Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancer Med ; 12(19): 19777-19793, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37732632

RESUMO

OBJECTIVES: Lung cancer is a major public health concern and represents the most common cause of cancer-related death worldwide. Among eukaryotes, the G protein-coupled receptor (GPCR) family stands as the largest group of membrane proteins. Alterations in GPCR gene expression and dysregulation of signal transduction have been recognized as the markers of malignancy. As a member of the GPCR family, G protein-coupled receptor 37 (GPR37) exhibits unknown functions in tumors, particularly in non-small-cell lung cancer (NSCLC) METHODS: We explored the expression and prognosis of GPR37 in NSCLC through TCGA, GTEx, GEO, and GEPIA2. We detected the expression of GPR37 in NSCLC tissues and cell lines. The study explored the influence of GPR37 on tumor cell proliferation. Furthermore, we examined the effects of GPR37 on tumor cell apoptosis and invasion. Most importantly, we investigated whether GPR37 affects cisplatin-induced drug resistance in NSCLC. Furthermore, by conducting animal experiments, we assessed the impact of GPR37 on NSCLC and delved into underlying mechanisms. RESULTS: (1) In NSCLC, the expression of GPR37 is markedly higher than that in corresponding normal tissues. We found that elevated GPR37 expression predicts an unfavorable prognosis. (2) It was demonstrated that GPR37 positively regulates NSCLC cell invasion, migration, and proliferation, suppresses cell apoptosis, heightens resistance to cisplatin, and promotes tumor formation and growth. Conversely, we observed that GPR37 knockdown suppresses NSCLC cell invasion, migration, and proliferation, promotes cell apoptosis, increases sensitivity to cisplatin, and affects tumor formation and growth. (3) GPR37 activates PI3K/Akt/mTOR signal transduction pathways to mediate epithelial-mesenchymal transition (EMT), thereby promoting the progression of NSCLC. CONCLUSIONS: It was suggested that GPR37 acts a crucial role in promoting the occurrence and development of NSCLC. Knockdown of GPR37 significantly inhibits the occurrence and development of NSCLC. Therefore, our findings demonstrated that GPR37 may represent a viable therapeutic target for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Animais , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Humanos
3.
Int Immunopharmacol ; 108: 108710, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35405595

RESUMO

Sepsis is a life-threatening medical condition caused by infection-triggered aberrant immune responses, leading to host tissue and organ injury. Despite advances in medical interventions, the mortality rate for septic shock remains high. Recent studies highlight the role of oxidative stress in the occurrence and development of sepsis, providing a potential therapeutic target for preventing sepsis-associated organ injury. In this study, we showed that Maackiain, a natural compound isolated from Sophora flavescens, exerted a protective role in a cecal ligation and puncture (CLP)-induced murine model of sepsis. Maackiain treatment reduced organ injury, and mitigated systematic inflammation and oxidative stress in septic mice. Maackiain also reduced the levels of inflammatory cytokines and reactive oxygen species (ROS) in RAW264.7 macrophage cells stimulated with lipopolysaccharide (LPS). We further demonstrated that Maackiain initiated activation of nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in RAW264.7 cells in an AMP-activated protein kinase (AMPK)-dependent way. Moreover, inhibition of AMPK/Nrf2 axis abrogated the anti-inflammatory and anti-oxidant effects of Maackiain both in vitro and in vivo. Collectively, our study indicates that Maackiain treatment inhibits inflammatory response and oxidative stress via activation of AMPK/Nrf2/HO-1 pathway, thus exerting a protective effect against sepsis, providing an alternative option for sepsis prevention.


Assuntos
Proteínas Quinases Ativadas por AMP , Heme Oxigenase-1 , Fator 2 Relacionado a NF-E2 , Pterocarpanos , Sepse , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Heme Oxigenase-1/metabolismo , Lipopolissacarídeos , Proteínas de Membrana , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Pterocarpanos/uso terapêutico , Células RAW 264.7 , Sepse/tratamento farmacológico , Sepse/metabolismo
4.
Oxid Med Cell Longev ; 2022: 3990607, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35126812

RESUMO

Neutrophils release chromatin and antimicrobial proteins to trap and kill microbes, which is termed as neutrophil extracellular trap (NET) formation. NETs play a pivotal role in host defense against infection. However, emerging evidence indicated that NETs also contribute to an exaggerated inflammatory response and organic injuries in sepsis. Zingerone, a natural compound extracted from Zingiber officinale, exerts antioxidant, anti-inflammatory, and antioncogenic properties. In this study, we found that treatment with zingerone reduced organ injury and improved the outcome in a cecal ligation puncture- (CLP-) induced polymicrobial sepsis model. Administration of zingerone also alleviates reactive oxygen species (ROS) accumulation and systematic inflammation in septic mice and inhibits neutrophil extracellular traps (NETs) formation in vivo and in vitro. Furthermore, inhibition of nuclear factor erythroid 2-related factor 2 (Nrf2) with its specific antagonist significantly counteracted the suppressive effects of zingerone on ROS and NETs and retarded the protective role of zingerone against sepsis-associated organ injury. In addition, exposure to zingerone does not affect phagocytic activity of neutrophils in vitro and bacterial dissemination in vivo. Above all, our results indicate that zingerone treatment obviously attenuates NET formation and inflammatory response via Nrf2-mediated ROS inhibition, thus providing a novel therapeutic strategy against sepsis-induced injury.


Assuntos
Armadilhas Extracelulares/metabolismo , Guaiacol/análogos & derivados , Fator 2 Relacionado a NF-E2/metabolismo , Neutrófilos/metabolismo , Substâncias Protetoras/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Sepse/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Animais , Doadores de Sangue , Células Cultivadas , Citocinas/sangue , Modelos Animais de Doenças , Armadilhas Extracelulares/efeitos dos fármacos , Guaiacol/administração & dosagem , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Fagocitose/efeitos dos fármacos , Sepse/sangue , Resultado do Tratamento
5.
Artigo em Inglês | MEDLINE | ID: mdl-34055025

RESUMO

METHODS: Phospho-AMP-activated protein kinase (p-AMPK) and AMP-activated protein kinase (AMPK) were detected by western blot. Immunofluorescence staining was used to validate changes in the levels of nuclear factor kappa B (NF-кB) p65 nuclear translocation. Mice were administered intraperitoneally with calycosin one hour before anaesthesia and endotracheal instillation of PM 2.5. The extent of lung injury was evaluated in the H&E-stained lung sections. Apoptotic cells were detected by TUNEL staining. RESULTS: Administration of calycosin was increased in PM 2.5-treated B2B cells in a dose-dependent manner in vitro. Fluorescence signals from anti-NF-кB p65 were increased in nuclei of cells pretreated with calycosin. The level of p-AMPK was increased by calycosin in vitro and in vivo. After pretreatment with compound C, the inhibitory effects of calycosin on cytotoxicity, levels of inflammatory cytokines and p-AMPK, and levels of NF-кB p65 nuclear translocation were not significantly decreased in vitro or in vivo. CONCLUSIONS: Calycosin effectively decreased the release of inflammatory cytokines and alleviated injury caused by PM 2.5. These effects were mediated through activation of AMPK to suppress NF-κB signalling.

6.
Oncotarget ; 11(34): 3263-3264, 2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32922665

RESUMO

[This corrects the article DOI: 10.18632/oncotarget.11888.].

7.
Front Pharmacol ; 11: 179, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32180727

RESUMO

Clostridium perfringens (C. perfringens) type A strains are the main cause of gas gangrene in humans and animals. Treatment of this lethal disease is limited, and the prognosis is not good. Alpha-toxin (CPA) and perfringolysin O (PFO) secreted by C. perfringens play irreplaceable roles in cytotoxicity to host cells, persistence in host tissues, and lethality of gas gangrene pathology. This work determined the influence of amentoflavone, a biflavonoid isolated from Selaginella tamariscina and other plants, on hemolysis and cytotoxicity mediated by CPA and PFO and evaluated the in vivo therapeutic effect on gas gangrene. Our data showed that amentoflavone could block the hemolysis and cytotoxicity induced by CPA and PFO in vitro, thereby mediating significant protection against mortality of infected mice in a mouse gas gangrene model, efficient bacterial clearance in tissues and alleviation of histological damage in vivo. Based on the above results, amentoflavone may be a potential candidate against C. perfringens infection by reducing CPA and PFO-mediated virulence.

8.
Clin Exp Hypertens ; 42(2): 181-189, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-30957546

RESUMO

Aim: We explored the role of histone modification in the association of depression-hypertension by comparing norepinephrine transporter (NET) gene levels in different depression-hypertensive patients. Then, we analyzed the expression of NET correlation with inflammatory cytokines to provide a new direction for detecting the association mechanism between depression and hypertension.Methods: NE expression levels in serum of diverse groups were detected by enzyme-linked immunosorbent assay. Then histone acetyltransferase (HAT), histone deacetylase (HDAC), H3K27ac, NET, TNF-α, and interleukin-6 (IL-6) were detected by western blot in nine female subjects in different depression and hypertension groups, and Chromatin immunoprecipitation-polymerase chain reaction (Chip-PCR) were used to confirm the degree of acetylation affecting on the transcription level of NET gene. Meanwhile, correlation between NET with TNF/IL-6 was analyzed by SPSS19.0 software program. Finally, Quantitative real-time polymerase chain reaction (qPCR) and western blot were used to detect TNF-α and IL-6 expression levels after NET overexpression or interference treatment in human umbilical vein endothelial cells and Neuro-2a cells.Results: The expression of HAT and H3K27ac had lower levels in D-H and nonD-H group than nonD-nonH group. The results showed that higher acetylation could promote expression of NET genes. Meanwhile, the expression of NET had a significant negative correlation with IL-6 (R = -0.933, p < 0.01) and tumor necrosis factor (TNF) (R = -0.817, p < 0.01) in subjects. In addition, the results confirmed that TNF-α and IL-6 mRNA and protein partial expressions could be inhibited by NET in both HUVECs and Neuronal cells (p < 0.01).Conclusion: In conclusion, differential expression of NET gene might function as an important factor in interaction between depression and hypertension by partially targeting TNF-α and IL-6.


Assuntos
Transtorno Depressivo/etiologia , Hipertensão/etiologia , Interleucina-6/metabolismo , Proteínas da Membrana Plasmática de Transporte de Norepinefrina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Acetilação , Citocinas/metabolismo , Feminino , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos
9.
Neural Regen Res ; 13(10): 1842-1850, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30136701

RESUMO

The mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in the regulation of cell growth, proliferation, differentiation, transformation and death. Mitogen-activated protein kinase phosphatase 1 (MKP1) has an inhibitory effect on the p38MAPK and JNK pathways, but it is unknown whether it plays a role in Aß-induced oxidative stress and neuronal inflammation. In this study, PC12 cells were infected with MKP1 shRNA, MKP1 lentivirus or control lentivirus for 12 hours, and then treated with 0.1, 1, 10 or 100 µM amyloid beta 42 (Aß42). The cell survival rate was measured using the cell counting kit-8 assay. MKP1, tumor necrosis factor-alpha (TNF-α) and interleukin-1ß (IL-1ß) mRNA expression levels were analyzed using quantitative real time-polymerase chain reaction. MKP1 and phospho-c-Jun N-terminal kinase (JNK) expression levels were assessed using western blot assay. Reactive oxygen species (ROS) levels were detected using 2',7'-dichlorofluorescein diacetate. Mitochondrial membrane potential was measured using flow cytometry. Superoxide dismutase activity and malondialdehyde levels were evaluated using the colorimetric method. Lactate dehydrogenase activity was measured using a microplate reader. Caspase-3 expression levels were assessed by enzyme-linked immunosorbent assay. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase dUTP nick end labeling method. MKP1 overexpression inhibited Aß-induced JNK phosphorylation and the increase in ROS levels. It also suppressed the Aß-induced increase in TNF-α and IL-1ß levels as well as apoptosis in PC12 cells. In contrast, MKP1 knockdown by RNA interference aggravated Aß-induced oxidative stress, inflammation and cell damage in PC12 cells. Furthermore, the JNK-specific inhibitor SP600125 abolished this effect of MKP1 knockdown on Aß-induced neurotoxicity. Collectively, these results show that MKP1 mitigates Aß-induced apoptosis, oxidative stress and neuroinflammation by inhibiting the JNK signaling pathway, thereby playing a neuroprotective role.

10.
Oxid Med Cell Longev ; 2018: 8678267, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29765506

RESUMO

6'-O-galloylpaeoniflorin (GPF), a galloylated derivative of paeoniflorin isolated from peony root, has been proven to possess antioxidant potential. In this present study, we revealed that GPF treatment exerted significant neuroprotection of PC12 cells following OGD, as evidenced by a reduction of oxidative stress, inflammatory response, cellular injury, and apoptosis in vitro. Furthermore, treatment with GPF increased the levels of phosphorylated Akt (p-Akt) and nuclear factor-erythroid 2-related factor 2 (Nrf2), as well as promoted Nrf2 translocation in PC12 cells, which could be inhibited by Ly294002, an inhibitor of phosphoinositide 3-kinase (PI3K). In addition, Nrf2 knockdown or Ly294002 treatment significantly attenuated the antioxidant, anti-inflammatory, and antiapoptotic activities of GPF in vitro. In vivo studies indicated that GPF treatment significantly reduced infarct volume and improved neurological deficits in rats subjected to CIRI, as well as decreased oxidative stress, inflammation, and apoptosis, which could be inhibited by administration of Ly294002. In conclusion, these results revealed that GPF possesses neuroprotective effects against oxidative stress, inflammation, and apoptosis after ischemia-reperfusion insult via activation of the PI3K/Akt/Nrf2 pathway.


Assuntos
Isquemia Encefálica/tratamento farmacológico , Compostos Bicíclicos Heterocíclicos com Pontes/uso terapêutico , Glucosídeos/uso terapêutico , Monoterpenos/uso terapêutico , Fármacos Neuroprotetores/uso terapêutico , Células PC12/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Isquemia Encefálica/patologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Linhagem Celular Tumoral , Glucosídeos/farmacologia , Masculino , Monoterpenos/farmacologia , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Ratos Wistar
11.
Neural Regen Res ; 12(11): 1870-1876, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29239334

RESUMO

The major pathological changes in Alzheimer's disease are beta amyloid deposits and cognitive impairment. Calycosin is a typical phytoestrogen derived from radix astragali that binds to estrogen receptors to produce estrogen-like effects. Radix astragali Calycosin has been shown to relieve cognitive impairment induced by diabetes mellitus, suggesting calycosin may improve the cognitive function of Alzheimer's disease patients. The protein kinase C pathway is upstream of the mitogen-activated protein kinase pathway and exerts a neuroprotective effect by regulating Alzheimer's disease-related beta amyloid degradation. We hypothesized that calycosin improves the cognitive function of a transgenic mouse model of Alzheimer's disease by activating the protein kinase C pathway. Various doses of calycosin (10, 20 and 40 mg/kg) were intraperitoneally injected into APP/PS1 transgenic mice that model Alzheimer's disease. Calycosin diminished hippocampal beta amyloid, Tau protein, interleukin-1beta, tumor necrosis factor-alpha, acetylcholinesterase and malondialdehyde levels in a dose-dependent manner, and increased acetylcholine and glutathione activities. The administration of a protein kinase C inhibitor, calphostin C, abolished the neuroprotective effects of calycosin including improving cognitive ability, and anti-oxidative and anti-inflammatory effects. Our data demonstrated that calycosin mitigated oxidative stress and inflammatory responses in the hippocampus of Alzheimer's disease model mice by activating the protein kinase C pathway, and thereby improving cognitive function.

12.
Oncol Rep ; 38(6): 3447-3454, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29039599

RESUMO

The function of miR-592 has been investigated in many types of cancer, however its roles in breast cancer remain unclear. We therefore investigated the biological function and underlying mechanism of miR-592 in breast cancer. In the present study, a marked downregulation of miR-592 was observed in breast cancer tissues and cell lines compared to the matched adjacent non-tumor tissues and normal breast cell line. Statistical analysis revealed that decreased miR-592 was negatively associated with advanced clinical stage, distant metastasis and lymph node metastases. Function analysis demonstrated that overexpression of miR-592 significantly inhibited cell proliferation, clone formation, migration and invasion in breast cancer cells in vitro, as well as suppressed tumor growth in vivo. Furthermore, transforming growth factor ß-2 (TGFß-2), a known oncogene, was identified as a direct target of miR-592, and its mRNA expression level was inversely correlated with the expression level of miR-592 in human breast cancer specimens. Restoration of TGFß-2 expression rescued the inhibitory effect in breast cancer cells caused by miR-592. Collectively, these data suggest that miR-592 may exert it suppressive role in breast cancer, at least in part, by targeting TGFß-2, and that miR-592 may be a novel target for breast cancer treatment.


Assuntos
Neoplasias da Mama/genética , Proliferação de Células/genética , MicroRNAs/genética , Fator de Crescimento Transformador beta1/genética , Adulto , Idoso , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cell Physiol Biochem ; 43(2): 757-767, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28950255

RESUMO

BACKGROUND/AIMS: Lung cancer is one of the most common causes of cancer related deaths worldwide. The role of several microRNAs (miRNAs) including miR-196b in different cancers has already been established. The study was aimed to explore the role of miR-196b in lung cancer and its possible underlying mechanism. METHODS: Human lung cancer cell line A549 was transfected with miR-196b mimic, miR-196b inhibitor and corresponding controls. Then cell viability, migration, invasion, and apoptosis of A549 lung cancer cells either with overexpression or with suppression of miR-196b were estimated sequentially. Next, dual luciferase activity assay was performed to clarify whether Runx2 was a direct target of miR-196b. Finally, the expressions of main factors associated with epithelial mesenchymal transition (EMT), PI3K/AKT/GSK3ß, Smad, and JNK pathways were detected by western blot. RESULTS: MiR-196b expression was significantly decreased in A549, H1650 and H1299 cell lines compared with in WI-38 and HEL-1 cell lines. Overexpression of miR-196b suppressed cell viability, migration, invasion, and induced apoptosis as well as inhibited TGF-ß induced EMT process in A549 cells. In addition, Runx2 was a putative target of miR-196b, and Runx2 silence remarkably increased cell apoptosis and abolished the promotive effects of miR-196b suppression on cell viability, migration and invasion. Finally, miR-196b also mediated its action by inactivation of PI3K/AKT/GSK3ß, Smad, and JNK pathways by down-regulation of Runx2. CONCLUSION: MiR-196b functions as a tumor suppressor that inhibited cell growth and metastasis of lung cancer cells by targeting Runx2. These findings provided further evidences for treatment of lung cancer.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Pulmão/patologia , MicroRNAs/genética , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal , Humanos , Pulmão/metabolismo , Metástase Neoplásica/genética , Metástase Neoplásica/patologia
14.
Afr J Tradit Complement Altern Med ; 14(2): 319-324, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28573248

RESUMO

BACKGROUND: The demand for the chemopreventive drug from the plant source is increasing in recent times, owing to its various biological activities without any adverse effect. The intention of this current study was to examine the anti-glioma effect of Withaferin A (WFA) on C6 glioma cell line model. MATERIALS AND METHODS: C6 glioma cells were administrated with different concentration of WFA (50, 100, 200 and 500 µg/mL) and DMSO (control) group to examine its anti-proliferative, anti-inflammatory and pro-apoptotic activities. RESULTS: Treatment with WFA showed a significant decline in the glioma cell count in a dose-dependent manner and thus proving its anti-proliferative effect. Similarly, inflammatory markers were also substantially lowered upon treatment with different concentration of WFA. However, DNA fragmentation and apoptotic markers like Caspase-3 and 9 were concomitantly enhanced after co-cultured with different concentration of WFA and thus exhibiting its cytotoxicity efficacy. Furthermore, the protein expression of Bcl2 and Bax were markedly downregulated and upregulated respectively; upon treatment with WFA on C6 glioma cells. CONCLUSION: The outcome of this study evidently demonstrates that C6 glioma cells co-cultured with increased concentration of WFA, showed an anti-proliferative, anti-inflammatory and pro-apoptotic effect in a dose-dependent fashion.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Apoptose , Caspases/metabolismo , Glioma/tratamento farmacológico , NF-kappa B/metabolismo , Fitoterapia , Vitanolídeos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Transporte Biológico , Linhagem Celular Tumoral , Proliferação de Células , Fragmentação do DNA/efeitos dos fármacos , Ativação Enzimática , Glioma/metabolismo , Inflamação/etiologia , Inflamação/prevenção & controle , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Withania/química , Vitanolídeos/farmacologia , Proteína X Associada a bcl-2/metabolismo
15.
Biomed Res Int ; 2017: 1893520, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28473977

RESUMO

Osteogenesis from preosteoblasts is important for bone tissue engineering. MicroRNAs are a class of endogenous small RNA molecules that potentially modulate osteogenesis. In this study, we found that miR-155 expression was downregulated in a time-dependent manner in cells of the preosteoblast cell line MC3T3-E1 after osteogenic induction using bone morphogenetic protein 2 (BMP2). Transfection with miR-155 decreased alkaline phosphatase (ALP) activity, ALP expression, and the staining intensity of Alizarin Red in MC3T3-E1 cells treated with BMP2, whereas treatment with miR-155 inhibitor promoted BMP2-induced osteoblast differentiation. The luciferase assay confirmed that miR-155 can bind to the 3' untranslated region of SMAD5 mRNA. miR-155 transfection significantly decreased the expression of SMAD5 protein and mRNA in MC3T3-E1 cells under control media and the p-SMAD5 protein level during osteogenesis. After transfecting cells with the SMAD5 overexpression plasmids, the inhibitory effect of miR-155 on osteogenesis was significantly attenuated. In conclusion, miR-155 inhibited osteoblast differentiation by downregulating the translation of SMAD5 in mouse preosteoblast cells. Inhibition of miR-155 promoted osteogenic potential and thus it can be used as a potential target in the treatment of bone defects.


Assuntos
Doenças Ósseas/genética , MicroRNAs/genética , Osteogênese/genética , Proteína Smad5/genética , Regiões 3' não Traduzidas , Células 3T3 , Fosfatase Alcalina/genética , Animais , Doenças Ósseas/tratamento farmacológico , Proteína Morfogenética Óssea 2/genética , Diferenciação Celular/genética , Regulação da Expressão Gênica/genética , Humanos , Camundongos , Osteoblastos/metabolismo , Transfecção
16.
Environ Toxicol Pharmacol ; 50: 192-199, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28192748

RESUMO

Exposure to particulate matter (PM) with an aerodynamic diameter≤2.5µm (PM2.5) induces reactive oxygen species (ROS) and pro-inflammatory cytokine production, leading to airway epithelial injury. However, the mechanisms underlying the toxicity of PM2.5 have not been clarified. Here, we show that exposure to PM2.5 induces sustained activation of the nuclear factor kappa B (NF-κB) signaling in human airway epithelial Beas-2B (B2B) cells. In addition, PM2.5 exposure significantly decreased miR-331 expression in B2B cells, which was abrogated by inhibition of ROS or phosphoinositide 3-kinase (PI3K)/Akt pathway. Induction of miR-331 overexpression attenuated the PM2.5 exposure-induced NF-kBp65 nuclear translocation, IL-6 and IL-8 expression in B2B cells. Furthermore, miR-331 targeted the inhibitor of NF-κB kinase beta (IKK-ß) by down-regulating the IKK-ß-regulated luciferase activity in HEK293 cells. Moreover, induction of miR-331 over-expression inhibited IKK-ß expression while induction of IKK-ß over-expression prevented the inhibition of miR-331 on the PM2.5 exposure-induced NF-kBp65 nuclear translocation, IL-6 and IL-8 expression in B2B cells. Therefore, PM2.5 exposure decreased miR-331 expression via the ROS/PI3K/Akt pathway, resulting in an increase in the IKK-ß expression and sustained NF-κB activation in human airway epithelial cells. Our findings may provide new insights into the molecular mechanisms underlying the toxicity of PM2.5 exposure and aid in design of new therapeutic strategies to prevent PM2.5-induced toxicity.


Assuntos
Células Epiteliais/efeitos dos fármacos , MicroRNAs/genética , NF-kappa B/metabolismo , Material Particulado/toxicidade , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Quinase I-kappa B/genética , NF-kappa B/genética , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
17.
Oncotarget ; 7(41): 66989-67003, 2016 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-27611950

RESUMO

This study aimed to explore the effects of miR-21 and PTEN/Akt signaling pathway on TGF-ß1-induced epithelial-mesenchymal transition (EMT) in gastric cancer (GC). GC tissues and adjacent tissues were collected from 83 patients. The qRT-PCR assay was performed to detect miR-21 expression. The expressions of PTEN, Akt and p-Akt were detected by immunohistochemistry. After 48 h of treatment with TGF-ß1 (10 ng/mL), the SGC-7901 and KATO-III cells were divided into the blank, negative control (NC), miR-21 inhibitors, PTEN-siRNA and miR-21 inhibitors + PTEN-siRNA groups. EMT related factors and PTEN expressions were detected by qRT-PCR assay and Western blotting. The scratch test was conducted to observe cell migration. Xenograft tumor model in nude mice was used to evaluate the effects of miR-21 on EMT of GC cells in vivo. In GC tissues, the expressions of miR-21, Akt and p-Akt were up-regulated, while PTEN expression was down-regulated. Gene and protein expressions of E-cadherin and PTEN in the miR-21 inhibitors group were higher than the blank, NC, PTEN-siRNA and miR-21 inhibitors + PTEN-siRNA groups, while the expressions of N-cadherin, ß-catenin, Vimentin and Slug in the miR-21 inhibitors group were lower than other groups. MiR-21 inhibitors significantly inhibit cell migration and invasion in GC cell lines. In vivo xenograft experiment revealed that miR-21 inhibitor inhibits the growth of transplanted tumor through up-regulating E-cadherin and PTEN expressions and down-regulating the expressions of N-cadherin, ß-catenin, Vimentin and Slug. These results suggest that miR-21 could promote TGF-ß1-induced EMT in GC cells through up-regulating PTEN expression.


Assuntos
Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , MicroRNAs/genética , PTEN Fosfo-Hidrolase/biossíntese , Neoplasias Gástricas/patologia , Adulto , Idoso , Animais , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Regulação para Cima
18.
Tumour Biol ; 37(6): 8317-25, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26729197

RESUMO

To verify the hypothesis that upregulation of microRNA-31 (miR-31) targeting integrin α5 (ITGA5) suppresses tumor cell invasion and metastasis by indirectly regulating phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway in human SGC7901 gastric cancer (GC) cells. The miRTarBase was used to predict whether ITGA5 is the target gene of miR-31, which was further confirmed by luciferase reporter gene assay. The SGC7901 GC cells were divided into five groups including the blank, miR-31 mimic, miR-31 mimic control, miR-31 inhibitor, and miR-31 inhibitor control groups. Reverse transcriptase-polymerase chain reaction (RT-PCR), western blotting, cell scratch test, and transwell assays were respectively performed in our study. TGA5 was found as the target gene of miR-31. The RT-PCR detection revealed that, compared with the blank group, ITGA5 messenger RNA (mRNA) expression decreased in the miR-31 mimic group, but increased in the miR-31 inhibitor group. The western blotting examination suggested that the expressions of ITGA5, PI3K, and AKT proteins reduced in the miR-31 mimic group, but enhanced in the miR-31 inhibitor group when compared to the blank group, respectively. The cell scratch and transwell assays indicated that the miR-31 expressions were negatively associated with GC cell migration and invasion. Besides, RT-PCR combined with western blotting demonstrated that the miR-31 expressions were higher in the normal tissues than those in the GC tissues, while the ITGA5 mRNA and protein showed lower expression in the normal tissues than they did in the GC tissues. Our study concluded that upregulation of miR-31 targeting ITGA5 may suppress tumor cell invasion and metastasis by indirectly regulating PI3K/AKT signaling pathway in human SGC7901 GC cells.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Integrina alfa5/metabolismo , MicroRNAs/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/genética , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Feminino , Humanos , Masculino , Invasividade Neoplásica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Regulação para Cima
19.
Tumour Biol ; 37(3): 2891-900, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26409449

RESUMO

Lung carcinoma is a deadly malignant disease with poor prognosis and increasing incidence in recent years. However, the molecular mechanism underlying the initiation and progression of lung cancer is still not completely elucidated. Recently, myocyte enhancer factor 2D (MEF2D) has been reported to promote the growth of liver cancer, but its implication in lung cancer is still unknown. This study is aimed to determine the role of MEF2D in lung carcinoma. Quantitative PCR (qPCR) and immunoblot assays showed that MEF2D was overexpressed in lung cancer tissues and cell lines, compared with the matched normal tissues and cell lines. Small interfering RNA (siRNA) suppression of MEF2D was able to reduce the proliferation, survival, and invasion of lung carcinoma cells. The transfection of MEF2D-expressing constructs into normal lung fibroblast cells promoted their proliferation and motility. The role of MEF2D in the growth of lung cancer was also confirmed in mice. Further study revealed that miR-218, which was underexpressed in lung carcinoma, was predicted to bind the 3'-untranslated region (UTR) of MEF2D mRNA. miR-218 was shown to suppress the activity of luciferase with MEF2D 3'-UTR. The changes in miR-218 levels affected the expression of MEF2D in lung cancer cells and normal fibroblast cells. There is also an inverse association between miR-218 abundance and MEF2D levels in the lung carcinoma specimen. Furthermore, the transfection of a plasmid that expressed MEF2D resistance to miR-218 regulation abolished the inhibitory effect of miR-218 on lung cancer cells. Collectively, MEF2D overexpression participated in the growth of lung cancers and its aberrant expression may result from the reduction of tumor suppressor miR-218.


Assuntos
Neoplasias Pulmonares/patologia , MicroRNAs/fisiologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Fatores de Transcrição MEF2/análise , Fatores de Transcrição MEF2/genética , Fatores de Transcrição MEF2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Invasividade Neoplásica
20.
Neural Regen Res ; 9(1): 41-50, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25206742

RESUMO

Transforming growth factor-beta (TGF-ß) type II receptor (TßRII) levels are extremely low in the brain tissue of patients with Alzheimer's disease. This receptor inhibits TGF-ß1/SMAD signaling and thereby aggravates amyolid-beta deposition and neuronal injury. Dab2, a specific adapter protein, protects TßRII from degradation and ensures the effective conduction of TGF-ß1/SMAD signaling. In this study, we used an adenoviral vector to overexpress the Dab2 gene in the mouse hippocampus and investigated the regulatory effect of Dab2 protein on TGF-ß1/SMAD signaling in a mouse model of Alzheimer's disease, and the potential neuroprotective effect. The results showed that the TßRII level was lower in APP/PS1 mouse hippocampus than in normal mouse hippocampus. After Dab2 expression, hippocampal TßRII and p-SMAD2/3 levels were significantly increased, while amyloid-beta deposition, microglia activation, tumor necrosis factor-α and interleulin-6 levels and neuronal loss were significantly attenuated in APP/PS1 mouse brain tissue. These results suggest that Dab2 can exhibit neuroprotective effects in Alzheimer's disease by regulating TGF-ß1/SMAD signaling.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...