Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
J Biomed Biotechnol ; 2010: 485468, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20671927

RESUMEN

In this study, we investigated the role of hyaluronan (HA) in non-small cell lung cancer (NSCLC) since close association between HA level and malignancy has been reported. HA is an abundant extracellular matrix component and its synthesis is regulated by growth factors and cytokines that include epidermal growth factor (EGF) and interleukin-1beta (IL-1beta). We showed that treatment with recombinant EGF and IL-1beta, alone or in combination with TGF-beta, was able to stimulate HA production in lung adenocarcinoma cell line A549. TGF-beta/IL-1beta treatment induced epithelial to mesenchymal-like phenotype transition (EMT), changing cell morphology and expression of vimentin and E-cadherin. We also overexpressed hyaluronan synthase-3 (HAS3) in epithelial lung adenocarcinoma cell line H358, resulting in induced HA expression, EMT phenotype, enhanced MMP9 and MMP2 activities and increased invasion. Furthermore, adding exogenous HA to A549 cells and inducing HA H358 cells resulted in increased resistance to epidermal growth factor receptor (EGFR) inhibitor, Iressa. Together, these results suggest that elevated HA production is able to induce EMT and increase resistance to Iressa in NSCLC. Therefore, regulation of HA level in NSCLC may be a new target for therapeutic intervention.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Citocinas/farmacología , Epitelio/patología , Ácido Hialurónico/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/farmacología , Neoplasias Pulmonares/patología , Mesodermo/patología , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Epitelio/efectos de los fármacos , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glucuronosiltransferasa/genética , Glucuronosiltransferasa/metabolismo , Humanos , Hialuronano Sintasas , Ácido Hialurónico/farmacología , Interleucina-1beta/farmacología , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Mesodermo/efectos de los fármacos , Invasividad Neoplásica , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Factor de Crecimiento Transformador beta1/farmacología
2.
Nat Commun ; 10(1): 2065, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061416

RESUMEN

N6-Methyladenosine (m6A) modification has been implicated in the progression of several cancers. We reveal that during epithelial-mesenchymal transition (EMT), one important step for cancer cell metastasis, m6A modification of mRNAs increases in cancer cells. Deletion of methyltransferase-like 3 (METTL3) down-regulates m6A, impairs the migration, invasion and EMT of cancer cells both in vitro and in vivo. m6A-sequencing and functional studies confirm that Snail, a key transcription factor of EMT, is involved in m6A-regulated EMT. m6A in Snail CDS, but not 3'UTR, triggers polysome-mediated translation of Snail mRNA in cancer cells. Loss and gain functional studies confirm that YTHDF1 mediates m6A-increased translation of Snail mRNA. Moreover, the upregulation of METTL3 and YTHDF1 act as adverse prognosis factors for overall survival (OS) rate of liver cancer patients. Our study highlights the critical roles of m6A on regulation of EMT in cancer cells and translation of Snail during this process.


Asunto(s)
Adenosina/análogos & derivados , Transición Epitelial-Mesenquimal/genética , Neoplasias Hepáticas/genética , ARN/metabolismo , Factores de Transcripción de la Familia Snail/genética , Adenosina/metabolismo , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Estimación de Kaplan-Meier , Hígado/patología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Metilación , Metiltransferasas/genética , Metiltransferasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Análisis de Matrices Tisulares , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Cancer Res ; 65(10): 4181-90, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15899809

RESUMEN

Arachidonic acid (AA) metabolizing enzymes and peroxisome proliferator-activated receptors (PPARs) have been shown to regulate the growth of epithelial cells. We have previously reported that exposure to the 5-lipoxygenase activating protein-directed inhibitor MK886 but not the cyclooxygenase inhibitor, indomethacin, reduced growth, increased apoptosis, and up-regulated PPARalpha and gamma expression in breast cancer cell lines. In the present study, we explore approaches to maximizing the proapoptotic effects of PPARgamma on lung cancer cell lines. Non-small-cell cancer cell line A549 revealed dose-dependent PPARgamma reporter activity after treatment with MK886. The addition of indomethacin in combination with MK886 further increases reporter activity. We also show increased growth inhibition and up-regulation of apoptosis after exposure to MK886 alone, or in combination with indomethacin and the PPAR ligand, 15-deoxy-Delta12,14-prostaglandin J2 compared with single drug exposures on the adenocarcinoma cell line A549 and small-cell cancer cell lines H345, N417, and H510. Real-time PCR analyses showed increased PPAR mRNA and retinoid X receptor (RXR)alpha mRNA expression after exposure to MK886 and indomethacin in a time-dependent fashion. The results suggest that the principal proapoptotic effect of these drugs may be mediated through the known antiproliferative effects of the PPARgamma-RXR interaction. We therefore explored a three-drug approach to attempt to maximize this effect. The combination of low-dose MK886, ciglitazone, and 13-cis-retinoic acid interacted at least in a superadditive fashion to inhibit the growth of lung cancer cell lines A549 and H1299, suggesting that targeting PPARgamma and AA action is a promising approach to lung cancer growth with a favorable therapeutic index.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Ácido Araquidónico/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Prostaglandina D2/análogos & derivados , Acetofenonas/administración & dosificación , Apoptosis/efectos de los fármacos , Ácido Araquidónico/metabolismo , Caspasas/metabolismo , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Humanos , Indoles/administración & dosificación , Isotretinoína/administración & dosificación , Ligandos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores Activados del Proliferador del Peroxisoma/biosíntesis , Receptores Activados del Proliferador del Peroxisoma/genética , Prostaglandina D2/administración & dosificación , Pirimidinas/administración & dosificación , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor alfa X Retinoide/biosíntesis , Receptor alfa X Retinoide/genética , Tetrazoles/administración & dosificación , Tiazolidinedionas/administración & dosificación
5.
J Vis Exp ; (92): e51997, 2014 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-25350748

RESUMEN

Protein arginine methylation is one of the most abundant post-translational modifications in the nucleus. Protein arginine methylation can be identified and/or determined via proteomic approaches, and/or immunoblotting with methyl-arginine specific antibodies. However, these techniques sometimes can be misleading and often provide false positive results. Most importantly, these techniques cannot provide direct evidence in support of the PRMT substrate specificity. In vitro methylation assays, on the other hand, are useful biochemical assays, which are sensitive, and consistently reveal if the identified proteins are indeed PRMT substrates. A typical in vitro methylation assay includes purified, active PRMTs, purified substrate and a radioisotope labeled methyl donor (S-adenosyl-L-[methyl-(3)H] methionine). Here we describe a step-by-step protocol to isolate catalytically active PRMT1, a ubiquitously expressed PRMT family member. The methyl transferase activities of the purified PRMT1 were later tested on Ras-GTPase activating protein binding protein 1 (G3BP1), a known PRMT substrate, in the presence of S-adenosyl-L-[methyl-(3)H] methionine as the methyl donor. This protocol can be employed not only for establishing the methylation status of novel physiological PRMT1 substrates, but also for understanding the basic mechanism of protein arginine methylation.


Asunto(s)
Arginina/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Arginina/química , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , ADN Helicasas , Humanos , Metilación , Proteínas de Unión a Poli-ADP-Ribosa , Proteína-Arginina N-Metiltransferasas/química , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , S-Adenosilmetionina/análogos & derivados , S-Adenosilmetionina/química , S-Adenosilmetionina/metabolismo
6.
J Vis Exp ; (92): e51998, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25408172

RESUMEN

Anchorage-independent growth is the ability of transformed cells to grow independently of a solid surface, and is a hallmark of carcinogenesis. The soft agar colony formation assay is a well-established method for characterizing this capability in vitro and is considered to be one of the most stringent tests for malignant transformation in cells. This assay also allows for semi-quantitative evaluation of this capability in response to various treatment conditions. Here, we will demonstrate the soft agar colony formation assay using a murine lung carcinoma cell line, CMT167, to demonstrate the tumor suppressive effects of two members of the Wnt signaling pathway, Wnt7A and Frizzled-9 (Fzd-9). Concurrent overexpression of Wnt7a and Fzd-9 caused an inhibition of colony formation in CMT167 cells. This shows that expression of Wnt7a ligand and its Frizzled-9 receptor is sufficient to suppress tumor growth in a murine lung carcinoma model.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias/métodos , Animales , Línea Celular Tumoral , Neoplasias Pulmonares/patología , Ratones , Ensayo de Tumor de Célula Madre/métodos
7.
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
8.
Neoplasia ; 12(11): 946-56, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21076620

RESUMEN

Heparanase-1 (HPR1), an endoglycosidase that specifically degrades heparan sulfate (HS) proteoglycans, is overexpressed in a variety of malignancies. Our present study sought to determine whether oncogene BRAF and RAS mutations lead to increased HPR1 expression. Reverse transcription-polymerase chain reaction analysis revealed that HPR1 gene expression was increased in HEK293 cells transiently transfected with a mutant BRAF or RAS gene. Flow cytometric analysis revealed that B-Raf activation led to loss of the cell surface HS, which could be blocked by two HPR1 inhibitors: heparin and PI-88. Cotransfection of a BRAF or RAS mutant gene with HPR1 promoter-driven luciferase reporters increased luciferase reporter gene expression in HEK293 cells. Knockdown of BRAF expression in a BRAF-mutated KAT-10 tumor cell line led to the suppression of HPR1 gene expression, subsequently leading to increased cell surface HS levels. Truncational and mutational analyses of the HPR1 promoter revealed that the Ets-relevant elements in the HPR1 promoter were critical for BRAF activation-induced HPR1 expression. Luciferase reporter gene expression driven by a four-copy GA binding protein (GABP) binding site was significantly lower in BRAF siRNA-transfected KAT-10 cells than in the control siRNA-transfected cells. We further showed that BRAF knockdown led to suppression of the expression of the GABPß, an Ets family transcription factor involved in regulating HPR1 promoter activity. Taken together, our study suggests that B-Raf kinase activation plays an important role in regulating HPR1 expression. Increased HPR1 expression may contribute to the aggressive behavior of BRAF-mutated cancer.


Asunto(s)
Glucuronidasa/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Sitios de Unión/genética , Western Blotting , Línea Celular Tumoral , Activación Enzimática , Factor de Transcripción de la Proteína de Unión a GA/genética , Factor de Transcripción de la Proteína de Unión a GA/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucuronidasa/antagonistas & inhibidores , Glucuronidasa/metabolismo , Células HEK293 , Heparina/farmacología , Heparitina Sulfato/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Microscopía Fluorescente , Oligosacáridos/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Interferencia de ARN , Elementos de Respuesta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas ras/genética , Proteínas ras/metabolismo
9.
Curr Opin Pharmacol ; 9(4): 384-8, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19570713

RESUMEN

The association between chronic inflammation and lung cancer has been extensively reported. Despite the differences in the clinical manifestations of chronic obstructive pulmonary disease (COPD) compared with lung cancer, the underlying inflammatory pathogenesis of these two diseases may involve shared biological features. Identifying the mechanisms that regulate specific inflammatory mediators shared by both diseases will represent a major breakthrough. Chemokines play an important role in the inflammatory response. Recent evidence suggests that post-transcriptional regulation, through control of mRNA stability, could be an important mechanism of regulation of chemokine expression. Therefore, it would be important to study the role of RNA-binding proteins, such as hnRNPs, in chemokine expression after initial inflammatory response. HnRNPs have shown the ability to modulate inflammatory mediators expression by affecting mRNA stability of COX-2, TNFalpha and IL-1beta and iNOS. Moreover, overexpression of hnRNPs has been reported in cancer, for example, hnRNP A2/B1 in lung cancer. Thus, investigating the role of hnRNPs in chemokine expression may allow us to understand the molecular mechanisms involved in the regulation of the inflammatory response before its progression to chronic inflammation and/or tumor development.


Asunto(s)
Quimiocinas/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Mediadores de Inflamación/fisiología , Neoplasias Pulmonares/metabolismo , Animales , Quimiocinas/fisiología , Ribonucleoproteínas Nucleares Heterogéneas/fisiología , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Neoplasias Pulmonares/inmunología , Unión Proteica/inmunología
10.
PPAR Res ; 2008: 750238, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19277204

RESUMEN

Lung cancer is the leading cause of cancer death in the United States and five-year survival remains low. Numerous studies have shown that chronic inflammation may lead to progression of carcinogenesis. As a result of inflammatory stimulation, arachidonic acid (AA) metabolism produces proliferation mediators through complex and dynamic interactions of the products of the LOX/COX enzymes. One important mediator in the activation of the AA pathways is the nuclear protein PPARgamma. Targeting LOX/COX enzymes and inducing activation of PPARgamma have resulted in significant reduction of cell growth in lung cancer cell lines. However, specific COX-inhibitors have been correlated with an increased cardiovascular risk. Clinical applications are still being explored with a novel generation of dual LOX/COX inhibitors. PPARgamma activation through synthetic ligands (TZDs) has revealed a great mechanistic complexity since effects are produced through PPARgamma-dependent and -independent mechanisms. Furthermore, PPARgamma could also be involved in regulation of COX-2. Overexpression of PPARgamma has reported to play a role in control of invasion and differentiation. Exploring the function of PPARgamma, in this new context, may provide a better mechanistic model of its role in cancer and give an opportunity to design a more efficient therapeutic approach in combination with LOX/COX inhibitors.

11.
Breast Cancer Res Treat ; 89(2): 199-208, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15692763

RESUMEN

BACKGROUND: Our previous studies revealed that a subset of mammary ductal carcinoma in situ (DCIS) contained focally disrupted myoepithelial (ME) cell layers that were predominantly overlain by estrogen receptor (ER) negative cells, which showed a substantially higher rate of cell proliferation and genetic alterations than adjacent ER positive cells within the same duct. This study attempted to assess whether these cells also had a different expression profile on tumor progression related genes. DESIGN: Consecutive sections were made from frozen tissues of 30 DCIS with focally disrupted ME cell layers and associated ER negative cell clusters. ER negative and adjacent ER positive cells within the same duct were microdissected for RNA extraction and amplification. Amplified RNA was converted to biotin-labeled cDNAs and interrogated with 'Cancer PathwayFinder' arrays. RESULTS: Cells within each or among ER negative clusters were immunohistochemically and morphologically similar, whereas they differed substantially from adjacent cells within the same duct. Of 20-paired informative ER negative and positive cells, 15 genes were differentially expressed. Of which, 11(73.3%) were higher in ER negative, 2 (13.3%) were higher in ER positive, and 2 (13.3%) were equal in these cells (p <0.01). Of 11 up-regulated genes in ER negative cells, 8 indirectly or directly promote proliferation and progression, and 3 promote apoptosis. CONCLUSION: ER negative cell clusters showed a significantly higher expressing frequency of multiple tumor progression related genes than their adjacent ER positive counterparts, suggesting that they are likely to be biologically more aggressive and have a greater potential for invasion.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Intraductal no Infiltrante/genética , Carcinoma Intraductal no Infiltrante/patología , Perfilación de la Expresión Génica , Invasividad Neoplásica/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Progresión de la Enfermedad , Células Epiteliales , Femenino , Humanos , Inmunohistoquímica , Receptores de Estrógenos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA