Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
FEBS Open Bio ; 14(6): 1011-1027, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38604998

RESUMO

Tumor immunotherapy can be a suitable cancer treatment option in certain instances. Here we investigated the potential immunomodulatory effect of oral glycyrrhiza polysaccharides (GCP) on the antitumor function of γδT cells in intestinal epithelial cells in mice. We found that GCP can inhibit tumor growth and was involved in the regulation of systemic immunosuppression. GCP administration also promoted the differentiation of gut epithelia γδT cells into IFN-γ-producing subtype through regulation of local cytokines in gut mucosa. GCP administration increased local cytokine levels through gut microbiota and the gut mucosa Toll-like receptors / nuclear factor kappa-B pathway. Taken together, our results suggest that GCP might be a suitable candidate for tumor immunotherapy, although further clinical research, including clinical trials, are required to validate these results.


Assuntos
Microbioma Gastrointestinal , Glycyrrhiza , NF-kappa B , Polissacarídeos , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Polissacarídeos/farmacologia , Glycyrrhiza/química , Receptores Toll-Like/metabolismo , Transdução de Sinais/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Antineoplásicos/farmacologia
2.
Braz J Med Biol Res ; 55: e12252, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36383801

RESUMO

Nanosized copper particles (nano Cu) have been incorporated into products in multiple industries, although studies have demonstrated that these particles are nephrotoxic. We investigated the cytotoxicity of nanosized copper particles on rat mesangial cells and measured rates of apoptosis, the expression of caspase-3, and generation of reactive oxygen species. We also measured autophagy through the acridine orange (AO) staining and expression of Beclin-1, microtubule-associated protein 1 light chain 3, and p62 to screen the underlying mechanism of toxicity. Nanosized copper particles inhibited mesangial cell viability, up-regulated the activity of caspase-3, and increased the rates of apoptosis and the generation of reactive oxygen species in a concentration-dependent manner. Exposure to nano Cu increased the formation of acidic vesicular organelles and the expression of Beclin-1, microtubule-associated protein 1 light chain 3, and p62, and treatment with an autophagy inhibitor reduced nephrotoxicity. This indicated that the autophagy pathway is involved in the toxicity induced by nanosized copper particles to mesangial cells. This finding can contribute to the development of safety guidelines for the evaluation of nanomaterials in the future.


Assuntos
Cobre , Células Mesangiais , Ratos , Animais , Caspase 3 , Cobre/toxicidade , Cobre/metabolismo , Células Mesangiais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Beclina-1/metabolismo , Autofagia , Apoptose , Proteínas Associadas aos Microtúbulos/metabolismo , Linhagem Celular Tumoral
3.
An Acad Bras Cienc ; 94(2): e20210938, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35946645

RESUMO

Triptolide is a natural active compound that has significant neuroprotective properties and shows promising effects in the treatment of Alzheimer's disease (AD). Recent studies have shown that autophagy occurs in AD. In this study, we determined whether autophagy regulated by triptolide ameliorates neuronal death caused by amyloid-Beta1-42 (Aß1-42). We examined the effects of triptolide on cell viability, autophagy, apoptosis, and the protein kinase B/mammalian target of the rapamysin/70 kDa ribosomal protein S6 kinase (Akt/mTOR/p70S6K) signaling pathway in PC12 cells. The results indicated that triptolide treatment exhibited a cytoprotective effect against cell injury induced by Aß1-42. Triptolide also reduced apoptosis and enhanced cell survival by decreasing autophagosome accumulation and inducing autophagic degradation. Furthermore, our results also showed that activating the Akt/mTOR/p70S6K mechanism was one reason for the protection of triptolide. Triptolide treatment protected against Aß1-42-induced cytotoxicity by decreasing autophagosome accumulation, and inducing autophagic degradation in PC12 cells. These findings also suggest that the reduction of autophagosome accumulation observed in triptolide-treated cells was Akt/mTOR/p70S6K pathway dependent. Overall, triptolide exhibits a neuron protective effect and this study provides new insight into AD prevention and treatment.


Assuntos
Doença de Alzheimer , Proteínas Proto-Oncogênicas c-akt , Animais , Autofagia , Diterpenos , Compostos de Epóxi , Humanos , Mamíferos , Neuroproteção , Fenantrenos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-akt/farmacologia , Ratos , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Serina-Treonina Quinases TOR/farmacologia
4.
Braz. j. med. biol. res ; 55: e12252, 2022. graf
Artigo em Inglês | LILACS-Express | LILACS | ID: biblio-1403899

RESUMO

Nanosized copper particles (nano Cu) have been incorporated into products in multiple industries, although studies have demonstrated that these particles are nephrotoxic. We investigated the cytotoxicity of nanosized copper particles on rat mesangial cells and measured rates of apoptosis, the expression of caspase-3, and generation of reactive oxygen species. We also measured autophagy through the acridine orange (AO) staining and expression of Beclin-1, microtubule-associated protein 1 light chain 3, and p62 to screen the underlying mechanism of toxicity. Nanosized copper particles inhibited mesangial cell viability, up-regulated the activity of caspase-3, and increased the rates of apoptosis and the generation of reactive oxygen species in a concentration-dependent manner. Exposure to nano Cu increased the formation of acidic vesicular organelles and the expression of Beclin-1, microtubule-associated protein 1 light chain 3, and p62, and treatment with an autophagy inhibitor reduced nephrotoxicity. This indicated that the autophagy pathway is involved in the toxicity induced by nanosized copper particles to mesangial cells. This finding can contribute to the development of safety guidelines for the evaluation of nanomaterials in the future.

5.
Cancer Lett ; 478: 82-92, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32171858

RESUMO

Cancer cells undergo metabolic reprogramming to sustain their own survival under an environment of increased energy demand; however, the mechanism by which cancer cells ensure survival under glucose deprivation stressed conditions remains elusive. Here, we show that deprivation of glucose, dramatically activated the glycogen pathway, accompanied by elevated phosphoglucomutase 1 (PGM1) expression. We further identified that AMP-activated protein kinase (AMPK) stimulated PGM1 expression by inducing histone deacetylase 8 (HDAC8) phosphorylation. Moreover, we demonstrated that glucose deprivation-induced AMPK activation stimulated the translocation of HDAC8 from the nucleus to the cytoplasm, consequently disrupting the binding between HDAC8 and histone 3. PGM1 expression was also found to be critical for lung cancer glycolysis, the oxidative pentose phosphate pathway, and oxidative phosphorylation under glucose deprivation conditions, and further led to the aberrant expression of metabolic enzymes involved in glucose metabolism mediated by ERK1/2. Finally, PGM1 was found to be highly expressed in lung cancer tissues from patients, which correlated with a poor prognosis. Taken together, these results revealed that AMPK activation by glucose deprivation leads to enhanced PGM1 expression, an essential component of the metabolic switch, to facilitate cancer progression, suggesting PGM1 as promising anti-cancer treatment targets.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Glucose/metabolismo , Histona Desacetilases/metabolismo , Neoplasias Pulmonares/metabolismo , Fosfoglucomutase/genética , Fosfoglucomutase/metabolismo , Proteínas Repressoras/metabolismo , Células A549 , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Sobrevivência Celular , Citoplasma/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicólise , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Fosforilação , Transdução de Sinais , Regulação para Cima
6.
Life Sci ; 145: 19-26, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26679104

RESUMO

BACKGROUND: Recently, an abnormal deposition of Amyloid-Beta (Aß) was considered the primary cause of the pathogenesis of Alzheimer's disease (AD). And how to inhibit the cytotoxicity is considered an important target for the treatment of AD. Triptolide (TP), a purified diterpenoid from the herb Tripterygium wilfordii Hook.f. (TWHF), has potential neuroprotective effects pertinent to disease of the nervous system. However, whether triptolide and its specific mechanisms have protective functions in differentiated PC12 cells treated with Aß25-35 remain unclear. AIMS: The purpose is to investigate the protective functions of triptolide in Aß25-35-stimulated differentiated PC12 cells. MAIN METHODS: In the study, we use 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) assay, flow cytometry assay, immunohistochemical staining and Western blot to observe the effects of triptolide on cytotoxicity induced by Aß25-35 and its mechanism of oxidative stress. KEY FINDINGS: The result of MTT and LDH assay indicates that triptolide protected PC12 cells against Aß25-35-induced cytotoxicity. The flow cytometry assay shows that triptolide attenuated Aß25-35-induced apoptosis in differentiated PC12 cells. Meanwhile, the results give a clear indication that triptolide could downregulate generation of reactive oxygen species (ROS), hydrogen peroxide (H2O2) and malondialdehyde (MDA) induced by Aß25-35. The apoptotic process triggered by triptolide involved the up-regulation of the activity of superoxide dismutase (SOD). SIGNIFICANCE: The results suggest that triptolide may serve as an important role in the inhibition of the cell apoptosis induced by Aß and the decreased oxidative stress is a key mechanism in the protective effect of triptolide in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Diterpenos/farmacologia , Neurônios/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fragmentos de Peptídeos/metabolismo , Fenantrenos/farmacologia , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Animais , Apoptose/efeitos dos fármacos , Diterpenos/química , Compostos de Epóxi/química , Compostos de Epóxi/farmacologia , Peróxido de Hidrogênio/metabolismo , Malondialdeído/metabolismo , Neurônios/metabolismo , Fármacos Neuroprotetores/química , Células PC12 , Fenantrenos/química , Ratos , Espécies Reativas de Oxigênio/metabolismo , Tripterygium/química
7.
PLoS One ; 10(11): e0142719, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26554937

RESUMO

Evidence shows that an abnormal deposition of amyloid beta-peptide25-35 (Aß25-35) was the primary cause of the pathogenesis of Alzheimer's disease (AD). And the elimination of Aß25-35 is considered an important target for the treatment of AD. Triptolide (TP), isolated from Tripterygium wilfordii Hook.f. (TWHF), has been shown to possess a broad spectrum of biological profiles, including neurotrophic and neuroprotective effects. In our study investigating the effect and potential mechanism of triptolide on cytotoxicity of differentiated rat pheochromocytoma cell line (the PC12 cell line is often used as a neuronal developmental model) induced by Amyloid-Beta25-35 (Aß25-35), we used 3-(4, 5-dimethylthiazol-2-yl)-2, 5- diphenyltetrazolium bromide (MTT) assay, flow cytometry, Western blot, and acridine orange staining to detect whether triptolide could inhibit Aß25-35-induced cell apoptosis. We focused on the potential role of the autophagy pathway in Aß25-35-treated differentiated PC12 cells. Our experiments show that cell viability is significantly decreased, and the apoptosis increased in Aß25-35-treated differentiated PC12 cells. Meanwhile, Aß25-35 treatment increased the expression of microtubule-associated protein light chain 3 II (LC3 II), which indicates an activation of autophagy. However, triptolide could protect differentiated PC12 cells against Aß25-35-induced cytotoxicity and attenuate Aß25-35-induced differentiated PC12 cell apoptosis. Triptolide could also suppress the level of autophagy. In order to assess the effect of autophagy on the protective effects of triptolide in differentiated PC12 cells treated with Aß25-35, we used 3-Methyladenine (3-MA, an autophagy inhibitor) and rapamycin (an autophagy activator). MTT assay showed that 3-MA elevated cell viability compared with the Aß25-35-treated group and rapamycin inhibits the protection of triptolide. These results suggest that triptolide will repair the neurological damage in AD caused by deposition of Aß25-35 via the autophagy pathway, all of which may provide an exciting view of the potential application of triptolide or TWHF as a future research for AD.


Assuntos
Peptídeos beta-Amiloides/fisiologia , Autofagia , Diterpenos/farmacologia , Fragmentos de Peptídeos/fisiologia , Fenantrenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Compostos de Epóxi/farmacologia , Células PC12 , Ratos , Espécies Reativas de Oxigênio/metabolismo
8.
Cell Mol Biol Lett ; 18(4): 494-506, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23990403

RESUMO

The aim of malignant glioma treatment is to inhibit tumor cell proliferation and induce tumor cell apoptosis. Remifentanil is a clinical anesthetic drug that can activate the N-methyl-D-aspartate (NMDA) receptor. NMDA receptor signaling activates glycogen synthase kinase-3ß (GSK-3ß). Discovered some 32 years ago, GSK-3ß was only recently considered as a therapeutic target in cancer treatment. The purpose of this study was to assess whether remifentanil can induce the apoptosis of C6 cells through GSK-3ß activation. 3-(4,5-Dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) was used to detect cell viability. Hoechst 33342 staining and flow cytometry were used to detect cell apoptosis. The effect of GSK-3ß activation was detected using a GSK-3ß activation assay kit and 4-benzyl-2-methyl-1,2,4-thiadiazolidine-3,5-dione (TDZD-8), a potent and selective small molecule inhibitor of GSK-3ß. The MTT assay indicated that remifentanil induced C6 cell death in a concentration- and time-dependent manner. Hoechst 33342 staining and flow cytometry showed that remifentanil significantly induced C6 cell apoptosis. The measurement of GSK-3ß activation showed that remifentanil increased the cellular level of GSK-3ß. All of these toxic effects can be attenuated by treatment with TDZD-8. These results suggest that remifentanil is able to induce C6 cell apoptosis through GSK-3ß activation, which provides a basis for its potential use in the treatment of malignant gliomas.


Assuntos
Analgésicos Opioides/farmacologia , Apoptose/efeitos dos fármacos , Glioma/tratamento farmacológico , Glioma/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Piperidinas/farmacologia , Animais , Linhagem Celular Tumoral , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Ratos , Remifentanil , Tiadiazóis/farmacologia
9.
Zhonghua Zhong Liu Za Zhi ; 34(4): 254-8, 2012 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-22781035

RESUMO

OBJECTIVE: To address the hypothesis that hydrogen sulfide (H(2)S) is a functionally significant stimulator in the development of glioblastoma (GBM) and explore the mechanism of stimulation. METHODS: Forty adult Sprague-Dawley (SD) rats were given intracerebral injection of rat C6 glioma cell suspension, and an intraperitoneal injection of sodium hydrosulfide (NaHS), an exogenous H(2)S donor. The 40 rats were randomly divided into 4 groups of 10 rats in each: the control group, NaHS group, C6 glioma group (intracerebral implantation of C6 glioma cells) and C6-NaHS group (intracerebral implantation of C6 glioma cells and intraperitoneal injection of NaHS). Food and water were freely available during all phases of the experiment. Physical symptoms were observed and the tumor size was measured. Histological changes were examined by pathology. Immunohistochemical staining was used to analyze the expression of HIF-1α and integrated optical density (IOD) was used to determine the tumor microvessel density (MVD). The H(2)S content in the brain was measured. RESULTS: The physical symptoms of tumor-bearing rats became more serious after NaHS injection. The H(2)S level in the C6 glioma group was higher than that in the control group [(35.25 ± 1.03) nmol/g vs. (29.12 ± 0.94) nmol/g, P < 0.05], and the highest H(2)S level was found in the C6-NaHS group. The pathological examination showed that the implanted tumors were predominantly spheroid with a distinct border and no capsule could be detected. Neovascular proliferation was also observed. Foci of tumor necrosis, intratumoral hemorrhage, pseudopalisades and tumor cavity were clearly observed. The glioma cells had scant eosinophilic cytoplasm and enlarged hyperchromatic nuclei. All these phenomena were more markedly in the C6-NaHS group compared with that in other three groups. The mean tumor volume was significantly different between the C6 and C6-NaHS rats [(32.0 ± 6.9) mm(3) vs. (67.8 ± 11.9) mm(3), P < 0.001]. Immunohistochemical analysis exhibited that the hypoxia-inducible factor-1alpha (HIF-1α) and CD34 expression were significantly increased after the intraperitoneal injection of NaHS in the C6-NaHS rats (comparing the IOD between C6-NaHS group and C6 group, HIF-1α: 133 962.9 ± 451.4 vs. 38 569.8 ± 408.6, P < 0.001; CD34: 73 368.6 ± 404.8 vs. 14 570.6 ± 748.7, P < 0.001). Moreover, compared with the C6 group, there were higher MVD in the C6-NaHS group [(41.2 ± 7.9)/mm(2) vs. (97.0 ± 10.8)/mm(2), P < 0.001]. CONCLUSIONS: H(2)S serves as a stimulator in the development of rat glioblastoma and exogenous H(2)S strongly promotes the tumor growth. The stimulating mechanisms include the increase of HIF-1α expression and neovascular formation. H(2)S may be a significant regulator in the development of tumor.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Sulfeto de Hidrogênio/metabolismo , Sulfetos/farmacologia , Animais , Antígenos CD34/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Injeções Intraperitoneais , Masculino , Transplante de Neoplasias , Neovascularização Patológica , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Sulfetos/administração & dosagem , Carga Tumoral
10.
Cell Stress Chaperones ; 17(4): 485-93, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22270613

RESUMO

Leukemia inhibitory factor (LIF) is a pleiotropic glycoprotein belonging to the interleukin-6 family of cytokines. In kidney, LIF regulates nephrogenesis, involves in tubular regeneration, responds to pro- and anti-inflammatory stimuli, and so on. LIF also plays an essential role in protective mechanisms triggered by preconditioning-induced oxidative stress. Although LIF shows a wide range of biologic activities, effects of LIF on high glucose-induced oxidative stress in podocytes remain unclear. The aim of the study was to assess whether LIF can attenuate high glucose-induced apoptosis in podocytes. The result of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay indicated that LIF protected podocytes against high glucose-induced cytotoxicity. The flow cytometry assay showed that LIF attenuated high glucose-induced apoptosis in podocytes. Meanwhile, the result of flow cytometric assay gave the clear indication that LIF decreased high glucose-induced elevated level of reactive oxygen species (ROS). The measurement of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, superoxide dismutase (SOD), malondialdehyde (MDA), and caspase-3 activity levels showed that LIF attenuated the high glucose-induced decreased level of SOD and elevated level of NADPH oxidase, MDA and caspase-3 activity. These results may provide potential therapy for diabetic nephropathy in the future.


Assuntos
Apoptose/efeitos dos fármacos , Glucose/farmacologia , Fator Inibidor de Leucemia/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Podócitos , Animais , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Espécies Reativas de Oxigênio/metabolismo
11.
J Biomed Mater Res A ; 100(3): 738-45, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22213249

RESUMO

Hydroxyapatite (HA) nanoparticles have been reported to exhibit anti-tumor effects on various human cancers, but the effects of HA on glioma cells remain unclear. The aim of this study was to explore whether HA can inhibit the proliferation and induce the apoptosis of C6 cells. Use of the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay indicated that HA induced C6 cell death in a concentration-dependent and time-dependent manner. Results from hoechst 33342 staining and flow cytometry assay showed that HA induced C6 cell apoptosis significantly. Meanwhile, the flow cytometric assay gave clear indication that HA induced intracellular accumulation of reactive oxygen species (ROS). The measurement of superoxide dismutase (SOD) generation showed that HA decreased the total SOD of cellular levels. Interestingly, pretreatment of N-(mercaptopropionyl)-glycine (N-MPG), known as a type of ROS scavenger formulations, could somehow inhibit C6 cell apoptosis induced by HA. These results may provide potential anti-glioma treatment in the future.


Assuntos
Apoptose/efeitos dos fármacos , Durapatita/química , Durapatita/farmacologia , Nanopartículas/química , Estresse Oxidativo/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Teste de Materiais , Ratos , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo , Tiopronina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA