Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Urol Oncol ; 39(3): 196.e1-196.e7, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33160845

RESUMEN

OBJECTIVE: We sought to identify heterogeneous nuclear ribonucleoprotein A3 (HNRNPA3) expression in bladder cancer and its relationship to clinicopathological findings and prognosis. METHODS: Immunohistochemical staining for HNRNPA3 was performed on 122 archived radical cystectomy specimens, with immunoreactivity being stratified on a 0 to 3 scale. The percentage of HNRNPA3 expressing tumor cells was calculated and multiplied by the staining score over an average of 5 areas to obtain a semiquantitative H-score (maximum value: 300). HNRNPA3 expression was categorized as high (≥80) or low (<80). RESULTS: The patients' median age was 70 years, and the median follow-up period was 39.4 months. High HNRNPA3 expression was significantly associated with lymph node metastasis (P= 0.014) and S100A8, S100A9 and uroplakin III expression (P= 0.028, 0.002, and 0.047, respectively). Log-rank tests indicated that high HNRNPA3 expression was significantly associated with disease progression and cancer-specific death (P= 0.013 and 0.006, respectively). In the Cox proportional hazards regression analysis, only lymph node metastasis was associated with disease progression and cancer-specific survival. CONCLUSION: HNRNPA3 may be a new biomarker to predict biologically aggressive cancers and determine the appropriate treatment modality in patients after radical cystectomy.


Asunto(s)
Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Metástasis Linfática , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología , Anciano , Cistectomía/métodos , Femenino , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/cirugía
2.
Eur Rev Med Pharmacol Sci ; 22(4): 993-1002, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29509247

RESUMEN

OBJECTIVE: In recent years, long non-coding RNAs (lncRNAs) have been identified to participate in tumor progression. The purpose of this study was to investigate the role of CACNA1G-AS1 in non-small cell lung cancer (NSCLC). PATIENTS AND METHODS: Quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) was used to detect the CACNA1G-AS1 expression level in 122 pairs of NSCLC and para-carcinoma normal tissue samples as well as in NSCLC cell lines. Moreover, the relationship of clinical pathological features with CACNA1G-AS1 was analyzed. Functional experiment cell lines were established using lentivirus and siRNA to study the effects of CACNA1G-AS1 on cell invasion and migration abilities. Several epithelial-mesenchymal transition (EMT) markers were measured using Western blotting. The expression level of HNRNPA2B1 was analyzed to further investigate the mechanism. RESULTS: The expression level of CACNA1G-AS1 in NSCLC tissues was significantly higher than that in para-carcinoma normal tissues, and the expression of CACNA1G-AS1 was higher in NSCLC cell lines than that in normal BEAS-2B cells. The higher CACNA1G-AS1 level was relative to more lymph node metastasis and distant metastasis. Function experiments revealed that CACNA1G-AS1 promoted cell invasion and migration. Also, CACNA1G-AS1 over-expression increased EMT in NSCLC cells. Besides, HNRNPA2B1 was regulated by CACNA1G-AS1 in NSCLC cells. CONCLUSIONS: CACNA1G-AS1 was identified as an oncogene in NSCLC for the first time, and could promote cell invasion, migration and EMT via increasing HNRNPA2B1 expression, providing a novel target for the biological therapy and prevention.


Asunto(s)
Canales de Calcio Tipo T/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Neoplasias Pulmonares/metabolismo , ARN Largo no Codificante/biosíntesis , Células A549 , Anciano , Canales de Calcio Tipo T/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Movimiento Celular/fisiología , Femenino , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Metástasis Linfática/genética , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , ARN Largo no Codificante/genética
3.
Tumour Biol ; 39(3): 1010428317694318, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28351333

RESUMEN

Increasing evidence has indicated that the splicing factor hnRNPA2B1 plays a direct role in cancer development, progression, gene expression, and signal transduction. Previous studies have shown that knocking down hnRNPA2B1 in breast cancer cells induces apoptosis, but the mechanism and other functions of hnRNPA2B1 in breast cancer are unknown. The goal of this study was to investigate the biological function, clinical significance, and mechanism of hnRNPA2B1 in breast cancer. The expression of hnRNPA2B1 in 92 breast cancer and adjacent normal tissue pairs was analyzed by immunohistochemical staining. Stable clones exhibiting knockdown of hnRNPA2B1 via small hairpin RNA expression were generated using RNA interference technology in breast cancer cell lines. The effects of hnRNPA2B1 on cell proliferation were examined by MTT and EdU assay, and cellular apoptosis and the cell cycle were examined by flow cytometry. A nude mouse xenograft model was established to elucidate the function of hnRNPA2B1 in tumorigenesis in vivo. The role of hnRNPA2B1 in signaling pathways was investigated in vitro. Our data revealed that hnRNPA2B1 was overexpressed in breast cancer tissue specimens and cell lines. Knockdown of hnRNPA2B1 reduced breast cancer cell proliferation, induced apoptosis, and prolonged the S phase of the cell cycle in vitro. In addition, hnRNPA2B1 knockdown suppressed subcutaneous tumorigenicity in vivo. On a molecular level, hnRNPA2B1 knockdown decreased signal transducer and activator of transcription 3 and extracellular-signal-regulated kinase 1/2 phosphorylation. We concluded that hnRNPA2B1 promotes the tumorigenic potential of breast cancer cells, MCF-7 and MDA-MB-231, through the extracellular-signal-regulated kinase 1/2 or signal transducer and activator of transcription 3 pathway, which may serve as a target for future therapies.


Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Factor de Transcripción STAT3/genética , Animales , Apoptosis/genética , Neoplasias de la Mama/patología , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/antagonistas & inhibidores , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Humanos , Sistema de Señalización de MAP Quinasas/genética , Células MCF-7 , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Mol Cell Biochem ; 427(1-2): 123-131, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28000042

RESUMEN

Heterogeneous nuclear ribonucleoprotein (hnRNP)A1, a member of the hnRNP family, is involved in a variety of RNA metabolisms. The hnRNPA1 expression is altered in some human diseases and mutations of the hnRNPA1 gene cause amyotrophic lateral sclerosis and multisystem proteinopathy. It has been therefore assumed that the dysregulation of hnRNPA1 is linked to the pathogenesis of the diseases. However, the mechanism underlying the regulation of the hnRNPA1 expression remains unknown. In this study, using cell-based models, we have found that hnRNPA1 negatively regulates its own mRNA expression by inhibiting the intron10 splicing of hnRNPA1 pre-mRNA. This mechanism likely serves as an autoregulation of the hnRNPA1 expression. We have also found that a low-grade excess of hnRNPA1 expression causes cytotoxicity by activating the mitochondrial apoptosis pathway. Collectively, these data suggest that the level of hnRNPA1 is strictly controlled to be within a certain range by the mRNA autoregulation in the physiological condition so that the cytotoxicity-causative alteration of hnRNPA1 expression does not take place.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Empalme del ARN/fisiología , ARN Mensajero/biosíntesis , Línea Celular , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , ARN Mensajero/genética
5.
BMC Cancer ; 16: 32, 2016 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-26791953

RESUMEN

BACKGROUND: The three members of the human heterochromatin protein 1 (HP1) family of proteins, HP1α, HP1ß, and HPγ, are involved in chromatin packing and epigenetic gene regulation. HP1α is encoded from the CBX5 gene and is a suppressor of metastasis. CBX5 is down-regulated at the transcriptional and protein level in metastatic compared to non-metastatic breast cancer. CBX5 shares a bi-directional promoter structure with the hnRNPA1 gene. But whereas CBX5 expression is down-regulated in metastatic cells, hnRNAP1 expression is constant. Here, we address the regulation of CBX5 in human breast cancer. METHODS: Transient transfection and transposon mediated integration of dual-reporter mini-genes containing the bi-directional hnRNPA1 and CBX5 promoter was performed to investigate transcriptional regulation in breast cancer cell lines. Bioinformatics and functional analysis were performed to characterize transcriptional events specifically regulating CBX5 expression. TSA treatment and Chromatin Immunoprecipitation (ChIP) were performed to investigate the chromatin structure along CBX5 in breast cancer cells. Finally, expression of hnRNPA1 and CBX5 mRNA isoforms were measured by quantitative reverse transcriptase PCR (qRT-PCR) in breast cancer tissue samples. RESULTS: We demonstrate that an hnRNPA1 and CBX5 bi-directional core promoter fragment does not comprise intrinsic capacity for specific CBX5 down-regulation in metastatic cells. Characterization of transcriptional events in the 20 kb CBX5 intron 1 revealed existence of several novel CBX5 transcripts. Two of these encode consensus HP1α protein but used autonomous promoters in intron 1 by which HP1α expression could be de-coupled from the bi-directional promoter. In addition, another CBX5 transcriptional isoform, STET, was discovered. This transcript includes CBX5 exon 1 and part of intron 1 sequences but lacks inclusion of HP1α encoding exons. Inverse correlation between STET and HP1α coding CBX5 mRNA expression was observed in breast cancer cell lines and tissue samples from breast cancer patients. CONCLUSION: We find that HP1α is down-regulated in a mechanism involving CBX5 promoter downstream sequences and that regulation through alternative polyadenylation and splicing generates a transcript, STET, with potential importance in carcinogenesis.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Cromosómicas no Histona/biosíntesis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Regiones Promotoras Genéticas , Neoplasias de la Mama/patología , Línea Celular Tumoral , Cromatina/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Exones/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Intrones/genética , Metástasis de la Neoplasia , Empalme del ARN/genética
6.
J Biomol Tech ; 26(4): 142-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26543439

RESUMEN

The Encyclopedia of DNA Elements (ENCODE) Project aims to identify all functional sequence elements in the human genome sequence by use of high-throughput DNA/cDNA sequencing approaches. To aid the standardization, comparison, and integration of data sets produced from different technologies and platforms, the ENCODE Consortium selected several standard human cell lines to be used by the ENCODE Projects. The Tier 1 ENCODE cell lines include GM12878, K562, and H1 human embryonic stem cell lines. GM12878 is a lymphoblastoid cell line, transformed with the Epstein-Barr virus, that was selected by the International HapMap Project for whole genome and transcriptome sequencing by use of the Illumina platform. K562 is an immortalized myelogenous leukemia cell line. The GM12878 cell line is attractive for the ENCODE Projects, as it offers potential synergy with the International HapMap Project. Despite the vast amount of sequencing data available on the GM12878 cell line through the ENCODE Project, including transcriptome, chromatin immunoprecipitation-sequencing for histone marks, and transcription factors, no small interfering siRNA-mediated knockdown studies have been performed in the GM12878 cell line, as cationic lipid-mediated transfection methods are inefficient for lymphoid cell lines. Here, we present an efficient and reproducible method for transfection of a variety of siRNAs into the GM12878 and K562 cell lines, which subsequently results in targeted protein depletion.


Asunto(s)
ARN Interferente Pequeño/genética , Transfección/métodos , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/genética , Electroporación , Expresión Génica , Técnicas de Silenciamiento del Gen , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Células K562 , Interferencia de ARN
7.
Oncol Rep ; 34(3): 1231-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26151392

RESUMEN

CD44 is a transmembrane receptor for hyaluronic acid. CD44 pre-mRNA contains 19 exons, 9 of which are alternatively spliced. Among the CD44 spliced variants, the v4-7 variant, one of the v6 exon-containing isoforms that contains variable exon 4, 5, 6 and 7, confers metastatic potential to non-metastatic cells. Splicing of CD44 and the function of CD44 isoforms are different in breast cancer cells. hnRNP A1 is a ubiquitously expressed protein with an inhibitory function in pre-mRNA splicing. We showed that CD44v6 isoform, which includes all of the v6-containing mRNA isoforms, had the highest expression level in non-metatatic breast cancer cells (MCF7) when compared to the level in metastatic breast cancer cells (MDA-MB-231) and normal breast cells (MCF10A). Furthermore we showed that hnRNP A1 knockdown regulated splicing of CD44 differently in breast cancer cells. We showed here that CD44 isoform expression is completely different in MDA-MB-231 cells than that in MCF7 and MCF10A cells, whereas MCF7 and MCF10A cells had a similar expression pattern of CD44 isoforms. RT-PCR analysis of CD44v6 showed that MCF7 and MCF10A cells predominantly expressed the c5v6v7v8v9v10c6 isoform. However, in addition to this isoform, MDA-MB-231 cells also expressed the c5v6v8v9v10c6 and c5v6c6 isoforms. We also found that knockdown of hnRNP A1 significantly reduced the expression of c5v6v7v8v9v10c6 and c5v6v8v9v10c6, and promoted the expression of c5v6c6. hnRNP A1 knockdown significantly induced cell death. In addition, hnRNP A1 knockdown induced a decrease in cell invasion in the MDA-MB-231 cells. Our results indicate that the knockdown of hnRNP A1 has a specific function on the splicing of CD44 in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Receptores de Hialuranos/genética , Invasividad Neoplásica/genética , Empalme Alternativo , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/genética , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Receptores de Hialuranos/biosíntesis , Immunoblotting , Invasividad Neoplásica/patología , Isoformas de Proteínas , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
8.
Inflamm Bowel Dis ; 21(7): 1541-52, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25901972

RESUMEN

BACKGROUND: The role of hnRNP A1 in the onset of intestinal inflammation remains unclear. This study investigated the function of hnRNP A1 in mice enteritis models. METHODS: C57Bl6/J mice were intraperitoneally injected with anti-CD3 antibodies to develop enteritis. In the DSS-induced colitis group, the mice were allowed free access to 3% DSS solution in their drinking water for 5 days. 3H-mannitol flux and complementary DNA array tests were used to assess the intestinal barrier function and messenger RNA (mRNA) expression, respectively. Real-time PCR was performed after immunoprecipitation with anti-hnRNP antibodies to determine the specific mRNA binding of hnRNP A1. RESULTS: The hnRNP A1 expression was increased in the intestine of the mouse at 24 hours after treatment with anti-CD3 antibodies and 5 days after starting DSS administration. Small interfering RNA (siRNA) against hnRNP A1 exacerbated the intestinal injuries in both models. According to the microarray analysis, trefoil factor 2 (TFF2) was identified as a candidate molecule targeted by hnRNP A1 in the anti-CD3 antibody-induced enteritis group. Moreover, the binding between hnRNP A1 and TFF2 mRNA significantly increased in the enteritis mice, and the administration of siRNA against either hnRNP A1 or TFF2 exacerbated the degree of intestinal injury. In the DSS-induced colitis group, treatment with the siRNA of hnRNP A1 worsened the intestinal injury, while the expression of TFF3 did not change. CONCLUSIONS: hnRNP A1 improves intestinal injury in anti-CD3 antibody-induced enteritis mice through the upregulation of TFF2, which regulates apoptosis and enhances epithelial restoration, whereas this molecule ameliorates DSS-induced colitis through a different pathway.


Asunto(s)
Apoptosis , Enteritis/genética , Regulación de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Mucinas/genética , Proteínas Musculares/genética , Péptidos/genética , ARN Mensajero/genética , Animales , Western Blotting , Complejo CD3/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Enteritis/inmunología , Enteritis/metabolismo , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Etiquetado Corte-Fin in Situ , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Mucinas/biosíntesis , Proteínas Musculares/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor Trefoil-2
9.
J Biol Chem ; 289(39): 26973-26988, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25124043

RESUMEN

The basic helix-loop-helix transcription factor hASH1, encoded by the ASCL1 gene, plays an important role in neurogenesis and tumor development. Recent findings indicate that local oxygen tension is a critical determinant for the progression of neuroblastomas. Here we investigated the molecular mechanisms underlying the oxygen-dependent expression of hASH1 in neuroblastoma cells. Exposure of human neuroblastoma-derived Kelly cells to 1% O2 significantly decreased ASCL1 mRNA and hASH1 protein levels. Using reporter gene assays, we show that the response of hASH1 to hypoxia is mediated mainly by post-transcriptional inhibition via the ASCL1 mRNA 5'- and 3'-UTRs, whereas additional inhibition of the ASCL1 promoter was observed under prolonged hypoxia. By RNA pulldown experiments followed by MALDI/TOF-MS analysis, we identified heterogeneous nuclear ribonucleoprotein (hnRNP)-A2/B1 and hnRNP-R as interactors binding directly to the ASCL1 mRNA 5'- and 3'-UTRs and influencing its expression. We further demonstrate that hnRNP-A2/B1 is a key positive regulator of ASCL1, findings that were also confirmed by analysis of a large compilation of gene expression data. Our data suggest that a prominent down-regulation of hnRNP-A2/B1 during hypoxia is associated with the post-transcriptional suppression of hASH1 synthesis. This novel post-transcriptional mechanism for regulating hASH1 levels will have important implications in neural cell fate development and disease.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Proteínas de Neoplasias/metabolismo , Neuroblastoma/metabolismo , Regiones no Traducidas 3' , Regiones no Traducidas 5' , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Proteínas de Neoplasias/genética , Neuroblastoma/genética , Regiones Promotoras Genéticas , Conejos , Ratas Wistar
10.
FASEB J ; 28(8): 3720-33, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24823364

RESUMEN

Cellular senescence is a permanent cell cycle arrest triggered by different stimuli. We recently identified up-regulation of microRNA (miR)-494 as a component of the genetic program leading to senescence of human diploid IMR90 fibroblasts. Here, we used 2-dimensional differential gel electrophoresis (2D-DIGE) coupled to mass spectrometry to profile protein expression changes induced by adoptive overexpression of miR-494 in IMR90 cells. miR-494 induced robust perturbation of the IMR90 proteome by significantly (P≤0.05) down-regulating a number of proteins. Combination of mass spectrometry-based identification of down-regulated proteins and bioinformatic prediction of the miR-494 binding sites on the relevant mRNAs identified 26 potential targets of miR-494. Among them, computational analysis identified 7 potential evolution-conserved miR-494 targets. Functional miR-494 binding sites were confirmed in 3'-untranslated regions (UTRs) of 4 of them [heterogeneous nuclear ribonucleoprotein A3 (hnRNPA3), protein disulfide isomerase A3 (PDIA3), UV excision repair protein RAD23 homolog B (RAD23B), and synaptotagmin-binding cytoplasmic RNA-interacting protein (SYNCRIP)/heterogeneous nuclear ribonucleoprotein Q (hnRNPQ)]. Their reduced expression correlated with miR-494 up-regulation in senescent cells. RNA interference-mediated knockdown of hnRNPA3 and, to a lesser extent, RAD23B mirrored the senescent phenotype induced by miR-494 overexpression, blunting cell proliferation and causing up-regulation of SA-ß-galactosidase and DNA damage. Ectopic expression of hnRNPA3 or RAD23B slowed the appearance of the senescent phenotype induced by miR-494. Overall, these findings identify novel miR-494 direct targets that are involved in cellular senescence.


Asunto(s)
Senescencia Celular/genética , Enzimas Reparadoras del ADN/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Fibroblastos/citología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Ribonucleoproteínas Nucleares Heterogéneas/biosíntesis , MicroARNs/fisiología , Proteína Disulfuro Isomerasas/biosíntesis , Línea Celular , Senescencia Celular/fisiología , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Células HEK293 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/fisiología , Ribonucleoproteínas Nucleares Heterogéneas/genética , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Humanos , Espectrometría de Masas , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/fisiología , Proteoma , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Transfección , Regulación hacia Arriba
11.
Int J Oncol ; 43(2): 548-60, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23708087

RESUMEN

Lung cancer remains the leading cause of cancer-related mortality for both men and women. Tumor recurrence and metastasis is the major cause of lung cancer treatment failure and death. The microRNA­200 (miR-200) family is a powerful regulator of the epithelial-mesenchymal transition (EMT) process, which is essential in tumor metastasis. Nevertheless, miR-200 family target genes that promote metastasis in non-small cell lung cancer (NSCLC) remain largely unknown. Here, we sought to investigate whether the microRNA-200 family regulates our previously identified NSCLC prognostic marker genes associated with metastasis, as potential molecular targets. Novel miRNA targets were predicted using bioinformatics tools based on correlation analyses of miRNA and mRNA expression in 57 squamous cell lung cancer tumor samples. The predicted target genes were validated with quantitative RT-PCR assays and western blot analysis following re-expression of miR-200a, -200b and -200c in the metastatic NSCLC H1299 cell line. The results show that restoring miR-200a or miR-200c in H1299 cells induces downregulation of DLC1, ATRX and HFE. Reinforced miR-200b expression results in downregulation of DLC1, HNRNPA3 and HFE. Additionally, miR-200 family downregulates HNRNPR3, HFE and ATRX in BEAS-2B immortalized lung epithelial cells in quantitative RT-PCR and western blot assays. The miR-200 family and these potential targets are functionally involved in canonical pathways of immune response, molecular mechanisms of cancer, metastasis signaling, cell-cell communication, proliferation and DNA repair in Ingenuity pathway analysis (IPA). These results indicate that re-expression of miR-200 downregulates our previously identified NSCLC prognostic biomarkers in metastatic NSCLC cells. These results provide new insights into miR-200 regulation in lung cancer metastasis and consequent clinical outcome, and may provide a potential basis for innovative therapeutic approaches for the treatment of this deadly disease.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , ADN Helicasas/biosíntesis , Transición Epitelial-Mesenquimal/genética , Femenino , Proteínas Activadoras de GTPasa/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteína de la Hemocromatosis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Antígenos de Histocompatibilidad Clase I/biosíntesis , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Proteínas de la Membrana/biosíntesis , Metástasis de la Neoplasia , Proteínas Nucleares/biosíntesis , Pronóstico , Proteínas Supresoras de Tumor/biosíntesis , Proteína Nuclear Ligada al Cromosoma X
12.
Stem Cells ; 31(12): 2647-58, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23495120

RESUMEN

Self-renewal and pluripotency of human embryonic stem cells (hESCs) are a complex biological process for maintaining hESC stemness. However, the molecular mechanisms underlying these special properties of hESCs are not fully understood. Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) is a multifunctional RNA-binding protein whose expression is related to cell proliferation and carcinogenesis. In this study, we found that hnRNP A2/B1 expression was localized to undifferentiated hESCs and decreased upon differentiation of hESCs. hnRNP A2/B1 knockdown reduced the number of alkaline phosphatase-positive colonies in hESCs and led to a decrease in the expression of pluripotency-associated transcription factors OCT4, NANOG, and SOX2, indicating that hnRNP A2/B1 is essential for hESC self-renewal and pluripotency. hnRNP A2/B1 knockdown increased the expression of gene markers associated with the early development of three germ layers, and promoted the process of epithelial-mesenchymal transition, suggesting that hnRNP A2/B1 is required for maintaining the undifferentiated and epithelial phenotypes of hESCs. hnRNP A2/B1 knockdown inhibited hESC proliferation and induced cell cycle arrest in the G0/G1 phase before differentiation via degradation of cyclin D1, cyclin E, and Cdc25A. hnRNP A2/B1 knockdown increased p27 expression and induced phosphorylation of p53 and Chk1, suggesting that hnRNP A2/B1 also regulates the G1/S transition of hESC cell cycle through the control of p27 expression and p53 and Chk1 activity. Analysis of signaling molecules further revealed that hnRNP A2/B1 regulated hESC proliferation in a PI3K/Akt-dependent manner. These findings provide for the first time mechanistic insights into how hnRNP A2/B1 regulates hESC self-renewal and pluripotency.


Asunto(s)
Células Madre Embrionarias/citología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Técnicas de Cocultivo , Regulación hacia Abajo , Células Madre Embrionarias/metabolismo , Fibroblastos/citología , Fase G1/fisiología , Técnicas de Silenciamiento del Gen , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Ratones , Células Madre Pluripotentes/metabolismo , Fase S/fisiología , Transfección
13.
Stem Cells ; 31(5): 906-17, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23335105

RESUMEN

To investigate the functional involvements of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1) in smooth muscle cell (SMC) differentiation from stem cells, embryonic stem cells were cultivated on collagen IV-coated plates to allow for SMC differentiation. We found that hnRNPA1 gene and protein expression was upregulated significantly during differentiation and coexpressed with SMC differentiation markers in the stem cell-derived SMCs as well as embryonic SMCs of 12.5 days of mouse embryos. hnRNPA1 knockdown resulted in downregulation of smooth muscle markers and transcription factors, while enforced expression of hnRNPA1 enhanced the expression of these genes. Importantly, knockdown of hnRNPA1 also resulted in impairment of SMC differentiation in vivo. Moreover, we demonstrated that hnRNPA1 could transcriptionally regulate SMC gene expression through direct binding to promoters of Acta2 and Tagln genes using luciferase and chromatin immunoprecipitation assays. We further demonstrated that the binding sites for serum response factor (SRF), a well-investigated SMC transcription factor, within the promoter region of the Acta2 and Tagln genes were responsible for hnRNPA1-mediated Acta2 and Tagln gene expression using in vitro site-specific mutagenesis and luciferase activity analyses. Finally, we also demonstrated that hnRNPA1 upregulated the expression of SRF, myocyte-specific enhancer factor 2c (MEF2c), and myocardin through transcriptional activation and direct binding to promoters of the SRF, MEF2c, and Myocd genes. Our findings demonstrated that hnRNPA1 plays a functional role in SMC differentiation from stem cells in vitro and in vivo. This indicates that hnRNPA1 is a potential modulating target for deriving SMCs from stem cells and cardiovascular regenerative medicine.


Asunto(s)
Células Madre Embrionarias/fisiología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Miocitos del Músculo Liso/fisiología , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Ratones , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Activación Transcripcional , Regulación hacia Arriba
14.
Int J Cancer ; 132(5): 1080-9, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-22821376

RESUMEN

Heterogeneous ribonucleoprotein (hnRNP) A1 is a member of the A/B subfamily of ubiquitously expressed hnRNPs, which have a wide variety of functions in gene expression and signal transduction. To investigate the biological function and clinical significance of hnRNP A1 in hepatocellular carcinoma (HCC), we measured hnRNP A1 expression in four HCC cell lines and two independent cohorts of HCC patients. We found that hnRNP A1 was overexpressed in the highly metastatic HCC cell lines and in tumor tissues of patients with recurrent HCC. Knockdown of hnRNP A1 in highly metastatic HCC cells caused a significant decrease in cell invasion, while upregulation of hnRNP A1 in poorly metastatic HCC cells led to a significant increase in their invasive capacity. We found that this effect may occur through the regulation of CD44v6 expression by hnRNP A1 in HCC cells. Both quantitative reverse transcription-polymerase chain reaction (qRT-RCR) and immunohistochemistry revealed that hnRNP A1 was upregulated in HCC tissues and coincided with overexpression of CD44v6. HCC patients with high hnRNP A1 tended to have higher levels of CD44v6, shorter overall survival (OS) and higher rates of tumor recurrence. Multivariate analyses revealed that hnRNP A1 alone or in combination with CD44v6 were independent prognostic indicators for OS and time to recurrence and have potential as therapeutic targets. In conclusion, overexpression of hnRNP A1 promotes HCC invasion by regulating the level of CD44v6 and indicates a poor prognosis for HCC patients after curative resection.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Receptores de Hialuranos/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Femenino , Estudios de Seguimiento , Ribonucleoproteína Nuclear Heterogénea A1 , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Humanos , Receptores de Hialuranos/genética , Inmunohistoquímica , Neoplasias Hepáticas/genética , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Pronóstico , Regulación hacia Arriba
15.
Brain Dev ; 34(3): 213-22, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21561730

RESUMEN

Spinal muscular atrophy (SMA) is a common autosomal recessive neuromuscular disorder that is caused by loss of the survival motor neuron gene, SMN1. SMA treatment strategies have focused on production of the SMN protein from the almost identical gene, SMN2. Valproic acid (VPA) is a histone deacetylase inhibitor that can increase SMN levels in some SMA cells or SMA patients through activation of SMN2 transcription or splicing correction of SMN2 exon 7. It remains to be clarified what concentration of VPA is required and by what mechanisms the SMN production from SMN2 is elicited. We observed that in two fibroblast cell lines from Japanese SMA patients, more than 1mM of VPA increased SMN2 expression at both the transcript and protein levels. VPA increased not only full-length (FL) transcript level but also exon 7-excluding (Δ7) transcript level in the cell lines and did not change the ratio of FL/Δ7, suggesting that SMN2 transcription was mainly activated. We also found that VPA modulated splicing factor expression: VPA increased the expression of splicing factor 2/alternative splicing factor (SF2/ASF) and decreased the expression of heterogeneous nuclear ribonucleoprotein A1 (hnRNPA1). In conclusion, more than 1mM of VPA activated SMN2 transcription and modulated the expression of splicing factors in our SMA fibroblast cell lines.


Asunto(s)
Expresión Génica/efectos de los fármacos , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Fármacos Neuroprotectores/farmacología , Proteínas Nucleares/biosíntesis , Proteínas de Unión al ARN/biosíntesis , Ácido Valproico/farmacología , Adulto , Western Blotting , Línea Celular , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Lactante , Atrofia Muscular Espinal/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factores de Empalme Serina-Arginina , Proteína 2 para la Supervivencia de la Neurona Motora/biosíntesis , Transcripción Genética/efectos de los fármacos
16.
J Neuroimmunol ; 235(1-2): 56-69, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21570130

RESUMEN

Multiple sclerosis (MS) patients make antibodies to heterogeneous nuclear ribonuclear protein A1 (hnRNP-A1), a nucleocytoplasmic protein. We hypothesized this autoimmune reaction might contribute to neurodegeneration. Antibodies from MS patients reacted with hnRNP-A1-'M9', its nuclear translocation sequence. Transfection of anti-M9 antibodies into neurons resulted in neuronal injury and changes in transcripts related to hnRNP-A1 function. Importantly, RNA levels for the spinal paraplegia genes (SPGs) decreased. Changes in SPG RNA levels were confirmed in neurons purified from MS brains. Also, we show molecular interactions between spastin (the encoded protein of SPG4) and hnRNP-A1. These data suggest a link between autoimmunity, clinical phenotype and neurodegeneration in MS.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Esclerosis Múltiple/inmunología , Esclerosis Múltiple/metabolismo , Degeneración Nerviosa/inmunología , Adulto , Anciano , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/patología , Línea Celular , Células Cultivadas , Corteza Cerebral/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Femenino , Ribonucleoproteína Nuclear Heterogénea A1 , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple/patología , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Adulto Joven
17.
Cancer Res ; 71(13): 4464-72, 2011 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-21586613

RESUMEN

The process of alternative splicing is widely misregulated in cancer, but the contribution of splicing regulators to cancer development is largely unknown. In this study, we found that the splicing factor hnRNP A2/B1 is overexpressed in glioblastomas and is correlated with poor prognosis. Conversely, patients who harbor deletions of the HNRNPA2B1 gene show better prognosis than average. Knockdown of hnRNP A2/B1 in glioblastoma cells inhibited tumor formation in mice. In contrast, overexpression of hnRNP A2/B1 in immortal cells led to malignant transformation, suggesting that HNRNPA2B1 is a putative proto-oncogene. We then identified several tumor suppressors and oncogenes that are regulated by HNRNPA2B1, among them are c-FLIP, BIN1, and WWOX, and the proto-oncogene RON. Knockdown of RON inhibited hnRNP A2/B1 mediated transformation, which implied that RON is one of the mediators of HNRNPA2B1 oncogenic activity. Together, our results indicate that HNRNPA2B1 is a novel oncogene in glioblastoma and a potential new target for glioblastoma therapy.


Asunto(s)
Neoplasias Encefálicas/genética , Genes Supresores de Tumor , Glioblastoma/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Empalme Alternativo , Animales , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Glioblastoma/metabolismo , Glioblastoma/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Humanos , Ratones , Células 3T3 NIH , Proto-Oncogenes Mas , Proteínas Tirosina Quinasas Receptoras/genética , Regulación hacia Arriba
18.
Cancer Res ; 70(18): 7137-47, 2010 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-20807810

RESUMEN

Heterogeneous nuclear ribonucleoprotein A2/B1 (hnRNP A2/B1) has been reported to be overexpressed in lung cancer and in other cancers such as breast, pancreas, and liver. However, a mechanism linking hnRNP A2/B1 overexpression and progression to cancer has not yet been definitively established. To elucidate this mechanism, we have silenced hnRNPA2/B1 mRNA in non-small-cell lung cancer cell lines A549, H1703, and H358. These cell lines present different levels of expression of epithelial-to-mesenchymal transition (EMT) markers such as E-cadherin, fibronectin, and vimentin. Microarray expression analysis was performed to evaluate the effect of silencing hnRNP A2/B1 in A549 cells. We identified a list of target genes, affected by silencing of hnRNP A2/B1, that are involved in regulation of migration, proliferation, survival, and apoptosis. Silencing hnRNP A2/B1 induced formation of cell clusters and increased proliferation. In the anchorage-independent assay, silencing hnRNP A2/B1 increased colony formation by 794% in A549 and 174% in H1703 compared with a 25% increase in proliferation, in both cell lines, in a two-dimensional proliferation assay. Silencing hnRNP A2/B1 decreased migration in intermediate cell line A549 and mesenchymal cell line H1703; however, no changes in proliferation were observed in epithelial cell line H358. Silencing hnRNP A2/B1 in A549 and H1703 cells correlated with an increase of E-cadherin expression and downregulation of the E-cadherin inhibitors Twist1 and Snai1. These data suggest that expression of hnRNP A2/B1 may play a role in EMT, in nonepithelial lung cancer cell lines A549 and H1703, through the regulation of E-cadherin expression.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Neoplasias Pulmonares/patología , Cadherinas/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Células Epiteliales/patología , Silenciador del Gen , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mesodermo/patología , Proteínas Nucleares/biosíntesis , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis , Proteína 1 Relacionada con Twist/biosíntesis
19.
Pathol Oncol Res ; 16(1): 55-9, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19609729

RESUMEN

This study was performed to compare osteopontin (OPN), beta-catenin and heterogeneous nuclear ribonucleoprotein B1 (hnRNP B1) immunreactivities in small cell lung carcinomas (SCLC) and non-small cell lung carcinomas (NSCLC). Correlation of these three antibodies with grade and clinicopathologic stage of the tumor in NSCLC was also investigated. Twenty-nine SCLC, 6 large cell carcinoma, 36 adenocarcinoma and 30 squamous cell carcinoma (SCC), totally 101 cases, were included in this study. OPN, beta-catenin and hnRNP B1 expressions were immunohistochemically evaluated. OPN positivity was 6.9% in SCLC and 67% in NSCLC. When NSCLC types were individually considered, OPN positivity was 66.7% in large cell carcinoma, 80% in SCC and 55.6% in adenocarcinomas. beta-catenin positivity was observed in 48.6% of NSCLC and none of SCLC cases. These results were statistically significant (p < 0.05). Neither grade nor stage of NSCLC was correlated with osteopontin, beta-catenin or hnRNP B1 immunreactivity. We observed that OPN and beta-catenin are useful in differentiating SCLC from NSCLC. This may be helpful in small lung biopsies where morphology is obscured by crush artifacts.


Asunto(s)
Biomarcadores de Tumor/análisis , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Neoplasias Pulmonares/metabolismo , Osteopontina/biosíntesis , beta Catenina/biosíntesis , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/patología , Estadificación de Neoplasias
20.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 39(5): 805-7, 2008 Sep.
Artículo en Chino | MEDLINE | ID: mdl-19024319

RESUMEN

OBJECTIVE: To test the effect of RNA interference of hnRNP Bx gene in human lung cell line A549. METHODS: The RNAs of cell line A549 were interfered by the RNA interference technique. The hnRNP B1 mRNAs were detected by fluorescence quantity RT-PCR. The proteins of hnRNP B1 were detected by Western blot technique. The growth rate of the cells was analysed by MTT. RESULTS: The growth of the cells was suppressed significantly. The mRNA and protein expressions of hnRNP B1 were inhibited. CONCLUSION: The expression of hnRNP B1 can be inhibited by RNA interference. RNA interference could become a possible clinical therapy for lung cancers.


Asunto(s)
Adenocarcinoma/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Neoplasias Pulmonares/genética , ARN Interferente Pequeño/genética , Adenocarcinoma/patología , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/biosíntesis , Humanos , Neoplasias Pulmonares/patología , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...