Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Front Oncol ; 13: 1099857, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36910598

RESUMO

Background: Patients with tumors generally present with accompanying activation of the coagulation system, which may be related to tumor stage. To our knowledge, few studies have examined the activation of the coagulation system in reference to lymph node metastasis within gastric cancer. This study aimed to investigate the correlation between multiple coagulation-related factors and lymph node metastasis in patients with gastric cancer after excluding the influence of tumor T stage. Materials and methods: We retrospectively evaluated the relationship between lymph node metastasis and coagulation-related factors in 516 patients with T4a stage gastric cancer. We further analyzed influencing factors for lymph node metastasis and verified the predictive value of maximum amplitude (MA, a parameter of thromboelastography which is widely used to assess the strength of platelet-fibrinogen interaction in forming clots) in reference to lymph node metastasis. Results: Platelet counts (P=0.011), fibrinogen levels (P=0.002) and MA values (P=0.006) were statistically significantly higher in patients with T4a stage gastric cancer presenting with lymph node metastasis than in those without lymph node metastasis. Moreover, tumor N stage was statistically significantly and positively correlated with platelet count (P<0.001), fibrinogen level (P=0.003), MA value (P<0.001), and D-dimer level (P=0.010). The MA value was an independent factor for lymph node metastasis (ß=0.098, 95% CI: 1.020-1.193, P=0.014) and tumor N stage (ß=0.059, 95% CI: 0.015-0.104, P=0.009), and could be used to predict the presence of lymph node metastasis in patients with gastric cancer (sensitivity 0.477, specificity 0.783, P=0.006). The independent influencing factors for MA value mainly included platelet levels, fibrinogen levels, D-dimer and hemoglobin levels; we found no statistically significant correlations with tumor diameter, tumor area, and other evaluated factors. Conclusion: We conclude that MA value is an independent influencing factor for lymph node metastasis and tumor N stage in patients with T4a stage gastric cancer. The MA value has important value in predicting the presence or absence of lymph node metastasis in patients with gastric cancer. Clinical trial registration: http://www.chictr.org.cn, identifier ChiCTR2200064936.

2.
BMC Cancer ; 23(1): 258, 2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-36941595

RESUMO

BACKGROUND: The lack of obvious symptoms of early gastric cancer (GC) as well as the absence of sensitive and specific biomarkers results in poor clinical outcomes. Tubulin is currently emerging as important regulators of the microtubule cytoskeleton and thus have a strong potential to be implicated in a number of disorders, however, its mechanism of action in gastric cancer is still unclear. Tubulin alpha-1 C (TUBA1C) is a subtype of α-tubulin, high TUBA1C expression has been shown to be closely related to a poor prognosis in various cancers, this study, for the first time, revealed the mechanism of TUBA1C promotes malignant progression of gastric cancer in vitro and in vivo. METHODS: The expression of lncRNA EGFR-AS1 was detected in human GC cell lines by qRT-PCR. Mass spectrometry experiments following RNA pulldown assays found that EGFR-AS1 directly binds to TUBA1C, the CCK8, EdU, transwell, wound-healing, cell cycle assays and animal experiments were conducted to investigate the function of TUBA1C in GC. Combined with bioinformatics analyses, reveal interaction between Ki-67, E2F1, PCNA and TUBA1C by western blot. Rescue experiments furtherly demonstrated the relationship of EGFR-AS1and TUBA1C. RESULTS: TUBA1C was proved to be a direct target of EGFR-AS1, and TUBA1C promotes gastric cancer proliferation, migration and invasion by accelerating the progression of the cell cycle from the G1 phase to the S phase and activating the expression of oncogenes: Ki-67, E2F1 and PCNA. CONCLUSION: TUBA1C is a new potential target of LncRNA EGFR-AS1 promotes gastric cancer progression and could be a novel biomarker and therapeutic target for GC.


Assuntos
RNA Longo não Codificante , Neoplasias Gástricas , Tubulina (Proteína) , Animais , Humanos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Regulação Neoplásica da Expressão Gênica , Antígeno Ki-67/metabolismo , Processos Neoplásicos , Prognóstico , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/patologia , Tubulina (Proteína)/metabolismo
3.
J Cancer Res Clin Oncol ; 149(9): 6329-6339, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36745222

RESUMO

PURPOSE: Radical gastrectomy with D2 lymphadenectomy can trigger a high incidence of postoperative pancreatic fistula (POPF), which produces a poor clinical prognosis. We sought to evaluate the effect of somatostatin analogs (SSA) on POPF and clinical prognosis after radical gastrectomy. METHODS: A total of 123 patients with a high risk of POPF after radical gastrectomy (drainage fluid amylase concentration on a postoperative day [POD] 1 > 3 times the upper limit of normal serum amylase value) were randomly divided into the SSA group (n = 61) and the control group (n = 62). The former received continuous intravenous SSA (0.3 mg/8 h) for 3 days from POD1, and the latter normal saline. The primary outcome was the incidence of POPF. RESULTS: The incidence of POPFs in the SSA group was significantly lower than that in the control group (3.3% vs. 14.5%, P = 0.029). The incidence of short-term postoperative complications was significantly lower in the SSA group than in the control group (9.8% vs. 24.2%, P = 0.034). The median white blood cell counts, neutrophil counts, and the percentage of neutrophils on POD4 were significantly lower in the SSA group than in the control group (all P < 0.05). The SSA group had a shorter mean time to the first liquid diet (87.33 ± 17.92 h vs. 93.97 ± 17.29 h, P = 0.039). And the SSA group had less median daily drainage volume (96.33 mL vs. 119.67 mL, P = 0.025) and shorter drainage duration (7.0 days vs. 10.0 days, P = 0.013). CONCLUSION: Postoperative treatment with a somatostatin analog reduced the incidence of POPF and short-term complications after radical gastrectomy. (TRN: ChiCTR2200056201, Reg. Date: 2022/2/1).


Assuntos
Fístula Pancreática , Somatostatina , Humanos , Fístula Pancreática/etiologia , Fístula Pancreática/epidemiologia , Fístula Pancreática/cirurgia , Somatostatina/uso terapêutico , Projetos Piloto , Fatores de Risco , Gastrectomia/efeitos adversos , Complicações Pós-Operatórias/etiologia , Amilases
4.
World J Gastroenterol ; 28(16): 1656-1670, 2022 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-35581960

RESUMO

BACKGROUND: Gastric cancer (GC) is considered as one of the most widespread malignancies. Emerging evidence has shown that lncRNAs can function as important oncogenes or tumor suppressors during GC progression. AIM: To investigate the effect and mechanism of lncRNA cancer susceptibility 20 (CASC20) in the proliferation and metastasis of GC cells. METHODS: Data mining and clinical samples were used to evaluate the expression of CASC20 in GC and adjacent tissues. CASC20 was down-regulated in GC cells by short-interfering RNA. Cell proliferation was evaluated by CCK-8 assay, and cell migration and invasion were detected by wound healing and Transwell assays. The expressions of proteins related to epithelial-mesenchymal transition were detected by western blot assay. RESULTS: The expression of CASC20 was increased in GC tumor tissues and various GC cell lines. High CASC20 expression was correlated with a high risk of lymphatic metastasis and poor prognosis in GC patients. In vitro assays showed that silencing CASC20 reduced cell proliferation, migration, and invasion in GC cells. Mechanistic studies revealed that CASC20 exhibits oncogenic functions by regulating MEMO1 expression through competitive endogenous binding to miR-143-5p, leading to induction of epithelial-mesenchymal transition. CONCLUSION: Our findings indicate that CASC20 serves as a tumor promoter by regulating metastasis in GC via the miR-143-5p/MEMO1 axis. CASC20 may be a potential therapeutic target for GC.


Assuntos
MicroRNAs , RNA Longo não Codificante , Neoplasias Gástricas , Proteínas Supressoras de Tumor/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , Neoplasias Gástricas/patologia
6.
Cell Death Dis ; 13(2): 187, 2022 02 25.
Artigo em Inglês | MEDLINE | ID: mdl-35217636

RESUMO

Integrative expression Quantitative Trait Loci (eQTL) analysis found that rs8180040 was significantly associated with Coiled-coil domain containing 12 (CCDC12) in colon adenocarcinoma (COAD) patients. Immunohistochemical staining and western blotting confirmed CCDC12 was highly expressed in COAD tissues, which was consistent with RNA-Seq data from the TCGA database. Knockdown of CCDC12 could significantly reduce proliferation, migration, invasion, and tumorigenicity of colon cancer cells, while exogenous overexpression of CCDC12 had the opposite effect. Four plex Isobaric Tags for Relative and Absolute Quantitation assays were performed to determine its function and potential regulatory mechanism and demonstrated that overexpression of CCDC12 would change proteins on the adherens junction pathway. Overexpressed Snail and knocked down CCDC12 subsequently in SW480 cells, and we found that overexpression of Snail did not significantly change CCDC12 levels in SW480 cells, while knockdown of CCDC12 reduced that of Snail. CCDC12 plays a significant role in tumorigenesis, development, and invasion of COAD and may affect the epithelial to mesenchymal transformation process of colon cancer cells by regulating the Snail pathway.


Assuntos
Neoplasias do Colo , Doença Pulmonar Obstrutiva Crônica , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias do Colo/metabolismo , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Humanos
7.
J Cancer Res Clin Oncol ; 147(9): 2709-2719, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-33606093

RESUMO

PURPOSE: Protective loop ileostomy is an effective diversion measure often used to reduce the risk of anastomotic leakage. The purpose of the present study was to evaluate the surgical outcomes of the one-stitch method (OM) of protective loop ileostomy in laparoscopic low anterior resection for BMI obesity patients with rectal cancer compared with the traditional method (TM). METHODS: The patients diagnosed as rectal adenocarcinoma cases by preoperative pathology were included in this retrospective study. The subjects underwent protective loop ileostomy in laparoscopic low anterior resection from January 2016 to June 2019 in the Shandong Provincial Hospital affiliated to Shandong University. The data of loop ileostomy and stoma closure operation were retrieved from the medical cases system of the hospital. RESULTS: 242 patients were included in the present study. In the BMI obese cohort, the OM group showed a shorter operative time both in the loop ileostomy (232.5 vs. 250.0 min, p = 0.04) and stoma closure operation (102.5 vs. 115.0 min, p = 0.001) and a lower peristomal adhesion extent (p = 0.02) and a shorter median postoperative stay (6 vs. 7 days, p = 0.03) during stoma closure operation than that of the TM group. In the TM group, obese cases showed a higher operative time of stoma closure operation (115.0 vs. 95.0, p < 0.001), a higher parastomal hernia rate (p = 0.04), a higher peristomal adhesion extent (p = 0.005) and a longer postoperative stay of stoma closure operation (p = 0.02) compared with the non-obese cases, while in the OM group, no significant differences were observed between the obese and non-obese cases in terms of the above-mentioned factors. CONCLUSIONS: The OM exhibited more advantages than TM, notably in BMI obesity patients.


Assuntos
Adenocarcinoma/cirurgia , Índice de Massa Corporal , Ileostomia/métodos , Laparoscopia/métodos , Tempo de Internação/estatística & dados numéricos , Obesidade/fisiopatologia , Neoplasias Retais/cirurgia , Adenocarcinoma/patologia , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Neoplasias Retais/patologia , Estudos Retrospectivos
8.
Cancer Cell Int ; 20: 497, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33061849

RESUMO

BACKGROUND: Long noncoding RNAs (LncRNAs) have been reported to critically regulate gastric cancer (GC). Recently, it was reported that LBX2 antisense RNA 1 (LBX2-AS1) is abnormally expressed in GC. However, the role of LBX2-AS1 in the malignancy of GC is worth further discussion. METHODS: Quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the LBX2-AS1, miR-4766-5p and C-X-C motif chemokine (CXCL5) expression in GC tissues and cells. Dual-luciferase reporter assay was applied to examine the target relationship between LBX2-AS1 and miR-4766-5p or miR-4766-5p and CXCL5. Cell counting kit-8 (CCK-8) and Transwell assays were used to detect cell proliferation, migration and invasion rates. The protein expression of CXCL5 was confirmed using western blot. The RNA pull down experiment was used to verify the specificity of LBX2-AS1 and miR-4766-5p on BGC-823 and SGC-7901 cells. RESULTS: LBX2-AS1 was up-regulated in GC tissues and cells, and its knockdown suppressed proliferation, migration and invasion of GC cells. While, overexpression of LBX2-AS1 increased proliferation and increased CXCL5 mRNA level. CXCL5 improved cell proliferation, migration and invasion of GC cells. LBX2-AS1 could bind to miR-4766-5p to regulate CXCL5 expression. Overexpression of CXCL5 overturned those effects of miR-4766-5p in GC cells. RNA Pull down shown that BGC-823 and SGC-7901 cells, miR-4766-5p specifically binds to LBX2-AS1. CONCLUSIONS: In short, this study demonstrated that LBX2-AS1 promoted proliferation, migration and invasion through up-regulation CXCL5 mediated by miR-4766-5p in GC. The LBX2-AS1/miR-4766-5p/CXCL5 regulatory axis provides a theoretical basis for the research on lncRNA-directed therapeutics in GC.

9.
J Cancer ; 11(23): 6841-6849, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33123275

RESUMO

Genome-wide association studies of colorectal cancer (CRC) have identified two risk SNPs. The characterization of these risk regions in diverse racial groups with different linkage disequilibrium structure would aid in localizing the causal variants. Herein, fine mapping of the established CRC loci was carried out in 1,508 cases and 1,482 controls obtained from the Han Chinese population. One distinct association signal was identified at these loci, where fine mapping implicated rs1010208 as a functional locus. Next, the candidate target genes of functional SNP rs1010208 were analyzed using data from TCGA databases by expression quantitative trait loci analysis method; the data from Peking University People's Hospital were utilized for verification. The dual-luciferase reporter system analysis confirmed that rs1010208 is a regulatory region that can be mutated to decrease the expression of HINT1, resulting in proliferation and invasiveness of CRC.

10.
Biosci Rep ; 40(8)2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32785606

RESUMO

OBJECTIVE: Recent studies have shown that distal-less homeobox 6 antisense 1 (DLX6-AS1) is aberrantly expressed in various cancers and is associated with poor prognosis. This meta-analysis is designed to investigate the effects of DLX6-AS1 expression on clinicopathological features and survival outcomes. METHODS: All eligible studies were searched from Pubmed, Web of Science, Embase, the Cochrane Library, and Wanfang database, up to August 2019. The literature was selected according to the inclusion and exclusion criteria listed in this work, and the quality of each eligible study was assessed. Each patient's clinicopathological features and survival data were analyzed using Stata12.0 software. Begg's test and sensitivity analysis were also conducted. RESULTS: A total of 12 articles were included, covering 841 patients. Results showed that high expression of DLX6-AS1 was significantly closely associated with poor overall survival in tumor patients (hazard ratio (HR) = 2.30, confidence interval (95% CI): 1.70-3.09, P<0.01). This meta-analysis also showed that overexpression of DLX6-AS1 was significantly associated with tumor stage (P<0.01), tumor size (P<0.01), lymph node metastasis (P<0.01), and distant metastasis (P<0.01). Begg's test suggested no publication bias. CONCLUSION: This meta-analysis revealed that high expression of DLX6-AS1 was related to the advanced clinicopathological characteristics of human digestive system cancers (gastric cancer, esophageal cancer, colon cancer, pancreatic cancer, and hepatocellular carcinoma) and other cancers such as ovarian cancer, osteosarcoma and non-small cell lung cancer, and DLX6-AS1 has important predictive value for poor prognosis. However, more studies are needed to further corroborate these findings.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias/genética , RNA Longo não Codificante/genética , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Neoplasias/diagnóstico , Neoplasias/mortalidade , Neoplasias/terapia , Fenótipo , Prognóstico , Medição de Risco , Fatores de Risco
11.
Biol Res ; 52(1): 60, 2019 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-31847887

RESUMO

BACKGROUND: Recent studies have confirmed that RASAL1 has an antitumor effect in many cancers, but its functional role and the molecular mechanism underlying in colon cancer has not been investigated. RESULTS: We collected human colon cancer tissues and adjacent non-tumor tissues, human colon cancer cell lines LoVo, CaCo2, SW1116, SW480 and HCT-116, and normal colonic mucosa cell line NCM460. RT-qPCR was used to detect the RASAL1 level in the clinical tissues and cell lines. In LoVo and HCT-116, RASAL1 was artificially overexpressed. Cell viability and proliferation were measured using CCK-8 assays, and cell cycle was detected via PI staining and flow cytometry analysis. RASAL1 significantly inhibited the cell proliferation via inducing cell cycle arrest, suppressed cell cycle associated protein expression, and decreased the lipid content and inhibited the SCD1 expression. Moreover, SCD1 overexpression induced and downregulation repressed cell proliferation by causing cell cycle arrest. Additionally, luciferase reporter assays were performed to confirm the direct binding between SREBP1c, LXRα and SCD1 promoter, we also demonstrated that RASAL1 inhibit SCD1 3'-UTR activity. RASAL1 inhibited tumor growth in xenograft nude mice models and shows inhibitory effect of SCD1 expression in vivo. CONCLUSION: Taken together, we concluded that RASAL1 inhibited colon cancer cell proliferation via modulating SCD1 activity through LXRα/SREBP1c pathway.


Assuntos
Proliferação de Células/fisiologia , Neoplasias do Colo/patologia , Proteínas Ativadoras de GTPase/metabolismo , Receptores X do Fígado/metabolismo , Estearoil-CoA Dessaturase/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Proteínas Ativadoras de GTPase/genética , Humanos , Receptores X do Fígado/genética , Camundongos , Estearoil-CoA Dessaturase/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
12.
Int J Clin Exp Pathol ; 12(9): 3171-3178, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31934161

RESUMO

Pulmonary fibrosis is a serious interstitial disease characterized by initial diffuse alveolar inflammation, fibroblast proliferation, ECM accumulation, and the destruction of normal pulmonary tissues, whose etiology remains unknown and therapeutic options remain limited. The prevalence of Vitamin D deficiency is increasing and has been linked to pulmonary fibrosis. In recent years, many studies focused on the mechanistic pathway of Vitamin D in the prevention of fibrosis. This review highlights the current evidence on the molecular mechanisms of Vitamin D in pulmonary fibrosis. We want to provide new clues to the clinical management of pulmonary fibrosis.

13.
J Laparoendosc Adv Surg Tech A ; 29(3): 316-322, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30088979

RESUMO

INTRODUCTION: The optimal approach of resection for mid-low rectal cancer after neoadjuvant chemoradiotherapy (nCRT) is still controversial. The aim of this meta-analysis was to clarify the safety and feasibility of laparoscopic surgery compared with open resection. MATERIALS AND METHODS: We performed a literature search for studies on PubMed, Embase and Cochrane Library up to March 1, 2018. Review Manager software was applied for data analysis. We used weighted mean difference (WMD) for continuous parameters and odds ratio (OR) for dichotomous variables. Confidence interval (CI) was set at 95% and a P value <.05 was considered statistically significant. RESULTS: A total of seven studies met the inclusion criteria for the meta-analysis: 466 patients in laparoscopic group and 491 in open group. The pooled result revealed that laparoscopic resection had a favorable blood loss (WMD = -116.88 mL; 95% CI: -189.78 to -43.99; P = .002), analogous lymph nodes harvest (WMD = -0.30; 95% CI: -1.29 to 0.70; P = .56), less postoperative complications (OR = 0.63; 95% CI: 0.46-0.88; P = .006), shorter time to pass first flatus (WMD = -0.76 day; 95% CI: -1.00 to -0.51; P < .00001), and stay in hospital (WMD = -2.71 days; 95% CI: -4.54 to -0.88; P = .004), despite similar operating time (WMD = 11.17 minutes; 95% CI: -14.37 to 36.70; P = .39). CONCLUSIONS: Laparoscopic resection might be a technically safe and feasible approach for mid-low rectal cancer patients after nCRT compared with open resection.


Assuntos
Laparoscopia , Neoplasias Retais/terapia , Quimiorradioterapia Adjuvante , Trato Gastrointestinal/fisiologia , Humanos , Laparoscopia/efeitos adversos , Tempo de Internação , Excisão de Linfonodo , Terapia Neoadjuvante , Duração da Cirurgia , Complicações Pós-Operatórias/etiologia , Recuperação de Função Fisiológica , Resultado do Tratamento
14.
Biol. Res ; 52: 60, 2019. graf
Artigo em Inglês | LILACS | ID: biblio-1100912

RESUMO

BACKGROUND: Recent studies have confirmed that RASAL1 has an antitumor effect in many cancers, but its functional role and the molecular mechanism underlying in colon cancer has not been investigated. RESULTS: We collected human colon cancer tissues and adjacent non-tumor tissues, human colon cancer cell lines LoVo, CaCo2, SW1116, SW480 and HCT-116, and normal colonic mucosa cell line NCM460. RT-qPCR was used to detect the RASAL1 level in the clinical tissues and cell lines. In LoVo and HCT-116, RASAL1 was artificially overexpressed. Cell viability and proliferation were measured using CCK-8 assays, and cell cycle was detected via PI staining and flow cytometry analysis. RASAL1 significantly inhibited the cell proliferation via inducing cell cycle arrest, suppressed cell cycle associated protein expression, and decreased the lipid content and inhibited the SCD1 expression. Moreover, SCD1 overexpression induced and downregulation repressed cell proliferation by causing cell cycle arrest. Additionally, luciferase reporter assays were performed to confirm the direct binding between SREBP1c, LXRα; and SCD1 promoter, we also demonstrated that RASAL1 inhibit SCD1 3'-UTR activity. RASAL1 inhibited tumor growth in xenograft nude mice models and shows inhibitory effect of SCD1 expression in vivo. CONCLUSION: Taken together, we concluded that RASAL1 inhibited colon cancer cell proliferation via modulating SCD1 activity through LXRα/SREBP1c pathway.


Assuntos
Humanos , Animais , Camundongos , Estearoil-CoA Dessaturase/metabolismo , Neoplasias do Colo/patologia , Proteínas Ativadoras de GTPase/metabolismo , Proliferação de Células/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Receptores X do Fígado/metabolismo , Estearoil-CoA Dessaturase/genética , Regulação para Baixo , Proteínas Ativadoras de GTPase/genética , Linhagem Celular Tumoral , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Receptores X do Fígado/genética
15.
Anticancer Drugs ; 29(10): 975-982, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30096129

RESUMO

Our study desired to investigate how miR-34c-3p regulates colon cancer cell proliferation and what is the relationship between miR-34c-3p and EIF3D. HCEpiC (normal human colonic epithelial cells), SW620, HT-29, SW480, and HCT-116 (human colon cancer cells lines) were used in our study. SW620 cells were chosen and divided into blank, miR-34c-3p mimics, miR-34c-3p NC, miR-34c-3p inhibitors, Lv-EIF3D, Lv-NC, and miR-34c-3p mimics+Lv-EIF3D groups. qRT-PCR was used for the detection of miR-34c-3p and EIF3D mRNA expressions. Dual-luciferase reporter assay was performed to investigate the effect of miR-34c-3p on EIF3D. Western blot was performed to detect EIF3D, cyclin D1, and c-Myc expressions. Clone formation and MTT assay were used to measure cell proliferation ability. colon cancer cells had lower miR-34c-3p expression, but higher EIF3D expression compared with HCEpiC. EIF3D mRNA expression was regulated negatively by miR-34c-3p. In the miR-34c-3p mimics group, colon cancer cell proliferation was significantly decreased, whereas c-Myc and cyclin D1 expressions were downregulated. Colon cancer cell proliferation in miR-34c-3p inhibitors and Lv-EIF3D groups was enhanced, and c-Myc and cyclin D1 expressions were decreased. The results suggested that by targeting EIF3D, miR-34c-3p inhibited colon cancer cell proliferation.


Assuntos
Proliferação de Células/genética , Neoplasias do Colo/patologia , Fator de Iniciação 3 em Eucariotos/genética , MicroRNAs/genética , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Ciclina D1/genética , Regulação para Baixo/genética , Células HCT116 , Células HT29 , Humanos , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima/genética
16.
Cell Physiol Biochem ; 49(1): 322-334, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30138934

RESUMO

BACKGROUND/AIMS: LncRNA EGFR-AS1 is an antisense transcript of EGFR, which plays a key role in gastric cancer progression. This study was aimed to explore the effects of lncRNA EGFR-AS1 on GC and the underling mechanisms. METHODS: The silencing of EGFR-AS1 expression was performed by using EGFR-AS1 shRNA lentivirus in MGC803 and SGC-7901 GC cell. The levels of lncRNA EGFR-AS1 and EGFR were detected by qPCR and western blot. Cell proliferation was assessed by CCK-8, EdU, and colony formation assays. The EGFR mRNA stability was explored by using RNA synthesis inhibitor α-amanitin. RESULTS: In our study, EGFR-AS1 significantly up-regulated in GC tissues and correlated with tumor size. And the expression of EGFR-AS1 positively correlated with EGFR in tissues. Moreover, knock-down of EGFR-AS1 inhibited the proliferation of GC cells via suppressing EGFR-dependent PI3K/AKT pathway in vitro and in vivo. Mechanismly, depletion of EGFR-AS1 was found to decrease EGFR expression by reduction of EGFR mRNA stability. CONCLUSION: Our findings suggested that EGFR-AS1 might have an oncogenic effect on GC and serve as a potential target of GC.


Assuntos
Receptores ErbB/metabolismo , RNA Longo não Codificante/metabolismo , RNA Mensageiro/metabolismo , Neoplasias Gástricas/patologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Receptores ErbB/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , Estabilidade de RNA , RNA Longo não Codificante/antagonistas & inibidores , RNA Longo não Codificante/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo
17.
Int J Clin Exp Pathol ; 11(6): 3157-3164, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-31938445

RESUMO

This study sought to verify the reactivation effect of dsP53-285 that can up-regulate P53 expression in vitro. In addition, we explored the reactivation effect that dsP53-285 has on the biological behavior of gastric cancer cells. The specific small activating RNA (saRNA), dsP53-285, targeting the P53 gene promoter was synthesized. Also, a double strained control RNA (dsControl) was synthesized as a negative control, and then siP53 was synthesized to exclude the off-target effect. Both BGC-823 and MGC-803 cells were transfected with the corresponding microRNA, or just treated with lipofectamine2000. RT-qPCR and Western blot were adopted to detect P53 mRNA or the protein content of each group. CCK-8 was adopted to detect the proliferation of each group. The migration ability was assessed using the scratch-wound assay. The results of RT-qPCR and Western blot showed that dsP53-285 caused a significant up-regulation of the P53 gene (P<0.01), and the expression level of the P21 gene changed with the reactivation. The CCK-8 showed that, compared to the control group, the proliferation ability of the dsP53-285 group was inhibited significantly (P<0.01). The reactivation effect was in a time-course manner. The wound scratch assay proved that, compared to the control group, the migration ability of dsP53-285 group was inhibited significantly (P<0.01). This phenomenon provides a theoretical basis for the carcinostatic activity of small activating RNA (saRNA) and might indicate a new targeted treatment option for gastric cancer.

18.
Oncol Lett ; 14(6): 6901-6906, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29163709

RESUMO

Mel-P15 is a peptide derived from melittin, the main toxic component in the venom of the European honeybee Apis mellifera. In the present study, the antitumor effects of Mel-P15 and the underlying molecular mechanisms of these effects in vivo were investigated. Mel-P15 directly stimulated natural killer (NK) cell cytotoxicity in vitro, which was increased to 55.45% at a 4 µg/ml dose of Mel-P15. In the mouse liver cancer (H22) xenograft mice model, Mel-P15 suppressed tumor growth in vivo; the tumor inhibitory rate was 61.15% following treatment with 2 mg/kg Mel-P15. In addition, the immune response was activated following Mel-P15 treatment. Mel-P15 treatment increased the spleen and thymus indices, promoted splenocyte proliferation, stimulated NK cytotoxicity and upregulated the secretion of cytokines, including interleukin-2, interferon-γ and tumor necrosis factor-α. In addition, the tumor inhibitory effect of Mel-P15 on BEL-7402-bearing nude mice was abrogated by the selective depletion of NK cells via the intraperitoneal injection of an anti-asialo GM-1 antibody. The results suggest that Mel-P15 inhibits tumor growth in vivo by promoting NK cell cytotoxicity. Mel-P15 may therefore be a potential immunotherapy candidate for the treatment of hepatocellular carcinoma.

19.
Biochem Biophys Res Commun ; 482(4): 542-548, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27856244

RESUMO

OBJECTIVE: To identify an effective saRNA sequence that can specifically up-regulate VEZT expression and to determine the influence of saRNA had on gastric cancer cell growth, proliferation, invasion and migration. METHODS: Three various saRNAs, that target the VEZT gene promoter at different locations relative to the transcription start site were synthesized. A dsControl saRNA was synthesized as a negative control, and a specific shRNA was synthesized to knockdown VEZT and eliminate any off-target effects of the saRNA. Both SGC-7901 and M-28 cells were either transfected with the different saRNAs, or treated with Lipofectamine2000 alone. To determine the most effective saRNA, real-time PCR and Western blot were used to determine the VEZT mRNA and protein content, respectively, of each treatment group. After selection, both cell lines were treated with the chosen saRNA, dsControl or Lipofectamine2000. The saRNA treated cells were divided into two groups: the first group was used immediately in the experiments, and the second group was transfected with shRNA by using RNAi-Mate. The proliferation of cells transfected with saRNA, or saRNA and shRNA, as well as the other control cells, was detected by CCK-8. The invasive and migratory abilities were determined using the transwell chamber assay. RESULTS: We identified the most effective saRNA via real-time PCR and Western blot. The selected saRNA inhibited the growth, invasion and migration of GC cells by specially reactivating VEZT. The real-time PCR and Western blot results showed that treatment with saRNA caused a significant up-regulation of VEZT, and an obvious decrease in the proliferative, invasive and migratory abilities; compared with the control groups (P < 0.01); furthermore, there were no significant differences among the control groups (P > 0.05). This phenomenon provides a theoretical basis for saRNA design and gene therapy for gastric cancer.


Assuntos
Proteínas de Transporte/genética , Proteínas de Membrana/genética , RNA/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Estômago/patologia , Regulação para Cima , Sequência de Bases , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Terapia Genética/métodos , Humanos , Invasividade Neoplásica/patologia , RNA/administração & dosagem , RNA Interferente Pequeno , Estômago/citologia , Neoplasias Gástricas/terapia
20.
Onco Targets Ther ; 9: 815-22, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26929650

RESUMO

PURPOSE: Liver metastasis is one of the leading causes of death in colorectal cancer (CRC) patients. The present study aimed to evaluate the value of eIF4E as a prognostic marker of colorectal liver metastasis (CLM) and identify the functional role of eIF4E in CRC metastasis. PATIENTS AND METHODS: The expression level of eIF4E in CRC tissues was analyzed by immunohistochemical staining and Western blot. Expression of eIF4E in CRC cell lines was evaluated by reverse transcription quantitative polymerase chain reaction (RT-qPCR) and Western blot. Cell Counting Kit-8 (CCK-8) and Transwell assays were performed to assess the effects of eIF4E on cell proliferation, migration, and invasion. Western blot was further used to investigate the mechanism of eIF4E in tumor metastasis. RESULTS: The upregulation frequency of eIF4E in the CLM group (82.5%) was higher than that in the non-CLM group (65.0%). Of the 80 patients recruited for the follow-up study, 23 were in the low eIF4E group (ratio of tumor to nontumor tissue

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...