Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Genet Genomic Med ; 11(1): e2093, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36369844

RESUMEN

BACKGROUND: Friedreich's ataxia (FRDA) is an autosomal recessive disease, whereby homozygous inheritance of an expanded GAA trinucleotide repeat expansion in the first intron of the FXN gene leads to transcriptional repression of the encoded protein frataxin. FRDA is a progressive neurodegenerative disorder, but the primary cause of death is heart disease which occurs in 60% of the patients. Several functions of frataxin have been proposed, but none of them fully explain why its deficiency causes the FRDA phenotypes nor why the most affected cell types are neurons and cardiomyocytes. METHODS: To investigate, we generated iPSC-derived neurons (iNs) and cardiomyocytes (iCMs) from an FRDA patient and upregulated FXN expression via lentivirus without altering genomic GAA repeats at the FXN locus. RESULTS: RNA-seq and differential gene expression enrichment analyses demonstrated that frataxin deficiency affected the expression of glycolytic pathway genes in neurons and extracellular matrix pathway genes in cardiomyocytes. Genes in these pathways were differentially expressed when compared to a control and restored to control levels when FRDA cells were supplemented with frataxin. CONCLUSIONS: These results offer novel insight into specific roles of frataxin deficiency pathogenesis in neurons and cardiomyocytes.


Asunto(s)
Ataxia de Friedreich , Células Madre Pluripotentes Inducidas , Humanos , Ataxia de Friedreich/genética , Ataxia de Friedreich/metabolismo , Ataxia de Friedreich/patología , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Expresión Génica , Neuronas/metabolismo , Neuronas/patología , Frataxina
3.
Alzheimers Dement ; 18(10): 1930-1942, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-34978147

RESUMEN

We previously demonstrated that in Alzheimer's disease (AD) patients, European apolipoprotein E (APOE) ε4 carriers express significantly more APOE ε4 in their brains than African AD carriers. We examined single nucleotide polymorphisms near APOE with significant frequency differences between African and European/Japanese APOE ε4 haplotypes that could contribute to this difference in expression through regulation. Two enhancer massively parallel reporter assay (MPRA) approaches were performed, supplemented with single fragment reporter assays. We used Capture C analyses to support interactions with the APOE promoter. Introns within TOMM40 showed increased enhancer activity in the European/Japanese versus African haplotypes in astrocytes and microglia. This region overlaps with APOE promoter interactions as assessed by Capture C analysis. Single variant analyses pinpoints rs2075650/rs157581, and rs59007384 as functionally different on these haplotypes. Identification of the mechanisms for differential regulatory function for APOE expression between African and European/Japanese haplotypes could lead to therapeutic targets for APOE ε4 carriers.


Asunto(s)
Enfermedad de Alzheimer , Apolipoproteína E4 , Humanos , Alelos , Enfermedad de Alzheimer/genética , Apolipoproteína E4/genética , Apolipoproteínas E/genética , Población Negra/genética , Genotipo , Haplotipos , Polimorfismo de Nucleótido Simple/genética
4.
Stem Cell Res ; 53: 102394, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34088019

RESUMEN

Dilated Cardiomyopathy (DCM) is one of the main causes of sudden cardiac death and heart failure and is the leading indication for cardiac transplantation worldwide. Mutations in different genes including TTN, MYH7, and LMNA, have been linked to the development of DCM. Here, we generated a human induced pluripotent stem cell (IPSC) line from a DCM patient with a familial history that carries a frameshift mutation in Filamin C (FLNC). The IPSCs show typical morphology of pluripotent cells, expression of pluripotency markers, normal karyotype, and in vitro capacity to differentiate into all three germ layers.


Asunto(s)
Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas , Cardiomiopatía Dilatada/genética , Filaminas/genética , Heterocigoto , Humanos , Mutación
5.
Stem Cell Res ; 54: 102399, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34034220

RESUMEN

Friedreich's Ataxia (FA) is an autosomal recessive disorder with an incidence of 1 in 50,000 in Caucasians. Most cases are caused by a biallelic GAA expansion in the first intron of the Frataxin (FXN) gene. FA is a neurodegenerative disease, but the leading cause of death is hypertrophic cardiomyopathy (HCM) that develops in 60% of the patients. We generated an induced pluripotent stem cell (iPSC) line from an FA patient with a homozygous GAA expansion in intron 1 of the FXN gene. The IPSCs display pluripotent cell morphology, expression of pluripotency markers, normal karyotype, and the capability to differentiate into all three germ layers.


Asunto(s)
Cardiomiopatía Hipertrófica , Ataxia de Friedreich , Células Madre Pluripotentes Inducidas , Enfermedades Neurodegenerativas , Ataxia de Friedreich/genética , Homocigoto , Humanos , Expansión de Repetición de Trinucleótido
6.
Stem Cell Res ; 54: 102396, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34029931

RESUMEN

Mutations in the gene that encodes the nuclear envelope proteins lamin A/C (LMNA) are considered to be a prominent cause of Dilated cardiomyopathy (DCM), a leading cause of heart failure and a prevalent indication for heart transplant. Here we described the generation of induced pluripotent stem cells (iPSCs) from a 53-year-old female with DCM plus progressive conduction disease who carry a heterozygous mutation in LMNA (c.1621C > T, p.R541C). PBMCs isolated from the patient were reprogrammed with Yamanaka factors KOS, KLF4, and c-MYC by the non-integrating sendai virus vector system. The obtained iPSC lines demonstrated normal karyotype and pluripotent identity.


Asunto(s)
Cardiomiopatía Dilatada , Células Madre Pluripotentes Inducidas , Cardiomiopatía Dilatada/genética , Línea Celular , Femenino , Humanos , Factor 4 Similar a Kruppel , Lamina Tipo A/genética , Persona de Mediana Edad , Mutación
7.
Prog Neurobiol ; 201: 102000, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33545232

RESUMEN

Neurodevelopmental disorders are thought to arise from interrupted development of the brain at an early age. Genome-wide association studies (GWAS) have identified hundreds of loci associated with susceptibility to neurodevelopmental disorders; however, which noncoding variants regulate which genes at these loci is often unclear. To implicate neuronal GWAS effector genes, we performed an integrated analysis of transcriptomics, epigenomics and chromatin conformation changes during the development from Induced pluripotent stem cell-derived neuronal progenitor cells (NPCs) into neurons using a combination of high-resolution promoter-focused Capture-C, ATAC-seq and RNA-seq. We observed that gene expression changes during the NPC-to-neuron transition were highly dependent on both promoter accessibility changes and long-range interactions which connect distal cis-regulatory elements (enhancer or silencers) to developmental-stage-specific genes. These genome-scale promoter-cis-regulatory-element atlases implicated 454 neurodevelopmental disorder-associated, putative causal variants mapping to 600 distal targets. These putative effector genes were significantly enriched for pathways involved in the regulation of neuronal development and chromatin organization, with 27 % expressed in a stage-specific manner. The intersection of open chromatin and chromatin conformation revealed development-stage-specific gene regulatory architectures during neuronal differentiation, providing a rich resource to aid characterization of the genetic and developmental basis of neurodevelopmental disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas , Trastornos del Neurodesarrollo , Diferenciación Celular , Cromatina , Estudio de Asociación del Genoma Completo , Humanos , Trastornos del Neurodesarrollo/genética , Neurogénesis , Impresión Tridimensional
8.
Front Physiol ; 12: 778982, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975533

RESUMEN

Mutations in the LMNA gene (encoding lamin A/C) are a significant cause of familial arrhythmogenic cardiomyopathy. Although the penetrance is high, there is considerable phenotypic variability in disease onset, rate of progression, arrhythmias, and severity of myopathy. To begin to address whether this variability stems from specific LMNA mutation sites and types, we generated seven patient-specific induced pluripotent stem cell (iPSC) lines with various LMNA mutations. IPSC-derived cardiomyocytes (iCMs) and cardiac fibroblasts (iCFs) were differentiated from each line for phenotypic analyses. LMNA expression and extracellular signal-regulated kinase pathway activation were perturbed to differing degrees in both iCMs and iCFs from the different lines. Enhanced apoptosis was observed in iCMs but not in iCFs. Markedly diverse irregularities of nuclear membrane morphology were present in iCFs but not iCMs, while iCMs demonstrated variable sarcomere disarray. Heterogenous electrophysiological aberrations assayed by calcium indicator imaging and multi-electrode array suggest differing substrates for arrhythmia that were accompanied by variable ion channel gene expression in the iCMs. Coculture studies suggest enhancement of the LMNA mutation effects on electrophysiological function exerted by iCFs. This study supports the utility of patient-specific iPSC experimental platform in the exploration of mechanistic and phenotypic heterogeneity of different mutations within a cardiac disease-associated gene. The addition of genetically defined coculture of cardiac-constituent non-myocytes further expands the capabilities of this approach.

9.
Front Physiol ; 10: 773, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31333477

RESUMEN

The relationship between cardiac excitability and contractility depends on when Ca2+ influx occurs during the ventricular action potential (AP). In mammals, it is accepted that Ca2+ influx through the L-type Ca2+ channels occurs during AP phase 2. However, in murine models, experimental evidence shows Ca2+ influx takes place during phase 1. Interestingly, Ca2+ influx that activates contraction is highly regulated by the autonomic nervous system. Indeed, autonomic regulation exerts multiple effects on Ca2+ handling and cardiac electrophysiology. In this paper, we explore autonomic regulation in endocardial and epicardial layers of intact beating mice hearts to evaluate their role on cardiac excitability and contractility. We hypothesize that in mouse cardiac ventricles the influx of Ca2+ that triggers excitation-contraction coupling (ECC) does not occur during phase 2. Using pulsed local field fluorescence microscopy and loose patch photolysis, we show sympathetic stimulation by isoproterenol increased the amplitude of Ca2+ transients in both layers. This increase in contractility was driven by an increase in amplitude and duration of the L-type Ca2+ current during phase 1. Interestingly, the ß-adrenergic increase of Ca2+ influx slowed the repolarization of phase 1, suggesting a competition between Ca2+ and K+ currents during this phase. In addition, cAMP activated L-type Ca2+ currents before SR Ca2+ release activated the Na+-Ca2+ exchanger currents, indicating Cav1.2 channels are the initial target of PKA phosphorylation. In contrast, parasympathetic stimulation by carbachol did not have a substantial effect on amplitude and kinetics of endocardial and epicardial Ca2+ transients. However, carbachol transiently decreased the duration of the AP late phase 2 repolarization. The carbachol-induced shortening of phase 2 did not have a considerable effect on ventricular pressure and systolic Ca2+ dynamics. Interestingly, blockade of muscarinic receptors by atropine prolonged the duration of phase 2 indicating that, in isolated hearts, there is an intrinsic release of acetylcholine. In addition, the acceleration of repolarization induced by carbachol was blocked by the acetylcholine-mediated K+ current inhibition. Our results reveal the transmural ramifications of autonomic regulation in intact mice hearts and support our hypothesis that Ca2+ influx that triggers ECC occurs in AP phase 1 and not in phase 2.

10.
PLoS One ; 14(5): e0216928, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31083689

RESUMEN

BACKGROUND: Heart failure (HF) is a leading cause of mortality and is associated with cardiac remodeling. Vulnerability to atrial fibrillation (AF) has been shown to be greater in the early stages of HF, whereas ventricular tachycardia/fibrillation develop during late stages. Here, we explore changes in gene expression that underlie the differential development of fibrosis and structural alterations that predispose to atrial and ventricular arrhythmias. OBJECTIVE: To study transcriptomic changes associated with the development of cardiac arrhythmias in early and late stages of heart failure. METHODS: Dogs were tachy-paced from right ventricle (RV) for 2-3 or 5-6 weeks (early and late HF). We performed transcriptomic analysis of right atria (RA) and RV isolated from control dogs and those in early and late HF. Transcripts with mean relative log2-fold change ≥2 were included in the differential analysis with significance threshold adjusted to p<0.05. RESULTS: Early HF remodeling was more prominent in RA with enrichment of extracellular matrix, circulatory system, wound healing and immune response pathways; many of these processes were not present in RA in late HF. RV showed no signs of remodeling in early HF but enrichment of extracellular matrix and wound healing in late HF. CONCLUSION: Our transcriptomic data indicate significant fibrosis-associated transcriptional changes in RA in early HF and in RV in late HF, with strong atrial predominance. These alterations in gene expression are consistent with the development of arrhythmogenesis in atria in early but not late HF and in the ventricle in late but not early HF.


Asunto(s)
Fibrilación Atrial/genética , Proteínas de la Matriz Extracelular/genética , Insuficiencia Cardíaca/genética , Marcapaso Artificial/veterinaria , Taquicardia Ventricular/genética , Transcriptoma , Animales , Fibrilación Atrial/diagnóstico por imagen , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Modelos Animales de Enfermedad , Perros , Ecocardiografía , Proteínas de la Matriz Extracelular/metabolismo , Fibrosis , Perfilación de la Expresión Génica , Atrios Cardíacos/diagnóstico por imagen , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Ventrículos Cardíacos/diagnóstico por imagen , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Hemodinámica/genética , Inmunidad Innata/genética , Análisis por Micromatrices , Taquicardia Ventricular/diagnóstico por imagen , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatología , Factores de Tiempo , Cicatrización de Heridas/genética
11.
Expert Rev Cardiovasc Ther ; 16(6): 387-404, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29757020

RESUMEN

INTRODUCTION: Brugada syndrome (BrS) is an inherited cardiac arrhythmia syndrome characterized by ST-segment elevation in right precordial ECG leads and associated with sudden cardiac death in young adults. The ECG manifestations of BrS are often concealed but can be unmasked by sodium channel blockers and fever. Areas covered: Implantation of a cardioverter defibrillator (ICD) is first-line therapy for BrS patients presenting with prior cardiac arrest or documented VT. A pharmacological approach to therapy is recommended in cases of electrical storm, as an adjunct to ICD and as preventative therapy. The goal of pharmacological therapy is to produce an inward shift to counter the genetically-induced outward shift of ion channel current flowing during the early phases of the ventricular epicardial action potential. This is accomplished by augmentation of ICa using □□adrenergic agents or phosphodiesterase III inhibitors or via inhibition of Ito. Radiofrequency ablation of the right ventricular outward flow tract epicardium is effective in suppressing arrhythmogenesis in BrS patients experiencing frequent appropriate ICD-shocks. Expert commentary: Understanding of the pathophysiology and approach to therapy of BrS has advanced considerably in recent years, but there remains an urgent need for development of cardio-selective and ion-channel-specific Ito blockers for treatment of BrS.


Asunto(s)
Síndrome de Brugada/terapia , Ablación por Catéter/métodos , Desfibriladores Implantables , Potenciales de Acción , Síndrome de Brugada/fisiopatología , Muerte Súbita Cardíaca , Electrocardiografía , Humanos , Bloqueadores de los Canales de Sodio/administración & dosificación
12.
Stem Cell Reports ; 10(6): 1867-1878, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29731429

RESUMEN

Human induced pluripotent stem cell (hiPSC)-derived atrial cardiomyocytes (CMs) hold great promise for elucidating underlying cellular mechanisms that cause atrial fibrillation (AF). In order to use atrial-like hiPSC-CMs for arrhythmia modeling, it is essential to better understand the molecular and electrophysiological phenotype of these cells. We performed comprehensive molecular, transcriptomic, and electrophysiologic analyses of retinoic acid (RA)-guided hiPSC atrial-like CMs and demonstrate that RA results in differential expression of genes involved in calcium ion homeostasis that directly interact with an RA receptor, chicken ovalbumin upstream promoter-transcription factor 2 (COUP-TFII). We report a mechanism by which RA generates an atrial-like electrophysiologic signature through the downstream regulation of calcium channel gene expression by COUP-TFII and modulation of calcium handling. Collectively, our results provide important insights into the underlying molecular mechanisms that regulate atrial-like hiPSC-CM electrophysiology and support the use of atrial-like CMs derived from hiPSCs to model AF.


Asunto(s)
Calcio/metabolismo , Fenómenos Electrofisiológicos , Atrios Cardíacos/citología , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Potenciales de Acción , Adenosina/metabolismo , Adenosina/farmacología , Biomarcadores , Carbacol/farmacología , Diferenciación Celular/efectos de los fármacos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Transcriptoma , Tretinoina/farmacología
13.
Handb Exp Pharmacol ; 246: 331-354, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28965168

RESUMEN

The voltage-gated cardiac sodium channel (Nav1.5) is a mega-complex comprised of a pore-forming α subunit and 4 ancillary ß-subunits together with numerous protein partners. Genetic defects in the form of rare variants in one or more sodium channel-related genes can cause a loss- or gain-of-function of sodium channel current (INa) leading to the manifestation of various disease phenotypes, including Brugada syndrome, long QT syndrome, progressive cardiac conduction disease, sick sinus syndrome, multifocal ectopic Purkinje-related premature contractions, and atrial fibrillation. Some sodium channelopathies have also been shown to be responsible for sudden infant death syndrome (SIDS). Although these genetic defects often present as pure electrical diseases, recent studies point to a contribution of structural abnormalities to the electrocardiographic and arrhythmic manifestation in some cases, such as dilated cardiomyopathy. The same rare variants in SCN5A or related genes may present with different clinical phenotypes in different individuals and sometimes in members of the same family. Genetic background and epigenetic and environmental factors contribute to the expression of these overlap syndromes. Our goal in this chapter is to review and discuss what is known about the clinical phenotype and genotype of each cardiac sodium channelopathy, and to briefly discuss the underlying mechanisms.


Asunto(s)
Arritmias Cardíacas/etiología , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Animales , Síndrome de Brugada/etiología , Humanos , Síndrome de QT Prolongado/etiología , Canal de Sodio Activado por Voltaje NAV1.5/fisiología
15.
J Physiol Sci ; 67(1): 217-225, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27241707

RESUMEN

In most species androgens shorten the cardiac action potential and reduce the risk of afterdepolarizations. Despite the central role of the rat model in physiological studies, the effects of androgens on the rat heart are still inconclusive. We therefore performed electrophysiological studies on the perfused rat right ventricular free wall. We found a correlation between androgenic activity and a propensity to generate ventricular ectopic action potentials. We also found that the testosterone treatment increased action potential duration at 90 % of repolarization (APD90), while androgenic inhibition increased the time to peak and decreased APD90. We observed that the voltage-gated potassium channel Kv4.3 and the bi-directional membrane ion transporter NCX in the rat myocardium were regulated by androgenic hormones. One possible explanation for these findings is that due to the expression of specific ion channels in the rat myocardium, the action potential response to its hormonal background is different from those described in other experimental models. Our results indicate that androgenic control of NCX expression plays a key role in determining arrhythmogenicity in the rat heart.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Corazón/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Testosterona/farmacología , Andrógenos/farmacología , Animales , Masculino , Miocardio/metabolismo , Ratas , Ratas Wistar
16.
J Mol Cell Cardiol ; 79: 69-78, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25451173

RESUMEN

Abnormal intracellular Ca(2+) cycling plays a key role in cardiac dysfunction, particularly during the setting of ischemia/reperfusion (I/R). During ischemia, there is an increase in cytosolic and sarcoplasmic reticulum (SR) Ca(2+). At the onset of reperfusion, there is a transient and abrupt increase in cytosolic Ca(2++), which occurs timely associated with reperfusion arrhythmias. However, little is known about the subcellular dynamics of Ca(2+) increase during I/R, and a possible role of the SR as a mechanism underlying this increase has been previously overlooked. The aim of the present work is to test two main hypotheses: (1) An increase diastolic Ca(2+) sparks frequency (cspf) constitutes a mayor substrate for the ischemia-induced diastolic Ca(2+) increase; (2) an increase in cytosolic Ca(2+) pro-arrhythmogenic events (Ca(2+) waves), mediates the abrupt diastolic Ca(2+) rise at the onset of reperfusion. We used confocal microscopy on mouse intact hearts loaded with Fluo-4. Hearts were submitted to global I/R (12/30 min) to assess epicardial Ca(2+) sparks in the whole heart. Intact heart sparks were faster than in isolated myocytes whereas cspf was not different. During ischemia, cspf significantly increased relative to preischemia (2.07±0.33 vs. 1.13±0.20 sp/s/100 µm, n=29/34, 7 hearts). Reperfusion significantly changed Ca(2+) sparks kinetics, by prolonging Ca(2+) sparks rise time and decreased cspf. However, it significantly increased Ca(2+) wave frequency relative to ischemia (0.71±0.14 vs. 0.38±0.06 w/s/100 µm, n=32/33, 7 hearts). The results show for the first time the assessment of intact perfused heart Ca(2+) sparks and provides direct evidence of increased Ca(2+) sparks in ischemia that transform into Ca(2+) waves during reperfusion. These waves may constitute a main trigger for reperfusion arrhythmias.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Corazón/fisiopatología , Daño por Reperfusión Miocárdica/metabolismo , Animales , Separación Celular , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Técnicas In Vitro , Cinética , Masculino , Ratones Endogámicos C57BL , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Perfusión , ARN Mensajero/genética , ARN Mensajero/metabolismo , Fracciones Subcelulares/metabolismo
17.
Rev. argent. cardiol ; 82(4): 310-315, ago. 2014. ilus, graf, tab
Artículo en Español | LILACS | ID: lil-734516

RESUMEN

Introducción El síndrome de Brugada es una canalopatía hereditaria con un patrón de transmisión autosómico dominante que presenta un marcado sesgo de género en la expresión del fenotipo, con una proporción hombre:mujer de 9:1. Un modelo celular de la enfermedad propone una distribución heterogénea de la amplitud de la fase 1 del potencial de acción ventricular como la base para el desarrollo del sustrato arritmogénico. Objetivo Investigar el papel de los andrógenos en la regulación de la fase 1 del potencial de acción cardíaco en ratas y sus consecuencias electrofisiológicas en un modelo experimental murino del síndrome de Brugada. Material y métodos Se estudió el control de la expresión génica por andrógenos en células HL-1 y en corazones de rata por reacción en cadena de la polimerasa (PCR) en tiempo real. Para los estudios de electrofisiología se reprodujo un modelo experimental del síndrome de Brugada en un sistema de Langendorff utilizando solución de Tyrode suplementada con pinacidil y terfenadina. Resultados El tratamiento de células HL-1 con dihidrotestosterona produjo un aumento en la expresión del canal del potasio Kv4.3 y del intercambiador de sodio/calcio (NCX). Se evaluó este efecto en ratas tratadas con testosterona y finasterida. La expresión de ambos genes se redujo con la finasterida, mientras que la testosterona aumentó el nivel de ácido ribonucleico mensajero (ARNm) del NCX. La testosterona produjo un acortamiento de la duración del potencial de acción a 90% de la repolarización (APD90) y del tiempo al pico (TTP), lo cual en modelos del síndrome de Brugada se correlaciona con un aumento de la arritmogenicidad. En nuestro modelo, este fenómeno se observó como un incremento en los potenciales de acción ventriculares ectópicos, esporádicos y sostenidos. La frecuencia de aparición de potenciales de acción ectópicos inducida con terfenadina y pinacidil en el grupo control se redujo en un orden de magnitud con el tratamiento con finasterida. Conclusiones: Los andrógenos controlan la expresión de componentes clave del potencial de acción cardíaco, con el resultado de un aumento de la arritmogenicidad. El tratamiento con finasterida revierte estos efectos.


Introduction The Brugada syndrome is an inherited channelopathy with autosomal dominant genotype transmission pattern presenting marked gender bias in phenotype expression, with a male to female ratio of 9:1. A cellular model of the disease suggests a heterogeneous distribution in the phase 1 amplitude of the ventricular action potential as the origin for the development of the arrhythmogenic substrate. Objective The aim of this study was to investigate the role of androgens on the cardiac action potential phase 1 regulation and its electrophysiological consequences in an experimental murine model of Brugada syndrome. Methods Androgen control of gene expression was studied in HL-1 cells and rat hearts using real time polymerase chain reaction (PCR). For the electrophysiological studies, an experimental model of the Brugada syndrome was reproduced in a Langendorff system using Tyrode solution supplemented with pinacidil and terfenadine. Results Treatment of HL-1 cells with di-hydro-testosterone increased the expression of the Kv4.3 potassium channel and the sodium/calcium exchanger (NCX). This effect was assessed in rats treated with testosterone and finasteride. The expression of both genes decreased with finasteride, whereas testosterone increased NCX messenger ribonucleic acid (mRNA) level. Testosterone produced action potential shortening at 90% repolarization (APD90) and decreased time to peak (TTP), which in Brugada syndrome models correlate with increased arrhythmogenesis. In our model, this phenomenon was observed both as an increase of sporadic and sustained ectopic ventricular action potentials. The frequency of ectopic action potentials induced with terfenadine and pinacidil in the control group was reduced by an order of magnitude with finasteride treatment. Conclusions Androgens control the expression of key components of the cardiac action potential resulting in increased arrhythmogenesis. Finasteride treatment reverses these effects.

18.
PLoS One ; 7(8): e43420, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952678

RESUMEN

Allergic asthma is an inflammatory disorder characterized by infiltration of the airway wall with inflammatory cells driven mostly by activation of Th2-lymphocytes, eosinophils and mast cells. There is a link between increased allergy and a reduction of some infections in Western countries. Epidemiological data also show that respiratory allergy is less frequent in people exposed to orofecal and foodborne microbes such as Toxoplasma gondii. We previously showed that both acute and chronic parasite T. gondii infection substantially blocked development of airway inflammation in adult BALB/c mice. Based on the high levels of IFN-γ along with the reduction of Th2 phenotype, we hypothesized that the protective effect might be related to the strong Th1 immune response elicited against the parasite. However, other mechanisms could also be implicated. The possibility that regulatory T cells inhibit allergic diseases has received growing support from both animal and human studies. Here we investigated the cellular mechanisms involved in T. gondii induced protection against allergy. Our results show for the first time that thoracic lymph node cells from mice sensitized during chronic T. gondii infection have suppressor activity. Suppression was detected both in vitro, on allergen specific T cell proliferation and in vivo, on allergic lung inflammation after adoptive transference from infected/sensitized mice to previously sensitized animals. This ability was found to be contact-independent and correlated with high levels of TGF-ß and CD4(+)FoxP3(+) cells.


Asunto(s)
Asma/metabolismo , Hipersensibilidad/metabolismo , Inflamación/patología , Toxoplasmosis/fisiopatología , Animales , Lavado Broncoalveolar , Antígenos CD4/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Interferón gamma/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Noqueados , Hipersensibilidad Respiratoria , Células Th2/citología , Células Th2/parasitología , Toxoplasma/metabolismo , Toxoplasmosis/inmunología , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA