Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 82: 40-50, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24705155

RESUMEN

Recent evidence has suggested that cigarette smoking is associated with an increased prevalence of heart diseases. Given that cigarette smoking triggers proinflammatory response via stimulation of the capsaicin-sensitive transient receptor potential cation channel TRPV1, this study was designed to evaluate the effect of an essential α,ß-unsaturated aldehyde from cigarette smoke crotonaldehyde on myocardial function and the underlying mechanism with a focus on TRPV1 and mitochondria. Cardiomyocyte mechanical and intracellular Ca2+ properties were evaluated including peak shortening (PS), maximal velocity of shortening/relengthening (±dL/dt), time-to-PS (TPS), time-to-90% relengthening (TR90), fura-2 fluorescence intensity (FFI), intracellular Ca2+ decay and SERCA activity. Apoptosis and TRPV1 were evaluated using Western blot analysis. Production of reactive oxygen species (ROS) and DNA damage were measured using the intracellular fluoroprobe 5-(6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate and 8-hydroxy-2'-deoxyguanosine (8-OHdG), respectively. Our data revealed that crotonaldehyde interrupted cardiomyocyte contractile and intracellular Ca2+ property including depressed PS, ±dL/dt, ΔFFI and SERCA activity, as well as prolonged TR90 and intracellular Ca2+ decay. Crotonaldehyde exposure increased TRPV1 and NADPH oxidase levels, promoted apoptosis, mitochondrial injury (decreased aconitase activity, PGC-1α and UCP-2) as well as production of ROS and 8-OHdG. Interestingly, crotonaldehyde-induced cardiac defect was obliterated by the ROS scavenger glutathione and the TRPV1 inhibitor capsazepine. Capsazepine (not glutathione) ablated crotonaldehyde-induced mitochondrial damage. Capsazepine, glutathione and the NADPH inhibitor apocynin negated crotonaldehyde-induced ROS accumulation. Our data suggest a role of crotonaldehyde compromises cardiomyocyte mechanical function possibly through a TRPV1- and mitochondria-dependent oxidative stress mechanism.


Asunto(s)
Aldehídos/farmacología , Mitocondrias Cardíacas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Aconitato Hidratasa/metabolismo , Animales , Calcio/metabolismo , Caspasa 3/metabolismo , Células Cultivadas , Daño del ADN , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Masculino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica , Miocitos Cardíacos/fisiología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Fumar
2.
Expert Opin Drug Saf ; 22(2): 133-140, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36803188

RESUMEN

BACKGROUND: Dapagliflozin has proven cardioprotective and nephroprotective effects. However, the risk of all-cause death with dapagliflozin remains unclear. RESEARCH DESIGN AND METHODS: We performed a meta-analysis of phase III randomized controlled trials (RCTs) for the risk of all-cause death and safety events with dapagliflozin compared to placebo. PubMed and EMBASE were searched from inception to 20 September 2022. RESULTS: Five trials were included in the final analysis. Compared with the placebo, dapagliflozin demonstrated an 11.2% reduction in the risk of all-cause death (OR 0.88, 95% CI 0.81-0.94). No statistically significant difference in urinary tract infection (OR: 0.95, 95% CI: 0.78 to 1.17), bone fracture (OR: 1.06, 95% CI: 0.94 to 1.20), and amputation (OR: 1.01, 95% CI: 0.82 to 1.23) was observed between patients treated with dapagliflozin and placebo. Compared with placebo, dapagliflozin was associated with a significant reduction in acute kidney injury (OR: 0.71, 95% CI: 0.60 to 0.83), and increased the risk of genital infection (OR: 8.21, 95% CI: 4.19 to 16.12). CONCLUSIONS: Dapagliflozin was associated with significantly reduced all-cause death and increased genital infection. Dapagliflozin was safe concerning urinary tract infection, bone fracture, amputation, and acute kidney injury, compared with the placebo.


Asunto(s)
Lesión Renal Aguda , Diabetes Mellitus Tipo 2 , Fracturas Óseas , Infecciones Urinarias , Humanos , Hipoglucemiantes/efectos adversos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Compuestos de Bencidrilo/efectos adversos , Infecciones Urinarias/inducido químicamente , Infecciones Urinarias/tratamiento farmacológico , Lesión Renal Aguda/inducido químicamente , Fracturas Óseas/epidemiología , Fracturas Óseas/prevención & control , Fracturas Óseas/inducido químicamente
3.
ESC Heart Fail ; 10(2): 1231-1241, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36702979

RESUMEN

AIMS: Head-to-head comparisons among SGLT2 inhibitors treatments in established heart failure remain absent. We conducted a systematic review of dedicated heart failure trials to assess indirectly the composite outcomes and individual clinical endpoints among SGLT2 inhibitor treatments. METHODS AND RESULTS: We systematically reviewed randomized controlled trials comparing SGLT2 inhibitors versus placebo in patients with established heart failure. A Bayesian approach to network meta-analysis was applied. Five trials including four treatment strategies were included in this study. The composite of cardiovascular death or hospitalization for heart failure showed no significant difference in the comparison between dapagliflozin and empagliflozin (OR 1.00, 95% CI 0.66-1.55), dapagliflozin and sotagliflozin (OR 1.54, 95% CI 0.91-2.65), and empagliflozin and sotagliflozin (OR 1.53, 95% CI 0.90-2.69). All-cause mortality showed no significant difference in the comparison between dapagliflozin and empagliflozin (OR 0.92, 95% CI 0.711-1.18), dapagliflozin and sotagliflozin (OR 1.05, 95% CI 0.68-1.59), and empagliflozin and sotagliflozin (OR 1.14, 95% CI 0.74-1.73). Cardiovascular death showed no significant difference in the comparison between dapagliflozin and empagliflozin (OR 0.94, 95% CI 0.71-1.23), dapagliflozin and sotagliflozin (OR 0.96, 95% CI 0.61-1.55), and empagliflozin and sotagliflozin (OR 1.03, 95% CI 0.64-1.66). Hospitalization for heart failure showed no significant difference in the comparison between dapagliflozin and empagliflozin (OR 1.13, 95% CI 0.64-1.97), dapagliflozin and sotagliflozin (OR 1.56, 95% CI 0.74-3.15), and empagliflozin and sotagliflozin (OR 1.39, 95% CI 0.68-2.78). CONCLUSIONS: In patients with established heart failure, there was no significant difference of the major efficacy outcomes among SGLT2 inhibitor treatments; however, sotagliflozin may be associated with the lowest risk of the composite of cardiovascular death or hospitalization for heart failure, and dapagliflozin may be associated with the lowest risk of all-cause and cardiovascular mortality.


Asunto(s)
Diabetes Mellitus Tipo 2 , Insuficiencia Cardíaca , Inhibidores del Cotransportador de Sodio-Glucosa 2 , Humanos , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico , Teorema de Bayes , Diabetes Mellitus Tipo 2/complicaciones , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/complicaciones
4.
Arch Biochem Biophys ; 511(1-2): 1-7, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21530483

RESUMEN

Activation of adenosine monophosphate-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy through peroxisome proliferators-activated receptor-α (PPARα) signaling pathway, but the detailed mechanism remains unclear. A rat model of cardiac hypertrophy created by transaortic constriction (TAC) was used to investigate the mechanism involved in regulation of PPARα activity by AMPK. It was observed that treatment with AICAR (5-aminoimidazole 1 carboxamide ribonucleoside), an AMPK activator, significantly inhibited cardiac hypertrophy in vivo and in vitro. Phosphorylated extracellular signal regulated protein kinase (phospho-ERK1/2) and phospho-p38 mitogen-activated protein kinase (MAPK) protein levels were significantly up-regulated, while PPARα protein level was down-regulated in TAC rats. AICAR treatment reversed the changes of PPARα and phospho-ERK1/2, but increased phospho-p38 MAPK protein level in TAC rats. Similar changes of PPARα and phospho-ERK1/2 protein levels were observed in the hypertrophied cardiomyocytes induced by phenylephrine treatment. Epidermal growth factor (EGF, ERK1/2 activator), but not SB203580 (p38 inhibitor) blocked the up-regulation of PPARα protein level induced by AICAR. Luciferase assay showed that AICAR increased PPARα transcriptional activity which was abrogated by EGF, but not by SB203580. These results demonstrate that AMPK activation enhances the activity of PPARα to inhibit cardiac hypertrophy through ERK1/2, but not p38 MAPK, signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/metabolismo , Cardiomegalia/prevención & control , Sistema de Señalización de MAP Quinasas , PPAR alfa/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Cardiomegalia/etiología , Cardiomegalia/patología , Células Cultivadas , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Clin Exp Pharmacol Physiol ; 38(1): 55-62, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21083698

RESUMEN

1. Metformin is an activator of AMP-activated protein kinase (AMPK). Recent studies suggest that pharmacological activation of AMPK inhibits cardiac hypertrophy. In the present study, we examined whether long-term treatment with metformin could attenuate ventricular hypertrophy in a rat model. The potential involvement of nitric oxide (NO) in the effects of metformin was also investigated. 2. Ventricular hypertrophy was established in rats by transaortic constriction (TAC). Starting 1 week after the TAC procedure, rats were treated with metformin (300 mg/kg per day, p.o.), N(G)-nitro-L-arginine methyl ester (L-NAME; 50 mg/kg per day, p.o.) or both for 8 weeks prior to the assessment of haemodynamic function and cardiac hypertrophy. 3. Cultured cardiomyocytes were used to examine the effects of metformin on the AMPK-endothelial NO synthase (eNOS) pathway. Cells were exposed to angiotensin (Ang) II (10⁻6 mol/L) for 24 h under serum-free conditions in the presence or absence of metformin (10⁻³ mol/L), compound C (10⁻6 mol/L), L-NAME (10⁻6 mol/L) or their combination. The rate of incorporation of [³H]-leucine was determined, western blotting analyses of AMPK-eNOS, neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS) were undertaken and the concentration of NO in culture media was determined. 4. Transaortic constriction resulted in significant haemodynamic dysfunction and ventricular hypertrophy. Myocardial fibrosis was also evident. Treatment with metformin improved haemodynamic function and significantly attenuated ventricular hypertrophy. Most of the effects of metformin were abolished by concomitant L-NAME treatment. L-NAME on its own had no effect on haemodynamic function and ventricular hypertrophy in TAC rats. 5. In cardiomyocytes, metformin inhibited AngII-induced protein synthesis, an effect that was suppressed by the AMPK inhibitor compound C or the eNOS inhibitor L-NAME. The improvement in cardiac structure and function following metformin treatment was associated with enhanced phosphorylation of AMPK and eNOS and increased NO production. 6. The findings of the present study indicate that long-term treatment with metformin could attenuate ventricular hypertrophy induced by pressure overload via activation of AMPK and a downstream signalling pathway involving eNOS-NO.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Hipertrofia Ventricular Izquierda/prevención & control , Metformina/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Activación Enzimática/efectos de los fármacos , Hemodinámica , Hipertrofia Ventricular Izquierda/metabolismo , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Masculino , Metformina/uso terapéutico , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
6.
Exp Ther Med ; 21(4): 323, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33732296

RESUMEN

Endothelial-cell (EC) apoptosis serves a vital role in the pathogenesis of atherosclerosis. Accumulating evidence has implicated microRNA (miRNA/miR) dysregulation in EC apoptosis. Although the role of miR-454-3p in carcinogenesis has been well documented, its role and underlying mechanism in EC apoptosis remain unclear. In the present study, the results revealed that miR-454-3p expression was substantially downregulated in human aortic endothelial cells (HAECs) following oxidized low-density lipoprotein (ox-LDL) treatment. miR-454-3p suppression significantly attenuated the viability of HAECs, while miR-454-3p overexpression repressed ox-LDL-induced HAEC apoptosis. Bioinformatics analysis and luciferase reporter assays revealed that transient receptor potential canonical 3 (TRPC3), a key regulator of atherosclerosis development, was the direct target of miR-454-3p. Furthermore, TRPC3 overexpression abolished the anti-apoptotic effect of miR-454-3p on HAECs. These results revealed a novel role of miR-454-3p in ox-LDL-induced apoptosis in HAECs.

7.
Acta Pharmacol Sin ; 31(7): 798-804, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581852

RESUMEN

AIM: To examine the inhibitory effects of adenosine monophosphate-activated protein kinase (AMPK) activation on cardiac hypertrophy in vitro and to investigate the underlying molecular mechanisms. METHODS: Cultured neonatal rat cardiomyocytes were treated with the specific AMPK activator 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR) and the specific AMPK antagonist Compound C, and then stimulated with phenylephrine (PE). The Muscle RING finger 1 (MuRF1)-small interfering RNA (siRNA) was transfected into cardiomyocytes using Lipofectamine 2000. The surface area of cultured cardiomyocytes was measured using planimetry. The protein degradation was determined using high performance liquid chromatography (HPLC). The expression of beta-myosin heavy chain (beta-MHC) and MuRF1, as well as the phosphorylation levels of AMPK and Forkhead box O 1 (FOXO1), were separately measured using Western blot or real-time polymerase chain reaction. RESULTS: Activation of AMPK by AICAR 0.5 mmol/L inhibited PE-induced increase in cardiomyocyte area and beta-MHC protein expression and PE-induced decrease in protein degradation. Furthermore, AMPK activation increased the activity of transcription factor FOXO1 and up-regulated downstream atrogene MuRF1 mRNA and protein expression. Treatment of hypertrophied cardiomyocytes with Compound C 1 micromol/L blunted the effects of AMPK on cardiomyocyte hypertrophy and changes to the FOXO1/MuRF1 pathway. The effects of AICAR on cardiomyocyte hypertrophy were also blocked after MuRF1 was silenced by transfection of cardiomyocytes with MuRF1-siRNA. CONCLUSION: The present study demonstrates that AMPK activation attenuates cardiomyocyte hypertrophy by modulating the atrophy-related FOXO1/MuRF1 signaling pathway in vitro.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/enzimología , Factores de Transcripción Forkhead/metabolismo , Proteínas Musculares/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Quinasas Activadas por AMP/efectos de los fármacos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Cardiomegalia/tratamiento farmacológico , Células Cultivadas , Activación Enzimática , Silenciador del Gen , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fenilefrina/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos , Transfección , Proteínas de Motivos Tripartitos
8.
Acta Biochim Biophys Sin (Shanghai) ; 42(4): 253-8, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20383463

RESUMEN

Proteasome inhibitors are involved in cell cycle control, growth and inflammatory signaling, and transcriptional regulation of mitotic cells. A recent study has suggested that specific proteasome inhibitor MG132 may suppress cardiomyocyte hypertrophy in vitro. However, the underlying molecular mechanisms are not clear. In this study, we investigated the effects of long-term MG132 treatment on cardiac hypertrophy and the related molecular mechanisms in vivo. MG132 (0.1 mg/kg/day) was intraperitoneally injected to rats with abdominal aortic banding (AAB) for 8 weeks. Results showed that treatment with MG132 significantly attenuated left ventricular (LV) myocyte area, LV weight/body weight, and lung weight/body weight ratios, decreased LV diastolic diameter and wall thickness, and increased fractional shortening in AAB rats. AAB induced the phosphorylation of ERK1/2, JNK1, and p38 in cardiac myocytes. The elevated phosphorylation levels of ERK1/2 and JNK1 in AAB rats were significantly reversed by MG132 treatment. In conclusion, our results suggested that long-term treatment with MG132 attenuates pressureoverload-induced cardiac hypertrophy and improves cardiac function in AAB rats through regulation of ERK1/2 and JNK1 signaling pathways.


Asunto(s)
Cardiomegalia/patología , Leupeptinas/farmacología , Sistema de Señalización de MAP Quinasas , Inhibidores de Proteasas/farmacología , Inhibidores de Proteasoma , Animales , Aorta/metabolismo , Aorta/patología , Ecocardiografía/métodos , Ventrículos Cardíacos/patología , Masculino , Fosforilación , Presión , Ratas , Ratas Sprague-Dawley , Transducción de Señal
9.
Acta Cardiol ; 64(6): 735-40, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20128148

RESUMEN

OBJECTIVE: Vascular endothelial growth factor (VEGF), angiopoietin (Ang)-1 and -2 regulate angiogenesis and might be important in myocardial collateral development. Elevated levels of angiogenic growth factors in patients with coronary artery disease (CAD) have been reported. However, the age-related change of angiogenic growth factors in patients with CAD remains unclear. METHODS AND RESULTS: Serum VEGF, Ang-1 and -2 levels were measured by enzyme-linked immunosorbent assay kits. Serum VEGF, Ang-1 and -2 levels in patients with CAD were significantly higher than those in healthy control subjects. In patients aged <61 years and 61 to 70 years, serum Ang-1 and -2 levels were significantly higher than in patients aged >70 years, serum Ang-2 levels in patients aged <61 years were significantly higher than in patients aged from 61 to 70 years. Serum VEGF levels were not significantly different in the three age groups. Serum VEGF, Ang-1 and -2 levels were not significantly different across the age groups in healthy subjects. Regression analysis showed that there was a negative correlation between age and Ang-1 and -2 in patients with CAD. CONCLUSION: Serum Ang-1 and -2 levels, but notVEGF levels in patients with CAD, are decreased along with advancing age.


Asunto(s)
Angiopoyetina 1/sangre , Angiopoyetina 2/sangre , Enfermedad de la Arteria Coronaria/sangre , Factor A de Crecimiento Endotelial Vascular/sangre , Adulto , Factores de Edad , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Modelos Lineales , Persona de Mediana Edad , Pronóstico
10.
Zhong Xi Yi Jie He Xue Bao ; 4(1): 48-51, 2006 Jan.
Artículo en Zh | MEDLINE | ID: mdl-16409970

RESUMEN

OBJECTIVE: To investigate the effects of Naohuandan Recipe on learning and memory abilities of SAM-P/8 mice and its role in anti-oxidation and anti-apoptosis. METHODS: Forty SAM-P/8 mice were randomly divided into four groups, which were untreated (normal saline-treated) group, Yinkeluo Tablets (extracts of gingko leaf)-treated group, low-dose Naohuandan Recipe-treated group and high-dose Naohuandan Recipe-treated group. Mice in these groups were given corresponding drugs orally for 105 days. Then the performances of learning and memory of mice were tested by a step-down passive avoidance task and a Y-maze test. The serum levels of superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were detected. The expression level of bcl-xl mRNA in cerebral cortex and hippocampus of mice was detected by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: The performances of learning and memory in the Yinkeluo Tablets-treated group, low- and high-dose Naohuandan Recipe-treated groups were significantly improved as compared with those in the untreated group (P<0.05 or P<0.01), and such performance was the best in the high-dose Naohuandan Recipe-treated group among these four groups (P<0.01). The serum levels of SOD and GSH-Px and the expression of bcl-xl mRNA in cerebral cortex and hippocampus of mice in the Yinkeluo Tablets-treated group, low- and high-dose Naohuandan Recipe-treated groups were also significantly higher than those in the untreated group (P<0.05 or P<0.01), while the serum level of MDA in the untreated group was higher than that in the other three groups (P<0.01). CONCLUSION: Naohuandan Recipe can improve learning and memory abilities of SAM-P/8 mice, and this effect may be related to its anti-oxidation efficacy and enhancement of expression level of bcl-xl mRNA.


Asunto(s)
Envejecimiento/metabolismo , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Aprendizaje/efectos de los fármacos , Envejecimiento/genética , Animales , Glutatión Peroxidasa/sangre , Memoria/efectos de los fármacos , Ratones , Ratones Mutantes/genética , Ratones Mutantes/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Superóxido Dismutasa/sangre , Proteína bcl-X/biosíntesis , Proteína bcl-X/genética
11.
Chin J Integr Med ; 11(2): 111-6, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16150197

RESUMEN

OBJECTIVE: To observe the therapeutic effect and mechanism of Naohuandan (NHD) in treating senile dementia (SD). METHODS: Clinical study: Fifty-eight patients with SD, whose diagnosis conforms to the Diagnostic Standard of DSM-IV issued by American Association of Psychiatry, were enrolled and randomly assigned into two groups. The 30 patients in the treated group were treated with NHD, 4 capsules each time, 3 times daily. The 28 patients in the control group were treated with Piracetam, 1.6 g each time, 3 times daily. The therapeutic course for both groups was 3 months. The therapeutic efficacy was estimated and compared by comprehensive scores of memory and cognition, scores of Mini-mental State Examination (MMSE) and Activities of Daily Living (ADL). Experimental study: Rats were divided into the control group, the model group and the high-dosage and low-dosage NHD treated groups. The protective effect of NHD on the per-oxidative damage of hippocampal neurons in beta-amyloid protein induced SD model was observed and the related criteria were determined. RESULTS: Clinical study showed that both NHD and Piracetam could improve the clinical symptoms of patients, the two medicines showing insignificant difference in total effective rate. But NHD was better in elevating MMSE score and lowering ADL score in patients than Piracetam (P < 0.05 and P < 0.01). Experimental study showed that (1) 24 and 72 hrs after modeling, the activity of SOD and GSH were lower and the level of MDA higher in the model group than those in the control group (P < 0.05 or P < 0.01). Compared with the model group at the corresponding time points, in the high-dosage NHD group, SOD and GSH were higher, MDA was lower (P < 0.05 or P < 0.01); but in the low-dosage NHD group, SOD at the 72nd hr was higher (P < 0.05) and MDA at 24th and 72nd hrs was lower (P < 0.01). And most of the criteria in the high-dosage NHD group was improved better than that in the low-dosage NHD group. (2) The survival rates of neurons in various groups were not different significantly (P > 0.05) 24 hrs after modeling, but that in the high-dosage NHD group was significantly higher than that in the model group (P < 0.01) and in the low-dosage NHD group 72 hrs after modeling (P < 0.05). CONCLUSION: NHD is an effective Chinese herbal preparation for treatment of SD, and its mechanism is related with its inhibition on peroxidative injury and protection on neurons.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Medicamentos Herbarios Chinos/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Anciano , Anciano de 80 o más Años , Animales , Supervivencia Celular/efectos de los fármacos , Femenino , Glutatión/metabolismo , Hipocampo/citología , Humanos , Masculino , Malondialdehído/metabolismo , Persona de Mediana Edad , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Pruebas Neuropsicológicas , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/metabolismo
12.
Cardiovasc Toxicol ; 13(4): 307-15, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23632742

RESUMEN

The aim of the present study was to examine the effects of acute infrasound exposure on oxidative damage and investigate the underlying mechanisms in rat cardiomyocytes. Neonatal rat cardiomyocytes were cultured and exposed to infrasound for several days. In the study, the expression of CAT, GPx, SOD1, and SOD2 and their activities in rat cardiomyocytes in infrasound exposure groups were significantly decreased compared to those in the various time controls, along with significantly higher levels of O2 (-) and H2O2. Decreased cardiac cell viability was not observed in various time controls. A significant reduction in cardiac cell viability was observed in the infrasound group compared to the control, while significantly increased cardiac cell viability was observed in the infrasound exposure and rosiglitazone pretreatment group. Compared to the control, rosiglitazone significantly upregulated CAT, GPx, SOD1, and SOD2 expression and their activities in rat cardiomyocytes exposed to infrasound, while the levels of O2 (-) or H2O2 were significantly decreased. A potential link between a significant downregulation of PPAR-γ expression in rat cardiomyocytes in the infrasound group was compared to the control and infrasound-induced oxidative stress. These findings indicate that infrasound can induce oxidative damage in rat cardiomyocytes by inactivating PPAR-γ.


Asunto(s)
Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/efectos de la radiación , Estrés Oxidativo/fisiología , Estrés Oxidativo/efectos de la radiación , PPAR gamma/biosíntesis , Sonido/efectos adversos , Animales , Células Cultivadas , Regulación de la Expresión Génica , Masculino , Ratas , Ratas Sprague-Dawley
13.
Toxicol Lett ; 199(2): 173-81, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-20837116

RESUMEN

Angiotensin-(1-7) displays antihypertensive and antiproliferative properties although its effect on cardiac remodeling and hypertrophy in hypertension has not been fully elucidated. The present study was designed to examine the effect of chronic angiotensin-(1-7) treatment on myocardial remodeling, cardiac hypertrophy and underlying mechanisms in spontaneous hypertension. Adult male spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto (WKY) rats were treated with or without angiotensin-(1-7) or the angiotensin-(1-7) antagonist A-779 for 24 weeks. Mean arterial pressure, left ventricular geometry, expression of the hypertrophic markers ANP and ß-MHC, collagen contents (type I and III), collagenase (MMP-1), matrix metalloproteinase-2 (MMP-2) and tissue inhibitor of MMPs-1 (TIMP-1) were evaluated in WKY and SHR rats with or without treatment. Our data revealed that chronic angiotensin-(1-7) treatment significantly suppressed hypertension, left ventricular hypertrophy, expression of ANP and ß-MHC as well as myocardial fibrosis in SHR rats, the effects of which were nullified by the angiotensin-(1-7) receptor antagonist A-779. In addition, angiotensin-(1-7) treatment significantly counteracted hypertension-induced changes in the mRNA expression of MMP-2 and TIMP-1 and collagenase activity, the effects of which were blunted by A-779. In vitro study revealed that angiotensin-(1-7) directly increased the activity of MMP-2 and MMP-9 while decreasing the content of TIMP-1 and TIMP-2. Taken together, our results revealed a protective effect of angiotensin-(1-7) against cardiac hypertrophy and collagen deposition, which may be related to concerted changes in MMPs and TIMPs levels. These data indicated the therapeutic potential of angiotensin-(1-7) in spontaneous hypertension-induced cardiac remodeling.


Asunto(s)
Angiotensina I/farmacología , Antihipertensivos/farmacología , Cardiomegalia/prevención & control , Hipertensión/tratamiento farmacológico , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Miocardio/patología , Fragmentos de Péptidos/farmacología , Inhibidores Tisulares de Metaloproteinasas/fisiología , Animales , Factor Natriurético Atrial/análisis , Factor Natriurético Atrial/genética , Presión Sanguínea/efectos de los fármacos , Colágeno/metabolismo , Colagenasas/metabolismo , Fibrosis , Hipertensión/metabolismo , Hipertensión/patología , Masculino , Metaloproteinasa 2 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Miocardio/metabolismo , Cadenas Pesadas de Miosina/análisis , Cadenas Pesadas de Miosina/genética , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Inhibidores Tisulares de Metaloproteinasas/análisis
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 30(11): 2419-22, 2010 Nov.
Artículo en Zh | MEDLINE | ID: mdl-21097394

RESUMEN

OBJECTIVE: To investigate the effects of AICAR on the activity of transcription factor FOXO1 and expression of ubiquitin ligase MuRF1 in rat cardiomyocytes, and explore the possible role of AMP-activated protein kinase (AMPK) in proteolysis pathways. METHODS: In vitro cultured neonatal rat cardiac myocytes were treated with AICAR, and Western blotting was used to detect the phosphorylation of FOXO1 and expression of MuRF1 in the cells. RESULTS: AICAR activated AMPK in rat cardiac myocytes. Activated AMPK significantly inhibited the phosphorylation of FOXO1 and increased MuRF1 protein expression. CONCLUSION: AMPK may regulate proteolysis by activating FOXO1 transcription factor and up-regulating MuRF1 expression.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Musculares/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Células Cultivadas , Ratas , Ratas Sprague-Dawley , Ribonucleótidos/farmacología , Proteínas de Motivos Tripartitos
15.
Eur J Pharmacol ; 620(1-3): 63-70, 2009 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-19699196

RESUMEN

The activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) has been shown to inhibit cardiac hypertrophy, however, the mechanism remains unclear. Rat models of cardiac hypertrophy were created with transaortic constriction (TAC) to investigate the mechanistic role of AMPK involved. RT-PCR and Western blot analyses indicated that hypertrophy marker genes ANP and beta-MHC expression were up-regulated in the myocardium of TAC rats. We also observed that the expressions of peroxisome proliferator-activated receptor-alpha (PPARalpha) and its target genes, carnitine palmitoyl transferase-capital I, Ukrainian (CPT-capital I, Ukrainian) and medium-chain acyl-COA dehydrogenases (MCAD), were down-regulated, and the fatty acid oxidation was decreased in TAC rats. Treatment of TAC animals with 5-aminoimidazole 1 carboxamide ribonucleoside (AICAR, 0.5 mg/g body wt), a specific activator of AMPK, inhibited cardiac hypertrophy in TAC and reversed PPARalpha, CPT-I and MCAD expression and fatty acid oxidation. Similar observations were made in hypertrophied cardiomyocytes induced by phenylephrine in vitro. Treatment of hypertrophied cardiomyocytes with Compound C, a specific AMPK inhibitor, showed an effect opposite to that of AICAR. The effect of AICAR on cardiac hypertrophy was blocked after PPARalpha was silenced by transfection of cardiomyocytes with PPARalpha-siRNA. Luciferase activity assay suggested that AICAR elevates PPARalpha transcriptional activity. These results indicate that AMPK plays an important role in the inhibition of cardiac hypertrophy by activating the PPARalpha signaling pathway.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiomegalia/enzimología , PPAR alfa/metabolismo , Transducción de Señal , Animales , Cardiomegalia/metabolismo , Activación Enzimática , Ácidos Grasos/metabolismo , Masculino , Ratones , Oxidación-Reducción , PPAR alfa/genética , Ratas , Ratas Sprague-Dawley
16.
Eur J Pharmacol ; 602(1): 132-7, 2009 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-19027731

RESUMEN

Although short-term B-type natriuretic peptide (BNP) treatment has been shown to be effective for decompensated congestive heart failure, little is known about the effects of long-term BNP treatment in ventricular remodeling and heart failure in response to myocardial infarction. The aim of the present study was to investigate the effects of long-term BNP treatment on ventricular remodeling and heart failure after myocardial infarction in rats. Myocardial infarction was induced by ligating the left anterior descending coronary artery. The surviving rats were randomly divided into four groups: 1) vehicle-treated myocardial infarction group ('vehicle-treated group'), 2) rats treated with low-dose BNP ('low BNP group'), 3) rats treated with high-dose BNP ('high BNP group'), 4) sham-operated group. Eight weeks after the operation, rats were sacrificed. Compared with the sham-operated group, the vehicle-treated group had significantly higher collagen deposition and angiotensin II levels (P<0.01) and a significantly lower cardiac function (P<0.05). Both BNP-treated groups had significant improvement of these indexes compared with the vehicle-treated group (P<0.01). The high BNP group had significantly less collagen deposition and better cardiac function than the untreated and low BNP groups. Moreover, the mRNA and protein expression of TGFbeta1 and Smad2 in the vehicle-treated group was significantly higher than in the sham-operated group (P<0.01). Both BNP-treated groups had a suppression of TGFbeta1 and Smad2 expression (P<0.01). In conclusion, long-term treatment with BNP prevents ventricular remodeling and deterioration of cardiac function in a dose-dependent fashion, a process that may be associated with the inhibition of TGFbeta1/ Smad2 signaling.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Natriuréticos/farmacología , Natriuréticos/uso terapéutico , Péptido Natriurético Encefálico/farmacología , Péptido Natriurético Encefálico/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Angiotensina II/metabolismo , Animales , Colágeno/metabolismo , Masculino , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA