on genetic etiology of pediatric hypertrophic cardiomyopathy (HCM) rarely aids in risk stratification and prediction of disease onset. Little data exist on the association between genetic modifiers and phenotypic expression of myocardial performance, hampering an individual precision medicine approach. METHODS: Single-nucleotide polymorphism genotyping for six previously established disease risk alleles in the hypoxia-inducible factor-1α-vascular endothelial growth factor pathway was performed in a pediatric cohort with HCM. Findings were correlated with echocardiographic parameters of systolic and diastolic myocardial deformation measured by two-dimensional (2-D) speckle-tracking strain. RESULTS: Twenty-five children (6.1 ± 4.5 years; 69% male) with phenotypic and genotypic (60%) HCM were included. Out of six risk alleles tested, one, VEGF1 963GG, showed an association with reduced regional systolic and diastolic left ventricular (LV) myocardial deformation. Moreover, LV average and segmental systolic and diastolic strain and strain rate were significantly reduced, as assessed by the standardized difference, in patients harboring the risk allele. CONCLUSIONS: This is the first study to identify an association between a risk allele in the VEGF pathway and regional LV myocardial function, with the VEGF1 963GG allele associated with reduced LV systolic and diastolic myocardial performance. While studies are needed to link this information to adverse clinical outcomes, this knowledge may help in risk stratification and patient management in HCM. IMPACT: Risk allele in the VEGF gene impacts on LV myocardial deformation phenotype in children with HCM. LV 2-D strain is significantly reduced in patients with risk allele compared to non-risk allele patients within HCM patient groups. Describes that deficiencies in LV myocardial performance in children with HCM are associated with a previously identified risk allele in the angiogenic transcription factor VEGF. First study to identify an association between a risk allele in the VEGF pathway and regional LV myocardial deformation measured by 2-D strain in children with HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Variación Genética , Ventrículos Cardíacos/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Polimorfismo de Nucleótido Simple , Factor A de Crecimiento Endotelial Vascular/genética , Disfunción Ventricular Izquierda/genética , Alelos , Niño , Preescolar , Ecocardiografía , Femenino , Genotipo , Humanos , Masculino , Miocardio/patología , Neovascularización Patológica , Fenotipo , Medicina de Precisión/métodos , Estudios Prospectivos , Riesgo
2.
Arterioscler Thromb Vasc Biol ; 40(5): 1325-1339, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32212852

RESUMEN

OBJECTIVE: Elastin gene deletion or mutation leads to arterial stenoses due to vascular smooth muscle cell (SMC) proliferation. Human induced pluripotent stem cells-derived SMCs can model the elastin insufficiency phenotype in vitro but show only partial rescue with rapamycin. Our objective was to identify drug candidates with superior efficacy in rescuing the SMC phenotype in elastin insufficiency patients. Approach and Results: SMCs generated from induced pluripotent stem cells from 5 elastin insufficiency patients with severe recurrent vascular stenoses (3 Williams syndrome and 2 elastin mutations) were phenotypically immature, hyperproliferative, poorly responsive to endothelin, and exerted reduced tension in 3-dimensional smooth muscle biowires. Elastin mRNA and protein were reduced in SMCs from patients compared to healthy control SMCs. Fourteen drug candidates were tested on patient SMCs. Of the mammalian target of rapamycin inhibitors studied, everolimus restored differentiation, rescued proliferation, and improved endothelin-induced calcium flux in all patient SMCs except one Williams syndrome. Of the calcium channel blockers, verapamil increased SMC differentiation and reduced proliferation in Williams syndrome patient cells but not in elastin mutation patients and had no effect on endothelin response. Combination treatment with everolimus and verapamil was not superior to everolimus alone. Other drug candidates had limited efficacy. CONCLUSIONS: Everolimus caused the most consistent improvement in SMC differentiation, proliferation and in SMC function in patients with both syndromic and nonsyndromic elastin insufficiency, and offers the best candidate for drug repurposing for treatment of elastin insufficiency associated vasculopathy.


Asunto(s)
Arteriopatías Oclusivas/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Elastina/deficiencia , Everolimus/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Síndrome de Williams/metabolismo , Arteriopatías Oclusivas/genética , Arteriopatías Oclusivas/metabolismo , Arteriopatías Oclusivas/patología , Estudios de Casos y Controles , Línea Celular , Constricción Patológica , Elastina/genética , Femenino , Heterocigoto , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Lactante , Masculino , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Síndrome de Williams/complicaciones , Síndrome de Williams/genética
3.
Prenat Diagn ; 39(1): 38-44, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30548283

RESUMEN

OBJECTIVE: To assess the cerebral and vascular development in fetuses with hypoplastic left heart syndrome (HLHS). METHODS: Pregnant women carrying fetuses diagnosed with HLHS who decided to interrupt their pregnancies were included in our study. Aortic size and blood flow were assessed based from fetal echocardiography. Immunohistochemical staining was performed in brain sections obtained from pathology in fetuses with HLHS and control fetuses without heart disease. RESULTS: Twenty-seven midgestation fetal HLHS were included (gestational age, 23.3 ± 3.4 weeks). Head circumference z scores were lower in HLHS fetuses. Middle cerebral artery pulsatility index, a measure of cerebrovascular resistance, was inversely correlated with the ascending aortic z score (P < 0.05). Fetuses with HLHS had lower capillary density in the germinal matrix and their capillaries were larger compared with control fetuses with (P < 0.05). The expression of neuronal differentiation marker, FGFR1, and oligodendrocyte precursor, O4, were lower in HLHS brains compared with controls (P < 0.05). CONCLUSION: Our study identified abnormalities of vascular flow and structural brain abnormalities in fetal HLHS associated with impaired neuronal and oligodendrocyte differentiation, as well as cerebral growth impairment, early in gestation. These findings may be related in part to early vascular abnormalities.


Asunto(s)
Vasos Sanguíneos/embriología , Encéfalo/embriología , Síndrome del Corazón Izquierdo Hipoplásico/embriología , Síndrome del Corazón Izquierdo Hipoplásico/fisiopatología , Encéfalo/anomalías , Encéfalo/patología , Capilares/embriología , Ecocardiografía , Femenino , Enfermedades Fetales , Corazón Fetal , Edad Gestacional , Humanos , Inmunohistoquímica , Arteria Cerebral Media/embriología , Embarazo , Flujo Pulsátil , Ultrasonografía Prenatal
4.
Am J Pathol ; 183(3): 720-34, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23871585

RESUMEN

Hypoplastic left heart syndrome (HLHS) is a severe cardiac malformation characterized by left ventricle (LV) hypoplasia and abnormal LV perfusion and oxygenation. We studied hypoxia-associated injury in fetal HLHS and human pluripotent stem cells during cardiac differentiation to assess the effect of microenvironmental perturbations on fetal cardiac reprogramming. We studied LV myocardial samples from 32 HLHS and 17 structurally normal midgestation fetuses. Compared with controls, the LV in fetal HLHS samples had higher nuclear expression of hypoxia-inducible factor-1α but lower angiogenic growth factor expression, higher expression of oncogenes and transforming growth factor (TGF)-ß1, more DNA damage and senescence with cell cycle arrest, fewer cardiac progenitors, myocytes and endothelial lineages, and increased myofibroblast population (P < 0.05 versus controls). Smooth muscle cells (SMCs) had less DNA damage compared with endothelial cells and myocytes. We recapitulated the fetal phenotype by subjecting human pluripotent stem cells to hypoxia during cardiac differentiation. DNA damage was prevented by treatment with a TGF-ß1 inhibitor (P < 0.05 versus nonhypoxic cells). The hypoplastic LV in fetal HLHS samples demonstrates hypoxia-inducible factor-1α up-regulation, oncogene-associated cellular senescence, TGF-ß1-associated fibrosis and impaired vasculogenesis. The phenotype is recapitulated by subjecting human pluripotent stem cells to hypoxia during cardiac differentiation and rescued by inhibition of TGF-ß1. This finding suggests that hypoxia may reprogram the immature heart and affect differentiation and development.


Asunto(s)
Reprogramación Celular , Senescencia Celular , Feto/patología , Síndrome del Corazón Izquierdo Hipoplásico/embriología , Síndrome del Corazón Izquierdo Hipoplásico/patología , Morfogénesis , Miocardio/patología , Células Madre Pluripotentes/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Transformada , Linaje de la Célula/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Daño del ADN , Cuerpos Embrioides/efectos de los fármacos , Cuerpos Embrioides/patología , Feto/efectos de los fármacos , Feto/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/patología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Morfogénesis/efectos de los fármacos , Mutágenos/toxicidad , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fenotipo , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba/efectos de los fármacos
5.
Cell Rep Med ; 5(5): 101520, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38642550

RESUMEN

Pathogenic variants in MYH7 and MYBPC3 account for the majority of hypertrophic cardiomyopathy (HCM). Targeted drugs like myosin ATPase inhibitors have not been evaluated in children. We generate patient and variant-corrected iPSC-cardiomyocytes (CMs) from pediatric HCM patients harboring single variants in MYH7 (V606M; R453C), MYBPC3 (G148R) or digenic variants (MYBPC3 P955fs, TNNI3 A157V). We also generate CMs harboring MYBPC3 mono- and biallelic variants using CRISPR editing of a healthy control. Compared with isogenic and healthy controls, variant-positive CMs show sarcomere disorganization, higher contractility, calcium transients, and ATPase activity. However, only MYH7 and biallelic MYBPC3 variant-positive CMs show stronger myosin-actin binding. Targeted myosin ATPase inhibitors show complete rescue of the phenotype in variant-positive CMs and in cardiac Biowires to mirror isogenic controls. The response is superior to verapamil or metoprolol. Myosin inhibitors can be effective in genotypically diverse HCM highlighting the need for myosin inhibitor drug trials in pediatric HCM.


Asunto(s)
Miosinas Cardíacas , Cardiomiopatía Hipertrófica , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Cadenas Pesadas de Miosina , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/metabolismo , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Niño , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Genotipo , Miosinas/metabolismo , Miosinas/genética , Masculino , Femenino , Sarcómeros/metabolismo , Sarcómeros/genética
6.
Pediatr Res ; 72(6): 583-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23007030

RESUMEN

BACKGROUND: Risk factors for diastolic dysfunction in hypertrophic cardiomyopathy (HCM) are poorly understood. We investigated the association of variants in hypoxia-response genes with phenotype severity in pediatric HCM. METHODS: A total of 80 unrelated patients <21 y and 14 related members from eight families with HCM were genotyped for six variants associated with vascular endothelial growth factor A (VEGFA) downregulation, or hypoxia-inducible factor A (HIF1A) upregulation. Associations between risk genotypes and left-ventricular (LV) hypertrophy, LV dysfunction, and freedom from myectomy were assessed. Tissue expression was measured in myocardial samples from 17 patients with HCM and 20 patients without HCM. RESULTS: Age at enrollment was 9 ± 5 y (follow-up, 3.1 ± 3.6 y). Risk allele frequency was 67% VEGFA and 92% HIF1A. Risk genotypes were associated with younger age at diagnosis (P < 0.001), septal hypertrophy (P < 0.01), prolonged E-wave deceleration time (EWDT) (P < 0.0001) and isovolumic relaxation time (IVRT) (P < 0.0001), and lower freedom from myectomy (P < 0.05). These associations were seen in sporadic and familial HCM independent of the disease-causing mutation. Risk genotypes were associated with higher myocardial HIF1A and transforming growth factor B1 (TGFB1) expression and increased endothelial-fibroblast transformation (P < 0.05). CONCLUSION: HIF1A-upregulation and/or VEGFA-downregulation genotypes were associated with more severe septal hypertrophy and diastolic dysfunction and may provide genetic markers to improve risk prediction in HCM.


Asunto(s)
Cardiomiopatía Hipertrófica/genética , Variación Genética , Hipoxia/genética , Niño , Preescolar , Humanos , Fenotipo , Polimorfismo de Nucleótido Simple
7.
Pediatr Res ; 72(4): 407-13, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22797143

RESUMEN

BACKGROUND: Hypoxia-inducible factor (HIF1A) regulates the myocardial response to hypoxia and hemodynamic load. We investigated the association of HIF1A variants with right-ventricular (RV) remodeling after tetralogy of Fallot (TOF) repair. METHODS: Children with TOF were genotyped for three single-nucleotide polymorphisms in HIF1A. Genotypes were analyzed for association with RV myocardial protein expression and fibrosis at complete repair (n = 42) and RV dilation, fractional area change, and freedom from pulmonary valve/conduit replacement on follow-up. RESULTS: In 180 TOF patients, mean age at repair was 1.0 ± 0.8 y with follow-up at 9.0 ± 3.5 y; 82% had moderate to severe pulmonary insufficiency. Freedom from RV reinterventions at 5, 10, and 15 y was 92, 84, and 67%, respectively. Patients with more functioning HIF1A alleles had higher transforming growth factor ß1 expression and more fibrosis at initial repair as compared with controls (P < 0.05). During follow-up, patients with more functioning HIF1A alleles showed less RV dilation, better preservation of RV function, and greater freedom from RV reinterventions (P < 0.05). This was confirmed in a replication cohort of 69 patients. CONCLUSION: In children who have had TOF repair, a lower number of functioning HIF1A alleles was associated with RV dilation and dysfunction, suggesting that hypoxia adaptation in unrepaired TOF may influence RV phenotype after repair.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Ventrículos Cardíacos/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Polimorfismo de Nucleótido Simple , Tetralogía de Fallot/cirugía , Remodelación Ventricular/genética , Adaptación Fisiológica , Proteínas Angiogénicas/genética , Proteínas Angiogénicas/metabolismo , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Preescolar , Fibrosis , Estudios de Seguimiento , Frecuencia de los Genes , Genotipo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/patología , Hipertrofia Ventricular Derecha/cirugía , Hipoxia/genética , Hipoxia/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Lactante , Estimación de Kaplan-Meier , Modelos Lineales , Modelos Logísticos , Fenotipo , Estudios Prospectivos , Sistema de Registros , Reoperación , Tetralogía de Fallot/genética , Tetralogía de Fallot/metabolismo , Tetralogía de Fallot/patología , Tetralogía de Fallot/fisiopatología , Factores de Tiempo , Factor de Crecimiento Transformador beta1/metabolismo , Resultado del Tratamiento , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/patología , Disfunción Ventricular Derecha/cirugía
8.
J Clin Invest ; 132(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34990407

RESUMEN

Obstructive arterial diseases, including supravalvular aortic stenosis (SVAS), atherosclerosis, and restenosis, share 2 important features: an abnormal or disrupted elastic lamellae structure and excessive smooth muscle cells (SMCs). However, the relationship between these pathological features is poorly delineated. SVAS is caused by heterozygous loss-of-function, hypomorphic, or deletion mutations in the elastin gene (ELN), and SVAS patients and elastin-mutant mice display increased arterial wall cellularity and luminal obstructions. Pharmacological treatments for SVAS are lacking, as the underlying pathobiology is inadequately defined. Herein, using human aortic vascular cells, mouse models, and aortic samples and SMCs derived from induced pluripotent stem cells of ELN-deficient patients, we demonstrated that elastin insufficiency induced epigenetic changes, upregulating the NOTCH pathway in SMCs. Specifically, reduced elastin increased levels of γ-secretase, activated NOTCH3 intracellular domain, and downstream genes. Notch3 deletion or pharmacological inhibition of γ-secretase attenuated aortic hypermuscularization and stenosis in Eln-/- mutants. Eln-/- mice expressed higher levels of NOTCH ligand JAGGED1 (JAG1) in aortic SMCs and endothelial cells (ECs). Finally, Jag1 deletion in SMCs, but not ECs, mitigated the hypermuscular and stenotic phenotype in the aorta of Eln-/- mice. Our findings reveal that NOTCH3 pathway upregulation induced pathological aortic SMC accumulation during elastin insufficiency and provide potential therapeutic targets for SVAS.


Asunto(s)
Estenosis Aórtica Supravalvular , Elastina , Proteína Jagged-1/metabolismo , Secretasas de la Proteína Precursora del Amiloide , Animales , Aorta/metabolismo , Estenosis Aórtica Supravalvular/genética , Estenosis Aórtica Supravalvular/metabolismo , Estenosis Aórtica Supravalvular/patología , Constricción Patológica , Elastina/genética , Elastina/metabolismo , Células Endoteliales/metabolismo , Humanos , Ratones , Receptor Notch3/genética
9.
Circ Genom Precis Med ; 13(6): e002971, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32960096

RESUMEN

BACKGROUND: Elastin insufficiency causes recurrent vascular stenoses. Hemizygous deletion of the elastin gene (ELN) causes Williams-Beuren syndrome (WBS), while single nucleotide variants in ELN cause nonsyndromic supravalvar aortic stenosis (SVAS). Our objective was to compare cardiovascular disease outcomes in patients with WBS and nonsyndromic SVAS. METHODS: Patients (81 WBS, 42 nonsyndromic SVAS) with cardiovascular disease were included in this retrospective single center study. Freedom from surgical and catheter interventions and reinterventions was compared. Vascular tissue from 8 patients and 6 controls was analyzed for arterial wall architecture. RESULTS: Patients with nonsyndromic SVAS presented at a younger age (median 0.3 [0.4-0.7] years) compared with patients with WBS (1.3 [0.2-3.0] years) and had lower freedom from surgical/catheter interventions compared with patients with WBS, with median event-free survival 1.1 (0.3-5.9) versus 4.7 (2.4-13.3) years, respectively (hazard ratio, 1.62 [95% CI, 1.02-2.56]; P=0.04). Patients with nonsyndromic SVAS also had a lower freedom from reinterventions (P=0.054 by log-rank test). This was related in part to a higher frequency of primary and reinterventions for concomitant valvar aortic stenosis. Histology revealed abnormal intimal and medial thickening, disorganized and fragmented elastic fibers, reduced smooth muscle calponin expression, and increased macrophage marker, CD68, expression in the arterial walls in patients with WBS and nonsyndromic SVAS compared with controls. CONCLUSIONS: Patients with nonsyndromic SVAS require early and more frequent vascular and valvular interventions and reinterventions, in particular for concomitant valvar aortic stenosis compared with patients with WBS. This provides important prognostic information to guide counseling of affected families with cardiovascular disease and may guide primary intervention strategies based on predicted risk of restenosis.


Asunto(s)
Arterias/patología , Sistema Cardiovascular/patología , Elastina/genética , Índice de Severidad de la Enfermedad , Enfermedades Vasculares/diagnóstico , Enfermedades Vasculares/genética , Adolescente , Estenosis Aórtica Supravalvular/genética , Catéteres , Niño , Preescolar , Femenino , Humanos , Lactante , Estimación de Kaplan-Meier , Masculino , Fenotipo , Estenosis de la Válvula Pulmonar/genética , Enfermedades Vasculares/patología , Enfermedades Vasculares/cirugía , Síndrome de Williams/genética
10.
JACC CardioOncol ; 2(5): 690-706, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34396283

RESUMEN

BACKGROUND: Despite known clinical risk factors, predicting anthracycline cardiotoxicity remains challenging. OBJECTIVES: This study sought to develop a clinical and genetic risk prediction model for anthracycline cardiotoxicity in childhood cancer survivors. METHODS: We performed exome sequencing in 289 childhood cancer survivors at least 3 years from anthracycline exposure. In a nested case-control design, 183 case patients with reduced left ventricular ejection fraction despite low-dose doxorubicin (≤250 mg/m2), and 106 control patients with preserved left ventricular ejection fraction despite doxorubicin >250 mg/m2 were selected as extreme phenotypes. Rare/low-frequency variants were collapsed to identify genes differentially enriched for variants between case patients and control patients. The expression levels of 5 top-ranked genes were evaluated in human induced pluripotent stem cell-derived cardiomyocytes, and variant enrichment was confirmed in a replication cohort. Using random forest, a risk prediction model that included genetic and clinical predictors was developed. RESULTS: Thirty-one genes were differentially enriched for variants between case patients and control patients (p < 0.001). Only 42.6% case patients harbored a variant in these genes compared to 89.6% control patients (odds ratio: 0.09; 95% confidence interval: 0.04 to 0.17; p = 3.98 × 10-15). A risk prediction model for cardiotoxicity that included clinical and genetic factors had a higher prediction accuracy and lower misclassification rate compared to the clinical-only model. In vitro inhibition of gene-associated pathways (PI3KR2, ZNF827) provided protection from cardiotoxicity in cardiomyocytes. CONCLUSIONS: Our study identified variants in cardiac injury pathway genes that protect against cardiotoxicity and informed the development of a prediction model for delayed anthracycline cardiotoxicity, and it also provided new targets in autophagy genes for the development of cardio-protective drugs. (Preventing Cardiac Sequelae in Pediatric Cancer Survivors [PCS2]; NCT01805778).

11.
Stem Cell Reports ; 13(6): 1126-1141, 2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31813827

RESUMEN

Induced pluripotent stem cells (iPSC) derived from healthy individuals are important controls for disease-modeling studies. Here we apply precision health to create a high-quality resource of control iPSCs. Footprint-free lines were reprogrammed from four volunteers of the Personal Genome Project Canada (PGPC). Multilineage-directed differentiation efficiently produced functional cortical neurons, cardiomyocytes and hepatocytes. Pilot users demonstrated versatility by generating kidney organoids, T lymphocytes, and sensory neurons. A frameshift knockout was introduced into MYBPC3 and these cardiomyocytes exhibited the expected hypertrophic phenotype. Whole-genome sequencing-based annotation of PGPC lines revealed on average 20 coding variants. Importantly, nearly all annotated PGPC and HipSci lines harbored at least one pre-existing or acquired variant with cardiac, neurological, or other disease associations. Overall, PGPC lines were efficiently differentiated by multiple users into cells from six tissues for disease modeling, and variant-preferred healthy control lines were identified for specific disease settings.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Sistemas CRISPR-Cas , Autorrenovación de las Células , Separación Celular , Ectodermo/citología , Ectodermo/metabolismo , Edición Génica , Humanos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Organoides , Fenotipo , Linfocitos T/metabolismo , Secuenciación Completa del Genoma
12.
Arterioscler Thromb Vasc Biol ; 23(12): 2172-7, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14551159

RESUMEN

OBJECTIVE: Elevated apolipoprotein D (apoD) levels are associated with reduced proliferation of cancer cells. We therefore investigated whether apoD, which occurs free or associated with HDL, suppresses vascular smooth muscle cell (VSMC) proliferation, which is related to the pathobiology of disease. METHODS AND RESULTS: Intense immunoreactivity for apoD was observed in human atherosclerotic plaque but not in normal coronary artery. However, an increase in apoD mRNA was seen in quiescent relative to proliferating fetal lamb aortic VSMCs, and in the rat aortic VSMC line (A10), we demonstrated uptake of apoD from serum. Stable transfection of apoD in A10 cells in the absence of serum did not influence VSMC proliferation assessed by [3H]-thymidine incorporation. ApoD, administered at a dose of 100 ng/mL, completely inhibited basal as well as platelet-derived growth factor (PDGF)-BB-induced VSMC proliferation (P<0.01) but had no effect on fibroblast growth factor-induced VSMC proliferation. ApoD did not suppress PDGF-BB or fibroblast growth factor-2-induced phosphorylation of extracellular signal regulated kinase (ERK) 1/2 but selectively inhibited PDGF-BB-mediated ERK1/2 nuclear translocation. CONCLUSIONS: Our data suggest that apoD selectively modulates the proliferative response of VSMC to growth factors by a mechanism related to nuclear translocation of ERK1/2.


Asunto(s)
Apolipoproteínas/fisiología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Factor de Crecimiento Derivado de Plaquetas/fisiología , Animales , Aorta , Apolipoproteínas/biosíntesis , Apolipoproteínas/genética , Apolipoproteínas/farmacología , Apolipoproteínas D , Arteriosclerosis/metabolismo , Becaplermina , División Celular/efectos de los fármacos , División Celular/genética , División Celular/fisiología , Células Cultivadas , Vasos Coronarios/química , Vasos Coronarios/patología , Vasos Coronarios/fisiología , Proteína Quinasa 3 Activada por Mitógenos , Músculo Liso Vascular/química , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/enzimología , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-sis , ARN Mensajero/biosíntesis , ARN Mensajero/metabolismo , Ratas , Ovinos , Transfección
13.
Stem Cells Transl Med ; 2(1): 2-15, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23283491

RESUMEN

Elastin haploinsufficiency in Williams-Beuren syndrome (WBS) leads to increased vascular smooth muscle cell (SMC) proliferation and stenoses. Our objective was to generate a human induced pluripotent stem (hiPS) cell model for in vitro assessment of the WBS phenotype and to test the ability of candidate agents to rescue the phenotype. hiPS cells were reprogrammed from skin fibroblasts of a WBS patient with aortic and pulmonary stenosis and healthy control BJ fibroblasts using four-factor retrovirus reprogramming and were differentiated into SMCs. Differentiated SMCs were treated with synthetic elastin-binding protein ligand 2 (EBPL2) (20 µg/ml) or the antiproliferative drug rapamycin (100 nM) for 5 days. We generated four WBS induced pluripotent stem (iPS) cell lines that expressed pluripotency genes and differentiated into all three germ layers. Directed differentiation of BJ iPS cells yielded an 85%-92% pure SMC population that expressed differentiated SMC markers, were functionally contractile, and formed tube-like structures on three-dimensional gel assay. Unlike BJ iPS cells, WBS iPS cells generated immature SMCs that were highly proliferative, showed lower expression of differentiated SMC markers, reduced response to the vasoactive agonists, carbachol and endothelin-1, impaired vascular tube formation, and reduced calcium flux. EBPL2 partially rescued and rapamycin fully rescued the abnormal SMC phenotype by decreasing the smooth muscle proliferation rate and enhancing differentiation and tube formation. WBS iPS cell-derived SMCs demonstrate an immature proliferative phenotype with reduced functional and contractile properties, thereby recapitulating the human disease phenotype. The ability of rapamycin to rescue the phenotype provides an attractive therapeutic candidate for patients with WBS and vascular stenoses.


Asunto(s)
Células Madre Pluripotentes Inducidas/fisiología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Síndrome de Williams/patología , Antígenos de Diferenciación/genética , Antígenos de Diferenciación/metabolismo , Señalización del Calcio , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Fibroblastos/metabolismo , Fibroblastos/fisiología , Hemicigoto , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/biosíntesis , Masculino , Contracción Muscular , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Fragmentos de Péptidos/farmacología , Fenotipo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Recombinantes/biosíntesis , Factores de Transcripción SOXB1/biosíntesis , Análisis de Secuencia de ADN , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Transcriptoma/efectos de los fármacos , Síndrome de Williams/genética
14.
J Cell Sci ; 122(Pt 9): 1441-51, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19366727

RESUMEN

Previously, we related fibronectin (Fn1) mRNA translation to an interaction between an AU-rich element in the Fn1 3' UTR and light chain 3 (LC3) of microtubule-associated proteins 1A and 1B. Since human fibrosarcoma (HT1080) cells produce little fibronectin and LC3, we used these cells to investigate how LC3-mediated Fn1 mRNA translation might alter tumor growth. Transfection of HT1080 cells with LC3 enhanced fibronectin mRNA translation. Using polysome analysis and RNA-binding assays, we show that elevated levels of translation depend on an interaction between a triple arginine motif in LC3 and the AU-rich element in Fn1 mRNA. Wild-type but not mutant LC3 accelerated HT1080 cell growth in culture and when implanted in SCID mice. Comparison of WT LC3 with vector-transfected HT1080 cells revealed increased fibronectin-dependent proliferation, adhesion and invasion. Microarray analysis of genes differentially expressed in WT and vector-transfected control cells indicated enhanced expression of connective tissue growth factor (CTGF). Using siRNA, we show that enhanced expression of CTGF is fibronectin dependent and that LC3-mediated adhesion, invasion and proliferation are CTGF dependent. Expression profiling of soft tissue tumors revealed increased expression of both LC3 and CTGF in some locally invasive tumor types.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Fibronectinas , Fibrosarcoma , Proteínas Asociadas a Microtúbulos/metabolismo , Biosíntesis de Proteínas , Animales , Adhesión Celular , Línea Celular , Proliferación Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Perfilación de la Expresión Génica , Humanos , Ratones , Proteínas Asociadas a Microtúbulos/genética , Invasividad Neoplásica , Análisis de Secuencia por Matrices de Oligonucleótidos , Polirribosomas/metabolismo , Ratas
15.
J Am Coll Cardiol ; 53(20): 1909-17, 2009 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-19442892

RESUMEN

OBJECTIVES: This study sought to investigate the influence of recipient renin-angiotensin-aldosterone system (RAAS) genotype on cardiac function, rejection, and outcomes after heart transplantation. BACKGROUND: The RAAS influences cardiac function and up-regulates inflammatory/immune pathways. Little is known about the effect of recipient RAAS polymorphisms in pediatric cardiac transplantation. METHODS: Patients <25 years of age, after cardiac transplantation, were enrolled (2003 to 2008) and genotyped for polymorphisms in genes associated with RAAS upregulation: AGT-G, ACE-D, AGTR1-C, CYP11B2-G, and CMA-A. Presence of at least 1 high-risk allele was defined as a high-risk genotype. Univariable and multivariable associations between genotypes and outcomes were assessed in time-dependent models using survival, logistic, or linear regression models. Biopsy samples were immunostained for interleukin (IL)-6, transforming growth factor (TGF)-beta, and tumor necrosis factor (TNF)-alpha during rejection and quiescence. RESULTS: A total of 145 patients were studied, 103 primary cohort and 42 replication cohort; 81% had rejection, 51% had graft dysfunction, and 13% had vasculopathy, 7% died and 8% underwent re-transplantation. A higher number of homozygous high-risk RAAS genotypes was associated with a higher risk of graft dysfunction (hazard ratio [HR]: 1.5, p = 0.02) and a higher probability of death (HR: 2.5, p = 0.04). The number of heterozygous high-risk RAAS genotypes was associated with frequency of rejection (+0.096 events/year, p < 0.001) and rejection-associated graft dysfunction (+0.37 events/year, p = 0.002). IL-6 and TGF-beta were markedly upregulated during rejection in patients with >/=2 high-risk RAAS genotypes. CONCLUSIONS: Recipient RAAS polymorphisms are associated with a higher risk of rejection, graft cytokine expression, graft dysfunction, and a higher mortality after cardiac transplantation. This may have implications for use of RAAS inhibitors in high-risk patients after transplantation.


Asunto(s)
ADN/genética , Regulación de la Expresión Génica , Rechazo de Injerto/genética , Trasplante de Corazón , Interleucina-6/genética , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador beta/genética , Biopsia , Cardiomiopatías/cirugía , Niño , Ecocardiografía , Femenino , Estudios de Seguimiento , Genotipo , Rechazo de Injerto/diagnóstico , Rechazo de Injerto/metabolismo , Cardiopatías Congénitas/cirugía , Humanos , Interleucina-6/biosíntesis , Masculino , Miocardio/metabolismo , Miocardio/patología , Reacción en Cadena de la Polimerasa , Estudios Retrospectivos , Factores de Riesgo , Factores de Tiempo , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Trasplante Homólogo
16.
Can J Physiol Pharmacol ; 86(9): 633-42, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18758513

RESUMEN

Bradykinin 2 receptor (B2R) deficiency predisposes to cardiac hypertrophy and hypertension. The pathways mediating these effects are not known. Two-month-old B2R knockout (KO) and wild-type (WT) mice were assigned to 4 treatment groups (n = 12-14/group): control (vehicle); nitro-L-arginine methyl ester (L-NAME) an NO synthase inhibitor; simvastatin (SIM), an NO synthase activator; and SIM+L-NAME. Serial echocardiography was performed and blood pressure (BP) at 6 weeks was recorded using a micromanometer. Myocardial eNOS and mitogen-activated protein kinase (MAPK, including ERK, p38, and JNK) protein expression were measured. Results showed that (i) B2RKO mice had significantly lower ejection fraction than did WT mice (61% +/- 1% vs. 73% +/- 1%), lower myocardial eNOS and phospho-eNOS, normal systolic BP, and higher LV mass, phospho-p38, and JNK; (ii) L-NAME increased systolic BP in KO mice (117 +/- 19 mm Hg) but not in WT mice and exacerbated LV hypertrophy and dysfunction; and (iii) in KO mice, SIM decreased hypertrophy, p38, and JNK, improved function, increased capillary eNOS and phospho-eNOS, and prevented L-NAME-induced LV hypertrophy without lowering BP. We conclude that disruption of the B2R causes maladaptive cardiac hypertrophy with myocardial eNOS downregulation and MAPK upregulation. SIM reverses these abnormalities and prevents the development of primary cardiac hypertrophy as well as hypertrophy secondary to L-NAME-induced hypertension.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Receptor de Bradiquinina B2/genética , Receptor de Bradiquinina B2/fisiología , Simvastatina/farmacología , Animales , Presión Sanguínea/fisiología , Cardiomegalia/diagnóstico por imagen , Inhibidores Enzimáticos/farmacología , Hipertrofia Ventricular Izquierda/diagnóstico por imagen , Hipertrofia Ventricular Izquierda/genética , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/fisiología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/ultraestructura , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Óxido Nítrico Sintasa de Tipo III/fisiología , Fosforilación , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Volumen Sistólico/fisiología , Ultrasonografía
17.
Ann Thorac Surg ; 81(5): 1817-23, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16631678

RESUMEN

BACKGROUND: Mitochondrial dysfunction may contribute to early postoperative neonatal heart dysfunction. Diazoxide, a mitochondrial-selective adenosine triphosphate-sensitive potassium-channel opener, is associated with mitochondrial preservation after cardioplegic arrest. We evaluated the mitochondrial-protective effect of diazoxide in terms of mitochondrial structure and function after neonatal cardioplegic arrest. METHODS: Newborn piglets (age, approximately 14 days) underwent cardiopulmonary bypass and 60 minutes of cardioplegic arrest using cold crystalloid cardioplegic solution (CCP, n = 5) or cold crystalloid cardioplegic solution with diazoxide (CCP+D, n = 5). After 6 hours of recovery, myocardium was harvested. Control myocardium from piglets that did not undergo cardiopulmonary bypass (non-CPB, n = 5) was obtained. RESULTS: Cardioplegic arrest was associated with translocation of Bax to the mitochondria, which was not prevented by diazoxide. Nevertheless, by electron microscopy, CCP-associated remodeling of mitochondrial structure was subjectively diminished in CCP+D hearts. In addition, CCP-associated mitochondrial permeabilization and cytochrome c release into the cytosol were prevented with CCP+D (p < 0.05). In vitro oxygen consumption of isolated mitochondria demonstrated deficient function of mitochondrial complex I in CCP, but it was preserved in the CCP+D myocardial mitochondria (p < 0.05). Complex II and IV activity was not different among groups. In parallel with impaired complex I function, the cardiac adenosine triphosphate content was diminished in CCP hearts, but well maintained in CCP+D hearts (p < 0.05). CONCLUSIONS: Although early apoptotic signaling events (Bax translocation) are not prevented by diazoxide, addition of the mitochondrial-selective adenosine triphosphate-sensitive potassium-channel opener to the cardioplegic solution is associated with protection of mitochondrial structural and functional integrity in a clinically relevant model of neonatal cardiac surgery. The mitochondrial-protective effects of diazoxide may contribute to improved postoperative myocardial function in the neonate.


Asunto(s)
Diazóxido/farmacología , Paro Cardíaco Inducido , Mitocondrias Cardíacas , Vasodilatadores/farmacología , Adenosina Trifosfato/análisis , Animales , Animales Recién Nacidos , Apoptosis/fisiología , Citocromos c/metabolismo , Hemodinámica , Mitocondrias Cardíacas/fisiología , Mitocondrias Cardíacas/ultraestructura , Consumo de Oxígeno , Canales de Potasio , Porcinos , Translocación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA