Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biomed Res Int ; 2023: 2942402, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37124931

RESUMEN

Background: Esophageal cancer remains one of the most lethal malignant diseases globally. Previous studies indicated that TRIM9 (Tripartite Motif Containing 9) is a potential marker in breast cancer patients. Therefore, in the current research, we intended to clarify the regulatory network of TRIM9 and its relative role in esophageal cancer patients. We aimed to elucidate the regulatory role of TRIM9 in esophageal cancer. Methods: Clinical tumor tissue samples combined with cancer cell line models were utilized to explore the TRIM9 expression pattern. Functional experiments including transwell assay, cell viability assay, and ubiquitination blocking experiments were performed to evaluate the role of the TRIM9/ZEB1 (zinc finger E-box binding homeobox 1) axis and UPP pathway in esophageal cancer progression and exacerbation. Results: Both esophageal cancer samples and cell line models showed significantly suppressed levels of TRIM9. Functional experiments confirmed that TRIM9 overexpression inhibited the cell viability, invasiveness, and stem-like phenotype of cancer cells. Subsequent investigations suggested that TRIM9-ZEB1 interaction accelerated ZEB1 protein degradation through the modulation of the UPP pathway, which confirmed the protective role of TRIM9 in esophageal cancer progression and metastasis. Conclusion: This study concluded that TRIM9 was a tumor suppressor that interacted with ZEB1 and accelerated ZEB1 protein degradation via the ubiquitin-proteasome pathway (UPP). Our research emphasized TRIM9-ZEB1 interaction as a valuable target for esophageal cancer treatment in future development. More detailed studies are needed to further consolidate our findings.


Asunto(s)
Neoplasias Esofágicas , MicroARNs , Proteínas de Motivos Tripartitos , Homeobox 1 de Unión a la E-Box con Dedos de Zinc , Humanos , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/genética , Proteolisis , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteínas de Motivos Tripartitos/metabolismo
2.
Gene ; 851: 147053, 2023 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-36395972

RESUMEN

FANCI, a member of the Fanconi anemia (FA) complementation group, normally associates with FANCD2 to play an important role in ribosome biogenesis and DNA repair. However, the correlation of FANCI with prognostic value and the molecular mechanism in patients with lung adenocarcinoma (LUAD) remains unclear. In the present study, bioinformatics analysis was performed on LUAD data from TCGA and GEO databases, and further confirmed by in vitro experiments. We found that a high level of FANCI was significantly correlated with a worse survival probability in patients with LUAD. Moreover, the results from in vitro experiments revealed high levels of FANCI in LUAD specimens and cell lines. Knockdown of FANCI expression in A549 and H460 cells significantly inhibited cell viability and clone formation of LUAD cells in vitro and in vivo. Furthermore, high FANCI levels were negatively correlated with a variety of tumor-infiltrating immune cells. Importantly, the overexpression of FANCI significantly inhibited the activation of M1 macrophages. All the data demonstrated that FANCI was a useful prognostic biomarker in patients with LUAD, and knockdown of FANCI inhibited tumor growth of LUAD cells in vitro and in vivo, partly by suppressing the activation of M1 macrophages.


Asunto(s)
Adenocarcinoma del Pulmón , Anemia de Fanconi , Neoplasias Pulmonares , Humanos , Adenocarcinoma del Pulmón/genética , Macrófagos , Neoplasias Pulmonares/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética
3.
Oncol Rep ; 48(2)2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35703356

RESUMEN

Fanconi anemia complementation group I (FANCI) is a critical protein for maintaining DNA stability. However, the exact role of FANCI in tumors remains to be elucidated. The present study aimed to explore the role and potential mechanism of action of FANCI in non­small cell lung cancer (NSCLC). To quantify the expression levels of FANCI and ubiquitin­conjugating enzyme E2T (UBE2T) in NSCLC tissues, reverse­transcription quantitative PCR and western blotting were employed. Cell Counting Kit­8, wound healing and Transwell assays along with flow cytometry analysis and tumor xenograft were used to investigate the biological effects of FANCI in NSCLC in vitro and in vivo. The binding of FANCI with UBE2T was confirmed using a co­immunoprecipitation assay. Epithelial­to­mesenchymal transition (EMT) protein markers were quantified via western blotting. The results showed that FANCI expression level was higher in NSCLC tumor tissues, compared with adjacent tissues. In A549 and H1299 cells, knockdown of FANCI inhibited cell proliferation, migration, invasion, cell cycle and EMT in vitro. Tumor growth was repressed in vitro, upon downregulation of FANCI expression. UBE2T was observed to directly bind to FANCI and regulate its monoubiquitination. Overexpression of UBE2T reversed the effects induced by FANCI knockdown in NSCLC cells. Furthermore, it was noted that FANCI interacted with WD repeat domain 48 (WDR48). Overexpression of WDR48 reversed the effects of FANCI on cell proliferation, migration and EMT. In conclusion, FANCI was identified to be a putative oncogene in NSCLC, wherein FANCI was monouniubiquitinated by UBE2T to regulate cell growth, migration and EMT through WDR48. The findings suggested that FANCI could be used as a prognostic biomarker and therapeutic target for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Anemia de Fanconi , Neoplasias Pulmonares , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo
4.
Cell Death Dis ; 13(5): 425, 2022 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-35501353

RESUMEN

The purpose of the current study was to define the role of MAX interactor 1 (Mxi1) in the pathogenesis of lung cancer and its underlying molecular mechanism. Bioinformatics analysis was performed to identify important regulatory pathway related to lung cancer. Dual luciferase reporter and ChIP assays were adopted to validate the interaction among Mxi1, miR-300 and KLF9. Loss- and gain-of-function studies were conducted to determine the roles of Mxi1, miR-300, and KLF9 in cell proliferation, migration, and invasion in vitro and their effects on myeloid-derived suppressor cell (MDSC) recruitment in vivo. Mxi1 was poorly expressed in lung cancer tissues and cells and its poor expression was associated with poor prognosis. Mxi1 inhibited miR-300 by suppressing its transcription. miR-300 suppressed the expression of KLF9, and KLF9 negatively regulated GADD34 expression in lung cancer cells. Mxi1 or KLF9 elevation or miR-300 repression inhibited lung cancer cell proliferation, as evidenced by reduced Ki67 and PCNA expression, and lowered invasion and migration. In vivo findings revealed that silencing KLF9 induced tumor growth by enhancing MDSC-mediated immunosuppression through upregulation of GADD34. Collectively, these findings suggest that Mxi1 can inhibit lung cancer progression by regulating the miR-300/KLF9 axis and GADD34-mediated immunosuppression.


Asunto(s)
Neoplasias Pulmonares , MicroARNs , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Neoplasias Pulmonares/patología , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Supresoras de Tumor/metabolismo
5.
Bioengineered ; 13(3): 4991-5004, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35156900

RESUMEN

Circular RNAs play important roles in cancer biology. In this research, we explored the underlying function and mechanism of cirMMD_007 in lung adenocarcinoma (LC). Clinical lung adenocarcinoma samples were obtained from surgery. Bioinformatic databases were used to predict miRNAs that can potentially target circRNAs and miRNA target genes. hsa_circMMD_007, miR-197-3p, and PTPN9 mRNA expressions were investigated by qRT-PCR. Protein expressions were examined using Western blot. The proliferation abilities were assessed by Cell Counting Kit-8 assays. Wound healing cell migration assay was applied to evaluate cell migration ability. Luciferase reporter assay and rescue experiments were then performed to elucidate the underlying mechanism. We found that the expression of circMMD_007 was abnormally increased in LC. The expression of circMMD_007 was higher in advanced stages. Knockout of circMMD_007 hindered the tumorigenesis of LC in vivo and in vitro. circMMD_007 could negatively regulate the expression of miR-197-3p. PTPN9 behaved to be a molecular target of miR-197-3p. In summary, this research demonstrated that circular RNA circMMD_007 could promote the oncogenic effects in the progression of LC through miR-197-3p/PTPN9 axis.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , MicroARNs , Proteínas Tirosina Fosfatasas no Receptoras , ARN Circular , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/genética , Proteínas Tirosina Fosfatasas no Receptoras/genética , ARN Circular/genética
6.
Surg Endosc ; 36(9): 6456-6463, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35024927

RESUMEN

OBJECTIVES: The blood supply of the transposed jejunum was assessed by ICG fluorescence imaging in jejunal interposition, and the correlation with anastomotic leakage or transposed jejunal necrosis was analyzed, aim to explore the value of the application ICG fluorescence imaging technology. METHODS: 84 esophageal reconstructions with jejunal interposition without supercharging were retrospectively analyzed. Intraoperatively, the blood supply of transposed jejunal was observed using ICG fluorescence endoscopy. ROC curve of T1/2 was constructed to calculate the corresponding T1/2max value of the region where the transposed jejunal want to be anastomosed with esophageal stump, the relationship between T1/2max value and anastomotic leakage or transposed jejunal necrosis was analyzed. RESULTS: The occurrence of anastomotic leakage and transposed jejunal necrosis was 9.5%, In the ROC curve, the maximum value of the Youden index was 0.691, the T1/2max value was 5.35 s. When T1/2max value > 5.35 s correspondingly, the probability of anastomotic leakage or transposition jejunal necrosis was 33.3% (7/21); when T1/2max value ≤ 5.35 s, the probability of anastomotic leakage or transposition jejunal necrosis was 1.6% (1/63). The difference between the two groups was statistically significant (P < 0.05). CONCLUSION: ICG fluorescent imaging can effectively assess the blood supply of transposed jejunum. When T1/2max > 5.35, the possibility of the incidence rate of anastomotic leakage or transposed jejunum necrosis increases, this will remind the operators to take corresponding remedial measures during operation.


Asunto(s)
Fuga Anastomótica , Verde de Indocianina , Anastomosis Quirúrgica/métodos , Fuga Anastomótica/etiología , Fuga Anastomótica/prevención & control , Humanos , Yeyuno/diagnóstico por imagen , Yeyuno/cirugía , Necrosis/etiología , Imagen Óptica , Estudios Retrospectivos
7.
J Biochem ; 170(6): 689-698, 2022 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-34117764

RESUMEN

The study aims to explore the interaction between miR-133a-3p and cell division cycle associated 8 (CDCA8) in oesophageal cancer (EC) and their effect on malignant behaviour of EC cells. Differential miRNAs and mRNAs were obtained from The Cancer Genome Atlas (TCGA) database. Quantitative real-time PCR (qRT-PCR) was used to detect the expression levels of miR-133a-3p and CDCA8 mRNA in EC cells. Western blot was used to detect the expression of CDCA8 protein. CCK-8, flow cytometry and Transwell assays were conducted to detect cell proliferation, cell cycle and apoptosis, as well as migration and invasion, respectively. The targeting relationship between miR-133a-3p and CDCA8 was verified by dual-luciferase reporter gene assay. In EC, miR-133a-3p expression was evidently low and CDCA8 expression was prominently high. MiR-133a-3p downregulated CDCA8 expression. A range of cell function experiments revealed that CDCA8 promoted the proliferation, migration and invasion of EC cells, reduced cell cycle arrest in G0/G1 phase and inhibited cell apoptosis, while miR-133a-3p could reverse the above effects by regulating CDCA8. MiR-133a-3p is a crucial tumour suppressor miRNA in EC, playing a tumour suppressor role by targeting CDCA8.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proliferación Celular , Neoplasias Esofágicas/metabolismo , MicroARNs/metabolismo , Proteínas de Neoplasias/metabolismo , ARN Neoplásico/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Neoplasias Esofágicas/genética , Humanos , MicroARNs/genética , Proteínas de Neoplasias/genética , ARN Neoplásico/genética
8.
Immunol Invest ; 51(1): 199-219, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32954867

RESUMEN

BACKGROUND: MicroRNA (miR)-146a might participate in the occurrence of malignant tumor. The aim of the current investigation was to evaluate the relationship of microRNA-146a (miR-146a) rs2910164 C > G locus to the development of digestive system cancer (DSC). METHODS: We retrieved publications from PubMed, China Biology Medicine and EMBASE databases up to August 29, 2019. Finally, 56 independent case-control studies with 59,098 participants were included. The strength of the relationship between rs2910164 locus and a risk of DSC was assessed. The power value was also calculated in this study. RESULTS: We identified a correlation of rs2910164 locus in miR-146a with DSC development in dominant model (P = .035; power value = 0.994). MiR-146a rs2910164 locus was also identified to be correlated with a risk of DSC in Asians (GG/CG vs. CC: P = .033; power value = 0.989). Sensitivity analysis revealed that any individual study could not alter the final decision. In our study, no significant bias was found among these included studies (P > .1). The results of heterogeneity analysis suggested that small sample size (<1000 subjects), colorectal carcinoma, Asians, gastric carcinoma, esophageal squamous cell carcinoma, hepatocellular cancer, hospital-based study and high-quality score (≥7.0) subgroups contributed the heterogeneity to our findings. Galbraith radial plot determined that eleven outliers contributed to the main heterogeneity. CONCLUSION: In summary, this meta-analysis highlights that rs2910164 locus might be implicated in the risk of DSC. More studies are, therefore, needed to confirm our results.


Asunto(s)
Neoplasias del Sistema Digestivo , MicroARNs , Estudios de Casos y Controles , Neoplasias del Sistema Digestivo/genética , Predisposición Genética a la Enfermedad , Humanos , MicroARNs/genética , Polimorfismo de Nucleótido Simple
9.
Cancer Cell Int ; 20: 228, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32536816

RESUMEN

BACKGROUND: Lung adenocarcinoma has surpassed lung squamous cell carcinoma as the most common type of non-small cell lung cancer. In this study, we had tested the biological role of TRIM2 in lung adenocarcinoma. METHODS: TRIM2 abundance in clinical tissues and six cell lines were examined with quantitative real-time PCR test (qRT-PCR) and western blot. TRIM2 overexpression treated H322 cells and TRIM2 knockdown treated A549 cells were used to study cell proliferation, migration, colony formation, invasion, and the expression of epithelial mesenchymal transformation (EMT) biomarkers. Moreover, ubiquitination related Snail1 degradation were studied with qRT-PCR and western blot. The relationships between TRIM2 and Snail1 were investigated with western blot, co-immunoprecipitation, migration, and invasion. RESULTS: TRIM2 was highly expressed in lung adenocarcinoma tissues. TRIM2 overexpression and knockdown treatments could affect cell proliferation, colony formation, migration, invasion, and the expression of EMT associated biomarkers. Moreover, TRIM2 can regulate the ubiquitination related Snail1 degradation. In addition, TRIM2 can regulate Snail1 degradation in lung adenocarcinoma via ubiquitination pathway. TRIM2 could promote the proliferation, migration, and invasion of lung adenocarcinoma. Meanwhile, TRIM2 can deubiquitinate and stabilize Snail1 protein, which play important role in the function of lung adenocarcinoma. CONCLUSION: A high TRIM2 expression could be detected in lung adenocarcinoma tissues and cells. TRIM2 could aggravate cell proliferation, invasion, and migration in colorectal cancer by regulating Snail1 ubiquitylation degradation. Our results could provide detailed information for further studies in lung adenocarcinoma.

10.
IUBMB Life ; 70(10): 1032-1039, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30194893

RESUMEN

Pancreatic cancer (PC) is one of the most malign human cancers, with its underlying molecular mechanisms largely unknown. In this work, we investigated the mechanistic role of protein arginine methyltransferase 1 (PRMT1) gene in PC. Expression of PRMT1 in immortal PC cell lines and clinical human PC tumors was evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. In PANC-1 and SW1990 cells, PRMT1 was either downregulated by lentiviral-mediated short hairpin RNA (shRNA) or upregulated by overexpression plasmid. The effects of PRMT1 downregulation or upregulation on PC proliferation and invasion in vitro, and xenograft in vivo, were evaluated. Gene expression of PRMT1 downstream target, zinc finger E-box binding homeobox 1 (ZEB1) was measured in PRMT1-downregulated PC cells. ZEB1 was also upregulated in PRMT1-downregulated PC cells to evaluate its functional role in PRMT1-mediated regulation in PC. PRMT1 was downregulated in both PC cell lines and human tumors. PRMT1 downregulation in PANC-1 and SW1990 cells significantly suppressed cancer proliferation and invasion in vitro and xenograft in vivo. However, PRMT1 overexpression did not have function impact in PC cells. ZEB1 gene expression was suppressed in PRMT1-downregulated PC cells. Subsequently, overexpressing ZEB1 reversed the antitumor effects of PRMT1 downregulation in PC cells. PRMT1 was aberrantly upregulated in PC. PRMT1 inhibition, possibly inversely acting through ZEB1, might be an effective molecular intervention to inhibit PC growth and invasion. © 2018 IUBMB Life, 70(10):1032-1039, 2018.


Asunto(s)
Proliferación Celular/genética , Neoplasias Pancreáticas/genética , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Invasividad Neoplásica/genética , Neoplasias Pancreáticas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...