Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Hepatocell Carcinoma ; 9: 171-188, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35308603

RESUMO

Background and Aims: Hepatocellular carcinoma (HCC) is a common malignant disease with high morbidity and mortality throughout the world. While Borealin is a putative oncogene that is dysregulated in multiple tumors, its exact role in HCC remains less investigated. Methods: Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC) assays were employed to examine the relative amount of Borealin. Gene set enrichment analysis (GSEA) and other bioinformatic analyses were implemented to probe into the potential functions of Borealin. The biological roles and mechanisms of Borealin in the tumorigenesis and development of HCC were further evaluated using a battery of functional assays in vivo and in vitro. Results: Borealin was enhanced in the HCC tissue samples and hepatoma cells when compared with the nontumor tissues and normal liver cells. Higher Borealin expression was positively linked with advanced pathological phenotypes and inferior overall survival. The overexpression of Borealin promoted the cells' abilities on proliferation, invasion and epithelial-mesenchymal transition (EMT) in vitro, facilitated tumor growth and lung metastasis in vivo, whereas the silencing of Borealin inhibited these capabilities in vitro. Furthermore, Borealin interacted with ß-catenin and further activated the Wnt/ß-catenin signaling pathway, which endowed HCC cells with highly aggressive and metastatic capabilities. Conclusion: Borealin was identified as an oncogene that could promote HCC growth and metastasis by activating the WNT/ß-catenin signaling pathway. These findings extended the understanding of Borealin in HCC tumorigenesis and development and highlighted the significance of Borealin in HCC diagnosis and treatment.

2.
Int Immunopharmacol ; 99: 108038, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34364304

RESUMO

BACKGROUND: Cannabinoid receptor 2 (CB2), whose activities are upregulated during sepsis, may be related to the regulation of inflammatory programmed cell death called pyroptosis. The aim of this study is to investigate the role of CB2 activation in attenuation of inflammation through inhibiting pyroptosis in cecal ligation puncture (CLP)-induced sepsis andlipopolysaccharide (LPS) + ATP-stimulated macrophages. METHODS: C57BL/6 mice were subjected to CLP procedure and treated with CB2 agonist HU308 and CB2 antagonist AM630. Lung tissues were collected for analyses of lung W/D ratio, inflammatory factors levels, and pyroptosis-related protein expression. Murine bone-marrow-derived macrophages (BMDM) were treated with LPS and ATP to construct a septic model in vitro in the presence of HU308 and AM630 for assessment of cell injury, cytokine levels and pyroptosis-related protein expression accordingly. To verify the relationship between CB2 receptors and pyroptosis in the process of inflammatory response, BMDM were transduced with CB2 receptors knockdown lentiviral vectors in the presence of HU308 and AM630 for assessment of pyroptosis-related protein expression. RESULTS: CB2 activation ameliorated the release of inflammatory mediators. The results showed that CLP-induced pyroptosis was elevated, and CB2 agonist HU308 treatment inhibited the pyroptosis activity through a decrease of the protein levels of NLRP3 as well as caspase-1 and GSDMD activation. Similar results were obtained in BMDM after LPS and ATP treatment. Treatment with CB2 knockdown lentiviral particles prevented the HU308-induced decreases in cell pyroptosis, demonstrating that endogenous CB2 receptors are required for the cannabinoid-induced cell protection. CONCLUSIONS: CB2 receptors activation plays a protective role in sepsis through inhibition of pyroptosis. The effect of CB2 receptors against pyroptosis depends on the existence of endogenous CB2 receptors.


Assuntos
Piroptose/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Receptor CB2 de Canabinoide/metabolismo , Sepse/tratamento farmacológico , Trifosfato de Adenosina/toxicidade , Animais , Canabinoides/farmacologia , Ceco/lesões , Modelos Animais de Doenças , Indóis/farmacologia , Inflamação/tratamento farmacológico , Inflamação/patologia , Ligadura/métodos , Lipopolissacarídeos/toxicidade , Pulmão/efeitos dos fármacos , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Punções/efeitos adversos , Punções/métodos , Receptor CB2 de Canabinoide/antagonistas & inibidores , Sepse/etiologia
3.
Inflammation ; 41(5): 1610-1620, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29766401

RESUMO

Penehyclidine hydrochloride (PHC), a type of hyoscyamus drug, has both antimuscarinic and antinicotinic activities and retains potent central and peripheral anticholinergic activities. Compared with other hyoscyamine, the notable advantage of PHC is that it has few M2 receptor-associated cardiovascular side effects. Recent studies and clinical trials have suggested that treatment with penehyclidine hydrochloride may also possess good effects in the treatment of lung injury. The mechanism responsible for this effect has yet to be determined; however, one possibility is that they might do so by a direct effect on pulmonary vascular endothelium. Since inflammatory reactions of the endothelium are signs of endothelial injury in the pathogenesis of lung injury, we determined the effects of penehyclidine hydrochloride on endothelial inflammatory injury in cultured human pulmonary microvascular endothelial cells (HPMVEC). Furthermore, human pulmonary microvascular endothelial cells were transfected with a shRNA-containing plasmid that specifically targets beta-arrestin-1 mRNA, to test whether the effect of penehyclidine hydrochloride on lipopolysaccharide (LPS)-induced endothelial cell injury is dependent on its upregulation of beta-arrestin-1 or not. Penehyclidine hydrochloride reduced the inflammatory responses to LPS stimulation, as evidenced by reduced lactate dehydrogenase (LDH), tumor necrosis factor-alpha (TNF-α), and interleukelin-6 (IL-6) levels, as well as vascular cell adhesion molecule 1 (VCAM-1) and intercellular cell adhesion molecule-1 (ICAM-1) expressions. This was found to result from increased beta-arrestin-1 expression and decreased nuclear transcription factor-κB (NF-κB) activation. Expression of a shRNA-containing plasmid that specifically targets beta-arrestin-1 mRNA nullified these effects of penehyclidine hydrochloride. The results indicate that penehyclidine hydrochloride exerts a protective effect on pulmonary microvascular endothelial inflammatory injury induced by LPS. We also demonstrate that this is due to its ability to increase beta-arrestin-1, which in turn inhibits NF-κB activation.


Assuntos
Células Endoteliais/patologia , Inflamação/tratamento farmacológico , Lesão Pulmonar/tratamento farmacológico , Quinuclidinas/farmacologia , beta-Arrestina 1/metabolismo , Células Cultivadas , Humanos , Inflamação/induzido quimicamente , Lipopolissacarídeos , Lesão Pulmonar/induzido quimicamente , Microvasos/patologia , NF-kappa B/antagonistas & inibidores , Quinuclidinas/uso terapêutico , Regulação para Cima/efeitos dos fármacos
4.
Pulm Pharmacol Ther ; 48: 144-150, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29158153

RESUMO

LPS has been recently shown to induce muscarinic acetylcholine 3 receptor (M3 receptor) expression and penehyclidine hydrochloride (PHC) is an anticholinergic drug which could block the expression of M3 receptor. PHC has been demonstrated to perform protective effect on cell injury. This study is to investigate whether the effect of PHC on microvascular endothelial injury is related to its inhibition of M3 receptor or not. HPMVECs were treated with specific M3 receptor shRNA or PBS, and randomly divided into LPS group (A group), LPS+PHC group (B group), LPS + M3 shRNA group (C group) and LPS + PHC + M3 shRNA group (D group). Cells were collected at 60 min after LPS treatment to measure levels of LDH, endothelial permeability, TNF-α and IL-6 levels, NF-κB p65 activation, I-κB protein expression, p38MAPK, and ERK1/2 activations as well as M3 mRNA expression. PHC could decrease LDH levels, cell permeability, TNF-α and IL-6 levels, p38 MAPK, ERK1/2, NF-κB p65 activations and M3 mRNA expressions compared with LPS group. When M3 receptor was silence, the changes of these indices were much more obvious. These findings suggest that M3 receptor plays an important role in LPS-induced pulmonary microvascular endothelial injury, which is regulated through NF-κB p65 and MAPK activation. And knockout of M3 receptor could attenuate LPS-induced pulmonary microvascular endothelial injury. Regulative effects of PHC on pulmonary microvascular permeability and NF-κB p65 as well as MAPK activations are including but not limited to inhibition of M3 receptor.


Assuntos
Antagonistas Colinérgicos/farmacologia , Endotélio Vascular/efeitos dos fármacos , Quinuclidinas/farmacologia , Receptor Muscarínico M3/genética , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/fisiopatologia , Permeabilidade Capilar/efeitos dos fármacos , Linhagem Celular , Endotélio Vascular/patologia , Técnicas de Silenciamento de Genes , Humanos , Lipopolissacarídeos/toxicidade , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Receptor Muscarínico M3/antagonistas & inibidores , Fator de Transcrição RelA/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...