Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Biochem Funct ; 38(1): 28-37, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31710389

RESUMO

Gastric cancer (GC) is one of the most common causes of cancer-related death worldwide, and outstanding biomarkers for therapeutic targets or predicting GC survival are still lacking. Increasing evidence indicated that nucleolar and spindle associated protein 1 (NUSAP1) involved in regulating the progression of various cancers; however, its specific role in GC remained unclear. In this study, we found that NUSAP1 was upregulated in the GC tissues and cell lines via analysing data from The Cancer Genome Atlas (TCGA), gene expression omnibus (GEO), qRT-PCR, and western blot assays. Patients with high NUSAP1 expression levels showed shorter free-progression survival (FPS), larger tumour size, and higher lymphatic metastasis rate compared with those with low NUSAP1 expression. Further functional experiments revealed knockdown of NUSAP1 could inhibit the growth, migration, and invasion of GC cells in vitro and vivo. Additionally, silencing NUSAP1 induced G0/G1 phase arrest, apoptosis, and suppressed the epithelial-mesenchymal transition (EMT) process. Finally, we performed gene set enrichment analysis (GSEA) and observed NUSAP1 was positive with mTORC1 signalling pathway, which was verified by the subsequent immunoblotting. In conclusion, our findings suggested that NUSAP1 contributed to GC progression and may act as a potential therapeutic target for GC. SIGNIFICANCE OF THE STUDY: Our results firstly illuminated that NUSAP1 expression was significantly upregulated in GC tissues and predicted poor FPS. Silencing it could attenuate GC progression via inhibiting mTORC1 signalling pathway. Hence, NUSAP1 may act as a promising therapy target for GC.


Assuntos
Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/biossíntese , Neoplasias Experimentais/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Biologia Computacional , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Relação Estrutura-Atividade
2.
Clin Lab ; 66(8)2020 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-32776742

RESUMO

BACKGROUND: Recently, accumulating evidence has suggested that the long noncoding RNA small nucleolar RNA host gene 15 (lncRNA SNHG15) was elevated in various malignancies and correlated to poor clinical outcome of patients. However, the prognosis value of SNHG15 in tumors remains not well understood. METHODS: The PubMed, Web of Science, Embase, Ovid, Cochrane Library databases were used to search for eligible articles. Stata MP14.0 software was applied in the systematic meta-analysis. The Cancer Genome Atlas (TCGA) dataset was adopted to verify the results. RESULTS: A total of 13 studies including 1,190 patients were enrolled in this meta-analysis. SNHG15 high expression predicted shorter overall survival (OS) (hazard ratio (HR) = 2.34, 95% confidence interval (CI): 1.75 - 3.12, p < 0.001) with no statistical heterogeneity, which was validated by the data of TCGA. The subgroup analyses stratified according to OS analysis method, cancer type, sample size, and follow-up time showed similar results. Additionally, SNHG15 expression was positively associated with TNM stage (III + IV vs. I + II, odds ratio (OR) = 2.23, 95% CI: 1.14 - 4.38, p = 0.020) and poor differentiation (low + undifferentiated vs. well + moderate, OR = 2.89, 95% CI: 1.89 - 4.42, p < 0.001). CONCLUSIONS: IncRNA SNHG15 may act as a useful and potential biomarker for prognosis and clinical parameters in human cancers.


Assuntos
Neoplasias , RNA Longo não Codificante , Biomarcadores Tumorais/genética , Biologia Computacional , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Prognóstico , RNA Longo não Codificante/genética
3.
J Cell Biochem ; 120(9): 15190-15201, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31021450

RESUMO

Gastric cancer (GC) is one of the most common cancers worldwide and has especially high morbidity and mortality in China. LEM domain containing 1 (LEMD1), an important cancer-testis antigen, has been reported to be overexpressed in various cancers and promotes the progression of cancers. However, the biological characteristics of LEMD1 remain to be explored in GC. The connection between LEMD1 expression and GC progression was analyzed by using The Cancer Genome Atlas datasets and our human microarray datasets. A Kaplan-Meier plot was used to analyze the relationship between LEMD1 expression and prognosis. The expression of LEMD1 was analyzed by quantitative real-time polymerase chain reaction and Western blot, and the proliferation ability of GC cells was analyzed by cell proliferation and colony formation assays and 5-ethynyl-2'-deoxyuridine analysis. The cell cycle and apoptosis were analyzed by flow cytometry. Furthermore, subcutaneously implanted tumor models in nude mice were used to demonstrate the role of LEMD1 in promoting tumor proliferation in vivo. In this study, we demonstrated that the LEMD1 expression level was increased in GC tissues and cells compared with normal tissues and GES-1. The in vivo and in vitro assays showed that LEMD1 promoted GC cell proliferation by regulating the cell cycle and apoptosis. Moreover, we showed that LEMD1 regulated cell proliferation by activating the phosphatidylinositol 3 kinase (PI3K) / protein kinase B (AKT) signaling pathway. Overall, the results of our study suggest that LEMD1 contributes to GC proliferation by regulating the cell cycle and apoptosis via activation of the PI3K/AKT signaling pathway. LEMD1 may act as a potential target for GC treatment.


Assuntos
Proteínas de Neoplasias/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Animais , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Prognóstico , Neoplasias Gástricas/genética , Regulação para Cima/genética
4.
Clin Lab ; 65(1)2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30775879

RESUMO

BACKGROUND: Procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3 (PLOD3) has been proven to be involved in various human cancers; however, the function of PLOD3 in gastric cancer (GC) remains unclear. In this study, the role of PLOD3 in GC was evaluated. METHODS: The expression of PLOD3 in GC tissues and normal tissues was predicted by The Cancer Genome Atlas (TCGA). The kmplot online tool was performed to evaluate the impact of PLOD3 expression on GC patients' survival. Real-time PCR was conducted to verify PLOD3 expression in our own clinical samples and GC cells. The Cell Counting Kit-8 and the colony formation assay were used to detect GC cell proliferation ability. RESULTS: PLOD3 was upregulated in human GC tissues (compared to adjacent normal tissues, p < 0.001) and GC cells. High expression of PLOD3 was significantly correlated with larger tumor size (p = 0.007) and poor prognosis. Inhibition of PLOD3 could suppress cell proliferation in GC. CONCLUSIONS: These results revealed that PLOD3 may promote the progression of GC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Neoplasias Gástricas/genética , Regulação para Cima , Linhagem Celular Tumoral , Proliferação de Células/genética , Progressão da Doença , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Prognóstico , Interferência de RNA , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia
5.
Cell Physiol Biochem ; 47(4): 1465-1481, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29949784

RESUMO

BACKGROUND/AIMS: Gastric cancer (GC) is one of the most prevalent digestive malignancies. MicroRNAs (miRNAs) are involved in multiple cellular processes, including oncogenesis, and miR-592 itself participates in many malignancies; however, its role in GC remains unknown. In this study, we investigated the expression and molecular mechanisms of miR-592 in GC. METHODS: Quantitative real-time PCR and immunohistochemistry were performed to determine the expression of miR-592 and its putative targets in human tissues and cell lines. Proliferation, migration, and invasion were evaluated by Cell Counting Kit-8, population doubling time, colony formation, Transwell, and wound-healing assays in transfected GC cells in vitro. A dual-luciferase reporter assay was used to determine whether miR-592 could directly bind its target. A tumorigenesis assay was used to study whether miR-592 affected GC growth in vivo. Proteins involved in signaling pathways and the epithelial-mesenchymal transition (EMT) were detected with western blot. RESULTS: The ectopic expression of miR-592 promoted GC proliferation, migration, and invasion in vitro and facilitated tumorigenesis in vivo. Spry2 was a direct target of miR-592 and Spry2 overexpression partially counteracted the effects of miR-592. miR-592 induced the EMT and promoted its progression in GC via the PI3K/AKT and MAPK/ERK signaling pathways by inhibiting Spry2. CONCLUSIONS: Overexpression of miR-592 promotes GC proliferation, migration, and invasion and induces the EMT via the PI3K/AKT and MAPK/ERK signaling pathways by inhibiting Spry2, suggesting a potential therapeutic target for GC.


Assuntos
Movimento Celular , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , MicroRNAs/metabolismo , RNA Neoplásico/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Neoplásico/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
6.
Cell Death Discov ; 7(1): 156, 2021 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-34226533

RESUMO

Accumulating evidence has suggested that long noncoding RNAs (lncRNAs) exert crucial modulation roles in the biological behaviors of multiple malignancies. Nonetheless, the specific function of lncRNA LINC00284 in papillary thyroid cancer (PTC) remains not fully understood. The objective of this research was to explore the influence of LINC00284 in PTC and elucidate its potential mechanism. The Cancer Genome Atlas (TCGA), gene expression omnibus (GEO) datasets were used to analyze LINC00284 expression differences in thyroid cancer and normal samples, followed by the verification of qRT-PCR in our own PTC and adjacent non-tumor tissues. The impacts of LINC00284 on PTC cell growth were detected in vitro via CCK-8, colony formation, EdU assays, and in vivo via a xenograft tumor model. Bioinformatics analyses and biological experiments were conducted to illuminate the molecular mechanism. We found that LINC00284 expression was remarkably increased in PTC tissues and its overexpression was closely correlated with larger tumor size. In addition, silencing LINC00284 could effectively attenuate PTC cell proliferation, induce apoptosis and G1 arrest in vitro, as well as suppress tumorigenesis in mouse xenografts. Mechanistic investigations showed that LINC00284 acted as a competing endogenous RNA (ceRNA) for miR-3127-5p, thus resulting in the disinhibition of its endogenous target E2F7. In short, our findings indicated that LINC00284-miR-3127-5p-E2F7 axis exerted oncogenic properties in PTC and may offer a new promising target for the diagnosis and therapy of PTC.

7.
Front Oncol ; 11: 664242, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33987098

RESUMO

So far, many existing evidences indicate that microRNAs (miRNA) are closely associated with the tumorigenesis and progression of various tumors. It has been reported that miR-1301-3p is abnormally expressed in several malignant tumors. However, the role of miR-1301-3p in gastric cancer (GC) remains unclear and is worth studying. Through qRT-PCR, the expression of miR-1301-3p and SIRT1 were detected in GC tissues and cells. The cell proliferation and cell cycle were measured through CCK-8 assay and clone formation assay. Dual luciferase reporter assay was used to determine the target of miR-1301-3p. Though tumorigenesis assay, we monitored the effect of miR-1301-3p on GC cell growth in vivo. miR-1301-3p was upregulated in GC tissues and cells in our study. Overexpression of miR-1301-3p accelerated GC cell proliferation, cell cycle progression and tumorigenesis. Notably, altering the expression miR-1301-3p caused deregulation of Cyclin D1, CDK4, c-Myc and P21. Furthermore, SIRT1 was the direct target of miR-1301-3p by luciferase reporter assay. After transfecting with miR-1301-3p inhibitor, we found that knockdown of SIRT1 could enhance the ability of proliferation. Our results identify miR-1301-3p as a novel potential therapeutic target that is associated with the tumorigenesis and progression of gastric cancer.

8.
Cell Death Dis ; 11(7): 521, 2020 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-32647147

RESUMO

Gastric cancer (GC) has been one of the most leading cause of cancer-death worldwide. Long non-coding RNAs (lncRNAs) have been found to be related with the carcinogenesis and the development of various cancers, including GC. However, there are still many GC-related lncRNAs functional roles and molecular mechanisms that have not yet been clearly studied. Herein, we report lncRNA CCDC144NL-AS1, which has not been explored in GC, and it is markedly upregulated in GC tissues, which may serve as an independent predictor of poor prognosis. We found that CCDC144NL-AS1 expression was significantly positively associated with a larger tumor size and more pronounced lymph node metastasis. Through a series of in vivo and in vitro functional experiments, we observed that CCDC144NL-AS1 could facilitate cell proliferation, invasion and migration and inhibit cell apoptosis in GC. Further mechanism investigation revealed that CCDC144NL-AS1 acted as a competing endogenous RNA (ceRNA) for sponging miR-143-3p and upregulated the expression of its direct endogenous target MAP3K7 in GC. Taken together, our results elucidate the oncogenic roles of CCDC144NL-AS1/miR-143-3p/MAP3K7 axis in GC progression, providing inspiration for further understanding of the mechanism of GC and making CCDC144NL-AS1 as a potential novel diagnostic and therapeutic target for GC.


Assuntos
MAP Quinase Quinase Quinases/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , RNA Interferente Pequeno/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Progressão da Doença , Células HEK293 , Xenoenxertos , Humanos , MAP Quinase Quinase Quinases/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , RNA Interferente Pequeno/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Transfecção , Regulação para Cima
9.
J Exp Clin Cancer Res ; 38(1): 452, 2019 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-31694721

RESUMO

BACKGROUND: Increasing evidence shows that stimulated by retinoic acid 6 (STRA6) participates in regulating multiple cancers. However, the biological roles of STRA6 in gastric cancer (GC) remain unknown. This study aimed to investigate the biological function of STRA6 and reveal the underlying mechanism of its dysregulation in GC. METHODS: The expression level of STRA6 was detected through quantitative real-time PCR and Western blot analysis. The effects of STRA6 on the proliferation of GC cells were studied through CCK-8 proliferation, colony formation and 5-ethynyl-2'-deoxyuridine (EdU) assays. The effects of STRA6 on migration and invasion were detected via wound healing and Transwell assays. Upstream miRNAs, which might regulate STRA6 expression, was predicted through bioinformatics analysis. Their interaction was further confirmed through dual-luciferase reporter assays and rescue experiments. RESULTS: STRA6 was up-regulated in GC and enhanced the proliferation and metastasis of GC cells in vitro and in vivo. STRA6 knockdown could inhibit the Wnt/ß-catenin signalling pathway. STRA6 was confirmed as an miR-873 target, which acted as a tumour suppressor in GC. Rescue assays showed that the repressing effect of miR-873 could be partially reversed by overexpressing STRA6. CONCLUSIONS: STRA6 is down-regulated by miR-873 and plays an oncogenic role by activating Wnt/ß-catenin signalling in GC.


Assuntos
Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica , Proteínas de Membrana/genética , MicroRNAs/genética , Oncogenes , Interferência de RNA , Neoplasias Gástricas/etiologia , Regiões 3' não Traduzidas , Adulto , Idoso , Animais , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Proteínas de Membrana/metabolismo , Camundongos , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/mortalidade , Neoplasias Gástricas/patologia , Carga Tumoral , Via de Sinalização Wnt
10.
Artif Cells Nanomed Biotechnol ; 47(1): 3814-3822, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31549851

RESUMO

It was investigated that TP73-AS1(TP73 antisense RNA 1) could function as an oncogene in gastric cancer (GC). The expression and function of long noncoding RNAs (lncRNAs) could be impacted by single nucleotide polymorphisms (SNPs), which are related to cancer susceptibility and prognosis. This study was to reveal the association between lncRNAs TP73-AS1 polymorphisms (rs1181865 A > G, rs9800 G > C, rs3737589 A > G, rs2298222 G > A, rs7515164 C > A) and GC in 1000 GC cases and 1000 controls in a Chinese Han population. Rs3737589 G allele had significant associations with the increasing risk of GC (G vs. A: p = .005). Rs3737589 variant genotypes (AG + GG) were related to an increased risk of GC in the elder population (age ≥60), females, nonsmokers, nondrinkers, individuals living in urban, and individuals without family history of GC in stratified analyses. Rs3737589 variant genotypes (AG + GG) were related to the advanced depth of tumor invasion (T3 + T4). Besides, we found that GC patients with AG or GG genotype of rs3737589 had poorer overall survival (OS) than those with AA genotype (p < .05). Our findings showed that the lncRNA TP73-AS1 rs3737589 polymorphism might increase the risk of GC, and rs3737589 polymorphism could be a potential biomarker to predict the prognosis of GC patients.


Assuntos
Etnicidade/genética , Predisposição Genética para Doença/genética , Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Estudos de Casos e Controles , China/epidemiologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/patologia , Análise de Sobrevida
11.
J Cancer ; 10(23): 5705-5713, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31737107

RESUMO

Purpose: Gastric adenosquamous carcinoma (ASC) is a rare pathological type with poorly understood clinicopathological features. The purpose of this study is to identify the characteristics of gastric ASC patients. Methods: Using the Surveillance, Epidemiology, and End Results (SEER) database (2000 to 2014), patients with ASC (N=93) or adenocarcinoma (AC) (N=41794) of the stomach were included. The epidemiology, tumor features, treatment, and outcomes between these two groups were compared. Results: The incidences of ASC from 1983 to 2014 [annual percentage change (APC) = -3.5%, 95% confidence interval (CI) -4.9 to -2.1] and AC from 1973-2014 [APC = -1.8%, 95%CI -2.0 to -1.6] decreased over time. Compared to AC cases, patients with ASC were more likely to present poor differentiation (74.2% vs 52.4%) and later summary stage (distant: 46.2% vs 33.6%) or later T stage (T4: 15.1%% vs 9.0%). Besides, the proportion of patients with distant metastasis (33.3% vs 23.9%), and chemotherapy (44.1% vs 34.0%) in ASC group was higher. The Kaplan-Meier analyses showed ASC cases had worse overall survival (OS) (p=0.017) than that of AC after propensity score matching (PSM), but not the cancer-specific survival (CSS) (p=0.849). The further subgroup analyses suggested no statistical significance between gastric ASC patients and AC patients for CSS. The multivariate cox proportional hazard analyses indicated that patients with distant summary stage (HR=2.11, p=0.014), no surgery (HR=2.22, p=0.016), and no/unknown chemotherapy (HR=3.33, p<0.001) were associated with poor OS for ASC population alone. However, for CSS, only ASC cases with no/unknown chemotherapy (HR=2.22, p=0.018) indicated worse outcomes. Conclusions: Gastric ASC presented more aggressive clinicopathologic characteristics and poorer OS compared with AC. The localized/regional summary stages and undergoing surgery suggested favorable OS for gastric ASC patients. ASC cases receiving chemotherary showed both better OS and CSS.

12.
Cell Death Dis ; 10(1): 32, 2019 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-30631050

RESUMO

Gastric cancer (GC) is one of the most frequent malignancies, and increasing evidence supports the contribution of microRNA (miRNAs) to cancer progression. miR-1254 has been confirmed to participate in the regulation of various cancers, while the function of miR-1254 in GC remains unknown. In this study, we investigated the role of miR-1254 in GC. The expression of miR-1254 was detected in human GC specimens and cell lines by miRNA RT-PCR. The effects of miR-1254 on GC proliferation were determined by CCK-8 proliferation assays, colony formation assays, 5-ethynyl-2'-deoxyuridine (EdU) incorporation, and cell-cycle assays. The ability of migration and invasion was examined by transwell and wound-healing assay. Dual Luciferase reporter assay was used to validate the interaction of miR-1254 with its target gene. The xenograft mouse models were conducted to investigate the effects of miR-1254 in vivo. The signaling pathways and epithelial-mesenchymal transition (EMT)-related proteins were detected with western blot. The results showed that miR-1254 inhibited the proliferation, migration and invasion in vitro and suppressed tumorigenesis in vivo. Smurf1 was shown to be the direct target of miR-1254. Overexpressing Smurf1 could partially counteract the effects caused by miR-1254. Similarly, the effects of the miR-1254-inhibitor were also rescued by Smurf1-shRNA. Furthermore, we found that miR-1254 inhibited EMT and decreased the PI3K/AKT signaling pathway through downregulating Smurf1. In summary, overexpression of miR-1254 could suppress proliferation, migration, invasion, and EMT via PI3K/AKT signaling pathways by downregulation of Smurf1 in GC, which suggests a potential therapeutic target for GC.


Assuntos
Movimento Celular , Proliferação de Células , Regulação para Baixo , MicroRNAs , Neoplasias Gástricas , Ubiquitina-Proteína Ligases , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Regiões 3' não Traduzidas , Sítios de Ligação , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Invasividade Neoplásica , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Transfecção , Carga Tumoral/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
13.
Dis Markers ; 2017: 6807452, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28931965

RESUMO

Long noncoding RNA (lncRNA) phosphatase and tensin homolog pseudogene 1 (PTENP1) is significantly downregulated in gastric cancer (GC), playing critical roles in GC progression. However, the association between PTENP1 genetic variants and GC risk has not yet been reported. Using TaqMan technology, three lncRNA PTENP1 tag single nucleotide polymorphisms (tagSNPs) (rs7853346 C>G, rs865005 C>T, and rs10971638 G>A) were genotyped in 768 GC patients and 768 cancer-free controls in a Chinese population. We found that subjects with rs7853346 G allele had a remarkably decreased risk of GC, compared with those carrying C allele (P = 0.011 in an additive model, P = 0.033 after Bonferroni's correction). The further stratified analyses showed that the link between variant genotypes of rs7853346 and decreased GC risk was more obvious in older subjects (≥60 years), nonsmokers, nondrinkers, and subjects without family history of GC. We also found that relative PTENP1 mRNA expression levels were higher in rs7853346 CG/GG genotype carriers than those with common genotype in both GC and normal tissues (P < 0.05). Besides, bioinformatics analyses revealed that rs7853346 may change the local folding structure and alter the target microRNAs (miRNAs) of PTENP1. In conclusion, our results suggested that lncRNA PTENP1 polymorphism rs7853346 may predict GC susceptibility.


Assuntos
Polimorfismo de Nucleotídeo Único , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
14.
Oncotarget ; 8(37): 62445-62453, 2017 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-28977958

RESUMO

Low molecular mass protein (LMP) gene performs a critical role in the foreign antigen processing machine via the major histocompatibility complex-I (MHC-I) complex CD8+ cytotoxic T lymphocytes (CTL) pathway. Recent studies have reported the association of LMP2-60 G>A (rs17587) and LMP7-145 C>A (rs2071543) polymorphisms with various types of cancers, but the outcomes remained inconsistent. To obtain a reliable conclusion, we summarized available data and conducted a meta-analysis involving a total of 19 published studies. Evidences were obtained from the PubMed, Google Scholar, Web of Science and Chinese National Knowledge Infrastructure (CNKI) databases. The results demonstrated that the rs17587 and rs2071543 polymorphisms were associated with an increased cancer risk in the recessive and homozygote models. Stratified analyses by ethnicity indicated a significant association only in Asian population. Furthermore, rs17587 showed a greater susceptibility to gynecological cancers, while rs2071543 increased the risk of gastrointestinal and gynecological cancers. Our results indicate that the LMP2 rs17587 and LMP7 rs2071543 polymorphisms may act as risk factors for cancer, especially for Asian populations. Additional larger-scale multicenter studies should be performed to validate our results.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA