Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 194
Filtrar
1.
Circ Res ; 135(3): 434-449, 2024 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-38864216

RESUMEN

BACKGROUND: Cardiac hypertrophy is an adaptive response to pressure overload aimed at maintaining cardiac function. However, prolonged hypertrophy significantly increases the risk of maladaptive cardiac remodeling and heart failure. Recent studies have implicated long noncoding RNAs in cardiac hypertrophy and cardiomyopathy, but their significance and mechanism(s) of action are not well understood. METHODS: We measured lincRNA-p21 RNA and H3K27ac levels in the hearts of dilated cardiomyopathy patients. We assessed the functional role of lincRNA-p21 in basal and surgical pressure-overload conditions using loss-of-function mice. Genome-wide transcriptome analysis revealed dysregulated genes and pathways. We labeled proteins in proximity to full-length lincRNA-p21 using a novel BioID2-based system. We immunoprecipitated lincRNA-p21-interacting proteins and performed cell fractionation, ChIP-seq (chromatin immunoprecipitation followed by sequencing), and co-immunoprecipitation to investigate molecular interactions and underlying mechanisms. We used GapmeR antisense oligonucleotides to evaluate the therapeutic potential of lincRNA-p21 inhibition in cardiac hypertrophy and associated heart failure. RESULTS: lincRNA-p21 was induced in mice and humans with cardiomyopathy. Global and cardiac-specific lincRNA-p21 knockout significantly suppressed pressure overload-induced ventricular wall thickening, stress marker elevation, and deterioration of cardiac function. Genome-wide transcriptome analysis and transcriptional network analysis revealed that lincRNA-p21 acts in trans to stimulate the NFAT/MEF2 (nuclear factor of activated T cells/myocyte enhancer factor-2) pathway. Mechanistically, lincRNA-p21 is bound to the scaffold protein KAP1 (KRAB-associated protein-1). lincRNA-p21 cardiac-specific knockout suppressed stress-induced nuclear accumulation of KAP1, and KAP1 knockdown attenuated cardiac hypertrophy and NFAT activation. KAP1 positively regulates pathological hypertrophy by physically interacting with NFATC4 to promote the overactive status of NFAT/MEF2 signaling. GapmeR antisense oligonucleotide depletion of lincRNA-p21 similarly inhibited cardiac hypertrophy and adverse remodeling, highlighting the therapeutic potential of inhibiting lincRNA-p21. CONCLUSIONS: These findings advance our understanding of the functional significance of stress-induced long noncoding RNA in cardiac hypertrophy and demonstrate the potential of lincRNA-p21 as a novel therapeutic target for cardiac hypertrophy and subsequent heart failure.


Asunto(s)
Cardiomegalia , Ratones Noqueados , ARN Largo no Codificante , Animales , Humanos , Masculino , Ratones , Cardiomegalia/metabolismo , Cardiomegalia/genética , Cardiomegalia/prevención & control , Cardiomegalia/patología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Remodelación Ventricular
3.
Circ Heart Fail ; 15(5): e008547, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35418250

RESUMEN

BACKGROUND: High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS: Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS: Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS: Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada , Doxorrubicina , Estrés Oxidativo , Sirtuina 3 , Acetilación/efectos de los fármacos , Animales , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Sirtuina 3/genética , Sirtuina 3/metabolismo
4.
Pharmacol Rep ; 74(1): 248-256, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34427908

RESUMEN

BACKGROUND: Doxorubicin is the chemotherapeutic drug of choice in osteosarcoma treatment, but its cumulative administration causes dilated cardiomyopathy. Combination therapy represents a potential strategy to reduce the therapeutic dosage of the chemotherapeutic agent and minimize its side effects. The aim of this study was to evaluate the potential of oridonin, a natural product from the medicinal herb Rabdosia rubescens, to act in combination with doxorubicin for osteosarcoma treatment. To date, there are no reports of the simultaneous administration of both drugs in osteosarcoma therapy. METHODS: The combined administration of different doses of oridonin and doxorubicin, as compared with the drugs alone, were tested in an in vitro model of osteosarcoma. The synergistic effect of the drugs on cell death was assessed by alamarBlue™ and by CompuSyn software. Early and late apoptosis markers (JC-1 fluorescence and Annexin V immunofluorescence), as well as the production of reactive oxygen species, were evaluated by flow cytometry. Western blot was used to assess the expression of anti-apoptotic proteins. RESULTS: Oridonin and doxorubicin presented a synergistic cytotoxic effect in osteosarcoma cells. In the presence of sub-cytotoxic concentrations of the natural product, there was an increased accumulation of intracellular doxorubicin, increased levels of reactive oxygen species (ROS), alteration of mitochondria membrane potential and a higher rate of apoptosis. CONCLUSION: The combined use of oridonin and doxorubicin could help to reduce the clinical dosage of doxorubicin and its dangerous side effects.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Diterpenos de Tipo Kaurano/farmacología , Doxorrubicina/farmacología , Isodon , Osteosarcoma , Transducción de Señal/efectos de los fármacos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/prevención & control , Cardiotónicos/farmacología , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/patología
5.
Mol Med Rep ; 24(6)2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34651661

RESUMEN

Cytochrome P450 family 2 subfamily E member 1 (CYP2E1) is a member of the cytochrome P450 enzyme family and catalyzes the metabolism of various substrates. CYP2E1 is upregulated in multiple heart diseases and causes damage mainly via the production of reactive oxygen species (ROS). In mice, increased CYP2E1 expression induces cardiac myocyte apoptosis, and knockdown of endogenous CYP2E1 can attenuate the pathological development of dilated cardiomyopathy (DCM). Nevertheless, targeted inhibition of CYP2E1 via the administration of drugs for the treatment of DCM remains elusive. Therefore, the present study aimed to investigate whether diallyl sulfide (DAS), a competitive inhibitor of CYP2E1, can be used to inhibit the development of the pathological process of DCM and identify its possible mechanism. Here, cTnTR141W transgenic mice, which developed typical DCM phenotypes, were used. Following treatment with DAS for 6 weeks, echocardiography, histological analysis and molecular marker detection were conducted to investigate the DAS­induced improvement on myocardial function and morphology. Biochemical analysis, western blotting and TUNEL assays were used to detected ROS production and myocyte apoptosis. It was found that DAS improved the typical DCM phenotypes, including chamber dilation, wall thinning, fibrosis, poor myofibril organization and decreased ventricular blood ejection, as determined using echocardiographic and histopathological analyses. Furthermore, the regulatory mechanisms, including inhibition both of the oxidative stress levels and the mitochondria­dependent apoptosis pathways, were involved in the effects of DAS. In particular, DAS showed advantages in terms of improved chamber dilation and dysfunction in model mice, and the improvement occurred in the early stage of the treatment compared with enalaprilat, an angiotensin­converting enzyme inhibitor that has been widely used in the clinical treatment of DCM and HF. The current results demonstrated that DAS could protect against DCM via inhibition of oxidative stress and apoptosis. These findings also suggest that inhibition of CYP2E1 may be a valuable therapeutic strategy to control the development of heart diseases, especially those associated with CYP2E1 upregulation. Moreover, the development of DAS analogues with lower cytotoxicity and metabolic rate for CYP2E1 may be beneficial.


Asunto(s)
Compuestos Alílicos/farmacología , Apoptosis/efectos de los fármacos , Cardiomiopatía Dilatada/prevención & control , Cardiotónicos/farmacología , Estrés Oxidativo/efectos de los fármacos , Sulfuros/farmacología , Compuestos Alílicos/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Cardiomiopatía Dilatada/patología , Cardiotónicos/uso terapéutico , Línea Celular , Citocromo P-450 CYP2E1/metabolismo , Modelos Animales de Enfermedad , Enalaprilato/farmacología , Enalaprilato/uso terapéutico , Femenino , Masculino , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Ratas , Sulfuros/uso terapéutico , Troponina T/metabolismo
6.
Int. j. cardiovasc. sci. (Impr.) ; 34(5): 588-592, Sept.-Oct. 2021. graf
Artículo en Inglés | LILACS | ID: biblio-1340056

RESUMEN

Abstract Heart failure (HF) is the most common cause of pulmonary hypertension (PH), and reduced exercise capacity and exertional dyspnea are the most frequent concerns in patients with PH-HF. Indeed, carbon dioxide end-tidal partial pressure (PETCO 2 ) during exercise is a well-established noninvasive marker of ventilation/perfusion ratio in PH. We aimed to evaluate the effect of aerobic exercise training on PETCO 2 response during exercise in a 59-year-old woman with PH secondary to idiopathic dilated cardiomyopathy. The patient with chronic fatigue and dyspnea at mild-to-moderate efforts was admitted to a cardiorespiratory rehabilitation program and had her cardiorespiratory response to exercise assessed during a cardiopulmonary exercise testing performed before and after three months of a thrice-weekly aerobic exercise training program. Improvements in aerobic capacity (23.9%) and endurance time (37.5%) and reduction in ventilatory inefficiency (-20.2%) was found after intervention. Post-intervention improvements in PETCO 2 at ventilatory anaerobic threshold (23.3%) and change in PETCO 2 kinetics pattern, with progressive increases from rest to peak of exercise, were also found. Patient also improved breathing pattern and timing of ventilation. This case report demonstrated for the first time that aerobic exercise training might be able to improve PETCO 2 response during exercise in a patient with PH-HF.


Asunto(s)
Humanos , Masculino , Femenino , Persona de Mediana Edad , Cardiomiopatía Dilatada/rehabilitación , Entrenamiento Aeróbico , Hipertensión Pulmonar/rehabilitación , Ventilación de Alta Frecuencia , Cardiomiopatía Dilatada/prevención & control , Intercambio Gaseoso Pulmonar , Prueba de Esfuerzo , Rehabilitación Cardiaca/métodos , Hipertensión Pulmonar/prevención & control
7.
Circ Res ; 128(10): 1514-1532, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33983834

RESUMEN

Our insight into the diverse and complex nature of dilated cardiomyopathy (DCM) genetic architecture continues to evolve rapidly. The foundations of DCM genetics rest on marked locus and allelic heterogeneity. While DCM exhibits a Mendelian, monogenic architecture in some families, preliminary data from our studies and others suggests that at least 20% to 30% of DCM may have an oligogenic basis, meaning that multiple rare variants from different, unlinked loci, determine the DCM phenotype. It is also likely that low-frequency and common genetic variation contribute to DCM complexity, but neither has been examined within a rare variant context. Other types of genetic variation are also likely relevant for DCM, along with gene-by-environment interaction, now established for alcohol- and chemotherapy-related DCM. Collectively, this suggests that the genetic architecture of DCM is broader in scope and more complex than previously understood. All of this elevates the impact of DCM genetics research, as greater insight into the causes of DCM can lead to interventions to mitigate or even prevent it and thus avoid the morbid and mortal scourge of human heart failure.


Asunto(s)
Alelos , Cardiomiopatía Dilatada/genética , Sitios Genéticos , Variación Genética , Cardiomiopatía Dilatada/prevención & control , Estudios de Cohortes , Conectina/química , Estudios Transversales , Interacción Gen-Ambiente , Humanos , Modelos Estadísticos , Fenotipo , Sarcómeros/química
8.
J Recept Signal Transduct Res ; 41(5): 494-503, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33092439

RESUMEN

Down-regulated lncRNA AC061961.2 in dilated cardiomyopathy (DCM) patients was previous reported. Whether AC061961.2 has regulatory effect on DCM still need exploration. Here, we tried to investigate the effect of AC061961.2 on DCM. After DCM model rat was established through injecting Adriamycin, left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD), left ventricular ejection fraction (LVEF), and left ventricular fractional shortening (LVFS) were measured by echocardiography. Histopathological changes and apoptosis were detected by hematoxylin-eosin, Masson staining, and TUNEL. After cardiomyocytes were isolated and identified by immunofluorescence, DCM cell model was established by injecting adriamycin. After transfected with overexpressed-AC061961.2 plasmids, cell apoptosis was detected by flow cytometry. The expressions of AC061961.2, ß-catenin, Axin2, c-Myc, CRP78, CHOP, Caspase-3, Bcl-2, and Bax in cardiomyocytes and heart tissues were detected by RT-qPCR or western blot. LVEDD and LVESD were increased while LVEF and LVFS were decreased in DCM rats. The histopathological of heart tissues showed a typical sign of DCM. Apoptosis were increased in heart tissues of DCM rats. In DCM rats, the expressions of AC061961.2, ß-catenin, Axin2, c-Myc, and Bcl-2 were decreased, the expressions of CRP78, CHOP, Caspase-3, and Bax were increased. After the overexpression of AC061961.2, levels of ß-catenin, Axin2, c-Myc, and Bcl-2 were increased, while levels of CRP78, CHOP, Caspase-3, and Bax were decreased, compared with that in DCM cardiomyocytes. LncRNA AC061961.2 overexpression inhibited endoplasmic reticulum stress induced apoptosis in DCM rats and cardiomyocytes via activating Wnt/ß-catenin pathway.


Asunto(s)
Apoptosis , Cardiomiopatía Dilatada/prevención & control , Estrés del Retículo Endoplásmico , Miocitos Cardíacos/patología , ARN Largo no Codificante/genética , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Proliferación Celular , Células Cultivadas , Regulación de la Expresión Génica , Masculino , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Wnt1/genética , beta Catenina/genética
9.
Int J Mol Sci ; 21(15)2020 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-32751757

RESUMEN

Nearly one in three people in the U.S. will develop heart failure (HF), characterized by fluid retention (edema) in the lungs and elsewhere. This leads to difficult breathing, deterioration of physical capacity, restriction of normal activities and death. There is little data about the safety and effects of sexual interactions in patients with HF. We tested whether a lack of sexual interactions affected pathophysiological outcomes in a pre-clinical mouse model of dilated cardiomyopathy that recapitulates the progressive stages of human HF. Male mice were randomly given access to, or deprived from, sexual interactions with female mice, which were confirmed by videography and generation of offspring. Cohousing with access to sexual interactions markedly prolonged survival, while cohousing without access to sexual activity did not. Sexual interactions improved systolic function, reduced HF-associated edema, altered transcription of heart contractile protein genes and decreased plasma testosterone levels. To determine whether testosterone levels contributed to survival, testosterone levels were experimentally reduced. Reduction of testosterone levels significantly prolonged survival. Taken together, in mice with dilated cardiomyopathy, sexual activity altered cardiac contractile gene transcription, improved systolic function, reduced edema and prolonged survival which may be in part due to lower testosterone levels.


Asunto(s)
Cardiomiopatía Dilatada/prevención & control , Coito/fisiología , Insuficiencia Cardíaca/prevención & control , Conducta Sexual/fisiología , Animales , Cardiomiopatía Dilatada/fisiopatología , Modelos Animales de Enfermedad , Femenino , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones , Contracción Miocárdica , Sobrevida/fisiología
10.
JCI Insight ; 5(15)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32603312

RESUMEN

The bromodomain and extraterminal (BET) family comprises epigenetic reader proteins that are important regulators of inflammatory and hypertrophic gene expression in the heart. We previously identified the activation of proinflammatory gene networks as a key early driver of dilated cardiomyopathy (DCM) in transgenic mice expressing a mutant form of phospholamban (PLNR9C) - a genetic cause of DCM in humans. We hypothesized that BETs coactivate this inflammatory process, representing a critical node in the progression of DCM. To test this hypothesis, we treated PLNR9C or age-matched WT mice longitudinally with the small molecule BET bromodomain inhibitor JQ1 or vehicle. BET inhibition abrogated adverse cardiac remodeling, reduced cardiac fibrosis, and prolonged survival in PLNR9C mice by inhibiting expression of proinflammatory gene networks at all stages of disease. Specifically, JQ1 had profound effects on proinflammatory gene network expression in cardiac fibroblasts, while having little effect on gene expression in cardiomyocytes. Cardiac fibroblast proliferation was also substantially reduced by JQ1. Mechanistically, we demonstrated that BRD4 serves as a direct and essential regulator of NF-κB-mediated proinflammatory gene expression in cardiac fibroblasts. Suppressing proinflammatory gene expression via BET bromodomain inhibition could be a novel therapeutic strategy for chronic DCM in humans.


Asunto(s)
Azepinas/farmacología , Proteínas de Unión al Calcio/fisiología , Cardiomiopatía Dilatada/prevención & control , Fibrosis/prevención & control , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Proteínas Nucleares/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Animales , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/patología , Fibrosis/etiología , Fibrosis/metabolismo , Fibrosis/patología , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
11.
Circ Res ; 127(5): 610-627, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32466726

RESUMEN

RATIONALE: Doxorubicin-induced cardiomyopathy (DiCM) is a primary cause of heart failure and mortality in cancer patients, in which macrophage-orchestrated inflammation serves as an essential pathological mechanism. However, the specific roles of tissue-resident and monocyte-derived macrophages in DiCM remain poorly understood. OBJECTIVE: Uncovering the origins, phenotypes, and functions of proliferative cardiac resident macrophages and mechanistic insights into the self-maintenance of cardiac macrophage during DiCM progression. METHODS AND RESULTS: Mice were administrated with doxorubicin to induce cardiomyopathy. Dynamic changes of resident and monocyte-derived macrophages were examined by lineage tracing, parabiosis, and bone marrow transplantation. We found that the monocyte-derived macrophages primarily exhibited a proinflammatory phenotype that dominated the whole DiCM pathological process and impaired cardiac function. In contrast, cardiac resident macrophages were vulnerable to doxorubicin insult. The survived resident macrophages exhibited enhanced proliferation and conferred a reparative role. Global or myeloid specifically ablation of SR-A1 (class A1 scavenger receptor) inhibited proliferation of cardiac resident reparative macrophages and, therefore, exacerbated cardiomyopathy in DiCM mice. Importantly, the detrimental effect of macrophage SR-A1 deficiency was confirmed by transplantation of bone marrow. At the mechanistic level, we show that c-Myc (Avian myelocytomatosis virus oncogene cellular homolog), a key transcriptional factor for the SR-A1-P38-SIRT1 (Sirtuin 1) pathway, mediated the effect of SR-A1 in reparative macrophage proliferation in DiCM. CONCLUSIONS: The SR-A1-c-Myc axis may represent a promising target to treat DiCM through augmentation of cardiac resident reparative macrophage proliferation.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Proliferación Celular , Autorrenovación de las Células , Macrófagos/enzimología , Miocardio/enzimología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores Depuradores de Clase A/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Células Cultivadas , Modelos Animales de Enfermedad , Doxorrubicina , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/patología , Fenotipo , Proteínas Proto-Oncogénicas c-myc/genética , Receptores Depuradores de Clase A/deficiencia , Receptores Depuradores de Clase A/genética , Transducción de Señal , Remodelación Ventricular
12.
Int J Mol Sci ; 21(9)2020 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-32344926

RESUMEN

AIMS: Coxsackievirus B3 (CVB3) is known to be an important cause of myocarditis and dilated cardiomyopathy. Enterovirus-2C (E2C) is a viral RNA helicase. It inhibits host protein synthesis. Based on these facts, we hypothesize that the inhibition of 2C may suppress virus replication and prevent enterovirus-mediated cardiomyopathy. METHODS AND RESULTS: We generated a chemically modified enterovirus-2C inhibitor (E2CI). From the in vitro assay, E2CI was showed strong antiviral effects. For in vivo testing, mice were treated with E2CI intraperitoneally injected daily for three consecutive days at a dose of 8mg/kg per day, after CVB3 post-infection (p.i) (CVB3 + E2CI, n = 33). For the infected controls (CVB3 only, n = 35), mice were injected with PBS (phosphate buffered saline) in a DBA/2 strain to establish chronic myocarditis. The four-week survival rate of E2CI-treated mice was significantly higher than that of controls (92% vs. 71%; p < 0.05). Virus titers and myocardial damage were significantly reduced in the E2CI treated group. In addition, echocardiography indicated that E2CI administration dramatically maintained mouse heart function compared to control at day 28 p.i chronic stage (LVIDD, 3.1 ± 0.08 vs. 3.9 ± 0.09, p < 0.01; LVDS, 2.0 ± 0.07 vs. 2.5 ± 0.07, p < 0.001; FS, 34.8 ± 1.6% vs. 28.5 ± 1.5%; EF, 67. 9 ± 2.9% vs. 54.7 ± 4.7%, p < 0.05; CVB3 + E2CI, n = 6 vs. CVB3, n = 4). Moreover, E2CI is effectively worked in human iPS (induced pluripotent stem cell) derived cardiomyocytes. CONCLUSION: Enterovirus-2C inhibitor (E2CI) was significantly reduced viral replication, chronic myocardium damage, and CVB3-induced mortality in DBA/2 mice. These results suggested that E2CI is a novel therapeutic agent for the treatment of enterovirus-mediated diseases.


Asunto(s)
Antivirales/farmacología , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/enzimología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocarditis/prevención & control , Miocitos Cardíacos/efectos de los fármacos , ARN Helicasas/antagonistas & inhibidores , Proteínas Virales/antagonistas & inhibidores , Animales , Antivirales/síntesis química , Antivirales/uso terapéutico , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/prevención & control , Enfermedad Crónica , Infecciones por Coxsackievirus/complicaciones , Enterovirus Humano B/efectos de los fármacos , Enterovirus Humano B/fisiología , Células HeLa , Humanos , Células Madre Pluripotentes Inducidas/virología , Luciferasas de Renilla/análisis , Masculino , Ratones , Ratones Endogámicos DBA , Miocarditis/etiología , Miocarditis/virología , Miocitos Cardíacos/patología , Miocitos Cardíacos/virología , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Oxazoles/farmacología , Oxazoles/uso terapéutico , Proteínas Recombinantes de Fusión/metabolismo , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/prevención & control , Replicación Viral/efectos de los fármacos
13.
Microb Pathog ; 144: 104169, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32205210

RESUMEN

Viral myocarditis (VMC) is a type of inflammation affecting myocardial cells caused by viral infection and has been an important cause of dilated cardiomyopathy (DCM) worldwide. Type B3 coxsackievirus (CVB3), a non-enveloped positive-strand RNA virus of the Enterovirus genus, is one of most common agent of viral myocarditis. Till now, effective treatments for VMC are lacking due to lack of drugs or vaccine. Lithium chloride (LiCl) is applied in the clinical management of manic depressive disorders. Accumulating evidence have demonstrated that LiCl, also as an effective antiviral drug, exhibited antiviral effects for specific viruses. However, there are few reports of evaluating LiCl's antiviral effect in mice model. Here, we investigated the inhibitory influence of LiCl on the CVB3 replication in vitro and in vivo and the development of CVB3-induced VMC. We found that LiCl significantly suppressed CVB3 replication in HeLa via inhibiting virus-induced cell apoptosis. Moreover, LiCl treatment in vivo obviously inhibited virus replication within the myocardium and alleviated CVB3-induced acute myocarditis. Collectively, our data demonstrated that LiCl inhibited CVB3 replication and negatively regulated virus-triggered inflammatory responses. Our finding further expands the antiviral targets of LiCl and provides an alternative agent for viral myocarditis.


Asunto(s)
Antivirales/farmacología , Cardiomiopatía Dilatada/tratamiento farmacológico , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B/efectos de los fármacos , Cloruro de Litio/farmacología , Miocarditis/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Cardiomiopatía Dilatada/prevención & control , Cardiomiopatía Dilatada/virología , Línea Celular , Infecciones por Coxsackievirus/prevención & control , Infecciones por Coxsackievirus/virología , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Células HEK293 , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Miocarditis/prevención & control , Miocarditis/virología , Miocardio/patología , Replicación Viral/efectos de los fármacos
14.
FASEB J ; 34(2): 2987-3005, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31908029

RESUMEN

The expression of α-cardiac actin, a major constituent of the cytoskeleton of cardiomyocytes, is dramatically decreased in a mouse model of dilated cardiomyopathy triggered by inducible cardiac-specific serum response factor (Srf) gene disruption that could mimic some forms of human dilated cardiomyopathy. To investigate the consequences of the maintenance of α-cardiac actin expression in this model, we developed a new transgenic mouse based on Cre/LoxP strategy, allowing together the induction of SRF loss and a compensatory expression of α-cardiac actin. Here, we report that maintenance of α-cardiac actin within cardiomyocytes temporally preserved cytoarchitecture from adverse cardiac remodeling through a positive impact on both structural and transcriptional levels. These protective effects were accompanied in vivo by the decrease of ROS generation and protein carbonylation and the downregulation of NADPH oxidases NOX2 and NOX4. We also show that ectopic expression of α-cardiac actin protects HEK293 cells against oxidative stress induced by H2 O2 . Oxidative stress plays an important role in the development of cardiac remodeling and contributes also to the pathogenesis of heart failure. Taken together, these findings indicate that α-cardiac actin could be involved in the regulation of oxidative stress that is a leading cause of adverse remodeling during dilated cardiomyopathy development.


Asunto(s)
Actinas/metabolismo , Cardiomiopatía Dilatada/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Actinas/genética , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Modelos Animales de Enfermedad , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/patología , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo
15.
Stem Cell Rev Rep ; 15(6): 827-841, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31612351

RESUMEN

Duchenne Muscular Dystrophy (DMD) is a progressive lethal disease caused by X-linked mutations of the dystrophin gene. Dystrophin deficiency clinically manifests as skeletal and cardiac muscle weakness, leading to muscle wasting and premature death due to cardiac and respiratory failure. Currently, no cure exists. Since heart disease is becoming a leading cause of death in DMD patients, there is an urgent need to develop new more effective therapeutic strategies for protection and improvement of cardiac function. We previously reported functional improvements correlating with dystrophin restoration following transplantation of Dystrophin Expressing Chimeric Cells (DEC) of myoblast origin in the mdx and mdx/scid mouse models. Here, we confirm positive effect of DEC of myoblast (MBwt/MBmdx) and mesenchymal stem cells (MBwt/MSCmdx) origin on protection of cardiac function after systemic DEC transplant. Therapeutic effect of DEC transplant (0.5 × 106) was assessed by echocardiography at 30 and 90 days after systemic-intraosseous injection to the mdx mice. At 90 days post-transplant, dystrophin expression in cardiac muscles of DEC injected mice significantly increased (15.73% ± 5.70 -MBwt/MBmdx and 5.22% ± 1.10 - MBwt/MSCmdx DEC) when compared to vehicle injected controls (2.01% ± 1.36) and, correlated with improved ejection fraction and fractional shortening on echocardiography. DEC lines of MB and MSC origin introduce a new promising approach based on the combined effects of normal myoblasts with dystrophin delivery capacities and MSC with immunomodulatory properties. Our study confirms feasibility and efficacy of DEC therapy on cardiac function and represents a novel therapeutic strategy for cardiac protection and muscle regeneration in DMD.


Asunto(s)
Cardiomiopatía Dilatada/prevención & control , Modelos Animales de Enfermedad , Distrofina/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Distrofia Muscular de Duchenne/complicaciones , Mioblastos/citología , Animales , Cardiomiopatía Dilatada/etiología , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Distrofina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Miocardio/citología
16.
Circulation ; 139(24): 2778-2792, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-30922078

RESUMEN

BACKGROUND: Enhancers are genomic regulatory elements conferring spatiotemporal and signal-dependent control of gene expression. Recent evidence suggests that enhancers can generate noncoding enhancer RNAs, but their (patho)biological functions remain largely elusive. METHODS: We performed chromatin immunoprecipitation-coupled sequencing of histone marks combined with RNA sequencing of left ventricular biopsies from experimental and genetic mouse models of human cardiac hypertrophy to identify transcripts revealing enhancer localization, conservation with the human genome, and hypoxia-inducible factor 1α dependence. The most promising candidate, hypoxia-inducible enhancer RNA ( HERNA)1, was further examined by investigating its capacity to modulate neighboring coding gene expression by binding to their gene promoters by using chromatin isolation by RNA purification and λN-BoxB tethering-based reporter assays. The role of HERNA1 and its neighboring genes for pathological stress-induced growth and contractile dysfunction, and the therapeutic potential of HERNA1 inhibition was studied in gapmer-mediated loss-of-function studies in vitro using human induced pluripotent stem cell-derived cardiomyocytes and various in vivo models of human pathological cardiac hypertrophy. RESULTS: HERNA1 is robustly induced on pathological stress. Production of HERNA1 is initiated by direct hypoxia-inducible factor 1α binding to a hypoxia-response element in the histoneH3-lysine27acetylation marks-enriched promoter of the enhancer and confers hypoxia responsiveness to nearby genes including synaptotagmin XVII, a member of the family of membrane-trafficking and Ca2+-sensing proteins and SMG1, encoding a phosphatidylinositol 3-kinase-related kinase. Consequently, a substrate of SMG1, ATP-dependent RNA helicase upframeshift 1, is hyperphoshorylated in a HERNA1- and SMG1-dependent manner. In vitro and in vivo inactivation of SMG1 and SYT17 revealed overlapping and distinct roles in modulating cardiac hypertrophy. Finally, in vivo administration of antisense oligonucleotides targeting HERNA1 protected mice from stress-induced pathological hypertrophy. The inhibition of HERNA1 postdisease development reversed left ventricular growth and dysfunction, resulting in increased overall survival. CONCLUSIONS: HERNA1 is a novel heart-specific noncoding RNA with key regulatory functions in modulating the growth, metabolic, and contractile gene program in disease, and reveals a molecular target amenable to therapeutic exploitation.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Cardiomiopatía Dilatada/prevención & control , Cardiomiopatía Hipertrófica/prevención & control , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos Cardíacos/metabolismo , Oligonucleótidos Antisentido/administración & dosificación , ARN no Traducido/metabolismo , Animales , Sitios de Unión , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Cardiomiopatía Hipertrófica/patología , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Regiones Promotoras Genéticas , ARN no Traducido/genética , Transducción de Señal , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo
19.
Nutr Metab Cardiovasc Dis ; 28(11): 1188-1195, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30143409

RESUMEN

BACKGROUND AND AIMS: Several studies propose that (-)-epicatechin, a flavonol present in high concentration in the cocoa, has cardioprotective effects. This study aimed to evaluate the impact of (-)-epicatechin on the development of dilated cardiomyopathy in a δ sarcoglycan null mouse model. METHODS AND RESULTS: δ Sarcoglycan null mice were treated for 15 days with (-)-epicatechin. Histological and morphometric analysis of the hearts treated mutant mice showed significant reduction of the vasoconstrictions in the coronary arteries as well as fewer areas with fibrosis and a reduction in the loss of the ventricular wall. On the contrary, it was observed a thickening of this region. By Western blot analysis, it was shown, and increment in the phosphorylation level of eNOS and PI3K/AKT/mTOR/p70S6K proteins in the heart of the (-)-epicatechin treated animals. On the other hand, we observed a significantly decreased level of the atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) heart failure markers. CONCLUSION: All the results indicate that (-)-epicatechin has the potential to prevent the development of dilated cardiomyopathy of genetic origin and encourages the use of this flavonol as a pharmacological therapy for dilated cardiomyopathy and heart failure diseases.


Asunto(s)
Cardiomiopatía Dilatada/prevención & control , Catequina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Sarcoglicanos/deficiencia , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Factor Natriurético Atrial/metabolismo , Cardiomiopatía Dilatada/enzimología , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/enzimología , Vasos Coronarios/fisiopatología , Modelos Animales de Enfermedad , Fibrosis , Masculino , Ratones Noqueados , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Péptido Natriurético Encefálico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sarcoglicanos/genética , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Vasoconstricción/efectos de los fármacos
20.
Basic Res Cardiol ; 113(3): 19, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29564567

RESUMEN

Previous studies have suggested the involvement of CD4 + T lymphocytes in cardiac remodelling. T-bet can direct Th1 lineage commitment. This study aimed to investigate the functional significance of T-bet in cardiac remodelling induced by pressure overload using T-bet global knockout rats. Increased T-bet levels were observed in rodent and human hypertrophied hearts. T-bet deficiency resulted in a less severe hypertrophic phenotype in rats. CD4 + T-lymphocyte reconstitution in T-bet-/- rats resulted in aggravated cardiac remodelling. T-cell homing molecule expression and cytokine secretion were altered in T-bet-deficient rat hearts. Administration of exogenous interferon-γ (IFN-γ) offset T-bet deficiency-mediated cardioprotection. Cardiomyocytes cultured in T-bet-/- CD4 + T-cell-conditioned media showed a reduced hypertrophic response after hypertrophic stimuli, which was abolished by an IFN-γ-neutralizing antibody. Taken together, our findings show that T-bet deficiency attenuates pressure overload-induced cardiac remodelling in rats. Specifically, targeting T-bet in T cells may be of great importance for the treatment of pathological cardiac remodelling and heart failure.


Asunto(s)
Cardiomegalia/metabolismo , Cardiomiopatía Dilatada/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/deficiencia , Células TH1/metabolismo , Remodelación Ventricular , Traslado Adoptivo , Animales , Cardiomegalia/inmunología , Cardiomegalia/fisiopatología , Cardiomegalia/prevención & control , Cardiomiopatía Dilatada/inmunología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Dilatada/prevención & control , Células Cultivadas , Quimiotaxis de Leucocito , Citocinas/inmunología , Citocinas/metabolismo , Técnicas de Silenciamiento del Gen , Genotipo , Humanos , Interferón gamma/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/inmunología , Comunicación Paracrina , Fenotipo , Ratas Sprague-Dawley , Ratas Transgénicas , Transducción de Señal , Proteínas de Dominio T Box/genética , Células TH1/efectos de los fármacos , Células TH1/inmunología , Células TH1/trasplante , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA