Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(4): 167085, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38369216

RESUMO

Mechanical ventilation (MV) has the potential to induce extra-pulmonary organ damage by adversely affecting the lungs and promoting the secretion of inflammatory cytokines. High-mobility group box 1 protein (HMGB1) is a pro-inflammatory mediator in ventilator-induced lung injury (VILI), but its effect on MV-associated liver injury and the mechanisms are poorly understood. In the present study, mice were subjected to high-volume MV (20 ml/kg) to induce VILI. MV-induced HMGB1 prompted neutrophil extracellular traps (NETs) formation and PANoptosis within the liver. Inhibiting NETs formation by DNase I or PAD4 inhibitor, or by HMGB1 neutralizing ameliorated the liver injury. HMGB1 activated neutrophils to form NETs through TLR4/MyD88/TRAF6 pathway. Importantly, Importin7 siRNA nanoparticles inhibited HMGB1 release and protected against MV-associated liver injury. These data provide evidence of MV-induced HMGB1 prompted NETs formation and PANoptosis in the liver via the TLR4/MyD88/TRAF6 pathway. HMGB1 is a potential therapeutic target for MV-associated liver injury.


Assuntos
Armadilhas Extracelulares , Proteína HMGB1 , Lesão Pulmonar Induzida por Ventilação Mecânica , Camundongos , Animais , Armadilhas Extracelulares/metabolismo , Respiração Artificial , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , RNA Interferente Pequeno/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Fígado/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/prevenção & controle , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo
2.
Biomed Pharmacother ; 165: 115237, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37516020

RESUMO

Mechanical ventilation (MV) may negatively affect the lungs and cause the release of inflammatory mediators, resulting in extra-pulmonary organ dysfunction. Studies have revealed systemically elevated levels of proinflammatory cytokines in animal models of ventilator-induced lung injury (VILI); however, whether these cytokines have an effect on gut injury and the mechanisms involved remain unknown. In this study, VILI was generated in mice with high tidal volume mechanical ventilation (20 ml/kg). Tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and IL-6 concentrations in serum and gut measured by ELISA showed significant elevation in the VILI mice. Significant increases in gut injury and PANoptosis were observed in the VILI mice, which were positively correlated with the serum levels of TNF-α, IL-1ß, and IL-6. The VILI mice displayed intestinal barrier defects, decreased expressions of occludin and zonula occludin-1 (ZO-1), and increased expression of claudin-2 and the activation of myosin light chain (MLC). Importantly, intratracheal administration of Imp7 siRNA nanoparticle effectively inhibited cytokines production and protected mice from VILI-induced gut injury. These data provide evidence of systemic cytokines contributing to gut injury following VILI and highlight the possibility of targeting cytokines inhibition via Imp7 siRNA nanoparticle as a potential therapeutic intervention for alleviating gut injury following VILI.


Assuntos
Citocinas , Lesão Pulmonar Induzida por Ventilação Mecânica , Camundongos , Animais , Citocinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Interleucina-6/metabolismo , RNA Interferente Pequeno/metabolismo , Ocludina/metabolismo , Pulmão/patologia , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Lesão Pulmonar Induzida por Ventilação Mecânica/patologia , Camundongos Endogâmicos C57BL
3.
Inflamm Res ; 72(5): 971-988, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37004548

RESUMO

BACKGROUND: The ability of p38 to phosphorylate substrates in the nucleus and the role of nuclear p38 in the regulation of inflammation have focused attention on the subcellular localization of the kinase. Although it is clear that p38 shuttles to the nucleus upon stimulation, the mechanisms that regulate p38 nuclear input in response to mechanical stretch remain to be determined. METHODS: Cyclic stretch (CS)-induced nuclear translocation of p38 was determined by Western blotting and immunofluorescence. The p38 interacting protein was identified using endogenous pull-down and protein binding assays. The potential role of importin-7 (Imp7) in CS-induced nuclear translocation of p38 and p38-dependent gene expression was confirmed using a series of in vitro and in vivo experiments. Furthermore, we tested the therapeutic potential of intratracheal administration of Imp7 siRNA-loaded nanoparticles in the ventilator-induced lung injury (VILI) mouse model. RESULTS: We show that CS induced phosphorylation-dependent nuclear translocation of p38, which required the involvement of microtubules and dynein. Endogenous pull-down assay revealed Imp7 to be a potential p38-interacting protein, and the direct interaction between p38 and Imp7 was confirmed by in vitro and in vivo binding assays. Furthermore, silencing Imp7 inhibited CS-induced nuclear translocation of p38 and subsequent cytokine production. Notably, intratracheal administration of Imp7 siRNA nanoparticles attenuated lung inflammation and histological damage in the VILI mouse model. CONCLUSIONS: Our findings uncover a key role for Imp7 in the process of p38 nuclear import after CS stimulation and highlight the potential of preventing p38 nuclear translocation in treatment of VILI.


Assuntos
Núcleo Celular , Lesão Pulmonar Induzida por Ventilação Mecânica , Camundongos , Animais , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , RNA Interferente Pequeno/metabolismo , Carioferinas/metabolismo , Lesão Pulmonar Induzida por Ventilação Mecânica/tratamento farmacológico , Lesão Pulmonar Induzida por Ventilação Mecânica/metabolismo
4.
Saudi Med J ; 43(11): 1209-1216, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36379536

RESUMO

OBJECTIVES: To determine whether sevoflurane regulates cisplatin resistance in neuroblastoma cells. METHODS: The SH-SY5Y cell line with cisplatin-resistant phenotype (SH-SY5Y-SR) was generated. Cells were co-treated with sevoflurane and cisplatin to seek the sevoflurane function on cisplatin resistance. Key targets of sevoflurane treatment were determined using sequencing (ribonucleic acid [RNA-seq]). Cells were then transfected with specific vectors. Linc00473 and microRNA-490-5p (miR-490-5p) levels were detected using reverse transcriptase quantitative real-time reverse transcription PCR (RT-qPCR). Linc00473-miR-490-5p binding was confirmed using a luciferase reporter-gene assay. After treatment, cell proliferation, viability, and caspase-3 activity were measured to determine the effects of treatment on tumor cells. Each experimental result is based on three independent experiments. RESULTS: Co-treatment with sevoflurane and cisplatin markedly improved the sensitivity of SH-SY5Y-SR cells to cisplatin, which inhibited the occurrence of cisplatin resistance. The RNA-sequencing analysis and RT-qPCR showed that sevoflurane inhibited linc00473 expression. Overexpression of linc00473 promoted cell proliferation, inhibited apoptosis, and promoted cisplatin resistance. The linc00473/miR-490-5p/V-akt murine thymoma viral oncogene homolog 1 (AKT1) axis was found to mediate the regulatory effects of sevoflurane on cisplatin resistance. CONCLUSION: Sevoflurane has great clinical potential against cisplatin-resistant tumors. Further animal experiments and clinical trials are required to achieve this goal.


Assuntos
MicroRNAs , Neuroblastoma , Camundongos , Animais , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Sevoflurano/farmacologia , MicroRNAs/genética , Linhagem Celular Tumoral , Neuroblastoma/tratamento farmacológico , Neuroblastoma/genética , Neuroblastoma/patologia , Proliferação de Células/genética , Proteínas Proto-Oncogênicas c-akt
5.
Cell Commun Signal ; 20(1): 65, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551614

RESUMO

BACKGROUND: Ezrin/radixin/moesin (ERM) proteins are members of the protein 4.1 superfamily and function as linkers that connect the actin cytoskeleton to the plasma membrane of cells. ERM also play critical role in the Lipopolysaccharide (LPS)-induced inflammatory response. However, the signaling mechanisms involved in this process remain unclear. In this study, we aimed to investigate the potential role of the rho-associated coiled-coil containing protein kinase (ROCK) pathway in LPS-induced ezrin phosphorylation and cytokine production in pulmonary alveolar epithelial cells. METHODS: Cultured A549 and HPAEpiC cells were treated with LPS. The expression and localization of ezrin in A549 and HPAEpiC cells were then analyzed by western blotting and immunoflurescence. Activation of RhoA/ROCK was assessed by western blotting and RhoA activity assays. The interaction of ezrin with Syk and myeloid differentiation factor 88 (MyD88)/IL-1R-associated kinase 1 (IRAK-1) was investigated by co-immunoprecipitation. The activation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) was measured with electrophoretic mobility shift assays and by western blotting. ELISA and western blotting were performed to detect the levels of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß), and high mobility group box 1 protein (HMGB1) release into the culture supernatant, and cellular HMGB1 levels. RESULTS: LPS induced ezrin phosphorylation in a concentration- and time-dependent manner. The blockade of RhoA/ROCK inhibited LPS-induced ezrin phosphorylation and its translocation from the cytoplasm to the cell membrane. Co-immunoprecipitation assays further revealed that ezrin associated with Syk constitutively, but only associated with MyD88/IRAK-1 upon LPS challenge. Moreover, LPS-induced p38 and nuclear NF-κB activation was found to be ezrin dependent. The suppression of ezrin by siRNA or the blockade of ROCK activation with Y-27632 reduced the production of TNF-α, IL-1ß, and HMGB1 in response to LPS. CONCLUSIONS: Our findings reveal a novel regulatory mechanism involving ezrin in the LPS-induced production of pro-inflammatory cytokines, and highlight the importance of the RhoA/ROCK-ezrin/Syk-MyD88/IRAK1 axis. Data presented in this manuscript provide novel insights into the signaling pathways activated in pulmonary alveolar epithelial cells by LPS. Video Abstract.


Assuntos
Proteína HMGB1 , Lipopolissacarídeos , Células Epiteliais Alveolares/metabolismo , Citocinas/metabolismo , Proteínas do Citoesqueleto , Proteína HMGB1/metabolismo , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
7.
Taiwan J Obstet Gynecol ; 58(3): 401-408, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31122533

RESUMO

OBJECTIVE: The aim of this study was to explore the effect of miR-146b expression and variants on endometriosis and its associated pain symptom. MATERIALS AND METHODS: Genotyping and expression of miR-146b was performed on 74 endometriosis patients and 23 healthy controls. ESCs were subsequently co-cultured with peripheral blood (PB)-derived monocytes (PBMC)-driven macrophages. After overexpression and inhibition of miR-146b, cytokine production from the macrophages were determined by enzyme-linked immunosorbent assay (ELISA). Western blot were done to measure the regulation of IRF5 by miR-146b. RESULTS: We found that miR-146b expression was increased in PF supernatant and PF CD14 + monocytes/Macrophages of endometriosis patients, with endometriosis patients with pain (EPWP) showing higher miR-146b expression compared with the endometriosis patients without pain (EPNP). CT/CC genotype of miR-146b rs1536309 was associated with the risk of pain symptom of endometriosis. For the function studies, we found that miR-146b was involved in the negative regulation of inflammation through attenuating IRF5 expression. Macrophages from patients who carries CT/CC genotype of miR-146b rs1536309 showed decreasing miR-146b expression and enhancement of the ability of pro-inflammation. CONCLUSIONS: Our findings suggest an important role of miR-146b level and variants in endometriosis that helps to regulate the process of endometriosis and its associated pain.


Assuntos
Endometriose/genética , MicroRNAs , Dor/genética , Adulto , Estudos de Casos e Controles , Feminino , Humanos , Fatores Reguladores de Interferon , Macrófagos/metabolismo , Células Estromais/metabolismo , Inquéritos e Questionários
8.
Oncol Res ; 26(6): 959-966, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29295731

RESUMO

Tbx3, a member of the T-box family of transcription factors, contributes directly to tumor formation, migration, and invasion. However, the role of Tbx3 in the metastasis of HCC remains unclear. In the present study, Tbx3 expression was detected in HCC tissues and cells by Western blot, and Tbx3 expression was regulated by use of siRNAs or lentivirus-mediated vectors. Here we found that Tbx3 protein expression increased in HCC tissues and cell lines. Tbx3 expression was positively associated with multiple tumor nodes, venous infiltration, and advanced TNM tumor stage. Survival analysis demonstrated that Tbx3 expression was an independent prognostic factor for HCC patients. In vitro assays further validated that Tbx3 indeed prompted HCC cell migration and invasion. In addition, Tbx3 expression was negatively related with E-cadherin expression in HCC tissues. Mechanically, Tbx3 inhibited the expression of E-cadherin, and then facilitated epithelial-mesenchymal transition (EMT) of HCC cells. Furthermore, the effect of Tbx3 knockdown on HCC cells was attenuated by E-cadherin knockdown. In conclusion, Tbx3 may be a novel prognostic factor, and it contributes to HCC cell migration, invasion, and EMT by repressing E-cadherin expression. Thus, Tbx3 may be recommended as a therapeutic target for HCC patients.


Assuntos
Antígenos CD/metabolismo , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Carcinoma Hepatocelular/patologia , Movimento Celular , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/patologia , Proteínas com Domínio T/metabolismo , Antígenos CD/genética , Apoptose , Biomarcadores Tumorais/genética , Caderinas/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Invasividade Neoplásica , Prognóstico , Taxa de Sobrevida , Proteínas com Domínio T/genética , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA