Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Otolaryngol Head Neck Surg ; 147(4): 726-32, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22568942

RESUMEN

OBJECTIVE: The presence of regional metastases in patients with head and neck squamous cell carcinoma (HNSCC) is a common and adverse event associated with poor prognosis. The authors' recent work on human HNSCC tissues underlies Snail's role as a molecular prognostic marker for HNSCC. Snail positivity is significantly predictive of poorly differentiated, lymphovascular invasive, and regionally metastatic tumors. Here, the authors investigate the capacity of Snail to drive epithelial-mesenchymal transition (EMT) in human oral epithelial cell lines and its ability to confer drug resistance. STUDY DESIGN: Snail was overexpressed in HNSCC and oral epithelial cell lines. Anchorage independent growth assays, wound healing assays, invasion and migration assays, spheroid modeling, and cell survival assays were performed. SETTING: Academic tertiary medical center. SUBJECTS AND METHODS: Snail overexpressing HNSCC (OSC, Tu212, Tu686) and oral epithelial cell lines (HOK 16-B, OKF-6) were evaluated using assays for wound healing, invasion and migration, 3-dimensional growth, Western blot, and immunofluorescence. RESULTS: The overexpression of Snail in human HNSCC and oral epithelial cell lines drives EMT. The transfection of Snail confers the expression of a mesenchymal molecular signature, including downregulation of the epithelial adherens, such as E-cadherin and ß-catenin, and induction of mesenchymal markers. Snail-overexpressing cell lines demonstrate rapid growth in Anchorage-independent growth assays, a decreased capacity to form tight spheroids, an increased resistance to erlotinib, and an increased capacity for invasion. CONCLUSION: Snail controls the mesenchymal phenotype and drives erlotinib resistance in HNSCC cells. Snail may prove to be a useful marker in predicting epidermal growth factor receptor inhibitor responsiveness.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Quinazolinas/farmacología , Factores de Transcripción/metabolismo , Biomarcadores de Tumor/metabolismo , Western Blotting , Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Supervivencia Celular , Distribución de Chi-Cuadrado , Regulación hacia Abajo , Resistencia a Antineoplásicos , Clorhidrato de Erlotinib , Técnica del Anticuerpo Fluorescente , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Fenotipo , Factores de Transcripción de la Familia Snail , Cicatrización de Heridas , beta Catenina/metabolismo
2.
Otolaryngol Head Neck Surg ; 142(5): 753-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20416468

RESUMEN

OBJECTIVES: To determine the role of ZEB1 in the inflammation-induced promotion of the epithelial-mesenchymal transition (EMT) in head and neck squamous cell carcinoma (HNSCC). STUDY DESIGN: A molecular biology study. Real-time quantitative reverse-transcriptase polymerase chain reaction (RT-PCR), Western blot analysis, and immunohistochemical staining of human HNSCC tissue sections were used to determine how inflammation affects the transcriptional repressor, ZEB1. SETTING: An academic hospital laboratory. SUBJECTS AND METHODS: Relative ZEB1 RNA levels were determined by RT-PCR, and protein expression was evaluated in situ by immunohistochemical staining of human HNSCC tissue sections. RESULTS: IL-1beta-treated HNSCC cell lines demonstrated a significant decrease in E-cadherin mRNA and an increase in the mRNA expression of the transcriptional repressor ZEB1. IL-1beta exposure led to enhanced ZEB1 binding at the chromatin level, as determined by chromatin immunoprecipitation assays (ChIP). An inverse relationship between ZEB1 and E-cadherin was demonstrated in situ by immunohistochemical staining of human HNSCC tissue sections. CONCLUSIONS: Our recent investigations indicate that inflammatory mediators are potent regulators of EMT in HNSCC. This is the first report indicating the role of ZEB1 in the inflammation-induced promotion of EMT in HNSCC. This newly defined pathway for transcriptional regulation of E-cadherin in HNSCC has important implications for targeted chemoprevention and therapy.


Asunto(s)
Carcinoma de Células Escamosas/patología , Células Epiteliales/patología , Neoplasias de Cabeza y Cuello/patología , Inflamación/patología , Mesodermo/patología , Factores de Transcripción/fisiología , Cadherinas/análisis , Línea Celular Tumoral , Ciclooxigenasa 2/análisis , Humanos , Inmunohistoquímica , Inmunoprecipitación , Interleucina-1beta/farmacología , ARN/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Clin Cancer Res ; 15(22): 6820-9, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19887480

RESUMEN

PURPOSE: As a transcriptional repressor of E-cadherin, Snail has predominantly been associated with epithelial-mesenchymal transition, invasion, and metastasis. However, other important Snail-dependent malignant phenotypes have not been fully explored. Here, we investigate the contributions of Snail to the progression of non-small cell lung cancer (NSCLC). EXPERIMENTAL DESIGN: Immunohistochemistry was done to quantify and localize Snail in human lung cancer tissues, and tissue microarray analysis was used to correlate these findings with survival. NSCLC cell lines gene-modified to stably overexpress Snail were evaluated in vivo in two severe combined immunodeficiency murine tumor models. Differential gene expression between Snail-overexpressing and control cell lines was evaluated using gene expression microarray analysis. RESULTS: Snail is upregulated in human NSCLC tissue, and high levels of Snail expression correlate with decreased survival (P < 0.026). In a heterotopic model, mice bearing Snail-overexpressing tumors developed increased primary tumor burden (P = 0.008). In an orthotopic model, mice bearing Snail-overexpressing tumors also showed a trend toward increased metastases. In addition, Snail overexpression led to increased angiogenesis in primary tumors as measured by MECA-32 (P < 0.05) positivity and CXCL8 (P = 0.002) and CXCL5 (P = 0.0003) concentrations in tumor homogenates. Demonstrating the importance of these proangiogenic chemokines, the Snail-mediated increase in tumor burden was abrogated with CXCR2 blockade. Gene expression analysis also revealed Snail-associated differential gene expression with the potential to affect angiogenesis and diverse aspects of lung cancer progression. CONCLUSION: Snail upregulation plays a role in human NSCLC by promoting tumor progression mediated by CXCR2 ligands.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Receptores de Interleucina-8B/metabolismo , Factores de Transcripción/metabolismo , Animales , Cadherinas/metabolismo , Progresión de la Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica/métodos , Ligandos , Ratones , Ratones SCID , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción de la Familia Snail , Transcripción Genética
4.
Clin Cancer Res ; 15(19): 6018-27, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19789323

RESUMEN

PURPOSE: Inflammatory cytokines have been implicated in the progression of head and neck squamous cell carcinoma (HNSCC). Herein we investigate the mechanisms by which interleukin-1beta (IL-1beta) might contribute to Epithelial-Mesenchymal Transition (EMT) in HNSCC. EXPERIMENTAL DESIGN: We evaluated the effect of IL-1beta on the molecular events of EMT in surgical specimens and HNSCC cell lines. We examined the correlation with tumor histologic features, and a SCID xenograft model was used to assess the effects of Snail overexpression. RESULTS: Cyclooxygenase-2 (COX-2)-dependent pathways contribute to the modulation of E-cadherin expression in HNSCC. An inverse relationship between COX-2 and E-cadherin was shown in situ by double immunohistochemical staining of human HNSCC tissue sections. Treatment of HNSCC cells with IL-1beta caused the downregulation of E-cadherin expression and upregulation of COX-2 expression. This effect was blocked in the presence of COX-2 small hairpin RNA. IL-1beta-treated HNSCC cell lines showed a significant decrease in E-cadherin mRNA and an increase in the mRNA expression of the transcriptional repressor Snail. IL-1beta exposure led to enhanced Snail binding at the chromatin level. Small hairpin RNA-mediated knockdown of Snail interrupted the capacity of IL-1beta to downregulate E-cadherin. In a SCID xenograft model, HNSCC Snail-overexpressing cells showed significantly increased primary and metastatic tumor burdens. CONCLUSIONS: IL-1beta modulates Snail and thereby regulates COX-2-dependent E-cadherin expression in HNSCC. This is the first report indicating the role of Snail in the inflammation-induced promotion of EMT in HNSCC. This newly defined pathway for transcriptional regulation of E-cadherin in HNSCC has important implications for targeted chemoprevention and therapy.


Asunto(s)
Carcinoma de Células Escamosas/genética , Neoplasias de Cabeza y Cuello/genética , Mediadores de Inflamación/farmacología , Interleucina-1beta/farmacología , Factores de Transcripción/genética , Animales , Cadherinas/genética , Carcinoma de Células Escamosas/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Células Cultivadas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/patología , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Regiones Promotoras Genéticas/efectos de los fármacos , Factores de Transcripción de la Familia Snail , Regulación hacia Arriba/efectos de los fármacos
5.
J Thorac Oncol ; 3(2): 107-10, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18303428

RESUMEN

Inflammation is an important contributor to lung tumor development and progression. In addition, inflammatory signaling may promote epithelial to mesenchymal transition, development of aggressive metastatic tumor phenotypes, and play a role in resistance to targeted therapies. New insights in inflammatory signaling have led to the evaluation of combination therapies that target these specific pathways. In addition to developing the optimal combination of targeted agents, biomarker-based selection of patients who will likely benefit will be critical to the success of this strategy. Here we focus on the potential contribution of inflammatory mediator-induced resistance to epidermal growth factor receptor tyrosine kinase inhibitors.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Receptores ErbB/antagonistas & inhibidores , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Transformación Celular Neoplásica , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Receptores ErbB/metabolismo , Humanos , Inflamación , Neoplasias Pulmonares/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Receptores Acoplados a Proteínas G/metabolismo
6.
J Thorac Oncol ; 3(2): 117-24, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18303430

RESUMEN

INTRODUCTION: Cyclooxygenase-2 overexpression may mediate resistance to epidermal growth factor receptor tyrosine kinase inhibition through prostaglandin E2-dependent promotion of epithelial to mesenchymal transition (EMT). Suppression of epithelial markers, such as E-cadherin, can lead to resistance to erlotinib. Prostaglandin E2 down-regulates E-cadherin expression by up-regulating transcriptional repressors, including ZEB1 and Snail. Furthermore, E-cadherin can be modulated by matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs), promoting tumor invasion and metastasis. Markers of EMT and tumor invasion were evaluated in patient serum from a phase I clinical trial investigating the combination of celecoxib and erlotinib in non-small cell lung cancer (NSCLC) patients. METHODS: Samples from 22 subjects were evaluated. Soluble E-cadherin (sEC) was evaluated by enzyme linked immunosorbent assay in patient serum at baseline, week 4, and week 8 of treatment. Other markers of EMT and angiogenesis were evaluated by enzyme linked immunosorbent assay, including MMP-9, TIMP-1, and CCL15. RESULTS: Serum sEC, MMP-9, TIMP-1, and CCL15 levels were determined at baseline and week 8. Patients with a partial response to therapy had a significant decrease in sEC, TIMP-1, and CCL15 at week 8. In patients who responded to the combination therapy, baseline MMP-9 was significantly lower compared with nonresponders (p = 0.006). CONCLUSIONS: sEC, MMP-9, TIMP-1, and CCL15 levels correlate with response to combination therapy with erlotinib and celecoxib in patients with NSCLC. A randomized phase II trial is planned comparing erlotinib and celecoxib with erlotinib plus placebo in advanced NSCLC. This study will prospectively assess these and other biomarkers in serum and tumor tissue.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Biomarcadores de Tumor/sangre , Cadherinas/sangre , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Neoplasias Pulmonares/tratamiento farmacológico , Metaloproteinasa 9 de la Matriz/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Celecoxib , Quimiocinas CC/sangre , Inhibidores de la Ciclooxigenasa/farmacología , Relación Dosis-Respuesta a Droga , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib , Humanos , Proteínas Inflamatorias de Macrófagos/sangre , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/administración & dosificación , Pirazoles/farmacología , Quinazolinas/administración & dosificación , Quinazolinas/farmacología , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Inhibidor Tisular de Metaloproteinasa-1/sangre
7.
Expert Rev Anticancer Ther ; 7(10): 1405-21, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17944566

RESUMEN

Lung carcinogenesis is a complex process requiring the acquisition of genetic mutations that confer the malignant phenotype as well as epigenetic alterations that may be manipulated in the course of therapy. Inflammatory signals in the lung cancer microenvironment can promote apoptosis resistance, proliferation, invasion, metastasis, and secretion of proangiogenic and immunosuppressive factors. Here, we discuss several prototypical inflammatory mediators controlling the malignant phenotype in lung cancer. Investigation into the detailed molecular mechanisms underlying the tumor-promoting effects of inflammation in lung cancer has revealed novel potential drug targets. Cytokines, growth factors and small-molecule inflammatory mediators released in the developing tumor microenvironment pave the way for epithelial-mesenchymal transition, the shift from a polarized, epithelial phenotype to a highly motile mesenchymal phenotype that becomes dysregulated during tumor invasion. Inflammatory mediators within the tumor microenvironment are derived from neoplastic cells as well as stromal and inflammatory cells; thus, lung cancer develops in a host environment in which the deregulated inflammatory response promotes tumor progression. Inflammation-related metabolic and catabolic enzymes (prostaglandin E(2) synthase, prostaglandin I(2) synthase and 15-hydroxyprostaglandin dehydrogenase), cell-surface receptors (E-type prostaglandin receptors) and transcription factors (ZEB1, SNAIL, PPARs, STATs and NF-kappaB) are differentially expressed in lung cancer cells compared with normal lung epithelial cells and, thus, may contribute to tumor initiation and progression. These newly discovered molecular mechanisms in the pathogenesis of lung cancer provide novel opportunities for targeted therapy and prevention in lung cancer.


Asunto(s)
Cocarcinogénesis , Inflamación/complicaciones , Neoplasias Pulmonares/prevención & control , Animales , Anticarcinógenos/administración & dosificación , Anticarcinógenos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinógenos Ambientales/efectos adversos , Diferenciación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Enfermedad Crónica , Ciclooxigenasa 2/fisiología , Inhibidores de la Ciclooxigenasa 2/administración & dosificación , Inhibidores de la Ciclooxigenasa 2/uso terapéutico , Citocinas/fisiología , Dinoprostona/metabolismo , Epigénesis Genética , Humanos , Inflamación/inducido químicamente , Inflamación/prevención & control , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/terapia , Ratones , Ensayos Clínicos Controlados Aleatorios como Asunto , Fumar/efectos adversos , Escape del Tumor
8.
Cancer Res ; 66(10): 5338-45, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707460

RESUMEN

Elevated tumor cyclooxygenase-2 (COX-2) expression is associated with tumor invasion, metastasis, and poor prognosis in non-small cell lung cancer (NSCLC). Here, we report that COX-2-dependent pathways contribute to the modulation of E-cadherin expression in NSCLC. First, whereas genetically modified COX-2-sense (COX-2-S) NSCLC cells expressed low E-cadherin and showed diminished capacity for cellular aggregation, genetic or pharmacologic inhibition of tumor COX-2 led to increased E-cadherin expression and resulted in augmented homotypic cellular aggregation among NSCLC cells in vitro. An inverse relationship between COX-2 and E-cadherin was shown in situ by double immunohistochemical staining of human lung adenocarcinoma tissue sections. Second, treatment of NSCLC cells with exogenous prostaglandin E(2) (PGE(2)) significantly decreased the expression of E-cadherin, whereas treatment of COX-2-S cells with celecoxib (1 mumol/L) led to increased E-cadherin expression. Third, the transcriptional suppressors of E-cadherin, ZEB1 and Snail, were up-regulated in COX-2-S cells or PGE(2)-treated NSCLC cells but decreased in COX-2-antisense cells. PGE(2) exposure led to enhanced ZEB1 and Snail binding at the chromatin level as determined by chromatin immunoprecipitation assays. Small interfering RNA-mediated knockdown of ZEB1 or Snail interrupted the capacity of PGE(2) to down-regulate E-cadherin. Fourth, an inverse relationship between E-cadherin and ZEB1 and a direct relationship between COX-2 and ZEB1 were shown by immunohistochemical staining of human lung adenocarcinoma tissue sections. These findings indicate that PGE(2), in autocrine or paracrine fashion, modulates transcriptional repressors of E-cadherin and thereby regulates COX-2-dependent E-cadherin expression in NSCLC. Thus, blocking PGE(2) production or activity may contribute to both prevention and treatment of NSCLC.


Asunto(s)
Cadherinas/biosíntesis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/farmacología , Proteínas de Homeodominio/biosíntesis , Neoplasias Pulmonares/metabolismo , Factores de Transcripción/biosíntesis , Cadherinas/genética , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Agregación Celular/fisiología , Línea Celular Tumoral , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/genética , Elementos E-Box , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Regiones Promotoras Genéticas , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Regulación hacia Arriba , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
9.
Clin Cancer Res ; 11(21): 7674-82, 2005 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16278387

RESUMEN

PURPOSE: Cyclooxygenase-2 (COX-2), phosphorylated signal transducers and activators of transcription 3 (STAT3), and interleukin-6 (IL-6) are elevated in non-small cell lung cancer (NSCLC). These molecules affect numerous cellular pathways, including angiogenesis and apoptosis resistance, and, therefore, may act in concert in NSCLC. EXPERIMENTAL DESIGN: We examined IL-6 and phosphorylated STAT3 in COX-2-overexpressing [COX-2 sense-oriented (COX-2-S)] NSCLC cells and control cells. The effect of IL-6, STAT3, phosphatidylinositol 3-kinase, and mitogen-activated protein/extracellular signal-regulated kinase kinase on vascular endothelial growth factor (VEGF) production and apoptosis resistance was assessed in COX-2-overexpresing cells. RESULTS: We report that NSCLC cells overexpressing COX-2 (COX-2-S) have increased IL-6 and phosphorylated STAT3 expression compared with control cells. IL-6 induced expression of VEGF in NSCLC cells. Moreover, blocking IL-6, mitogen-activated protein/extracellular signal-regulated kinase kinase, or phosphatidylinositol 3-kinase decreased VEGF production in COX-2-S cells. The addition of IL-6 to NSCLC cells resulted in increased apoptosis resistance. Furthermore, the inhibition of STAT3 or IL-6 induced apoptosis and reduced survivin expression, a member of the inhibitor of apoptosis protein family in COX-2-S cells. CONCLUSIONS: Overall, these findings suggest a novel pathway in which COX-2 activates STAT3 by inducing IL-6 expression. This pathway could contribute to tumor formation by promoting survivin-dependent apoptosis resistance and VEGF production. These findings provide a rationale for the future development of STAT3, IL-6, and/or COX-2-targeted therapies for the treatment of lung cancer.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Ciclooxigenasa 2/biosíntesis , Ciclooxigenasa 2/fisiología , Regulación Neoplásica de la Expresión Génica , Interleucina-6/metabolismo , Neoplasias Pulmonares/metabolismo , Factor de Transcripción STAT3/metabolismo , Secuencias de Aminoácidos , Apoptosis , Western Blotting , Diferenciación Celular , Línea Celular Tumoral , Supervivencia Celular , Citocinas/metabolismo , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Neovascularización Patológica , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , ARN/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
J Immunol ; 175(3): 1483-90, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16034085

RESUMEN

Naturally occurring CD4+CD25+ regulatory T cells (T reg) are pivotal in suppressing immune responses and maintaining tolerance. The identification of molecules controlling T reg differentiation and function is important in understanding host immune responses in malignancy and autoimmunity. In this study we show that PGE2 enhances the in vitro inhibitory function of human purified CD4+CD25+ T reg cells. Moreover, PGE2 induces a regulatory phenotype in CD4+CD25- T cells. PGE2-treated T cell-mediated inhibition of anti-CD3-stimulated lymphocyte proliferation did not require cell contact. Phenotypic analysis revealed that PGE2 diminished CD25 expression in both CD4+CD25dim T cells and CD4+CD25bright T reg cells. PGE2 exposure induced the T reg cell-specific transcription factor forkhead/winged helix transcription factor gene (FOXP3) in CD4+CD25- T cells and significantly up-regulated its expression in CD4+CD25+ T reg cells. Similarly, 24-h incubation with supernatants from cyclooxygenase-2-overexpressing lung cancer cells that secrete high levels of PGE2 significantly induced FOXP3 in CD4+CD25- T cells. Finally, PGE2 up-regulated FOXP3 at both mRNA and protein levels and enhanced FOXP3 promoter activity. This is the first report indicating that PGE2 can modulate FOXP3 expression and T reg function in human lymphocytes.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Dinoprostona/farmacología , Regulación de la Expresión Génica/inmunología , Receptores de Interleucina-2/biosíntesis , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Adyuvantes Inmunológicos/farmacología , Línea Celular Tumoral , Factores de Transcripción Forkhead , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunosupresores/farmacología , Células Jurkat , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/inmunología , ARN Mensajero/biosíntesis , Linfocitos T Reguladores/efectos de los fármacos , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
11.
Cancer Res ; 65(14): 6275-81, 2005 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16024629

RESUMEN

Cyclooxygenase 2 (COX-2) overexpression is found in a wide variety of human cancers and is linked to all stages of tumorigenesis. Elevated tumor COX-2 expression is associated with increased angiogenesis, tumor invasion, suppression of host immunity and promotes tumor cell resistance to apoptosis. Previous reports have linked the COX-2 product prostaglandin E2 (PGE2) to the abnormal activation of the mitogen-activated protein kinase/Erk kinase pathway. Here we show that PGE2 is able to rapidly stimulate Erk phosphorylation in a subset of non-small cell lung cancer (NSCLC) cell lines. This effect is not evident in bronchial epithelial cells. In contrast to previous reports in colon cancer, we found that Erk activation as well as cellular proliferation induced by PGE2 was not inhibited by pretreatment of the cells with epidermal growth factor receptor (EGFR) inhibitors. Activation of the Erk pathway by PGE2 was also resistant to src kinase inhibitors but sensitive to the protein kinase C inhibition. PGE2 effects are mediated through four G protein-coupled receptors. Selective inhibition of EP receptors revealed the possible involvement of Ca2+-dependent signaling in PGE2-mediated activation of Erk. Our data indicate the presence of an EGFR-independent activation of the mitogen-activated protein kinase/Erk pathway by PGE2 in NSCLC cells. These findings provide evidence for the possible link between tumor COX-2 overexpression and elevated Erk-mediated cancer cell proliferation and migration. Importantly, these findings suggest that COX-2 overexpression may contribute to EGFR inhibitor resistance in NSCLC.


Asunto(s)
Dinoprostona/farmacología , Receptores ErbB/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Células Escamosas/enzimología , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , AMP Cíclico/metabolismo , Ciclooxigenasa 2 , Activación Enzimática , Receptores ErbB/antagonistas & inhibidores , Humanos , Neoplasias Pulmonares/enzimología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas de la Membrana , Fosforilación/efectos de los fármacos , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores
12.
J Leukoc Biol ; 78(2): 555-64, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15908458

RESUMEN

Control of apoptosis is fundamental for dendritic cell (DC) homeostasis. Numerous factors maintain DC viability throughout their lifespan, including inhibitor of apoptosis proteins. Among them, survivin is overexpressed in many human malignancies, but its physiological function in normal cells has not been fully delineated. Prostaglandin E2 (PGE2), also overproduced in several malignancies, has shown to induce proapoptotic and antiapoptotic effects in different cell types, including immune cells. In DC, PGE2 predominantly affects maturation and modulates immune functions. Here, we show that exposure of monocyte-derived DC to PGE2 (10(-5) M) for 72 h significantly increased DC survivin mRNA and protein expression. In contrast, DC, matured with lipopolysaccharide or tumor necrosis factor alpha, did not reveal survivin induction in response to PGE2. Following exposure to apoptotic stimuli, DC treated with PGE2 exhibited an overall increased viability compared with control DC, and this effect was correlated inversely with caspase-3 activation. Moreover, PGE2-treated, survivin-deficient DC demonstrated reduced viability in response to apoptotic stimuli. Further analysis indicated that PGE2 induced DC survivin expression in an E prostanoid (EP)2/EP4 receptor and phosphatidylinositol-3 kinase-dependent manner. These findings suggest that PGE2-dependent regulation of survivin is important in modulating apoptosis resistance in human DC.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/metabolismo , Dinoprostona/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas de Neoplasias/metabolismo , Apoptosis/efectos de la radiación , Caspasa 3 , Caspasas/metabolismo , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Dinoprostona/metabolismo , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Regulación de la Expresión Génica/fisiología , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/fisiología , Proteínas Inhibidoras de la Apoptosis , Lipopolisacáridos/farmacología , Monocitos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , ARN Mensajero/biosíntesis , Receptores de Prostaglandina E/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E , Subtipo EP4 de Receptores de Prostaglandina E , Survivin , Factor de Necrosis Tumoral alfa/farmacología
13.
J Immunol ; 173(9): 5458-66, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15494493

RESUMEN

Dendritic cell (DC) migration is crucial for the initiation of immune responses. The balance between metalloproteinases (MMP) and tissue inhibitors of metalloproteinases (TIMP) has been shown to modulate DC migration. PGE2, which is overproduced in a wide variety of human malignancies, has been implicated in MMP and TIMP regulation in various cells, including monocytes. In the present study, we hypothesized that tumor-derived PGE2 would affect DC migratory capacity through the extracellular matrix (ECM) by altering MMP and TIMP balance. Treatment of monocyte-derived immature DC with exogenous PGE2 induced TIMP-1 secretion but not MMP-9 production and was correlated with reduced DC migration through ECM. Because recombinant TIMP-1 replicated PGE2 inhibition of DC migration while anti-TIMP-1 neutralizing Ab reversed it, we conclude that PGE2-mediated induction of TIMP-1 was responsible for the reduced migration of PGE2-treated DC. Similarly, DC cultured for 48 h in supernatants from cyclooxygenase-2 overexpressing lung cancer cells that secrete high levels of PGE2, exhibited decreased migration through ECM. Finally, analysis of E prostanoid receptor expression and their selective inhibition revealed that the enhanced TIMP-1 secretion in PGE2-treated DC was mediated predominantly by the E prostanoid receptor 2. These findings indicate that PGE2-dependent enhancement of TIMP-1 production causes reduced migration of DC through ECM.


Asunto(s)
Adyuvantes Inmunológicos/fisiología , Inhibición de Migración Celular , Células Dendríticas/citología , Células Dendríticas/enzimología , Dinoprostona/fisiología , Matriz Extracelular/enzimología , Matriz Extracelular/inmunología , Inhibidor Tisular de Metaloproteinasa-1/biosíntesis , 16,16-Dimetilprostaglandina E2/metabolismo , 16,16-Dimetilprostaglandina E2/farmacología , Adyuvantes Inmunológicos/metabolismo , Adyuvantes Inmunológicos/farmacología , Diferenciación Celular/inmunología , Línea Celular Tumoral , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Dinoprostona/metabolismo , Relación Dosis-Respuesta Inmunológica , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Receptores CCR7 , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/biosíntesis , Receptores de Prostaglandina E/fisiología , Subtipo EP2 de Receptores de Prostaglandina E , Transducción de Señal/inmunología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Transfección , Regulación hacia Arriba/inmunología
14.
Cancer Res ; 64(18): 6377-80, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15374942

RESUMEN

The Coxsackie Adenovirus Receptor (CAR) has primarily been studied in its role as the initial cell surface attachment receptor for Coxsackie and group C adenoviruses. Recent reports suggest that CAR mediates homotypic intercellular adhesion as part of the tight and/or adherens junction. Thus, CAR is well positioned to participate in intercellular interactions and signaling. Using an antisense (AS)-CAR plasmid vector, we silenced surface CAR expression in lung cancer cells that possessed a high basal expression of this molecule and monitored the resultant tumorigenesis. AS-CAR transfectants exhibit a profound loss in the ability to generate xenografts in scid/scid mice. The emergence of delayed-onset tumors in animals that received injection with AS-CAR transfectants correlates with the resurfacing of CAR expression, suggesting that such expression and tumor emergence are temporally related. To study the mechanism underlying the differences in tumorigenicity, control and AS-CAR cells were compared in terms of their in vitro growth potential. Whereas only subtle differences in the proliferative capacity of the two populations were evident when assayed with growth on plastic, significant differences became apparent when one compared the relative ability of these populations to form colonies in soft agar. These data indicate that silencing surface CAR expression abrogates xenograft tumorigenesis in vivo and colony formation in vitro and invoke the novel possibility that CAR expression is needed for the efficient formation of tumors by a subset of lung cancer cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores Virales/fisiología , Adenoviridae , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , ADN sin Sentido , Enterovirus , Femenino , Humanos , Neoplasias Pulmonares/genética , Ratones , Ratones SCID , Receptores Virales/antagonistas & inhibidores , Receptores Virales/biosíntesis , Receptores Virales/genética , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cancer Res ; 64(18): 6549-55, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15374967

RESUMEN

Constitutive overexpression of cyclooxygenase-2 (COX-2) occurs frequently in several different malignancies, including lung, colon, breast, and prostate cancer. Clinical studies have established elevated serum insulin-like growth factor (IGF-I) content and IGF-I:IGF-binding protein 3 (IGFBP-3) ratio as risk factors for these same malignancies. Therefore, we sought to determine the link between COX-2 expression and the IGF axis in COX-2 gene-modified human non-small-cell lung cancer (NSCLC) cells. Overexpression of COX-2 in NSCLC cells enhanced the antiapoptotic and mitogenic effects of IGF-I and IGF-II, facilitated the autophosphorylation of the type 1 IGF receptor, increased class IA phosphatidylinositol 3'-kinase activity, and decreased expression of IGFBP-3. Thus, these findings show that COX-2 augments the stimulatory arm of the IGF axis.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Factor II del Crecimiento Similar a la Insulina/fisiología , Factor I del Crecimiento Similar a la Insulina/fisiología , Isoenzimas/fisiología , Neoplasias Pulmonares/enzimología , Prostaglandina-Endoperóxido Sintasas/fisiología , Receptor IGF Tipo 1/metabolismo , Apoptosis/fisiología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Celecoxib , División Celular/fisiología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Ciclooxigenasa 2 , ADN sin Sentido/genética , Regulación hacia Abajo/efectos de los fármacos , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/biosíntesis , Factor I del Crecimiento Similar a la Insulina/farmacología , Factor II del Crecimiento Similar a la Insulina/farmacología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/biosíntesis , Isoenzimas/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas de la Membrana , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Prostaglandina-Endoperóxido Sintasas/biosíntesis , Prostaglandina-Endoperóxido Sintasas/genética , Pirazoles , Transducción de Señal/fisiología , Sulfonamidas/farmacología
16.
Drug Resist Updat ; 7(3): 169-84, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15296859

RESUMEN

Lung cancer is the leading cause of cancer death in the United States. Although the low 5-year survival rate (under 15%) has changed minimally in the last 25 years, new agents and combinations of agents that target tumor proliferation, invasion, and survival may lead to improvement in patient outcomes. There is evidence that cyclooxygenase-2 (COX-2) is overexpressed in lung cancer and promotes tumor proliferation, invasion, angiogenesis, and resistance to apoptosis. COX-2 inhibitors have been found to inhibit tumor growth in animal models and have demonstrated responses when combined with conventional therapy in phase II clinical trials. Further understanding of the mechanisms involved in COX-2-mediated tumorigenesis and its interaction with other molecules in lung cancer may lead to improved therapeutic strategies for this disease. In addition, delineation of how COX-2-dependent genes modulate the malignant phenotype will provide novel insights in lung cancer pathogenesis.


Asunto(s)
Isoenzimas/fisiología , Neoplasias Pulmonares/etiología , Prostaglandina-Endoperóxido Sintasas/fisiología , Células Presentadoras de Antígenos/fisiología , Apoptosis , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/uso terapéutico , Citocinas/fisiología , Receptores ErbB/antagonistas & inhibidores , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/prevención & control , Proteínas de la Membrana , Neovascularización Patológica/etiología
18.
Cancer Res ; 64(5): 1853-60, 2004 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-14996749

RESUMEN

Elevated tumor cyclooxygenase (COX)-2 activity plays a multifaceted role in non-small cell lung cancer (NSCLC). To elucidate the role of COX-2 in the in vitro and in vivo expression of two known NSCLC angiogenic peptides, CXC ligand (CXCL) 8 and CXCL5, we studied two COX-2 gene-modified NSCLC cell lines, A549 and H157. COX-2 overexpression enhanced the in vitro expression of both CXCL8 and CXCL5. In contrast, specific COX-2 inhibition decreased the production of both peptides as well as nuclear translocation of nuclear factor kappaB. In a severe combined immunodeficient mouse model of human NSCLC, the enhanced tumor growth of COX-2-overexpressing tumors was inhibited by neutralizing anti-CXCL5 and anti-CXCL8 antisera. We conclude that COX-2 contributes to the progression of NSCLC tumorigenesis by enhancing the expression of angiogenic chemokines CXCL8 and CXCL5.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/irrigación sanguínea , Quimiocinas CXC/análisis , Péptidos y Proteínas de Señalización Intercelular/análisis , Isoenzimas/fisiología , Neoplasias Pulmonares/irrigación sanguínea , Prostaglandina-Endoperóxido Sintasas/fisiología , Transporte Activo de Núcleo Celular , Animales , Carcinoma de Pulmón de Células no Pequeñas/química , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Quimiocina CXCL5 , Ciclooxigenasa 2 , Dinoprostona/fisiología , Humanos , Isoenzimas/antagonistas & inhibidores , Neoplasias Pulmonares/química , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas de la Membrana , Ratones , Ratones SCID , FN-kappa B/metabolismo , Neovascularización Patológica/etiología
19.
FASEB J ; 18(1): 206-8, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14597555

RESUMEN

Elevated tumor cyclooxygenase 2 (COX-2) expression is associated with increased angiogenesis, tumor invasion and promotion of tumor cell resistance to apoptosis. The mechanism(s) by which COX-2 exerts its cytoprotective effects are not completely understood but may be due to an imbalance of pro- and anti-apoptotic gene expression. To analyze COX-2-dependent gene expression and apoptosis, we created cell lines constitutively expressing COX-2 cDNA in sense and antisense orientations. Whereas COX-2 sense cells have significantly heightened resistance to radiation and drug-induced apoptosis, COX-2 antisense cells are highly sensitive to apoptosis induction. We found that the expression of the anti-apoptotic protein survivin correlated positively with COX-2 expression. A COX-2-dependent modulation of survivin ubiquitination led to its stabilization in COX-2 overexpressing cells, and this effect was replicated by exogenous PGE2 treatment of parental tumor cells. In contrast to previous studies in other cell types, in nonsmall cell lung cancer cells survivin was expressed in a cell cycle-independent manner. When established in SCID mice in vivo, COX-2 antisense-derived tumors had significantly decreased survivin levels while COX-2 sense-derived tumors demonstrated elevated levels compared with controls. In accord with these findings, survivin and COX-2 were frequently upregulated and co-expressed in human lung cancers in situ.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Isoenzimas/metabolismo , Neoplasias Pulmonares/enzimología , Proteínas Asociadas a Microtúbulos/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Animales , Elementos sin Sentido (Genética) , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Ciclo Celular , Línea Celular Tumoral , Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa 2 , Inhibidores de la Ciclooxigenasa/farmacología , Dinoprostona/farmacología , Humanos , Proteínas Inhibidoras de la Apoptosis , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteínas de la Membrana , Ratones , Proteínas de Neoplasias , Prostaglandina-Endoperóxido Sintasas/genética , Survivin , Ubiquitinas/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...