Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
BMC Cancer ; 22(1): 1091, 2022 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-36284263

RESUMO

BACKGROUND: Neuro-oncological ventral antigen 1 (NOVA1) is a neuron-specific RNA-binding protein which regulates alternative splicing in the developing nervous system. Recent research has found that NOVA1 plays a significant role in carcinogenesis. In this paper, we examine the role of NOVA1 in non-small cell lung cancer (NSCLC) and its underlying molecular mechanisms. METHODS: The expression of NOVA1 in NSCLC was detected by immunohistochemistry and correlations between NOVA1 expression and clinicopathological factors were analyzed by chi-square tests. Kaplan-Meier survival analysis and the Cox regression model were used to evaluate the predictive effect of prognostic factors. Western blotting, Cell Counting Kit-8, colony formation, apoptosis, migration and invasion assays were used to detect the effects of silencing (si)NOVA1 RNA on Wnt/ß-catenin signaling and biological behavior in NSCLC cell lines. RESULTS: Our study showed that expression of NOVA1 was up-regulated and significantly correlated with poor differentiation (p = 0.020), advanced TNM stage (P = 0.001), T stage (P = 0.001) and lymph node metastasis (P = 0.000) as well as the expression of ß-catenin (P = 0.012) in NSCLC. The down-regulation of NSCLC by siRNA significantly inhibited proliferation, migration and invasion and promoted apoptosis in NSCLC cells. Expression of Wnt signaling molecules, including ß-catenin, activated ß-catenin, cyclin D1, matrix metalloproteinase (MMP)-2 and MMP-7, was also significantly reduced by siNOVA1. The inhibition of Wnt/ß-catenin signaling in A549 and H1299 cells by siNOVA1 was reversed after treatment with a ß-catenin expression plasmid. CONCLUSION: The present study suggests that NOVA1 may serve as a potential prognosis biomarker in NSCLC. High NOVA1 expression was associated with poor survival rate. Finally, in vitro experiments verified that NOVA1 promotes NSCLC cell proliferation and invasion by regulating Wnt/ß-catenin signaling.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Via de Sinalização Wnt , Ciclina D1/genética , Metaloproteinase 7 da Matriz/metabolismo , beta Catenina/genética , beta Catenina/metabolismo , Neoplasias Pulmonares/patologia , RNA Interferente Pequeno/farmacologia , Antígeno Neuro-Oncológico Ventral , Proliferação de Células/genética , Prognóstico , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Biomarcadores , Movimento Celular/genética , Linhagem Celular Tumoral
2.
Int J Med Sci ; 19(2): 338-351, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35165519

RESUMO

Mitogen-inducible gene 6 (Mig-6) is a tumor suppressor gene that plays an important role in many types of cancers by interacting with EGFR. However, its molecular mechanism in hepatocellular carcinoma (HCC) and its relationship with miRNAs need to be elucidated. Therefore, this study aimed to explore whether Mig-6 could promote apoptosis and the inhibition of autophagy via its downstream miRNA in HCC cell lines. We used two cell lines, HepG2 and HLE, to establish Mig-6 overexpression and knockdown experiments, as well as miR-193a mimic and inhibitor experiments. The miRNA microarray profiling was also used to verify Mig-6-regulated miRNA. We found that Mig-6 induced apoptosis and reduced autophagy of HCC cell lines. miR-193a-3p is a Mig-6-regulated miRNA in the Mig-6-overexpression model. It affected the apoptosis and autophagy of HCC cells, at least partly by regulating the expression of TGF-ß2. Additionally, the relationship between Mig-6 and transforming growth factor TGF-ß2 was explored in depth for the first time. These findings revealed an important role of Mig-6 in the apoptosis and autophagy of HCC cells by regulating miR-193a-3p, providing a novel insight into the therapeutic target in HCC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia/genética , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Apoptose/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Fator de Crescimento Transformador beta2/metabolismo
3.
Arch Gynecol Obstet ; 303(2): 285-311, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33222040

RESUMO

PURPOSE: To evaluate the efficacy and toxicity of angiogenesis inhibitors for the treatment of ovarian cancer patients, we conducted a meta-analysis of the published literature on this subject. METHODS: In this meta-analysis, we searched PubMed, EMBASE, Web of Science, and Cochrane Library databases for randomized controlled trials (RCTs). The literature search was performed up to August 12, 2019. The risk of bias of the included studies was evaluated using The Cochrane Collaboration's tool, and the statistical analyses were performed using RevMan 5.3 software. The sensitivity analysis was performed with Stata 12.0 software. RESULTS: 22 RCTs with 11,254 patients were included. Our meta-analysis demonstrates that angiogenesis inhibitors therapy can significantly improve progression-free survival (PFS) (hazard ratio [HR] 0.71, 95% CI 0.63-0.79, I2 = 80%, P < 0.00001) and overall survival (OS) (HR 0.95, 95% CI 0.90-0.99, I2 = 0%, P = 0.03) in ovarian cancer patients. The subgroups results suggest differences in the benefit in OS in first-line treatment (HR 1.00, 95% CI 0.93-1.08, I2 = 0%, P = 0.90) compared with treatment at relapse (HR 0.87, 95% CI 0.81-0.95, I2 = 0%, P = 0.0008). The PFS improved both in first-line treatment (HR 0.87, 95% CI 0.79-0.95, I2 = 60%, P = 0.003) and recurrent treatment (HR 0.60, 95% CI 0.53-0.67, I2 = 57% P < 0.0001) patients. The PFS and OS in recurrent group were prolonged both in the platinum-resistant group(PFS: HR 0.50, 95% CI 0.42-0.60, I2 = 0%, P < 0.00001; OS: HR 0.76, 95% CI 0.62-0.93, I2 = 0%, P = 0.007) and the platinum-sensitive group (PFS: HR 0.58, 95% CI 0.49-0.69, I2 = 64%, P < 0.00001; OS: HR 0.88, 95% CI 0.79-0.99, I2 = 0%, P = 0.03). However, this therapy is associated with a higher risk of common adverse events of grade ≥ 3 (risk ratio [RR]: 1.12; 95% CI 1.07-1.17; I2 = 0%, P = 0.68) such as arterial thromboembolic disease, ascites, diarrhea, gastrointestinal perforations, headache, hemorrhagic, hypertension, hypokalemia, leucopenia, pain, proteinuria, thrombocytopenia, and thrombosis or embolism. CONCLUSION: This meta-analysis suggests angiogenesis inhibitors may significantly improve PFS and OS of ovarian cancer patients and increase the incidence of common adverse events.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Antineoplásicos/administração & dosagem , Carcinoma Epitelial do Ovário/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Ensaios Clínicos Controlados Aleatórios como Assunto , Inibidores da Angiogênese/efeitos adversos , Antineoplásicos/efeitos adversos , Carcinoma Epitelial do Ovário/mortalidade , Intervalo Livre de Doença , Feminino , Humanos , Recidiva Local de Neoplasia , Neoplasias Ovarianas/mortalidade , Intervalo Livre de Progressão , Análise de Sobrevida
4.
Exp Mol Pathol ; 102(3): 492-499, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28506767

RESUMO

The ablation of Mig-6 has been shown to induce tumor formation in various tissues. However, the relationships between Mig-6 expression, clinical pathological factors, and prognosis have not been clarified in hepatocellular carcinoma (HCC), and the mechanism by which Mig-6 regulates the proliferation of HCC cells has not been reported. In this study, we investigated the clinical significance of the loss of Mig-6 expression in HCC and the mechanism underlying the inhibition of cell proliferation by Mig-6. The down-regulation of Mig-6 correlated significantly with large tumors, a more advanced BCLC stage, and a more advanced TNM stage, and low Mig-6 expression predicted significantly reduced survival. Low Mig-6 expression and high Cyclin D1 expression were independent predictors for survival. The overexpression of Mig-6 led to significant G1 arrest and growth inhibition in HCC cells, possibly through the inhibition P-ERK and Cyclin D1. These results indicate that Mig-6 expression is low in HCC, which predicts a poor prognosis. Mig-6 may regulate cell proliferation and the cell cycle through the P-ERK/Cyclin D1 pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Hepatocelular/diagnóstico , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/diagnóstico , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Ciclina D1/genética , Ciclina D1/metabolismo , Regulação para Baixo , Feminino , Células Hep G2 , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Pessoa de Meia-Idade , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Prognóstico , Modelos de Riscos Proporcionais , Transfecção , Proteínas Supressoras de Tumor/genética
5.
Pharm Biol ; 54(12): 2802-2806, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27251880

RESUMO

CONTEXT: Augmented renal clearance (ARC) refers to enhanced renal elimination of circulating solute, and has attracted wide attention in recent years. OBJECTIVE: This study evaluates the effects of ARC on serum vancomycin concentration in patients administered vancomycin. MATERIALS AND METHODS: This was a retrospective study in patients receiving vancomycin treatment at a dose of 1000 mg in every 12 h and undergoing serum monitoring admitted over a 2-year period (May 2013 to May 2015), in order to estimate the influence of ARC on serum vancomycin concentration. In this study, statistical comparisons were made on the results from patients grouped according to creatinine clearance (CLcr). RESULTS: One hundred forty-eight patients were enrolled in our study. The results showed that ARC patients were significantly younger, with a significantly lower Scr and higher GFR. The CLcr and steady-state trough concentrations of serum vancomycin exhibited a logarithmic correlation (Rs = -0.699, R2 = 0.488, p < 0.01) in the patients included in our study. The trough vancomycin concentrations of 62.9% patients in high CLcr group were under 10 µg/mL. DISCUSSION AND CONCLUSION: Since ARC was significantly associated with subtherapeutic serum vancomycin concentration, it was necessary to devise adjusted dosage regimens for these patients based on their CLcr.


Assuntos
Antibacterianos/sangue , Nefropatias/sangue , Nefropatias/tratamento farmacológico , Rim/efeitos dos fármacos , Taxa de Depuração Metabólica/efeitos dos fármacos , Vancomicina/sangue , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Criança , Feminino , Taxa de Filtração Glomerular , Humanos , Rim/metabolismo , Nefropatias/diagnóstico , Masculino , Taxa de Depuração Metabólica/fisiologia , Pessoa de Meia-Idade , Estudos Retrospectivos , Vancomicina/farmacologia , Vancomicina/uso terapêutico , Adulto Jovem
6.
Mol Carcinog ; 51(7): 522-34, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21739478

RESUMO

Downregulation of Mig-6 expression has been implicated in several human cancers and its loss can lead to prolonged activation of EGFR and carcinogenesis. The present study aimed to investigate the clinical significance of loss of Mig-6 expression in nonsmall-cell lung cancer (NSCLC) and the biological functions of Mig-6 in NSCLC cell lines. Mig-6 expression was downregulated in 47/91 (51.6%) cases of NSCLC that were examined. Mig-6 downregulation significantly correlated with poor differentiation (P = 0.0131), histological type (P = 0.0021), and EGFR expression (P = 0.003). In addition, knockdown of Mig-6 expression in H1299 and BE1 cells promoted EGF-induced tumor cell proliferation and migration. Furthermore, Mig-6 knockdown led to a significant increase in phospho-AKT, phospho-ERK, phospho-EGFR as well as MMP-2 and MMP-9 levels. These results indicate that downregulated Mig-6 in NSCLC tissues may serve as a new marker that can predict the activation of EGFR signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Regulação para Baixo , Receptores ErbB/metabolismo , Neoplasias Pulmonares/metabolismo , Transdução de Sinais , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Técnicas de Silenciamento de Genes , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real , Proteínas Supressoras de Tumor/genética
7.
J Cancer ; 10(4): 1060-1069, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30854112

RESUMO

The characterization of high recurrence rate of HCC after TAE provides insights into persistent issues surrounding the role of adjunct therapies administered with TAE. As a regulator of the HER family, Mig-6 is down-regulated in HCC and predicts the prognosis of HCC. In this study, we found up-regulation the expression of Mig-6 enhances autophagy in HCC cells. This function of Mig-6 is related to the activation of the JNK pathway. Next AAV-9 encoding Mitogen inducible gene 6 (Mig-6) was delivered into VX2 liver transplant tumor of rabbits by using hepatic artery catheter. Wild-type AAV is not associated with any human or animal disease and has very low immunogenicity. There are over 100 different AAV serotypes that vary in the amino acid sequence of their capsid protein. We also describe a novel combination therapy coupling AAV-Mig-6 and TAE in a rabbit model resulted in a growth rate decrease in tumor compared with TAE alone. Furthermore, we show that the changes of LC3b and p62, as well as the p-JNK were consistent with changes in vitro experiments. These results suggest that Mig-6 efficiently inhibits tumor progression in vivo. Our findings suggest that Mig-6 induced autophagy inhibition may become a necessary target for adjunct therapy in TAE.

8.
Zhongguo Fei Ai Za Zhi ; 17(11): 783-8, 2014 Nov.
Artigo em Zh | MEDLINE | ID: mdl-25404268

RESUMO

BACKGROUND AND OBJECTIVE: Thymosin beta 10 (Tß10) is one of ß-thymosin family members, has a highly conserved polar 5 kDa peptides. This peptide is now regarded to be a small actin-binding protein and thereby induce depolymerization of the intracellular F-actin networks. Alteration of Tß10 expression may alter the balance of cell growth, cell death, cell attachment and cell migration. Tß10 also affects cell metastasis as well as proliferation, apoptosis and vascularization of cancer cells. But function of Tß10 appear to be rather different between cancer cells, and the molecular mechanisms of ß-thymosins to regulate cell apoptosis and proliferation in NSCLC (non-small cell lung cancer) cell lines are unclear. In this study, we used lung adenocarcinoma cell line A549, added Tß10 or down-regulated the expression of Tß10. We observed the change of apoptosis, proliferation and cell cyclin ability in A549 and the mechanisms underline them were also identified. METHODS: After A549 was treated with 100 ng/mL recombinant human Tß10 or siTß10, apoptosis rate of A549 and cell cycle distribution were detected by flow cytometry (FCM). CCK-8 assay was employed to determine the proliferation of A549. The mRNA level of P53, Caspase-3, Cyclin A and Cyclin E were determined by real-time PCR. The protein level of P53, Caspase-3, Cyclin A and Cyclin E were detected by Western blot. RESULTS: Add Tß10 can inhibit the apoptosis and prompt the proliferation of A549. It can also increase the cell rates of S-phrase and G2/M-phrase, decrease the expression of P53 and Caspase-3, but increase the expression of Cyclin A and Cyclin E. Interferance of Tß10 can prompt the apoptosis and inhibit the proliferation of A549. It can also increase the cell rates of G0/G1-phrase, increase the expression of P53 and Caspase-3, but decrease the expression of Cyclin A and Cyclin E. CONCLUSIONS: In lung cancer cell line, Tß10 can inhibit the apoptosis by increase P53, drive cells into the S and G2/M-phase, prompt cell proliferation by increase the expression of Cyclin A and Cyclin E. Tß10 may become a potential biomarker and therapy target for non-small cell lung cancer.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Timosina/farmacologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Apoptose/genética , Western Blotting , Caspase 3/genética , Caspase 3/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Ciclina A/genética , Ciclina A/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timosina/genética , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Zhongguo Fei Ai Za Zhi ; 17(5): 378-83, 2014 May.
Artigo em Zh | MEDLINE | ID: mdl-24854554

RESUMO

BACKGROUND AND OBJECTIVE: Our previous study found that thymosin ß10 overexpressed in lung cancer and positively correlated with differentiation, lymph node metastasis and stage of lung cancer. In this reasearch we aim to study the effects and mechanism of exogenous human recombinant Tß10 on the expression of VEGF-C on non-small cell lung cancer. METHODS: After SPC, A549 and LK2 cells were treated with 100 ng/mL recombinant human Tß10, the mRNA level of VEGF-C were detected by RT-PCR. The mean while the protein expression of VEGF-C, P-AKT and AKT were determined by Western blot assay. RESULTS: Exogenous recombinant human Tß10 were significantly promote the expression levels of VEGF-C mRNA and protein while promoting the phosphorylation of AKT. Exogenous Tß10 can promote the expression of VEGF-C mRNA and protein in lung cancer cell lines A549 and LK2 (P<0.05), and this effect can be inhibited by use AKT inhibitor LY294002 (P<0.05). CONCLUSIONS: Tß10 human recombinant proteins can promote the expression of VEGF-C by activating AKT phosphorylation in lung cancer cell lines.


Assuntos
Neoplasias Pulmonares/genética , Timosina/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/metabolismo , Proteína Oncogênica v-akt/genética , Proteína Oncogênica v-akt/metabolismo , Fosforilação , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/metabolismo
10.
Oncol Rep ; 31(4): 1707-14, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24573418

RESUMO

Mitogen-inducible gene-6 (Mig-6), an immediate early response gene, is a specific negative regulator of epidermal growth factor receptor (EGFR). Ablation of Mig-6 has been shown to induce tumor formation in various tissues, supporting the tumor suppressor function of Mig-6. However, little is known about the role of Mig-6 in non-small cell lung cancer (NSCLC) apoptosis, nor has the contribution of upregulated Mig-6 on biological behaviors of A549 and H157 cells previously been reported. The aim of the present study was to investigate the effects of exogenously transfected Mig-6 on proliferation, invasion and apoptosis of A549 and H157 cells and to identify novel underlying mechanisms of Mig-6-induced apoptosis. We used immunohistochemical staining to examine the expression of Mig-6 protein in NSCLC tissues. For evaluation of the prognostic value of Mig-6 expression to each clinicopathologic factor, Kaplan-Meier method and Cox's proportional hazards model were employed. Mig-6 low expression was correlated with a poor prognosis in patients with lung cancer. Patients with high expression of Mig-6 had a statistically significantly longer survival than those with low expression of Mig-6. Cox's regression analysis indicated that loss of Mig-6 expression was an independent, unfavorable prognostic factors. We utilized siRNA-targeting Mig-6 and Mig-6 overexpression plasmid to determine the effect of Mig-6 on lung cancer cells. Flow cytometry studies revealed Mig-6 overexpression promoted apoptosis in NSCLC cell lines. siRNA-mediated Mig-6 knockdown inhibited apoptosis of cancer cells, but this anti-apoptotic effect was abolished by inhibition of ERK. Upregulation of Mig-6 decreased the proliferation and invasive potential of transfected cells. Moreover, upregulation of Mig-6 inhibited proliferation and invasion of A549 and H157 cells. Collectively, our results showed that Mig-6 is a potential biomarker for evaluation of tumor prognosis of lung cancer. Mig-6 promotes apoptosis in lung cancer cells via the ERK pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/genética , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/genética , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Western Blotting , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Linhagem Celular Tumoral , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Prognóstico , Modelos de Riscos Proporcionais , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Regulação para Cima
11.
Artigo em Inglês | MEDLINE | ID: mdl-25168794

RESUMO

Valproate sodium is one of the most prescribed antiepileptic drugs. However, valproate sodium has various side effects, especially its toxicity on liver. Current markers for toxicity reflect mostly the late stages of tissue damage; thus, more efficient methods for toxicity evaluation are desired. To evaluate the toxicity of valproate sodium on liver, we performed both UPLC-MS and (1)HNMR-based metabonomics analysis of serum samples from 34 epileptic patients (age: 42.0±18.6, 18 male/16 female) after valproate sodium treatment. Compared to conventional markers, the serum metabolic profiles provided clear distinction of the valproate sodium induced normal liver function and abnormal liver function in epileptic patients. Through multivariate statistical analysis, we identified marker metabolites associated with the hepatotoxicity induced by valproate sodium, such as glucose, lactate, acetoacetate, VLDL/LDL, lysophosphatidylcholines, phosphatidylcholines, choline, creatine, amino acids, N-acetyl glycoprotein, pyruvate and uric acid. This metabonomics approach may provide effective way to evaluate the valproate sodium-induced toxicity in a manner that can complement current measures. This approach is expected to find broader application in other drug-induced toxicity assessment.


Assuntos
Anticonvulsivantes/efeitos adversos , Anticonvulsivantes/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Metabolômica/métodos , Ácido Valproico/efeitos adversos , Ácido Valproico/metabolismo , Adulto , Anticonvulsivantes/química , Anticonvulsivantes/uso terapêutico , Cromatografia Líquida de Alta Pressão/métodos , Epilepsia/tratamento farmacológico , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Espectrometria de Massas/métodos , Redes e Vias Metabólicas , Pessoa de Meia-Idade , Análise Multivariada , Ressonância Magnética Nuclear Biomolecular/métodos , Ácido Valproico/química , Ácido Valproico/uso terapêutico , Adulto Jovem
12.
Int J Clin Exp Pathol ; 7(10): 7304-11, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25400829

RESUMO

Non small cell lung cancer (NSCLC) accounts for 85% of all lung cancers and is the most common cause of lung cancer death. Currently, the epidermal growth factor receptor inhibitor gefitinib is widely used for patients with advanced NSCLC. However, drug resistance is a major obstacle. Mig-6 is a feedback inhibitor of EGFR and its down-stream pathway; it has been shown to play a role in gefitinib sensitivity. There is neither systematical research on the relationship between Mig-6 expression and gefitinib sensitivity, nor has the contribution of up-regulated Mig-6 on the gefitinib-resistant cell lines. In the present work, four NSCLC cell lines (H1299, A549, PC-9, and PC-9/AB11) with different sensitivities to gefitinib were subjected to analysis of the expression of Mig-6. We found that Mig-6 is over-expressed in gefitinib-sensitive NSCLC cell lines, but is low in gefitinib-resistant NSCLC cell lines. Further analysis revealed that over-expression of Mig-6 increased cell apoptosis and inhibited proliferation of gefitinib-resistant NSCLC cells treated with gefitinib, whereas lowering the expression of Mig-6 decreased cell apoptosis and promoted cell proliferation after treatment with gefitinib in gefitinib-sensitive NSCLC cell lines. These results suggest that Mig-6 is involved in mediating the response to gefitinib in NSCLC cell lines. Additionally we demonstrated that Mig-6 could reverse gefitinib resistance through inhibition of EGFR/ERK pathway in NSCLC cells. Our work uncovered that Mig-6 may be an effective therapeutic target in gefitinib-resistant lung cancer patients.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Neoplasias Pulmonares/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Gefitinibe , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Fatores de Tempo , Transfecção , Proteínas Supressoras de Tumor/genética
13.
PLoS One ; 7(5): e36903, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22615840

RESUMO

We aimed to investigate the clinical significance of the expression of novel scaffold protein CARMA3 in non-small-cell lung cancer (NSCLC) and the biological function of CARMA3 in NSCLC cell lines. We observed moderate to high CARMA3 staining in 68.8% of 141 NSCLC specimens compared to corresponding normal tissues. The overexpression of CARMA3 was significantly correlated with TNM stage (P = 0.022) and tumor status (P = 0.013). CARMA3 upregulation also correlated with a shorter survival rate of patients of nodal status N0 (P = 0.042)as well as the expression of epidermal growth factor receptor (EGFR) (P = 0.009). In EGFR mutation positive cases, CARMA3 expression was much higher (87.5%) compared to non-mutation cases (66.1%). In addition, we observed that knockdown of CARMA3 inhibits tumor cell proliferation and invasion, and induces cell cycle arrest at the boundary between the G1 and S phase. We further demonstrated a direct link between CARMA3 and NF-κB activation. The change of biological behavior in CARMA3 knockdown cells may be NF-κB-related. Our findings demonstrated, for the first time, that CARMA3 was overexpressed in NSCLC and correlated with lung cancer progression, EGFR expression, and EGFR mutation. CARMA3 could serve as a potential companion drug target, along with NF-kB and EGFR in EGFR-mutant lung cancers.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Proteínas Adaptadoras de Sinalização CARD/biossíntese , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular Tumoral , Progressão da Doença , Receptores ErbB/biossíntese , Receptores ErbB/genética , Fase G1/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Invasividade Neoplásica , Estadiamento de Neoplasias , Fase S/genética , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA