Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
PLoS Genet ; 14(5): e1007329, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29795570

RESUMEN

As part of a broader collaborative network of exome sequencing studies, we developed a jointly called data set of 5,685 Ashkenazi Jewish exomes. We make publicly available a resource of site and allele frequencies, which should serve as a reference for medical genetics in the Ashkenazim (hosted in part at https://ibd.broadinstitute.org, also available in gnomAD at http://gnomad.broadinstitute.org). We estimate that 34% of protein-coding alleles present in the Ashkenazi Jewish population at frequencies greater than 0.2% are significantly more frequent (mean 15-fold) than their maximum frequency observed in other reference populations. Arising via a well-described founder effect approximately 30 generations ago, this catalog of enriched alleles can contribute to differences in genetic risk and overall prevalence of diseases between populations. As validation we document 148 AJ enriched protein-altering alleles that overlap with "pathogenic" ClinVar alleles (table available at https://github.com/macarthur-lab/clinvar/blob/master/output/clinvar.tsv), including those that account for 10-100 fold differences in prevalence between AJ and non-AJ populations of some rare diseases, especially recessive conditions, including Gaucher disease (GBA, p.Asn409Ser, 8-fold enrichment); Canavan disease (ASPA, p.Glu285Ala, 12-fold enrichment); and Tay-Sachs disease (HEXA, c.1421+1G>C, 27-fold enrichment; p.Tyr427IlefsTer5, 12-fold enrichment). We next sought to use this catalog, of well-established relevance to Mendelian disease, to explore Crohn's disease, a common disease with an estimated two to four-fold excess prevalence in AJ. We specifically attempt to evaluate whether strong acting rare alleles, particularly protein-truncating or otherwise large effect-size alleles, enriched by the same founder-effect, contribute excess genetic risk to Crohn's disease in AJ, and find that ten rare genetic risk factors in NOD2 and LRRK2 are enriched in AJ (p < 0.005), including several novel contributing alleles, show evidence of association to CD. Independently, we find that genomewide common variant risk defined by GWAS shows a strong difference between AJ and non-AJ European control population samples (0.97 s.d. higher, p<10-16). Taken together, the results suggest coordinated selection in AJ population for higher CD risk alleles in general. The results and approach illustrate the value of exome sequencing data in case-control studies along with reference data sets like ExAC (sites VCF available via FTP at ftp.broadinstitute.org/pub/ExAC_release/release0.3/) to pinpoint genetic variation that contributes to variable disease predisposition across populations.


Asunto(s)
Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad/genética , Judíos/genética , Enfermedades Raras/genética , Algoritmos , Enfermedad de Crohn/epidemiología , Genética de Población , Estudio de Asociación del Genoma Completo , Haplotipos , Humanos , Modelos Genéticos , Epidemiología Molecular , Polimorfismo de Nucleótido Simple , Enfermedades Raras/epidemiología
3.
Blood Press ; 28(4): 239-249, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31044621

RESUMEN

Background: White-coat effect (WCE) confounds diagnosis and treatment of hypertension. The prevalence of white-coat hypertension is higher in Europe and Asia compared to other continents suggesting that genetic factors could play a role. Methods: To study genetic variation affecting WCE, we conducted a two-stage genome-wide association study involving 1343 Finnish subjects. For the discovery stage, we used Genetics of Drug Responsiveness in Essential Hypertension (GENRES) cohort (n = 206), providing the mean WCE values from up to four separate office/ambulatory recordings conducted on placebo. Associations with p values <1 × 10-5 were included in the replication step in three independent cohorts: Haemodynamics in Primary and Secondary Hypertension (DYNAMIC) (n = 182), Finn-Home study (n = 773) and Dietary, Lifestyle and Genetic Determinants of Obesity and Metabolic Syndrome (DILGOM) (n = 182). Results: No single nucleotide polymorphisms reached genome-wide significance for association with either systolic or diastolic WCE. However, two loci provided suggestive evidence for association. A known coronary artery disease risk locus rs2292954 in SPG7 associated with systolic WCE (discovery p value = 2.2 × 10-6, replication p value = 0.03 in Finn-Home, meta-analysis p value 2.6 × 10-4), and rs10033652 in RASGEF1B with diastolic WCE (discovery p value = 4.9 × 10-6, replication p value = 0.04 in DILGOM, meta-analysis p value = 5.0 × 10-3). Conclusion: This study provides evidence for two novel candidate genes, SPG7 and RASGEF1B, associating with WCE. Our results need to be validated in even larger studies carried out in other populations.


Asunto(s)
Estudio de Asociación del Genoma Completo , Hipertensión de la Bata Blanca/genética , ATPasas Asociadas con Actividades Celulares Diversas/genética , Hipertensión Esencial/genética , Femenino , Finlandia , Humanos , Masculino , Síndrome Metabólico/genética , Metaloendopeptidasas/genética , Persona de Mediana Edad , Obesidad/genética , Factores de Intercambio de Guanina Nucleótido ras/genética
4.
BMC Med Genet ; 19(1): 110, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973135

RESUMEN

BACKGROUND: Reduced nocturnal fall (non-dipping) of blood pressure (BP) is a predictor of cardiovascular target organ damage. No genome-wide association studies (GWAS) on BP dipping have been previously reported. METHODS: To study genetic variation affecting BP dipping, we conducted a GWAS in Genetics of Drug Responsiveness in Essential Hypertension (GENRES) cohort (n = 204) using the mean night-to-day BP ratio from up to four ambulatory BP recordings conducted on placebo. Associations with P < 1 × 10- 5 were further tested in two independent cohorts: Haemodynamics in Primary and Secondary Hypertension (DYNAMIC) (n = 183) and Dietary, Lifestyle and Genetic determinants of Obesity and Metabolic Syndrome (DILGOM) (n = 180). We also tested the genome-wide significant single nucleotide polymorphism (SNP) for association with left ventricular hypertrophy in GENRES. RESULTS: In GENRES GWAS, rs4905794 near BCL11B achieved genome-wide significance (ß = - 4.8%, P = 9.6 × 10- 9 for systolic and ß = - 4.3%, P = 2.2 × 10- 6 for diastolic night-to-day BP ratio). Seven additional SNPs in five loci had P values < 1 × 10- 5. The association of rs4905794 did not significantly replicate, even though in DYNAMIC the effect was in the same direction (ß = - 0.8%, P = 0.4 for systolic and ß = - 1.6%, P = 0.13 for diastolic night-to-day BP ratio). In GENRES, the associations remained significant even during administration of four different antihypertensive drugs. In separate analysis in GENRES, rs4905794 was associated with echocardiographic left ventricular mass (ß = - 7.6 g/m2, P = 0.02). CONCLUSIONS: rs4905794 near BCL11B showed evidence for association with nocturnal BP dipping. It also associated with left ventricular mass in GENRES. Combined with earlier data, our results provide support to the idea that BCL11B could play a role in cardiovascular pathophysiology.


Asunto(s)
Presión Sanguínea/genética , Hipertensión/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Ensayos Clínicos como Asunto , Estudios Cruzados , Método Doble Ciego , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Hipertrofia Ventricular Izquierda/genética , Masculino , Persona de Mediana Edad , Obesidad/genética , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Represoras/genética
5.
BMC Med Genet ; 19(1): 56, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29622001

RESUMEN

BACKGROUND: Long QT syndrome (LQTS) is an inherited cardiac disorder predisposing to sudden cardiac death (SCD). We studied factors affecting the clinical course of genetically confirmed patients, in particular those not receiving ß-blocker treatment. In addition, an attempt was made to associate risk of events to specific types of KCNQ1 and KCNH2 mutations. METHODS: A follow-up study covering a mean of 18.6 ± 6.1 years was conducted in 867 genetically confirmed LQT1 and LQT2 patients and 654 non-carrier relatives aged 18-40 years. Cox regression models were used to evaluate the contribution of clinical and genetic risk factors to cardiac events. RESULTS: In mutation carriers, risk factors for cardiac events before initiation of ß-blocker included LQT2 genotype (hazard ratio [HR] = 2.1, p = 0.002), female gender (HR = 3.2, p < 0.001), a cardiac event before the age of 18 years (HR = 5.9, p < 0.001), and QTc ≥500 ms (vs < 470 ms, HR = 2.7, p = 0.001). LQT1 patients carrying the KCNQ1 D317N mutation were at higher risk (HR = 3.0-3.9, p < 0.001-0.03) compared to G589D, c.1129-2A > G and other KCNQ1 mutation carriers after adjusting for gender, QTc duration, and cardiac events before age 18. KCNH2 c.453delC, L552S and R176W mutations associated with lower risk (HR = 0.11-0.23, p < 0.001) than other KCNH2 mutations. CONCLUSIONS: LQT2 (compared to LQT1), female gender, a cardiac event before age 18, and long QT interval increased the risk of cardiac events in LQTS patients aged 18 to 40 years. The nature of the underlying mutation may be associated with risk variation in both LQT1 and LQT2. The identification of high-risk and low-risk mutations may enhance risk stratification.


Asunto(s)
Canal de Potasio ERG1/genética , Canal de Potasio KCNQ1/genética , Síndrome de QT Prolongado/genética , Mutación , Síndrome de Romano-Ward/genética , Antagonistas Adrenérgicos beta/uso terapéutico , Adulto , Femenino , Estudios de Seguimiento , Predisposición Genética a la Enfermedad , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Masculino , Pronóstico , Análisis de Regresión , Factores de Riesgo , Síndrome de Romano-Ward/tratamiento farmacológico , Adulto Joven
6.
Nucleic Acids Res ; 44(7): 3095-104, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26681690

RESUMEN

As the formation of ribonucleoprotein complexes is a major mechanism of angiotensin II type 1 receptor (AT1R) regulation, we sought to identify novel AT1R mRNA binding proteins. By affinity purification and mass spectroscopy, we identified TIA-1. This interaction was confirmed by colocalization of AT1R mRNA and TIA-1 by FISH and immunofluorescence microscopy. In immunoprecipitates of endogenous TIA- 1, reverse transcription-PCR amplified AT1R mRNA. TIA-1 has two binding sites within AT1R 3'-UTR. The binding site proximal to the coding region is glyceraldehyde-3-phosphate dehydrogenase (GAPDH)-dependent whereas the distal binding site is not. TIA-1 functions as a part of endoplasmic reticulum (ER) stress response leading to stress granule (SG) formation and translational silencing. We and others have shown that AT1R expression is increased by ER stress-inducing factors. In unstressed cells, TIA-1 binds to AT1R mRNA and decreases AT1R protein expression. Fluorescence microscopy shows that ER stress induced by thapsigargin leads to the transfer of TIA-1 to SGs. In FISH analysis AT1R mRNA remains in the cytoplasm and no longer colocalizes with TIA-1. Thus, release of TIA-1-mediated suppression by ER stress increases AT1R protein expression. In conclusion, AT1R mRNA is regulated by TIA-1 in a ER stress-dependent manner.


Asunto(s)
Regiones no Traducidas 3' , Estrés del Retículo Endoplásmico , Regulación de la Expresión Génica , Proteínas de Unión a Poli(A)/metabolismo , Receptor de Angiotensina Tipo 1/genética , Sitios de Unión , Células Cultivadas , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Células HEK293 , Humanos , ARN Mensajero/metabolismo , Antígeno Intracelular 1 de las Células T
7.
Europace ; 18(10): 1599-1607, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26705554

RESUMEN

AIMS: Spontaneous Ca2+ release leads to afterdepolarizations and triggered arrhythmia in catecholaminergic polymorphic ventricular tachycardia (CPVT). Irregular Ca2+ release is hypothesized to manifest as slowed depolarization and irregular repolarization. Our goal was to study depolarization and repolarization abnormalities in CPVT, as they remain largely uninvestigated. METHODS AND RESULTS: We studied intracellular Ca2+ handling and action potentials (APs) in an induced pluripotent stem cell (iPSC) model of CPVT. Induced pluripotent stem cell cardiomyocytes from a RyR2-P2328S patient showed increased non-alternating variability of Ca2+ transients in response to isoproterenol. ß-Agonists decreased AP upslope velocity in CPVT cells and in monophasic AP recordings of CPVT patients. We compared 24 h electrocardiograms (ECGs) of 19 CPVT patients carrying RyR2 mutations and 19 healthy controls. Short-term variability (STV) of the QT interval was 6.9 ± 0.5 ms in CPVT patients vs. 5.5 ± 0.4 ms in controls (P < 0.05) and associated with a history of arrhythmic events. Mean T-wave alternans (TWA) was 25 ± 1.4 µV in CPVT patients vs. 31 ± 2.0 µV in controls (P < 0.05). Older CPVT patients showed lower maximal upslope velocity of the ECG R-spike than control patients. CONCLUSION: Catecholaminergic polymorphic ventricular tachycardia patients show higher STV of repolarization but lower TWA on the 24 h ECG than control patients, which is likely to reflect increased non-alternating variability of Ca2+ release by mutant RyR2s as observed in vitro. ß-Agonists slow depolarization in RyR2-mutant cells and in CPVT patients. These findings may constitute a marker of arrhythmogenicity.


Asunto(s)
Potenciales de Acción , Señalización del Calcio , Miocitos Cardíacos/citología , Taquicardia Ventricular/fisiopatología , Agonistas Adrenérgicos beta/uso terapéutico , Adulto , Estudios de Casos y Controles , Electrocardiografía Ambulatoria , Femenino , Finlandia , Humanos , Células Madre Pluripotentes Inducidas/citología , Isoproterenol/uso terapéutico , Masculino , Persona de Mediana Edad , Mutación , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/genética
8.
Ann Noninvasive Electrocardiol ; 21(4): 343-51, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26965448

RESUMEN

BACKGROUND: The cardiac sodium channel Nav 1.5, encoded by the gene SCN5A, is associated with a wide spectrum of hereditary arrhythmias. The gain-of-function mutation p.I141V in SCN5A was identified in a large multigenerational family with exercise-induced polymorphic ventricular arrhythmias. The purpose of this study was to evaluate the molecular and clinical effects of flecainide administration on patients with this syndrome. METHODS: Eleven p.I141V carriers who exhibited frequent multiformic premature ventricular complexes (PVCs) during exercise were subjected to exercise stress tests, both before and after intravenous infusion of 2 mg/kg flecainide. The in vitro effects of flecainide were evaluated using the patch-clamp technique with HEK293 cells expressing the Nav 1.5 channel. RESULTS: The flecainide treatment significantly reduced the frequency of PVCs during and after exercise. Next, the sensitivity of the p.I141V mutant channel to flecainide was compared to that of the wild type channel. Perfusion of flecainide inhibited the peak and window currents in both groups. CONCLUSION: The clinical investigations of the affected patients, as well as the molecular and pharmacological characterization of the SCN5A p.I141V mutation, provide new evidence supporting the association of this mutation with exercise-induced polymorphic ventricular arrhythmias. These data also demonstrate that flecainide may serve as an effective treatment for the defect in Nav 1.5 that leads to an increased sodium window current.


Asunto(s)
Antiarrítmicos/uso terapéutico , Flecainida/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.5/genética , Complejos Prematuros Ventriculares/tratamiento farmacológico , Adulto , Anciano , Electrocardiografía , Prueba de Esfuerzo , Femenino , Células HEK293 , Humanos , Masculino , Persona de Mediana Edad , Mutación , Técnicas de Placa-Clamp
9.
Nucleic Acids Res ; 40(12): 5250-61, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22362742

RESUMEN

Angiotensin II type 1 receptor (AT1R) has a pathophysiological role in hypertension, atherosclerosis and heart failure. Type 2 diabetes is hyperinsulinemic state and a major risk factor for atherosclerosis and hypertension. It is known that hyperinsulinemia upregulates AT1R expression post-transcriptionally by increasing the half-life of AT1R mRNA, but little is known about the mechanism of this effect. In the present study, we first identified AT1R 3'-UTR as a mediator of insulin effect. Using 3'-UTR as a bait, we identified through analysis of insulin-stimulated cell lysates by affinity purification and mass spectrometry HuR as an insulin-regulated AT1R mRNA binding protein. By ribonucleoprotein immunoprecipitation, we found HuR binding to AT1R to be increased by insulin. Overexpression of HuR leads to increased AT1R expression in a 3'-UTR-dependent manner. Both insulin and HuR overexpression stabilize AT1R 3'-UTR and their responsive element within 3'-UTR are located within the same region. Cell fractionation demonstrated that insulin induced HuR translocation from nucleus to cytoplasm increased HuR binding to cytoplasmic AT1R 3'-UTR. Consistent with HuR translocation playing a mechanistic role in HuR effect, a reduction in the cytoplasmic levels of HuR either by silencing of HuR expression or by inhibition of HuR translocation into cytoplasm attenuated insulin response. These results show that HuR translocation to cytoplasm is enhanced by insulin leading to AT1R upregulation through HuR-mediated stabilization of AT1R mRNA.


Asunto(s)
Proteínas ELAV/metabolismo , Regulación de la Expresión Génica , Insulina/farmacología , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/genética , Regiones no Traducidas 3' , Sitios de Unión , Células Cultivadas , Ácidos Grasos Insaturados/farmacología , Células HEK293 , Humanos , Cinética , Transporte de Proteínas/efectos de los fármacos , Receptor de Angiotensina Tipo 1/metabolismo
10.
PLoS One ; 18(4): e0284364, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37053203

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF) plays a key role in diabetic retinopathy (DR). Previously, we have reported an association between mutations in a gene coding for the L-type calcium channel subunit, VEGF and DR. L-type calcium channel blockers (LTCCBs) have been widely used as antihypertensive medication (AHM), but their association with VEGF and DR is still unclear. Therefore, we explored the effect of LTCCBs compared to other AHMs on VEGF concentrations in retinal cells and human serum. Furthermore, we evaluated the association between the use of LTCCBs and the risk of severe diabetic eye disease (SDED). RESEARCH DESIGN AND METHODS: Müller cells (MIO-M1) were cultured as per recommended protocol and treated with LTCCBs and other AHMs. VEGF secreted from cells were collected at 24 hours intervals. In an interventional study, 39 individuals received LTCCBs or other AHM for four weeks with a four-week wash-out placebo period between treatments. VEGF was measured during the medication and placebo periods. Finally, we evaluated the risk of SDED associated with LTCCB usage in 192 individuals from the FinnDiane Study in an observational setting. RESULTS: In the cell cultures, the medium VEGF concentration increased time-dependently after amlodipine (P<0.01) treatment, but not after losartan (P>0.01), or lisinopril (P>0.01). Amlodipine, but no other AHM, increased the serum VEGF concentration (P<0.05) during the interventional clinical study. The usage of LTCCB was not associated with the risk of SDED in the observational study. CONCLUSIONS: LTCCB increases VEGF concentrations in retinal cells and human serum. However, the usage of LTCCBs does not appear to be associated with SDED in adults with type 1 diabetes.


Asunto(s)
Retinopatía Diabética , Factor A de Crecimiento Endotelial Vascular , Adulto , Humanos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Bloqueadores de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/uso terapéutico , Retinopatía Diabética/metabolismo , Antihipertensivos/uso terapéutico , Amlodipino/farmacología
11.
J Cardiovasc Electrophysiol ; 23(2): 194-9, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21954897

RESUMEN

INTRODUCTION: A test involving intravenous infusion of epinephrine has been proposed as a method alternative to exercise stress test in diagnosis of catecholaminergic polymorphic ventricular tachycardia (CPVT). We aimed at estimating the predictive value of intravenous epinephrine administration in CPVT patients with frequent exercise-induced ventricular ectopy. METHODS AND RESULTS: We recruited 81 subjects, including 25 CPVT-linked ryanodine receptor 2 (RYR2) mutation carriers, 11 genetically undefined CPVT patients, and 45 unaffected family members. All subjects underwent a maximal exercise stress test and an intravenous epinephrine infusion test. Exercise stress test was positive in 25 (31%) patients including 14 of 25 (56%) established RYR2-mutation carriers and all 11 (100%) genetically undefined CPVT patients. Epinephrine infusion induced arrhythmias in 3 (12%) RYR2-mutation carriers, 4 (36%) genetically undefined CPVT patients, and 1 (2%) unaffected family member. A total of 18 exercise stress test positive patients did not respond to intravenous epinephrine administration, whereas only 1 epinephrine test responder had a normal exercise stress test. Thus, if exercise stress test is used as a standard, the sensitivity of the epinephrine infusion test is 28% and specificity is 98%. CONCLUSIONS: Intravenous epinephrine infusion has low sensitivity and may not be considered as an alternative method for a maximal exercise stress test in diagnosis of CPVT.


Asunto(s)
Epinefrina , Prueba de Esfuerzo/normas , Polimorfismo Genético/genética , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/genética , Adulto , Epinefrina/administración & dosificación , Prueba de Esfuerzo/métodos , Femenino , Estudios de Seguimiento , Heterocigoto , Humanos , Infusiones Intravenosas , Masculino , Persona de Mediana Edad , Mutación/genética , Valor Predictivo de las Pruebas , Canal Liberador de Calcio Receptor de Rianodina/genética , Taquicardia Ventricular/fisiopatología , Adulto Joven
12.
Heart Rhythm ; 19(9): 1491-1498, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35525424

RESUMEN

BACKGROUND: Long QT syndrome (LQTS) is an inherited arrhythmia disorder characterized by ventricular repolarization abnormalities and a risk of sudden cardiac death. The electrophysiological components generating the high risk of arrhythmias in LQTS are prolonged repolarization, increased dispersion of repolarization, and early afterdepolarizations, which are clinically estimated as QT interval, T-wave peak to T-wave end (TPE) interval, and T2/T1-wave amplitude ratio, respectively. In experimental LQTS type 2 (LQT2) models, ß-blockers decrease dispersion of repolarization and prevent early afterdepolarizations. In clinical studies in patients with LQT2 , ß-blockers are more effective against exercise-induced than arousal-induced cardiac events. OBJECTIVES: The aim of the study was to investigate the effects of ß-blocker therapy on repolarization properties in LQT2. METHODS: QT and TPE intervals and maximal T2/T1-wave amplitude ratios recorded by 24-hour electrocardiograms before and during ß-blocker therapy were evaluated in 25 patients with LQT2. RESULTS: ß-Blocker therapy decreased the maximal T2/T1-wave amplitude ratio from 2.9 ± 1.1 to 1.8 ± 0.7 (P < .001), but did not change the pause-induced T2/T1-wave amplitude ratio. Under medication, abrupt maximal TPE intervals were shorter at heart rates of ≥75 beats/min and maximal QT intervals were shorter at a heart rate of 100 beats/min. CONCLUSION: ß-Blockers stabilize ventricular repolarization in LQT2 by reducing electrocardiographic early afterdepolarizations and by reducing abrupt prolongation of electrocardiographic dispersion of repolarization and ventricular repolarization duration at elevated heart rates. The effect of ß-blockers on pause-induced electrocardiographic early afterdepolarizations is weak. The findings provide electrocardiographic explanation for the protective effects of ß-blockers against exercise-induced cardiac events in LQT2.


Asunto(s)
Electrocardiografía , Síndrome de QT Prolongado , Antagonistas Adrenérgicos beta/farmacología , Antagonistas Adrenérgicos beta/uso terapéutico , Arritmias Cardíacas/tratamiento farmacológico , Electrocardiografía Ambulatoria , Frecuencia Cardíaca , Humanos , Síndrome de QT Prolongado/diagnóstico , Síndrome de QT Prolongado/tratamiento farmacológico
13.
Epigenetics ; 17(11): 1432-1445, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35213289

RESUMEN

Essential hypertension remains the leading risk factor of global disease burden, but its treatment goals are often not met. We investigated whether DNA methylation is associated with antihypertensive responses to a diuretic, a beta-blocker, a calcium channel blocker or an angiotensin receptor antagonist. In addition, since we previously showed an SNP at the transcription start site (TSS) of the catecholamine biosynthesis-related ACY3 gene to associate with blood pressure (BP) response to beta-blockers, we specifically analysed the association of methylation sites close to the ACY3 TSS with BP responses to beta-blockers. We conducted an epigenome-wide association study between leukocyte DNA methylation and BP responses to antihypertensive monotherapies in two hypertensive Finnish cohorts: the GENRES (https://clinicaltrials.gov/ct2/show/NCT03276598; amlodipine 5 mg, bisoprolol 5 mg, hydrochlorothiazide 25 mg, or losartan 50 mg daily) and the LIFE-Fin studies (https://clinicaltrials.gov/ct2/show/NCT00338260; atenolol 50 mg or losartan 50 mg daily). The monotherapy groups consisted of approximately 200 individuals each. We identified 64 methylation sites to suggestively associate (P < 1E-5) with either systolic or diastolic BP responses to a particular study drug in GENRES. These associations did not replicate in LIFE-Fin . Three methylation sites close to the ACY3 TSS were associated with systolic BP responses to bisoprolol in GENRES but not genome-wide significantly (P < 0.05). No robust associations between DNA methylation and BP responses to four different antihypertensive drugs were identified. However, the findings on the methylation sites close to the ACY3 TSS may support the role of ACY3 genetic and epigenetic variation in BP response to bisoprolol.


Asunto(s)
Antihipertensivos , Hipertensión , Humanos , Antihipertensivos/uso terapéutico , Estudios Cruzados , Losartán/uso terapéutico , Bisoprolol/uso terapéutico , Bloqueadores de los Canales de Calcio/uso terapéutico , Atenolol/farmacología , Atenolol/uso terapéutico , Metilación de ADN , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Hidroclorotiazida/uso terapéutico , Amlodipino/uso terapéutico , Diuréticos/uso terapéutico , Antagonistas de Receptores de Angiotensina/uso terapéutico , Método Doble Ciego , Catecolaminas/uso terapéutico , Resultado del Tratamiento
14.
Pharmgenomics Pers Med ; 15: 249-260, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35356681

RESUMEN

Purpose: Bisoprolol is a widely used beta-blocker in patients with cardiovascular diseases. As with other beta-blockers, there is variability in response to bisoprolol, but the underlying reasons for this have not been clearly elucidated. Our aim was to investigate genetic factors that affect bisoprolol pharmacokinetics (PK) and pharmacodynamics (PD), and potentially the clinical outcomes. Patients and Methods: Patients with non-ST elevation acute coronary syndrome were recruited prospectively on admission to hospital and followed up for up to 2 years. Patients from this cohort who were on treatment with bisoprolol, at any dose, had bisoprolol adherence data and a plasma sample, one month after discharge from index hospitalisation were included in the study. Individual bisoprolol clearance values were estimated using population pharmacokinetic modeling. Genome-wide association analysis after genotyping was undertaken using an Illumina HumanOmniExpressExome-8 v1.0 BeadChip array, while CYP2D6 copy number variations were determined by PCR techniques and phenotypes for CYP2D6 and CYP3A were inferred from the genotype. GWAS significant SNPs were analysed for heart rate response to bisoprolol in an independent cohort of hypertensive subjects. Results: Six hundred twenty-two patients on bisoprolol underwent both PK and genome wide analysis. The mean (IQR) of the estimated clearance in this population was 13.6 (10.0-18.0) L/h. Bisoprolol clearance was associated with rs11029955 (p=7.17×10-9) mapped to the region of coiled-coil domain containing 34 region (CCDC34) on chromosome 11, and with rs116702638 (p=2.54×10-8). Each copy of the minor allele of rs11029955 was associated with 2.2 L/h increase in clearance. In an independent cohort of hypertensive subjects, rs11029955 was associated with 24-hour heart rate response to 4-week treatment with bisoprolol (p= 9.3×10-5), but not with rs116702638. Conclusion: A novel locus on the chromosomal region 11p14.1 was associated with bisoprolol clearance in a real-world cohort of patients and was validated in independent cohort with a pharmacodynamic association.

15.
BMC Med Genet ; 12: 11, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21244686

RESUMEN

BACKGROUND: Long QT syndrome (LQTS) is an inherited ion channel disorder manifesting with prolongation of the cardiac repolarization phase and severe ventricular arrhythmias. The common KCNE1 D85N potassium channel variant prolongs QT interval by inhibiting IKs (KCNQ1) and IKr (KCNH2) currents and is therefore a suitable candidate for a modifier gene in LQTS. METHODS: We studied the effect of D85N on age-, sex-, and heart rate-adjusted QT-interval duration by linear regression in LQTS patients carrying the Finnish founder mutations KCNQ1 G589D (n = 492), KCNQ1 IVS7-2A>G (n = 66), KCNH2 L552S (n = 73), and KCNH2 R176W (n = 88). We also investigated the association between D85N and clinical variables reflecting the severity of the disease. RESULTS: D85N was associated with a QT prolongation by 26 ms (SE 8.6, p = 0.003) in males with KCNQ1 G589D (n = 213), but not in females with G589D (n = 279). In linear regression, the interaction between D85N genotype and sex was significant (p = 0.028). Within the KCNQ1 G589D mutation group, KCNE1 D85N carriers were more often probands of the family (p = 0.042) and were more likely to use beta blocker medication (p = 0.010) than non-carriers. The number of D85N carriers in other founder mutation groups was too small to assess its effects. CONCLUSIONS: We propose that KCNE1 D85N is a sex-specific QT-interval modifier in type 1 LQTS and may also associate with increased severity of disease. Our data warrant additional studies on the role of KCNE1 D85N in other genetically homogeneous groups of LQTS patients.


Asunto(s)
Síndrome de QT Prolongado/genética , Polimorfismo Genético , Canales de Potasio con Entrada de Voltaje/genética , Caracteres Sexuales , Antagonistas Adrenérgicos beta/uso terapéutico , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/genética , Niño , Estudios de Cohortes , Electrocardiografía , Femenino , Pruebas Genéticas , Genotipo , Frecuencia Cardíaca/genética , Heterocigoto , Homocigoto , Humanos , Modelos Lineales , Síndrome de QT Prolongado/complicaciones , Síndrome de QT Prolongado/fisiopatología , Masculino , Persona de Mediana Edad , Mutación , Canales de Potasio/genética , Índice de Severidad de la Enfermedad , Factores Sexuales
16.
Nucleic Acids Res ; 37(7): 2346-58, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19246543

RESUMEN

Regulation of angiotensin II type 1 receptor (AT1R) has a pathophysiological role in hypertension, atherosclerosis and heart failure. We started from an observation that the 3'-untranslated region (3'-UTR) of AT1R mRNA suppressed AT1R translation. Using affinity purification for the separation of 3'-UTR-binding proteins and mass spectrometry for their identification, we describe glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as an AT1R 3'-UTR-binding protein. RNA electrophoretic mobility shift analysis with purified GAPDH further demonstrated a direct interaction with the 3'-UTR while GAPDH immunoprecipitation confirmed this interaction with endogenous AT1R mRNA. GAPDH-binding site was mapped to 1-100 of 3'-UTR. GAPDH-bound target mRNAs were identified by expression array hybridization. Analysis of secondary structures shared among GAPDH targets led to the identification of a RNA motif rich in adenines and uracils. Silencing of GAPDH increased the expression of both endogenous and transfected AT1R. Similarly, a decrease in GAPDH expression by H(2)O(2) led to an increased level of AT1R expression. Consistent with GAPDH having a central role in H(2)O(2)-mediated AT1R regulation, both the deletion of GAPDH-binding site and GAPDH overexpression attenuated the effect of H(2)O(2) on AT1R mRNA. Taken together, GAPDH is a translational suppressor of AT1R and mediates the effect of H(2)O(2) on AT1R mRNA.


Asunto(s)
Regiones no Traducidas 3'/metabolismo , Regulación de la Expresión Génica , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Biosíntesis de Proteínas , Receptor de Angiotensina Tipo 1/genética , Secuencia de Bases , Línea Celular , Gliceraldehído-3-Fosfato Deshidrogenasas/antagonistas & inhibidores , Gliceraldehído-3-Fosfato Deshidrogenasas/genética , Humanos , Peróxido de Hidrógeno/farmacología , Datos de Secuencia Molecular , Interferencia de ARN , ARN Mensajero/química , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo
17.
Clin Pharmacol Ther ; 110(3): 723-732, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34231218

RESUMEN

We sought to identify genome-wide variants influencing antihypertensive drug response and adverse cardiovascular outcomes, utilizing data from four randomized controlled trials in the International Consortium for Antihypertensive Pharmacogenomics Studies (ICAPS). Genome-wide antihypertensive drug-single nucleotide polymorphism (SNP) interaction tests for four drug classes (ß-blockers, n = 9,195; calcium channel blockers (CCBs), n = 10,511; thiazide/thiazide-like diuretics, n = 3,516; ACE-inhibitors/ARBs, n = 2,559) and cardiovascular outcomes (incident myocardial infarction, stroke, or death) were analyzed among patients with hypertension of European ancestry. Top SNPs from the meta-analyses were tested for replication of cardiovascular outcomes in an independent Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) study (n = 21,267), blood pressure (BP) response in independent ICAPS studies (n = 1,552), and ethnic validation in African Americans from the Genetics of Hypertension Associated Treatment study (GenHAT; n = 5,115). One signal reached genome-wide significance in the ß-blocker-SNP interaction analysis (rs139945292, Interaction P = 1.56 × 10-8 ). rs139945292 was validated through BP response to ß-blockers, with the T-allele associated with less BP reduction (systolic BP response P = 6 × 10-4 , Beta = 3.09, diastolic BP response P = 5 × 10-3 , Beta = 1.53). The T-allele was also associated with increased adverse cardiovascular risk within the ß-blocker treated patients' subgroup (P = 2.35 × 10-4 , odds ratio = 1.57, 95% confidence interval = 1.23-1.99). The locus showed nominal replication in CHARGE, and consistent directional trends in ß-blocker treated African Americans. rs139945292 is an expression quantitative trait locus for the 50 kb upstream gene NTM (neurotrimin). No SNPs attained genome-wide significance for any other drugs classes. Top SNPs were located near CALB1 (CCB), FLJ367777 (ACE-inhibitor), and CES5AP1 (thiazide). The NTM region is associated with increased risk for adverse cardiovascular outcomes and less BP reduction in ß-blocker treated patients. Further investigation into this region is warranted.


Asunto(s)
Antihipertensivos/uso terapéutico , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/genética , Sistema Cardiovascular/efectos de los fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Hipertensión/tratamiento farmacológico , Negro o Afroamericano/genética , Anciano , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Femenino , Estudio de Asociación del Genoma Completo/métodos , Humanos , Hipertensión/genética , Masculino , Persona de Mediana Edad , Pruebas de Farmacogenómica/métodos , Polimorfismo de Nucleótido Simple/genética
18.
Pharmacogenet Genomics ; 20(5): 342-5, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20300048

RESUMEN

Varying results have been reported on the association of beta-adrenergic receptor polymorphisms with blood pressure (BP) response to beta-blockers. We investigated the influence of ADRB1 Ser49Gly and Arg389Gly, and ADRB2 Gly16Arg and Glu27Gln polymorphisms on ambulatory BP response to bisoprolol and three other antihypertensive drug monotherapies in a placebo-controlled, double-blind, cross-over study with 233 moderately hypertensive men. ADRB1 Ser49Ser homozygotes tended to have a better ambulatory BP response to bisoprolol but the difference was statistically nonsignificant. ADRB1 Arg389Arg homozygotes did not show better BP response to bisoprolol than the other genotypes. There were no significant associations of ADRB2 polymorphisms with BP responses to any of the study drugs. The results from this controlled study in hypertensive men do not support clinical use of common polymorphisms in ADRB1 and ADRB2 in predicting BP responses to beta-blockers or to three other antihypertensive drugs.


Asunto(s)
Antihipertensivos/farmacología , Variación Genética , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Farmacogenética/métodos , Receptores Adrenérgicos beta 1/genética , Receptores Adrenérgicos beta 2/genética , Alelos , Estudios Cruzados , Método Doble Ciego , Homocigoto , Humanos , Masculino , Placebos , Polimorfismo Genético
19.
Pharmacogenet Genomics ; 20(2): 77-85, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20065889

RESUMEN

OBJECTIVE: This pharmacogenetics substudy from the Losartan Intervention for Endpoint reduction in Hypertension study in patients with hypertension and left ventricular hypertrophy (LVH) treated with the angiotensin receptor blocker losartan versus the beta-blocker atenolol for 4.8 years tested whether the insertion/deletion (I/D) polymorphism of the angiotensin-converting enzyme (ACE) gene and 12 other previously well-characterized polymorphisms of hypertension susceptibility genes affected blood pressure reduction, heart rate reduction, cardiovascular events, and/or response to treatment. These polymorphisms were chosen because they could affect blood pressure control or the pharmacological action of losartan or atenolol. METHODS: We genotyped 3503 patients, 1774 on losartan and 1729 on atenolol. RESULTS: ACE and the 12 other genotypes did not affect the reduction in systolic blood pressure, diastolic blood pressure, pulse pressure, mean arterial pressure, or heart rate, or treatment differences between losartan and atenolol on these endpoints, as assessed by general linear models. Also, ACE and the 12 other genotypes did not affect risk of the primary composite endpoint or its components stroke, myocardial infarction, and cardiovascular death, or treatment differences between losartan and atenolol on these endpoints, as assessed by Cox proportional hazards models including baseline Framingham risk score and LVH. CONCLUSION: ACE insertion/deletion and 12 other polymorphisms of hypertension susceptibility genes did not affect blood pressure reduction, heart rate reduction, or cardiovascular events in patients with hypertension and LVH, or treatment differences between losartan and atenolol on these endpoints. These results suggest that the observed effects of losartan versus atenolol in the Losartan Intervention for Endpoint reduction in hypertension study do not depend on ACE and 12 other polymorphisms of hypertension susceptibility genes.


Asunto(s)
Antihipertensivos/uso terapéutico , Hipertensión/tratamiento farmacológico , Hipertrofia Ventricular Izquierda/tratamiento farmacológico , Mutación INDEL/genética , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Polimorfismo Genético , Anciano , Anciano de 80 o más Años , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Determinación de Punto Final , Femenino , Genotipo , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Hipertensión/complicaciones , Hipertensión/fisiopatología , Hipertrofia Ventricular Izquierda/complicaciones , Hipertrofia Ventricular Izquierda/fisiopatología , Losartán/farmacología , Losartán/uso terapéutico , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
20.
Nucleic Acids Res ; 36(13): 4474-87, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18603592

RESUMEN

p100 protein (SND1, Tudor-SN) is a multifunctional protein that functions as a co-activator for several transcription factors, has a role in mRNA processing and participates in RNAi-induced silencing complex (RISC) with yet unknown function. In this study we identified a novel function for p100 as a regulator of angiotensin II type 1 receptor (AT1R) expression. The binding of p100 to AT1R 3'-untranslated region (3'-UTR) via staphylococcal nuclease-like (SN-like) domains increased receptor expression by decreasing the rate of mRNA decay and enhancing its translation. Overexpression of p100 increased AT1R expression, whereas decrease in p100 binding to 3'-UTR either by p100 silencing or by the deletion of p100 binding site downregulated receptor expression. The effect of p100 through AT1R 3'-UTR was independent of Argonaute2 (Ago2), a known p100 partner, and was thus RISC-independent. Nucleotides 118 to 120 of the AT1R 3'-UTR were found to be critical for the binding of p100 to 3'-UTR. In summary, p100 is a multifunctional regulator of gene expression that regulates transcription, mRNA maturation, and as described in this article, also mRNA stability and translation.


Asunto(s)
Regiones no Traducidas 3'/metabolismo , Proteínas Nucleares/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Receptor de Angiotensina Tipo 1/genética , Regiones no Traducidas 3'/química , Animales , Proteínas Argonautas , Sitios de Unión , Línea Celular , Regulación hacia Abajo , Ensayo de Cambio de Movilidad Electroforética , Endonucleasas , Factor 2 Eucariótico de Iniciación/metabolismo , Genes Reporteros , Humanos , Inmunoprecipitación , Luciferasas/análisis , Luciferasas/genética , MicroARNs/metabolismo , Proteínas Nucleares/química , Biosíntesis de Proteínas , Estructura Terciaria de Proteína , Complejo Silenciador Inducido por ARN/metabolismo , Receptor de Angiotensina Tipo 1/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA