Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Probiotics Antimicrob Proteins ; 15(5): 1287-1297, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36044175

RESUMEN

Diabetes-related brain complications have been reported in clinical patients and experimental models. The objective of the present study was to investigate the neuroprotective mechanisms of Lactobacillus reuteri GMNL-263 in streptozotocin (STZ)-induced diabetic rats. In this study, three different groups, namely control group, STZ-induced (55 mg/kg streptozotocin intraperitoneally) diabetic rats (DM), and DM rats treated with Lactobacillus reuteri GMNL-263 (1 × 109 CFU/rat/day), were utilized to study the protective effect of GMNL-263 in the hippocampus of STZ-induced diabetic rats. The results demonstrated that GMNL-263 attenuated diabetes-induced hippocampal damage by enhancing the cell survival pathways and repressing both inflammatory and apoptotic pathways. Histopathological analysis revealed that GMNL-263 prevented structural changes in the hippocampus in the DM group and decreased the level of inflammation and apoptosis in the hippocampus of DM rats. The IGF1R cell survival signaling pathway also improved after GMNL-263 treatment. These results indicate that probiotic GMNL-263 exerts beneficial effects in the brain of diabetic rats and has potential ability for clinical application.


Asunto(s)
Diabetes Mellitus Experimental , Limosilactobacillus reuteri , Fármacos Neuroprotectores , Probióticos , Ratas , Animales , Fármacos Neuroprotectores/farmacología , Estreptozocina/efectos adversos , Estreptozocina/metabolismo , Hipocampo
2.
J Biochem Mol Toxicol ; 36(9): e23128, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35698875

RESUMEN

Hypertension is a common chronic cardiovascular disease reported among both men and women. Hypertension in males affects the testis and reproduction function; however, the pathogenesis is poorly understood. Rapamycin has been reported to have a variety of beneficial pharmacological effects; however, high-doses rapamycin does have side effects such as immunosuppression. The present study investigates whether low-dose rapamycin can reduce the damage caused by hypertension to the testis of spontaneously hypertensive rats (SHRs) and further examines molecular mechanism of low-dose rapamycin in preventing testicular toxicity induced by angiotensin II (Ang II). Low rapamycin dose restores the testicle size, histological alterations, 3ß-hydroxysteroid dehydrogenase (3ß-HSD) expression, and prevents apoptosis in SHR rats. Ang II downregulates angiotensin-converting enzyme-2 (ACE2) expression through AT1R, p-ERK, and MAS receptor in LC-540 Leydig cells in a dose-dependent manner. Low doses of rapamycin effectively upregulate steroidogenic enzymes, steroidogenic acute regulatory protein and 3ß-HSD expression in Leydig cells. Rapamycin upregulates ACE2 expression through p-PKAc and p-PI3k in Ang II-treated cells. Further, rapamycin curbs mitochondrial superoxide generation and depleted mitochondrial membrane potential induced by Ang II through activation of Nrf2-mediated Gpx4 and superoxide dismutase 2 expression. Our results revealed the involvement of ACE2, AT1R, AT2R, PKAc, and oxidative stress in Ang-II-induced testicular toxicity, suggesting low-dose rapamycin could be a potential therapeutic candidate to attenuate testicular toxicity.


Asunto(s)
Angiotensina II , Hipertensión , Angiotensina II/farmacología , Enzima Convertidora de Angiotensina 2 , Animales , Femenino , Humanos , Hidroxiesteroide Deshidrogenasas , Hipertensión/inducido químicamente , Hipertensión/tratamiento farmacológico , Hipertensión/metabolismo , Células Intersticiales del Testículo/metabolismo , Masculino , Factor 2 Relacionado con NF-E2 , Fosfatidilinositol 3-Quinasas , Ratas , Ratas Endogámicas SHR , Sirolimus/farmacología , Sirolimus/uso terapéutico , Superóxidos
3.
Biogerontology ; 22(5): 495-506, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34251569

RESUMEN

Ageing is a complex biological process that increases the probability of disease and death, which affects the organs of all species. The accumulation of oxidative damage in the brain contributes to a progressive loss of cognitive functions or even declined the energy metabolism. In this study, we tested the effects of exercise training on the apoptosis, survival, and antioxidant signaling pathways in the cerebral cortex of three age groups of male rats; 3, 12, and 18 months. We observed that H2S and the expression of Nrf2-related antioxidant pathways declined with age and increased after exercise training. IGF1R survival pathway was less increased in middle-aged rats; however, significantly increased after exercise training. The expression of mitochondrial-dependent apoptotic pathway components, such as Bak, cytochrome C, and caspase 3 in the ageing control group, were much higher than those of the exercise training groups. This study demonstrated that exercise training could reduce the apoptosis and oxidative stress that accrues throughout ageing, which causes brain damage.


Asunto(s)
Antioxidantes , Factor 2 Relacionado con NF-E2 , Envejecimiento , Animales , Apoptosis , Corteza Cerebral/metabolismo , Masculino , Factor 2 Relacionado con NF-E2/metabolismo , Ratas
5.
Free Radic Biol Med ; 160: 141-148, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-32745770

RESUMEN

Doxorubicin (DOX) is a widely used antitumor drug that causes severe neurotoxicity in patients. Diallyl trisulfide (DATS) is an organosulfur compound with established potent antioxidant and anti-inflammatory properties. Herein, we investigated the neuroprotective efficacy of DATS in preventing DOX-induced neurotoxicity in a rat model. Specifically, DATS (40 mg/kg) was administered to rats 24 h after DOX treatment, once a week for 8 weeks. Our results showed that DATS treatment led to a decrease in plasma levels of tumor necrosis factor-alpha (TNF-α) induced by DOX. DATS restored cerebral cortex and hippocampus histopathological architecture and neuronal loss. Immunohistochemical staining indicated that DATS decreased the expression of glial fibrillar acidic protein (GFAP) in DOX treated rats. Components of stress-related inflammatory proteins (TNF-α, phospho nuclear factor kappa B (NF-κB), inducible nitricoxide synthase (iNOS) and cyclooxygenase-2 (COX-2)) were all significantly increased in the DOX group, in comparison with the control group, whereas they were decreased after DATS treatment. In addition, the mRNA of antioxidant enzymes (superoxide dismutase 2 (SOD2), catalase, glutathione peroxidase 1, 4 (GPx1 and GPx4)) and antioxidant proteins (heme oxygenase-1 (HO-1), superoxide dismutase 1, 2 (SOD1 and SOD2), Γ-glutamylcysteine synthase (Γ-GCSc)) were markedly increased in DOX group compared with the control group, which were significantly attenuated by DATS treatment. The upregulation of antioxidants enzymes in DOX group was probably a compensatory effect against elevated oxidative stress induced by DOX. DATS treatment could ameliorate this oxidative stress in brain. Our results suggested that DATS has potential clinical applications in the prevention of DOX-induced neurotoxicity by ameliorating inflammatory insults and oxidative stress.


Asunto(s)
Compuestos Alílicos , Antibióticos Antineoplásicos , Apoptosis , Doxorrubicina , Estrés Oxidativo , Sulfuros , Compuestos Alílicos/farmacología , Animales , Antibióticos Antineoplásicos/toxicidad , Antioxidantes , Encéfalo , Doxorrubicina/toxicidad , Humanos , Inflamación , Estrés Oxidativo/efectos de los fármacos , Ratas , Sulfuros/farmacología
6.
Aging (Albany NY) ; 12(8): 6852-6864, 2020 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-32320382

RESUMEN

Hippocampus is one of the most vulnerable brain regions in terms of age-related pathological change. Exercise is presumed to delay the aging process and promote health because it seems to improve the function of most of the aging mechanisms. The purpose of this study is to evaluate the effects of swimming exercise training on brain inflammation, apoptotic and survival pathways in the hippocampus of D-galactose-induced aging in SD rats. The rats were allocated to the following groups: (1) control; (2) swimming exercise; (3) induced-aging by injecting D-galactose; (4) induced-aging rats with swimming exercise. The longevity-related AMPK/SIRT1/PGC-1α signaling pathway and brain IGF1/PI3K/Akt survival pathway were significantly reduced in D-galactose-induced aging group compared to non-aging control group and increased after exercise training. The inflammation pathway markers were over-expressed in induced-aging hippocampus, exercise significantly inhibited the inflammatory signaling activity. Fas-dependent and mitochondrial-dependent apoptotic pathways were significantly increased in the induced-aging group relative to the control group whereas they were decreased in the aging-exercise group. This study demonstrated that swimming exercise not only reduced aging-induced brain apoptosis and inflammatory signaling activity, but also enhanced the survival pathways in the hippocampus, which provides one of the new beneficial effects for exercise training in aging brain.


Asunto(s)
Envejecimiento/fisiología , Hipocampo/fisiología , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Transducción de Señal , Natación/fisiología , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Envejecimiento/patología , Animales , Apoptosis , Caspasas/metabolismo , Ciclooxigenasa 2/metabolismo , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Expresión Génica , Hipocampo/patología , Inflamación/metabolismo , Masculino , FN-kappa B/metabolismo , Neuronas/patología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosfatidilinositol 3-Quinasa/genética , Fosfatidilinositol 3-Quinasa/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Receptor IGF Tipo 1/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Proteína bcl-X/metabolismo
7.
J Mol Cell Cardiol ; 122: 58-68, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30098987

RESUMEN

Mitochondrial dysfunction is a major contributor to myocyte loss and the development of heart failure. Myocytes have quality control mechanisms to retain functional mitochondria by removing damaged mitochondria via specialized autophagy, i.e., mitophagy. The underlying mechanisms of fission affect the survival of cardiomyocytes, and left ventricular function in the heart is poorly understood. Here, we demonstrated the direct effect and potential mechanisms of mitochondrial functional defects associated with abnormal mitochondrial dynamics in heart failure. We observed that IGF-IIR signaling produced significant changes in mitochondrial morphology and function; such changes were associated with the altered expression and distribution of dynamin-related protein (Drp1) and mitofusin (Mfn2). IGF-IIR signaled extracellular signal-regulated kinase (ERK) activation to promote Drp1 phosphorylation and translocation to mitochondria for mitochondrial fission and mitochondrial dysfunction. Moreover, IGF-IIR signaling triggered Rab9-dependent autophagosome formation by the JNK-mediated phosphorylation of Bcl-2 at serine 87 and promoted ULK1/Beclin 1-dependent autophagic membrane formation. Excessive mitochondrial fission by Drp1 enhanced the Rab9-dependent autophagosome recognition and engulfing of damaged mitochondria and eventually decreased cardiomyocyte viability. Therefore, these results demonstrated the connection between Rab9-dependent autophagosomes and mitochondrial fission in cardiac myocytes, which provides a potential therapeutic strategy for treating heart disease.


Asunto(s)
Dinaminas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Receptor IGF Tipo 2/metabolismo , Análisis de Varianza , Animales , Autofagosomas/metabolismo , Autofagia , Línea Celular , Femenino , Sistema de Señalización de MAP Quinasas , Dinámicas Mitocondriales , Mitofagia , Miocitos Cardíacos/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas de Unión al GTP rab/metabolismo
8.
J Cell Physiol ; 233(9): 7080-7091, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29574782

RESUMEN

Mitochondria dysfunction is the major characteristic of mitophagy, which is essential in mitochondrial quality control. However, excessive mitophagy contributes to cell death in a number of diseases, including ischemic stroke and hepatotoxicity. Insulin-like growth factor II (IGF-II) and its receptor (IGF-IIR) play vital roles in the development of heart failure during hypertension. We found that IGF-II triggers IGF-IIR receptor activation, causing mitochondria dysfunction, resulting in mitophagy, and cardiomyocyte cell death. These results indicated that IGF-IIR activation triggers mitochondria fragmentation, leading to autophagosome formation, and loss of mitochondria content. These results are associated with Parkin-dependent mitophagy. Additionally, autophagic proteins Atg5, and Atg7 deficiency did not suppress IGF-IIR-induced mitophagy. However, Rab9 knockdown reduced mitophagy and maintained mitochondrial function. These constitutive mitophagies through IGF-IIR activation trigger mitochondria loss and mitochondrial ROS accumulation for cardiomyocyte viability decrease. Together, our results indicate that IGF-IIR predominantly induces mitophagy through the Rab9-dependent alternative autophagy.


Asunto(s)
Autofagia , Mitocondrias/metabolismo , Mitofagia , Receptor IGF Tipo 2/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Animales , Animales Recién Nacidos , Comunicación Autocrina , Autofagosomas/metabolismo , Autofagosomas/ultraestructura , Dependovirus/metabolismo , Femenino , Corazón/fisiopatología , Humanos , Factor II del Crecimiento Similar a la Insulina/metabolismo , Mitocondrias/ultraestructura , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/ultraestructura , Especificidad de Órganos , Comunicación Paracrina , Ratas Sprague-Dawley , Ubiquitina-Proteína Ligasas/metabolismo
9.
Cell Death Dis ; 8(8): e2990, 2017 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-28796250

RESUMEN

Hypertension-induced cardiac hypertrophy and attenuated cardiac function are the major characteristics of early stage heart failure. Cardiomyocyte death in pathological cardiac conditions is the primary cause of heart failure and mortality. Our previous studies found that heat shock factor 1 (HSF1) protected cardiomyocytes from death by suppressing the IGF-IIR signaling pathway, which is critical for hypertensive angiotensin II-induced cardiomyocyte apoptosis. However, the role of heat shock factor 2 (HSF2) in hypertension-induced cardiac hypertrophy is unknown. We identified HSF2 as a miR-18 target for cardiac hypertrophy. p53 activation in angiotensin II (ANG II)-stimulated NRVMs is responsible for miR-18 downregulation both in vitro and in vivo, which triggers HSF2 expression and the activation of IGF-IIR-induced cardiomyocyte hypertrophy. Finally, we provide genetic evidence that miR-18 is required for cardiomyocyte functions in the heart based on the gene transfer of cardiac-specific miR-18 via adenovirus-associated virus 2 (AAV2). Transgenic overexpression of miR-18 in cardiomyocytes is sufficient to protect against dilated cardiomyopathy during hypertension-induced heart failure. Our results demonstrated that the p53-miR-18-HSF2-IGF-IIR axis was a critical regulatory pathway of cardiomyocyte hypertrophy in vitro and in vivo, suggesting that miR-18 could be a therapeutic target for the control of cardiac functions and the alleviation of cardiomyopathy during hypertension-induced heart failure.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Proteínas de Choque Térmico/genética , Hipertrofia/metabolismo , MicroARNs/genética , Miocitos Cardíacos/metabolismo , Receptor IGF Tipo 2/metabolismo , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Células Cultivadas , Femenino , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/genética , Proteínas de Choque Térmico/metabolismo , Hipertensión/complicaciones , Hipertensión/metabolismo , Ratones , MicroARNs/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Sprague-Dawley , Receptor IGF Tipo 2/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
10.
Environ Toxicol ; 32(3): 754-763, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27098997

RESUMEN

Severe and potentially fatal hypotension and cardiac contractile dysfunction are common symptoms in patients with sepsis. LPS was previously found to dramatically upregulate expression of fibrosis-related factors FGF-2, uPA, MMP-2, and MMP-9 in primary cardiac fibroblasts. MMPs are capable of denaturing and degrading fibrillar collagens and other components of the extracellular matrix (ECM). Studies have shown that dysregulation of expression of MMPs is associated with development of myocardial extracellular matrix remodeling and cardiac fibrosis, which contribute to progression of heart failure. In this study, H9c2 cells and cardiac fibroblasts were divided into five treatment groups: control, LPS (1 µg/mL) and three concentrations of FCEtOH (Carthami Flos ethanolic extract) (31.25, 62.5, and 125 µg/mL). Phosphorylation of ERK-1/2 was observed to be rapidly induced upon treatment with LPS. In contrast, it was significantly suppressed by the administration of FCEtOH (125 µg/mL). Effects of FCEtOH on LPS-induced MMP-2 and MMP-9 expression in H9c2 cells occurred directly through ERK1/2 were determined. H9c2 cells were therefore pretreated with EGF-R to activate ERK pathway. Both protein levels of MMP-2 and MMP-9 and immunefluorescent signals of MMP-9 were significantly enhanced by EGFR. In contrast, MMP-2 and MMP-9 were significantly reduced after FCEtOH administration. Based on these findings, the authors concluded that FCEtOH elicits a protective effect against LPS-induced cardio-fibrosis through the ERK1/2 pathway. Carthamus tinctorius L may potentially serve as a cardio-protective agent against LPS- induced cardiac fibrosis. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 754-763, 2017.


Asunto(s)
Carthamus tinctorius/química , Lipopolisacáridos/toxicidad , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Extractos Vegetales/farmacología , Regulación hacia Arriba/efectos de los fármacos , Animales , Carthamus tinctorius/metabolismo , Células Cultivadas , Regulación hacia Abajo/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Microscopía Fluorescente , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Fosforilación/efectos de los fármacos , Extractos Vegetales/química , Ratas , Ratas Sprague-Dawley , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
11.
Growth Factors ; 34(3-4): 73-86, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27366871

RESUMEN

The HIF-1α transcriptional factor and the BH-3 only protein BNIP3 are known to play fundamental roles in response to hypoxia. The objective of this research is to investigate the molecular mechanisms and the correlation of HIF-1α, BNIP3 and IGFBP-3 in hypoxia-induced cardiomyocytes injuries. Heart-derived H9c2 cells and neonatal rat ventricular myocytes (NRVMs) were incubated in normoxic or hypoxic conditions. Hypoxia increased HIF-1α expression and activated the downstream BNIP3 and IGFBP-3 thereby triggered mitochondria-dependent apoptosis. Moreover, IGF1R/PI3K/Akt signaling was attenuated by HIF-1α-dependent IGFBP-3 expression to enhance hypoxia-induced apoptosis. Autophagy suppression with 3-methyladenine or siATG5 or siBeclin-1 significantly decreased myocardial apoptosis under hypoxia. Knockdown of FoxO3a or BNIP3 significantly abrogated hypoxia-induced autophagy and mitochondria-dependent apoptosis. Moreover, prolonged-hypoxia induced HIF-1α stimulated BNIP3 and enhanced IGFBP-3 activation to inhibit IGF1R/PI3K/Akt survival pathway and mediate mitochondria-dependent cardiomyocyte apoptosis. HIF-1α and FoxO3a blockage are sufficient to annul the change of excessive hypoxia of hearts.


Asunto(s)
Apoptosis , Autofagia , Proteína Forkhead Box O3/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Hipoxia de la Célula , Línea Celular , Células Cultivadas , Femenino , Proteína Forkhead Box O3/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Sprague-Dawley , Transducción de Señal
12.
Environ Toxicol ; 31(7): 769-81, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25727812

RESUMEN

Consumption of deep sea minerals (DSM), such as magnesium, calcium, and potassium, is known to reduce hypercholesterolemia-induced myocardial hypertrophy and cardiac-apoptosis and provide protection against cardiovascular diseases. Heart diseases develop as a lethal complication among diabetic patients usually due to hyperglycemia-induced cardiac-apoptosis that causes severe cardiac-damages, heart failure, and reduced life expectancy. In this study, we investigated the potential of DSM and its related cardio-protection to increase the life expectancy in diabetic rats. In this study, a heart failure rat model was developed by using streptozotocin (65 mg kg(-1) ) IP injection. Different doses of DSM-1× (37 mg kg(-1) day(-1) ), 2× (74 mg kg(-1) day(-1) ) and 3× (111 mg kg(-1) day(-1) ), were administered to the rats through gavages for 4 weeks. The positive effects of DSM on the survival rate of diabetes rats were determined with respect to the corresponding effects of MgSO4 . Further, to understand the mechanism by which DSM enhances the survival of diabetic rats, their potential to regulate cardiac-apoptosis and control cardiac-dysfunction were examined. Echocardiogram, tissue staining, TUNEL assay, and Western blotting assay were used to investigate modulations in the myocardial contractile function and related signaling protein expression. The results showed that DSM regulate apoptosis and complement the cardiomyocyte proliferation by enhancing survival mechanisms. Moreover DSM significantly reduced the mortality rate and enhanced the survival rate of diabetic rats. Experimental results show that DSM administration can be an effective strategy to improve the life expectancy of diabetic subjects by improving cardiac-cell proliferation and by controlling cardiac-apoptosis and associated cardiac-dysfunction. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 769-781, 2016.


Asunto(s)
Apoptosis/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Longevidad/efectos de los fármacos , Minerales/uso terapéutico , Agua de Mar/química , Animales , Glucemia/metabolismo , Proliferación Celular/efectos de los fármacos , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Ecocardiografía , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/prevención & control , Sulfato de Magnesio/farmacología , Masculino , Minerales/química , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Análisis de Supervivencia
13.
Chin J Physiol ; 58(4): 244-53, 2015 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-26211648

RESUMEN

Magnesium sulfate (MgSO4) ameliorates hypoxia/ischemia-induced neuronal apoptosis in a rat model. This study aimed to investigate the mechanisms governing the anti-apoptotic effect of MgSO4 on cobalt chloride (CoCl2)-exposed NB41A3 mouse neuroblastoma cells. MgSO4 increased the viability of NB41A3 cells treated with CoCl2 in a dose-dependent manner. MgSO4 treatment was shown to lead to an increase in the anti-apoptotic Bcl-2 family proteins, with a concomitant decrease in the pro-apoptotic proteins. MgSO4 also attenuated the CoCl2-induced disruption of mitochondrial membrane potential (ΔΨ(m)) and reduced the release of cytochrome c form the mitochondria to the cytosol. Furthermore, exposure to CoCl2 caused activation of the hypoxia-inducible factor 1α (HIF-1α). On the other hand, MgSO4 markedly reduced CoCl2-induced HIF-1α activation and suppressed HIF-1α downstream protein BNIP3. MgSO4 treatment induced ERK1/2 activation and attenuated CoCl2-induced activation of p38 and JNK. Addition of the ERK1/2 inhibitor U0126 significantly reduced the ability of MgSO4 to protect neurons from CoCl2-induced mitochondrial apoptotic events. However, incubation of cultures with the p38 and JNK inhibitors did not significantly affect MgSO4-mediated neuroprotection. MgSO4 appears to suppress CoCl2-induced NB41A3 cell death by activating ERK1/2/ MAPK pathways, which further modulates the role of Bcl-2 family proteins and mitochondria in NB41A3 cells. Our data suggest that MgSO4 may act as a survival factor that preserves mitochondrial integrity and inhibits apoptotic pathways.


Asunto(s)
Apoptosis/efectos de los fármacos , Cobalto/farmacología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Sulfato de Magnesio/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Línea Celular Tumoral , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Especies Reactivas de Oxígeno/metabolismo
14.
Cell Physiol Biochem ; 36(1): 274-84, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25967966

RESUMEN

BACKGROUND/AIMS: The risk of heart disease is higher in males than in females. However, this advantage of females declines with increasing age, presumably a consequence of decreased estrogen secretion and malfunctioning of the estrogen receptor. We previously demonstrated that 17ß-estradiol (E2) prevents cardiomyocyte hypertrophy, autophagy and apoptosis via estrogen receptor α (ERα), but the effects of ERß on myocardial injury remained elusive. The present paper thus, investigated the cardioprotective effects of estrogen (E2) and ERß against hypoxia-induced cell death. METHODS: Transient transfection of Tet-On ERß gene construct was used to overexpress ERß in hypoxia-treated H9c2 cardiomyoblast cells. RESULTS: Our data revealed that IGF1R, Akt phosphorylation and Bcl-2 expression are enhanced by ERß in H9c2 cells. Moreover, ERß overexpression reduced accumulation of hypoxia-related proteins, autophagy-related proteins and mitochondria-apoptotic proteins and enhanced the protein levels of Bcl-2, pAkt and Bad under hypoxic condition. In neonatal rat ventricular myocytes (NRVMs), we observed that hypoxia induced cell apoptosis as measured by TUNEL staining, and E2 and/or ERß could totally abolish hypoxia-induced apoptosis. The suppressive effects of E2 and/or ERß in hypoxia-treated NRVMs were totally reversed by ER antagonist, ICI. Taken together, E2 and/or ERß exert the protective effect through repressed hypoxia-inducible HIF-1α, BNIP3 and IGFBP-3 levels to restrain the hypoxia-induced autophagy and apoptosis effects in H9c2 cardiomyoblast cells. CONCLUSION: The results suggest that females probably could tolerate better prolonged hypoxia condition than males, and E2/ERß treatment could be a potential therapy to prevent hypoxia-induced heart damage."


Asunto(s)
Estradiol/farmacología , Receptor beta de Estrógeno/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Receptor beta de Estrógeno/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Miocitos Cardíacos/citología , Ratas , Ratas Sprague-Dawley
15.
Cell Physiol Biochem ; 36(1): 334-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25967972

RESUMEN

BACKGROUND: Hemorrhagic shock (HS) is the major cause of death from trauma. Hemorrhagic shock may lead to cellular hypoxia and organ damage. Our previous findings showed that HS induced a cardiac apoptosis pathway and synergistically caused myocardial cell damage in diabetic rats under trauma-induced HS. Tetramethylpyrazine (TMP) is a major biologically active ingredient purified from the rhizome of Ligusticum wallichii (called Chuang Xiong in Chinese). Chuan Xiong rescued cells from synergistic cardiomyoblast cell injury under high-glucose (HG) conditions plus hypoxia. TMP is one of the most important active ingredients that elevated the survival rate in ischemic brain injury and prevented inducible NO synthase expression to have anti-inflammatory effects against cell damage in different cell types. METHOD: Here, we further investigate whether TMP can protect against hypoxic (<1% oxygen) conditions in H9c2 cardiomyoblast cells for 24 hrs. RESULTS: Our results showed that hypoxia mediated through HIF-1α/JNK/p38 activation significantly elevated the levels of the hypoxia-related proteins HIF-1α, BNIP3 and IGFBP3, further enhanced the pro-apoptotic protein Bak and upregulated downstream Caspase 9 and 3, resulting in cell death. All of these phenomena were fully recovered under TMP treatment. We observed that TMP exerted this effect by activating the IGF1 receptor survival pathway, dependent primarily on PI3K/Akt. When PI3K (class I) was blocked by specific siRNA, the hypoxia-induced activated caspase 3 and cell apoptosis could not be reversed by TMP treatment. CONCLUSION: Our results strongly suggest that TMP could be used to restore hypoxia-induced myocardial cell apoptosis and cardiac hypoxic damage.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Mioblastos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Pirazinas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Supervivencia Celular , Células Cultivadas , Mioblastos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Ratas
16.
Chin J Physiol ; 58(1): 38-45, 2015 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-25687490

RESUMEN

Cirrhotic cardiomyopathy (CCM) is a common cardiac dysfunction in patients waiting for orthotopic liver transplantation (OLT). Carbon tetrachloride (CCl4) intraperitoneal (IP) injection has been reported as successful in a cirrhosis-induced CCM model. In this work, we used the same assay for CCM induction using CCl4 (0.2 mg/kg) IP injection twice per day for 14 days during the cardiac protection drugs treatment process. The cardiac protection drugs were silymarin (100 mg/kg/day), baicalein (30 mg/kg/day), San Huang Shel Shin Tang (SHSST, 30 mg/kg/day) and ß-cyclodextrin modified SHSST (SHSSTc, 30 mg/kg/day and 300 mg/kg/day). After 4 weeks of treatment, the SHSSTc cardiac protection effects were determined through activation of the IGF1R cell survival pathway and inhibition of Fas-FADD death domain induced-apoptosis. SHSSTc cardiac protection was enhanced through ß-cyclodextrin modification, which increased bio-availability, and displayed stronger protective effects than silymarin and baicalein, both of which are well-known liver protection drugs. Thus, SHSSTc might provide the best therapeutic benefit in CCM treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Cardiomiopatías/tratamiento farmacológico , Cardiotónicos/farmacología , Ciclodextrinas/farmacología , Corazón/efectos de los fármacos , Cirrosis Hepática Experimental/complicaciones , Animales , Tetracloruro de Carbono , Cardiomiopatías/patología , Flavanonas/farmacología , Ratas , Ratas Sprague-Dawley , Silimarina/farmacología
17.
Cell Transplant ; 23(4-5): 513-29, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24816448

RESUMEN

Ischemic heart damage usually triggers cardiomyopathological remodeling and fibrosis, thus promoting the development of heart functional failure. Mesenchymal stem cells (MSCs) are a heterogeneous group of cells in culture, with multipotent and hypoimmunogenic characters to aid tissue repair and avoid immune responses, respectively. Numerous experimental findings have proven the feasibility, safety, and efficiency of MSC therapy for cardiac regeneration. Despite that the exact mechanism remains unclear, the therapeutic ability of MSCs to treat ischemia heart diseases has been tested in phase I/II clinical trials. Based on encouraging preliminary findings, MSCs might become a potentially efficacious tool in the therapeutic options available to treat ischemic and nonischemic cardiovascular disorders. The molecular mechanism behind the efficacy of MSCs on promoting engraftment and accelerating the speed of heart functional recovery is still waiting for clarification. It is hypothesized that cardiomyocyte regeneration, paracrine mechanisms for cardiac repair, optimization of the niche for cell survival, and cardiac remodeling by inflammatory control are involved in the interaction between MSCs and the damaged myocardial environment. This review focuses on recent experimental and clinical findings related to cellular cardiomyoplasticity. We focus on MSCs, highlighting their roles in cardiac tissue repair, transdifferentiation, the MSC niche in myocardial tissues, discuss their therapeutic efficacy that has been tested for cardiac therapy, and the current bottleneck of MSC-based cardiac therapies.


Asunto(s)
Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Infarto del Miocardio/terapia , Animales , Diferenciación Celular , Corazón/fisiología , Humanos , Células Madre Mesenquimatosas/metabolismo , Comunicación Paracrina , Regeneración , Nicho de Células Madre
18.
Chin J Physiol ; 57(3): 111-20, 2014 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-24826779

RESUMEN

Obesity is often associated with the development of cardiac hypertrophy but the hypertrophy-related pathways in obesity remain unknown. The purpose of this study was to evaluate cardiac hypertrophy-related markers, atrial natriuretic peptide (ANP), B-type natriuretic peptide (BNP), tumor necrosis factor-alpha (TNFα) and hypertrophy-related pathways, interleukin (IL)-6-STAT3, IL-6-MEK5-ERK5 and calcineurin-nuclear factor of activated T-cells (NFAT)3 in the excised hearts from obese rats. Twelve obese Zucker rats were studied at 5-6 months of age and twelve age-matched lean Zucker rats served as the control group. The cardiac characteristics, myocardial architecture, ANP, BNP, TNFα levels, IL-6, STAT3, p-STAT3, MEK5, ERK-5, p-ERK-5, calcineurin and NFAT3 in the left ventricle from the rats were measured by heart weight index, echocardiography, vertical cross section, histological analysis, reverse transcription polymerase chain reaction and western blotting. Compared with the lean control, the whole heart weight, the left ventricule weight, the ratio of the whole heart weight to tibia length, echocardiographic interventricular septum, left ventricular posterior wall thickness, myocardial morphological changes and systolic blood pressure were found to increase in the obese rats. The protein levels of ANP, BNP, TNFα, IL-6, STAT3, p-STAT3, MEK-5, ERK-5, p-ERK 5, calcineurin and NFAT3 were also significantly increased in the hearts of the obese rats. The results showed that the hypertrophy-related markers, ANP, BNP and TNFα, the hypertrophy-related pathways IL-6-STAT3 and IL-6-MEK5-ERK5, and the calcineurin-NFAT3 hypertrophy-related pathways were more active in obese Zucker rats, which may provide possible hypertrophic mechanisms for developing cardiac hypertrophy and pathological changes in obesity.


Asunto(s)
Cardiomegalia/complicaciones , Cardiomegalia/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Obesidad Mórbida/complicaciones , Obesidad Mórbida/metabolismo , Animales , Factor Natriurético Atrial/metabolismo , Cardiomegalia/genética , Modelos Animales de Enfermedad , Interleucina-6/metabolismo , MAP Quinasa Quinasa 5/metabolismo , Masculino , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Péptido Natriurético Encefálico/metabolismo , Obesidad Mórbida/genética , ARN Mensajero/metabolismo , Ratas Zucker , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
19.
Chem Res Toxicol ; 26(5): 654-61, 2013 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-23682761

RESUMEN

Recent studies have found that cigarette smoke is epidemiologically linked to an increased risk for impaired cognitive development in adolescents. This study evaluated the influence of side stream smoke (SSS) exposure on hippocampal apoptosis and of the lumbrokinase (LK) effects on SSS induced apoptosis in young hamster hippocampus. Twenty male hamsters at six weeks of age were randomly divided into control group, SSS group (exposed to tobacco cigarettes smoke at doses of 10 cigarettes for 30 min twice a day for 1 month), and SSS hamsters with LK treatment (1.2 mg/kg, ip) for twice a week for 1 month. TUNEL assay and Western blotting were performed. The TUNEL-positive apoptotic cells, as well as Fas-dependent activity and mitochondria-dependent apoptotic pathways, such as Fas, FADD, activated caspase-8, t-Bid, activated caspase-9, and activated caspase-3, were significantly increased in the SSS-exposed hippocampus compared to the control and highly attenuated in the LK treatment group. Additionally, SSS exposure significantly increased the autophagy marker proteins, Beclin-1, ATG7, and LC3-II levels, in the hippocampus compared to those in the control group and obviously attenuated after LK treatment. LK also reduced hippocampus injury by enhancing eNOS expression and remarkably inhibited the proinflammatory NFκB/iNOS/COX-2 signaling activity. We found that the detrimental effects of SSS on the hippocampus are truly mediated by cell apoptosis and autophagy. However, LK reduced the hippocampus apoptosis and autophagy related injuries induced by SSS in a widespread manner. We suggest that LK presents protective effects on hippocampus apoptosis and has therapeutic potential against abnormal hippocampal function.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Endopeptidasas/metabolismo , Hipocampo/efectos de los fármacos , Nicotiana/efectos adversos , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Transducción de Señal/efectos de los fármacos , Humo/efectos adversos , Animales , Cricetinae , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Hipocampo/metabolismo , Hipocampo/patología , Masculino , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Relación Estructura-Actividad
20.
Cell Transplant ; 22(4): 711-21, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23190451

RESUMEN

Adult stem cells have been proven to possess tremendous potential in the treatment of hematological disorders, possibly in transplantation. Mesenchymal stem cells (MSCs) are a heterogeneous group of cells in culture, with hypoimmunogenic character to avoid alloreactive T-cell recognition as well as inhibition of T-cell proliferation. Numerous experimental findings have shown that MSCs also possess the ability to promote engraftment of donor cells and to accelerate the speed of hematological recovery. Despite that the exact mechanism remains unclear, the therapeutic ability of MSCs on hematologic transplantation have been tested in preclinical trials. Based on encouraging preliminary findings, MSCs might become a potentially efficacious tool in the therapeutic options available to treat and cure hematological malignancies and nonmalignant disorders. The molecular mechanisms behind the real efficacy of MSCs on promoting engraftment and accelerating hematological recovery are awaiting clarification. It is hypothesized that direct cell-to-cell contact, paracrine factors, extracellular matrix scaffold, BM homing capability, and endogenous metabolites of immunologic and nonimmunologic elements are involved in the interactions between MSCs and HSCs. This review focuses on recent experimental and clinical findings related to MSCs, highlighting their roles in promoting engraftment, hematopoietic recovery, and GvHD/graft rejection prevention after HSCT, discussing the potential clinical applications of MSC-based treatment strategies in the context of hematological transplantation.


Asunto(s)
Neoplasias Hematológicas/terapia , Trasplante de Células Madre Hematopoyéticas , Células Madre Mesenquimatosas/citología , Animales , Hematopoyesis , Humanos , Inmunomodulación , Nicho de Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA