Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 610
Filtrar
1.
Curr Eye Res ; 46(2): 232-238, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32757684

RESUMEN

Purpose: This work aimed to investigate the influences of microRNA-340 (miR-340) on proliferation and apoptosis of retinoblastoma (RB) cells and explore its regulatory mechanism. MATERIALS AND METHODS: miR-340 mimic and inhibitor were applied for up-regulating or inhibiting the expression of miR-340 in RB cell lines. Then, CCK-8 and AnnexinV-FITC/PI staining were used to measure cell proliferation and apoptosis, respectively. After that, luciferase assay was performed to affirm the direct targets of miR-340. Furthermore, qRT-PCR and western blotting assay were carried out to detect the levels of miR-340 and KIF14. RESULTS: Our results indicated that the miR-340 was lowly expressed in RB cell lines, and up-regulation of miR-340 can decrease the proliferation and induce the apoptosis of RB cells. Moreover, we verified that miR-340 controls KIF14 expression, either directly or through a subsequent molecular cascade, and inversely related to its expression. The results obtained from the rescue assays presented that over-expression of KIF14 reversed the miR-340-mediated inhibition on malignant phenotype of RB cells. CONCLUSIONS: Overall, we proved that miR-340 can decrease the proliferation and increase the apoptosis of RB cells, and its function in RB cells was at least partially achieved via down-regulation of KIF14, prompting that miR-340 was expected to supply a new direction for clinical therapy of RB in the future.


Asunto(s)
Apoptosis/genética , Regulación Neoplásica de la Expresión Génica , Cinesinas/genética , MicroARNs/genética , Proteínas Oncogénicas/genética , Neoplasias de la Retina/genética , Retinoblastoma/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Cinesinas/biosíntesis , MicroARNs/biosíntesis , Proteínas Oncogénicas/biosíntesis , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Neoplasias de la Retina/metabolismo , Neoplasias de la Retina/patología , Retinoblastoma/metabolismo , Retinoblastoma/patología
2.
Sci Signal ; 13(661)2020 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-33293463

RESUMEN

Small, genetically determined differences in transcription [expression quantitative trait loci (eQTLs)] are implicated in complex diseases through unknown molecular mechanisms. Here, we showed that a small, persistent increase in the abundance of the innate pathogen sensor NOD1 precipitated large changes in the transcriptional state of monocytes. A ~1.2- to 1.3-fold increase in NOD1 protein abundance resulting from loss of regulation by the microRNA cluster miR-15b/16 lowered the threshold for ligand-induced activation of the transcription factor NF-κB and the MAPK p38. An additional sustained increase in NOD1 abundance to 1.5-fold over basal amounts bypassed this low ligand concentration requirement, resulting in robust ligand-independent induction of proinflammatory genes and oncogenes. These findings reveal that tight regulation of NOD1 abundance prevents this sensor from exceeding a physiological switching checkpoint that promotes persistent inflammation and oncogene expression. Furthermore, our data provide insight into how a quantitatively small change in protein abundance can produce marked changes in cell state that can serve as the initiator of disease.


Asunto(s)
Regulación de la Expresión Génica , Monocitos/metabolismo , Proteína Adaptadora de Señalización NOD1/biosíntesis , Proteínas Oncogénicas/biosíntesis , Transducción de Señal , Transcripción Genética , Humanos , Inflamación/metabolismo , Células THP-1
3.
PLoS One ; 15(11): e0242780, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33232371

RESUMEN

As the genomic profile across cancers varies from person to person, patient prognosis and treatment may differ based on the mutational signature of each tumour. Thus, it is critical to understand genomic drivers of cancer and identify potential mutational commonalities across tumors originating at diverse anatomical sites. Large-scale cancer genomics initiatives, such as TCGA, ICGC and GENIE have enabled the analysis of thousands of tumour genomes. Our goal was to identify new cancer-causing mutations that may be common across tumour sites using mutational and gene expression profiles. Genomic and transcriptomic data from breast, ovarian, and prostate cancers were aggregated and analysed using differential gene expression methods to identify the effect of specific mutations on the expression of multiple genes. Mutated genes associated with the most differentially expressed genes were considered to be novel candidates for driver mutations, and were validated through literature mining, pathway analysis and clinical data investigation. Our driver selection method successfully identified 116 probable novel cancer-causing genes, with 4 discovered in patients having no alterations in any known driver genes: MXRA5, OBSCN, RYR1, and TG. The candidate genes previously not officially classified as cancer-causing showed enrichment in cancer pathways and in cancer diseases. They also matched expectations pertaining to properties of cancer genes, for instance, showing larger gene and protein lengths, and having mutation patterns suggesting oncogenic or tumor suppressor properties. Our approach allows for the identification of novel putative driver genes that are common across cancer sites using an unbiased approach without any a priori knowledge on pathways or gene interactions and is therefore an agnostic approach to the identification of putative common driver genes acting at multiple cancer sites.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Regulación Neoplásica de la Expresión Génica , Mutación , Proteínas Oncogénicas , Lesiones Precancerosas , Transcriptoma , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo
4.
Biomed Res Int ; 2020: 1648419, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32420320

RESUMEN

Abnormal expression of neuroepithelial cell transforming gene 1 (NET1) has been authenticated in many human cancers, including lung cancer. We have previously reported that NET1 functioned as an oncogene and promoted human non-small-cell lung cancer (NSCLC) growth and migration. However, the correlation between NET1 and its upstream miRNAs needed further illustration. Our present work demonstrated that miR-22 had a relatively low expression, and NET1 had a relatively high expression in both NSCLC samples and lung adenocarcinoma cell lines compared with corresponding normal controls. Moreover, miR-22 directly regulated NET1 and was verified to weaken cancer cell proliferation and migration, as well as enhance cell apoptosis by suppressing NET1. Furthermore, the inhibitory effect of miR-22 can be reversed via overexpressing NET1 using an ectopic expression vector in NSCLC cells. Our findings showed that miR-22/NET-1 axis may contribute to the inhibition of NSCLC growth and migration and represents a promising therapeutic target for NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/metabolismo , MicroARNs/biosíntesis , Proteínas Oncogénicas/biosíntesis , ARN Neoplásico/biosíntesis , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Células HEK293 , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , MicroARNs/genética , Proteínas Oncogénicas/genética , ARN Neoplásico/genética
5.
Int Ophthalmol ; 40(2): 343-350, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31571090

RESUMEN

PURPOSE: To investigate the overexpression of genes in sebaceous gland carcinoma (SGC) of the eyelid compared to sebaceous adenoma of the eyelid in order to elucidate the molecular mechanism underlying pathogenesis. METHODS: We performed histopathological examination of eyelid tissues surgically removed from four patients diagnosed with SGC (cases 1-3) and sebaceous adenoma (case 4) of the eyelid. Next, we performed global gene expression analysis of surgical tissue samples using a GeneChip® system and the Ingenuity Pathways Knowledge Base. The results of the GeneChip® analysis were explored with quantitative real-time polymerase chain reaction (qRT-PCR) analysis. RESULTS: In the SGC samples, we found that 211, 199, and 199 genes, respectively, showed ≥ 2.0-fold higher expression than those in the sebaceous adenoma sample (case 4); 194 genes were common to all three SGC samples. For the 194 genes with upregulated expression, functional category analysis showed that SGC of the eyelid employed a unique gene network, including cyclin-dependent kinase inhibitor 2A (CDKN2A), cyclin-dependent kinase 1 (CDK1), and cyclin E1 (CCNE1), which are related to cell cycle progression, incidence of tumor, and cell viability. Furthermore, qRT-PCR analysis showed that the expression levels of CDKN2A, CDK1, and CCNE1 were significantly upregulated in all SGC cases compared to those in the sebaceous adenoma case. These data were similar to the results of microarray analysis. CONCLUSION: Overexpression of cell cycle-related genes CDKN2A, CDK1, CCNE1, and their gene network may help elucidate the pathogenic pathway of SGC of the eyelid at the molecular level.


Asunto(s)
Adenocarcinoma Sebáceo/genética , Proteína Quinasa CDC2/genética , Ciclina E/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias de los Párpados/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Oncogénicas/genética , Neoplasias de las Glándulas Sebáceas/genética , Adenocarcinoma Sebáceo/metabolismo , Adenocarcinoma Sebáceo/patología , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Proteína Quinasa CDC2/biosíntesis , Ciclina E/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Neoplasias de los Párpados/metabolismo , Neoplasias de los Párpados/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Proteínas Oncogénicas/biosíntesis , ARN Neoplásico/genética , Neoplasias de las Glándulas Sebáceas/metabolismo , Neoplasias de las Glándulas Sebáceas/patología , Glándulas Sebáceas/metabolismo , Glándulas Sebáceas/patología
6.
Appl Immunohistochem Mol Morphol ; 28(1): 62-66, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31809312

RESUMEN

Most prostate cancers (PC) overexpress the ERG oncogene and karyopherin α 2 (KPNA2). These genes play a role in prostatic carcinogenesis, but their prognostic significance is still debated. The aim of this study was to determine the prognostic significance of ERG and KPNA2 expression, and their association to early prostate-specific antigen (PSA) biochemical recurrence in advanced PC with lymph node metastases. A series of 65 consecutive pN1 M0 R0 PC samples obtained by radical prostatectomy with lymphadenectomy has been analyzed for ERG and KPNA2 expression by immunohistochemistry. For each case, the following clinical data were collected: age, preoperative serum PSA levels, Gleason grade group, TNM stage, and follow-up. PC recurrence was investigated by serum PSA assay and defined by a PSA concentration >0.2 ng/mL after a nadir of <0.1 ng/mL following radical prostatectomy. ERG-positive staining was found in 25/65 cases (38%), and KPNA2 in 56/65 cases (86%); neither was detected in normal prostatic tissue. Immunohistochemical concordance was found between primary tumor and lymph node metastases in 24/25 (96%) of ERG and 53/56 (95%) of KPNA2-positive cases. The follow-up was known in all cases, and early PSA recurrence occurred in 25/65 cases (38%). ERG positivity, both alone and in conjunction with KPNA2 positivity, was strongly associated with early PSA recurrence [both ERG+ and KPNA+, odds ratio: 22.2 (95% confidence interval, 6.0-82.3); ERG+ alone odds ratio: 17.9 (95% confidence interval, 5.1-63.5); P<0.0001 for both]. KPNA2 expression was significantly associated with the tumor stage (P<0.00001). The results suggest that the ERG+ phenotype might be selected in metastasis-initiating clones. ERG and KPNA2 may have a prognostic value, and their positivity in PC might warrant more aggressive treatments.


Asunto(s)
Recurrencia Local de Neoplasia/metabolismo , Proteínas Oncogénicas/biosíntesis , Neoplasias de la Próstata/metabolismo , alfa Carioferinas/biosíntesis , Anciano , Humanos , Calicreínas/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/patología , Regulador Transcripcional ERG/biosíntesis
7.
J Exp Clin Cancer Res ; 38(1): 376, 2019 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-31455378

RESUMEN

BACKGROUND: Metformin has been reported to function as the anti-tumor inhibiting the growth of different types of cancers, including bladder cancer. But there are few reports on the roles of Yap1, the key molecule of Hippo pathway, in the metformin induced inhibition of bladder cancer (BLCA). We are wondering if the inhibitory effect of metformin on bladder cancer is fulfilled via Yap1 and exploring the related mechanism. METHODS: MTS and colony formation assays were used to explore the cellular viabilities and proliferation of BLCA cells challenged by metformin at different concentrations, in vitro. Flow Cytometry (FCM) was used to analyze the cell cycle and the cellular apoptosis of the BLCA cells. Western Blot was performed to detect the expressions of AMPKα, Yap1, CCND1, CCNE1/2 and CDK2/4/6 in the metformin-treated BLCA cell lines. RNAi method was used for the related genetic functional analysis. The relationships among Yap1, TEADs and CCNE1/2 were predicted and evaluated using bioinformatics, dual-luciferase reporter and co-immunoprecipitation (Co-IP) assays. For in vivo experiments, a xenograft model was used to investigate the effects of metformin on the proliferation of BLCA cells. And Immunohistochemistry (IHC) assay was performed to assess the expressions of CCNE1/2 and Yap1 proteins in the tumor tissues from the model. RESULTS: Metformin could inhibit the proliferation of the BLCA cells via inducing the G1 cell cycle arrest without apoptosis. And metformin upregulated the phosphorylated AMPKα and decreased the expressions of Yap1 and CCND1, CCNE1/2 and CDK4/6. AMPK inhibition by compound C (CC) restored the cell proliferation and the G1 cell cycle arrest induced by metformin, in vivo. Knockdown of YAP1 inhibited the proliferation of BLCA cells and caused the cell cycle arrest at G1 phase by decreasing the expressions of CCNE1/2 and other G1 phase related molecules, which has been restored by the Yap 5SA mutant. Bioinformatics analysis showed that trans-factor TEAD4 was highly expressed and positively associated with the expressions of CCNE1 and CCNE2 in BLCA and only TEAD4 was precipitated by Yap1 in the BLCA cells. Further studies demonstrated that Yap1 positively regulated both CCNE1 and CCNE2 expressions via forming complex with TEAD4. Furthermore, we observed that metformin inhibited the cell proliferation by decreasing the expressions of Yap1 and both CCNE1 and CCNE2 in xenograft model. CONCLUSIONS: The results of our study reveal a new potential regulatory pathway in which metformin inhibits cell proliferation via AMPKα/Yap1/TEAD4/CCNE1/2 axis in BLCA cells, providing new insights into novel molecular therapeutic targets for BLCA.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Ciclina E/antagonistas & inhibidores , Ciclinas/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Metformina/farmacología , Proteínas Musculares/metabolismo , Proteínas Oncogénicas/antagonistas & inhibidores , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina E/biosíntesis , Ciclina E/genética , Ciclina E/metabolismo , Ciclinas/biosíntesis , Ciclinas/genética , Ciclinas/metabolismo , Proteínas de Unión al ADN/genética , Femenino , Fase G1/efectos de los fármacos , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Musculares/genética , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Factores de Transcripción de Dominio TEA , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transfección , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Proteínas Señalizadoras YAP
8.
Eur Rev Med Pharmacol Sci ; 23(15): 6445-6452, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31378883

RESUMEN

OBJECTIVE: This work aimed to analyze the relative expression level of long intergenic non-protein coding ribonucleic acid 1503 (LINC01503) in cholangiocarcinoma tissues and cells, and to explore the effects of LINC01503 on cell proliferation, migration and invasion. PATIENTS AND METHODS: Logarithmic growth phase cholangiocarcinoma cells were selected and transfected with Lipofectamine 2000 (si-LINC01503, si-NC). The expression of LINC01503 was detected by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR). The proliferation of cells was observed by 3-(4, 5-dimethyl-2-thiazolyl)-2, 5-diphenyl-2-H-tetrazolium bromide (MTT) assay. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay was used to detect cell apoptosis. Transwell assay was used to observe the cell migration and invasion ability. RESULTS: The expression of LINC01503 was significantly increased in cholangiocarcinoma tissues compared with adjacent tissues (p<0.05), and the up-regulated expression of LINC01503 was associated with lymph node metastasis. Compared with normal bile duct cells (HIBEC), cholangiocarcinoma cells (RBE, QBC939) have higher expression of LINC01503, and si-LINC01503 transfection can effectively reduce the proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) of cholangiocarcinoma cells. CONCLUSIONS: LINC01503 is highly expressed in cholangiocarcinoma and can effectively promote the proliferation, migration, invasion and EMT process of cancer cells, and LINC01503 is expected to be a potential biomarker for cholangiocarcinoma.


Asunto(s)
Proliferación Celular/fisiología , Colangiocarcinoma/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Proteínas Oncogénicas/biosíntesis , ARN Largo no Codificante/biosíntesis , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas Oncogénicas/genética , ARN Largo no Codificante/genética
9.
Sci Adv ; 5(7): eaax2887, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31281901

RESUMEN

Using biochemical characterization of fusion proteins associated with endometrial stromal sarcoma, we identified JAZF1 as a new subunit of the NuA4 acetyltransferase complex and CXORF67 as a subunit of the Polycomb Repressive Complex 2 (PRC2). Since CXORF67's interaction with PRC2 leads to decreased PRC2-dependent H3K27me2/3 deposition, we propose a new name for this gene: CATACOMB (catalytic antagonist of Polycomb; official gene name: EZHIP ). We map CATACOMB's inhibitory function to a short highly conserved region and identify a single methionine residue essential for diminution of H3K27me2/3 levels. Remarkably, the amino acid sequence surrounding this critical methionine resembles the oncogenic histone H3 Lys27-to-methionine (H3K27M) mutation found in high-grade pediatric gliomas. As CATACOMB expression is regulated through DNA methylation/demethylation, we propose CATACOMB as the potential interlocutor between DNA methylation and PRC2 activity. We raise the possibility that similar regulatory mechanisms could exist for other methyltransferase complexes such as Trithorax/COMPASS.


Asunto(s)
Glioma/metabolismo , Histonas/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Oncogénicas/biosíntesis , Complejo Represivo Polycomb 2/metabolismo , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Metilación de ADN , ADN de Neoplasias , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/patología , Células HCT116 , Histonas/genética , Humanos , Metilación , Proteínas de Neoplasias/genética , Proteínas Oncogénicas/genética , Complejo Represivo Polycomb 2/genética
10.
Biomed Res Int ; 2019: 2582416, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31183364

RESUMEN

INTRODUCTION: Glioblastoma (GBM) is the most frequent and malignant type of primary brain tumors in adults. The valuable prognostic biomarkers and therapeutic targets for GBM remain to be elucidated. The association of adipokines with cancer has been well documented. The C1q/TNF-related protein 1 (CTRP1), a novel adipokine, belongs to the CTRP family. METHODS: In the present study, the expression and potential roles of CTRP1 in GBM were explored based on in silico evaluation, including GEPIA, the Pathology Atlas of the Human Protein Atlas, cBioPortal, TIMER, and SurvExpress. The CCK8, transwell, and wound healing assays were used to detect cell proliferation and migration. RESULTS: It was found that mRNA expression levels of CTRP1 were significantly upregulated in GBM tissues compared with those in nontumor tissues according to the analysis on public dataset and immunohistochemical results of GBM tissues (P<0.05). CTRP1 was mainly localized in the cytoplasm and cell membrane of GBM cells. The genetic alterations of CTRP1 occurred at a low rate in GBM (2 of 591 sequenced cases/patients, 0.33%). The mRNA expression levels of CTRP1 were positively associated with the tumor-infiltrating macrophages and CCL2 in GBM (P<0.05, respectively). The higher mRNA expression levels of CTRP1 were significantly correlated with higher risk and shorter overall survival time in GBM (P<0.05). CTRP1 knockdown significantly inhibited the proliferation and migration in human GBM cells, suggesting the inhibition of CTRP1 on human GMB progression. Moreover, CTRP1 knockdown inhibited CCL2 expression, and CCL2 overexpression reversed the inhibition of cell proliferation and migration induced by CTRP1 knockdown, suggesting that CTRP1 promoted tumor progression by regulating CCL2 expression. CONCLUSIONS: These findings suggest that CTRP1 potentially indicates poor prognosis in GBM and promotes the progression of human GBM.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias Encefálicas , Regulación Neoplásica de la Expresión Génica , Glioblastoma , Proteínas Oncogénicas/biosíntesis , Proteínas/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Femenino , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Masculino , Tasa de Supervivencia
11.
Biomed Res Int ; 2019: 9085435, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31205947

RESUMEN

Interferon-induced transmembrane protein 1 (IFITM1), a 17-kDa membrane protein, is generally known as a modulator in many cellular functions. Recent studies showed overexpression of IFITM1 in cancers and relationship between IFITM1 overexpression and tumor progression. However, the role of IFITM1 in lung cancer remains unclear. Here, we presented the overexpression of IFITM1 in lung cancer tissues and cell lines A549 and H460 using quantitative Real-Time RT-PCR. In vitro assay indicated IFITM1 silencing inhibited lung cancer cell proliferation, migration, and invasion. Further, in vivo assay showed that IFITM1 silencing markedly suppressed cell growth and metastasis of lung cancer in tumor-bearing BALB/c nude mice. Mechanistically, we found that IFITM1 silencing significantly alleviated the protein levels of ß-catenin, cyclin D1, and c-Mycin lung cancer cells and tumor samples. Taken together, our study revealed the role of IFITM1 as a tumor promoter during lung cancer development and the possible molecular mechanism.


Asunto(s)
Antígenos de Diferenciación/biosíntesis , Movimiento Celular , Proliferación Celular , Neoplasias Pulmonares/metabolismo , Proteínas Oncogénicas/biosíntesis , Células A549 , Animales , Antígenos de Diferenciación/genética , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Trasplante de Neoplasias , Proteínas Oncogénicas/genética
12.
Int J Cancer ; 145(5): 1299-1311, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31093963

RESUMEN

Molecular subtypes of breast cancer are defined on the basis of gene expression and genomic/epigenetic pattern differences. Different subtypes are thought to originate from distinct cell lineages, but the early activation of an oncogene could also play a role. It is difficult to discriminate the respective inputs of oncogene activation or cell type of origin. In this work, we wished to determine whether activation of distinct oncogenic pathways in human mammary epithelial cells (HMEC) could lead to different patterns of genetic and epigenetic changes. To this aim, we transduced shp53 immortalized HMECs in parallel with the CCNE1, WNT1 and RASv12 oncogenes which activate distinct oncogenic pathways and characterized them at sequential stages of transformation for changes in their genetic and epigenetic profiles. We show that initial activation of CCNE1, WNT1 and RASv12, in shp53 HMECs results in different and reproducible changes in mRNA and micro-RNA expression, copy number alterations (CNA) and DNA methylation profiles. Noticeably, HMECs transformed by RAS bore very specific profiles of CNAs and DNA methylation, clearly distinct from those shown by CCNE1 and WNT1 transformed HMECs. Genes impacted by CNAs and CpG methylation in the RAS and the CCNE1/WNT1 clusters showed clear differences, illustrating the activation of distinct pathways. Our data show that early activation of distinct oncogenic pathways leads to active adaptive events resulting in specific sets of CNAs and DNA methylation changes. We, thus, propose that activation of different oncogenes could have a role in reshaping the genetic landscape of breast cancer subtypes.


Asunto(s)
Neoplasias de la Mama/genética , Glándulas Mamarias Humanas/fisiología , Oncogenes , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Ciclina E/biosíntesis , Ciclina E/genética , Metilación de ADN , Epigénesis Genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/fisiología , Femenino , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Genoma Humano , Xenoinjertos , Humanos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Desnudos , Ratones SCID , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína Wnt1/biosíntesis , Proteína Wnt1/genética
13.
J Ovarian Res ; 12(1): 41, 2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31077237

RESUMEN

BACKGROUND AND OBJECTIVE: Ovarian cancer is a common gynaecological cancer with a poor prognosis that poses a serious threat to human life and health. It is essential to explore the possible prognostic biomarkers of ovarian cancer. As an important tumour suppressor gene, BCL7A actively participates in the growth of tumours. We aimed to study the prognostic role of BCL7A in ovarian cancer. RESULTS: Through data mining of RNAseq data from the Cancer Genome Atlas database (TCGA), we explored the clinical relevance of BCL7A mRNA expression. As a result, we found that BCL7A is expressed at low levels in ovarian cancer tissues and is correlated with survival status. Survival analysis showed that, compared with those who had higher levels of BCL7A expression, patients with ovarian cancer and low levels of BCL7A generally had shorter overall/relapse-free survival times. Cox regression models showed that low BCL7A expression could be used as an independent prognostication factor for ovarian cancer patients. CONCLUSIONS: Low BCL7A expression is an independent risk factor for poor prognosis in ovarian cancer patients.


Asunto(s)
Proteínas de Microfilamentos/biosíntesis , Proteínas Oncogénicas/biosíntesis , Neoplasias Ováricas/genética , Femenino , Humanos , Proteínas de Microfilamentos/genética , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Oncogénicas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/mortalidad , Neoplasias Ováricas/patología , Pronóstico , ARN Mensajero/metabolismo , Factores de Riesgo , Análisis de Supervivencia
14.
J Clin Oncol ; 37(14): 1169-1178, 2019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-30807234

RESUMEN

PURPOSE: A large-panel gene expression analysis was conducted to identify biomarkers associated with the effectiveness of adding palbociclib to fulvestrant. METHODS: The PALOMA-3 ( ClinicalTrials.gov identifier: NCT01942135) trial randomly assigned 521 endocrine-pretreated patients with metastatic breast cancer to receive palbociclib plus fulvestrant or placebo plus fulvestrant. Primary analysis was first conducted on 10 genes on the basis of pathway biology and evidence from previous studies followed by a systematic panel-wide search among 2,534 cancer-related genes. The association of gene expression with the effect of palbociclib on progression-free survival (PFS) was evaluated using Cox proportional hazards regression analysis, with gene expression as a continuous variable or dichotomized by median. An independent breast cancer cohort from the Preoperative Palbociclib (POP) Clinical Trial ( ClinicalTrials.gov identifier: NCT02008734) was used for validation, in 61 patients with primary breast cancer treated with 2 weeks of palbociclib. RESULTS: In the PALOMA-3 trial, 302 patients had tumor tissue analyzed (palbociclib arm, 194 patients; placebo arm, 108 patients). Palbociclib efficacy was lower in patients with high versus low cyclin E1 (CCNE1) mRNA expression (median PFS: palbociclib arm, 7.6 v 14.1 months; placebo arm, 4.0 v 4.8 months, respectively; interaction P unadjusted = .00238; false discovery rate-adjusted P = .0238). CCNE1 mRNA was more predictive in metastatic than in archival primary biopsy tissue samples. No significant interaction was found between treatment and expression levels of CDK4, CDK6, cyclin D1, and RB1. Palbociclib was efficacious in both luminal A and luminal B tumors. High CCNE1 mRNA expression was associated with poor antiproliferative activity of palbociclib in the POP trial (P = .005). CONCLUSION: Addition of palbociclib to fulvestrant demonstrated efficacy in all biomarker groups, although high CCNE1 mRNA expression was associated with relative resistance to palbociclib.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclina E/genética , Fulvestrant/uso terapéutico , Proteínas Oncogénicas/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclina E/biosíntesis , Femenino , Fulvestrant/administración & dosificación , Perfilación de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Proteínas Oncogénicas/biosíntesis , Piperazinas/administración & dosificación , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Piridinas/administración & dosificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo
15.
Pathol Res Pract ; 215(3): 539-545, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30638950

RESUMEN

BACKGROUND: GPR110 is a member of the adhesion G protein-coupled receptor family, which has been identified as an oncogene in various cancers, including hepatocellular carcinoma, lung cancer, prostatic cancer and glioma. Whereas the expression and the clinical relevance of GPR110 in gastric cancer has not been investigated. The research purpose of this study was to explore the expression pattern of GPR110 and evaluate its clinical-pathological and prognostic value in gastric cancer. METHODS: In this study, the expression of GPR110 was detected in 117 paired gastric cancer tissues and adjacent non-tumorous tissues by using qRT-PCR and immunohistochemical assays. Univariate Kaplan-Meier and multivariate Cox analysis were used to determine the prognostic value of GPR110 in GC. RESULTS: We demonstrated that the mRNA and protein levels of GPR110 in GC tissues were overexpressed than the adjacent non-tumorous tissues. Furthermore, elevated GPR110 protein expression was correlated with decreased overall and recurrence-free survival (P = 0.001 and P = 0.000, respectively). Univariate and multivariate analysis indicated that GPR110 protein level may serve as an independent prognostic indicator for determining prognosis of GC patients. CONCLUSIONS: Our study revealed that high expression of GPR110 predicts the poor prognosis of GC patients, and GPTR110 may function as a potential biomarker for the diagnosis of GC.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas Oncogénicas/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Neoplasias Gástricas/patología , Adulto , Anciano , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Pronóstico , Modelos de Riesgos Proporcionales , Neoplasias Gástricas/mortalidad
16.
J Cell Biochem ; 120(3): 4634-4643, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30320920

RESUMEN

A better understanding of breast cancer pathogenesis would contribute to improved diagnosis and therapy and potentially decreased mortality rates. Here, we found that the MORC family CW-type zinc finger 4 (MORC4) overexpression in breast cancer tissues is associated with poor survival, and the short-interfering RNA knockdown of MORC4 suppresses the growth of breast cancer cells by promoting apoptosis. To investigate the mechanisms associated with MORC4 upregulation, microRNAs potentially targeting MORC4 were analyzed, with miR-193b-3p identified as the regulator and a negative correlation between miR-193b-3p and MORC4 expression determined in both breast cancer cell lines and tissues. Further analysis verified that MORC4 silencing did not affect miR-193b-3p expression, although altered miR-193b-3p expression attenuated MORC4 protein levels. Moreover, dual-luciferase reporter assays verified miR-193b-3p binding to the 3' untranslated region of MORC4. Furthermore, restoration of miR-193b-3p expression in breast cancer cells led to decreased growth and activation of apoptosis, which was consistent with results associated with MORC4 silencing in breast cancer cells. These results identified MORC4 as differentially expressed in breast cancer cells and tissues and its downregulation by miR-193b-3p, as well as its roles in regulating the growth of breast cancer cells via regulation of apoptosis. Our findings offer novel insights into potential mechanisms associated with breast cancer pathogenesis.


Asunto(s)
Apoptosis , Neoplasias de la Mama/metabolismo , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/biosíntesis , Proteínas Oncogénicas/biosíntesis , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Femenino , Humanos , Células MCF-7 , MicroARNs/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , ARN Neoplásico/genética
17.
Biochimie ; 157: 1-9, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30391286

RESUMEN

Neoplastic cells display altered biosynthetic and bioenergetic machinery to support cell survival. Therefore, cancer cells optimally utilize all available fuel resources to pump their highly upregulated metabolic pathways. While glucose is the main carbon source, transformed cells also utilize other molecules, which can be utilized in metabolic pathways, designated as alternative fuels. Acetate is one of such alternative metabolic fuels, which is mainly consumed in carbohydrate and lipid metabolism. However, studies demonstrate the contradictory effects of acetate on tumor cell survival. Moreover, the mechanisms of its antitumor actions remain poorly understood. Further, the spectrum of acetate susceptible tumor targets needs to be characterized in order to optimize the use of acetate in maneuvering tumor progression as a therapeutic strategy. As the effect of acetate on survival properties of the tumor cells of thymic origin is not worked out, in the present study the effect of acetate was investigated against tumor cells derived from a murine thymoma designated as Dalton's Lymphoma (DL). Acetate treatment of tumor cells inhibited tumor cell survival accompanied by induction of apoptotic cell death, associated with modulated expression of cell survival regulatory HIF1α, ROS, p53, Caspase 3, Bax and HSP70 along with the elevated level of cytosolic cytochrome c. Acetate treatment also modulated the expression of pH regulators MCT-1 and V-ATPase accompanied by altered pH homeostasis. Expression of MDR and lipid metabolism regulatory molecules was also inhibited in tumor cells upon acetate exposure. Further, pre-exposure of tumor cells to α-CHC (α-cyano-4-hydroxycinnamate), an inhibitor of MCT-1, partially abrogated the cytotoxic action of acetate. These findings shed a new light regarding the effect and mechanisms of the exogenous acetate on the biology of tumor cells of thymic origin.


Asunto(s)
Acetatos/farmacología , Citotoxinas/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Homeostasis/efectos de los fármacos , Proteínas Oncogénicas/biosíntesis , Timoma , Neoplasias del Timo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Concentración de Iones de Hidrógeno , Metabolismo de los Lípidos/efectos de los fármacos , Ratones , Timoma/tratamiento farmacológico , Timoma/metabolismo , Timoma/patología , Neoplasias del Timo/tratamiento farmacológico , Neoplasias del Timo/metabolismo , Neoplasias del Timo/patología
18.
Gynecol Oncol ; 152(2): 387-395, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30442385

RESUMEN

OBJECTIVE: The study was to explore the roles of Nck1 in the angiogenesis of cervical squamous cell carcinoma (CSCC). METHODS: mRNA and protein levels were evaluated with real-time quantitative PCR and immunohistochemisty/western blotting respectively. The cancer microvessel density (MVD) was assayed with CD34 endothelial labeling. Nck1 gene knock-in (SiHa-Nck1+) and knock-down (SiHa-Nck1-) were achieved by gene transfection and siRNA respectively. Protein level from cellular supernatant was measured with ELISA. Proliferation, migration and tube formation of the Human Umbilical Vein Endothelial cells (HUVECs) were evaluated by CCK-8 cell viability assay, transwell chamber assay and in vitro Matrigel tubulation assay respectively. RESULTS: Nck1 level gradually increased from normal cervical epithelia to high-grade CIN, overexpressed in CSCC and was associated with cancer MVD. The ability of proliferation, migration and tube formation of HUVECs was enhanced in SiHa-Nck1+-treated while decreased in SiHa-NcK1--treated cells compared to SiHa-control-treated cells. Mechanistically, RAC1-GTP, p-PAK1 and MMP2 were increased in SiHa-NCK1+ cells and pretreatment with the Rac1 inhibitor (NSC23766) significantly decreased their levels. Furthermore, inhibition of PAK1 reduced MMP2 level in SiHa-Nck1+ cells whereas the level of Rac1-GTP was unaltered. Also, inhibition of Rac1 or PAK1 impaired angiogenesis-inducing capacity of cancer cells. CONCLUSIONS: Nck1 promotes the angiogenesis-inducing capacity of CSCC via the Rac1/PAK1/MMP2 signal pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma de Células Escamosas/irrigación sanguínea , Metaloproteinasa 2 de la Matriz/metabolismo , Proteínas Oncogénicas/metabolismo , Neoplasias del Cuello Uterino/irrigación sanguínea , Quinasas p21 Activadas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Persona de Mediana Edad , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Regulación hacia Arriba , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
19.
Gynecol Oncol ; 151(2): 327-336, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30209015

RESUMEN

OBJECTIVES: Readily apparent cyclin E1 expression occurs in 50% of HGSOC, but only half are linked to 19q12 locus amplification. The amplified/cyclin E1hi subset has intact BRCA1/2, unfavorable outcome, and is potentially therapeutically targetable. We studied whether non-amplified/cyclin E1hi HGSOC has similar characteristics. We also assessed the expression of cyclin E1 degradation-associated proteins, FBXW7 and USP28, as potential drivers of high cyclin E1 expression in both subsets. METHODS: 262 HGSOC cases were analyzed by in situ hybridization for 19q12 locus amplification and immunohistochemistry for cyclin E1, URI1 (another protein encoded by the 19q12 locus), FBXW7 and USP28 expression. Tumors were classified by 19q12 amplification status and correlated to cyclin E1 and URI1 expression, BRCA1/2 germline mutation, FBXW7 and USP28 expression, and clinical outcomes. Additionally, we assessed the relative genomic instability of amplified/cyclin E1hi and non-amplified/cyclin E1hi groups of HGSOC datasets from The Cancer Genome Atlas. RESULTS: Of the 82 cyclin E1hi cases, 43 (52%) were amplified and 39 (48%) were non-amplified. Unlike amplified tumors, non-amplified/cyclin E1hi tumor status was not mutually exclusive with gBRCA1/2 mutation. The non-amplified/cyclin E1hi group had significantly increased USP28, while the amplified/cyclin E1hi cancers had significantly lower FBXW7 expression consistent with a role for both in stabilizing cyclin E1. Notably, only the amplified/cyclin E1hi subset was associated with genomic instability and had a worse outcome than non-amplified/cyclin E1hi group. CONCLUSIONS: Amplified/cyclin E1hi and non-amplified/cyclin E1hi tumors have different pathological and biological characteristics and clinical outcomes indicating that they are separate subsets of cyclin E1hi HGSOC.


Asunto(s)
Cromosomas Humanos Par 19 , Ciclina E/genética , Cistadenocarcinoma Seroso/genética , Proteínas Oncogénicas/genética , Neoplasias Ováricas/genética , Adulto , Anciano , Anciano de 80 o más Años , Proteína BRCA1/biosíntesis , Proteína BRCA1/genética , Proteína BRCA2/biosíntesis , Proteína BRCA2/genética , Ciclina E/biosíntesis , Cistadenocarcinoma Seroso/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/biosíntesis , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Femenino , Amplificación de Genes , Inestabilidad Genómica , Humanos , Persona de Mediana Edad , Proteínas Oncogénicas/biosíntesis , Neoplasias Ováricas/metabolismo , Ubiquitina Tiolesterasa/biosíntesis , Ubiquitina Tiolesterasa/genética
20.
J Hum Genet ; 63(10): 1015-1028, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30082847

RESUMEN

Our original microRNA (miRNA) expression signatures (based on RNA sequencing) revealed that both strands of the miR-145 duplex (miR-145-5p, the guide strand, and miR-145-3p, the passenger strand) were downregulated in several types of cancer tissues. Involvement of passenger strands of miRNAs in cancer pathogenesis is a new concept in miRNA biogenesis. In our continuing analysis of lung adenocarcinoma (LUAD) pathogenesis, we aimed here to identify important oncogenes that were controlled by miR-145-5p and miR-145-3p. Downregulation of miR-145-5p and miR-145-3p was confirmed in LUAD clinical specimens. Functional assays showed that miR-145-3p significantly blocked the malignant abilities in LUAD cells, e.g., cancer cell proliferation, migration and invasion. Thus, the data showed that expression of the passenger strand of the miR-145-duplex acted as an anti-tumor miRNA. In LUAD cells, we identified four possible target genes (LMNB2, NLN, SIX4, and DDC) that might be regulated by both strands of miR-145. Among the possible targets, high expression of LMNB2 predicted a significantly poorer prognosis of LUAD patients (disease-free survival, p = 0.0353 and overall survival, p = 0.0017). Overexpression of LMNB2 was detected in LUAD clinical specimens and its aberrant expression promoted malignant transformation of LUAD cells. Genes regulated by anti-tumor miR-145-5p and miR-145-3p are closely involved in the molecular pathogenesis of LUAD. We suggest that they are promising prognostic markers for this disease. Our approach, based on the roles of anti-tumor miRNAs, will contribute to improved understanding of the molecular pathogenesis of LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Regulación Neoplásica de la Expresión Génica , MicroARNs , Proteínas Oncogénicas , ARN Neoplásico , Células A549 , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , MicroARNs/biosíntesis , MicroARNs/genética , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...