Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Europace ; 26(4)2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38546222

RESUMEN

AIMS: Right heart disease (RHD), characterized by right ventricular (RV) and atrial (RA) hypertrophy, and cardiomyocytes' (CM) dysfunctions have been described to be associated with the incidence of atrial fibrillation (AF). Right heart disease and AF have in common, an inflammatory status, but the mechanisms relating RHD, inflammation, and AF remain unclear. We hypothesized that right heart disease generates electrophysiological and morphological remodelling affecting the CM, leading to atrial inflammation and increased AF susceptibility. METHODS AND RESULTS: Pulmonary artery banding (PAB) was surgically performed (except for sham) on male Wistar rats (225-275 g) to provoke an RHD. Twenty-one days (D21) post-surgery, all rats underwent echocardiography and electrophysiological studies (EPS). Optical mapping was performed in situ, on Langendorff-perfused hearts. The contractility of freshly isolated CM was evaluated and recorded during 1 Hz pacing in vitro. Histological analyses were performed on formalin-fixed RA to assess myocardial fibrosis, connexin-43 levels, and CM morphology. Right atrial levels of selected genes and proteins were obtained by qPCR and Western blot, respectively. Pulmonary artery banding induced severe RHD identified by RV and RA hypertrophy. Pulmonary artery banding rats were significantly more susceptible to AF than sham. Compared to sham RA CM from PAB rats were significantly elongated and hypercontractile. Right atrial CM from PAB animals showed significant augmentation of mRNA and protein levels of pro-inflammatory interleukin (IL)-6 and IL1ß. Sarcoplasmic-endoplasmic reticulum Ca2+-ATPase-2a (SERCA2a) and junctophilin-2 were decreased in RA CM from PAB compared to sham rats. CONCLUSIONS: Right heart disease-induced arrhythmogenicity may occur due to dysfunctional SERCA2a and inflammatory signalling generated from injured RA CM, which leads to an increased risk of AF.


Asunto(s)
Fibrilación Atrial , Cardiopatías , Masculino , Ratas , Animales , Miocitos Cardíacos/metabolismo , Ratas Wistar , Atrios Cardíacos , Hipertrofia/metabolismo , Hipertrofia/patología , Inflamación/metabolismo
2.
Cardiovasc Res ; 120(5): 506-518, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38181429

RESUMEN

AIMS: Cellular senescence is a stress-related or aging response believed to contribute to many cardiac conditions; however, its role in atrial fibrillation (AF) is unknown. Age is the single most important determinant of the risk of AF. The present study was designed to (i) evaluate AF susceptibility and senescence marker expression in rat models of aging and myocardial infarction (MI), (ii) study the effect of reducing senescent-cell burden with senolytic therapy on the atrial substrate in MI rats, and (iii) assess senescence markers in human atrial tissue as a function of age and the presence of AF. METHODS AND RESULTS: AF susceptibility was studied with programmed electrical stimulation. Gene and protein expression was evaluated by immunoblot or immunofluorescence (protein) and digital polymerase chain reaction (PCR) or reverse transcriptase quantitative PCR (messenger RNA). A previously validated senolytic combination, dasatinib and quercetin, (D+Q; or corresponding vehicle) was administered from the time of sham or MI surgery through 28 days later. Experiments were performed blinded to treatment assignment. Burst pacing-induced AF was seen in 100% of aged (18-month old) rats, 87.5% of young MI rats, and 10% of young control (3-month old) rats (P ≤ 0.001 vs. each). Conduction velocity was slower in aged [both left atrium (LA) and right atrium (RA)] and young MI (LA) rats vs. young control rats (P ≤ 0.001 vs. each). Atrial fibrosis was greater in aged (LA and RA) and young MI (LA) vs. young control rats (P < 0.05 for each). Senolytic therapy reduced AF inducibility in MI rats (from 8/9 rats, 89% in MI vehicle, to 0/9 rats, 0% in MI D + Q, P < 0.001) and attenuated LA fibrosis. Double staining suggested that D + Q acts by clearing senescent myofibroblasts and endothelial cells. In human atria, senescence markers were upregulated in older (≥70 years) and long-standing AF patients vs. individuals ≤60 and sinus rhythm controls, respectively. CONCLUSION: Our results point to a potentially significant role of cellular senescence in AF pathophysiology. Modulating cell senescence might provide a basis for novel therapeutic approaches to AF.


Asunto(s)
Fibrilación Atrial , Remodelación Atrial , Senescencia Celular , Modelos Animales de Enfermedad , Fibrosis , Atrios Cardíacos , Infarto del Miocardio , Animales , Fibrilación Atrial/fisiopatología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Fibrilación Atrial/genética , Humanos , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Atrios Cardíacos/patología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/genética , Masculino , Quercetina/farmacología , Senoterapéuticos/farmacología , Factores de Edad , Femenino , Anciano , Persona de Mediana Edad , Estimulación Cardíaca Artificial
3.
Cardiovasc Res ; 120(4): 345-359, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38091977

RESUMEN

AIMS: Recent studies suggest that bioactive mediators called resolvins promote an active resolution of inflammation. Inflammatory signalling is involved in the development of the substrate for atrial fibrillation (AF). The aim of this study is to evaluate the effects of resolvin-D1 on atrial arrhythmogenic remodelling resulting from left ventricular (LV) dysfunction induced by myocardial infarction (MI) in rats. METHODS AND RESULTS: MI was produced by left anterior descending coronary artery ligation. Intervention groups received daily intraperitoneal resolvin-D1, beginning before MI surgery (early-RvD1) or Day 7 post-MI (late-RvD1) and continued until Day 21 post-MI. AF vulnerability was evaluated by performing an electrophysiological study. Atrial conduction was analysed by using optical mapping. Fibrosis was quantified by Masson's trichrome staining and gene expression by quantitative polymerase chain reaction and RNA sequencing. Investigators were blinded to group identity. Early-RvD1 significantly reduced MI size (17 ± 6%, vs. 39 ± 6% in vehicle-MI) and preserved LV ejection fraction; these were unaffected by late-RvD1. Transoesophageal pacing induced atrial tachyarrhythmia in 2/18 (11%) sham-operated rats, vs. 18/18 (100%) MI-only rats, in 5/18 (28%, P < 0.001 vs. MI) early-RvD1 MI rats, and in 7/12 (58%, P < 0.01) late-RvD1 MI rats. Atrial conduction velocity significantly decreased post-MI, an effect suppressed by RvD1 treatment. Both early-RvD1 and late-RvD1 limited MI-induced atrial fibrosis and prevented MI-induced increases in the atrial expression of inflammation-related and fibrosis-related biomarkers and pathways. CONCLUSIONS: RvD1 suppressed MI-related atrial arrhythmogenic remodelling. Early-RvD1 had MI sparing and atrial remodelling suppressant effects, whereas late-RvD1 attenuated atrial remodelling and AF promotion without ventricular protection, revealing atrial-protective actions unrelated to ventricular function changes. These results point to inflammation resolution-promoting compounds as novel cardio-protective interventions with a particular interest in attenuating AF substrate development.


Asunto(s)
Fibrilación Atrial , Remodelación Atrial , Cardiomiopatías , Infarto del Miocardio , Disfunción Ventricular Izquierda , Ratas , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/prevención & control , Infarto del Miocardio/metabolismo , Inflamación/prevención & control , Inflamación/complicaciones , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/prevención & control , Fibrosis
4.
Nat Commun ; 12(1): 2551, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33953173

RESUMEN

Endogenous cardiac pacemaker function regulates the rate and rhythm of cardiac contraction. The mutation p.Lys23Glu in the cohesin protein Shugoshin-1 causes severe heart arrhythmias due to sinoatrial node dysfunction and a debilitating gastrointestinal motility disorder, collectively termed the Chronic Atrial and Intestinal Dysrhythmia Syndrome, linking Shugoshin-1 and pacemaker activity. Hyperpolarization-activated, cyclic nucleotide-gated cation channel 4 (HCN4) is the predominant pacemaker ion-channel in the adult heart and carries the majority of the "funny" current, which strongly contributes to diastolic depolarization in pacemaker cells. Here, we study the mechanism by which Shugoshin-1 affects cardiac pacing activity with two cell models: neonatal rat ventricular myocytes and Chronic Atrial and Intestinal Dysrhythmia Syndrome patient-specific human induced pluripotent stem cell derived cardiomyocytes. We find that Shugoshin-1 interacts directly with HCN4 to promote and stabilize cardiac pacing. This interaction enhances funny-current by optimizing HCN4 cell-surface expression and function. The clinical p.Lys23Glu mutation leads to an impairment in the interaction between Shugoshin-1 and HCN4, along with depressed funny-current and dysrhythmic activity in induced pluripotent stem cell derived cardiomyocytes derived from Chronic Atrial and Intestinal Dysrhythmia Syndrome patients. Our work reveals a critical non-canonical, cohesin-independent role for Shugoshin-1 in maintaining cardiac automaticity and identifies potential therapeutic avenues for cardiac pacemaking disorders, in particular Chronic Atrial and Intestinal Dysrhythmia Syndrome.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Animales , Arritmias Cardíacas , Proteínas de Ciclo Celular/genética , Línea Celular , Supervivencia Celular , Proteínas Cromosómicas no Histona/genética , Técnicas de Silenciamiento del Gen , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/genética , Células Madre Pluripotentes Inducidas/metabolismo , Transporte Iónico/fisiología , Proteínas Musculares/genética , Contracción Miocárdica , Miocitos Cardíacos/metabolismo , Canales de Potasio/genética , Ratas , Cohesinas
5.
Circ Res ; 128(5): 619-635, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33375812

RESUMEN

RATIONALE: The mechanisms underlying atrial fibrillation (AF), the most common clinical arrhythmia, are poorly understood. Nucleoplasmic Ca2+ regulates gene expression, but the nature and significance of nuclear Ca2+-changes in AF are largely unknown. OBJECTIVE: To elucidate mechanisms by which AF alters atrial-cardiomyocyte nuclear Ca2+ ([Ca2+]Nuc) and CaMKII (Ca2+/calmodulin-dependent protein kinase-II)-related signaling. METHODS AND RESULTS: Atrial cardiomyocytes were isolated from control and AF dogs (kept in AF by atrial tachypacing [600 bpm × 1 week]). [Ca2+]Nuc and cytosolic [Ca2+] ([Ca2+]Cyto) were recorded via confocal microscopy. Diastolic [Ca2+]Nuc was greater than [Ca2+]Cyto under control conditions, while resting [Ca2+]Nuc was similar to [Ca2+]Cyto; both diastolic and resting [Ca2+]Nuc increased with AF. IP3R (Inositol-trisphosphate receptor) stimulation produced larger [Ca2+]Nuc increases in AF versus control cardiomyocytes, and IP3R-blockade suppressed the AF-related [Ca2+]Nuc differences. AF upregulated nuclear protein expression of IP3R1 (IP3R-type 1) and of phosphorylated CaMKII (immunohistochemistry and immunoblot) while decreasing the nuclear/cytosolic expression ratio for HDAC4 (histone deacetylase type-4). Isolated atrial cardiomyocytes tachypaced at 3 Hz for 24 hours mimicked AF-type [Ca2+]Nuc changes and L-type calcium current decreases versus 1-Hz-paced cardiomyocytes; these changes were prevented by IP3R knockdown with short-interfering RNA directed against IP3R1. Nuclear/cytosolic HDAC4 expression ratio was decreased by 3-Hz pacing, while nuclear CaMKII phosphorylation was increased. Either CaMKII-inhibition (by autocamtide-2-related peptide) or IP3R-knockdown prevented the CaMKII-hyperphosphorylation and nuclear-to-cytosolic HDAC4 shift caused by 3-Hz pacing. In human atrial cardiomyocytes from AF patients, nuclear IP3R1-expression was significantly increased, with decreased nuclear/nonnuclear HDAC4 ratio. MicroRNA-26a was predicted to target ITPR1 (confirmed by luciferase assay) and was downregulated in AF atrial cardiomyocytes; microRNA-26a silencing reproduced AF-induced IP3R1 upregulation and nuclear diastolic Ca2+-loading. CONCLUSIONS: AF increases atrial-cardiomyocyte nucleoplasmic [Ca2+] by IP3R1-upregulation involving miR-26a, leading to enhanced IP3R1-CaMKII-HDAC4 signaling and L-type calcium current downregulation. Graphic Abstract: A graphic abstract is available for this article.


Asunto(s)
Fibrilación Atrial/metabolismo , Calcio/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Miocitos Cardíacos/metabolismo , Potenciales de Acción , Animales , Fibrilación Atrial/fisiopatología , Canales de Calcio Tipo L/metabolismo , Señalización del Calcio , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Perros , Histona Desacetilasas/metabolismo , Humanos , Receptores de Inositol 1,4,5-Trifosfato/genética , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/fisiología
6.
Cardiovasc Res ; 117(2): 462-471, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-31977017

RESUMEN

AIMS: No studies have assessed the specific contributions of atrial fibrillation (AF)-related atrial vs. associated ventricular arrhythmia to remodelling. This study assessed the roles of atrial arrhythmia vs. high ventricular rate in AF-associated remodelling. METHODS AND RESULTS: Four primary dog-groups (12/group) were subjected to 3-week pacing: 600-b.p.m. atrial tachypacing maintaining AF [AF w/o- atrioventricular block (AVB)]; atrial tachypacing with atrioventricular-node ablation (AF+AVB) and ventricular-demand pacing (80 b.p.m.); 160-b.p.m. ventricular-tachypacing (V160) reproducing the response rate during AF; and sinus rhythm with AVB/ventricular-pacing at 80-b.p.m. (control group). At terminal study, left-atrial (LA) effective refractory period (ERP) was reduced equally in both AF groups (w/o-AVB and AF+AVB). AF-inducibility was increased strongly in AF groups (w/o-AVB and AF+AVB) and modestly in V160. AF duration was significantly increased in AF w/o-AVB but not in AF+AVB or V160. Conduction velocity was decreased in AF w/o-AVB, to a greater extent than in AF+AVB and V160. Atrial fibrous-tissue content was increased in AF w/o-AVB, AF+AVB and V160, with collagen-gene up-regulation only in AF w/o-AVB. Connexin43 gene expression was reduced only in AF w/o-AVB. An additional group of 240-b.p.m. ventricular tachypacing dogs (VTP240; to induce heart failure) was studied: vs. other tachypaced groups, VTP240 caused greater fibrosis, but no change in LA-ERP or AF-inducibility. VTP240 also increased AF duration, strongly decreased left ventricular ejection fraction, and was the only group with LA natriuretic-peptide activation. CONCLUSION: The atrial tachyarrhythmia and rapid ventricular response during AF produce distinct atrial remodelling; both contribute to the arrhythmogenic substrate, providing new insights into AF-related remodelling and novel considerations for ventricular rate-control.


Asunto(s)
Fibrilación Atrial/fisiopatología , Función del Atrio Izquierdo , Remodelación Atrial , Insuficiencia Cardíaca/fisiopatología , Frecuencia Cardíaca , Función Ventricular Izquierda , Potenciales de Acción , Animales , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/etiología , Fibrilación Atrial/metabolismo , Estimulación Cardíaca Artificial , Colágeno/genética , Colágeno/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perros , Fibrosis , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Factores de Tiempo
7.
Cardiovasc Res ; 117(7): 1776-1789, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32866246

RESUMEN

AIMS: Inflammation plays a role in atrial fibrillation (AF), but classical anti-inflammatory molecules are ineffective. Recent evidence suggests that failure of inflammation-resolution causes persistent inflammatory signalling and that a novel drug-family called resolvins promotes inflammation-resolution. Right heart disease (RHD) is associated with AF; experimental RHD shows signs of atrial inflammatory-pathway activation. Here, we evaluated resolvin-therapy effects on atrial arrhythmogenic remodelling in experimental RHD. METHODS AND RESULTS: Pulmonary hypertension and RHD were induced in rats with an intraperitoneal injection of 60 mg/kg monocrotaline (MCT). An intervention group received daily resolvin-D1 (RvD1), starting 1 day before MCT administration. Right atrial (RA) conduction and gene-expression were analysed respectively by optical mapping and qPCR/gene-microarray. RvD1 had no or minimal effects on MCT-induced pulmonary artery or right ventricular remodelling. Nevertheless, in vivo transoesophageal pacing induced atrial tachyarrhythmias in no CTRL rats vs. 100% MCT-only rats, and only 33% RvD1-treated MCT rats (P < 0.001 vs. MCT-only). Conduction velocity was significantly decreased by MCT, an effect prevented by RvD1. RHD caused RA dilation and fibrosis. RvD1 strongly attenuated RA fibrosis but had no effect on RA dilation. MCT increased RA expression of inflammation- and fibrosis-related gene-expression pathways on gene-microarray transcriptomic analysis, effects significantly attenuated by RvD1 (334 pathways enriched in MCT-rats vs. control; only 177 dysregulated by MCT with RvD1 treatment). MCT significantly increased RA content of type 1 (proinflammatory) CD68-positive M1 macrophages without affecting type 2 (anti-inflammatory) M2 macrophages. RvD1-treated MCT-rat RA showed significant reductions in proinflammatory M1 macrophages and increases in anti-inflammatory M2 macrophages vs. MCT-only. MCT caused statistically significant increases in protein-expression (western blot) of COL3A1, ASC, CASP1, CASP8, IL1ß, TGFß3, CXCL1, and CXCL2, and decreases in MMP2, vs. control. RvD1-treatment suppressed all these MCT-induced protein-expression changes. CONCLUSION: The inflammation-resolution enhancing molecule RvD1 prevents AF-promoting RA remodelling, while suppressing inflammatory changes and fibrotic/electrical remodelling, in RHD. Resolvins show potential promise in combating atrial arrhythmogenic remodelling by suppressing ongoing inflammatory signalling.


Asunto(s)
Antiarrítmicos/farmacología , Antiinflamatorios/farmacología , Fibrilación Atrial/prevención & control , Ácidos Docosahexaenoicos/farmacología , Atrios Cardíacos/efectos de los fármacos , Hipertensión Pulmonar/prevención & control , Mediadores de Inflamación/metabolismo , Disfunción Ventricular Derecha/prevención & control , Potenciales de Acción/efectos de los fármacos , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Remodelación Atrial/efectos de los fármacos , Modelos Animales de Enfermedad , Fibrosis , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Fenotipo , Ratas Wistar , Transducción de Señal , Transcriptoma , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/fisiopatología
8.
JCI Insight ; 5(8)2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32255765

RESUMEN

Atrial fibrillation (AF) alters atrial cardiomyocyte (ACM) Ca2+ handling, promoting ectopic beat formation. We examined the effects of AF-associated remodeling on Ca2+-related action potential dynamics and consequences for AF susceptibility. AF was maintained electrically in dogs by right atrial (RA) tachypacing. ACMs isolated from AF dogs showed increased Ca2+ release refractoriness, spontaneous Ca2+ spark frequency, and cycle length (CL) threshold for Ca2+ and action potential duration (APD) alternans versus controls. AF increased the in situ CL threshold for Ca2+/APD alternans and spatial dispersion in Ca2+ release recovery kinetics, leading to spatially discordant alternans associated with reentrant rotor formation and susceptibility to AF induction/maintenance. The clinically available agent dantrolene reduced Ca2+ leak and CL threshold for Ca2+/APD alternans in ACMs and AF dog right atrium, while suppressing AF susceptibility; caffeine increased Ca2+ leak and CL threshold for Ca2+/APD alternans in control dog ACMs and RA tissues. In vivo, the atrial repolarization alternans CL threshold was increased in AF versus control, as was AF vulnerability. Intravenous dantrolene restored repolarization alternans threshold and reduced AF vulnerability. Immunoblots showed reduced expression of total and phosphorylated ryanodine receptors and calsequestrin in AF and unchanged phospholamban/SERCA expression. Thus, along with promoting spontaneous ectopy, AF-induced Ca2+ handling abnormalities favor AF by enhancing vulnerability to repolarization alternans, promoting initiation and maintenance of reentrant activity; dantrolene provides a lead molecule to target this mechanism.


Asunto(s)
Potenciales de Acción , Fibrilación Atrial/metabolismo , Señalización del Calcio , Calcio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Fibrilación Atrial/fisiopatología , Perros , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Masculino
9.
J Physiol ; 596(12): 2359-2379, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29574723

RESUMEN

KEY POINTS: Ex vivo proliferated c-Kit+ endogenous cardiac progenitor cells (eCPCs) obtained from mouse and human cardiac tissues have been reported to express a wide range of functional ion channels. In contrast to previous reports in cultured c-Kit+ eCPCs, we found that ion currents were minimal in freshly isolated cells. However, inclusion of free Ca2+ intracellularly revealed a prominent inwardly rectifying current identified as the intermediate conductance Ca2+ -activated K+ current (KCa3.1) Electrical function of both c-Kit+ eCPCs and bone marrow-derived mesenchymal stem cells is critically governed by KCa3.1 calcium-dependent potassium channels. Ca2+ -induced increases in KCa3.1 conductance are necessary to optimize membrane potential during Ca2+ entry. Membrane hyperpolarization due to KCa3.1 activation maintains the driving force for Ca2+ entry that activates stem cell proliferation. Cardiac disease downregulates KCa3.1 channels in resident cardiac progenitor cells. Alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine. ABSTRACT: Endogenous c-Kit+ cardiac progenitor cells (eCPCs) and bone marrow (BM)-derived mesenchymal stem cells (MSCs) are being developed for cardiac regenerative therapy, but a better understanding of their physiology is needed. Here, we addressed the unknown functional role of ion channels in freshly isolated eCPCs and expanded BM-MSCs using patch-clamp, microfluorometry and confocal microscopy. Isolated c-Kit+ eCPCs were purified from dog hearts by immunomagnetic selection. Ion currents were barely detectable in freshly isolated c-Kit+ eCPCs with buffering of intracellular calcium (Ca2+i ). Under conditions allowing free intracellular Ca2+ , freshly isolated c-Kit+ eCPCs and ex vivo proliferated BM-MSCs showed prominent voltage-independent conductances that were sensitive to intermediate-conductance K+ -channel (KCa3.1 current, IKCa3.1 ) blockers and corresponding gene (KCNN4)-expression knockdown. Depletion of Ca2+i induced membrane-potential (Vmem ) depolarization, while store-operated Ca2+ entry (SOCE) hyperpolarized Vmem in both cell types. The hyperpolarizing SOCE effect was substantially reduced by IKCa3.1 or SOCE blockade (TRAM-34, 2-APB), and IKCa3.1 blockade (TRAM-34) or KCNN4-knockdown decreased the Ca2+ entry resulting from SOCE. IKCa3.1 suppression reduced c-Kit+ eCPC and BM-MSC proliferation, while significantly altering the profile of cyclin expression. IKCa3.1 was reduced in c-Kit+ eCPCs isolated from dogs with congestive heart failure (CHF), along with corresponding KCNN4 mRNA. Under perforated-patch conditions to maintain physiological [Ca2+ ]i , c-Kit+ eCPCs from CHF dogs had less negative resting membrane potentials (-58 ± 7 mV) versus c-Kit+ eCPCs from control dogs (-73 ± 3 mV, P < 0.05), along with slower proliferation. Our study suggests that Ca2+ -induced increases in IKCa3.1 are necessary to optimize membrane potential during the Ca2+ entry that activates progenitor cell proliferation, and that alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine.


Asunto(s)
Proliferación Celular , Ventrículos Cardíacos/citología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre/citología , Animales , Calcio/metabolismo , Células Cultivadas , Perros , Femenino , Ventrículos Cardíacos/fisiopatología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Transporte Iónico , Masculino , Potenciales de la Membrana , Células Madre Mesenquimatosas/fisiología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Células Madre/fisiología
10.
J Am Heart Assoc ; 6(4)2017 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-28381466

RESUMEN

BACKGROUND: Cardiac fibroblasts play important functional and pathophysiological roles. Intracellular ("intracrine") angiotensin-II (Ang-II) signaling regulates intercellular communication, excitability, and gene expression in cardiomyocytes; however, the existence and role of intracrine Ang-II signaling in cardiac fibroblasts is unstudied. Here, we evaluated the localization of Ang-II receptors on atrial fibroblast nuclei and associated intracrine effects of potential functional significance. METHODS AND RESULTS: Immunoblots of subcellular protein-fractions from isolated canine atrial fibroblasts indicated the presence of nuclear Ang-II type 1 receptors (AT1Rs) and Ang-II type 2 receptors (AT2Rs). Fluorescein isothiocyanate-Ang-II binding displaceable by AT1R- and AT2R-blockers was present on isolated fibroblast nuclei. G-protein subunits, including Gαq/11, Gαi/3, and Gß, were observed in purified fibroblast nuclear fractions by immunoblotting and intact-fibroblast nuclei by confocal immunocytofluorescence microscopy. Nuclear AT1Rs and AT2Rs regulated de novo RNA synthesis ([α32P]UTP incorporation) via IP3R- and NO-dependent pathways, respectively. In intact cultured fibroblasts, intracellular Ang-II release by photolysis of a membrane-permeable caged Ang-II analog led to IP3R-dependent nucleoplasmic Ca2+-liberation, with IP3R3 being the predominant nuclear isoform. Intracellular Ang-II regulated fibroblast proliferation ([3H]thymidine incorporation), collagen-1A1 mRNA-expression, and collagen secretion. Intracellular Ang-II and nuclear AT1R protein levels were significantly increased in a heart failure model in which atrial fibrosis underlies atrial fibrillation. CONCLUSIONS: Fibroblast nuclei possess AT1R and AT2R binding sites that are coupled to intranuclear Ca2+-mobilization and NO liberation, respectively. Intracellular Ang-II signaling regulates fibroblast proliferation, collagen gene expression, and collagen secretion. Heart failure upregulates Ang-II intracrine signaling-components in atrial fibroblasts. These results show for the first time that nuclear angiotensin-II receptor activation and intracrine Ang-II signaling control fibroblast function and may have pathophysiological significance.


Asunto(s)
Angiotensina II/fisiología , Proliferación Celular , Colágeno/metabolismo , Fibroblastos/metabolismo , Atrios Cardíacos/citología , Insuficiencia Cardíaca/metabolismo , Transcripción Genética , Bloqueadores del Receptor Tipo 1 de Angiotensina II/farmacología , Bloqueadores del Receptor Tipo 2 de Angiotensina II/farmacología , Animales , Calcio/metabolismo , Núcleo Celular/metabolismo , Colágeno Tipo I/genética , Modelos Animales de Enfermedad , Perros , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Immunoblotting , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Microscopía Fluorescente , Óxido Nítrico/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Receptor de Angiotensina Tipo 2/metabolismo
11.
Heart Rhythm ; 14(5): 739-748, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28188930

RESUMEN

BACKGROUND: Complete atrioventricular block (CAVB) causes arrhythmogenic remodeling and increases the risk of torsades de pointes arrhythmias. MicroRNAs (miRNAs) are key regulators of gene expression that contribute to cardiac remodeling. OBJECTIVE: The purpose of this study was to assess miRNA changes after CAVB and identify novel candidates potentially involved in arrhythmogenic cardiac remodeling. METHODS: CAVB was induced in mice via His-bundle ablation. Expression of miRNAs was evaluated by pan-miRNA microarray with quantitative polymerase chain reaction (qPCR) confirmation, on samples obtained 24 hours and 4 weeks post-CAVB. MiRNA target prediction algorithms were used to identify potential target genes. Targets confirmed by luciferase assays in HEK293 cells were followed up with overexpression studies in neonatal rat ventricular myocytes to evaluate regulation using real time- quantitative polymerase chain reaction (RT-qPCR), western blots, cell shortening measurements, and fura-2 Ca2+ fluorescence imaging. RESULTS: Of >400 miRNAs assayed, only miRNA-135a (miR-135a) was altered at 24 hours, down-regulated 78% (P <.001). Algorithms predicted miR-135a regulation of the sodium-calcium exchanger type 1 (NCX1). miR-135a transfection suppressed NCX1 3'UTR reporter activity by 42% (P <.001), mRNA expression by 34% (P <.001), and protein levels by 45% (P <.001) vs noncoding miRNA control. miR-135a overexpression reduced spontaneous beating frequency of neonatal rat ventricular myocytes by 63% (P <.001) while slowing decay (by 56%, P <.05) of caffeine-induced Ca2+ transients. miR-135a also suppressed the Ca2+ loading effects of ouabain and ouabain-induced spontaneous Ca2+ release events. CONCLUSION: NCX1 is negatively regulated by miR-135a, a microRNA that is down-regulated in the heart after CAVB in mice. By controlling NCX1 expression, miR-135a modulates cardiomyocyte automaticity, Ca2+ extrusion, and arrhythmogenic Ca2+ loading/spontaneous Ca2+ release events. Therefore, miR-135a may contribute to proarrhythmic remodeling after CAVB.


Asunto(s)
Bloqueo Atrioventricular/genética , Bloqueo Atrioventricular/fisiopatología , Sistema de Conducción Cardíaco/fisiopatología , MicroARNs/genética , Intercambiador de Sodio-Calcio/genética , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo , Expresión Génica , Regulación de la Expresión Génica , Células HEK293 , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Humanos , Ratones , MicroARNs/fisiología , Miocitos Cardíacos/fisiología , Ratas , Intercambiador de Sodio-Calcio/biosíntesis , Intercambiador de Sodio-Calcio/metabolismo , Torsades de Pointes/fisiopatología
12.
J Am Coll Cardiol ; 66(1): 47-58, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26139058

RESUMEN

BACKGROUND: Atrial fibrillation (AF) is associated with metabolic stress, which activates adenosine monophosphate-regulated protein kinase (AMPK). OBJECTIVES: This study sought to examine AMPK response to AF and associated metabolic stress, along with consequences for atrial cardiomyocyte Ca(2+) handling. METHODS: Calcium ion (Ca(2+)) transients (CaTs) and cell shortening (CS) were measured in dog and human atrial cardiomyocytes. AMPK phosphorylation and AMPK association with Ca(2+)-handling proteins were evaluated by immunoblotting and immunoprecipitation. RESULTS: CaT amplitude and CS decreased at 4-min glycolysis inhibition (GI) but returned to baseline at 8 min, suggesting cellular adaptation to metabolic stress, potentially due to AMPK activation. GI increased AMPK-activating phosphorylation, and an AMPK inhibitor, compound C (CompC), abolished the adaptation of CaT and CS to GI. The AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR) increased CaT amplitude and CS, restoring CompC-induced CaT and CS decreases. CompC decreased L-type calcium channel current (ICa,L), along with ICa,L-triggered CaT amplitude and sarcoplasmic reticulum (SR) Ca(2+) content under voltage clamp conditions in dog cells and suppressed CaT and ICa,L in human cardiomyocytes. Small interfering ribonucleic acid-based AMPK knockdown decreased CaT amplitude in neonatal rat cardiomyocytes. L-type Ca(2+) channel α subunits coimmunoprecipitated with AMPKα. Atrial AMPK-activating phosphorylation was enhanced by 1 week of electrically maintained AF in dogs; fractional AMPK phosphorylation was increased in paroxysmal AF and reduced in longstanding persistent AF patients. CONCLUSIONS: AMPK is activated by metabolic stress and AF, and helps maintain the intactness of atrial ICa,L, Ca(2+) handling, and cell contractility. AMPK contributes to the atrial compensatory response to AF-related metabolic stress; AF-related metabolic responses may be an interesting new therapeutic target.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Fibrilación Atrial/enzimología , Calcio/metabolismo , Miocitos Cardíacos/enzimología , Estrés Fisiológico , Animales , Células Cultivadas , Perros , Activación Enzimática , Humanos , Ratas
13.
J Mol Cell Cardiol ; 77: 113-24, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25315712

RESUMEN

Heart failure (HF) causes left-atrial (LA) and left-ventricular (LV) remodeling, with particularly-prominent changes in LA that create a substrate for atrial fibrillation (AF). MicroRNAs (miRs) are potential regulators in cardiac remodeling. This study evaluated time-dependent miR expression-changes in LA and LV tissue, fibroblasts and cardiomyocytes in experimental HF. HF was induced in dogs by ventricular tachypacing (varying periods, up to 2weeks). Following screening-microarray, 15 miRs were selected for detailed real-time qPCR assay. Extracellular matrix mRNA-expression was assessed by qPCR. Tachypacing time-dependently reduced LV ejection-fraction, increased LV-volume and AF-duration, and caused tissue-fibrosis with LA changes greater than LV. Tissue miR-expression significantly changed in LA for 10 miRs; in LV for none. Cell-selective analysis showed significant time-dependent changes in LA-fibroblasts for 10/15 miRs, LV-fibroblasts 8/15, LA-cardiomyocytes in 6/15 and LV-cardiomyocytes 3/15. Cell-expression specificity did not predict cell-specificity of VTP-induced expression-changes, e.g. 4/6 cardiomyocyte-selective miRs changed almost exclusively in fibroblasts (miR-1, miR-208b, miR133a/b). Thirteen miRs directly implicated in fibrosis/extracellular-matrix regulation were prominently changed: 9/13 showed fibroblast-selective alterations and 5/13 LA-selective. Multiple miRs changed in relation to associated extracellular-matrix targets. Experimental HF causes tissue and cell-type selective, time-dependent changes in cardiac miR-expression. Expression-changes are greater in LA versus LV, and greater in fibroblasts than cardiomyocytes, even for most cardiomyocyte-enriched miRs. This study, the first to examine time, chamber and cell-type selective changes in an experimental model of HF, suggests that multiple miR-changes underlie the atrial-selective fibrotic response and emphasize the importance of considering cell-specificity of miR expression-changes in cardiac remodeling paradigms.


Asunto(s)
Arritmias Cardíacas/metabolismo , Insuficiencia Cardíaca/metabolismo , MicroARNs/metabolismo , Miocardio/metabolismo , Animales , Colágeno Tipo I/metabolismo , Perros , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Atrios Cardíacos/patología , Ventrículos Cardíacos/patología , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Especificidad de Órganos , Transcriptoma , Remodelación Ventricular
14.
J Clin Invest ; 123(5): 1939-51, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23543060

RESUMEN

Atrial fibrillation (AF) is a highly prevalent arrhythmia with pronounced morbidity and mortality. Inward-rectifier K+ current (IK1) is believed to be an important regulator of reentrant-spiral dynamics and a major component of AF-related electrical remodeling. MicroRNA-26 (miR-26) is predicted to target the gene encoding KIR2.1, KCNJ2. We found that miR-26 was downregulated in atrial samples from AF animals and patients and this downregulation was accompanied by upregulation of IK1/KIR2.1 protein. miR-26 overexpression suppressed expression of KCNJ2/KIR2.1. In contrast, miR-26 knockdown, inhibition, or binding-site mutation enhanced KCNJ2/KIR2.1 expression, establishing KCNJ2 as a miR-26 target. Knockdown of endogenous miR-26 promoted AF in mice, whereas adenovirus-mediated expression of miR-26 reduced AF vulnerability. Kcnj2-specific miR-masks eliminated miR-26-mediated reductions in Kcnj2, abolishing miR-26's protective effects, while coinjection of a Kcnj2-specific miR-mimic prevented miR-26 knockdown-associated AF in mice. Nuclear factor of activated T cells (NFAT), a known actor in AF-associated remodeling, was found to negatively regulate miR-26 transcription. Our results demonstrate that miR-26 controls the expression of KCNJ2 and suggest that this downregulation may promote AF.


Asunto(s)
Fibrilación Atrial/metabolismo , Regulación de la Expresión Génica , MicroARNs/metabolismo , Canales de Potasio de Rectificación Interna/metabolismo , Animales , Sitios de Unión , Células Cultivadas , Perros , Regulación hacia Abajo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Miocitos Cardíacos/citología , Factores de Transcripción NFATC/metabolismo , Potasio/química , Ratas , Transcripción Genética
16.
PLoS One ; 6(5): e20362, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21655246

RESUMEN

The human ether-à-go-go-1 (h-eag1) K(+) channel is expressed in a variety of cell lines derived from human malignant tumors and in clinical samples of several different cancers, but is otherwise absent in normal tissues. It was found to be necessary for cell cycle progression and tumorigenesis. Specific inhibition of h-eag1 expression leads to inhibition of tumor cell proliferation. We report here that h-eag1 expression is controlled by the p53-miR-34-E2F1 pathway through a negative feed-forward mechanism. We first established E2F1 as a transactivator of h-eag1 gene through characterizing its promoter region. We then revealed that miR-34, a known transcriptional target of p53, is an important negative regulator of h-eag1 through dual mechanisms by directly repressing h-eag1 at the post-transcriptional level and indirectly silencing h-eag1 at the transcriptional level via repressing E2F1. There is a strong inverse relationship between the expression levels of miR-34 and h-eag1 protein. H-eag1antisense antagonized the growth-stimulating effects and the upregulation of h-eag1 expression in SHSY5Y cells, induced by knockdown of miR-34, E2F1 overexpression, or inhibition of p53 activity. Therefore, p53 negatively regulates h-eag1 expression by a negative feed-forward mechanism through the p53-miR-34-E2F1 pathway. Inactivation of p53 activity, as is the case in many cancers, can thus cause oncogenic overexpression of h-eag1 by relieving the negative feed-forward regulation. These findings not only help us understand the molecular mechanisms for oncogenic overexpression of h-eag1 in tumorigenesis but also uncover the cell-cycle regulation through the p53-miR-34-E2F1-h-eag1 pathway. Moreover, these findings place h-eag1 in the p53-miR-34-E2F1-h-eag1 pathway with h-eag as a terminal effecter component and with miR-34 (and E2F1) as a linker between p53 and h-eag1. Our study therefore fills the gap between p53 pathway and its cellular function mediated by h-eag1.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go/metabolismo , Benzotiazoles/farmacología , Western Blotting , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Imidazoles/farmacología , MicroARNs/genética , MicroARNs/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Piperazinas/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tolueno/análogos & derivados , Tolueno/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
17.
EMBO J ; 30(3): 524-32, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21217645

RESUMEN

In cancers with wild-type (WT) p53 status, the function of p53 is inhibited through direct interaction with Mdm2 oncoprotein, a negative feedback loop to limit the function of p53. In response to cellular stress, p53 escapes the p53:Mdm2 negative feedback to accumulate rapidly to induce cell cycle arrest and apoptosis. We demonstrate herein that an microRNA miR-605 is a new component in the p53 gene network, being transcriptionally activated by p53 and post-transcriptionally repressing Mdm2. Activation of p53 upregulated miR-605 via interacting with the promoter region of the gene. Overexpression of miR-605 directly decreased Mdm2 expression at the post-transcriptional level but indirectly increased the transcriptional activity of p53 on miR-34a via downregulating Mdm2; knockdown of miR-605 did the opposite. Mdm2 inhibitor upregulated expression of both miR-34a and miR-605, which was mitigated by p53 inhibitor. miR-605 preferentially induced apoptosis in WT p53-expressing cells, an effect abolished by p53 inhibition. These results indicate that miR-605 acts to interrupt p53:Mdm2 interaction to create a positive feedback loop aiding rapid accumulation of p53 to facilitate its function in response to stress.


Asunto(s)
Apoptosis/fisiología , Retroalimentación Fisiológica/fisiología , Regulación de la Expresión Génica/fisiología , MicroARNs/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Estrés Fisiológico/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Análisis de Varianza , Apoptosis/genética , Western Blotting , Caspasa 3/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Ensayo de Cambio de Movilidad Electroforética , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica/genética , Humanos , Etiquetado Corte-Fin in Situ , Luciferasas , MicroARNs/genética , Plásmidos/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico/genética , Sales de Tetrazolio , Tiazoles
18.
Cell Physiol Biochem ; 25(6): 571-86, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20511702

RESUMEN

Excitability is a fundamental characteristic of cardiac cells, which is delicately determined by ion channel activities modulated by many factors. MicroRNA (miRNA) expression is dynamically regulated and altered miRNA expression can render expression deregulation of ion channel genes leading to channelopathies-arrhythmogenesis. Indeed, evidence has emerged indicating the crucial role of miRNAs in controlling cardiac excitability by regulating expression of ion channel genes at the post-transcriptional level. However, the very limited experimental data in the literature hinder our understanding of the role of miRNAs and the often one-to-one interaction between miRNA and ion-channel gene in the published studies also casts a doubt about fullness of our view. Unfortunately, currently available techniques do not permit thorough characterization of miRNA targeting; computational prediction programs remain the only source for rapid identification of a putative miRNA target in silico. We conducted a rationally designed bioinformatics analysis in conjunction with experimental approaches to identify the miRNAs from the currently available miRNA databases which have the potential to regulate human cardiac ion channel genes and to validate the analysis with several pathological settings associated with the deregulated miRNAs and ion channel genes in the heart. We established a matrix of miRNAs that are expressed in cardiac cells and have the potential to regulate the genes encoding cardiac ion channels and transporters. We were able to explain a particular ionic remodeling process in hypertrophy/heart failure, myocardial ischemia, or atrial fibrillation with the corresponding deregulated miRNAs under that pathological condition; the changes of miRNAs appear to have anti-correlation with the changes of many of the genes encoding cardiac ion channels under these situations. These results indicate that multiple miRNAs might be critically involved in the electrical/ionic remodeling processes of cardiac diseases through altering their expression in cardiac cells, which has not been uncovered by previous experimental studies.


Asunto(s)
Regulación de la Expresión Génica , Cardiopatías/genética , Canales Iónicos/genética , MicroARNs/genética , Miocardio/metabolismo , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/fisiopatología , Biología Computacional , Perros , Femenino , Perfilación de la Expresión Génica , Cardiopatías/fisiopatología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/fisiopatología , Ratas , Ratas Wistar
19.
Cell Physiol Biochem ; 23(4-6): 317-26, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19471099

RESUMEN

Cardiac hypertrophy is characterized by electrical remolding with increased risk of arrhythmogenesis. Enhanced abnormal automaticity of ventricular cells may contribute to hypertrophic arrhythmias. The pacemaker current I(f), carried by the hyperpolarization-activated channels encoded mainly by the HCN2 and HCN4 genes in the heart, plays an important role in rhythmogenesis. Their expressions reportedly increase in hypertrophic and failing hearts, contributing to arrhythmogenesis under these conditions. However, how their expressions are controlled remained unclear. We performed a study to characterize the regulatory elements and transcriptional control of HCN2 and HCN4 genes. We identified the transcription start sites by 5'RACE and core promoter regions of these genes using luciferase reporter assay, and revealed the ubiquitous Sp1 protein as a common transactivator of HCN2 and HCN4 genes. We further unraveled robust increases in HCN2/HCN4 transcripts and protein levels, using real-time RT-PCR and Western blot analyses, in a rat model of left ventricular hypertrophy and in angiotensin II-induced neonatal ventricular hypertrophy. The upregulation of HCN2 and HCN4 transcription was accompanied by pronounced elevations of Sp1 and silencing of Sp1 by siRNA prevented overexpression of HCN2/HCN4 in hypertrophic cardiomyocytes. Our data indicate that Sp1 drives HCN2/HCN4 transcription and determines the functional level of HCN2/HCN4 mRNAs, and upregulation of Sp1 underlie the abnormal re-expression of HCN2/HCN4 genes in hypertrophied myocytes. This study also provides the first evidence for the role of Sp1 in the reactivation of 'fetal' cardiac genes, HCN2 and HCN4, in ventricular myocytes, and thereby in the pathological electrical remodeling in hypertrophied myocytes.


Asunto(s)
Canales Catiónicos Regulados por Nucleótidos Cíclicos/genética , Hipertrofia Ventricular Izquierda/genética , Inmunoglobulinas/metabolismo , Canales Iónicos/genética , Proteínas Musculares/genética , Miocitos Cardíacos/metabolismo , Canales de Potasio/genética , Angiotensina II/metabolismo , Angiotensina II/farmacología , Animales , Secuencia de Bases , Línea Celular , Canales Catiónicos Regulados por Nucleótidos Cíclicos/metabolismo , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Inmunoglobulinas/genética , Canales Iónicos/metabolismo , Datos de Secuencia Molecular , Proteínas Musculares/metabolismo , Canales de Potasio/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Transcripción Genética , Regulación hacia Arriba
20.
Nucleic Acids Res ; 37(3): e24, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19136465

RESUMEN

Anti-miRNA antisense inhibitors (AMOs) have demonstrated their utility in miRNA research and potential in miRNA therapy. Here we report a modified AMO approach in which multiple antisense units are engineered into a single unit that is able to simultaneously silence multiple-target miRNAs, the multiple-target AMO or MTg-AMO. We validated the technique with two separate MTg-AMOs: anti-miR-21/anti-miR-155/anti-miR-17-5p and anti-miR-1/anti-miR-133. We first verified the ability of the MTg-AMOs to antagonize the repressive actions of their target miRNAs using luciferase reporter activity assays and to specifically knock down the levels of their target miRNAs using real-time RT-PCR methods. We then used the MTg-AMO approach to identify several tumor suppressors-TGFBI, APC and BCL2L11 as the target genes for oncogenic miR-21, miR-155 and miR-17-5p, respectively, and two cardiac ion channel genes HCN2 (encoding a subunit of cardiac pacemaker channel) and CACNA1C (encoding the alpha-subunit of cardiac L-type Ca(2+) channel) for the muscle-specific miR-1 and miR-133. We further demonstrated that the MTg-AMO targeting miR-21, miR-155 and miR-17-5p produced a greater inhibitory effect on cancer cell growth, compared with the regular single-target AMOs. Moreover, while using the regular single-target AMOs excluded HCN2 as a target gene for either miR-1 or miR-133, the MTg-AMO approach is able to reveal HCN2 as the target for both miR-1 and miR-133. Our findings suggest the MTg-AMO as an improved approach for miRNA target finding and for studying function of miRNAs. This approach may find its broad application for exploring biological processes involving multiple miRNAs and multiple genes.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Oligodesoxirribonucleótidos Antisentido/química , Interferencia de ARN , Animales , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Genes Supresores de Tumor , Ingeniería Genética , Humanos , Canales Iónicos/genética , MicroARNs/química , Neoplasias/patología , Neoplasias/terapia , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA