Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
2.
Int J Med Sci ; 19(10): 1557-1566, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36185329

RESUMO

Extra-proliferation and increased migration of vascular smooth cells con-tribute to the formation of atherosclerosis. Ras small G proteins play a critical role in the prolif-eration and migration of a wide range of cells. Mulberry, an economic fruit in Asia, exhibits anti-inflammation, anti-migration, and anti-oxidant properties. The mechanisms of action of mulberry extracts on K-Ras small G protein-induced proliferation and migration of vascular smooth muscle cell have not been extensively investigated. In this study, we explored the effects of mulberry polyphenol extracts (MPE) on the proliferation and migration of K-Ras-overexpressing A7r5 smooth muscle cells. The overexpression of K-Ras enhanced the ex-pression and activity of matrix metalloproteinase (MMP)-2, promoted vascular endothelial growth factor (VEGF) production, and eventually triggered the migration of A7r5 cells. Treatment with MPE attenuated K-Ras-induced phenomenon. In addition, MPE blocked K-Ras-induced actin fibril stress. MPE dose-dependently diminished K-Ras-induced Rho A, Rac1, CDC42, and phosphorylated focal adhesion kinase (FAK) expression. MPE elevated Rho B ex-pression. Phosphorylated AKT and glycogen synthase kinase (GSK) induced by K-Ras were also repressed by MPE treatment. MPE enhanced the interaction of IκB with NFκB. MPE restored the G0/G1 population and p21 and p27 expressions, which were repressed by K-Ras. Finally, MPE triggered the degradation of K-Ras by ubiquitination. MPE inhibited the migration and proliferation of vascular smooth cell through K-Ras-induced pathways and eventually pre-vented atherosclerosis.


Assuntos
Aterosclerose , Proteínas Monoméricas de Ligação ao GTP , Morus , Actinas/metabolismo , Antioxidantes/farmacologia , Aterosclerose/tratamento farmacológico , Aterosclerose/genética , Aterosclerose/metabolismo , Movimento Celular/genética , Proliferação de Células/genética , Células Cultivadas , Regulação para Baixo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Frutas/metabolismo , Quinases da Glicogênio Sintase/metabolismo , Humanos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Proteínas Monoméricas de Ligação ao GTP/farmacologia , Músculo Liso Vascular , Miócitos de Músculo Liso , Polifenóis/metabolismo , Polifenóis/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
J Invest Dermatol ; 142(7): 1882-1892.e5, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34883044

RESUMO

The upregulation of the adaptor protein NUMB triggers melanocytic differentiation from multipotent skin stem cells, which share many properties with aggressive melanoma cells. Although NUMB acts as a tumor suppressor in various human cancer types, little is known about its role in melanoma. In this study, we investigated the role of NUMB in melanoma progression and its regulatory mechanism. Analysis of The Cancer Genome Atlas melanoma datasets revealed that high NUMB expression in melanoma tissues correlates with improved patient survival. Moreover, NUMB expression is downregulated in metastatic melanoma cells. NUMB knockdown significantly increased the invasion potential of melanoma cells in a three-dimensional collagen matrix in vitro and in the lungs of a mouse model in vivo; it also significantly upregulated the expression of the NOTCH target gene CCNE. Previous studies suggested that Wnt signaling increases NUMB expression. By mimicking Wnt stimulation through glycogen synthase kinase-3 inhibition, we increased NUMB expression in melanoma cells. Furthermore, a glycogen synthase kinase-3 inhibitor reduced the invasion of melanoma cells in a NUMB-dependent manner. Together, our results suggest that NUMB suppresses invasion and metastasis in melanoma, potentially through its regulation of the NOTCH‒CCNE axis and that the inhibitors that upregulate NUMB can exert therapeutic effects in melanoma.


Assuntos
Melanoma , Proteínas de Membrana , Proteínas do Tecido Nervoso , Animais , Linhagem Celular Tumoral , Quinases da Glicogênio Sintase/metabolismo , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Via de Sinalização Wnt
4.
Mol Med Rep ; 21(5): 2202-2208, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32323765

RESUMO

The plant extract piperine is used as a traditional Chinese medicine due to its anti­inflammatory effects and efficacy against numerous types of cancer. The aim of the present study was to investigate the antitumor mechanism of piperine in human osteosarcoma U2OS and 143B cell lines. The effects of piperine on cell apoptosis and invasion of human osteosarcoma cells were assessed using flow cytometry and Transwell assays. Moreover, western blotting was used to measure the effects of piperine on the protein expression levels of the metastasis markers matrix metalloproteinase­2 (MMP­2) and vascular endothelial growth factor (VEGF). In addition, the involvement of the Wnt/ß­catenin signaling pathway in modulating the effects of piperine was examined via western blot analysis. The results of MTT and Transwell invasion assays indicated that piperine treatment dose­dependently reduced U2OS and 143B cell viability and invasion. Furthermore, a significant reduction was identified in MMP­2, VEGF, glycogen synthase kinase­3ß and ß­catenin protein expression levels, as well as the expression levels of their target proteins cyclooxygenase­2, cyclin D1 and c­myc, in U2OS cells after piperine treatment. In addition, similar results were observed in 143B cells. Therefore, the present study demonstrated the efficacy of piperine in osteosarcoma, and identified that the Wnt/ß­catenin signaling pathway may modulate the antitumor effects of piperine on human U2OS and 143B cells.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzodioxóis/farmacologia , Proliferação de Células/efeitos dos fármacos , Osteossarcoma/tratamento farmacológico , Piperidinas/farmacologia , Alcamidas Poli-Insaturadas/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular , Ciclina D1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citometria de Fluxo , Quinases da Glicogênio Sintase/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , beta Catenina/metabolismo
5.
J Biol Chem ; 294(49): 18836-18845, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31690628

RESUMO

Heterotrimeric G proteins are important molecular switches that facilitate transmission of a variety of signals from the outside to the inside of cells. G proteins are highly conserved, enabling study of their regulatory mechanisms in model organisms such as the budding yeast Saccharomyces cerevisiae Gpa2 is a yeast Gα protein that functions in the nutrient signaling pathway. Using Phos-tag, a highly specific phosphate binding tag for separating phosphorylated proteins, we found that Gpa2 undergoes phosphorylation and that its level of phosphorylation is markedly increased upon nitrogen starvation. We also observed that phosphorylation of Gpa2 depends on glycogen synthase kinase (GSK). Disrupting GSK activity diminishes Gpa2 phosphorylation levels in vivo, and the purified GSK isoforms Mck1 and Ygk3 are capable of phosphorylating Gpa2 in vitro Functionally, phosphorylation enhanced plasma membrane localization of Gpa2 and promoted nitrogen starvation-induced activation of protein kinase A. Together, the findings of our study reveal a mechanism by which GSK- and nutrient-dependent phosphorylation regulates subcellular localization of Gpa2 and its ability to activate downstream signaling.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Membrana Celular/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Regulação Fúngica da Expressão Gênica , Quinases da Glicogênio Sintase/genética , Quinases da Glicogênio Sintase/metabolismo , Fosforilação , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transdução de Sinais
6.
Sci Rep ; 9(1): 10049, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31296906

RESUMO

Cancer stem cells are associated with chemoresistance and rapid recurrence of malignant tumors, including glioblastoma (GBM). Although temozolomide (TMZ) is the most effective drug treatment for GBM, GBM cells acquire resistance and become refractory to TMZ during treatment. Therefore, glioma stem cell (GSC)-targeted therapy and TMZ-enhancing therapy may be effective approaches to improve GBM prognosis. Many drugs that suppress the signaling pathways that maintain GSC or enhance the effects of TMZ have been reported. However, there are no established therapies beyond TMZ treatment currently in use. In this study, we screened drug libraries composed of 1,301 existing drugs using cell viability assays to evaluate effects on GSCs, which led to selection of kenpaullone, a kinase inhibitor, as a TMZ enhancer targeting GSCs. Kenpaullone efficiently suppressed activity of glycogen synthase kinase (GSK) 3ß. Combination therapy with kenpaullone and TMZ suppressed stem cell phenotype and viability of both GSCs and glioma cell lines. Combination therapy in mouse models significantly prolonged survival time compared with TMZ monotherapy. Taken together, kenpaullone is a promising drug for treatment of GBM by targeting GSCs and overcoming chemoresistance to TMZ.


Assuntos
Benzazepinas/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Quimioterapia Adjuvante/métodos , Glioblastoma/tratamento farmacológico , Quinases da Glicogênio Sintase/metabolismo , Indóis/uso terapêutico , Células-Tronco Neoplásicas/patologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Glioblastoma/patologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Transdução de Sinais , Temozolomida/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Cell Death Dis ; 10(6): 448, 2019 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-31171769

RESUMO

Aberrant microRNA-708 (miR-708) expression is frequently reported in cancer studies; however, its role in glioma has not been examined in detail. We investigated miR-708 function in glioma and revealed that miR-708 expression was significantly down-regulated in glioma tissues and cell lines. Restoration of miR-708 inhibited glioma cell growth and invasion both in vitro and in vivo. The oncogene SPHK2 (sphingosine kinase 2) was identified as a downstream target of miR-708 using luciferase and western blot assays. miR-708 inhibited AKT/ß-catenin signaling, which is activated by SPHK2. In addition, we revealed that miR-708 was transcriptionally repressed by EZH2 (enhancer of zeste homolog 2)-induced histone H3 lysine 27 trimethylation and promoter methylation. In summary, our findings revealed that miR-708 is a glioma tumor suppressor and suggest that miR-708 is a potential therapeutic target for glioma patients.


Assuntos
Neoplasias Encefálicas/metabolismo , Transição Epitelial-Mesenquimal/genética , Glioma/metabolismo , MicroRNAs/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , beta Catenina/metabolismo , Animais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica/genética , Glioma/enzimologia , Glioma/genética , Quinases da Glicogênio Sintase/química , Quinases da Glicogênio Sintase/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Metilação , Camundongos , Camundongos Nus , MicroRNAs/genética , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Prognóstico , Proteínas Proto-Oncogênicas c-akt/química , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/genética , Transplante Heterólogo , beta Catenina/genética
8.
J Neurol Sci ; 402: 121-130, 2019 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31129265

RESUMO

Several kinases have been implicated in the pathogenesis of Parkinson's disease (PD), most notably leucine-rich repeat kinase 2 (LRRK2), as LRRK2 mutations are the most common genetic cause of a late-onset parkinsonism that is clinically indistinguishable from sporadic PD. More recently, several other kinases have emerged as promising disease-modifying targets in PD based on both preclinical studies and clinical reports on exenatide, the urate precursor inosine, nilotinib and lithium use in PD patients. These kinases include protein kinase B (Akt), glycogen synthase kinases-3ß and -3α (GSK-3ß and GSK-3α), c-Abelson kinase (c-Abl) and cyclin-dependent kinase 5 (cdk5). Activities of each of these kinases are involved either directly or indirectly in phosphorylating tau or increasing α-synuclein levels, intracellular proteins whose toxic oligomeric forms are strongly implicated in the pathogenesis of PD. GSK-3ß, GSK-3α and cdk5 are the principle kinases involved in phosphorylating tau at sites critical for the formation of tau oligomers. Exenatide analogues, urate, nilotinib and lithium have been shown to affect one or more of the above kinases, actions that can decrease the formation and increase the clearance of intraneuronal phosphorylated tau and α-synuclein. Here we review the current preclinical and clinical evidence supporting kinase-targeting agents as potential disease-modifying therapies for PD patients enriched with these therapeutic targets and incorporate LRRK2 physiology into this novel model.


Assuntos
Antiparkinsonianos/uso terapêutico , Quinase 5 Dependente de Ciclina/metabolismo , Quinases da Glicogênio Sintase/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Doença de Parkinson/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Humanos , Doença de Parkinson/tratamento farmacológico
9.
Semin Cancer Biol ; 56: 25-36, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-29309927

RESUMO

Glycogen synthase kinase-3 (GSK-3), a serine/threonine kinase is an archetypal multifunctional moonlighting protein involved in diverse cellular processes including metabolism, insulin signaling, proliferation, differentiation, apoptosis, neuronal function and embryonic development. The two known isoforms, GSK-3α and GSK-3ß that undergo activation/inactivation by post-translational, site-specific phosphorylation incorporate a vast number of substrates in their repertoire. Dysregulation of GSK-3 has been linked to diverse disease entities including cancer. The role of GSK-3 in cancer is paradoxical and enigmatic. The enzyme functions as a tumour promoter or suppressor based on the context, cell type and phosphorylation status. GSK-3 is the central hub that orchestrates signals from the Wnt/ß-catenin, PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, hedgehog, Notch and TP53 pathways to elicit regulatory influences on cancer initiation, epithelial-mesenchymal transition, and resistance to therapy. As a direct target of several microRNAs, GSK-3 influences hallmark attributes of cancer, cancer stemness and treatment resistance. There is overwhelming evidence to indicate that GSK-3 is aberrantly regulated in different cancer types. Consequently, GSK-3 has emerged as a potential therapeutic target in cancer. A plethora of natural and synthetic GSK-3 modulators have been discovered and the number of patents published for GSK-3 inhibitors has also been steadily increasing in recent years. This review focuses on the intricate interactions between GSK-3 and oncogenic signalling circuits as well as the feasibility of targeting GSK-3 for the treatment of cancer.


Assuntos
Quinases da Glicogênio Sintase/genética , Quinases da Glicogênio Sintase/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos , Biomarcadores Tumorais , Suscetibilidade a Doenças , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/química , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Quinases da Glicogênio Sintase/antagonistas & inibidores , Quinases da Glicogênio Sintase/química , Humanos , Isoenzimas , MicroRNAs/genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
10.
Ann Rheum Dis ; 77(7): 1053-1062, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29431119

RESUMO

OBJECTIVES: Accelerated atherosclerotic disease typically complicates rheumatoid arthritis (RA), leading to premature cardiovascular death. Inflammatory macrophages are key effector cells in both rheumatoid synovitis and the plaques of coronary artery disease (CAD). Whether both diseases share macrophage-dependent pathogenic mechanisms is unknown. METHODS: Patients with RA or CAD (at least one myocardial infarction) and healthy age-matched controls were recruited into the study. Peripheral blood CD14+ monocytes were differentiated into macrophages. Metabolic profiles were assessed by Seahorse Analyzer, intracellular ATP concentrations were quantified and mitochondrial protein localisation was determined by confocal image analysis. RESULTS: In macrophages from patients with RA or CAD, mitochondria consumed more oxygen, generated more ATP and built tight interorganelle connections with the endoplasmic reticulum, forming mitochondria-associated membranes (MAM). Calcium transfer through MAM sites sustained mitochondrial hyperactivity and was dependent on inactivation of glycogen synthase kinase 3b (GSK3b), a serine/threonine kinase functioning as a metabolic switch. In patient-derived macrophages, inactivated pGSK3b-Ser9 co-precipitated with the mitochondrial fraction. Immunostaining of atherosclerotic plaques and synovial lesions confirmed that most macrophages had inactivated GSK3b. MAM formation and GSK3b inactivation sustained production of the collagenase cathepsin K, a macrophage effector function closely correlated with clinical disease activity in RA and CAD. CONCLUSIONS: Re-organisation of the macrophage metabolism in patients with RA and CAD drives unopposed oxygen consumption and ultimately, excessive production of tissue-destructive enzymes. The underlying molecular defect relates to the deactivation of GSK3b, which controls mitochondrial fuel influx and as such represents a potential therapeutic target for anti-inflammatory therapy.


Assuntos
Artrite Reumatoide/patologia , Doença da Artéria Coronariana/patologia , Quinases da Glicogênio Sintase/metabolismo , Macrófagos/metabolismo , Sinovite/patologia , Pesquisa Translacional Biomédica , Artrite Reumatoide/sangue , Artrite Reumatoide/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/fisiopatologia , Feminino , Humanos , Macrófagos/enzimologia , Masculino , Pessoa de Meia-Idade , Tamanho Mitocondrial/fisiologia , Monócitos/metabolismo , Consumo de Oxigênio/fisiologia , Fatores de Risco , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Sinovite/metabolismo
11.
Kaohsiung J Med Sci ; 34(3): 142-149, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29475461

RESUMO

This study examined the effects of eugenosedin-A (Eu-A) in a streptozotocin (STZ)/nicotinamide-induced rat model of type II diabetes mellitus (T2DM). Six-week-old Sprague-Dawley rats were randomly divided into three groups: (1) RD group, normal rats fed a regular diet (RD), (2) DM group, T2DM rats fed a high-fat diet, and (3) Eu-A group, T2DM rats fed a high fat diet plus oral Eu-A (5 mg/kg/day). After 30 days, the DM group had higher body weight, higher blood glucose and lower insulin levels than the RD group. The DM group also had increased protein expression of glycogen synthase kinase (GSK) in liver and skeletal muscle and decreased protein expression of insulin receptor (IR), insulin receptor substrate-1 (IRS-1), IRS-2, AMP-activated protein kinase (AMPK), glucose transporter-4 (GLUT-4), glucokinase (GCK), and peroxisome proliferator-activated receptor γ (PPAR-γ). STZ/nicotinamide-induced T2DM increased the expression of mitogen-activated protein kinases (MAPKs: p38, ERK, JNK) and inflammatory p65 protein. In the Eu-A treated T2DM rats, however, blood glucose was attenuated and the insulin concentration stimulated. Changes in IR, IRS-1 and IRS-2 proteins as well as AMPK, GLUT-4, GCK, GSK, PPAR-γ, MAPKs, and inflammatory p65 proteins were ameliorated. These results suggested that Eu-A alleviates STZ/nicotinamide-induced hyperglycemia by improving insulin levels and glucose metabolism, and inhibiting the MAPKs- and p65-mediated inflammatory pathway.


Assuntos
Diabetes Mellitus Experimental/tratamento farmacológico , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/farmacologia , Proteínas Quinases Ativadas por Mitógeno/genética , Piperazinas/farmacologia , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Glucoquinase/genética , Glucoquinase/metabolismo , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Quinases da Glicogênio Sintase/genética , Quinases da Glicogênio Sintase/metabolismo , Hiperglicemia/induzido quimicamente , Hiperglicemia/genética , Hiperglicemia/patologia , Insulina/sangue , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Niacinamida , PPAR gama/genética , PPAR gama/metabolismo , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transdução de Sinais , Estreptozocina , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo
12.
J Med Food ; 21(4): 380-389, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29271701

RESUMO

Red ginseng oil (RGO) has been shown to possess anti-inflammatory and hepatoprotective activity. In this study, we evaluated the inhibitory effect of RGO on 12-O-tetradecanoylphorbol-13-acetate (TPA)-stimulated neoplastic transformation of JB6 P+ cells. RGO pretreatment abolished the transformation of JB6 P+ cells challenged by TPA. RGO suppressed the transactivation of activator protein-1 (AP-1) and nuclear factor kappa B (NF-κB) transcription factors as well as protein levels of cyclooxygenase-2, cyclin D1, cyclin E, and Bcl-2 in the TPA-treated cells. Additionally, TPA-induced phosphorylations of extracellular signal-regulated kinases, 90 kDa ribosomal S6 kinase 2, c-Jun N-terminal kinases, and glycogen synthase kinase 3ß were downregulated in the presence of RGO. Furthermore, RGO induced the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated antioxidant enzyme heme oxygenase-1 (HO-1) expression, and effectively blocked the overproduction of TPA-induced reactive oxygen species. These results suggest that RGO exerts a potent chemopreventive activity in skin cell model.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Epiderme/efeitos dos fármacos , Panax/química , Óleos de Plantas/farmacologia , Neoplasias Cutâneas/metabolismo , Pele/efeitos dos fármacos , Animais , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Linhagem Celular , Transformação Celular Neoplásica/induzido quimicamente , Transformação Celular Neoplásica/metabolismo , Ciclinas/metabolismo , Ciclo-Oxigenase 2/metabolismo , Epiderme/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinases da Glicogênio Sintase/metabolismo , Heme Oxigenase-1/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Óleos de Plantas/uso terapêutico , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/prevenção & controle , Acetato de Tetradecanoilforbol , Fator de Transcrição AP-1/metabolismo
13.
Mol Cell Biochem ; 441(1-2): 89-98, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28887744

RESUMO

Primary liver cancer (hepatocellular carcinoma, HCC) is a leading cause of cancer-related deaths, and alternative ways to treat this disease are urgently needed. In recent years, novel approaches to cancer treatment have been based on microRNAs, small non-coding RNA molecules that play a crucial role in cancer progression by regulating gene expression. Overexpression of some microRNAs has shown therapeutic potential, but whether or not this was the case for microRNA-203 (miR-203) in liver cancer was unknown. Therefore, the aim of this study was to investigate the effect of miR-203 overexpression in liver cancer and explore the related mechanisms. Liver cancer cells from the HepG2 and Hep3B cell lines were transfected with either miR-203 mimics or negative control RNA, and then the cells were subjected to cell viability, cell proliferation, and Western blotting assays. As a result of microRNA-203 overexpression, HepG2 and Hep3B cell viability and cell proliferation significantly declined. Furthermore, microRNA-203 overexpression led to inhibited expression of phosphatidylinositol-4,5-bisphosphate 3-kinase (PIK3)/protein kinase B (Akt), c-Jun, and p38 mitogen-activated protein kinases (p38 MAPK), and restored glycogen synthase kinase 3 (GSK 3) activity in HepG2 cells. Our results suggest that c-Jun, p38 MAPK, PIK3CA/Akt, and GSK3 signaling involved in the effect of miR-203 on the proliferation of HCC cells.


Assuntos
Carcinoma Hepatocelular/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Quinases da Glicogênio Sintase/metabolismo , Neoplasias Hepáticas/metabolismo , Sistema de Sinalização das MAP Quinases , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , RNA Neoplásico/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Carcinoma Hepatocelular/genética , Proliferação de Células , Classe I de Fosfatidilinositol 3-Quinases/genética , Quinases da Glicogênio Sintase/genética , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Proteínas Proto-Oncogênicas c-jun/genética , RNA Neoplásico/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
14.
Artigo em Inglês | MEDLINE | ID: mdl-29250491

RESUMO

Porphyromonas gingivalis is a host-adapted oral pathogen associated with chronic periodontitis that successfully survives and persists in the oral epithelium. Recent studies have positively correlated periodontitis with increased risk and severity of oral squamous cell carcinoma (OSCC). Intriguingly, the presence of P. gingivalis enhances tumorigenic properties independently of periodontitis and has therefore been proposed as a potential etiological agent for OSCC. However, the initial host molecular changes induced by P. gingivalis infection which promote predisposition to cancerous transformation through EMT (epithelial-mesenchymal-transition), has never been studied in human primary cells which more closely mimic the physiological state of cells in vivo. In this study, we examine for the first time in primary oral epithelial cells (OECs) the expression and activation of key EMT mediators during long-term P. gingivalis infection in vitro. We examined the inactive phosphorylated state of glycogen synthase kinase-3 beta (p-GSK3ß) over 120 h P. gingivalis infection and found p-GSK3ß, an important EMT regulator, significantly increases over the course of infection (p < 0.01). Furthermore, we examined the expression of EMT-associated transcription factors, Slug, Snail, and Zeb1 and found significant increases (p < 0.01) over long-term P. gingivalis infection in protein and mRNA expression. Additionally, the protein expression of mesenchymal intermediate filament, Vimentin, was substantially increased over 120 h of P. gingivalis infection. Analysis of adhesion molecule E-cadherin showed a significant decrease (p < 0.05) in expression and a loss of membrane localization along with ß-catenin in OECs. Matrix metalloproteinases (MMPs) 2, 7, and 9 are all markedly increased with long-term P. gingivalis infection. Finally, migration of P. gingivalis infected cells was evaluated using scratch assay in which primary OEC monolayers were wounded and treated with proliferation inhibitor, Mitomycin C. The cellular movement was determined by microscopy. Results displayed P. gingivalis infection promoted cell migration which was slightly enhanced by co-infection with Fusobacterium nucleatum, another oral opportunistic pathogen. Therefore, this study demonstrates human primary OECs acquire initial molecular/cellular changes that are consistent with EMT induction during long-term infection by P. gingivalis and provides a critically novel framework for future mechanistic studies.


Assuntos
Células Epiteliais/microbiologia , Mesoderma/microbiologia , Fenótipo , Porphyromonas gingivalis/patogenicidade , Apoptose , Caderinas/metabolismo , Carcinoma de Células Escamosas/etiologia , Linhagem Celular Tumoral , Movimento Celular , Coinfecção , Fusobacterium nucleatum/patogenicidade , Regulação da Expressão Gênica , Quinases da Glicogênio Sintase/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Mitomicina/farmacologia , Neoplasias Bucais/microbiologia , Infecções Oportunistas/complicações , Periodontite/complicações , Periodontite/microbiologia , Fatores de Transcrição da Família Snail/genética , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Tempo , Fatores de Transcrição/genética , Vimentina/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , beta Catenina/metabolismo
15.
Biochemistry (Mosc) ; 81(7): 739-47, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27449620

RESUMO

Increasing evidence has shown that pseudogenes can widely regulate gene expression. However, little is known about the specific role of PTENP1 and miR-499-5p in insulin resistance. The relative transcription level of PTENP1 was examined in db/db mice and high fat diet (HFD)-fed mice by real-time PCR. To explore the effect of PTENP1 on insulin resistance, adenovirus overexpressing or inhibiting vectors were injected through the tail vein. Bioinformatics predictions and a luciferase reporter assay were used to explore the interaction between PTENP1 and miR-499-5p. The relative transcription level of PTENP1 was largely enhanced in db/db mice and HFD-fed mice. Furthermore, the overexpression of PTENP1 resulted in impaired Akt/GSK activation as well as glycogen synthesis, while PTENP1 inhibition led to the improved activation of Akt/GSK and enhanced glycogen contents. More importantly, PTENP1 could directly bind miR-499-5p, thereby becoming a sink for miR-499-5p. PTENP1 overexpression results in the impairment of the insulin-signaling pathway and may function as a competing endogenous RNA for miR-499-5p, thereby contributing to insulin resistance.


Assuntos
MicroRNAs/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Pseudogenes/genética , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Sítios de Ligação , Western Blotting , Linhagem Celular , Dieta Hiperlipídica , Genes Reporter , Glicogênio/biossíntese , Quinases da Glicogênio Sintase/metabolismo , Insulina/metabolismo , Resistência à Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , MicroRNAs/genética , Oligonucleotídeos Antissenso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Alinhamento de Sequência
16.
Sci Rep ; 6: 26277, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27195613

RESUMO

Retrospective studies indicate that the use of regional anesthesia can reduce cancer recurrence after surgery which could be due to ranging from immune function preservation to direct molecular mechanisms. This study was to investigate the effects of bupivacaine on ovarian and prostate cancer cell biology and the underlying molecular mechanisms. Cell viability, proliferation and migration of ovarian carcinoma (SKOV-3) and prostate carcinoma (PC-3) were examined following treatment with bupivacaine. Cleaved caspase 3, 8 and 9, and GSK-3ß, pGSK-3ß(tyr216) and pGSK-3ß(ser9) expression were assessed by immunofluorescence. FAS ligand neutralization, caspase and GSK-3 inhibitors and GSK-3ß siRNA were applied to further explore underlying mechanisms. Clinically relevant concentrations of bupivacaine reduced cell viability and inhibited cellular proliferation and migration in both cell lines. Caspase 8 and 9 inhibition generated partial cell death reversal in SKOV-3, whilst only caspase 9 was effective in PC-3. Bupivacaine increased the phosphorylation of GSK-3ß(Tyr216) in SKOV-3 but without measurable effect in PC3. GSK-3ß inhibition and siRNA gene knockdown decreased bupivacaine induced cell death in SKOV-3 but not in PC3. Our data suggests that bupivacaine has direct 'anti-cancer' properties through the activation of intrinsic and extrinsic apoptotic pathways in ovarian cancer but only the intrinsic pathway in prostate cancer.


Assuntos
Anestésicos Locais/farmacologia , Apoptose/efeitos dos fármacos , Bupivacaína/farmacologia , Caspases/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Quinases da Glicogênio Sintase/metabolismo , Humanos , Masculino , Neoplasias Ovarianas , Neoplasias da Próstata , Interferência de RNA
17.
Psychoneuroendocrinology ; 68: 202-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26999723

RESUMO

Stress exposure during early development is known to produce long-term mental health deficits. Stress promotes poor lifestyle choices such as poor diet. Early life adversity and diets high in fat and sugar (HFHS) are known to affect anxiety and memory. However additive effects of HFHS and stress during early development are less explored. Here, we examined whether early life stress (ELS) simulated by limited nesting (LN) induces anxiety-like behaviour and cognitive deficits that are modulated by HFHS diet. We examined key hippocampal markers involved in anxiety and cognition, testing the hypothesis that post-weaning HFHS following ELS would ameliorate anxiety-like behaviour but worsen memory and associated hippocampal changes. Sprague-Dawley rats were exposed to LN, postnatal days 2-9, and at weaning, male siblings were given unlimited access to chow or HFHS resulting in (Con-Chow, Con-HFHS, LN-Chow, LN-HFHS, n=11-15/group). Anxiety-like behaviour was assessed by Elevated Plus Maze (EPM) at 10 weeks and spatial and object recognition tested at 11 weeks of age. Rats were culled at 13 weeks. Hippocampal mRNA expression was measured using TaqMan(®) Array Micro Fluidic cards (Life Technologies). As expected HFHS diet increased body weight; LN and control rats had similar weights at 13 weeks, energy intake was also similar across groups. LN-Chow rats showed increased anxiety-like behaviour relative to control rats, but this was reversed by HFHS diet. Spatial and object recognition memory were unaltered by LN exposure or consumption of HFHS diet. Hippocampal glucocorticoid receptor (GR) protein was not affected by LN exposure in chow rats, but was increased by 45% in HFHS rats relative to controls. Hippocampal genes involved in plasticity and mood regulation, GSKα and GSKß were affected, with reductions in GSKß under both diet conditions, and reduced GSKα only in LN-HFHS versus Con-HFHS. Interestingly, HFHS diet and LN exposure independently reduced expression of Akt3 mRNA, a key gene involved post-natal brain development. In summary, while an energy rich diet ameliorated anxiety-like behaviour induced by LN exposure, it significantly altered key genes that are essential for hippocampal development.


Assuntos
Ansiedade/dietoterapia , Ansiedade/metabolismo , Gorduras na Dieta/administração & dosagem , Sacarose Alimentar/administração & dosagem , Animais , Peso Corporal/fisiologia , Dieta Hiperlipídica , Ingestão de Energia/fisiologia , Comportamento Alimentar , Quinases da Glicogênio Sintase/metabolismo , Hipocampo/metabolismo , Masculino , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/psicologia
18.
J Mol Endocrinol ; 55(3): 245-62, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26386043

RESUMO

Activation of apoptosis in cardiomyocytes by saturated palmitic acids contributes to cardiac dysfunction in diabetic cardiomyopathy. Beta-catenin (b-catenin) is a transcriptional regulator of several genes involved in survival/anti-apoptosis. However, its role in palmitate-induced cardiomyocyte apoptosis remains unclear. Glucagon-like peptide 1 (GLP1) has been shown to exhibit potential cardioprotective properties. This study was designed to evaluate the role of b-catenin signalling in palmitate-induced cardiomyocyte apoptosis and the molecular mechanism underlying the protective effects of GLP1 on palmitate-stressed cardiomyocytes. Exposure of neonatal rat cardiomyocytes to palmitate increased the fatty acid transporter CD36-mediated intracellular lipid accumulation and cardiomyocyte apoptosis, decreased accumulation and nuclear translocation of active b-catenin, and reduced expression of b-catenin target protein survivin and BCL2. These detrimental effects of palmitate were significantly attenuated by GLP1 co-treatment. However, the anti-apoptotic effects of GLP1 were markedly abolished when b-catenin was silenced with a specific short hairpin RNA. Furthermore, analysis of the upstream molecules and mechanisms responsible for GLP1-associated cardiac protection revealed that GLP1 restored the decreased phosphorylation of protein kinase B (Akt) and glycogen synthase kinase-3b (GSK3b) in palmitate-stimulated cardiomyocytes. In contrast, inhibition of Akt with an Akt-specific inhibitor MK2206 or blockade of GLP1 receptor (GLP1R) with a competitive antagonist exendin-(9-39) significantly abrogated the GLP1-mediated activation of GSK3b/b-catenin signalling, leading to increased apoptosis in palmitate-stressed cardiomyocytes. Collectively, our results demonstrated for the first time that the attenuated b-catenin signalling may contribute to palmitate-induced cardiomyocyte apoptosis, while GLP1 can protect cardiomyocytes from palmitate-induced apoptosis through activation of GLP1R/Akt/GSK3b-mediated b-catenin signalling.


Assuntos
Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Quinases da Glicogênio Sintase/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta Catenina/metabolismo , Animais , Apoptose/genética , Antígenos CD36/metabolismo , Sobrevivência Celular/genética , Técnicas de Silenciamento de Genes , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Metabolismo dos Lipídeos , Palmitatos/farmacologia , RNA Interferente Pequeno/genética , Ratos , beta Catenina/genética
19.
PLoS One ; 10(6): e0130008, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26091260

RESUMO

RNA interference-mediated gene silencing was shown to be an efficient tool for validation of targets that may become anti-tick vaccine components. Here, we demonstrate the application of this approach in the validation of components of molecular signaling cascades, such as the Protein Kinase B (AKT)/Glycogen Synthase Kinase (GSK) axis during tick embryogenesis. It was shown that heptane and hypochlorite treatment of tick eggs can remove wax, affecting corium integrity and but not embryo development. Evidence of AKT and GSK dsRNA delivery into de-waxed eggs of via electroporation is provided. Primers designed to amplify part of the dsRNA delivered into the electroporated eggs dsRNA confirmed its entry in eggs. In addition, it was shown that electroporation is able to deliver the fluorescent stain, 4',6-diamidino-2-phenylindole (DAPI). To confirm gene silencing, a second set of primers was designed outside the dsRNA sequence of target gene. In this assay, the suppression of AKT and GSK transcripts (approximately 50% reduction in both genes) was demonstrated in 7-day-old eggs. Interestingly, silencing of GSK in 7-day-old eggs caused 25% reduction in hatching. Additionally, the effect of silencing AKT and GSK on embryo energy metabolism was evaluated. As expected, knockdown of AKT, which down regulates GSK, the suppressor of glycogen synthesis, decreased glycogen content in electroporated eggs. These data demonstrate that electroporation of de-waxed R. microplus eggs could be used for gene silencing in tick embryos, and improve the knowledge about arthropod embryogenesis.


Assuntos
Técnicas de Silenciamento de Genes , RNA de Cadeia Dupla/genética , Rhipicephalus/genética , Animais , Proteínas de Artrópodes/genética , Proteínas de Artrópodes/metabolismo , Eletroporação , Feminino , Expressão Gênica , Quinases da Glicogênio Sintase/genética , Quinases da Glicogênio Sintase/metabolismo , Heptanos/química , Óvulo/química , Óvulo/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Solventes/química , Técnicas de Cultura de Tecidos , Ceras/química
20.
Sci Rep ; 5: 11602, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26111969

RESUMO

MiR-19a, a member of mir-17-92 microRNA clusters, has been demonstrated to promote cell proliferation and angiogenesis via regulating the PI3K/AKT pathway, the major insulin signaling pathway. However, whether miR-19a plays an important role in glycogen synthesis in hepatocytes remains unknown. Here, we define the impact of miR-19a on glycogen synthesis and IL-6-induced reduced glycogenesis in hepatocytes and its underlying mechanisms. Our studies indicate that miR-19a was down-regulated in the livers of db/db mice and mice injected with IL-6, as well as mouse NCTC 1469 hepatocytes and HEP 1-6 hepatocytes treated by IL-6. We found that over-expression of miR-19a in NCTC 1469 cells and HEP 1-6 cells led to increased activation of the AKT/GSK pathway and synthesis of glycogen, whereas down-regulation of miR-19a impaired AKT/GSK phosphorylation and glycogenesis. Over-expression of miR-19a ameliorated IL-6-induced reduced glycogen synthesis in hepatocytes. Moreover, we identified PTEN as the target of miR-19a by a luciferase assay. Down-regulation of PTEN rescued the effects of miR-19a suppression on the activation of the AKT/GSK pathway and improved glycogenesis in NTC 1469 cells. These findings show for the first time that miR-19a might activate the AKT/GSK pathway and glycogenesis via down-regulation of PTEN expression.


Assuntos
Regulação da Expressão Gênica , Glicogênio/biossíntese , Hepatócitos/metabolismo , MicroRNAs/genética , PTEN Fosfo-Hidrolase/genética , Animais , Western Blotting , Linhagem Celular , Perfilação da Expressão Gênica/métodos , Quinases da Glicogênio Sintase/metabolismo , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Interleucina-6/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Análise de Sequência com Séries de Oligonucleotídeos , PTEN Fosfo-Hidrolase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA