Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Mol Pharmacol ; 92(3): 188-192, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28404616

RESUMEN

Pharmacology is the science that investigates the interactions between organisms and drugs and their mechanisms. Pharmacology plays a translational role in modern medicine, bridging basic research and the clinic. With its economy booming, China has invested an enormous amount of financial and human resources in pharmacological research in the recent decade. As a result, major breakthroughs have been achieved in both basic and clinical research, with the discovery of many potential drug targets and biomarkers that has made a sizable contribution to the overall advancement of pharmacological sciences. In this article, we review recent research efforts and representative scientific achievements and discuss future challenges and directions for the pharmacological sciences in China.


Asunto(s)
Farmacología , Animales , China , Descubrimiento de Drogas , Humanos , Investigación
2.
Glia ; 63(12): 2208-19, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26200696

RESUMEN

Oxidative stress plays an important role in the progression of Alzheimer's disease (AD) and other neurodegenerative conditions. Glutathione (GSH), the major antioxidant in the central nervous system, is primarily synthesized and released by astrocytes. We determined if ß-amyloid (Aß42), crucially involved in Alzheimer's disease, affected GSH release. Monomeric Aß (mAß) stimulated GSH release from cultured cortical astrocytes more effectively than oligomeric Aß (oAß) or fibrillary Aß (fAß). Monomeric Aß increased the expression of the transporter ABCC1 (also referred to as MRP1) that is the main pathway for GSH release. GSH release from astrocytes, with or without mAß stimulation, was reduced by pharmacological inhibition of ABCC1. Astrocytes robustly express connexin proteins, especially connexin43 (Cx43), and mAß also stimulated Cx43 hemichannel-mediated glutamate and GSH release. Aß-stimulation facilitated hemichannel opening in the presence of normal extracellular calcium by reducing astrocyte cholesterol level. Aß treatment did not alter the intracellular concentration of reduced or oxidized glutathione. Using a mouse model of AD with early onset Aß deposition (5xFAD), we found that cortical ABCC1 was significantly increased in temporal register with the surge of Aß levels in these mice. ABCC1 levels remained elevated from 1.5 to 3.5 months of age in 5xFAD mice, before plunging to subcontrol levels when amyloid plaques appeared. Similarly, in cultured astrocytes, prolonged incubation with aggregated Aß, but not mAß, reduced induction of ABCC1 expression. These results support the hypothesis that in the early stage of AD pathogenesis, less aggregated Aß increases GSH release from astrocytes (via ABCC1 transporters and Cx43 hemichannels) providing temporary protection from oxidative stress which promotes AD development.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Glutatión/metabolismo , Envejecimiento/metabolismo , Enfermedad de Alzheimer/metabolismo , Animales , Astrocitos/efectos de los fármacos , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/metabolismo , Colesterol/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Oxidación-Reducción , Placa Amiloide/metabolismo
3.
J Neurochem ; 133(1): 38-52, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25661995

RESUMEN

Alzheimer's disease (AD) is characterized by early impairments in memory and progressive neurodegeneration. Disruption of synaptic plasticity processes that underlie learning and memory contribute partly to this pathophysiology. Tripchlorolide (T4 ), an extract from a traditional Chinese herbal Tripterygium wilfordii Hook F, has been shown to be neuroprotective in animal models of Parkinson's disease and to improve cognitive deficits in senescence-accelerated mouse P8. In this study, we investigated the effect of T4 on cognitive decline and synaptic plasticity in five times familial AD (5XFAD) mice co-expressing mutated amyloid precursor protein and presenilin-1. Five-month-old 5XFAD mice and wild type littermates were intraperitoneally injected with T4 , 5 µg/kg or 25 µg/kg, every other day for 60 days. T4 treatment significantly improved spatial learning and memory, alleviated synaptic ultrastructure degradation, up-regulated expression of synapse-related proteins, including synaptophysin, post-synaptic density-95, N-methyl-D-aspartate receptor subunit 1, phosphorylation of calcium/calmodulin dependent protein kinase II α, and phosphorylation of cyclic AMP-response element binding protein, and promoted activation of the phophoinositide-3-kinase-Akt-mammalian target of rapamycin signaling pathway in 5XFAD mice. Accumulation of amyloid ß (Aß) may contribute to synapse dysfunction and memory impairment in AD. We found that T4 treatment significantly reduced cerebral Aß deposits and lowered Aß levels in brain homogenates. These effects coincided with a reduction in cleavage of ß-carboxyl-terminal amyloid precursor protein (APP) fragment, levels of soluble APPß, and protein expression of ß-site APP cleaving enzyme 1. Taken together, our findings identify T4 as a potent negative regulator of brain Aß levels and show that it significantly ameliorates synaptic degeneration and cognitive deficits in a mouse model of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Cognición/efectos de los fármacos , Diterpenos/uso terapéutico , Inmunosupresores/uso terapéutico , Fenantrenos/uso terapéutico , Sinapsis/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Animales , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Ratones Transgénicos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Serina-Treonina Quinasas TOR/metabolismo
4.
Am Surg ; 90(11): 3102-3112, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38835213

RESUMEN

BACKGROUND: This study aimed to evaluate the effectiveness and safety of 2D laparoscopy vs 3D laparoscopy for the treatment of colorectal cancer. METHODS: A literature search was conducted through PubMed, Web of Science, and Embase from their inception to January 2024. Studies investigating different outcomes of colorectal surgery were included. Results are presented as odds ratios (ORs) or mean differences (MDs) with 95% confidence intervals (CIs). The protocol for this review has been registered on PROSPERO (CRD42024504902). RESULTS: A total of 10 publications were retrieved in this article. The 3D group is associated with a significant improvement in intraoperative blood loss (MD = -8.04, 95% CI = -14.18 to -1.89, P = 0.01, I2 = 55%), operative time (MD = -17.33, 95% CI = -29.15 to -5.51, P = 0.004, I2 = 90%), and postoperative hospital stay (MD = -0.23, 95% CI = -0.43 to -0.04, P = 0.02, I2 = 48%) compared to that of patients treated in the 2D group, particularly for rectal cancer patients above three results (MD = -10.36, 95% CI = -15.00 to -5.73, P < 0.001, I2 = 0%), (MD = -18.85, 95% CI = -34.88 to -2.82, P = 0.02, I2 = 57%), and (MD = -0.93, 95% CI = -1.53 to -0.34, P = 0.002, I2 = 0%), respectively. There was no significant statistical difference in the time of pass flatus (MD = -0.14, 95% CI = -0.49 to 0.21, P = 0.44, I2 = 79%) and the number of dissected lymph nodes (MD = 0.36, 95% CI = -0.49 to 1.21, P = 0.41, I2 = 45%), but the 3D group had an earlier postoperative pass flatus for rectal cancer patients (MD = -0.46, 95% CI = -0.66 to -0.27, P<0.001, I2 = 0%) and the more number of dissected lymph nodes for colon cancer patients (MD = 1.54, 95% CI = 0.05 to 3.03, P = 0.04, I2 = 69%) than the 2D group. There was no significant difference in postoperative overall complication (OR = 0.94, 95% CI = 0.67 to 1.31, P = 0.71, I2 = 0%) and anastomotic leakage (OR = 0.93, 95% CI = 0.48 to 1.80, P = 0.83, I2 = 0%) in the two groups, regardless of rectal cancer and colon surgery patients. CONCLUSION: This meta-analysis demonstrates that 3D laparoscopy could reduce the amount of blood loss, accelerate postoperative pass flatus, and shorten the operation time and postoperative hospital stay over 2D for radical rectal cancer surgery, without obvious advantage for radical colon cancer surgery. Moreover, 3D laparoscopy increases the number of dissected lymph nodes for radical colon cancer surgery but may not be observed in rectal cancer surgery.


Asunto(s)
Neoplasias Colorrectales , Laparoscopía , Humanos , Laparoscopía/métodos , Neoplasias Colorrectales/cirugía , Tiempo de Internación/estadística & datos numéricos , Tempo Operativo , Pérdida de Sangre Quirúrgica/estadística & datos numéricos , Resultado del Tratamiento , Complicaciones Posoperatorias/epidemiología
6.
Mar Drugs ; 11(4): 1019-34, 2013 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-23538867

RESUMEN

OBJECTIVE: To investigate astaxanthin (ATX) neuroprotection, and its mechanism, on a 1-methyl-4-phenyl-pyridine ion (MPP+)-induced cell model of Parkinson's disease. METHODS: Mature, differentiated PC12 cells treated with MPP+ were used as an in vitro cell model. The MTT assay was used to investigate cell viability after ATX treatment, and western blot analysis was used to observe Sp1 (activated transcription factor 1) and NR1 (NMDA receptor subunit 1) protein expression, real-time PCR was used to monitor Sp1 and NR1 mRNA, and cell immunofluorescence was used to determine the location of Sp1 and NR1 protein and the nuclear translocation of Sp1. RESULTS: PC12 cell viability was significantly reduced by MPP+ treatment. The expression of Sp1 and NR1 mRNA and protein were increased compared with the control (p < 0.01). Following co-treatment with ATX and MPP+, cell viability was significantly increased, and Sp1 and NR1 mRNA and protein were decreased, compared with the MPP+ groups (p < 0.01). In addition, mithracycin A protected PC12 cells from oxidative stress caused by MPP+ by specifically inhibiting the expression of Sp1. Moreover, cell immunofluorescence revealed that ATX could suppress Sp1 nuclear transfer. CONCLUSION: ATX inhibited oxidative stress induced by MPP+ in PC12 cells, via the SP1/NR1 signaling pathway.


Asunto(s)
Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/tratamiento farmacológico , 1-Metil-4-fenilpiridinio/toxicidad , Animales , Western Blotting , Técnica del Anticuerpo Fluorescente , Células PC12 , Enfermedad de Parkinson/fisiopatología , Plicamicina/análogos & derivados , Plicamicina/farmacología , ARN Mensajero/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Xantófilas/farmacología
7.
Glia ; 60(4): 559-69, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22228589

RESUMEN

The ε4 allele of the Apolipoprotein E (APOE) gene is the strongest genetic risk factor for late-onset Alzheimer's disease (AD), and affects clinical outcomes of chronic and acute brain damages. The mechanisms by which apoE affect diverse diseases and disorders may involve modulation of the glial response to various types of brain damage. We examined glial activation in a mouse model where each of the human APOE alleles are expressed under the endogenous mouse APOE promoter, as well as in APOE knock-out mice. APOE4 mice displayed increased glial activation in response to intracerebroventricular lipopolysaccharide (LPS) compared to APOE2 and APOE3 mice by several measures. There were higher levels of microglia/macrophage, astrocytes, and invading T-cells after LPS injection in APOE4 mice. APOE4 mice also displayed greater and more prolonged increases of cytokines (IL-1ß, IL-6, TNF-α) than APOE2 and APOE3 mice. We found that APOE4 mice had greater synaptic protein loss after LPS injection, as measured by three markers: PSD-95, drebin, and synaptophysin. In all assays, APOE knock-out mice responded similar to APOE4 mice, suggesting that the apoE4 protein may lack anti-inflammatory characteristics of apoE2 and apoE3. Together, these findings demonstrate that APOE4 predisposes to inflammation, which could contribute to its association with Alzheimer's disease and other disorders.


Asunto(s)
Encéfalo/citología , Regulación de la Expresión Génica/genética , Neuroglía/fisiología , Sinapsis/patología , Análisis de Varianza , Animales , Antígenos de Diferenciación/metabolismo , Apolipoproteína E2/genética , Apolipoproteína E3/genética , Apolipoproteína E4/genética , Apolipoproteínas E/deficiencia , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Citocinas/metabolismo , Homólogo 4 de la Proteína Discs Large , Ensayo de Inmunoadsorción Enzimática , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Genotipo , Proteína Ácida Fibrilar de la Glía/metabolismo , Guanilato-Quinasas/metabolismo , Humanos , Inyecciones Intraventriculares , Lipopolisacáridos/farmacología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Neuroglía/efectos de los fármacos , Sinapsis/metabolismo
8.
BMC Neurosci ; 13: 156, 2012 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-23272707

RESUMEN

BACKGROUND: Although the etiology of PD remains unclear, increasing evidence has shown that oxidative stress plays an important role in its pathogenesis and that of other neurodegenerative disorders. NOX2, a cytochrome subunit of NOX, transports electrons across the plasma membrane to generate ROS, leading to physiological and pathological processes. Heme oxygenase-1 (HO-1) can be rapidly induced by oxidative stress and other noxious stimuli in the brain or other tissues. Astaxanthin (ATX), a carotenoid with antioxidant properties, is 100-1000 times more effective than vitamin E. The present study investigated the neuroprotective effects of ATX on MPP(+)-induced oxidative stress in PC12 cells. RESULTS: MPP(+) significantly decreased MTT levels in a concentration-dependent manner. Hemin, SnPPIX and ATX didn't exhibit any cytotoxic effects on PC12 cells. Pretreatment with ATX (5, 10, 20 µM), caused intracellular ROS production in the MPP(+) group to decrease by 13.06%, 22.13%, and 27.86%, respectively. MPP(+) increased NOX2, NRF2 and HO-1 protein expression compared with control (p < 0.05). Co-treatment with hemin or ATX suppressed NOX2 expression (p < 0.01), and greatly increased NRF2 and HO-1 expression (p < 0.01). MPP(+) treatment up-regulated both NOX2 (p < 0.01) and HO-1 (p < 0.01) mRNA levels. Co-treatment with hemin or ATX significantly increased HO-1 mRNA levels (p < 0.01), and decreased NOX2 mRNA levels (p < 0.01). MPP(+) increased NOX2 and HO-1 expression with considerable fluorescence extending out from the perinuclear region toward the periphery; this was attenuated by DPI. Co-treatment with hemin or ATX significantly up-regulated HO-1 expression and decreased NOX2 expression with considerable fluorescence intensity (stronger than the control and MPP(+) groups). CONCLUSIONS: ATX suppresses MPP(+)-induced oxidative stress in PC12 cells via the HO-1/NOX2 axis. ATX should be strongly considered as a potential neuroprotectant and adjuvant therapy for patients with Parkinson's disease.


Asunto(s)
1-Metil-4-fenilpiridinio/antagonistas & inhibidores , Antioxidantes/farmacología , Hemo-Oxigenasa 1/biosíntesis , Glicoproteínas de Membrana/biosíntesis , NADPH Oxidasas/biosíntesis , Estrés Oxidativo/efectos de los fármacos , Catecolaminas/farmacología , Supervivencia Celular/efectos de los fármacos , Agonistas de Dopamina/farmacología , Interacciones Farmacológicas , Regulación de la Expresión Génica/efectos de los fármacos , Hemina/farmacología , Imidazolinas/farmacología , NADPH Oxidasa 2 , Factor 2 Relacionado con NF-E2/biosíntesis , Fármacos Neuroprotectores/farmacología , Células PC12 , Especies Reactivas de Oxígeno/metabolismo , Xantófilas/farmacología
9.
Acta Pharmacol Sin ; 33(4): 438-44, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22407229

RESUMEN

AIM: To investigate the effect of ginsenoside Rb1 on voltage-gated calcium currents in cultured rat hippocampal neurons and the modulatory mechanism. METHODS: Cultured hippocampal neurons were prepared from Sprague Dawley rat embryos. Whole-cell configuration of the patch-clamp technique was used to record the voltage-gated calcium currents (VGCCs) from the hippocampal neurons,and the effect of Rb1 was examined. RESULTS: Rb1 (2-100 µmol/L) inhibited VGCCs in a concentration-dependent manner, and the current was mostly recovered upon wash-out. The specific L-type Ca(2+) channel inhibitor nifedipine (10 µmol/L) occluded Rb1-induced inhibition on VGCCs. Neither the selective N-type Ca(2+) channel blocker ω-conotoxin-GVIA (1 µmol/L), nor the selective P/Q-type Ca(2+) channel blocker ω-agatoxin IVA (30 nmol/L) diminished Rb1-sensitive VGCCs. Rb1 induced a leftward shift of the steady-state inactivation curve of I(Ca) to a negative potential without affecting its activation kinetics or reversal potential in the I-V curve. The inhibitory effect of Rb1 was neither abolished by the adenylyl cyclase activator forskolin (10 µmol/L), nor by the PKA inhibitor H-89 (10 µmol/L). CONCLUSION: Ginsenoside Rb1 selectively inhibits the activity of L-type voltage-gated calcium channels, without affecting the N-type or P/Q-type Ca(2+) channels in hippocampal neurons. cAMP-PKA signaling pathway is not involved in this effect.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Ginsenósidos/farmacología , Hipocampo/citología , Neuronas/efectos de los fármacos , Animales , Células Cultivadas , Neuronas/metabolismo , Panax/química , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley
10.
BMC Complement Altern Med ; 12: 82, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22742579

RESUMEN

BACKGROUND: Parkinson's disease is a high incidence neurodegenerative disease in elderly people, and oxidative stress plays an important role in the pathogenesis. Oxygen metabolism in the brain is high, which lacks an antioxidative protection mechanism. Recently, it has been found that polyphenols play an important role in antioxidation. (-)-epigallocatechin-3-gallate (EGCG) is an important component of tea polyphenols and its biological effects, such as strong antioxidation, scavenging of free radicals and anti-apoptosis, can pass through the blood brain barrier. The SIRT1/PGC-1α signaling pathway has not been reported in PC12 cells. Therefore, research of the protective mechanism of EGCG in PC12 cells damaged by -methyl-4-phenyl-pyridine (MMP+) may provide a new insight into protect against and treatment of Parkinson's disease. METHODS: MPP(+)-treated highly differentiated PC12 cells were used as the in vitro cell model. An MTT assay was used to investigate cell viability after EGCG treatment, a dichlorofluorescin diacetate assay was used to measure reactive oxygen species (ROS) production, western blot analysis was used to observe PGC-1α and SIRT1 protein expression, and real-time PCR to observe PGC-1α, SOD1 and GPX1 mRNA expression. RESULTS: PC12 cell viability was significantly reduced after MPP(+) treatment by 11.46% compared with that of the control (P < 0.05). However, cell viability was unchanged by 10 µmol/L EGCG treatment. In co-treatments with EGCG and MPP(+), cell viability was significantly increased by 12.92% (P < 0.05) and MPP(+)-induced ROS production was markedly decreased. PGC-1α mRNA expression was obviously upregulated by 21.51% (P < 0.05), and SOD1 and GPX1 mRNA expression was slightly increased by 12.94% and 15.63% (P > 0.05), respectively, by treatment with EGCG and then MPP(+) for 12 h. The mRNA expression of PGC-1α, SOD1 and GPX1 was increased by 25.17%, 40% and 146% (all P < 0.05), respectively, by treatment with EGCG and then MPP(+) for 24 h. Such effects were not observed with MPP(+) treatment alone. CONCLUSION: The SIRT1/PGC-1α pathway is one of the mechanisms of EGCG suppression of MPP(+)-induced injury of PC12 cells.


Asunto(s)
Antioxidantes/farmacología , Camellia sinensis/química , Catequina/análogos & derivados , Supervivencia Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Fitoterapia , 1-Metil-4-fenilpiridinio , Animales , Antioxidantes/metabolismo , Antioxidantes/uso terapéutico , Catequina/farmacología , Catequina/uso terapéutico , Glutatión Peroxidasa/metabolismo , Células PC12 , Enfermedad de Parkinson/tratamiento farmacológico , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Sirtuina 1/metabolismo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Té/química , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Glutatión Peroxidasa GPX1
11.
Zhonghua Yi Xue Za Zhi ; 92(5): 330-5, 2012 Feb 07.
Artículo en Zh | MEDLINE | ID: mdl-22490838

RESUMEN

OBJECTIVE: To explore whether or not ginsenoside Rg1 can modify the metabolism of amyloid precursor protein (APP) and the generation of amyloid beta (Aß) by nuclear factor-kappa B (NF-κB). METHODS: N2a/APP695 cells, a mutated APP-overexpressing neuronal cell line, was used to mimic the APP metabolism and Aß generation in vitro. The BACE1 mRNA and protein levels were detected by RT-PCR (reverse transcription-polymerase chain reaction) and Western blot respectively. Then the expression levels and subcellular localization of NF-κB were detected by Western blot and confocal laser scanning microscope respectively. RESULTS: The treatment of ginsenoside Rg1 at a dose of 2.5 µmol/L decreased the levels of Aß1-40 and Aß1-42 (13.3 ± 4.3) ng/ml vs (12.0 ± 5.4) ng/ml in N2a/APP695 cells, decreased the protein level of BACE1 (BACE1/ß-actin 0.26 ± 0.05), increased the protein level of NF-κB p65 (p-p65/p65 0.93 ± 0.02) and resulted in the translocation of NF-κB from cytoplasm to nucleus. Quinazoline inhibited the activation of NF-κB with a reduction of p-p65 and p-p65/p65 in N2a/APP695 cells and increased the BACE1 protein level. And the treatment of ginsenoside Rg1 showed similar changes in N2a/APP695 cells when compared with the treatment of quinazoline alone. CONCLUSION: Ginsenoside Rg1 may modify the metabolism of APP by enhancing the nuclear binding of NF-κB to BACE1 promoter and inhibiting the transcription and translation of BACE1.


Asunto(s)
Ginsenósidos/farmacología , FN-kappa B/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Línea Celular Tumoral , Ratones , Neuroblastoma , Regiones Promotoras Genéticas
12.
Yao Xue Xue Bao ; 46(9): 1065-71, 2011 Sep.
Artículo en Zh | MEDLINE | ID: mdl-22121776

RESUMEN

This study is to observe the effect of ilexonin A (IA) on the expression of basic fibroblast growth factor (bFGF) and growth associated protein-43 (GAP-43), and neurogenesis after cerebral ischemia-reperfusion in rats and explore its possible mechanism of protecting neuronal injury. Models of middle cerebral artery occlusion (MCAO) were established in SD rats. Before and after two hours ischemia-reperfusion, IA (20 and 40 mg x kg(-1)) was injected immediately and on 3, 7, 14, and 28 d once a day. The neurological severity was evaluated by neurological severity scores (NSS); neuronal injury in the boundary zone of the infarction area was evaluated by TUNEL and Niss1 staining. The expressions of bFGF and GAP-43 and neurogenesis were evaluated by Western blotting and 5-bromodeoxyuridine (Brdu) fluorescence staining, respectively. After treatment with IA, the NSS of treatment groups were lower than that of the models (3 and 7 d). The number of TUNEL positive neurons decreased and Nissl positive neurons increased at the same time (3 d). The expressions of bFGF and GAP-43 increased significantly in the boundary zone of the infarction area when compared to model group. Moreover, IA markedly enhanced the neurogenesis in the brain after ischemia-reperfusion, which revealed an increase of Brdu/NeuN positive cells in the boundary zone of the infarction area. The possible mechanism of protecting neuronal injury of IA may be related to inhibition on neuronal apoptosis, upregulation of bFGF and GAP-43, and neurogenesis in boundary zone of infarction after cerebral ischemia-reperfusion.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/metabolismo , Proteína GAP-43/metabolismo , Neurogénesis/efectos de los fármacos , Daño por Reperfusión/metabolismo , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/etiología , Bromodesoxiuridina/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Compuestos Orgánicos/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/etiología
13.
Yao Xue Xue Bao ; 45(7): 853-9, 2010 Jul.
Artículo en Zh | MEDLINE | ID: mdl-20931782

RESUMEN

This study is to explore whether the Wnt/beta-catenin signaling pathway is involved in the process of tripchlorolide (T4) protecting against oligomeric Abeta(1-42)-induced neuronal apoptosis. Primary cultured cortical neurons were used for the experiments on day 6 or 7. The oligomeric Abeta(1-42) (5 micromol x L(-1) for 24 h) was applied to induce neuronal apoptosis. Prior to treatment with Abeta(1-42) for 24 h, the cultured neurons were pre-incubated with T4 (2.5, 10, and 40 nmol x L(-1)), Wnt3a (Wnt signaling agonists) and Dkk1 (inhibitors) for indicated time. Then the cell viability, neuronal apoptosis, and protein levels of Wnt, glycogen synthase kinase 3beta (GSK3beta), beta-catenin and phospho-beta-catenin were measured by MTT assay, TUNEL staining and Western blotting, respectively. The result demonstrated that oligomeric Abeta(1-42) induced apoptotic neuronal cell death in a time- and dose-dependent manner. Pretreatment with T4 significantly increased the neuronal cell survival and attenuated neuronal apoptosis. Moreover, oligomeric Abeta(1-42)-induced phosphorylation of beta-catenin and GSK3beta was markedly inhibited by T4. Additionally, T4 stabilized cytoplasmic beta-catenin. These results indicate that tripchlorolide protects against the neurotoxicity of Abeta by regulating Wnt/beta-catenin signaling pathway. This may provide insight into the clinical application of tripchlorolide to Alzheimer's disease.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Apoptosis/efectos de los fármacos , Diterpenos/farmacología , Fragmentos de Péptidos/toxicidad , Fenantrenos/farmacología , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/citología , Diterpenos/aislamiento & purificación , Femenino , Feto , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Neuronas/citología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/aislamiento & purificación , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/antagonistas & inhibidores , Fenantrenos/aislamiento & purificación , Fosforilación , Plantas Medicinales/química , Embarazo , Ratas , Ratas Sprague-Dawley , Tripterygium/química
14.
Glia ; 57(11): 1227-38, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19170180

RESUMEN

Recent research has focused on soluble oligomeric assemblies of beta-amyloid peptides (Abeta) as the proximate cause of neuroinflammation, synaptic loss, and the eventual dementia associated with Alzheimer's disease (AD). In this study, tripchlorolide (T4), an extract of Tripterygium wilfordii Hook. F (TWHF), was studied as a novel agent to suppress neuroinflammatory process in microglial cells and to protect neuronal cells against microglia-mediated oligomeric Abeta toxicity. T4 significantly attenuated oligomeric Abeta(1-42)-induced release of inflammatory productions such as tumor necrosis factor-alpha, interleukin-1beta, nitric oxide (NO), and prostaglandin E2. It also downregulated the protein levels of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) in microglial cells. Further molecular mechanism study demonstrated that T4 inhibited the nuclear translocation of nuclear factor-kappaB (NF-kappaB) without affecting I-kappaBalpha phosphorylation. It repressed Abeta-induced JNK phosphorylation but not ERK or p38 MAPK. The inhibition of NF-kappaB and JNK by T4 is correlated with the suppression of inflammatory mediators in Abeta-stimulated microglial cells. These results suggest that T4 protects neuronal cells by blocking inflammatory responses of microglial cells to oligomeric Abeta(1-42) and that T4 acts on the signaling of NF-kappaB and JNK, which are involved in the modulation of inflammatory response. Therefore, T4 may be an effective agent in modulating neuroinflammatory process in AD.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Diterpenos/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Microglía/efectos de los fármacos , Subunidad p50 de NF-kappa B/metabolismo , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Fenantrenos/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/fisiología , Línea Celular , Células Cultivadas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía/inmunología , Microglía/fisiología , Neuroinmunomodulación/efectos de los fármacos , Neuronas/fisiología
15.
J Gerontol A Biol Sci Med Sci ; 63(3): 253-64, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18375874

RESUMEN

Tert-butyl hydroperoxide (t-BHP), an analog of hydroperoxide, induced characteristic changes of senescence in human diploid fibroblasts WI-38 cells. It was reported that ginsenoside Rg1, an active ingredient of ginseng, ameliorated learning deficits in aged rats. The present study was aimed to investigate whether ginsenoside Rg1 can delay the premature senescence of WI-38 cells induced by t-BHP and to explore the underlying molecular mechanisms. First, Rg1 pretreatment markedly reversed senescent morphological changes in WI-38 cells induced by t-BHP. Second, t-BHP treatment alone resulted in an increase in the protein levels of P16 and P21, and a decline in intracellular adenosine 5'-triphosphate (ATP) level and mitochondrial complex IV activity. Ginsenoside Rg1 pretreatment had significant effects of attenuating these changes. These data indicate that ginsenoside Rg1 has an anti-aging effect on t-BHP-induced premature senescence in WI-38 cells. This effect may be mediated by regulating cell cycle proteins and enhancing mitochondrial functioning.


Asunto(s)
Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Ginsenósidos/farmacología , terc-Butilhidroperóxido/farmacología , Adenosina Trifosfato/metabolismo , Western Blotting , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Diploidia , Relación Dosis-Respuesta a Droga , Transporte de Electrón/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Fibroblastos/citología , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Ginsenósidos/química , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/ultraestructura , Microscopía Electrónica de Transmisión , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Estructura Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/genética , Telomerasa/metabolismo
16.
Brain Res ; 1200: 99-106, 2008 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-18289510

RESUMEN

Increasing evidence have shown that beta-amyloid (Abeta) induced hyperphosphorylation of tau, which eventually resulted in the disruption of microtubule (MT) integrity. Cyclin-dependent kinase 5 (CDK5) and its activator p35 are required for neurite outgrowth. The cleavage of p35 to p25, mediated by calpain and calcium, caused CDK5 dislocation and subsequently p25/CDK5-induced tau hyperphosphorylation, which disrupted the cytoskeleton and resulted in neuronal death. In the present study we investigated the effects of ginsenoside Rb1 on fibrillar Abeta(25-35)-induced tau hyperphosphorylation in primary cultured cortical neurons and also the potential involvement of Ca(2+)-calpain-CDK5 signal pathway. The present study suggests that Ca(2+), calpain, and p25 in CDK5 pathway may play important roles in Abeta(25-35)-induced tau hyperphosphorylation.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Calpaína/efectos de los fármacos , Ginsenósidos/farmacología , Proteínas del Tejido Nervioso/efectos de los fármacos , Neuronas/efectos de los fármacos , Fragmentos de Péptidos/antagonistas & inhibidores , Proteínas tau/efectos de los fármacos , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Animales , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Calpaína/metabolismo , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Citoesqueleto/efectos de los fármacos , Citoesqueleto/metabolismo , Citoesqueleto/patología , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Ginsenósidos/uso terapéutico , Microtúbulos/efectos de los fármacos , Microtúbulos/metabolismo , Microtúbulos/patología , Proteínas del Tejido Nervioso/metabolismo , Ovillos Neurofibrilares/efectos de los fármacos , Ovillos Neurofibrilares/metabolismo , Ovillos Neurofibrilares/patología , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/toxicidad , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas tau/metabolismo
17.
Neurochem Int ; 50(4): 671-80, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17291629

RESUMEN

We previously reported that injection of bacterial lipopolysaccharide (LPS) into gravid female rats at embryonic day 10.5 resulted in a birth of offspring with fewer than normal dopamine (DA) neurons along with innate immunity dysfunction and many characteristics seen in Parkinson's disease (PD) patients. The LPS-exposed animals were also more susceptible to secondary toxin exposure as indicated by an accelerated DA neuron loss. Glutathione (GSH) is an important antioxidant in the brain. A disturbance in glutathione homeostasis has been proposed for the pathogenesis of PD. In this study, animals prenatally exposed to LPS were studied along with an acute intranigral LPS injection model for the status of glutathione homeostasis, lipid peroxidation, and related enzyme activities. Both prenatal LPS exposure and acute LPS injection produced a significant GSH reduction and increase in oxidized GSH (GSSG) and lipid peroxide (LPO) production. Activity of gamma-glutamylcysteine synthetase (GCS), the rate-limiting enzyme in de novo GSH synthesis, was up-regulated in acute supranigral LPS model but was reduced in the prenatal LPS model. The GCS light subunit protein expression was also down-regulated in prenatal LPS model. GSH redox recycling enzyme activities (glutathione peroxidase, GPx and glutathione reducdase, GR) and glutathione-S-transferase (GST), gamma-glutamyl transpeptidase (gamma-GT) activities were all increased in prenatal LPS model. Prenatal LPS exposure and aging synergized in GSH level and GSH-related enzyme activities except for those (GR, GST, and gamma-GT) with significant regional variations. Additionally, prenatal LPS exposure produced a reduction of DA neuron count in the substantia nigra (SN). These results suggest that prenatal LPS exposure may cause glutathione homeostasis disturbance in offspring brain and render DA neurons susceptible to the secondary neurotoxin insult.


Asunto(s)
Encefalitis/metabolismo , Glutatión/metabolismo , Degeneración Nerviosa/metabolismo , Estrés Oxidativo/fisiología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Sustancia Negra/metabolismo , Envejecimiento/metabolismo , Animales , Animales Recién Nacidos , Causalidad , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Dopamina/metabolismo , Encefalitis/inducido químicamente , Encefalitis/fisiopatología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Femenino , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Mediadores de Inflamación/toxicidad , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/fisiología , Lipopolisacáridos/toxicidad , Masculino , Degeneración Nerviosa/inducido químicamente , Degeneración Nerviosa/fisiopatología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Enfermedad de Parkinson/fisiopatología , Embarazo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Ratas , Ratas Sprague-Dawley , Sustancia Negra/efectos de los fármacos , Sustancia Negra/fisiopatología
19.
Mol Neurobiol ; 54(3): 1967-1977, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-26910813

RESUMEN

Alzheimer's disease (AD) is the most common dementia and the trigger of its pathological cascade is widely believed to be the overproduction and accumulation of ß-amyloid protein (Aß) in the affected brain. However, effective AD remedies are still anxiously awaited. Recent evidence suggests that curcumin may be a potential agent for AD treatment. In this study, we used 5×FAD transgenic mice as an AD model to investigate the effects of curcumin on AD. Our results showed that curcumin administration (150 or 300 mg/kg/day, intragastrically, for 60 days) dramatically reduced Aß production by downregulating BACE1 expression, preventing synaptic degradation, and improving spatial learning and memory impairment of 5×FAD mice. These findings suggest that curcumin is a potential candidate for AD treatment.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/biosíntesis , Curcumina/uso terapéutico , Trastornos de la Memoria/metabolismo , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Ácido Aspártico Endopeptidasas/genética , Curcumina/farmacología , Expresión Génica , Masculino , Trastornos de la Memoria/tratamiento farmacológico , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
Brain Res ; 1090(1): 35-44, 2006 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-16647047

RESUMEN

The most reliable and robust risk factor for some neurodegenerative diseases is aging. It has been proposed that processes of aging are associated with the generation of reactive oxygen species and a disturbance of glutathione homeostasis in the brain. Yet, aged animals have rarely been used to model the diseases that are considered to be age-related such as Parkinson's or Alzheimer's disease. This suggests that the results from these studies would be more valuable if aged animals were used. The present study was designed to provide insight into the glutathione redox state in young and aged rat siblings of both genders by studying the enzyme activities related to glutathione synthesis, cycling, and usage. The results suggested a significant age-related reduction of reduced glutathione (GSH) level in all brain regions examined, associated with an increase of GSH oxidation to glutathione disulfide (GSSG) and decrease of the GSH/GSSG ratio. These changes were accompanied by diminished gamma-glutamylcysteine synthetase activity in de novo glutathione synthesis and increased lipid peroxidation. In addition, these changes were associated with increased enzyme activities related to the GSH usage (glutathione peroxidase, gamma-glutamyl transpeptidase, and glutathione S-transferase). The results indicate that aged animals are likely more vulnerable to oxidative stress and insinuate the roles of aged animals in modeling age-related neurodegeneration diseases.


Asunto(s)
Envejecimiento/fisiología , Química Encefálica/fisiología , Encéfalo/enzimología , Enzimas/metabolismo , Glutatión/metabolismo , Estrés Oxidativo/fisiología , Animales , Regulación hacia Abajo/fisiología , Femenino , Glutamato-Cisteína Ligasa/metabolismo , Glutatión Peroxidasa/metabolismo , Glutatión Transferasa/metabolismo , Masculino , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/fisiopatología , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Caracteres Sexuales , gamma-Glutamiltransferasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA