Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Int J Med Sci ; 19(1): 47-64, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34975298

RESUMO

Background: Clear cell renal cell carcinoma (ccRCC) is a cell metabolic disease with high metastasis rate and poor prognosis. Our previous studies demonstrate that glucose-6-phosphate dehydrogenase (G6PD), the first and rate-limiting enzyme of the pentose phosphate pathway, is highly expressed in ccRCC and predicts poor outcomes of ccRCC patients. The aims of this study were to confirm the oncogenic role of G6PD in ccRCC and unravels novel mechanisms involving Cyclin E1 and MMP9 in G6PD-mediated ccRCC progression. Methods: Real-time RT-PCR, Western blot and immunohistochemistry were used to determine the expression patterns of G6PD, Cyclin E1 and MMP9 in ccRCC. TCGA dataset mining was used to identify Cyclin E1 and MMP9 correlations with G6PD expression, relationships between clinicopathological characteristics of ccRCC and the genes of interest, as well as the prognosis of ccRCC patients. The role of G6PD in ccRCC progression and the regulatory effect of G6PD on Cyclin E1 and MMP9 expression were investigated by using a series of cytological function assays in vitro. To verify this mechanism in vivo, xenografted mice models were established. Results: G6PD, Cyclin E1 and MMP9 were overexpressed and positively correlated in ccRCC, and they were associated with poor prognosis of ccRCC patients. Moreover, G6PD changed cell cycle dynamics, facilitated cells proliferation, promoted migration in vitro, and enhanced ccRCC development in vivo, more likely through enhancing Cyclin E1 and MMP9 expression. Conclusion: These findings present G6PD, Cyclin E1 and MMP9, which contribute to ccRCC progression, as novel biomarkers and potential therapeutic targets for ccRCC treatment.


Assuntos
Carcinoma de Células Renais/genética , Ciclina E/genética , Regulação Neoplásica da Expressão Gênica , Glucosefosfato Desidrogenase/fisiologia , Neoplasias Renais/genética , Metaloproteinase 9 da Matriz/genética , Proteínas Oncogênicas/genética , Regulação para Cima , Animais , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Ciclina E/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Proteínas Oncogênicas/metabolismo
2.
Cancer Sci ; 112(10): 4075-4086, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34310804

RESUMO

The regulatory relationship between silent information regulator 2 (SIRT2) and glucose 6-phosphate dehydrogenase (G6PD) in clear cell renal cell carcinoma (ccRCC) is still unclear. The present study aimed to explore the function of SIRT2 and its regulatory effect on G6PD in ccRCC. The Cancer Genome Atlas data mining of SIRT2 was first analyzed. Quantitative real-time PCR and western blot analyses were used to assess the mRNA and protein expression levels, respectively. Cell viability, colony formation, cell cycle, cell apoptosis, and TUNEL assays and EdU staining were used to investigate the roles of SIRT2 in ccRCC proliferation and apoptosis. The coimmunoprecipitation (Co-IP) assay was used to analyze the association between SIRT2 and G6PD in ccRCC cells. Quantitative Co-IP assay was used to detect the levels of G6PD ubiquitination and small ubiquitin-related modifier 1 (SUMO1). An in vivo experiment was also carried out to confirm in vitro findings. The results indicated that SIRT2 promoted ccRCC proliferation and inhibited apoptosis by regulating cell cycle and apoptosis related proteins. Silent information regulator 2 interacted with G6PD, facilitated its activity through deacetylation, and increased its stability by reducing its ubiquitination and enhancing its SUMO1 modification. Silent information regulator 2 also promoted ccRCC tumor development in vivo. Taken together, the present study indicated that SIRT2 promoted ccRCC progression by increasing G6PD activity and stability, and it could be a potential new diagnostic and therapeutic target for ccRCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Cisteína Endopeptidases/metabolismo , Glucosefosfato Desidrogenase/metabolismo , Neoplasias Renais/metabolismo , Sirtuína 2/fisiologia , Acetilação , Animais , Apoptose , Western Blotting , Carcinoma de Células Renais/patologia , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular , Bases de Dados Genéticas , Progressão da Doença , Feminino , Humanos , Imunoprecipitação , Neoplasias Renais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Modificação Traducional de Proteínas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco , Ubiquitinação
3.
Eur J Clin Invest ; 50(10): e13332, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32589285

RESUMO

BACKGROUND: This study aimed to summarize the association between diabetes mellitus (DM) and the incidence of lung cancer using a meta-analysis of cohort studies. MATERIALS AND METHODS: We systematically searched PubMed, Embase and the Cochrane Library to identify potential cohort studies. Relative risk (RR) was used to calculate the association between DM and the risk of lung cancer. Subgroup analysis, sensitivity analysis and test for publication bias were performed. Twenty cohort studies were selected. RESULTS: The participants with DM showed little or no significant effect on the risk of lung cancer (RR: 1.10; 95% CI: 0.99-1.23; P = .087). DM was not associated with the risk of lung cancer in men (RR: 1.11; 95%CI: 0.92-1.35; P = .270), but a significant association was observed in women (RR: 1.18; 95%CI: 1.10-1.28; P < .001). Subgroup analysis suggested that smoker status was confounding variables that could bias the relationship between DM and the incidence of lung cancer. CONCLUSIONS: This meta-analysis suggests that DM has no significant impact on the incidence of lung cancer in men but has a harmful effect on women.


Assuntos
Diabetes Mellitus/epidemiologia , Neoplasias Pulmonares/epidemiologia , Humanos
4.
Cancer Cell Int ; 20(1): 565, 2020 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-33292264

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

5.
Cancer Cell Int ; 20: 483, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33041664

RESUMO

BACKGROUND: Glucose 6-phosphate dehydrogenase (G6PD) serves key roles in cancer cell metabolic reprogramming, and has been reported to be involved in certain carcinogenesis. Previous results from our laboratory demonstrated that overexpressed G6PD was a potential prognostic biomarker in clear cell renal cell carcinoma (ccRCC), the most common subtype of kidney cancer. G6PD could stimulate ccRCC growth and invasion through facilitating reactive oxygen species (ROS)-phosphorylated signal transducer and activator of transcription 3 (pSTAT3) activation and ROS-MAPK-MMP2 axis pathway, respectively. However, the reasons for ectopic G6PD overexpression and the proliferation repressive effect of G6PD inhibition in ccRCC are still unclear. METHODS: The impact of ROS accumulation on NF-κB signaling pathway and G6PD expression was determined by real-time RT-PCR and Western blot in ccRCC cells following treatment with ROS stimulator or scavenger. The regulatory function of NF-κB signaling pathway in G6PD transcription was analyzed by real-time RT-PCR, Western blot, luciferase and ChIP assay in ccRCC cells following treatment with NF-κB signaling activator/inhibitor or lentivirus infection. ChIP and Co-IP assay was performed to demonstrate protein-DNA and protein-protein interaction of NF-κB and pSTAT3, respectively. MTS assay, human tissue detection and xenograft model were conducted to characterize the association between NF-κB, pSTAT3, G6PD expression level and proliferation functions. RESULTS: ROS-stimulated NF-κB and pSTAT3 signaling over-activation could activate each other, and exhibit cross-talks in G6PD aberrant transcriptional regulation. The underlying mechanism was that NF-κB signaling pathway facilitated G6PD transcription via direct DNA-protein interaction with p65 instead of p50. p65 and pSTAT3 formed a p65/pSTAT3 complex, occupied the pSTAT3-binding site on G6PD promoter, and contributed to ccRCC proliferation following facilitated G6PD overexpression. G6PD, pSTAT3, and p65 were highly expressed and positively correlated with each other in ccRCC tissues, confirming that NF-κB and pSTAT3 synergistically promote G6PD overexpression. Moreover, G6PD inhibitor exhibited tumor-suppressor activities in ccRCC and attenuated the growth of ccRCC cells both in vitro and in vivo. CONCLUSION: ROS-stimulated aberrations of NF-κB and pSTAT3 signaling pathway synergistically drive G6PD transcription through forming a p65/pSTAT3 complex. Moreover, G6PD activity inhibition may be a promising therapeutic strategy for ccRCC treatment.

6.
BMC Cancer ; 20(1): 682, 2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32698787

RESUMO

BACKGROUND: Long intergenic non-coding RNA 00511 (LINC00511) is highly expressed in diverse cancers and has a correlation with poor clinical outcomes for cancer patients. In view of contradictory data among published data, we aim to evaluate the prognostic role of LINC00511 for cancer patients. METHODS: In the present study, a meta-analysis of related studies has been performed to investigate the prognostic significance of LINC00511 in cancer patients. Relevant studies published before December 22, 2019 were systematically searched online in PubMed, EMBASE, Web of Science, and the Cochrane Library databases. The relationship between LINC00511 expression and cancer patients' survival, including overall survival (OS), disease-free survival (DFS)/relapse-free survival (RFS) and progression-free survival (PFS), was evaluated using pooled hazard ratios (HRs) with their corresponding 95% confidence intervals (CIs). The association between LINC00511 expression and clinicopathological features was assessed using odd ratios (ORs) and their corresponding 95% CIs. RESULTS: A total of 14 eligible studies with 1883 patients were enrolled in the present meta-analysis. The results demonstrated that elevated expression of LINC00511 was significantly associated with poor OS (HR = 2.62; 95% CI: 2.00-3.45; p <  0.001), PFS (HR = 1.80; 95% CI: 1.29-2.51; p = 0.001) and DFS/RFS (HR = 2.90; 95% CI: 1.04-8.12; p = 0.04). Additionally, High LINC00511 expression was associated with large tumor size (OR = 3.10; 95% CI: 1.97-4.86; p <  0.00001), lymph node metastasis (OR = 3.11; 95% CI: 2.30-4.21; p <  0.00001), advanced clinical stage (OR = 3.95; 95% CI: 2.68-5.81; p <  0.00001), distant metastasis (OR = 2.39; 95% CI: 1.16-4.93; p = 0.02), and disease recurrence (OR = 4.62; 95% CI: 2.47-8.65; p <  0.00001). Meanwhile, no correlation was found between LINC00511 expression and age, gender, and histological grade. These findings were consolidated by the results of bioinformatics analysis. CONCLUSIONS: Based on our findings, LINC00511 may serve as a novel prognostic biomarker for cancer patients.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/metabolismo , Neoplasias/mortalidade , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Humanos , Prognóstico , Viés de Publicação
7.
J Cell Biochem ; 120(11): 19124-19136, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31310363

RESUMO

In this study, we aimed to explore the mechanism of glutathione peroxidase 3 (GPX3) in the growth of malignant melanoma (MM) cells by hypoxia-inducible factor-1α (HIF1-α) and HIF2-α regulating the metabolism through reactive oxygen species (ROS). The messenger RNA and protein expression of GPX3, HIF1-α, HIF2-α in tissues, and cell lines were measured by reverse transcription-quantitative PCR and Western blot analysis. A375 cells were transfected with GPX3 overexpression plasmid, small interfering RNA (siRNA) targeting GPX3, or siRNA targeting HIF1-α/HIF2-α to upregulate or downregulate the expression of GPX3 or HIF1-α/HIF2-α. The effects of H2 O2 and N-acetylcysteine (NAC) on the levels of HIF1-α and HIF2-α after overexpression of GPX3 were studied. The cell viability was detected by Cell Counting Kit-8. The levels of ROS, glucose uptake and lactic acid production, oxidative phosphorylation, and glycolysis of cells were measured for assessment of cellular metabolism. The expression of GPX3 decreased, while ROS, HIF1-α, and HIF2-α increased in MM tissues and cells. Overexpression of GPX3 inhibited the viability of MM cells and the growth of melanoma xenografts. The overexpression of GPX3 reduced the glucose uptake, extracellular lactic acid content, and extracellular acidification rate and increased the oxygen consumption rate level. Overexpression of GPX3 could reduce the levels of HIF1-α and HIF2-α, which could regulate metabolic levels. GPX3 reduced ROS level in MM to inhibit HIF1-α and HIF2-α. The addition of H2 O2 increased while NAC reduced the protein levels of HIF1-α and HIF2-α in the cells overexpressing GPX3. Our study demonstrates that GPX3 inhibits the growth of MM cells through its inhibitory effect on cell metabolic disorder by inhibiting HIF1-α via regulating ROS.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Glutationa Peroxidase/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Melanoma/metabolismo , Proteínas de Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Idoso , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular Tumoral , Feminino , Glutationa Peroxidase/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Melanoma/genética , Melanoma/patologia , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Estabilidade Proteica
8.
Artigo em Inglês | MEDLINE | ID: mdl-36612384

RESUMO

Rural tourism is an important income generation method for farmers post-pandemic. However, few studies have focused on how pandemic fatigue has affected their willingness to participate in rural tourism development. We conducted a quasi-experiment to test these effects using data from two Chinese villages. Shanlian village, which was more severely affected by COVID-19, was the experimental group, while Huashu village was set as the control group. Our results reveal that both physical and mental fatigue hinder farmers' intention to engage in rural tourism. Further, there were significant interaction effects between physical and mental fatigue on the farmers' participation in rural tourism. For farmers with low physical fatigue, the higher their mental fatigue, the less willing they were to participate in rural development. Conversely, for the higher physical fatigue group, farmers with low levels of mental fatigue were still more willing to participate in rural tourism development. These findings reduce the current research gap concerning the relationship between pandemic fatigue and farmers' participation in rural tourism and indicate that practitioners and policymakers should consider farmers' fatigue management as an important factor for the sustainability of rural tourism during the ongoing COVID-19 crisis.


Assuntos
Agricultura , COVID-19 , Humanos , China/epidemiologia , COVID-19/epidemiologia , COVID-19/prevenção & controle , Fazendeiros , Pandemias/prevenção & controle , População Rural , Turismo
9.
J Cancer ; 12(9): 2550-2559, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33854616

RESUMO

Pancreatic cancer is associated with poor prognosis due to limited therapeutic options. Excision repair cross-complementing 3 (ERCC3) is an important member of nucleotide excision repair (NER) that is overexpressed in some cancers and may be regarded as a poor prognostic factor. Yet, its role in pancreatic cancer remains unclear. This study aimed to investigate the expression and functions of ERCC3 in pancreatic cancer patients and its relation with clinicopathological features. Our data suggested that the protein expression level of ERCC3 was higher in tumor tissues than in adjacent tissues. In addition, the expression of ERCC3 has shown to be associated with the tumor extent (p=0.035). Besides, analysis of the dataset in The Cancer Genome Atlas (TCGA) revealed that high expression of ERCC3 was associated with poor overall survival in pancreatic cancer patients (p=0.0136). In Cox regression analysis, ERCC3 was an independent prognostic factor for overall survival in pancreatic cancer (p<0.001). Furthermore, our in vitro data further suggested that the overexpression of ERCC3 significantly promoted pancreatic cancer (BxPC-3, CFPAC-1, and PANC-1 cells) proliferation, invasion, and migration. Taken together, this study suggested that high expression of ERCC3 might be a poor prognostic factor in human pancreatic cancer and might be used as a promising therapeutic target for pancreatic cancer treatment.

10.
Int J Oncol ; 57(1): 197-212, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32319593

RESUMO

Glucose­6­phosphate dehydrogenase (G6PD) is crucial rate­limiting enzyme of the pentose phosphate pathway (PPP). G6PD dysregulation has been reported in various types of human cancer, and the role of G6PD in cancer progression was demonstrated in numerous studies. A previous study from our laboratory described the prognostic significance of G6PD in clear cell renal cell carcinoma (ccRCC), and demonstrated its proliferative role through positive feedback regulation of the phosphorylated form of signal transducer and activator of transcription 3. However, the role of G6PD in ccRCC invasion remains unclear. In the present study, reverse transcription­quantitative (RT­q) PCR, western blotting, enzyme activity assay, transwell assay and immunohistochemistry analysis in cell model, xenograft mice model and human specimen studies were performed to evaluate the role of G6PD in ccRCC invasion. The results from the present study demonstrated that G6PD may promote ccRCC cell invasive ability by increasing matrix metalloproteinase 2 (MMP2) mRNA and protein expression both in vitro and in vivo. In addition, a positive correlation between G6PD and MMP2 expression was demonstrated by RT­qPCR and western blotting in twenty pairs of ccRCC tumor specimens and matched adjacent normal tissues. Furthermore, G6PD promoted reactive oxygen species (ROS) generation and activated the MAPK signaling pathway in ccRCC cells. In addition, ROS significantly promoted the MAPK signaling pathway activation, which in turn contributed to MMP2 overexpression in ccRCC cells. In conclusion, the present study demonstrated that G6PD may facilitate ccRCC cell invasive ability by enhancing MMP2 expression through ROS­MAPK axis pathway.


Assuntos
Carcinoma de Células Renais/patologia , Glucosefosfato Desidrogenase/metabolismo , Neoplasias Renais/patologia , Metaloproteinase 2 da Matriz/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glucosefosfato Desidrogenase/genética , Humanos , Rim/patologia , Sistema de Sinalização das MAP Quinases/genética , Invasividade Neoplásica/patologia , Via de Pentose Fosfato/genética , Espécies Reativas de Oxigênio/metabolismo , Organismos Livres de Patógenos Específicos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Nutr Res ; 73: 1-14, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31835095

RESUMO

The purpose of this review is to discuss the molecular mechanisms underlying the anticancer properties of S-allylcysteine (SAC). Over the decades, evidence derived from in vitro and in vivo studies has shown that this predominant organosulfur component of aged garlic extract has multiple anticancer properties; hence, some potential mechanisms responsible for the anticarcinogenic action have been suggested. These mechanisms include induction of carcinogen detoxification, inhibition of cell proliferation and growth, mediation of cell cycle arrest, induction of cell death, inhibition of epithelial-mesenchymal transition and cell invasion, suppression of metastasis, and induction of immunomodulation in cancer cells. However, the actions and mechanisms are not comprehensive, and important aspects of the anticancer activities of SAC still need to be explored. In light of the current evidence, more specific studies, specifically clinical and epidemiological, are required to advance the promising use of SAC as a chemopreventive and therapeutic agent in cancer.


Assuntos
Antineoplásicos/farmacologia , Cisteína/análogos & derivados , Alho , Neoplasias/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisteína/farmacologia , Modelos Animais de Doenças , Humanos , Camundongos , Ratos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA