Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Transl Med ; 21(1): 290, 2023 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120549

RESUMEN

BACKGROUND: Oncostatin M (OSM) is a secreted cytokine of the interleukin (IL)-6 family that induces biological effects by activating functional receptor complexes of the common signal transducing component glycoprotein 130 (gp130) and OSM receptor ß (OSMR) or leukaemia inhibitory factor receptor (LIFR), which are mainly involved in chronic inflammatory and cardiovascular diseases. The effect and underlying mechanism of OSM/OSMR/LIFR on the development of cardiac hypertrophy remains unclear. METHODS AND RESULTS: OSMR-knockout (OSMR-KO) mice were subjected to aortic banding (AB) surgery to establish a model of pressure overload-induced cardiac hypertrophy. Echocardiographic, histological, biochemical and immunological analyses of the myocardium and the adoptive transfer of bone marrow-derived macrophages (BMDMs) were conducted for in vivo studies. BMDMs were isolated and stimulated with lipopolysaccharide (LPS) for the in vitro study. OSMR deficiency aggravated cardiac hypertrophy, fibrotic remodelling and cardiac dysfunction after AB surgery in mice. Mechanistically, the loss of OSMR activated OSM/LIFR/STAT3 signalling and promoted a proresolving macrophage phenotype that exacerbated inflammation and impaired cardiac repair during remodelling. In addition, adoptive transfer of OSMR-KO BMDMs to WT mice after AB surgery resulted in a consistent hypertrophic phenotype. Moreover, knockdown of LIFR in myocardial tissue with Ad-shLIFR ameliorated the effects of OSMR deletion on the phenotype and STAT3 activation. CONCLUSIONS: OSMR deficiency aggravated pressure overload-induced cardiac hypertrophy by modulating macrophages and OSM/LIFR/STAT3 signalling, which provided evidence that OSMR might be an attractive target for treating pathological cardiac hypertrophy and heart failure.


Asunto(s)
Interleucina-6 , Receptores OSM-LIF , Receptores de Oncostatina M , Transducción de Señal , Animales , Ratones , Cardiomegalia , Macrófagos , Oncostatina M/genética , Receptores OSM-LIF/genética , Receptores de Oncostatina M/genética
2.
Front Pharmacol ; 12: 581833, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34276351

RESUMEN

Background: The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may persist in patients with coronavirus disease 2019 (COVID-19) despite receiving standard care. Methods: In this pilot study of hospitalized adult patients (≥18 years of age), with radiologically confirmed pneumonia who were SARS-CoV-2 positive for more than 28 days despite standard care, were assigned to receive standard of care (SOC, grp I) or leflunomide + SOC (grp 2). After 2 weeks, grp 1 and grp 2 patients who continued to be SARS-CoV-2-positive received leflunomide for 14 days while continuing SOC. The primary outcomes were the rate of and time to SARS-CoV-2 clearance and the 14-day and 30-day hospital discharge rate. Results: 12 patients were enrolled in grp 1 and 15 patients were in grp 2. The 14 days SARS-CoV-2 viral clearance rate was 80.0% (12/15) for grp 2 patients receiving leflunomide vs. 16.7% for grp 1 patients (2/12) (p = 0.002). By day 14, the median time to SARS-CoV-2 clearance was 6.0 days (range 1-12, IQR 1-12) for grp 2 patients. In grp 1, two patients converted to viral negative on days 1 and 6 (p = 0.002). The 14-day discharge rate was 73.3% (11/15) for the grp 2 vs. 8.3% (1/12) for grp 1 (p = 0.001). The 30 days discharge rate was 100% (15/15) for the grp 2 vs. 66.7% (8/12) for grp 1. No severe adverse events or deaths were reported. Conclusion: Leflunomide may improve the SARS-CoV-2 clearance rate and discharge rate in patients with refractory COVID-19. The tolerability of the 14-28 days course of treatment with leflunomide is acceptable. These preliminary observations need to be verified by a large sample size and randomized controlled trial.

3.
Front Pharmacol ; 12: 648688, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34054527

RESUMEN

Background: Liquiritin (LIQ) is a traditional Chinese medicine that has been reported to regulate inflammation, oxidative stress and cell apoptosis. However, the beneficial effects of LIQ in lipopolysaccharides (LPS)-induced septic cardiomyopathy (SCM) has not been reported. The primary goal of this study was to investigate the effects of LIQ in LPS-induced SCM model. Methods: Mice were pre-treated with LIQ for 7 days before they were injected with LPS (10 mg/kg) for inducing SCM model. Echocardiographic analysis was used to evaluate cardiac function after 12 h of LPS injection. Thereafter, mice were sacrificed to collect hearts for molecular and histopathologic assays by RT-PCR, western-blots, immunohistochemical and terminal deoxynucleotidyl transferase nick-end labeling (TUNEL) staining analysis respectively. AMPKα2 knockout (AMPKα2-/-) mice were used to elucidate the mechanism of LIQ Neonatal rat cardiomyocytes (NRCMs) treated with or without LPS were used to further investigate the roles and mechanisms of LIQ in vitro experiments. Results: LIQ administration attenuated LPS-induced mouse cardiac dysfunction and reduced mortality, based upon the restoration of EF, FS, LVEDs, heart rate, dp/dt max and dp/dt min deteriorated by LPS treatment. LIQ treatment also reduced mRNA expression of TNFα, IL-6 and IL-1ß, inhibited inflammatory cell migration, suppressed cardiac oxidative stress and apoptosis, and improved metabolism. Mechanistically, LIQ enhanced the phosphorylation of AMP-activated protein kinase α2 (AMPKα2) and decreased the phosphorylation of mTORC1, IκBα and NFκB/p65. Importantly, the beneficial roles of LIQ were not observed in AMPKα2 knockout model, nor were they observed in vitro model after inhibiting AMPK activity with an AMPK inhibitor. Conclusion: We have demonstrated that LIQ exerts its protective effects in an SCM model induced by LPS administration. LIQ reduced inflammation, oxidative stress, apoptosis and metabolic alterations via regulating AMPKα2 dependent signaling pathway. Thus, LIQ might be a potential treatment or adjuvant for SCM treatment.

4.
Front Pharmacol ; 12: 610792, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33716740

RESUMEN

Cold-inducible RNA-binding protein (CIRP) is an intracellular stress-response protein that can respond to various stress conditions by changing its expression and regulating mRNA stability. As an RNA-binding protein, CIRP modulates gene expression at the post-transcriptional level, including those genes involved in DNA repair, cellular redox metabolism, circadian rhythms, telomere maintenance, and cell survival. CIRP is expressed in a large variety of tissues, including testis, brain, lung, kidney, liver, stomach, bone marrow, and heart. Recent studies have observed the important role of CIRP in cardiac physiology and diseases. CIRP regulates cardiac electrophysiological properties such as the repolarization of cardiomyocytes, the susceptibility of atrial fibrillation, and the function of the sinoatrial node in response to stress. CIRP has also been suggested to protect cardiomyocytes from apoptosis under various stress conditions, including heart failure, high glucose conditions, as well as during extended heart preservation under hypothermic conditions. This review summarizes the findings of CIRP investigations in cardiac physiology and diseases and the underlying molecular mechanism.

5.
Oxid Med Cell Longev ; 2020: 2647807, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908628

RESUMEN

AIMS: The aim of this study was to investigate whether resveratrol (RSV) could ameliorate ischemia- and hypoxia-associated cardiomyocyte apoptosis and injury via inhibiting senescence signaling and inflammasome activation. MATERIALS AND METHODS: Mice were treated with RSV by gastric tube (320 mg/kg/day) or vehicle one week before left coronary artery ligation or sham surgery until the end of the experiments. After pressure-volume loop analysis, mouse hearts were harvested for histopathological (including PSR, TTC, TUNEL staining, immunohistochemistry, and immunofluorescence) and molecular analysis by western blotting and RT-PCR. In addition, neonatal rat cardiomyocytes (NRCMs), cardiac fibroblasts (CFs), and macrophages were isolated for in vitro experiments. Key Findings. RSV treatment decreased mortality and improved cardiac hemodynamics. RSV inhibited the expression of senescence markers (p53, p16, and p19), inflammasome markers (NLRP3 and Cas1 p20), and nuclear translocation of NF-κB, hence alleviating infarction area, fibrosis, and cell apoptosis. RSV also inhibited expression of interleukin- (IL-) 1ß, IL-6, tumor necrosis factor-α, and IL-18 in vivo. In in vitro experiment, RSV prevented hypoxia-induced NRCM senescence and apoptosis. After inhibition of sirtuin 1 (Sirt1) by EX27, RSV failed to inhibit p53 acetylation and expression. Moreover, RSV could inhibit expression of NLRP3 and caspase 1 p20 in NRCMs, CFs, and macrophages, respectively, in in vitro experiments. Significance. Our findings revealed that RSV protected against ischemia-induced mouse heart injury in vivo and hypoxia-induced NRCM injury in vitro via regulating Sirt1/p53-mediated cell senescence and inhibiting NLRP3-mediated inflammasome activation.


Asunto(s)
Inflamasomas/metabolismo , Isquemia Miocárdica/complicaciones , Miocardio/patología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Resveratrol/farmacología , Transducción de Señal , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Hipoxia de la Célula/efectos de los fármacos , Pruebas de Función Cardíaca/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Isquemia Miocárdica/tratamiento farmacológico , Isquemia Miocárdica/patología , Isquemia Miocárdica/fisiopatología , Miocardio/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas Sprague-Dawley , Resveratrol/uso terapéutico , Factores de Riesgo , Transducción de Señal/efectos de los fármacos
6.
J Emerg Nurs ; 46(6): 791-801.e7, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32994038

RESUMEN

INTRODUCTION: The purpose of this study was to rapidly quantify the safety measures regarding donning and doffing personal protective equipment, complaints of discomfort caused by wearing personal protective equipment, and the psychological perceptions of health care workers in hospitals in Wuhan, China, responding to the outbreak. METHODS: A cross-sectional online questionnaire design was used Data were collected from March 14, 2020, to March 16, 2020, in Wuhan, China. Descriptive statistics and χ2 analyses testing were used. RESULTS: Standard nosocomial infection training could significantly decrease the occurrence of infection (3.6% vs 13.0%, χ2 = 4.47, P < 0.05). Discomfort can be classified into 7 categories. Female sex (66.0% vs 50.5%, χ2 = 6.37), occupation (62.7% vs 30.8%, χ2 = 5.33), working at designated hospitals (44.8% vs 26.7%, χ2 = 5.17) or in intensive care units (70.4% vs 57.9%, χ2 = 3.88), and working in personal protective equipment for > 4 hours (62.2% vs 39.2%, χ2 = 9.17) led to more complaints about physical discomfort or increased occurrence of pressure sores (all P < 0.05). Psychologically, health care workers at designated hospitals (60.0% vs 42.1%, χ2 = 4.97) or intensive care units (55.9% vs 41.5%, χ2 = 4.40) (all P < 0.05) expressed different rates of pride. DISCUSSION: Active training on infection and protective equipment could reduce the infection risk. Working for long hours increased the occurrence of discomfort and skin erosion. Reducing the working hours and having adequate protective products and proper psychological interventions may be beneficial to relieve discomfort.


Asunto(s)
COVID-19/prevención & control , Infección Hospitalaria/prevención & control , Personal de Salud/psicología , Transmisión de Enfermedad Infecciosa de Paciente a Profesional/prevención & control , Equipo de Protección Personal/efectos adversos , Neumonía Viral/prevención & control , Adulto , COVID-19/epidemiología , China/epidemiología , Estudios Transversales , Femenino , Humanos , Masculino , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/virología , SARS-CoV-2 , Encuestas y Cuestionarios
7.
J Mol Med (Berl) ; 98(5): 691-705, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32280997

RESUMEN

Cardiac hypertrophy is a complex pathological process, and the molecular mechanisms underlying hypertrophic remodeling have not been clearly elucidated. Leukocyte immunoglobulin-like receptor B4 (lilrb4) is an inhibitory transmembrane protein that is necessary for the regulation of various cellular signaling pathways. To investigate whether lilrb4 plays a role in cardiac hypertrophy, we performed aortic banding in lilrb4 knockout mice, lilrb4 cardiac-specific transgenic mice, and their wild-type littermates. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. We found that lilrb4 was expressed both in myocardial tissue and on cultured cardiomyocytes under basal conditions, but the expression was obviously decreased in mouse hearts following aortic banding and in cardiomyocytes treated with angiotensin II. Lilrb4 disruption aggravated cardiac hypertrophy, fibrosis, and dysfunction in response to pressure overload. Conversely, the cardiac overexpression of lilrb4 led to the opposite effects. Moreover, lilrb4 overexpression inhibited angiotensin II-induced cardiomyocyte hypertrophy in vitro. Mechanistically, we determined that the cardioprotective effect of lilrb4 was mediated through an interaction with SHP-2, the preservation of phosphorylated SHP-2, and the inhibition of the NF-κB pathway. In addition, SHP-2 knockdown in cardiomyocytes eliminated the inhibitory effects of lilrb4 on angiotensin II-induced hypertrophy and NF-κB activation. Our results suggest that lilrb4 protects against pathological cardiac hypertrophy via the SHP-2-dependent inhibition of the NF-κB pathway and may act as a potential therapeutic target for cardiac hypertrophy. KEY MESSAGES: Lilrb4 expression is decreased by hypertrophic stimuli. Lilrb4 protects against pathological cardiac hypertrophy. Lilrb4 interacts with SHP-2 and inhibits NF-κB pathway.


Asunto(s)
Cardiomegalia/etiología , Cardiomegalia/metabolismo , Susceptibilidad a Enfermedades , Glicoproteínas de Membrana/genética , FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores Inmunológicos/genética , Animales , Biomarcadores , Biopsia , Cardiomegalia/diagnóstico , Modelos Animales de Enfermedad , Ecocardiografía , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Hemodinámica , Inmunohistoquímica , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Receptores Inmunológicos/metabolismo
8.
PPAR Res ; 2019: 2601408, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31933619

RESUMEN

BACKGROUND: Oxidative stress, inflammation and cardiac apoptosis were closely involved in doxorubicin (DOX)-induced cardiac injury. Piperine has been reported to suppress inflammatory response and pyroptosis in macrophages. However, whether piperine could protect the mice against DOX-related cardiac injury remain unclear. This study aimed to investigate whether piperine inhibited DOX-related cardiac injury in mice. METHODS: To induce DOX-related acute cardiac injury, mice in DOX group were intraperitoneally injected with a single dose of DOX (15 mg/kg). To investigate the protective effects of piperine, mice were orally treated for 3 weeks with piperine (50 mg/kg, 18:00 every day) beginning two weeks before DOX injection. RESULTS: Piperine treatment significantly alleviated DOX-induced cardiac injury, and improved cardiac function. Piperine also reduced myocardial oxidative stress, inflammation and apoptosis in mice with DOX injection. Piperine also improved cell viability, and reduced oxidative damage and inflammatory factors in cardiomyocytes. We also found that piperine activated peroxisome proliferator-activated receptor-γ (PPAR-γ), and the protective effects of piperine were abolished by the treatment of the PPAR-γ antagonist in vivo and in vitro. CONCLUSIONS: Piperine could suppress DOX-related cardiac injury via activation of PPAR-γ in mice.

9.
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
11.
Mol Cell Biochem ; 430(1-2): 81-90, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28303407

RESUMEN

The aim of this study is to investigate the effect of evodiamine on fibroblast activation in cardiac fibroblasts and endothelial to mesenchymal transition (EndMT) in human umbilical vein endothelial cells (HUVECs). Neonatal rat cardiac fibroblasts were stimulated with transforming growth factor beta 1 (TGF-ß1) to induce fibroblast activation. After co-cultured with evodiamine (5, 10 µM), the proliferation and pro-fibrotic proteins expression of cardiac fibroblasts were evaluated. HUVECs were also stimulated with TGF-ß1 to induce EndMT and treated with evodiamine (5, 10 µM) at the same time. The EndMT response in the HUVECs was evaluated as well as the capacity of the transitioned endothelial cells migrating to surrounding tissue. As a result, Evodiamine-blunted TGF-ß1 induced activation of cardiac fibroblast into myofibroblast as assessed by the decreased expressions of α-SMA. Furthermore, evodiamine reduced the increased protein expression of fibrosis markers in neonatal and adult rat cardiac fibroblasts induced by TGF-ß1. HUVECs stimulated with TGF-ß1 exhibited lower expression levels of CD31, CD34, and higher levels of α-SMA, vimentin than the control cells. This phenotype was eliminated in the HUVECs treated with both 5 and 10 µM evodiamine. Evodiamine significantly reduced the increase in migration ability that occurred in response to TGF-ß1 in HUVECs. In addition, the activation of Smad2, Smad3, ERK1/2, and Akt, and the nuclear translocation of Smad4 in both cardiac fibroblasts and HUVEC were blocked by evodiamine treatment. Thus, evodiamine could prevent cardiac fibroblasts from activation into myofibroblast and protect HUVEC against EndMT. These effects may be mediated by inhibition of the TGFß pathway in both cardiac fibroblasts and HUVECs.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Alcaloides Indólicos/farmacología , Miofibroblastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Miofibroblastos/citología , Ratas , Ratas Sprague-Dawley
12.
Planta Med ; 83(9): 761-769, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28010025

RESUMEN

Evodiamine, a major component of Evodia rutaecarpa, can protect the myocardium against injury induced by atherosclerosis and ischemia-reperfusion. However, the effect of evodiamine against cardiac fibrosis remains unclear. This study aims to investigate the possible effect and mechanism involved in the function of evodiamine on isoproterenol-induced cardiac fibrosis and endothelial-to-mesenchymal transition. Isoproterenol was used to induce cardiac fibrosis in mice, and evodiamine was gavaged simultaneously. After 14 days, cardiac function was accessed by echocardiography. The extent of cardiac fibrosis and hypertrophy was evaluated by pathological and molecular analyses. The extent of endothelial-to-mesenchymal transition was evaluated by the expression levels of CD31, CD34, α-smooth muscle actin, and vimentin by immunofluorescence staining and Western blot analysis. After 14 days, the heart weight/body weight ratio and heart weight/tibia length ratio revealed no significant difference between the isoproterenol group and the isoproterenol/evodiamine-treated groups, whereas the increased heart weight was reduced in the isoproterenol/evodiamine-treated groups. Echocardiography revealed that interventricular septal thickness and left ventricular posterior wall thickness at the end diastole decreased in the evodiamine-treated groups. Evodiamine reduced isoproterenol-induced cardiac fibrosis as accessed by normalization in collagen deposition and gene expression of hypertrophic and fibrotic markers. Evodiamine also prevented endothelial-to-mesenchymal transition as evidenced by the increased expression levels of CD31 and CD34, decreased expression levels of α-smooth muscle actin and vimentin, and increased microvascular density in the isoproterenol/evodiamine-treated mice hearts. Furthermore, isoproterenol-induced activation of transforming growth factor-ß1/Smad signal was also blunted by evodiamine. Therefore, evodiamine may prevent isoproterenol-induced cardiac fibrosis by regulating endothelial-to-mesenchymal transition, which is probably mediated by the blockage of the transforming growth factor-ß1/Smad pathway.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Fibrosis/prevención & control , Mesodermo/citología , Miocardio/patología , Extractos Vegetales/uso terapéutico , Quinazolinas/uso terapéutico , Animales , Ecocardiografía , Endotelio Vascular/citología , Evodia , Fibrosis/inducido químicamente , Corazón/efectos de los fármacos , Isoproterenol , Masculino , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/biosíntesis
13.
Hypertension ; 68(6): 1393-1399, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27698061

RESUMEN

Identifying the key factor involved in cardiac remodeling is critically important for developing novel strategies to protect against heart failure. Here, the role of Mnk1 (mitogen-activated protein kinase-interacting kinase 1) in cardiac remodeling was clarified. Cardiac remodeling was induced by transverse aortic constriction in Mnk1-knockout mice and their wild-type control mice. After 4 weeks of transverse aortic constriction, Mnk1-knockout mice developed exaggerated cardiac hypertrophy, fibrosis, dysfunction, and cardiomyocyte apoptosis and showed increased ERK1/2 (extracellular signal-regulated kinase 1/2) activation along with reduced sprouty2 expression. In line with the in vivo studies, Mnk1 knockdown by Mnk1 siRNA transfection induced exaggerated angiotensin II-induced cardiomyocyte hypertrophy in neonatal rat ventricular myocytes (NRVMs). Moreover, adenovirus-mediated overexpression of Mnk1 in NRVMs protected cardiomyocytes from angiotensin II-induced hypertrophy. In addition, overexpression of sprouty2 rescued NRVMs with Mnk1 knockdown from angiotensin II-induced hypertrophy. In accordance with the in vivo studies, as compared with the control group, Mnk1 knockdown led to hyperphosphorylation of ERK1/2 and suppression of the sprouty2 expression in angiotensin II-treated NRVMs; furthermore, Mnk1 overexpression led to hypophosphorylation of ERK1/2 in angiotensin II-treated NRVMs. In addition, sprouty2 overexpression suppressed the activation of ERK1/2 in angiotensin II-treated NRVMs with Mnk1 knockdown. Impressively, MnK1-knockout mice with overexpression of sprouty2 exhibited signs of a blunted cardiac hypertrophic response. Mnk1 likely carries out a suppressive function in cardiac hypertrophy via regulating the sprouty2/ERK1/2 pathway. It implicates Mnk1 in the development of cardiac remodeling.


Asunto(s)
Cardiomegalia/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Remodelación Ventricular/fisiología , Análisis de Varianza , Angiotensina II/farmacología , Animales , Biomarcadores/metabolismo , Cardiomegalia/fisiopatología , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/deficiencia , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Distribución Aleatoria , Transducción de Señal
14.
Exp Ther Med ; 12(4): 2027-2038, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27698689

RESUMEN

Bisoprolol is a drug that acts via the mechanism of specifically and selectively inhibiting the ß1-adrenoreceptor in cardiac myocytes, and provides a pure reduction of heart rate without changing other cardiac parameters. It has long been clinically used to treat cerebrovascular and cardiovascular illnesses. However, there is little information available on whether the role of bisoprolol in the attenuation of ventricular remodeling is dependent upon the achievement of a target dose, and whether it must be used as a preferred option. The aim of the present study was to clarify the underlying benefits of bisoprolol in the attenuation of pressure overload-induced cardiac hypertrophy and fibrosis at different doses. C57BL/6J male mice, aged 6-8 weeks, were treated with saline or one of three different doses of bisoprolol (Biso: 2.5, 5 or 10 mg/kg/day) for 8 weeks from day 1 after aortic banding (AB). A number of mice underwent sham surgery and were treated with saline or bisoprolol. The mice were randomly assigned into the sham (n=24) and AB (n=62) groups. The results revealed that bisoprolol had a protective role against the cardiac hypertrophy, fibrosis and dysfunction caused by AB. This was determined on the basis of heart/body and lung/body weight ratios and heart weight/tibia length ratios, as well as echocardiographic and hemodynamic parameters, histological analysis, and the gene expression levels of hypertrophic and fibrotic markers. The present study revealed that administration of bisoprolol for a long time period may enhance its role in the prevention of cardiac hypertrophy and fibrosis induced by AB, whereas no statistically significant difference was observed between the middle- and high-doses. These observations indicated that the function of bisoprolol in protecting against cardiac hypertrophy, fibrosis and dysfunction is time-dependent. Furthermore, it is proposed that a middle dose of bisoprolol may be a better option for patients with cardiovascular illnesses, particularly those undertaking coronary artery bypass graft and cardiac pacemaker surgeries. These promising results require further clinical investigation.

15.
Clin Sci (Lond) ; 130(22): 2061-2071, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27580926

RESUMEN

OX40, which belongs to the tumour necrosis factor (TNF)-receptor family, is a costimulatory receptor that can potentiate T-cell receptor signalling on the surface of T-lymphocytes. The role of OX40 in non-immune systems, particularly the cardiovascular system, has not been defined. In the present study, we observed a noticeable increase in OX40 expression during cardiac remodelling in rodent heart. In the present study, cardiac hypertrophy was induced by aortic banding (AB) in OX40 knockout (KO) mice and wild-type (WT) mice. After 8 weeks, the OX40 KO mice showed significantly attenuated cardiac hypertrophy, fibrosis and inflammation as well as preserved cardiac function compared with the WT mice. Follow-up in vitro studies suggested that CD4+ T-lymphocyte proliferation and pro-inflammatory cytokine release were significantly decreased, whereas anti-inflammatory cytokine release was considerably increased in OX40 KO mice compared with WT mice as assessed by Cell Counting Kit-8 (CCK-8) assay and ELISA. Co-culturing neonatal rat cardiomyocytes with the activated supernatant of CD4+ T-lymphocytes from OX40 KO mice reduced the hypertrophy response. Interestingly, OX40 KO mice with reconstituted CD4+ T-lymphocytes presented deteriorated cardiac remodelling. Collectively, our data indicate that OX40 regulates cardiac remodelling via the modulation of CD4+ T-lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Cardiomegalia/metabolismo , Receptores OX40/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Proliferación Celular , Humanos , Ratones , Ratones Noqueados , Miocitos Cardíacos/metabolismo , Ratas , Receptores OX40/genética
16.
Mol Cell Biochem ; 417(1-2): 87-96, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27160937

RESUMEN

Diabetic cardiomyopathy, characterized by the presence of diastolic and/or systolic myocardial dysfunction, is one of the major causes of heart failure. Nobiletin, which is extracted from the fruit peel of citrus, is reported to possess anti-inflammatory, anti-oxidative, and hypolipidemic properties. The purpose of this study was to investigate whether nobiletin exerts the therapeutic effect on streptozotocin-induced diabetic cardiomyopathy (DCM) in mice. 80 experimental male C57BL mice were randomly assigned into four groups: sham + vehicle (VEH/SH), sham + nobiletin (NOB/SH), DCM + vehicle (VEH/DM), and DCM + nobiletin (NOB/DM). Nobiletin treatment ameliorated cardiac dysfunction in the DCM group, as shown by the result of echocardiography and hemodynamic measurements. Nobiletin treatment also blunted the mRNA expression of NADPH oxidase isoforms p67(phox), p22(phox), and p91(phox), and abated oxidative stress. Although administration of diabetic mice with nobiletin did not significantly effect the level of blood glucose, it decreased the TGF-ß1, CTGF, fibronectin, and collagen Iα expressions and blunted cardiac fibrosis. In addition, nobiletin inhibited the activation of c-Jun NH2-terminal kinase (JNK), P38, and NF-κB in the cardiac tissue of diabetic mice. Collectively, our study indicates that treatment with nobiletin mitigates cardiac dysfunction and interstitial fibrosis, and these beneficial of nobiletin may belong to the suppression of JNK, P38, and NF-κB signaling pathways.


Asunto(s)
Diabetes Mellitus Experimental/prevención & control , Cardiomiopatías Diabéticas/prevención & control , Flavonas/farmacología , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Masculino , Ratones , Miocardio/patología
17.
Br J Pharmacol ; 173(9): 1502-16, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26845648

RESUMEN

BACKGROUND AND PURPOSE: Activation of glucagon-like peptide-1 (GLP-1) receptor exerts a range of cardioprotective effects. Geniposide is an agonist of GLP-1 receptor, but its role in cardiac hypertrophy remains completely unknown. Here, we have investigated its protective effects and clarified the underlying molecular mechanisms. EXPERIMENTAL APPROACH: The transverse aorta was constricted in C57/B6 mice and then geniposide was given orally for 7 weeks. Morphological changes, echocardiographic parameters, histological analyses and hypertrophic markers were used to evaluate hypertrophy. KEY RESULTS: Geniposide inhibited the hypertrophic response induced by constriction of the transverse aorta or by isoprenaline. Activation of 5'-AMP-activated protein kinase-α (AMPKα) and inhibition of mammalian target of rapamycin, ERK and endoplasmic reticulum stress were observed in hypertrophic hearts that were treated with geniposide. Furthermore, Compound C (CpC) or knock-down of AMPKα restricted protection of geniposide against cell hypertrophy and activation of mammalian target of rapamycin and ERK induced by hypertrophic stimuli. CpC or shAMPKα also abolished the protection of geniposide against endoplasmic reticulum stress induced by thapsigargin or dihtiothreitol. The cardio-protective effects of geniposide were ablated in mice subjected to CpC. GLP-1receptor blockade counteracted the anti-hypertrophic response and activation of AMPKα by geniposide. Knock-down of GLP-1 receptor also offset the inhibitory effects of geniposide on cardiac hypertrophy in vivo. CONCLUSIONS AND IMPLICATIONS: Geniposide protected against cardiac hypertrophy via activation of the GLP-1 receptor/AMPKα pathway. Geniposide is a potential therapeutic drug for cardiac hypertrophy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/prevención & control , Péptido 1 Similar al Glucagón/metabolismo , Iridoides/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Cardiomegalia/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Péptido 1 Similar al Glucagón/agonistas , Masculino , Ratones , Ratones Endogámicos C57BL , Relación Estructura-Actividad
18.
Exp Ther Med ; 11(2): 415-420, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26893624

RESUMEN

Patients with septic shock suffer from high mortality rates, particularly when complicated by severe myocardial depression which is characterized by hypotension and a reduction in cardiac output. Inflammation is an important factor involved in the early stages of sepsis. The aim of the present study was to investigate the effect of the Chinese herbal compound puerarin (1, 5, 10, 20 and 40 µM) on cardiomyocyte inflammatory response in a sepsis model using H9c2 cardiomyocytes stimulated with 1 µg/ml lipopolysaccharide (LPS). The mRNA expression levels of tumor necrosis factor (TNF)-α and interleukin (IL)-ß were evaluated using reverse transcription-quantitative polymerase chain reaction. In addition, the protein expression levels of various factors were determined using western blot analysis. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling was used to evaluate the apoptosis rates in the various groups, and immunocytochemical analysis was employed to determine the effect of puerarin on the nuclear translocation of p65 protein. The present study demonstrated that LPS stimulation increased IL-1ß and TNF-α mRNA expression levels, as compared with the controls (P<0.05). Following treatment with various concentrations of puerarin, the expression levels of IL-1ß and TNF-α were markedly blunted, particularly in the LPS + 40 µM puerarin group (P<0.05 vs. the LPS group). Furthermore, puerarin administration significantly inhibited LPS-induced apoptosis in H9c2 cardiomyocytes, as determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) staining (TUNEL positive cells: LPS + 40 µM puerarin group, 5.5% vs. LPS group, 10.5%; P<0.01). In addition, puerarin significantly decreased LPS-induced phosphorylated nuclear factor (p-NF)-κB p65 and Bax expression levels, and increased the expression levels of Bcl-2, as compared with the LPS group (P<0.05). These data indicated that puerarin may serve as a valuable protective agent against cardiovascular inflammatory diseases.

20.
Mol Med Rep ; 13(1): 980-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26648261

RESUMEN

Shensongyangxin (SSYX) is a medicinal herb, which has long been used in traditional Chinese medicine. Various pharmacological activities of SSYX have been identified. However, the role of SSYX in cardiac hypertrophy remains to be fully elucidated. In present study, aortic banding (AB) was performed to induce cardiac hypertrophy in mice. SSYX (520 mg/kg) was administered by daily gavage between 1 and 8 weeks following surgery. The extent of cardiac hypertrophy was then evaluated by pathological and molecular analyses of heart tissue samples. In addition, in vitro experiments were performed to confirm the in vivo results. The data of the present study demonstrated that SSYX prevented the cardiac hypertrophy and fibrosis induced by AB, as assessed by measurements of heart weight and gross heart size, hematoxylin and eosin staining, cross­sectional cardiomyocyte area and the mRNA expression levels of hypertrophic markers. SSYX also inhibited collagen deposition and suppressed the expression of transforming growth factor ß (TGFß), connective tissue growth factor, fibronectin, collagen Ⅰα and collagen Ⅲα, which was mediated by the inhibition of the TGFß/small mothers against decapentaplegic (Smad) signaling pathway. The inhibitory action of SSYX on cardiac hypertrophy was mediated by the inhibition of Akt signaling. In vitro investigations in the rat H9c2 cardiac cells also demonstrated that SSYX attenuated angiotensin II­induced cardiomyocyte hypertrophy. These findings suggested that SSYX attenuated cardiac hypertrophy and fibrosis in the pressure overloaded mouse heart. Therefore, the cardioprotective effect of SSYX is associated with inhibition of the Akt and TGFß/Smad signaling pathways.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Medicamentos Herbarios Chinos/administración & dosificación , Medicina Tradicional China , Miocitos Cardíacos/efectos de los fármacos , Animales , Cardiomegalia/metabolismo , Cardiomegalia/patología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Miocitos Cardíacos/metabolismo , Presión , Transducción de Señal/efectos de los fármacos , Proteína Smad2/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...