Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Neurosci ; 52(12): 4890-4907, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32638449

RESUMO

The cellular and molecular mechanisms underlying leptin-mediated brain protection against cerebral ischemia were investigated at the blood-brain barrier (BBB) and neutrophil level. Through the ischemia/reperfusion (I/R) animal model, we found that leptin expression level was significantly decreased in ischemic hemisphere. Brain injection with leptin (15 µg/kg, intracisternally) could block the I/R-increased BBB permeability, activation of matrix metallopeptidase 9 (MMP-9) and brain infiltration of blood-borne neutrophils to reduce the infarct volume of ischemic brain. The brain expression level of tight junction protein ZO-1 as well as number and motility of neutrophils in blood was all increased by the same injection, indicating BBB stability (rather than reduction in neutrophils) played a major role in the leptin-inhibited brain infiltration of neutrophils. Leptin-mediated protection of BBB was further confirmed in vitro, through a BBB cellular model under the in vitro ischemic condition (G/R: glucose-oxygen-serum deprivation followed by GOS restoration). The results showed that leptin again could block the G/R-increased neutrophil adherence to EC layer as well as BBB permeability, likely by stimulating the endothelial expression of ZO-1 and VE-Cadherin. The study has demonstrated that leptin could protect ischemic brain via multiple ways (other than neuronal protection), by inhibiting the BBB permeability, brain infiltration of the blood-borne neutrophils and neutrophil adherence to vascular ECs. The role of leptin in vascular biology of stroke could further support its therapeutic potential in other neurodegenerative diseases, associated with BBB disorder.


Assuntos
Isquemia Encefálica , Traumatismo por Reperfusão , Animais , Barreira Hematoencefálica , Isquemia Encefálica/tratamento farmacológico , Infarto , Leptina , Neutrófilos , Ratos , Ratos Sprague-Dawley , Reperfusão , Traumatismo por Reperfusão/tratamento farmacológico
2.
Cell Mol Neurobiol ; 39(5): 591-604, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30852719

RESUMO

It is known that cerebral ischemia can cause brain inflammation and adiposome can serve as a depot of inflammatory mediators. In the study, the pro-inflammatory and pro-death role of adiposome in ischemic microglia and ischemic brain was newly investigated. The contribution of PPARγ to adiposome formation was also evaluated for the first time in ischemic microglia. Focal cerebral ischemia/reperfusion (I/R) animal model and the in vitro glucose-oxygen-serum deprivation (GOSD) cell model were both applied in the study. GOSD- or I/R-induced adiposome formation, inflammatory activity, cell death of microglia, and brain infarction were, respectively, determined, in the absence or presence of NS-398 (adiposome inhibitor) or GW9662 (PPARγ antagonist). GOSD-increased adiposome formation played a critical role in stimulating the inflammatory activity (production of TNF-α and IL-1ß) and cell death of microglia. Similar results were also found in ischemic brain tissues. GOSD-induced PPARγ partially contributed to the increase of adiposomes and adiposome-mediated inflammatory responses of microglia. Blockade of adiposome formation with NS-398 or GW9662 significantly reduced not only the inflammatory activity and death rate of GOSD-treated microglia but also the brain infarct volume and motor function deficit of ischemic rats. The pathological role of microglia-derived adiposome in cerebral ischemia has been confirmed and attributed to its pro-inflammatory and/or pro-death effect upon ischemic brain cells and tissues. Adiposome and its upstream regulator PPARγ were therefore as potential targets for the treatment of ischemic stroke. Therapeutic values of NS-398 and GW9662 have been suggested.


Assuntos
Isquemia Encefálica/terapia , Gotículas Lipídicas/metabolismo , Microglia/metabolismo , Acidente Vascular Cerebral/terapia , Anilidas/farmacologia , Animais , Animais Recém-Nascidos , Isquemia Encefálica/complicações , Morte Celular , Meios de Cultura Livres de Soro , Ciclo-Oxigenase 2/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glucose/deficiência , Inflamação/patologia , Interleucina-1beta/metabolismo , Masculino , Microglia/patologia , Atividade Motora/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Nitrobenzenos/farmacologia , Oxigênio , PPAR gama/metabolismo , Ratos Sprague-Dawley , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/terapia , Acidente Vascular Cerebral/complicações , Sulfonamidas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
3.
Chin J Physiol ; 59(6): 331-347, 2016 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-27817195

RESUMO

Lung resistance-related protein (LRP) is a human major vault protein (MVP) implicated in drug resistance of cancer cells in a cell-type dependent manner. The primary goal of the study was to understand the role(s) of LRP in doxorubicin (DOX) resistance of non-small cell lung cancer (NSCLC) cells and the underlying working mechanisms. In the study, the roles of LRP in the regulation of DOX dynamics, nuclear import of minor vault proteins (vault poly (ADP-ribose) polymerase, vPARP and telomerase associated protein-1, TEP-1) and DOX-mediated cytotoxicity were examined in CH27 and H460 cells. Our results were the first to show that the CH27 cells with higher LRP expression levels were more resistant to DOX-induced cytotoxicity as shown in apoptosis experiments. LRP at the nuclear membrane could regulate DOX efflux from the nucleus to the cytosol, and also the reverse vPARP/TEP1 influx from the cytosol, to protect NSCLC cells from DOX-induced apoptosis. Cytosolic LRP could bind to DOX, vPARP and TEP1 to clear DOX away from the nucleus and promote the assembly of vaults for cell protection again. Based on the data obtained, the molecular mechanisms responsible for DOX resistance of NSCLC were delineated to demonstrate that LRP, vPARP and TEP1 were potential targets for NSCLC therapy. Inhibitors of these proteins, including small interfering LRP (siLRP), wheat-germ agglutenin (WGA) (WGA), 3-aminobenzamide (3-AB) and 3,6,9-trisubstituted acridine 9-[4-(N,N-dimethylamino) phenylamino]-3,6-bis(3-pyrrolodinopropionamido) acridine (BRACO-19), break down the DOX resistance of NSCLC cells, particularly in CH27 cells, and may have therapeutic values in the control of NSCLC.


Assuntos
Antibióticos Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Doxorrubicina , Resistencia a Medicamentos Antineoplásicos/fisiologia , Partículas de Ribonucleoproteínas em Forma de Abóbada/metabolismo , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Humanos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas de Ligação a RNA
4.
Cell Oncol (Dordr) ; 39(5): 411-433, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27306525

RESUMO

BACKGROUND: Non-small cell lung cancers (NSCLCs) frequently exhibit resistance to therapeutic drugs, which seriously hampers their treatment. Here, we set out to assess the roles of the multidrug resistance protein 1 (MRP1) and P-glycoprotein (P-gp) in the doxorubicin (DOX) resistance of NSCLC cells, as well as the putative therapeutic efficacy of MRP1 and P-gp blockers on DOX-treated NSCLC cells. METHODS: The impact of DOX on cell survival, DOX efflux and MRP1 and P-gp expression was assessed in 5 different NSCLC-derived cell lines (parental CH27, A549, H1299, H460, and DOX resistant CH27) in the absence or presence of MK571 (MRP1 inhibitor) or Verapamil (P-gp inhibitor), under both normoxic and hypoxic conditions. RESULTS: We found that in response to DOX treatment, NSCLC cells that express high levels of MRP1 and P-gp (such as CH27) showed a better DOX efflux and a higher DOX resistance. MK571 and Verapamil were found to abolish DOX resistance and to act as chemosensitizers for DOX therapy in all cell lines tested. We also found that hypoxia could inhibit MRP1 and P-gp expression in a HIF-1α-dependent manner, abolish DOX resistance and boost the chemosensitizer effect of MK571 and Verapamil on DOX treatment of all the NSCLC cells tested, except the DOX-resistant CH27 cells. CONCLUSIONS: From our data we conclude that MRP1 and P-gp play critical roles in the DOX resistance of the NSCLC cells tested. MRP1 and P-gp targeted therapy using MK571, Verapamil, CoCl2 or ambient hypoxia appeared to be promising in abolishing the DOX efflux and DOX resistance of the NSCLC cells. The putative therapeutic efficacies of MRP1 and/or P-gp blockers on NSCLC cells are worthy of note.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Pulmonares/patologia , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Antineoplásicos/farmacologia , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Resistência a Múltiplos Medicamentos/fisiologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Citometria de Fluxo , Humanos , Neoplasias Pulmonares/metabolismo , Propionatos/farmacologia , Quinolinas/farmacologia , Verapamil/farmacologia
5.
PLoS One ; 11(1): e0146692, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26745377

RESUMO

The protective value of neuron-derived conditioned medium (NCM) in cerebral ischemia and the underlying mechanism(s) responsible for NCM-mediated brain protection against cerebral ischemia were investigated in the study. NCM was first collected from the neuronal culture growing under the in vitro ischemic condition (glucose-, oxygen- and serum-deprivation or GOSD) for 2, 4 or 6 h. Through the focal cerebral ischemia (bilateral CCAO/unilateral MCAO) animal model, we discovered that ischemia/reperfusion (I/R)-induced brain infarction was significantly reduced by NCM, given directly into the cistern magna at the end of 90 min of CCAO/MCAO. Immunoblocking and chemical blocking strategies were applied in the in vitro ischemic studies to show that NCM supplement could protect microglia, astrocytes and neurons from GOSD-induced cell death, in a growth factor (TGFß1, NT-3 and GDNF) and p-ERK dependent manner. Brain injection with TGFß1, NT3, GDNF and ERK agonist (DADS) alone or in combination, therefore also significantly decreased the infarct volume of ischemic brain. Moreover, NCM could inhibit ROS but stimulate IL-1ß release from GOSD-treated microglia and limit the infiltration of IL-ß-positive microglia into the core area of ischemic brain, revealing the anti-oxidant and anti-inflammatory activities of NCM. In overall, NCM-mediated brain protection against cerebral ischemia has been demonstrated for the first time in S.D. rats, due to its anti-apoptotic, anti-oxidant and potentially anti-glutamate activities (NCM-induced IL-1ß can inhibit the glutamate-mediated neurotoxicity) and restriction upon the infiltration of inflammatory microglia into the core area of ischemic brain. The therapeutic potentials of NCM, TGFß1, GDNF, NT-3 and DADS in the control of cerebral ischemia in human therefore have been suggested and require further investigation.


Assuntos
Infarto da Artéria Cerebral Média/tratamento farmacológico , Neurônios/metabolismo , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/prevenção & controle , Animais , Apoptose , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/patologia , Movimento Celular , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Avaliação Pré-Clínica de Medicamentos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/fisiologia , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Microglia/fisiologia , Fatores de Crescimento Neural/fisiologia , Estresse Oxidativo , Cultura Primária de Células , Fatores de Proteção , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fator de Crescimento Transformador beta1/fisiologia
6.
Clin Exp Metastasis ; 31(8): 881-95, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25168821

RESUMO

Signaling pathway(s) responsible for transforming growth factor ß (TGFß)-induced epithelial mesenchymal transition (EMT), invasion and migration of H460 cells (non-small cell lung cancer/NSCLC) was identified in the study. The results showed that TGFß-induced p(38)/ß-catenin/PPARγ signaling pathway played a critical role in the promotion of EMT, invasion and migration of H460 cells. All these pathological outcomes attributed to PPARγ-increased expression of p-EGFR, p-c-MET and Vimentin and the decrease of E-cadherin. Transforming growth factor ß and p(38)-induced ß-catenin not only stimulated the expression of PPARγ but also physically interacted with it. Blocking the ligand binding domain of PPARγ (with GW9662) could significantly interfere the binding between PPARγ and ß-catenin, and interrupt the nuclear infiltration of both factors. These findings suggested that ß-catenin was an upstream regulator and a ligand of PPARγ, and the binding between these two molecules was critical for their nuclear infiltration. Transforming growth factor ß-induced tumor invasion and migration was also seen in U373 cells (brain glioma, with high inducible PPARγ) in a PPARγ-dependent manner, but not in CH27 cells (squamous NSCLC, with low PPARγ). PPARγ shRNA, GW9662, JW67 and 2,4-diaminoquinazoline were all revealed to have important values in the control of the intrinsic and TGFß-induced EMT, tumor invasion and migration of H460 cells. The results further suggested that PPARγ and ß-catenin may be the potential markers for the early diagnosis and/or treatment of metastatic tumors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Transição Epitelial-Mesenquimal , Neoplasias Pulmonares/patologia , PPAR gama/metabolismo , Fator de Crescimento Transformador beta/farmacologia , beta Catenina/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Adesão Celular , Ciclo Celular , Movimento Celular , Proliferação de Células , Imunofluorescência , Humanos , Imunoprecipitação , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , PPAR gama/antagonistas & inibidores , PPAR gama/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Células Tumorais Cultivadas
7.
Food Chem Toxicol ; 71: 197-206, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24994563

RESUMO

Lauryl gallate (LG) is a gallic acid derivative that has been widely used as an antioxidant food additive. In this study, we examined the anticancer effects of LG on the human acute myeloid leukemia (AML) HL60 and KG-1 cells. Our results showed that LG inhibited cell proliferation in a concentration- and time-dependent manner in both HL60 and KG-1 cells. The IC50s of LG in HL60 and KG-1 cells were 3.5 and 8.0 µM, respectively. Treatment with LG increased the proportions of annexin V-stained and sub-G1-phase HL60 and KG-1 cells. Moreover, activation of both extrinsic and intrinsic apoptotic pathways was involved in LG-induced AML cell apoptosis, accompanied by dissipation of mitochondrial membrane potential, downregulation of anti-apoptotic proteins (Bcl-2, Mcl-1, and Bcl-xL), upregulation of pro-apoptotic proteins (Bak, PUMA, DR4, and DR5), and increased caspase-2, -3, -8, and -9 activation. Our results also indicated that LG could induce monocytic differentiation in both HL60 and KG-1 cells, confirmed by morphological changes, nitroblue tetrazolium reduction assays, nonspecific esterase assays, and increased CD14 expression. After blocking LG-induced ERK and Sp1 expression using the ERK-specific inhibitor PD98059, monocytic differentiation in both HL60 and KG-1 cells decreased, suggesting that LG-induced differentiation proceeded through an ERK/Sp1 signaling axis.


Assuntos
Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Ácido Gálico/análogos & derivados , Leucemia Mieloide Aguda/patologia , Ácido Gálico/farmacologia , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Proteínas de Neoplasias/metabolismo
8.
Life Sci ; 102(1): 55-64, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24631138

RESUMO

AIMS: The activation of hepatic stellate cells (HSCs) in response to liver injury is critical to the development of liver fibrosis, thus, the blockage of the activation of HSCs is considered as a rational approach for anti-fibrotic treatment. In this report, we investigated the effects and the underlying mechanisms of gallic acid (GA) in interfering with the activation of HSCs. MAIN METHODS: The primary cultured rat HSCs were treated with various doses of GA for different time intervals. The morphology, viability, caspase activity, calcium ion flux, calpain I activity, reactive oxygen species generation and lysosomal functions were then investigated. KEY FINDINGS: GA selectively killed HSCs in both dose- and time-dependent manners, while remained no harm to hepatocytes. Besides, caspases were not involved in GA-induced cell death of HSCs. Further results showed that GA toxicity was associated with a rapid burst of reactive oxygen species (ROS) and a subsequent increase of intracellular Ca(2+) and calpain activity. Addition of calpain I but not calpain II inhibitor rescued HSCs from GA-induced death. In parallel, pretreatment with antioxidants or an intracellular Ca(2+) chelator eradicated GA responses, implying that GA-mediated cytotoxicity was dependent on its pro-oxidative properties and its effect on Ca(2+) flux. Furthermore, application of ROS scavengers also reversed Ca(2+) release and the disruption of lysosomal membranes in GA-treated HSCs. SIGNIFICANCE: These results provide evidence for the first time that GA causes selective HSC death through a Ca(2+)/calpain I-mediated necrosis cascade, suggesting that GA may represent a potential therapeutic agent to combat liver fibrosis.


Assuntos
Antioxidantes/farmacologia , Cálcio/metabolismo , Calpaína/metabolismo , Ácido Gálico/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Animais , Antioxidantes/administração & dosagem , Relação Dose-Resposta a Droga , Ácido Gálico/administração & dosagem , Células Estreladas do Fígado/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Masculino , Necrose/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Fatores de Tempo
9.
Biochem Biophys Res Commun ; 437(1): 55-61, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23791833

RESUMO

The aim of this study is to investigate the relationship between paraoxonase 1 (PON1) and atherosclerosis-related inflammation. In this study, human hepatoma HepG2 cell line was used as a hepatocyte model to examine the effects of the pro-inflammatory cytokines on PON1 expression. The results showed that IL-6, but not TNF-α and IL-1ß, significantly increased both the function and protein level of PON1; data from real-time RT-PCR analysis revealed that the IL-6-induced PON1 expression occurred at the transcriptional level. Increase of IκB kinase activity and IκB phosphorylation, and reduction of IκB protein level were also observed in IL-6-treated HepG2 cells compared with untreated culture. This event was accompanied by increase of NF-κB-p50 and -p65 nuclear translocation. Moreover, treatment with IL-6 augmented the DNA binding activity of NF-κB. Furthermore, pharmacological inhibition of NF-κB activation by PDTC and BAY 11-7082, markedly suppressed the IL-6-mediated PON1 expression. In addition, IL-6 increased the levels of phosphorylated protein kinase B (PKB, AKT). An AKT inhibitor LY294002 effectively suppressed IKK/IκB/NF-κB signaling and PON1 gene expression induced by IL-6. Our findings demonstrate that IL-6 upregulates PON1 gene expression through an AKT/NF-κB signaling axis in human hepatocyte-derived HepG2 cell line.


Assuntos
Arildialquilfosfatase/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Interleucina-6/farmacologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Arildialquilfosfatase/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células Hep G2 , Humanos , Interleucina-1beta/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
10.
Artigo em Inglês | MEDLINE | ID: mdl-22754582

RESUMO

Alzheimer's disease (AD) is an irreversible neurodegenerative disorder characterized by amyloid accumulation, neuronal death, and cognitive impairments. Yi-Chi-Tsung-Ming-Tang (YCTMT) is a traditional Chinese medicine and has never been used to enhance cognitive function and treat neurodegenerative disorders such as senile dementia. Whether YCTMT has a beneficial role in improving learning and memory in AD patients remains unclear. The present study showed that oral administration of YCTMT ameliorated amyloid-ß- (Aß(1-40)) injection-induced learning and memory impairments in rats, examined using passive avoidance and Morris water-maze tests. Immunostaining and Western Blot results showed that continuous Aß(1-40) infusion caused amyloid accumulation and decreased acetylcholine level in hippocampus. Oral administration of medium and high dose of YCTMT 7 days after the Aß(1-40) infusion decreased amyloid accumulation area and reversed acetylcholine decline in the Aß(1-40)-injected hippocampus, suggesting that YCTMT might inhibit Aß plague accumulation and rescue reduced acetylcholine expression. This study has provided evidence on the beneficial role of YCTMT in ameliorating amyloid-induced AD-like symptom, indicating that YCTMT may offer an alternative strategy for treating AD.

11.
Am J Chin Med ; 40(2): 357-72, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22419429

RESUMO

Human serum paraoxonase 1 (PON1), a calcium-dependent ester hydrolase, protects against the oxidative modification of low-density lipoprotein (LDL) and is a major anti-atherosclerotic component of high-density lipoprotein (HDL). Graptopetalum paraguayense, a folk herbal medicine commonly used in Taiwan, has antioxidative, anti-inflammatory, anti-hypertensive, and anti-atherogenic properties. The effects of G. paraguayense on the activity and/or expression of PON1 were examined using various extracts of the plant; extracts were made in water (GPWE), 50% ethanol (GP50E), and 95% ethanol (GP95E). Of these extracts, GP50E was found to be the most effective at increasing the function and expression of PON1 in a human hepatoma HepG2 cell line. Data from electrophoretic mobility shift assays and promoter-reporter luciferase analyses demonstrated that the DNA binding activity and transactivation ability of NF-κB were enhanced by GP50E. Treatment with NF-κB inhibitors, pyrrolidine dithiocarbamate, and BAY 11-7082 significantly attenuated GP50E-induced PON1 production and NF-κB transactivation activity. In addition, GP50E increased the levels of phosphorylated protein kinase B (PKB/AKT). Pharmacological inhibition of AKT by LY294002 effectively suppressed NF-κB activation and PON1 gene expression, suggesting that AKT was an upstream regulator of GP50E-mediated biological events. Overall, the results show that GP50E up-regulated PON1 gene expression via an AKT/NF-κB-dependent signaling pathway in human hepatoma HepG2 cells. This observation led to the conclusion that the anti-atherogenic characteristics of G. paraguayense are modulated, at least in part, via the up-regulation of hepatocyte PON1 gene expression.


Assuntos
Arildialquilfosfatase/genética , Crassulaceae/química , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , NF-kappa B/metabolismo , Extratos Vegetais/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Arildialquilfosfatase/metabolismo , Células Hep G2 , Humanos , NF-kappa B/genética , Proteínas Proto-Oncogênicas c-akt/genética
12.
Cell Signal ; 23(10): 1640-50, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21664967

RESUMO

The primary goal of the study was to investigate how peroxisome proliferator-activated receptor γ (PPARγ) played a critical role in the protection of H460 cell, one of the non-small cell lung cancer (NSCLC) cells with multidrug resistance, from transforming growth factor ß (TGFß)-mediated mitoinhibition. In the study, TGFß resistance of H460 cell was first confirmed by analyses of PPARγ expression, its interaction with TGFß-induced Smad3 and phospho-Smad3 (p-Smad3) and survival of H460. Results showed that enable to escape from G2/M phase arrest, H460 cell had higher resistance to TGFß-mediated mitoinhibition than CH27 (a drug sensitive control). TGFß significantly increased PPARγ expression of H460 but not of CH27 cell whereas nuclear accumulation of p-Smad3 was only limited to CH27, the latter was believed to contribute to the induction of P(21 waf1/cip1) and cyclin B1, cell cycle arrest at G2/M phase and TGFß-mediated mitoinhibition of CH27 cell. TGFß-induced PPARγ of H460 cell was further demonstrated to bind to Smad3 and p-Smad3, and GW9662 (PPARγ inhibitor) or PPARγ-specific shRNA could disrupt the binding. GW9662 also increased the nuclear accumulation of p-Smad3 that eventually led to the reduction of TGFß resistance of H460. A transient knockdown of PPARγ with shRNA revealed a similar effect as GW9662. In addition, activation of P(38) instead of ERK played a critical role in TGFß-induced expression of PPARγ, which subsequently activated RhoA in H460 cell.


Assuntos
Ciclo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , PPAR gama/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Anilidas/farmacologia , Apoptose , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase 2/farmacologia , Flavonoides/farmacologia , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Imidazóis/farmacologia , Imunoprecipitação , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , PPAR gama/antagonistas & inibidores , Ligação Proteica , Piridinas/farmacologia , RNA Interferente Pequeno/metabolismo , Proteína Smad3/metabolismo , Azul Tripano/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
13.
Eur J Pharmacol ; 650(2-3): 519-25, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21044622

RESUMO

Human serum paraoxonase 1 (PON1), an arylesterase, is associated with high-density lipoprotein (HDL) and can inhibit the oxidative modification of low-density lipoprotein (LDL), implying that PON1 may prevent atherosclerosis. Berberine, a botanical alkaloid, lowers the cholesterol level in serum and is thought to display cardioprotective properties. However, the effect of berberine on PON1 gene expression remains unclear. Thus, we evaluated how berberine regulates PON1 gene expression. In human hepatoma HepG2 and Huh7 cells, the PON1 protein levels were increased by berberine in a dose- and time-dependent manner. Data from real time PCR analysis indicated that berberine could up-regulate PON1 expression at the transcriptional level. Additionally, treating HepG2 cells with berberine increased the levels of phosphorylated JNK and its downstream target c-Jun. The PON1 upstream region contained a consensus binding site for AP1, and the electrophoretic mobility shift assay and chromatin immunoprecipitation analysis indicated that the AP1 factors, especially c-Jun, bind to the upstream sequence of the PON1 promoter upon berberine treatment. Moreover, pretreatment with SP600125 (JNK inhibitor) or curcumin (AP-1 inhibitor) markedly attenuated the berberine-induced PON1 promoter activity and protein expression. This is the first study to suggest that JNK/c-Jun signalling pathway plays a crucial role in berberine-regulated PON1 transcription in human hepatoma cells. The induction of PON1 by berberine elucidates a potential mechanism through which berberine may protect against atherosclerosis.


Assuntos
Arildialquilfosfatase/biossíntese , Berberina/farmacologia , Hepatócitos/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Arildialquilfosfatase/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Relação Dose-Resposta a Droga , Ensaio de Desvio de Mobilidade Eletroforética , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Fosforilação , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-jun/genética , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Regulação para Cima
14.
Curr Neurovasc Res ; 7(3): 223-37, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20560877

RESUMO

Molecular mechanism underlying leptin-mediated neuronal protection against glucose-oxygen-serum deprivation (GOSD) insult was investigated by focusing on the interactions among leptin, Interleukin-1beta (IL-1beta) and glutamate and their impacts on the growth of neurons under GOSD. The trypan blue dye exclusion assay, 4', 6-diamidino-2-phenylindole (DAPI) assay, cytokine antibody array assay, immunocytochemical staining assay, glutamate determination kit, immunoblocking and chemical blocking strategies were applied to serve the study goal. Results showed that in response to 6 h of GOSD, cortical neurons can secrete significant amounts of leptin and IL-1beta to protect neurons from GOSD-induced cell damage. Serine/threonine kinase Akt (Akt) and extracellular signal-related kinase (ERK) inhibitors significantly reversed leptin-mediated neuroprotection. GOSD-induced IL-1beta was further enhanced by leptin in Akt/ERK-dependent manner. Blockade of endogenous leptin with specific antibodies significantly inhibited GOSD-induced IL-1beta expression and increased glutamate release from GOSD neurons. IL-1 blockade with IL-1 receptor antagonist (IL-1ra) on the other hand, inhibited leptin-mediated neuroprotection and suppression of glutamate release from GOSD neurons. Pre-treating GOSD neurons with leptin and IL-1beta in combined significantly increased their survival but decreased their releases of glutamate. The results indicate that leptin may act through Akt and ERK signaling pathways to protect neurons from GOSD insult; the protection was in part IL-1beta dependent and through which the glutamate release from GOSD neurons was inhibited. Therapeutic values of leptin and IL-1beta were suggested in the treatment of cerebral ischemia at early stage.


Assuntos
Córtex Cerebral/metabolismo , Ácido Glutâmico/metabolismo , Hipóxia/prevenção & controle , Interleucina-1beta/metabolismo , Leptina/metabolismo , Neurônios/metabolismo , Análise de Variância , Animais , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/efeitos dos fármacos , Glucose/deficiência , Ácido Glutâmico/farmacologia , Imuno-Histoquímica , Interleucina-1beta/farmacologia , Leptina/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
15.
Nucleic Acids Res ; 38(18): 6148-58, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20494979

RESUMO

Carcinogenesis is determined based on both cell proliferation and death rates. Recent studies demonstrate that heat shock proteins (HSPs) regulate apoptosis. HLJ1, a member of the DnaJ-like Hsp40 family, is a newly identified tumor suppressor protein closely related to relapse and survival in non-small cell lung cancer (NSCLC) patients. However, its role in apoptosis is currently unknown. In this study, NSCLC cell lines displaying varying HLJ1 expression levels were subjected to ultraviolet (UV) irradiation, followed by flow cytometry. Interestingly, the percentages of apoptotic cells in the seven cell lines examined were positively correlated with HLJ1 expression. Enforcing expression of HLJ1 in low-HLJ1 expressing highly invasive cells promoted UV-induced apoptosis through enhancing JNK and caspase-3 activation in NSCLC. Additionally, UV irradiation led to reduced levels of HLJ1 predominantly in apoptotic cells. The pan-caspase inhibitor, zVAD-fmk and caspase-3-specific inhibitor, DEVD-fmk, prevented UV-induced degradation of HLJ1 by the late stage of apoptosis. Further experiments revealed a non-typical caspase-3 cleavage site (MEID) at amino acid 125-128 of HLJ1. Our results collectively suggest that HLJ1 is a novel substrate of caspase-3 during the UV-induced apoptotic process.


Assuntos
Apoptose , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Caspase 3/metabolismo , Proteínas de Choque Térmico HSP40/metabolismo , Neoplasias Pulmonares/enzimologia , Raios Ultravioleta , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia
16.
Eur J Pharmacol ; 627(1-3): 75-84, 2010 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-19903469

RESUMO

Molecular mechanism(s) responsible for drug resistance of non-small cell lung cancer (NSCLC) cells to cisplatin was investigated. Results showed that cisplatin (50muM)-induced cell death (apoptosis) was more significant in CH27 and A549 cell lines than in H460. The high protein levels of X-linked inhibitor-of-apoptosis protein (XIAP) observed in H460 cells appeared to play a key role in the regulation of cisplatin resistance of H460 cells. XIAP can bind to and suppress the activities of caspase 3 in H460 cells and lead to apoptosis inhibition of these cells. Blockade of XIAP activity by Embelin (XIAP inhibitor) or siRNA has increased caspase 3 activities and promoted cisplatin-induced cell death of H460 cells. The results indicate a therapeutic value of Embelin and/or XIAP siRNA in the control of cisplatin-resistant NSCLC cells (H460).


Assuntos
Cisplatino/farmacologia , Neoplasias Pulmonares/patologia , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Apoptose/efeitos dos fármacos , Benzoquinonas/farmacologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , RNA Interferente Pequeno/genética , Fatores de Tempo , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/deficiência , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética
17.
Brain Res Bull ; 79(3-4): 169-76, 2009 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-19429187

RESUMO

Molecules involved in self-protection of neurons against glucose/oxygen/serum deprivation (GOSD) were investigated. Trypan blue dye exclusion assay, Western blotting, ELISA, cytokine antibody array and chemical blocking assay were applied in the study. Results showed that early induction (at 6h of GOSD) of cyclooxygenase-2 (COX-2), leptin, transforming growth factor-beta1 (TGF-beta1), glial-cell-line-derived neurotrophic factor (GDNF) and neurotrophin-3 (NT-3) all played a compensatory role in the protection of neurons against GOSD. Decline of these molecules and peroxisome proliferators-activated receptor (PPAR)-gamma and -alpha since 12h of GOSD may lead to an irreversible neuronal death. Nitric oxide (NO) and superoxide at low concentrations were neuroprotective whereas at high concentrations were detrimental to neurons. Accumulation of NO and superoxide at late stage of GOSD should therefore be prevented. The study provided a useful platform for screening of potential anti-ischemic drugs and also explained why GOSD neuron derived conditioned medium (NCM) only exerted a time-restricted neuroprotection.


Assuntos
Glucose/metabolismo , Fármacos Neuroprotetores/metabolismo , Oxigênio/metabolismo , Soro/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados/química , Ciclo-Oxigenase 2/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Leptina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Neurotrofina 3/metabolismo , Óxido Nítrico/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxidos/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
18.
Respirology ; 14(4): 511-21, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19386070

RESUMO

BACKGROUND AND OBJECTIVE: The aim of this study was to characterize the changes in neutrophils and cytokines in BAL fluid following acute lung injury (ALI), and to determine the protective effect of post-injury treatment with IL-10. METHODS: A rat model of ALI was established by evenly spraying LPS (16 mg/kg) into the lungs followed by observation for 48 h. Histological changes and the kinetics of neutrophil infiltration were evaluated in the injured lungs. The cytokines (TNF-alpha, IL-6, IL-10 and interferon-gamma) and macrophage-inflammatory protein (MIP-2) were measured in BAL fluid by ELISA. The activation of BAL fluid neutrophils was investigated after treatment with IL-10 in vitro. The protective effect on histology and MIP-2 levels of intra-tracheal instillation of IL-10 12 and 16 h after LPS treatment was studied in vivo. RESULTS: Intra-tracheal instillation of LPS caused significant lung injury and the activation of neutrophils. The levels of TNF-alpha and IL-6 in BAL fluid peaked at 8 and 16 h after LPS instillation respectively. IL-10 levels reached a maximum at 16-24 h, at the beginning of resolution of tissue injury. IL-10 inhibited the activation of neutrophils in vitro and MIP-2 induction in vivo. IL-10 had a protective effect if it was administered 12 but not 16 h after LPS. CONCLUSIONS: Neutrophils appeared to play an important role in ALI. Time-dependent treatment with IL-10 after intra-tracheal instillation of LPS was effective in protecting rats from ALI, probably by suppressing pulmonary infiltration with activated neutrophils.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/patologia , Interleucina-10/uso terapêutico , Lesão Pulmonar Aguda/metabolismo , Animais , Técnicas de Cultura de Células , Citocinas/metabolismo , Modelos Animais de Doenças , Instilação de Medicamentos , Interleucina-10/administração & dosagem , Lipopolissacarídeos , Masculino , Ativação de Neutrófilo/efeitos dos fármacos , Infiltração de Neutrófilos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traqueia
19.
Shock ; 32(3): 276-85, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19106808

RESUMO

The pathophysiological role of soluble immune complexes (SICXs) and its relationship with neutrophils were investigated in LPS-induced acute lung injury (ALI) animal model (Sprague-Dawley rat) and through the in vitro studies. Results showed that LPS-induced SICX was timely related to changes of tumor necrosis factor alpha and macrophage inflammatory protein 2 (inflammatory cytokines) in bronchoalveolar lavage fluid. In vitro study showed that SICX can bind to Fc gammaR (CD64 and CD32 or CD16) to prevent the apoptosis of neutrophils. The SICX-mediated apoptosis inhibition was extracellular signal-regulated kinase (ERK) or phosphoinositide 3 kinase dependent and was interrupted by PD98059 and LY294002. In vivo, additional amount of SICX exacerbated the lung injury caused by LPS. LPS-induced lung injury and macrophage inflammatory protein 2 release, however, were prevented by CD64 and CD32 blockers (decoy antibodies). In conclusion, excessive amount of SICX in lung can act through Fc gammaRs to protect bronchoalveolar lavage fluid neutrophils from apoptosis that eventually lead to delayed resolution of ALI caused by LPS. Blockade of SICX engagement of CD32 and CD64 (with decoy antibodies) could interrupt SICX-mediated protection of neutrophils and protect lung from LPS-induced injury. The decoy antibodies may therefore have therapeutic utility in ALI.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Complexo Antígeno-Anticorpo/fisiologia , Lipopolissacarídeos/farmacologia , Neutrófilos/citologia , Lesão Pulmonar Aguda/induzido quimicamente , Animais , Complexo Antígeno-Anticorpo/metabolismo , Apoptose/efeitos dos fármacos , Líquido da Lavagem Broncoalveolar/química , Quimiocina CXCL2/metabolismo , Cromonas/farmacologia , Inibidores Enzimáticos , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Flavonoides/farmacologia , Masculino , Morfolinas/farmacologia , Neutrófilos/efeitos dos fármacos , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptores de IgG/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
20.
Brain Res ; 1239: 24-35, 2008 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-18804095

RESUMO

Conditioned medium (CM) collected from cultures of ischemic microglia, astrocytes, and neurons were protective to astrocytes under the in vitro ischemic condition (deprivation of oxygen, glucose and serum). Molecular and signaling pathway(s) responsible for the CMs protective activity were investigated. Results showed that CMs from the ischemic microglia (MCM), astrocytes (ACM) and neurons (NCM) contained glial cell line-derived neurotrophic factor (GDNF), which protects astrocytes against the in vitro ischemia. Expression of extra cellular signal-regulated kinase (ERK1/2) and nuclear factor-kappa B (NF-kB) by GDNF led to the inhibition of apoptosis of the ischemic astrocytes in a caspase 3-independent manner. However, CMs- and GDNF-mediated protection of the ischemic astrocytes was protein kinase B (Akt) independent. These results provided mechanistic data regarding how GDNF- and CMs-mediated protection of the ischemic astrocytes is taking place. These observations provide information for the use of GDNF and GDNF containing CMs in the control of cerebral ischemia.


Assuntos
Astrócitos/fisiologia , Isquemia Encefálica/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Isquemia/fisiopatologia , NF-kappa B/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Astrócitos/efeitos dos fármacos , Caspase 3/metabolismo , Células Cultivadas , Meios de Cultivo Condicionados , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacologia , Isquemia/tratamento farmacológico , Microglia/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...