Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443158

RESUMEN

The sinus node (SAN) is the primary pacemaker of the human heart, and abnormalities in its structure or function cause sick sinus syndrome, the most common reason for electronic pacemaker implantation. Here we report that transcription factor GATA6, whose mutations in humans are linked to arrhythmia, is highly expressed in the SAN and its haploinsufficiency in mice results in hypoplastic SANs and rhythm abnormalities. Cell-specific deletion reveals a requirement for GATA6 in various SAN lineages. Mechanistically, GATA6 directly activates key regulators of the SAN genetic program in conduction and nonconduction cells, such as TBX3 and EDN1, respectively. The data identify GATA6 as an important regulator of the SAN and provide a molecular basis for understanding the conduction abnormalities associated with GATA6 mutations in humans. They also suggest that GATA6 may be a potential modifier of the cardiac pacemaker.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Frecuencia Cardíaca/fisiología , Nodo Sinoatrial/embriología , Animales , Arritmias Cardíacas/fisiopatología , Diferenciación Celular/genética , Factor de Transcripción GATA6/genética , Regulación del Desarrollo de la Expresión Génica/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis , Nodo Sinoatrial/fisiología , Proteínas de Dominio T Box/genética
2.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892161

RESUMEN

Males and females exhibit intrinsic differences in the structure and function of the heart, while the prevalence and severity of cardiovascular disease vary in the two sexes. However, the mechanisms of this sex-based dimorphism are yet to be elucidated. Sex chromosomes and sex hormones are the main contributors to sex-based differences in cardiac physiology and pathophysiology. In recent years, the advances in induced pluripotent stem cell-derived cardiac models and multi-omic approaches have enabled a more comprehensive understanding of the sex-specific differences in the human heart. Here, we provide an overview of the roles of these two factors throughout cardiac development and explore the sex hormone signaling pathways involved. We will also discuss how the employment of stem cell-based cardiac models and single-cell RNA sequencing help us further investigate sex differences in healthy and diseased hearts.


Asunto(s)
Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , Femenino , Masculino , Caracteres Sexuales , Hormonas Esteroides Gonadales/metabolismo , Diferenciación Celular , Animales , Corazón/fisiología , Cromosomas Sexuales/genética , Transducción de Señal
3.
IUBMB Life ; 75(9): 717-731, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-36988388

RESUMEN

Inhibition of the mammalian target of rapamycin (mTOR) with the macrolide rapamycin or pharmacological suppression of KATP channel opening translated to scar expansion of the myocardial infarcted (MI) adult female rodent heart. The present study tested the hypotheses that rapamycin-mediated scar expansion was sex-specific and that mTOR signaling directly influenced KATP channel subunit expression/activity. Scar size was significantly larger in post-MI male rats as compared to the previous data reported in post-MI female rats. The reported scar expansion of rapamycin-treated post-MI female rats was not observed following the administration of the macrolide to post-MI male rats. Protein levels of the KATP channel subunits Kir6.2 and SUR2A and phosphorylation of the serine2448 residue of mTOR were similar in the normal heart of adult male and female rats. By contrast, greater tuberin inactivation characterized by the increased phosphorylation of the threonine1462 residue and reduced raptor protein levels were identified in the normal heart of adult female rats. Rapamycin pretreatment of phorbol 12,13-dibutyrate (PDBu)-treated neonatal rat ventricular cardiomyocytes (NNVMs) suppressed hypertrophy, inhibited p70S6K phosphorylation, and attenuated SUR2A protein upregulation. In the presence of low ATP levels, KATP channel activity detected in untreated NNVMs was significantly attenuated in PDBu-induced hypertrophied NNVMs via a rapamycin-independent pathway. Thus, rapamycin administration to post-MI rats unmasked a sex-specific pattern of scar expansion and mTOR signaling in PDBu-induced hypertrophied NNVMs significantly increased SUR2A protein levels. However, the biological advantage associated with SUR2A protein upregulation was partially offset by an mTOR-independent pathway that attenuated KATP channel activity in PDBu-induced hypertrophied NNVMs.


Asunto(s)
Infarto del Miocardio , Sirolimus , Femenino , Masculino , Animales , Ratas , Sirolimus/farmacología , Cicatriz , Serina-Treonina Quinasas TOR/genética , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/genética , Macrólidos , Antibacterianos , Adenosina Trifosfato , Mamíferos
4.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36142639

RESUMEN

Male sex is one of the most important risk factors of atrial fibrillation (AF), with the incidence in men being almost double that in women. However, the reasons for this sex difference are unknown. Accordingly, in this study, we sought to determine whether there are sex differences in intracellular Ca2+ homeostasis in mouse atrial myocytes that might help explain male predisposition to AF. AF susceptibility was assessed in male (M) and female (F) mice (4-5 months old) using programmed electrical stimulation (EPS) protocols. Males were 50% more likely to develop AF. The Ca2+ transient amplitude was 28% higher in male atrial myocytes. Spontaneous systolic and diastolic Ca2+ releases, which are known sources of triggered activity, were significantly more frequent in males than females. The time to 90% decay of Ca2+ transient was faster in males. Males had 54% higher Na+-Ca2+ exchanger (NCX1) current density, and its expression was also more abundant. L-type Ca2+ current (ICaL) was recorded with and without BAPTA, a Ca2+ chelator. ICaL density was lower in males only in the absence of BAPTA, suggesting stronger Ca2+-dependent inactivation in males. CaV1.2 expression was similar between sexes. This study reports major sex differences in Ca2+ homeostasis in mouse atria, with larger Ca2+ transients and enhanced NCX1 function and expression in males resulting in more spontaneous Ca2+ releases. These sex differences may contribute to male susceptibility to AF by promoting triggered activity.


Asunto(s)
Fibrilación Atrial , Intercambiador de Sodio-Calcio/metabolismo , Animales , Fibrilación Atrial/metabolismo , Calcio/metabolismo , Quelantes/metabolismo , Ácido Egtácico/análogos & derivados , Femenino , Atrios Cardíacos/metabolismo , Humanos , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , Retículo Sarcoplasmático/metabolismo , Caracteres Sexuales
5.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36142603

RESUMEN

Men have a higher risk of developing atrial fibrillation (AF) than women, though the reason for this is unknown. Here, we compared atrial electrical and structural properties in male and female mice and explored the contribution of sex hormones. Cellular electrophysiological studies revealed that action potential configuration, Na+ and K+ currents were similar in atrial myocytes from male and female mice (4-5 months). Immunofluorescence showed that male atrial myocytes had more lateralization of connexins 40 (63 ± 4%) and 43 (66 ± 4%) than females (Cx40: 45 ± 4%, p = 0.006; Cx43: 44 ± 4%, p = 0.002), with no difference in mRNA expression. Atrial mass was significantly higher in males. Atrial myocyte dimensions were also larger in males. Atrial fibrosis was low and similar between sexes. Orchiectomy (ORC) abolished sex differences in AF susceptibility (M: 65%; ORC: 38%, p = 0.050) by reducing connexin lateralization and myocyte dimensions. Ovariectomy (OVX) did not influence AF susceptibility (F: 42%; OVX: 33%). This study shows that prior to the development of age-related remodeling, male mice have more connexin lateralization and larger atria and atrial myocyte than females. Orchiectomy reduced AF susceptibility in males by decreasing connexin lateralization and atrial myocyte size, supporting a role for androgens. These sex differences in AF substrates may contribute to male predisposition to AF.


Asunto(s)
Fibrilación Atrial , Conexina 43/metabolismo , Animales , Fibrilación Atrial/metabolismo , Conexina 43/genética , Conexinas/genética , Conexinas/metabolismo , Femenino , Atrios Cardíacos/metabolismo , Humanos , Masculino , Ratones , ARN Mensajero/metabolismo , Caracteres Sexuales
6.
J Mol Cell Cardiol ; 147: 27-34, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32798536

RESUMEN

BACKGROUND: The heart rate progressively increases throughout pregnancy, reaching a maximum in the third trimester. This elevated heart rate is also present in pregnant mice and is associated with accelerated automaticity, higher density of the pacemaker current If and changes in Ca2+ homeostasis in sinoatrial node (SAN) cells. Strong evidence has also been provided showing that 17ß-estradiol (E2) and estrogen receptor α (ERα) regulate heart rate. Accordingly, we sought to determine whether E2 levels found in late pregnancy cause the increased cardiac automaticity associated with pregnancy. METHODS AND RESULTS: Voltage- and current-clamp experiments were carried out on SAN cells isolated from female mice lacking estrogen receptor alpha (ERKOα) or beta (ERKOß) receiving chronic E2 treatment mimicking late pregnancy concentrations. E2 treatment significantly increased the action potential rate (284 ± 24 bpm, +E2 354 ± 23 bpm, p = 0.040) and the density of If (+52%) in SAN cells from ERKOß mice. However, If density remains unchanged in SAN cells from E2-treated ERKOα mice. Additionally, E2 also increased If density (+67%) in nodal-like human-induced pluripotent stem cell-derived cardiomyocytes (N-hiPSC-CM), recapitulating in a human SAN cell model the effect produced in mice. However, the L-type calcium current (ICaL) and Ca2+ transients, examined using N-hiPSC-CM and SAN cells respectively, were not affected by E2, indicating that other mechanisms contribute to changes observed in these parameters during pregnancy. CONCLUSION: The accelerated SAN automaticity observed in E2-treated ERKOß mice is explained by an increased If density mediated by ERα, demonstrating that E2 plays a major role in regulating SAN function during pregnancy.


Asunto(s)
Estrógenos/farmacología , Corazón/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Cafeína/farmacología , Canales de Calcio Tipo L/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Femenino , Corazón/efectos de los fármacos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Embarazo , Nodo Sinoatrial/efectos de los fármacos , Nodo Sinoatrial/metabolismo
7.
J Biol Chem ; 291(9): 4826-43, 2016 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-26742847

RESUMEN

Alteration in the L-type current density is one aspect of the electrical remodeling observed in patients suffering from cardiac arrhythmias. Changes in channel function could result from variations in the protein biogenesis, stability, post-translational modification, and/or trafficking in any of the regulatory subunits forming cardiac L-type Ca(2+) channel complexes. CaVα2δ1 is potentially the most heavily N-glycosylated subunit in the cardiac L-type CaV1.2 channel complex. Here, we show that enzymatic removal of N-glycans produced a 50-kDa shift in the mobility of cardiac and recombinant CaVα2δ1 proteins. This change was also observed upon simultaneous mutation of the 16 Asn sites. Nonetheless, the mutation of only 6/16 sites was sufficient to significantly 1) reduce the steady-state cell surface fluorescence of CaVα2δ1 as characterized by two-color flow cytometry assays and confocal imaging; 2) decrease protein stability estimated from cycloheximide chase assays; and 3) prevent the CaVα2δ1-mediated increase in the peak current density and voltage-dependent gating of CaV1.2. Reversing the N348Q and N812Q mutations in the non-operational sextuplet Asn mutant protein partially restored CaVα2δ1 function. Single mutation N663Q and double mutations N348Q/N468Q, N348Q/N812Q, and N468Q/N812Q decreased protein stability/synthesis and nearly abolished steady-state cell surface density of CaVα2δ1 as well as the CaVα2δ1-induced up-regulation of L-type currents. These results demonstrate that Asn-663 and to a lesser extent Asn-348, Asn-468, and Asn-812 contribute to protein stability/synthesis of CaVα2δ1, and furthermore that N-glycosylation of CaVα2δ1 is essential to produce functional L-type Ca(2+) channels.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Membrana Celular/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional , Sustitución de Aminoácidos , Animales , Animales Recién Nacidos , Canales de Calcio Tipo L/genética , Membrana Celular/química , Células Cultivadas , Glicosilación , Células HEK293 , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Peso Molecular , Mutagénesis Sitio-Dirigida , Miocitos Cardíacos/citología , Mutación Puntual , Estabilidad Proteica , Conejos , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/metabolismo , Propiedades de Superficie
8.
J Mol Cell Cardiol ; 86: 85-94, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26205295

RESUMEN

BACKGROUND AND OBJECTIVE: We have previously shown that androgens upregulate cardiac K(+) channels and shorten repolarization. However, the effects that estrogens (E2) and estrogen receptors (ER) might have on the various repolarizing K(+) currents and underlying ion channels remain incompletely understood. Accordingly, our objective was to verify whether and how E2 and its ERs subtypes influence these K(+) currents. METHODS AND RESULTS: In order to examine the influence of E2 and ERs on K(+) currents we drastically lowered the E2 level through ovariectomy (OVX; 74% reduction vs CTL) and in parallel, we used female mice lacking either ERα (ERαKO) or ERß (ERßKO). In OVX mice, results showed a specific increase of 35% in the density of the Ca(2+)-independent transient outward K(+) current (Ito) compared to CTL. Western blots showed increase in Kv4.2 and Kv4.3 sarcolemmal protein expression while qPCR revealed higher mRNA expression of only Kv4.3 in OVX mice. This upregulation of Ito was correlated with a shorter ventricular action potential duration and QTc interval. In ERαKO but not ERßKO mice, the mRNA of Kv4.3 was selectively increased. Furthermore, when ventricular myocytes obtained from ERαKO and ERßKO were cultured in the presence of E2, results showed that E2 reduced Ito density only in ERßKO myocytes confirming the repressive role of E2-ERα in regulating Ito. CONCLUSION: Altogether, these results suggest that E2 negatively regulates the density of Ito through ERα, this highlights a potential role for this female hormone and its α-subtype receptor in modulating cardiac electrical activity.


Asunto(s)
Receptor alfa de Estrógeno/genética , Estrógenos/metabolismo , Ventrículos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Canales de Potasio Shal/biosíntesis , Potenciales de Acción , Animales , Calcio/metabolismo , Receptor beta de Estrógeno/genética , Estrógenos/genética , Femenino , Ventrículos Cardíacos/patología , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , Ovariectomía , Técnicas de Placa-Clamp , ARN Mensajero/biosíntesis , Canales de Potasio Shal/genética
9.
J Biol Chem ; 289(32): 21896-908, 2014 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-24936064

RESUMEN

Inflammation is now widely recognized as a key component of heart disease. Patients suffering from arrhythmias and heart failure have increased levels of tumor necrosis factor-α (TNFα) and interleukin-1ß (IL-1ß). Evidence suggests that these cytokines are important mediators of cardiac remodeling; however, their effects on ion channels and arrhythmogenesis remain incompletely understood. The L-type Ca(2+) current (ICaL) is a major determinant of the plateau phase of cardiac action potential and has a critical excitation-contraction coupling role. Thus, altering its properties could have detrimental effects on cardiac electrical and contractile functions. Accordingly, the objective of this study was to elucidate the effect of TNFα and IL-1ß on ICaL, while exploring the underlying regulatory mechanisms. Neonatal mouse ventricular myocytes were treated with a pathophysiological concentration (30 pg/ml) of TNFα and IL-1ß for 24 h. Voltage-clamp recordings showed that TNFα had no effect on ICaL, whereas IL-1ß decreased the current density by 36%. Although both IL-1ß- and TNFα-treated myocytes showed significant increase in reactive oxidative species (ROS), Western blot experiments revealed that only IL-1ß increased PKCϵ membrane translocation. The antioxidant N-acetyl-L-cysteine normalized ROS levels and restored ICaL density. Furthermore, the PKCϵ translocation inhibitor ϵ-V1-2 blocked the effect of IL-1ß on ICaL. The reduction of ICaL by IL-1ß was also seen in cultured adult ventricular myocytes. Overall, chronic IL-1ß treatment decreased ICaL density in cardiomyocytes. These effects implicated ROS signaling and PKCϵ activation. These findings could contribute to explain the role of IL-1ß in the development of arrhythmia and heart failure.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Interleucina-1beta/metabolismo , Miocardio/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Acetilcisteína/farmacología , Animales , Animales Recién Nacidos , Antioxidantes/farmacología , Canales de Calcio Tipo L/genética , Células Cultivadas , Activación Enzimática , Acoplamiento Excitación-Contracción , Ratones , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Técnicas de Placa-Clamp , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Cell Physiol ; 230(1): 170-9, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24915827

RESUMEN

Upregulation of the intermediate filament protein nestin was identified in a subpopulation of fibroblasts during reactive and reparative fibrosis and directly contributed to the enhanced proliferative phenotype. The present study tested the hypothesis that nestin was expressed in lung fibroblasts and the pattern of expression represented a distinct marker of pulmonary remodeling secondary to myocardial infarction and type I diabetes. Nestin((+)) fibroblasts were detected in rat lungs and a subpopulation exhibited a myofibroblast phenotype delineated by the co-expression of smooth muscle α-actin. In the lungs of myocardial infarcted rats, interstitial collagen content and nestin mRNA/protein levels were significantly increased despite the absence of secondary pulmonary hypertension, whereas smooth muscle α-actin protein expression was unchanged. Exposure of rat pulmonary fibroblasts to pro-fibrotic stimuli angiotensin II and transforming growth factor-ß significantly increased nestin protein levels. In the lungs of type I diabetic rats, the absence of a reactive fibrotic response was associated with a significant downregulation of nestin mRNA/protein expression. Nestin was reported a target of miR-125b, albeit miR-125b levels were unchanged in pulmonary fibroblasts treated with pro-fibrotic stimuli. Nestin((+)) cells lacking smooth muscle α-actin/collagen staining were also identified in rodent lungs and a transgenic approach revealed that expression of the intermediate filament protein was driven by intron 2 of the nestin gene. The disparate regulation of nestin characterized a distinct pattern of pulmonary remodeling secondary to myocardial infarction and type I diabetes and upregulation of the intermediate filament protein in lung fibroblasts may have facilitated in part the reactive fibrotic response.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Diabetes Mellitus Tipo 1/patología , Pulmón/patología , Infarto del Miocardio/patología , Nestina/biosíntesis , Actinas/biosíntesis , Angiotensina II/farmacología , Animales , Biomarcadores , Diferenciación Celular , Colágeno Tipo I/biosíntesis , Fibroblastos/metabolismo , Insuficiencia Cardíaca/patología , Humanos , Hipertensión Pulmonar/patología , Hipertrofia Ventricular Derecha/patología , Pulmón/metabolismo , Masculino , MicroARNs/biosíntesis , MicroARNs/genética , Contracción Miocárdica/fisiología , Nestina/genética , Fibrosis Pulmonar/patología , ARN Mensajero/biosíntesis , Ratas , Ratas Sprague-Dawley , Estreptozocina , Factor de Crecimiento Transformador beta/farmacología
11.
Circulation ; 127(20): 2009-20, 2013 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-23609807

RESUMEN

BACKGROUND: Pregnancy is associated with a faster heart rate (HR), which is a risk factor for arrhythmias. However, the underlying mechanisms for this increased HR are poorly understood. Therefore, this study was performed to gain mechanistic insight into the pregnancy-induced increase in HR. METHODS AND RESULTS: Using surface ECG we observed that pregnant (P) mice have faster HR (531±14 beats per minute [bpm]) compared with nonpregnant (NP) mice (470±27 bpm; P<0.03). Results obtained with Langendorff-perfused hearts showed that this difference persisted in the absence of autonomic nervous innervation (NP, 327±16 bpm; P, 385±18 bpm; P<0.02). Spontaneous action potentials of sinoatrial node cells from pregnant mice exhibited higher automaticity (NP, 292±13 bpm; P, 330±12 bpm; P=0.047) and steeper diastolic depolarization (NP, 0.20±0.03 V/s; P, 0.40±0.06 V/s; P=0.004). Pregnancy increased the density of the hyperpolarization-activated current (If) (at -90mV: NP, -15.2±1.0 pA/pF; P, -28.6±2.9 pA/pF; P=0.0002) in sinoatrial node cells. Voltage dependence of the If activation curve and the intracellular cAMP levels were unchanged in sinoatrial node cells of pregnant mice. However, there was a significant increase in HCN2 channel protein expression with no change in HCN4 expression. Maximal depolarizing shift of the If activation curve induced by isoproterenol was attenuated in pregnancy. This reduced response to isoproterenol may be attributable to the lower cAMP sensitivity of HCN2 isoform compared with that of HCN4. CONCLUSIONS: This study shows that an increase in If current density contributes to the acceleration of sinoatrial node automaticity and explains, in part, the higher HR observed in pregnancy.


Asunto(s)
Sistema de Conducción Cardíaco/fisiología , Frecuencia Cardíaca/fisiología , Canales Iónicos/biosíntesis , Embarazo/fisiología , Nodo Sinoatrial/fisiología , Regulación hacia Arriba/fisiología , Animales , Canales Catiónicos Regulados por Nucleótidos Cíclicos/biosíntesis , Femenino , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Ratones , Canales de Potasio
12.
J Cardiovasc Electrophysiol ; 25(8): 896-904, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24678923

RESUMEN

INTRODUCTION: KV 4 together with KV Channel-Interacting Protein 2 (KChIP2) mediate the fast recovering transient outward potassium current (I(to,f)) in the heart. KChIP2 is downregulated in human heart failure (HF), potentially underlying the loss of I(to,f). We investigated remodeling associated with HF hypothesizing that KChIP2 plays a central role in the modulation of outward K(+) currents in HF. METHODS AND RESULTS: HF was induced by aortic banding in wild-type (WT) and KChIP2 deficient (KChIP2(-/-)) mice, evaluated by echocardiography. Action potentials were measured by floating microelectrodes in intact hearts. Ventricular cardiomyocytes were isolated and whole-cell currents were recorded by patch clamp. Left ventricular action potentials in KChIP2(-/-) mice were prolonged in a rate dependent manner, consistent with patch-clamp data showing loss of a fast recovering outward K(+) current and upregulation of the slow recovering I(to,s) and I(Kur). HF decreased all outward K(+) currents in WT mice and did not change the relative contribution of I(to,f) in WT mice. Compared to WT HF, KChIP2(-/-) HF had a larger reduction of K(+) -current density. However, the relative APD prolongation caused by HF was shorter for KChIP2(-/-) compared with WT, and the APs of the 2 HF mouse types were indistinguishable. CONCLUSION: I(to,f) is just one of many K(+) currents being downregulated in murine HF. The downregulation of repolarizing currents in HF is accentuated in KChIP2(-/-) mice. However, the prolongation of APs associated with HF is less in KChIP2(-/-) compared to WT, suggesting other compensatory mechanism(s) in the KChIP2(-/-) mouse.


Asunto(s)
Sistema de Conducción Cardíaco/metabolismo , Insuficiencia Cardíaca/metabolismo , Proteínas de Interacción con los Canales Kv/deficiencia , Miocitos Cardíacos/metabolismo , Potasio/metabolismo , Potenciales de Acción , Animales , Estimulación Cardíaca Artificial , Modelos Animales de Enfermedad , Regulación hacia Abajo , Genotipo , Sistema de Conducción Cardíaco/fisiopatología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Proteínas de Interacción con los Canales Kv/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores de Tiempo
13.
Cardiovasc Res ; 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-39259837

RESUMEN

AIMS: Pregnant women have a significantly elevated resting heart rate (HR), which makes cardiac arrhythmias more likely to occur. Although electrical remodeling of the sinoatrial node (SAN) has been documented, the underlying mechanism is not fully understood. The acetylcholine-activated potassium current (IKACh), one of the major repolarizing currents in the SAN, plays a critical role in HR control by hyperpolarizing the maximal diastolic potential (MDP) of the SAN action potential (AP), thereby reducing SAN automaticity and HR. Thus, considering its essential role in cardiac automaticity, this study aims to determine whether changes in IKACh are potentially involved in the increased HR associated with pregnancy. METHODS AND RESULTS: Experiments were conducted on non-pregnant (NP, 2-3 months old) and pregnant (P, 17-18 gestation days) female CD-1 mice. IKACh was recorded on spontaneously beating SAN cells using the muscarinic agonist carbachol (CCh). Voltage-clamp data showed a reduction in IKACh density during pregnancy, which returned to control values shortly after delivery. The reduction in IKACh was explained by a decrease in protein expression of Kir3.1 channel subunit and the muscarinic type 2 receptor. In agreement with these findings, current-clamp data shows that the MDP of SAN cells from P mice were less hyperpolarized following CCh administration. Surface electrocardiograms (ECGs) recorded on anesthetized mice revealed that the cholinergic antagonist atropine and the selective KACh channel blocker tertiapin-Q increased HR in NP mice and had only a minimal effect on P mice. AP and ECG data also showed that pregnancy is associated with a decrease in beating and heart rate variability, respectively. CONCLUSION: IKACh function and expression are decreased in the mouse SAN during pregnancy, strongly suggesting that, in addition to other electrical remodeling of the SAN, reduced IKACh also plays an important role in the pregnancy-induced increased HR.

14.
J Am Heart Assoc ; 11(8): e023974, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35435021

RESUMEN

Background Elevated angiotensin II levels are thought to play an important role in atrial electrical and structural remodeling associated with atrial fibrillation. However, the mechanisms by which this remodeling occurs are still unclear. Accordingly, we explored the effects of angiotensin II on atrial remodeling using transgenic mice overexpressing angiotensin II type 1 receptor (AT1R) specifically in cardiomyocytes. Methods and Results Voltage-clamp techniques, surface ECG, programmed electrical stimulations along with quantitative polymerase chain reaction, Western blot, and Picrosirius red staining were used to compare the atrial phenotype of AT1R mice and their controls at 50 days and 6 months. Atrial cell capacitance and fibrosis were increased only in AT1R mice at 6 months, indicating the presence of structural remodeling. Ca2+ (ICaL) and K+ currents were not altered by AT1R overexpression (AT1R at 50 days). However, ICaL density and CaV1.2 messenger RNA expression were reduced by structural remodeling (AT1R at 6 months). Conversely, Na+ current (INa) was reduced (-65%) by AT1R overexpression (AT1R at 50 days) and the presence of structural remodeling (AT1R at 6 months) yields no further effect. The reduced INa density was not explained by lower NaV1.5 expression but was rather associated with an increase in sarcolemmal protein kinase C alpha expression in the atria, suggesting that chronic AT1R activation reduced INa through protein kinase C alpha activation. Furthermore, connexin 40 expression was reduced in AT1R mice at 50 days and 6 months. These changes were associated with delayed atrial conduction time, as evidenced by prolonged P-wave duration. Conclusions Chronic AT1R activation leads to slower atrial conduction caused by reduced INa density and connexin 40 expression.


Asunto(s)
Remodelación Atrial , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Atrios Cardíacos , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Proteína Quinasa C-alfa/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo
15.
Nat Commun ; 13(1): 6504, 2022 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-36323663

RESUMEN

Unlike artificial nanosystems, biological systems are ideally engineered to respond to their environment. As such, natural molecular buffers ensure precise and quantitative delivery of specific molecules through self-regulated mechanisms based on Le Chatelier's principle. Here, we apply this principle to design self-regulated nucleic acid molecular buffers for the chemotherapeutic drug doxorubicin and the antimalarial agent quinine. We show that these aptamer-based buffers can be programmed to maintain any specific desired concentration of free drug both in vitro and in vivo and enable the optimization of the chemical stability, partition coefficient, pharmacokinetics and biodistribution of the drug. These programmable buffers can be built from any polymer and should improve patient therapeutic outcome by enhancing drug activity and minimizing adverse effects and dosage frequency.


Asunto(s)
Doxorrubicina , Polímeros , Humanos , Distribución Tisular , Preparaciones Farmacéuticas , Sistemas de Liberación de Medicamentos , Tampones (Química)
16.
J Clin Invest ; 118(12): 3845-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19033655

RESUMEN

Mutations in contractile proteins in heart muscle can cause anatomical changes that result in cardiac arrhythmias and sudden cardiac death. However, a conundrum has existed because mutations in one such contractile protein, a so-called Ca2+ sensor troponin T (TnT), can promote ventricular rhythm disturbances even in the absence of hypertrophy or fibrosis. Thus, these mutations must enhance abnormal electrophysiological events via alternative means. In this issue of the JCI, Baudenbacher et al. report a novel mechanism to explain this puzzle (see the related article beginning on page 3893). They show that a selected TnT mutation in the adult mouse heart can markedly increase the sensitivity of cardiac muscle myofilaments to Ca2+ and enhance the susceptibility to arrhythmia, even in the absence of anatomical deformities. As these same mutations can cause some forms of arrhythmias in humans, these findings are of both basic and translational significance.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Calcio/metabolismo , Cardiomiopatía Hipertrófica/metabolismo , Cardiotónicos/efectos adversos , Quinolinas/efectos adversos , Taquicardia Ventricular/metabolismo , Tiadiazinas/efectos adversos , Citoesqueleto de Actina/patología , Potenciales de Acción/efectos de los fármacos , Animales , Cardiomiopatía Hipertrófica/inducido químicamente , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/patología , Cardiomiopatía Hipertrófica/fisiopatología , Cardiotónicos/farmacología , Gatos , Muerte Súbita Cardíaca , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Fibrosis/inducido químicamente , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Fibrosis/fisiopatología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Masculino , Ratones , Ratones Mutantes , Quinolinas/farmacología , Factores de Riesgo , Taquicardia Ventricular/inducido químicamente , Taquicardia Ventricular/genética , Taquicardia Ventricular/patología , Taquicardia Ventricular/fisiopatología , Tiadiazinas/farmacología , Troponina T/genética , Troponina T/metabolismo
17.
Am J Physiol Heart Circ Physiol ; 301(5): H2018-27, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21856918

RESUMEN

Transgenic mice that overexpress human type 1 angiotensin II receptor (AT(1)R) in the heart develop cardiac hypertrophy. Previously, we have shown that in 6-mo AT(1)R mice, which exhibit significant cardiac remodeling, fractional shortening is decreased. However, it is not clear whether altered contractility is attributable to AT(1)R overexpression or is secondary to cardiac hypertrophy/remodeling. Thus the present study characterized the effects of AT(1)R overexpression on ventricular L-type Ca(2+) currents (I(CaL)), cell shortening, and Ca(2+) handling in 50-day and 6-mo-old male AT(1)R mice. Echocardiography showed there was no evidence of cardiac hypertrophy in 50-day AT(1)R mice but that fractional shortening was decreased. Cellular experiments showed that cell shortening, I(CaL), and Ca(v)1.2 mRNA expression were significantly reduced in 50-day and 6-mo-old AT(1)R mice compared with controls. In addition, Ca(2+) transients and caffeine-induced Ca(2+) transients were reduced whereas the time to 90% Ca(2+) transient decay was prolonged in both age groups of AT(1)R mice. Western blot analysis revealed that sarcoplasmic reticulum Ca(2+)-ATPase and Na(+)/Ca(2+) exchanger protein expression was significantly decreased in 50-day and 6-mo AT(1)R mice. Overall, the data show that cardiac contractility and the mechanisms that underlie excitation-contraction coupling are altered in AT(1)R mice. Furthermore, since the alterations in contractility occur before the development of cardiac hypertrophy, it is likely that these changes are attributable to the increased activity of the renin-angiotensin system brought about by AT(1)R overexpression. Thus it is possible that AT(1)R blockade may help maintain cardiac contractility in individuals with heart disease.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Acoplamiento Excitación-Contracción , Contracción Miocárdica , Miocardio/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Función Ventricular Izquierda , Factores de Edad , Envejecimiento , Animales , Western Blotting , Cafeína/farmacología , Canales de Calcio Tipo L/efectos de los fármacos , Señalización del Calcio/efectos de los fármacos , Acoplamiento Excitación-Contracción/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Contracción Miocárdica/efectos de los fármacos , Miocardio/patología , Receptor de Angiotensina Tipo 1/genética , Retículo Sarcoplasmático/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Factores de Tiempo , Regulación hacia Arriba , Función Ventricular Izquierda/efectos de los fármacos
18.
Heart Rhythm O2 ; 2(2): 168-173, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34113919

RESUMEN

BACKGROUND: The heart rate increases by 10-20 beats per minute (bpm) throughout pregnancy in women, reaching maximum heart rate in the third trimester. During pregnancy, important changes in thyroid hormones also occur, with increases of up to 50% in the levels of triiodothyronine (T3), the biological active thyroid hormone. In addition, T3 has been shown to regulate cardiac electrophysiology. OBJECTIVE: Thus, in the present study the potential contribution of T3 in pregnancy-induced increased heart rate was explored. METHODS: We compared the heart rate between nonpregnant and pregnant mice under control conditions and after altering thyroid hormone levels with T3 and propylthiouracil (PTU, an antithyroid drug) treatments. RESULTS: Consistent with the clinical data, we found a 58% rise in T3 levels during pregnancy in mice. Although pregnant mice had a higher baseline heart rate (607 ± 8 bpm, P = .004) and higher T3 levels (1.9 ± 0.4 nM, P = .0005) than nonpregnant mice (heart rate: 546 ± 16 bpm; T3 levels: 1.2 ± 0.1 nM), their heart rate responded similarly to T3 treatment as nonpregnant mice (nonpregnant: Δ130 ± 22 bpm; pregnant: Δ126 ± 17 bpm, P = .858). Additionally, the heart rate remained significantly elevated (607 ± 11 bpm, P = .038) and comparable to untreated pregnant mice, after the use of the antithyroid drug PTU, although T3 levels (1.3 ± 0.2 nM, P = .559) returned to nonpregnant values. CONCLUSION: Based on these results, it is unlikely that T3 contributes significantly to the pregnancy-induced increased heart rate.

19.
J Am Heart Assoc ; 10(4): e017791, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33533257

RESUMEN

Background Mitogen-activated protein kinase-activated protein kinase-2 (MK2) is a protein serine/threonine kinase activated by p38α/ß. Herein, we examine the cardiac phenotype of pan MK2-null (MK2-/-) mice. Methods and Results Survival curves for male MK2+/+ and MK2-/- mice did not differ (Mantel-Cox test, P=0.580). At 12 weeks of age, MK2-/- mice exhibited normal systolic function along with signs of possible early diastolic dysfunction; however, aging was not associated with an abnormal reduction in diastolic function. Both R-R interval and P-R segment durations were prolonged in MK2-deficient mice. However, heart rates normalized when isolated hearts were perfused ex vivo in working mode. Ca2+ transients evoked by field stimulation or caffeine were similar in ventricular myocytes from MK2+/+ and MK2-/- mice. MK2-/- mice had lower body temperature and an age-dependent reduction in body weight. mRNA levels of key metabolic genes, including Ppargc1a, Acadm, Lipe, and Ucp3, were increased in hearts from MK2-/- mice. For equivalent respiration rates, mitochondria from MK2-/- hearts showed a significant decrease in Ca2+ sensitivity to mitochondrial permeability transition pore opening. Eight weeks of pressure overload increased left ventricular mass in MK2+/+ and MK2-/- mice; however, after 2 weeks the increase was significant in MK2+/+ but not MK2-/- mice. Finally, the pressure overload-induced decrease in systolic function was attenuated in MK2-/- mice 2 weeks, but not 8 weeks, after constriction of the transverse aorta. Conclusions Collectively, these results implicate MK2 in (1) autonomic regulation of heart rate, (2) cardiac mitochondrial function, and (3) the early stages of myocardial remodeling in response to chronic pressure overload.


Asunto(s)
Presión Sanguínea/fisiología , Bradicardia/fisiopatología , Cardiomiopatía Hipertrófica/fisiopatología , Frecuencia Cardíaca/fisiología , Mitocondrias Cardíacas/metabolismo , Función Ventricular Izquierda/fisiología , Remodelación Ventricular , Animales , Bradicardia/diagnóstico , Bradicardia/metabolismo , Cardiomiopatía Hipertrófica/diagnóstico , Cardiomiopatía Hipertrófica/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/deficiencia
20.
J Cardiovasc Electrophysiol ; 21(8): 916-22, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20132381

RESUMEN

INTRODUCTION: Cardiac arrhythmias have been reported in AIDS patients. Arrhythmias can arise from alterations in ventricular Na(+) channel function. However, it is unknown whether HIV affects cardiac Na(+) channel function. Therefore, the purpose of this study was to characterize the effect of HIV on ventricular Na(+) current (I(Na)) in a transgenic model of HIV (CD4C/HIV mice), which exhibit a severe AIDS-like disease. METHODS AND RESULTS: Patch-clamp techniques were used to examine I(Na) and action potentials (AP) in ventricular myocytes isolated from HIV and wild-type (WT) mice. In HIV myocytes peak I(Na) was reduced (at -50 mV: HIV, -55.3 +/- 4.3 pA/pF, n = 15; WT, -79.4 +/- 5.2 pA/pF, n = 16, P < 0.05), whereas late I(Na) was similar in both groups (HIV, -4.3 +/- 0.4 pA/pF; WT, -4.4 +/- 0.4 pA/pF, n = 22/group). AP amplitude (HIV 91.5 +/- 4.7 mV, n = 12; WT 104.4 +/- 3.1 mV, n = 15, P < 0.05) and the maximal velocity of the AP upstroke (V(max); HIV, 57.2 +/- 9.3 mV/ms, n = 12; WT, 113.5 +/- 8 mV/ms, n = 15, P < 0.05) were decreased in HIV myocytes. ECG recordings revealed that the QRS complex was prolonged in HIV mice (HIV, 15.7 +/- 0.2 ms, n = 22; WT, 14.1 +/- 0.5 ms, n = 10, P < 0.05). The serum levels of interleukin-1beta were elevated in HIV mice (HIV, 18.1 +/- 3.1 pg/mL, n = 3; WT, 5.1 +/- 1.1 pg/mL, n = 4, P < 0.05) in line with previous evidence that suggests that elevated levels of cytokines can affect cardiac ion currents. CONCLUSION: Overall, our observations suggest that elevated levels of proinflammatory cytokines in CD4C/HIV mice could alter Na(+) channel function, thus altering cardiac depolarization and contribute to the generation of arrhythmias.


Asunto(s)
Arritmias Cardíacas/etiología , Infecciones por VIH/complicaciones , VIH-1/genética , Miocitos Cardíacos/metabolismo , Canales de Sodio/metabolismo , Sodio/metabolismo , Potenciales de Acción , Animales , Arritmias Cardíacas/inmunología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/virología , Biomarcadores/sangre , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Infecciones por VIH/inmunología , Infecciones por VIH/metabolismo , Infecciones por VIH/virología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Mediadores de Inflamación/sangre , Interleucina-1beta/sangre , Ratones , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.5 , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Canales de Sodio/genética , Factores de Tiempo , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA