Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Mol Cancer ; 17(1): 118, 2018 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-30098595

RESUMEN

BACKGROUND: Accumulating evidences show that long noncoding RNAs (lncRNA) play essential roles in the development and progression of various malignancies. However, their functions remains poorly understood and many lncRNAs have not been defined in colorectal cancer (CRC). In this study, we investigated the role of DLEU1 in CRC. METHODS: Quantitative real-time PCR was used to detect the expression of DLEU1 and survival analysis was adopted to explore the association between DLEU1 expression and the prognosis of CRC patients. CRC cells were stably transfected with lentivirus approach and cell proliferation, migration, invasion and cell apoptosis, as well as tumorigenesis in nude mice were performed to assess the effects of DLEU1 in BCa. Biotin-coupled probe pull down assay, RNA immunoprecipitation and Fluorescence in situ hybridization assays were conducted to confirm the relationship between DLEU1 and SMARCA1. RESULTS: Here we revealed that DLEU1 was crucial for activation of KPNA3 by recruiting SMARCA1, an essential subunit of the NURF chromatin remodeling complex, in CRC. DLEU1 was indispensible for the deposition of SMARCA1 at the promoter of KPNA3 gene. Increased expression of DLEU1 and KPNA3 was observed in human CRC tissues. And higher expression of DLEU1 or KPNA3 in patients indicates lower survival rate and poorer prognosis. DLEU1 knockdown remarkably inhibited CRC cell proliferation, migration and invasion in vitro and in vivo while overexpressing KPNA3 in the meantime reversed it. CONCLUSIONS: Our results identify DLEU1 as a key regulator by a novel DLEU1/SMARCA1/KPNA3 axis in CRC development and progression, which may provide a potential biomarker and therapeutic target for the management of CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , alfa Carioferinas/genética , Animales , Células CACO-2 , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HT29 , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Pronóstico , Regiones Promotoras Genéticas , ARN Largo no Codificante , Activación Transcripcional , Regulación hacia Arriba
2.
Eur J Med Res ; 29(1): 219, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38576045

RESUMEN

PURPOSE: Colorectal cancer (CRC) is a highly heterogeneous malignancy with an unfavorable prognosis. The purpose of this study was to address the heterogeneity of CRC by categorizing it into ion channel subtypes, and to develop a predictive modeling based on ion channel genes to predict the survival and immunological states of patients with CRC. The model will provide guidance for personalized immunotherapy and drug treatment. METHODS: A consistent clustering method was used to classify 619 CRC samples based on the expression of 279 ion channel genes. Such a method was allowed to investigate the relationship between molecular subtypes, prognosis, and immune infiltration. Furthermore, a predictive modeling was constructed for ion channels to evaluate the ion channel properties of individual tumors using the least absolute shrinkage and selection operator. The expression patterns of the characteristic genes were validated through molecular biology experiments. The effect of potassium channel tetramerization domain containing 9 (KCTD9) on CRC was verified by cellular functional experiments. RESULTS: Four distinct ion channel subtypes were identified in CRC, each characterized by unique prognosis and immune infiltration patterns. Notably, Ion Cluster3 exhibited high levels of immune infiltration and a favorable prognosis, while Ion Cluster4 showed relatively lower levels of immune infiltration and a poorer prognosis. The ion channel score could predict overall survival, with lower scores correlated with longer survival. This score served as an independent prognostic factor and presented an excellent predictive efficacy in the nomogram. In addition, the score was closely related to immune infiltration, immunotherapy response, and chemotherapy sensitivity. Experimental evidence further confirmed that low expression of KCTD9 in tumor tissues was associated with an unfavorable prognosis in patients with CRC. The cellular functional experiments demonstrated that KCTD9 inhibited the proliferation, migration and invasion capabilities of LOVO cells. CONCLUSIONS: Ion channel subtyping and scoring can effectively predict the prognosis and evaluate the immune microenvironment, immunotherapy response, and drug sensitivity in patients with CRC.


Asunto(s)
Neoplasias Colorrectales , Canales Iónicos , Humanos , Canales Iónicos/genética , Nomogramas , Inmunoterapia , Neoplasias Colorrectales/genética , Pronóstico , Microambiente Tumoral
3.
Front Immunol ; 13: 988303, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275690

RESUMEN

Background: Colon adenocarcinoma (COAD) is a prevalent malignancy that causes significant mortality. Microsatellite instability plays a pivotal function in COAD development and immunotherapy resistance. However, the detailed underlying mechanism requires further investigation. Consequently, identifying molecular biomarkers with prognostic significance and revealing the role of MSI in COAD is important for addressing key obstacles in the available treatments. Methods: CIBERSORT and ESTIMATE analyses were performed to evaluate immune infiltration in COAD samples, followed by correlation analysis for MSI and immune infiltration. Then, differentially expressed genes (DEGs) in MSI and microsatellite stability (MSS) samples were identified and subjected to weighted gene co-expression network analysis (WGCNA). A prognostic model was established with univariate cox regression and LASSO analyses, then evaluated with Kaplan-Meier analysis. The correlation between the prognostic model and immune checkpoint inhibitor (ICI) response was also analyzed. Results: In total, 701 significant DEGs related to MSI status were identified, and WGCNA revealed two modules associated with the immune score. Then, a seven-gene prognostic model was constructed using LASSO and univariate cox regression analyses to predict survival and ICI response. The high-risk score patients in TCGA and GEO cohorts presented a poor prognosis, as well as a high immune checkpoint expression, so they are more likely to benefit from ICI treatment. Conclusion: The seven-gene prognostic model constructed could predict the survival of COAD and ICI response and serve as a reference for immunotherapy decisions.


Asunto(s)
Adenocarcinoma , Neoplasias del Colon , Humanos , Pronóstico , Inestabilidad de Microsatélites , Neoplasias del Colon/patología , Adenocarcinoma/patología , Inhibidores de Puntos de Control Inmunológico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo
4.
Int Immunopharmacol ; 91: 107275, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33360085

RESUMEN

Periodontitis is a common chronic disease. Osteoclast differentiation contributes to alveolar bone resorption which is a distinct phenomenon during periodontitis. Syndecan 4 (SDC4), a member of the syndecan family, was found to be highly expressed during periodontitis. However, little is known about its role in periodontitis. Herein, we explored the role of SDC4 in osteoclast differentiation. An experimental periodontitis rat model was established by ligating the right first molar. The SDC4 expression in periodontium was detected by western blot and immunofluorescence. Our study demonstrated that SDC4 was highly expressed in the periodontium of periodontitis rats. It was positively transcriptionally regulated by NF-κB. SDC4 silencing abrogated osteoclast differentiation induced by RANKL, while SDC4 overexpression enhanced osteoclast differentiation. Moreover, SDC4 enhanced autophagy induced by RANKL. 3-MA, an autophagy inhibitor, was employed to explore whether SDC4 impacts osteoclast differentiation through activating autophagy. Treatment with 3-MA abolished osteoclast differentiation which was enhanced by SDC4, indicating that SDC4 promotes osteoclast differentiation through activating autophagy. This study reveals that SDC4 may contribute to osteoclast differentiation during periodontitis through activating autophagy. It sheds light on the important role of SDC4 in periodontitis.


Asunto(s)
Autofagia/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Periodontitis/metabolismo , Periodoncio/metabolismo , Ligando RANK/farmacología , Sindecano-4/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , FN-kappa B/metabolismo , Osteoclastos/metabolismo , Osteoclastos/patología , Periodontitis/genética , Periodontitis/patología , Periodoncio/patología , Células RAW 264.7 , Ratas Sprague-Dawley , Transducción de Señal , Sindecano-4/genética , Transcripción Genética
5.
J Exp Clin Cancer Res ; 37(1): 222, 2018 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-30201010

RESUMEN

BACKGROUND: Emerging evidence has shown long noncoding RNAs (lncRNAs) exert important roles in colorectal cancer (CRC) tumorigenesis. However, most lncRNAs involved in this process remain undefined and the underlying molecular mechanisms mediated by lncRNAs are largely unknown. METHODS: An unbiased screening was used to identify novel lncRNAs involved in CRC according to an online-available data dataset. In situ hybridization (ISH) and qRT-PCR was used to detect lncRNA expression patterns. CCK8, colony formation, fluorescence activated cell sorter (FACS), transwell, xenograft nude mouse model and western blot assays were used to analyze the functions of SLCO4A1-AS1. RNA-pulldown, western blot, RNA fluorescence in situ hybridization (RNA-FISH) and electrophoretic mobility shift assay (EMSA) assays were utilized to explore the molecular mechanism of SLCO4A1-AS1. RESULTS: LncRNA SLCO4A1-AS1 was significantly upregulated in CRC tissues and its overexpression was closely related with poor prognosis and tumor metastasis. By knocking down SLCO4A1-AS1, we found that SLCO4A1-AS1 promoted the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of CRC cells in vitro, as well as inhibited cell apoptosis. Moreover, SLCO4A1-AS1 dramatically delayed tumor propagation in vivo. Mechanistically, SLCO4A1-AS1 activates Wnt/ß-catenin signaling. SLCO4A1-AS1 enhanced the stability of ß-catenin by impairing the interaction of ß-catenin with GSKß and inhibiting its phosphorylation. Finally, restoration of ß-catenin protein level rescued the proliferation, migration and invasion in SLCO4A1-AS1-depleted CRC cells. CONCLUSION: SLCO4A1-AS1 serves as an oncogenic role in CRC through activating Wnt/ß-catenin signaling pathway. And SLCO4A1-AS1 might be a useful biomarker for CRC diagnosis and prognosis.


Asunto(s)
Carcinogénesis/genética , Proliferación Celular/genética , Neoplasias Colorrectales/genética , ARN Largo no Codificante/genética , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Ratones , Transportadores de Anión Orgánico/genética , Vía de Señalización Wnt/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA