Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Cell Death Discov ; 9(1): 421, 2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-37993419

RESUMO

S-palmitoylation is a reversible protein lipidation that controls the subcellular localization and function of targeted proteins, including oncogenes such as N-RAS. The depalmitoylation enzyme family ABHD17s can remove the S-palmitoylation from N-RAS to facilitate cancer development. We previously showed that ABHD17C has oncogenic roles in hepatocellular carcinoma (HCC) cells, and its mRNA stability is controlled by miR-145-5p. However, it is still unclear whether ABHD17C is regulated at the post-translational level. In the present study, we identified multiple ubiquitin-specific proteases (USPs) that can stabilize ABHD17C by inhibiting the ubiquitin-proteasome-mediated degradation. Among them, USP35 is the most potent stabilizer of ABHD17C. We found a positive correlation between the elevated expression levels of USP35 and ABHD17C, together with their association with increased PI3K/AKT pathway activity in HCCs. USP35 knockdown caused decreased ABHD17C protein level, impaired PI3K/AKT pathway, reduced proliferation, cell cycle arrest, increased apoptosis, and mitigated migration and invasion. USP35 can interact with and stabilize ABHD17C by inhibiting its ubiquitination. Overexpression of ABHD17C can rescue the defects caused by USP35 knockdown in HCC cells. In support of these in vitro observations, xenograft assay data also showed that USP35 deficiency repressed HCC development in vivo, characterized by reduced proliferation and disrupted PI3K/AKT signaling. Together, these findings demonstrate that USP35 may promote HCC development by stabilization of ABHD17C and activation of the PI3K/AKT pathway.

3.
Dig Dis ; 41(5): 685-694, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37579733

RESUMO

BACKGROUND: Mild cellular atypia of esophageal squamous epithelial dysplasia has a risk of progressing to cancer that poses great confusion for pathological diagnosis. There is no research on the diagnosis and differential diagnosis of esophageal squamous dysplasia by the expression of immunohistochemical (IHC) p53. The study aims to conduct a graded diagnosis of esophageal squamous epithelial hyperplasia by combining p53 expressions and microscopic histomorphological characteristics. METHODS: The study was conducted from January 2021 to January 2022 and included a total of 208 cases including 262 specimens with atypical hyperplasia or dysplasia of squamous epithelia discovered by esophageal mucosal biopsy. HE staining was used to grade the epithelial hyperplasia degree, and all cases underwent p53 IHC evaluation. RESULTS: Benign lesions: we did not find any p53 IHC mutant-phenotype (0/12 cases) in 12 cases of esophagitis. We found 10 cases (10/80 cases) of p53 IHC mutant-phenotype in 80 cases of low-grade dysplasia, and 158 cases (158/170 cases) of p53 IHC mutant-phenotype of high-grade lesions in 170 cases of high-grade dysplasia and early cancer based on the χ2 test results. We found statistically significant differences in p53 IHC mutant-phenotype between the high-grade squamous epithelial lesions and benign lesions. The sensitivity and specificity of p53 in detecting high-grade squamous epithelial lesions were 92.9% and 89.1%, respectively. The positive predictive value was 94.0%, and the negative predictive value was 87.2%. CONCLUSION: In this study, we found that p53 IHC had high sensitivity and specificity in detecting high-grade esophageal squamous epithelial lesions. Therefore, it has potential to be used as a routine item in pathological detection for auxiliary risk stratification of esophageal squamous epithelial lesions.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Esofágicas , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Hiperplasia/diagnóstico , Hiperplasia/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Esofágicas/diagnóstico , Neoplasias Esofágicas/genética , Fenótipo
4.
Cell Biol Int ; 47(8): 1368-1380, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37076992

RESUMO

Ubiquitin-specific protease 33 (USP33) has been implicated in various cancers, but its biological function and mechanism of action remain unknown in pancreatic cancer (PCa) as a deubiquitinating enzyme. Herein, we report that USP33 silencing inhibits PCa cell survival and self-renewal. USPs highly expressed in spherical PCa cells were screened by comparing the levels of ubiquitin-specific proteases in spherical PCa cells and adherent PCa cells. After silencing USP, the effect of USP on the proliferation of PCa cells was detected by CCK-8 and colony formation assay, and the effect of USP on cell stemness was detected by tumor sphere formation assay, flow analysis, and western blot analysis. The interaction of USP with CTNNB1 and the effect of USP on the ubiquitination of CTNNB1 were verified by coimmunoprecipitation assay. After replenishing CTNNB1, cell proliferation and cell stemness were examined. USP33 is upregulated in spheric BXPC-3, PCNA-1, and SW1990, compared with adherent BXPC-3, PCNA-1, and SW1990. USP33 interacts with CTNNB1, and stabilizes CTNNB1 by suppressing its degradation. Furthermore, cell proliferation, colony-forming, and self-renewal abilities of PCa cells in vitro, and the expression of stem cell markers EpCAM and CD44, C-myc, Nanog, and SOX2, were suppressed when USP33 was knocked down, which was reversed when CTNNB1 was ectopically expressed in PCa cells. Thus, USP33 promotes PCa cell proliferation and self-renewal by inhibiting the degradation of CTNNB1. USP33 inhibition may be a new treatment option for PCa patients.


Assuntos
Regulação Neoplásica da Expressão Gênica , Células-Tronco Neoplásicas , Humanos , Linhagem Celular Tumoral , Sobrevivência Celular , Antígeno Nuclear de Célula em Proliferação/metabolismo , Movimento Celular , Ubiquitinação , Proliferação de Células , Células-Tronco Neoplásicas/metabolismo , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , beta Catenina/metabolismo
6.
Surg Endosc ; 36(7): 4869-4877, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34724579

RESUMO

BACKGROUND: T-tube drainage after laparoscopic common bile duct exploration (LCBDE) has been demonstrated to be safe and effective for patients with acute cholangitis caused by common bile duct stones (CBDSs). The outcomes after LCBDE with primary closure in patients with CBDS-related acute cholangitis are unknown. The present study aimed to evaluate the efficacy and safety of LCBDE with primary closure for the management of acute cholangitis caused by CBDSs. METHODS: Between June 2015 and June 2020, 368 consecutive patients with choledocholithiasis combined with cholecystolithiasis, who underwent laparoscopic cholecystectomy (LC) + LCBDE in our department, were retrospectively reviewed. A total of 193 patients with CBDS-related acute cholangitis underwent LC + LCBDE with primary closure of the CBD (PC group) and 62 patients underwent LC + LCBDE followed by T-tube placement (T-tube group). A total of 113 patients who did not have cholangitis were excluded. The clinical data were compared and analyzed. RESULTS: There was no mortality in either group. No significant differences were noted in morbidity, bile leakage rate, retained CBD stones, or readmission rate within 30 days between the two groups. Compared with the T-tube group, the PC group avoided T-tube-related complications and had a shorter operative time (121.12 min vs. 143.37 min) and length of postoperative hospital stay (6.59 days vs. 8.81 days). Moreover, the hospital expenses in the PC group were significantly lower than those in the T-tube group ($4844.47 vs. $5717.22). No biliary stricture occurred during a median follow-up of 18 months in any patient. No significant difference between the two groups was observed in the rate of stone recurrence. CONCLUSIONS: LCBDE with primary closure is a safe and effective treatment for cholangitis caused by CBDSs. LCBDE with primary closure is not inferior to T-tube drainage for the management of CBDS-related acute cholangitis in suitable patients.


Assuntos
Colangite , Colecistectomia Laparoscópica , Coledocolitíase , Cálculos Biliares , Laparoscopia , Colangite/etiologia , Colangite/cirurgia , Colecistectomia Laparoscópica/efeitos adversos , Coledocolitíase/complicações , Coledocolitíase/cirurgia , Ducto Colédoco/cirurgia , Cálculos Biliares/complicações , Cálculos Biliares/cirurgia , Humanos , Laparoscopia/efeitos adversos , Tempo de Internação , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/cirurgia , Estudos Retrospectivos
7.
Water Sci Technol ; 84(6): 1452-1463, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34559079

RESUMO

The effect of light has raised attention on wastewater treatment. However, little research has concentrated on the influences of light on activated sludge. In this study, the influences of light on the performance, quorum sensing (QS) and metagenomic characteristics of anoxic/oxic reactors were investigated. The reactor without light (AO1) showed higher total nitrogen (TN) removal (79.15 ± 1.69%) than the reactor with light (AO2) (74.54 ± 1.30%), and significant differences were observed. It was observed that light facilitated the production of protein-like and tryptophan-like substances by employing parallel factor analysis for extracellular polymeric substance (EPS), resulting in more EPS production in AO2, indicating light was beneficial to EPS production. The concentrations of N-acyl-homoserine lactones (AHLs) were various in the two reactors, so the AHLs-mediated QS behaviors in both reactors were also different. These results revealed that light significantly influenced nitrogen removal, EPS, and QS. Metagenomic analysis based on Tax4Fun demonstrated that light reduced the denitrification, stimulated the polysaccharide and protein biosynthesis pathways and down-regulated the AHLs synthesis pathway, resulting in lower TN removal, more EPS production, and lower AHLs concentrations. Based on the above, the likely mechanism was proposed for the influences of light on the reactor.


Assuntos
Matriz Extracelular de Substâncias Poliméricas , Percepção de Quorum , Acil-Butirolactonas , Reatores Biológicos , Metagenoma , Esgotos
8.
J Exp Clin Cancer Res ; 40(1): 177, 2021 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-34039401

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) cells-secreted exosomes (exo) could stimulate M2 macrophage polarization and promote HCC progression, but the related mechanism of long non-coding RNA distal-less homeobox 6 antisense 1 (DLX6-AS1) with HCC-exo-mediated M2 macrophage polarization is largely ambiguous. Thereafter, this research was started to unearth the role of DLX6-AS1 in HCC-exo in HCC through M2 macrophage polarization and microRNA (miR)-15a-5p/C-X-C motif chemokine ligand 17 (CXCL17) axis. METHODS: DLX6-AS1, miR-15a-5p and CXCL17 expression in HCC tissues and cells were tested. Exosomes were isolated from HCC cells with overexpressed DLX6-AS1 and co-cultured with M2 macrophages. MiR-15a-5p/CXCL17 down-regulation assays were performed in macrophages. The treated M2 macrophages were co-cultured with HCC cells, after which cell migration, invasion and epithelial mesenchymal transition were examined. The targeting relationships between DLX6-AS1 and miR-15a-5p, and between miR-15a-5p and CXCL17 were explored. In vivo experiment was conducted to detect the effect of exosomal DLX6-AS1-induced M2 macrophage polarization on HCC metastasis. RESULTS: Promoted DLX6-AS1 and CXCL17 and reduced miR-15a-5p exhibited in HCC. HCC-exo induced M2 macrophage polarization to accelerate migration, invasion and epithelial mesenchymal transition in HCC, which was further enhanced by up-regulated DLX6-AS1 but impaired by silenced DLX6-AS1. Inhibition of miR-15a-5p promoted M2 macrophage polarization to stimulate the invasion and metastasis of HCC while that of CXCL17 had the opposite effects. DLX6-AS1 mediated miR-15a-5p to target CXCL17. DLX6-AS1 from HCC-exo promoted metastasis in the lung by inducing M2 macrophage polarization in vivo. CONCLUSION: DLX6-AS1 from HCC-exo regulates CXCL17 by competitively binding to miR-15a-5p to induce M2 macrophage polarization, thus promoting HCC migration, invasion and EMT.


Assuntos
Carcinoma Hepatocelular/metabolismo , Quimiocinas CXC/metabolismo , Neoplasias Hepáticas/metabolismo , Macrófagos/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Movimento Celular/fisiologia , Polaridade Celular/fisiologia , Quimiocinas CXC/genética , Exossomos/metabolismo , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , RNA Longo não Codificante/genética , Transdução de Sinais
9.
Biochem Biophys Res Commun ; 527(1): 90-97, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32446397

RESUMO

Previous studies suggest that genistein protects liver from acetaminophen (APAP)-induced injury, however, the detailed mechanism of the process is still incompletely. Therefore, present study was to investigate the potential mechanism of the genistein mediated protection against APAP-induced hepatotoxicity. As shown, supplementation with 150 mg/kg genistein greatly alleviated the increase in serum alanine aminotransferase (ALT) activity, aspartate aminotransferase (AST) activity, hepatic malondialdehyde (MDA) contents, and reversed the decrease in hepatic GSH levels in response to overdose APAP. At the same time, hepatic SIRT1 protein and activity were markedly upregulated in mouse receiving genistein. However, the amelioration was almost abolished by the knockdown of hepatic SIRT1 expression using lentivirus carrying specific shRNA targeting SIRT1. These results were further validated by histopathology examination. Moreover, depletion of hepatic SIRT1 prevented the accumulation of Nrf2 in nucleus and the upregulation of the antioxidant gene expression in the presence of genistein and/or APAP. Concomitantly, the induced mRNA expression of UDP-glucuronosyltransferases (UGTs) by genistein was largely dependent on the SIRT1 expression and activity. Together, our results support the notion that the strong elevation of SIRT1 expression and activity may represent a potential mechanism of protection against APAP-induced liver injury by genistein.


Assuntos
Acetaminofen/efeitos adversos , Analgésicos não Narcóticos/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Genisteína/uso terapêutico , Substâncias Protetoras/uso terapêutico , Sirtuína 1/metabolismo , Animais , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/análise , Fator 2 Relacionado a NF-E2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/análise
10.
Biochem Biophys Res Commun ; 517(2): 253-259, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31349968

RESUMO

Gastric cancer (GC) is a common malignant tumor, and many studies have shown that circular RNAs (circRNAs) play important roles in the progress of GC. This study showed that circ_SPECC1 was down-regulated in various GC cell lines, significantly inhibited GC cell proliferation and invasion, and promote apoptosis, which might play an anti-oncogene role. Circ_SPECC1 was mainly located in the cytoplasm, and its sequence contained multiple potential binding sites of miR-526b. Pull-down experiments with biotinylated miR-526b mimics and circ_SPECC1 probe showed that they could enrich each other. RIP experiments found hat anti-AGO2 antibody could significantly enrich circ_SPECC1. Further dual luciferase reporter gene assay also confirmed that miR-526b could bind directly to circ_SPECC1. miR-526b was also down-regulated in GC cells, and one of its important target genes was KDM4A. Both circ_SPECC1 and miR-526b inhibited the expression of KDM4A and its downstream effector YAP1, but miR-526b inhibitors terminated the above-mentioned inhibition of circ_SPECC1, and KDM4A overexpression reversed the inhibition of circ_SPECC1 and miR-526b on YAP1 expression. Both miR-526b and KDM4A siRNA inhibited GC cell proliferation and invasion, and promote apoptosis; KDM4A overexpression had the opposite effects, and significantly blocked the regulation of miR-526b on cell growth and invasion. Therefore, circ_SPECC1 can enhance miR-526b inhibitory effect on downstream KDM4A/YAP1 pathway by adsorbing it, thus inhibiting GC cell growth and invasion. These findings enrich the mechanism of circRNAs in GC and will provide more new targets for the prevention and treatment of GC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Histona Desmetilases com o Domínio Jumonji/genética , MicroRNAs/genética , Invasividade Neoplásica/genética , RNA Circular/genética , Neoplasias Gástricas/genética , Fatores de Transcrição/genética , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica/patologia , Transdução de Sinais , Neoplasias Gástricas/patologia , Proteínas de Sinalização YAP
11.
Gene ; 690: 129-136, 2019 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-30597237

RESUMO

As an innovative CXC chemokine, CXCL17 has a mysterious clinical significance and modulating influence on hepatocellular carcinoma (HCC). Our study examined the activity and mechanisms of CXCL17 on growth, autophagy, and metastasis of HCC. Upregulation of CXCL17 expression was observed in HCC, which is correlated with poorer histological stages and outcomes. Elevation of CXCL17 expression promoted proliferation, invasion, and migration and decreased LC-3B biosynthesis and p62 protein reduction, which are known to stimulate autophagy. However, silencing of CXCL17 inhibited the development of these cancerous phenotypes. Furthermore, AMPK was stimulated after knockdown of CXCL17. This stimulation, as well as stimulation of autophagy was caused by liver kinase B1 (LKB1), whose function is induced by knockdown CXCL17. Additionally, knockdown of CXCL17 enhanced nuclear translocation of LKB1. Altogether, these findings suggest that elevated CXCL17 expression in HCC promotes malignant reactions in malignant cells. Our research offers new evidence that chemokine CXCL17 reinforces malignant invasion and suppresses autophagy via the LKB1-AMPK pathway.


Assuntos
Carcinoma Hepatocelular/patologia , Quimiocinas/genética , Neoplasias Hepáticas/patologia , Transdução de Sinais , Regulação para Cima , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Autofagia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células , Quimiocinas/metabolismo , Quimiocinas CXC , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Invasividade Neoplásica , Transplante de Neoplasias , Proteínas Serina-Treonina Quinases/metabolismo
12.
J Transl Med ; 16(1): 216, 2018 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-30068360

RESUMO

BACKGROUND: Little has been known about the role of non-coding RNA regulatory network in the patterns of growth and invasiveness of gastric cancer (GC) development. METHODS: MicroRNAs (miRNAs) microarray was used to screen differential miRNA expression profiles in Ming's classification. The significant differential expressions of representative miRNAs and their interacting circular RNA (circRNA) were confirmed in GC cell line and 63 pairs of GC samples. Then, a circRNA/miRNA network was constructed by bioinformatics approaches to identify molecular pathways. Finally, we explored the clinical value of the common targets in the pathway by using receiver operating characteristic curve and survival analysis. RESULTS: Significantly differential expressed miRNAs were found in two pathological types of GC. Both of miR-124 and miR-29b were consistently down-regulated in GC. CircHIPK3 could play a negative regulatory role on miR-124/miR-29b expression and associated with T stage and Ming's classification in GC. The bioinformatics analyses showed that targets expression of circHIPK3-miR-124/miR-29b axes in cancer-related pathways was able to predict the status of GC and associated with individual survival time. CONCLUSIONS: The targets of circHIPK3-miR-124/miR-29b axes involved in the progression of GC. CircHIPK3 could take part in the proliferation process of GC cell and may be potential biomarker in histological classification of GC.


Assuntos
Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , MicroRNAs/genética , RNA/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Biomarcadores Tumorais/genética , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Modelos Biológicos , Anotação de Sequência Molecular , Invasividade Neoplásica , Prognóstico , RNA/metabolismo , RNA Circular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/genética
13.
Int J Oncol ; 53(3): 1183-1192, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30015866

RESUMO

Golgi phosphorylated protein (GOLPH)3 is overexpressed in colorectal cancer tissues and promotes the proliferation of colon cancer cells. A previous study by the authors demonstrated that GOLPH3 was associated with poor prognosis in colorectal cancer. However, the association between GOLPH3 gene overexpression and resistance to platinum-based drugs in colon cancer remains unknown. In the present study, the association between GOLPH3 overexpression and resistance of HT29 colon cancer cells to cisplatin and the mechanism underlying the development of chemoresistance were investigated. HT29 cells were divided into five groups. The expression of GOLPH3 mRNA was measured in the control and siRNA transfection groups. Reverse transcription-quantitative polymerase chain reaction analysis, cell proliferation, colony formation assay, tumor sphere formation and apoptosis (Annexin V) assays, western blotting and a nude mouse tumorigenicity assay were performed. HT29 cells were resistant to 10 µM cisplatin treatment, whereas the expression of GOLPH3, P-glycoprotein, phosphorylated extracellular signal-regulated kinase (pERK)1/2 and ß-catenin protein was significantly upregulated compared with the control group. With cisplatin treatment, silencing GOLPH3 gene expression downregulated the expression of these proteins, reduced cell proliferation and tumorigenicity, induced apoptosis and reversed the resistance of HT29 cells to cisplatin. In addition, the change in pERK1/2 and ß-catenin expression demonstrated that the mechanism of GOLPH3 overexpression involved in cisplatin resistance was associated with activation of the mitogen-activated protein kinase/ERK and Wnt/ß­catenin signaling pathways in HT29 cells. The tumorigenicity experiment in nude mice also demonstrated that silencing GOLPH3 expression increased the sensitivity of HT29 cells to cisplatin in vivo. Therefore, overexpression of GOLPH3 may be involved in the resistance of HT29 colon cancer cells to cisplatin chemotherapy by activating multiple cell signaling pathways.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/metabolismo , Animais , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cisplatino/uso terapêutico , Neoplasias Colorretais/patologia , Regulação para Baixo , Feminino , Inativação Gênica , Células HT29 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Esferoides Celulares , Regulação para Cima , Via de Sinalização Wnt/efeitos dos fármacos , Via de Sinalização Wnt/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
World J Gastroenterol ; 23(13): 2337-2345, 2017 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-28428713

RESUMO

AIM: To determine the potential roles of CD4 and microRNA (miR)-145 in gastric cancer. METHODS: The levels of CD44 and miR-145 were determined in gastric cancer cells. Quantitative real-time polymerase chain reaction was used to measure to the level of CD44 mRNA. A luciferase reporter assay and western blotting were performed to examine the effect of miR-145 on CD44 expression. Tumor sphere and MTT assays were carried out to evaluate the self-renewal and chemo-resistance properties of gastric cancer cells. RESULTS: The expression of CD44 was greatly increased and miR-145 was decreased in gastric cancer cells that were highly enriched in cancer stem cells (CSCs). The results demonstrated that miR-145 regulated CD44 by targeting directly the CD44 3'-untranslated region (3'-UTR). In gastric cancer cells, overexpression of miR-145 repressed the activity of the CD44 3'-UTR, and disruption of miR-145/CD44 3'-UTR interactions abrogated the silencing effects. In addition, miR-145 inhibition stimulated CD44 3'-UTR activity and disruption of miR-145/CD44 3'-UTR interactions abrogated this stimulatory effect. Enforced CD44 expression greatly increased tumor sphere formation and chemo-resistance in gastric cancer cells. Furthermore, the inhibition of CSCs and the chemo-sensitivity of gastric cancer cells treated with miR-145 were significantly abrogated by overexpression of CD44. CONCLUSION: miR-145 targeting of CD44 plays critical roles in the regulation of tumor growth and chemo-resistance in gastric cancer.


Assuntos
Receptores de Hialuronatos/metabolismo , MicroRNAs/metabolismo , Neoplasias Gástricas/metabolismo , Linhagem Celular Tumoral , Autorrenovação Celular , Resistencia a Medicamentos Antineoplásicos , Humanos
15.
Zhonghua Zhong Liu Za Zhi ; 35(5): 325-30, 2013 May.
Artigo em Chinês | MEDLINE | ID: mdl-24054006

RESUMO

OBJECTIVE: To study the function and clinicopathological significance of RNA-29c (miR-29c) in the carcinogenesis and development of gastric cancer. METHODS: MicroRNA microarray was applied to assess the miRNAs expression profile of gastric cancer. Quantitative real-time PCR was used to detect the expression of miR-29c in 64 cases of gastric cancer tissues and corresponding normal gastric epithelium, as well as cell lines GES-1, BGC-823 and SGC-7901 cells. MTT assay and flow cytometry were applied to detect the effects of forced expression of miR-29c in gastric cancer BGC-823 cells including cell proliferation, apoptosis, cell cycle and drug sensitivity. Quantitative real-time PCR, Western blot and luciferase reporter assay were used to explore the targeted relationship between miR-29c and myeloid cell leukemia-1 (Mcl-1). RESULTS: Compared with normal gastric epithelium, seven microRNAs (miR-374b*, miRPlus-E1212, miR-338-5p, miR-297, miR-21, miR-135b, miR-18a) were significantly up-regulated more than 2-folds, and nine microRNAs (miR-29b-2*, miR-1260, miRPlus-E1241, miR-S1-5p, miR-148a, miR-29c, miR-647, miR-196b*, ebv-miR-BART5) were significantly down-reguated in gastric cancer tissues. The average expression level of miR-29c in gastric cancer tissues was 0.70 ± 0.34 and in corresponding normal epithelium was 1.00 ± 0.06 (P < 0.05). miR-29c expression was related to tumor size, lymph node metastasis, clinical stage, Laurén classification, Borrmann classification and Ming classification (P < 0.05). The poorer differentiation degree of gastric cell lines, the lower was miR-29c expression level (P < 0.05). Overexpression of miR-29c in gastric cancer BGC-823 cells suppressed cell proliferation, stimulated cell apoptosis, induced cell cycle arrest in S phase and increased the chemotherapy sensitivity to drug docetaxel (all were P < 0.05). The average expression level of Mcl-1 mRNA in gastric cancer tissues was 3.47 ± 1.34 and corresponding epithelialium was 1.00 ± 0.20 (P < 0.05). The expression level of miR-29c was negatively related with that of Mcl-1 mRNA in gastric cancer tissues. miR-29c directly targeted to regulation of Mcl-1 expression. CONCLUSIONS: There are special miRNA expression profile in gastric cancer. The expression of miR-29c is closely related to biological behavior of human gastric cancer. miR-29c is involved in targeted regulation of Mcl-1, and may be one of mechanisms of the carcinogenesis of gastric cancer.


Assuntos
Proliferação de Células , MicroRNAs/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Neoplasias Gástricas , Antineoplásicos/uso terapêutico , Apoptose , Pontos de Checagem do Ciclo Celular , Diferenciação Celular , Linhagem Celular Tumoral , Docetaxel , Resistencia a Medicamentos Antineoplásicos , Regulação Neoplásica da Expressão Gênica , Humanos , Metástase Linfática , MicroRNAs/genética , Análise em Microsséries , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Estadiamento de Neoplasias , RNA Mensageiro/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Taxoides/uso terapêutico , Transcriptoma , Carga Tumoral
16.
Gut ; 62(5): 716-26, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22490519

RESUMO

OBJECTIVE: Deficiency or reduced expression of signal transduction and activation of RNA family protein Quaking (Qki) is associated with developmental defects in neural and vascular tissues and the development of debilitating human diseases including colorectal cancer (CRC). However, the mechanisms underlying the aberrant downregulation or deficiency of Qki were uncertain. DESIGN: Expression of miR-574-5p, Qki5/6/7/7b splicing variants, ß-catenin and p27(Kip1) was determined in mouse and human CRC cells and tissues to investigate the post-transcriptional regulation of Qki isoforms by miR-574-5p and its impact on ß-catenin/p27(Kip1) signalling, cell cycle progression, proliferation, migration, invasion and tumour growth. RESULTS: In the CRC tissues of C57BL/6-Apc(min/+) mice, miR-574-5p was found to be significantly upregulated and negatively correlated with the expression of Qki but positively correlated with the expression of ß-catenin. In mouse and human CRC cells, miR-574-5p was shown to regulate Qki isoforms (Qki6/7 in particular) post-transcriptionally and caused altered expression in ß-catenin and p27(Kip1) , increased proliferation, migration and invasion and decreased differentiation and cell cycle exit. Furthermore, in clinical CRC tissues, miR-574-5p was shown to be greatly upregulated and inversely correlated with the expression of Qkis. Finally, inhibition of miR-574-5p was shown to suppress the growth of tumours in the nude mice. CONCLUSIONS: Together, these novel findings suggest that miR-574-5p is a potent ribo-regulator for Qkis and that aberrant miR-574-5p upregulation can be oncogenic.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/metabolismo , MicroRNAs/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animais , Biomarcadores Tumorais/genética , Neoplasias Colorretais/patologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , MicroRNAs/genética , Neoplasias Experimentais , Inibidores de Proteínas Quinases/metabolismo , Proteínas de Ligação a RNA/genética , Reação em Cadeia da Polimerase em Tempo Real , Transplante Heterólogo , Proteínas Wnt/genética , beta Catenina/genética
17.
Zhonghua Yi Xue Za Zhi ; 92(46): 3269-73, 2012 Dec 11.
Artigo em Chinês | MEDLINE | ID: mdl-23328512

RESUMO

OBJECTIVE: To explore the function and clinicopathological significance of miR-135b in the occurrence and development of gastric cancer. METHODS: Seventy-two pairs of fresh gastric cancer tissues and corresponding normal gastric epithelium were collected at our department from September 2007 to March 2011. Six of them were randomly selected and miRNA microarray was applied to study the miRNA expression profiles of gastric cancer. Quantitative real-time PCR was used to validate the reliability of microarray and detect miR-135b expression in the above clinical samples, as well as cell lines GES-1, BGC-823 and SGC-7901. The methods of cell transfection, thiazolyl blue tetrazolium bromide (MTT) and flow cytometry were used to study the impact of miR-135 on cell proliferation, cell cycle and apoptosis of gastric cancer cells. Real-time quantitative PCR and Western blot were used to explore the relationship between miR-135b and adenomatous polyposis coli (APC). RESULTS: Compared with normal gastric tissues, 7 miRNA were significantly up-regulated and 9 miRNA significantly down-regulated for over 2 folds in gastric cancer tissues (P < 0.05). The results of microarray were verified by quantitative real-time PCR. MiR-135b expression was up-regulated in most clinical samples compared with their corresponding epithelium (n = 66, 91.67%). The average expression level of miR-135b in gastric cancer tissues was significantly higher than normal epithelium (3.42 ± 2.62 vs 1.00 ± 0.07, P < 0.05). MiR-135b was related to Laurén classification, tumor differentiation, invasion and pathologic tumor-node-metastasis (pTNM) stage of gastric cancer (all P < 0.05). The worse differentiation degree of gastric cell lines, the higher miR-135b expression level (P < 0.05). MiR-135b promoted gastric cancer cell proliferation, inhibited its apoptosis and directly regulated the expression of APC in gastric cancer cell. CONCLUSIONS: Special miRNA expression profiles of gastric cancer are found. MiR-135b is closely correlated with the biological behavior of human gastric cancer. And its regulation of APC may be one of the pathogenic mechanisms of gastric cancer.


Assuntos
MicroRNAs/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Proteína da Polipose Adenomatosa do Colo/metabolismo , Idoso , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , Análise em Microsséries , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Gástricas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA