Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
World J Gastrointest Oncol ; 16(5): 1925-1946, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38764837

RESUMO

BACKGROUND: The treatment of gastric cancer (GC) has caused an enormous social burden worldwide. Accumulating studies have reported that N6-methyladenosine (m6A) is closely related to tumor progression. METTL5 is a m6A methyltransferase that plays a pivotal role in maintaining the metabolic stability of cells. However, its aberrant regulation in GC has not been fully elucidated. AIM: To excavate the role of METTL5 in the development of GC. METHODS: METTL5 expression and clinicopathological characteristics were analyzed via The Cancer Genome Atlas dataset and further verified via immunohistochemistry, western blotting and real-time quantitative polymerase chain reaction in tissue microarrays and clinical samples. The tumor-promoting effect of METTL5 on HGC-27 and AGS cells was explored in vitro by Cell Counting Kit-8 assays, colony formation assays, scratch healing assays, transwell assays and flow cytometry. The tumor-promoting role of METTL5 in vivo was evaluated in a xenograft tumor model. The EpiQuik m6A RNA Methylation Quantification Kit was used for m6A quantification. Next, liquid chromatography-mass spectrometry was used to evaluate the association between METTL5 and sphingomyelin metabolism, which was confirmed by Enzyme-linked immunosorbent assay and rescue tests. In addition, we investigated whether METTL5 affects the sensitivity of GC cells to cisplatin via colony formation and transwell experiments. RESULTS: Our research revealed substantial upregulation of METTL5, which suggested a poor prognosis of GC patients. Increased METTL5 expression indicated distant lymph node metastasis, advanced cancer stage and pathological grade. An increased level of METTL5 correlated with a high degree of m6A methylation. METTL5 markedly promotes the proliferation, migration, and invasion of GC cells in vitro. METTL5 also promotes the growth of GC in animal models. METTL5 knockdown resulted in significant changes in sphingomyelin metabolism, which implies that METTL5 may impact the development of GC via sphingomyelin metabolism. In addition, high METTL5 expression led to cisplatin resistance. CONCLUSION: METTL5 was found to be an oncogenic driver of GC and may be a new target for therapy since it facilitates GC carcinogenesis through sphingomyelin metabolism and cisplatin resistance.

2.
Front Oncol ; 12: 1058028, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36561519

RESUMO

Objective: With the prevalence of next-generation sequencing (NGS) technology, a large number of long non-coding RNAs (lncRNAs) have attracted tremendous attention and have been the topic of extensive research on gastric cancer (GC). It was revealed that lncRNAs not only participate in the transduction of various signaling pathways, thus influencing GC genesis and development, but also have the potential for GC diagnosis. Therefore, we aimed to conduct a meta-analysis of previous studies on GC. Materials and methods: An electronic search was made before August 2021 on databases including PubMed, Embase, and Web of Science. Relevant articles that compare lncRNA expression in GC patients and healthy controls were summarized. We conducted a meta-analysis with the objective of evaluating the ability of lncRNAs in diagnosing GC. Results: A total of 40 original research studies including 6,772 participants were discussed in this meta-analysis. The overall sensitivity, specificity, and the area under the curve (AUC) were 0.78 (95% CI: 0.75-0.81), 0.79 (95% CI: 0.74-0.83), and 0.85 (95% CI: 0.81-0.87), respectively. The value of pooled diagnostic odds ratios (DORs) was 13.00 (95% CI: 10.00-17.00). Conclusions: This meta-analysis revealed that serum or plasma lncRNAs have high sensitivity and specificity, which makes lncRNAs clinically feasible in diagnosing GC. The results from this meta-analysis demonstrated that peripheral blood lncRNAs may become novel noninvasive biomarkers in the foreseeable future. At the same time, it should be noted that a greater number of blood samples and more evidence from rigorous multicenter clinical studies are necessary to justify their applicability as cancer biomarkers.

3.
Regen Ther ; 21: 25-33, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35663842

RESUMO

Background: Gastric cancer (GC) progression is related with gene regulations. Objectives: This study explored underlying regulatory axis of circRNA PVT1 (circPVT1) in GC. Methods: GC cell lines were detected for circPVT1 expression with the normal mucous epithelial cell GES-1 as control. After regulation of circPVT1, miR-423-5p and SMAD3 expression through transfection, CCK8 evaluated the cell viability, Transwell measured the migratory and invasive capability of cells. Luciferase verified the paired bindings between miR-423-5p and CircPVT1 or SMAD3. The functions of CircPVT1/miR-423-5p/SMAD3 were evaluated using RT-PCR, CCK8, Transwell assays. Western blot analyzed EMT-related proteins and phosphorylation of Smad3 in GC cells. Immunofluorescence method was used to evaluate the EMT-related proteins as well. Results: CircPVT1 displayed higher expression in GC cells and knockdown led to decrease in cell growth, invasion and migration. CircPVT1 was targeted by miR-423-5p as a ceRNA of SMAD3. miR-423-5p upregulation suppressed both cicRNA PVT1 and SMAD3 in GC cells. Decrease in SMAD3 expression suppressed CircPVT1 by releasing miR-423-5p in cells, inhibiting cell growth, invasion and migration and suppressing the EMT process. Conclusion: CircPVT1 modulated cell growth, invasion and migration through EMT mediation in gastric cancer through miR-423-5p/Smad3 pathway.

4.
Curr Mol Pharmacol ; 15(3): 557-569, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34139974

RESUMO

BACKGROUND: Gastric Cancer (GC) is the fifth most common malignancy tumor and the third cause of cancer-related death around the world. Immune checkpoint inhibitors (ICIs) such as programmed cell death-1 (PD-1) antibodies play an active role in tumor therapy. A recent study reveals that Wnt/ß-catenin signaling pathway is negatively correlated with T-cell infiltration in tumor microenvironment (TME), thereby influencing the therapeutic efficacy of PD-1 antibody. OBJECTIVE: In this study, we aimed to uncover the relationship of Wnt/ß-catenin pathway to CD8+ T cell activity as well as its effect on anti-PD-1 therapeutic efficacy in GC. METHODS AND RESULTS: We first collected clinical samples and went through an immunohistochemical analysis and found that a high ß-catenin expression in GC tissues was often associated with a significant absence of CD8+ T-cell infiltration. In addition, our data further indicated that disruption of the Wnt/ß-catenin pathway in GC cells inhibited their migratory and invasive ability. Meanwhile, enhanced sensitivity of GC cells to PD-1 blockade therapy was evident by decreased Jurkat cell apoptosis rate and increased GC cell apoptosis rate in a tumor and Jurkat cells co-culture system with the presence of Wnt/ß-catenin pathway inhibition. CONCLUSION: Collectively, these findings indicated Wnt/ß-catenin pathway may play a significant role in modulating the activity of Jurkat cells and downregulation of Wnt/ß-catenin may enhance the sensitivity of GC cells to PD-1 antibody in vitro. This result further indicated that ß-catenin and PD-1 targeted inhibition might become a potential and effective therapy for GC patients.


Assuntos
Neoplasias Gástricas , beta Catenina , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Receptor de Morte Celular Programada 1/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Microambiente Tumoral , Via de Sinalização Wnt , beta Catenina/metabolismo
5.
Front Immunol ; 12: 702615, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34322132

RESUMO

Immune checkpoint blockade has attracted a lot of attention in the treatment of human malignant tumors. We are trying to establish a prognostic model of gastric cancer (GC) based on the expression profile of immunoregulatory factor-related genes. Based on the TCGA database, we identified 234 differentially expressed immunoregulatory factors. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) conducted enrichment analysis to clarify the biological functions of differential expression of immunoregulatory factors. STRING database predicted the interaction network between 234 differently expressed immune regulatory factors. The expression of 11 immunoregulatory factors was significantly related to the overall survival of gastric cancer patients. Univariate Cox regression analysis, Kaplan-Meier analysis and multivariate Cox regression analysis found that immunomodulatory factors were involved in the progression of gastric cancer and promising biomarkers for predicting prognosis. Among them, CXCR4 was related to the low survival of GC patients and a key immunomodulatory factor in GC. Based on TCGA data, the high expression of CXCR4 in GC was positively correlated with the advanced stage and grade of gastric cancer and related to poor prognosis. Univariate analysis and multivariate analysis indicated that CXCR4 was an independent prognostic indicator for TCGA gastric cancer patients. In vitro functional studies had shown that CXCR4 promoted the proliferation, migration, and invasion of gastric cancer cells. In summary, this study has determined the prognostic value of 11 immunomodulatory factors in gastric cancer. CXCR4 is an independent prognostic indicator for gastric cancer patients, which may help to improve the individualized prognostic prediction of GC and provide candidates for the diagnosis and treatment of GC.


Assuntos
Biomarcadores Tumorais/imunologia , Receptores CXCR4 , Neoplasias Gástricas/imunologia , Adulto , Biomarcadores Tumorais/metabolismo , Feminino , Perfilação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Transcriptoma
6.
Aging (Albany NY) ; 12(1): 707-717, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31915311

RESUMO

LMX1A (LIM homeobox transcription factor 1α) is a tumor suppressor protein. Our previous study has shown that microRNA-9 ("miR-9"), being upregulated in human gastric cancer (GC), targets LMX1A to promote GC cell progression. Through searching long non-coding RNA (LncRNA) database, we identified that LncRNA KCNQ1OT1 is the competing endogenous RNA (ceRNA) of miR-9. KCNQ1OT1 putatively targets miR-9. Its level is downregulated in human GC tissues. In AGS cells and primary human GC cells, forced overexpression of KCNQ1OT1, by a lentiviral construct, induced miR-9 downregulation and LMX1A upregulation. Furthermore, KCNQ1OT1 overexpression inhibited GC cell survival, proliferation, migration and invasion, but inducing apoptosis activation. Contrarily, KCNQ1OT1 silencing, by targeted siRNAs, induced miR-9 accumulation and LMX1A downregulation. Consequently, GC cell proliferation, migration and invasion were enhanced. Importantly, KCNQ1OT1 overexpression or silencing was ineffective in LMX1A knockout AGC cells. Taken together, KCNQ1OT1 inhibits GC cell progression via regulating miR-9 and LMX1A expression.


Assuntos
Regulação Neoplásica da Expressão Gênica , Proteínas com Homeodomínio LIM/genética , MicroRNAs/genética , RNA Longo não Codificante , Neoplasias Gástricas/genética , Fatores de Transcrição/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/genética , Progressão da Doença , Humanos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Interferência de RNA , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
7.
J Cell Physiol ; 235(5): 4388-4398, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31637706

RESUMO

Gastric cancer (GC) is one of the most frequent malignancies worldwide. Long noncoding RNAs (lncRNAs) are found to be largely implicated in various cancers, including GC. However, the function of lncRNA VCAN antisense RNA 1 (VCAN-AS1) in GC remains unclear. Herein, we observed a low level of VCAN-AS1 in normal gastric tissues through NCBI and UCSC, and that VCAN-AS1 upregulation in GC tissues was related to poor prognosis by TCGA. Furthermore, VCAN-AS1 was found markedly enhanced in GC tissues and cell lines, while its upregulation was related with clinical outcomes of GC patients. Besides this, silencing VCAN-AS1 represses cell proliferation, migration, and invasion but enhances apoptosis. More important, we discovered that VCAN-AS1 expression was negatively correlated with wild-type p53 levels in GC tissues and that p53 was negatively modulated by VCAN-AS1 in GC cells. Furthermore, p53 suppression reversed the repression of VCAN-AS1 silence on the biological processes of AGS cells. Intriguingly, we identified that both VCAN-AS1 and TP53 can bind with eIF4A3, one of the core proteins in the exon junction complex. Also, we confirmed that VCAN-AS1 negatively regulates TP53 expression by competitively binding with eIF4A3. Our findings disclosed that VCAN-AS1 contributes to GC progression through interacting with eIF4A3 to downregulate TP53 expression, indicating that VCAN-AS1 is a novel therapeutic strategy for GC treatment.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Invasividade Neoplásica/fisiopatologia , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Adulto , Linhagem Celular Tumoral , Movimento Celular , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , RNA Longo não Codificante/genética , Proteína Supressora de Tumor p53/genética
8.
PLoS One ; 14(9): e0221640, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31557193

RESUMO

Our research group has showed that the LIM homeobox transcription factor 1 alpha (LMX1A) is inactivated in gastric cancers. Overexpression of LMX1A inhibits tumor growth. However, the mechanisms remains unclear. Considering LMX1A as a transcription factor, a comparison of RNA-seq between gastric cancer cells (GCCs) and GCCs with LMX1A overexpressed was performed to identify genes transcriptionally activated by LMX1A. Among the potential LMX1A target genes, angiopoietin-like 4 (ANGPTL4) has been reported to be an important tumor suppressor and thus was selected for further validation and research. Both LMX1A and ANGPTL4 showed downregulated expression in gastric cancer samples. More importantly, the expression of LMX1A is positively correlated with ANGPTL4, without including other family members in gastric cancer cell lines. What's more, knockdown of ANGPTL4 rescued the tumor suppressive phenotype of LMX1A overexpression, which indicated that LMX1A upregulates ANGPTL4 to exert its role. Mechanistically, we found that LMX1A inhibited the expression of the oncogene C-Myc, which is alleviated by ANGPTL4 knockdown. In general, our results showed that LMX1A exerts its tumor suppressive role by activating ANGPTL4 to inhibit C-Myc.


Assuntos
Proteína 4 Semelhante a Angiopoietina/genética , Proteína 4 Semelhante a Angiopoietina/metabolismo , Genes myc , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 4 Semelhante a Angiopoietina/antagonistas & inibidores , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Supressores de Tumor , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
9.
Cell Prolif ; 52(6): e12687, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31468594

RESUMO

OBJECTIVES: As one of the most life-threatening malignancies, gastric cancer is the third contributor of cancer mortalities globally. Increasing studies have proven the regulatory roles of lncRNAs in the development of diverse malignant tumours. But little is known about its function and molecular mechanism in gastric carcinoma. MATERIALS AND METHODS: RT-qPCR was performed to measure the expression pattern of LOXL1-AS1 in gastric cancer. To ascertain its definite role, CCK-8, EdU, Western blot, transwell and sphere formation assays were adopted. RNA pull-down, RIP, ChIP and luciferase reporter assays were carried out to investigate the molecular mechanism of LOXL1-AS1 in gastric carcinoma. RESULTS: LOXL1-AS1 was highly expressed in tissues and cells of gastric cancer. The upregulation of LOXL1-AS1 predicted poor prognosis in gastric carcinoma. Our findings demonstrated that LOXL1-AS1 accelerated the deterioration of gastric cancer by inducing cell proliferation, migration, EMT and stemness. Moreover, the expression of USF1 in gastric cancer was higher than in normal control and LOXL1-AS1 negatively modulated USF1. Functionally, LOXL1-AS1 acted as a ceRNA to upregulate USF1 via sponging miR-708-5p. Besides, we confirmed USF1 promoted the transcription of stemness marker SOX2. Rescue experiments testified the stimulative role of LOXL1-AS1/miR-708-5p/USF1 pathway in gastric cancer progression. It was also validated that LOXL1-AS1 facilitated cell growth of gastric carcinoma in vivo. CONCLUSIONS: Our study unravelled that LOXL1-AS1/miR-708-5p/USF1 pathway contributed to the development of gastric cancer.


Assuntos
Aminoácido Oxirredutases/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Fatores Estimuladores Upstream/genética , Carcinogênese/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos
10.
Biochem Biophys Res Commun ; 511(1): 13-20, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30765226

RESUMO

Gastric cancer (GC) is a common malignancy. Developing novel and efficient anti-GC agents is urgent. GSK1059615 is a PI3K (phosphatidylinositol 3-kinase) and mTOR (mammalian target of rapamycin) dual inhibitor. It activity in human GC cells is tested here. In AGS cells and primary human GC cells, GSK1059615 potently inhibited cell growth, survival, proliferation and cell cycle progression. Further, significant apoptosis activation was detected in GSK1059615-treated GC cells. Contrarily in the primary human gastric epithelial cells, GSK1059615 failed to induce significant cytotoxicity and apoptosis. GSK1059615 blocked PI3K-AKT-mTOR cascade activation, inducing microRNA-9 downregulation but LMX1A (LIM homeobox transcription factor 1α) upregulation in GC cells. Significantly, GSK1059615 administration (i.p., daily, at 10 or 30 mg/kg) in nude mice potently inhibited subcutaneous AGS xenograft growth. AKT-mTOR inhibition and LMX1A upregulation were detected in AGS xenograft tissues with GSK1059615 administration. Together, we conclude that GSK1059615 inhibits GC cell growth in vitro and in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Serina-Treonina Quinases TOR/metabolismo
11.
Biochem Biophys Res Commun ; 505(2): 405-412, 2018 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-30262143

RESUMO

LIM homeobox transcription factor 1, alpha (LMX1A) is downregulated in human gastric cancer (GC), functioning as a tumor suppressor. The current study aims to identify specific microRNA that can regulate LMX1A expression. By sequence analysis of LMX1A mRNA 3'-untranslated region (3'-UTR), we show that microRNA-9 (miR-9) putatively targets human LMX1A. In established (AGS cells) and primary human GC cells, ectopic overexpression of miR-9 by a lentiviral construct decreased LMX1A 3'-UTR activity, causing LMX1A mRNA and protein downregulation. Functional analyses show that miR-9 overexpression enhanced GC cell survival and proliferation. On the contrary, miR-9 inhibition by antagomir-9 lentivirus increased LMX1A 3'-UTR activity to upregulate LMX1A mRNA and protein expression, causing GC cell apoptosis. CRISPR/Cas9-mediated LMX1A knockout promoted AGS cell survival and proliferation. Importantly, miR-9 and antagomiR-9 were ineffective to the function of LMX1A-knockout AGS cells. In human GC tissues miR-9 is upregulated, which is negatively correlated with LMX1A downregulation. Together, we conclude that miR-9 selectively targets LMX1A to promote GC cell progression.


Assuntos
Proteínas com Homeodomínio LIM/antagonistas & inibidores , MicroRNAs/farmacologia , Neoplasias Gástricas/genética , Fatores de Transcrição/antagonistas & inibidores , Regiões 3' não Traduzidas/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas com Homeodomínio LIM/genética , Neoplasias Gástricas/patologia , Fatores de Transcrição/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...