Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 111
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cancer ; 11: 4, 2012 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-22239913

RESUMEN

BACKGROUND: Although cigarette smoking is the principal cause of lung carcinogenesis, chronic obstructive pulmonary disease (COPD), an inflammatory disease of the lung, has been identified as an independent risk factor for lung cancer. Bacterial colonization, particularly with non-typeable Haemophilus influenzae (NTHi), has been implicated as a cause of airway inflammation in COPD besides cigarette smoke. Accordingly, we hypothesized that lung cancer promotion may occur in a chronic inflammatory environment in the absence of concurrent carcinogen exposure. RESULTS: Herein, we investigated the effects of bacterial-induced COPD-like inflammation and tobacco carcinogen-enhanced tumorigenesis/inflammation in the retinoic acid inducible G protein coupled receptor knock out mouse model (Gprc5a-/- mouse) characterized by late-onset, low multiplicity tumor formation. Three-month-old Gprc5a-/- mice received 4 intraperitoneal injections of the tobacco-specific carcinogen, NNK, followed by weekly exposure to aerosolized NTHi lysate for 6 months. The numbers of inflammatory cells in the lungs and levels of several inflammatory mediators were increased in Gprc5a-/- mice treated with NTHi alone, and even more so in mice pretreated with NNK followed by NTHi. The incidence of spontaneous lung lesions in the Gprc5a-/- mice was low, but NTHi exposure led to enhanced development of hyperplastic lesions. Gprc5a-/- mice exposed to NNK alone developed multiple lung tumors, while NTHi exposure increased the number of hyperplastic foci 6-fold and the tumor multiplicity 2-fold. This was associated with increased microvessel density and HIF-1α expression. CONCLUSION: We conclude that chronic extrinsic lung inflammation induced by bacteria alone or in combination with NNK enhances lung tumorigenesis in Gprc5a-/- mice.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Pulmonares/etiología , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Receptores Acoplados a Proteínas G/genética , Animales , Transformación Celular Neoplásica/genética , Modelos Animales de Enfermedad , Infecciones por Haemophilus/complicaciones , Infecciones por Haemophilus/microbiología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/inducido químicamente , Neovascularización Patológica/microbiología , Nitrosaminas , Enfermedad Pulmonar Obstructiva Crónica/microbiología
2.
Gastroenterology ; 138(3): 1035-45.e1-2, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19818782

RESUMEN

BACKGROUND & AIMS: Galectin-3 is a beta-galactoside-binding protein that increases gastric cancer cell motility in response to integrin signaling and is highly expressed in gastric tumor cells. Galectin-3 induces cytoskeletal remodeling to increase cell motility, but the mechanisms of this process are not understood. We investigated the effects of galectin-3 on fascin-1, an actin-bundling protein. METHODS: We collected malignant and normal tissues from gastric cancer patients and examined the expression levels of galectin-3 and fascin-1. We silenced galectin-3 expression in human gastric cancer cell lines using small interfering RNA and lenti-viral constructs and determined the effects on fascin-1 expression, cell motility, and invasion. RESULTS: Malignant gastric tissues expressed high levels of galectin-3 and fascin-1, compared with normal gastric tissues. Silencing of galectin-3 resulted in altered cancer cell morphology, reduced fascin-1 expression, decreased cell motility, and reduced malignant cell invasion. Galectin-3 overexpression reversed these effects. Silencing of fascin-1 also reduced cell motility and caused changes in cell shape, as did silencing of galectin-3. Furthermore, galectin-3 silencing inhibited the interaction between glycogen synthase kinase (GSK)-3beta, beta-catenin, and T-cell factor (TCF) 4, and the binding of beta-catenin/TCF-4 to the fascin-1 promoter. Nuclear localization of GSK-3beta and beta-catenin were not detected when galectin-3 was silenced. Overexpression of mutated galectin-3 (with mutations in the GSK-3beta binding and phosphorylation motifs) did not increase fascin-1 levels, in contrast to overexpression of wild-type galectin-3. CONCLUSIONS: Galectin-3 increases cell motility by up-regulating fascin-1 expression. Galectin-3 might be a potential therapeutic target for the prevention and treatment of gastric cancer progression.


Asunto(s)
Proteínas Portadoras/metabolismo , Movimiento Celular , Galectina 3/metabolismo , Proteínas de Microfilamentos/metabolismo , Neoplasias Gástricas/metabolismo , Sitios de Unión , Proteínas Portadoras/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Forma de la Célula , Galectina 3/genética , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas de Microfilamentos/genética , Mutación , Invasividad Neoplásica , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/secundario , Factores de Transcripción TCF/metabolismo , Proteína 2 Similar al Factor de Transcripción 7 , Transfección , Regulación hacia Arriba , beta Catenina/metabolismo
3.
Carcinogenesis ; 31(12): 2118-23, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20819778

RESUMEN

Second primary tumor (SPT) and/or recurrence negatively impact the prognosis of patients with curatively treated early-stage head and neck cancer. MicroRNAs (miRNAs) play important roles in cancer development. We explored whether the variations of miRNA-related pathway were associated with the risk of SPT/recurrence in patients with early-stage head and neck cancer. This study includes 150 early-stage head and neck cancer patients with SPT/recurrence and 300 patients without SPT/recurrence. Two hundred and thirty-five tagging and potentially functional single-nucleotide polymorphisms (SNPs) were genotyped from eight miRNA biogenesis pathway genes and 135 miRNA-targeted genes. Eighteen miRNA-related SNPs were significantly associated with the risk of SPT/recurrence. The most significant SNP was rs3747238, a miRNA-binding site SNP in SMC1B. The variant homozygous genotype of this SNP was associated with a 1.74-fold increased risk [95% confidence interval (CI) 1.19-2.54; P = 0.004]. Cumulative effect analysis showed joint effects for the number of unfavorable genotype in patients. Survival tree analysis further identified the high-order gene-gene interactions and categorized the study subjects into low-, medium- and high-risk groups. Patients in the high-risk group had a 4.84-fold increased risk (95% CI: 3.11-7.51; P = 2.45 × 10(-12)) and a shorter event-free median survival time of 37.9 months (log rank P = 2.28 × 10(-13)). Our results suggested that miRNA-related genetic polymorphisms may be used individually and jointly to predict the risk of SPT/recurrence of early-stage head and neck cancer patients.


Asunto(s)
Neoplasias de Cabeza y Cuello/genética , MicroARNs/fisiología , Recurrencia Local de Neoplasia/genética , Neoplasias Primarias Secundarias/genética , Polimorfismo de Nucleótido Simple , Proteínas de Ciclo Celular/genética , Proteínas Cromosómicas no Histona/genética , Variación Genética , Genotipo , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/patología , Humanos , Carioferinas/genética , Recurrencia Local de Neoplasia/etiología , Estadificación de Neoplasias , Neoplasias Primarias Secundarias/etiología , Riesgo
4.
Carcinogenesis ; 31(10): 1755-61, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20627871

RESUMEN

Curatively treated patients with early-stage head and neck squamous cell carcinoma (HNSCC) are at high risks for second primary tumor (SPT) and recurrence. The regulator of G-protein signaling (RGS) is important in essential signaling transduction and cellular activities. We hypothesize that genetic variations of RGS may modulate the risk of SPT/recurrence in patients with early-stage HNSCC. In a nested case-control study, we evaluated 98 single-nucleotide polymorphisms (SNPs) in 17 RGS genes for the risk of SPT/recurrence among 450 HNSCC patients. Eight SNPs showed significant associations with the risk of SPT/recurrence, with the most significant one of rs2179653, which is located in the 5'-flanking region of RGS2 gene. Under a recessive genetic model, the homozygous variant genotype of this SNP was associated with 2.95-fold [95% confidence interval (CI): 1.52-5.74] increased risk of SPT/recurrence. This association remained significant after the adjustment for multiple comparisons. Cumulative effects analysis revealed that the risk increased significantly with the increasing numbers of unfavorable genotypes. Compared with subjects carrying 0-2 unfavorable genotypes, the hazard ratios (95% CIs) for those carrying 3 or 4+ were 1.73 (1.10-2.70) and 3.05 (1.92-4.83), respectively. Furthermore, survival tree analysis revealed potential higher order gene-gene interactions and indicated different outcomes based on distinct genotype profiles. Genetic variations of RGS genes may modulate the susceptibility to SPT/recurrence in early-stage HNSCC patients individually and cumulatively. Our results stressed the importance of taking a polygenic approach to evaluate the cumulative and interaction effects of genetic variations in the prediction of cancer risk and prognosis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Predisposición Genética a la Enfermedad , Neoplasias de Cabeza y Cuello/genética , Recurrencia Local de Neoplasia/genética , Neoplasias Primarias Secundarias/genética , Polimorfismo de Nucleótido Simple , Proteínas RGS/genética , Anciano , Dosificación de Gen , Variación Genética , Humanos , Persona de Mediana Edad , Análisis Multivariante
5.
Mol Cancer Ther ; 8(3): 521-32, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19276160

RESUMEN

The standard treatment for most advanced cancers is multidrug therapy. Unfortunately, combinations in the clinic often do not perform as predicted. Therefore, to complement identifying rational drug combinations based on biological assumptions, we hypothesized that a functional screen of drug combinations, without limits on combination sizes, will aid the identification of effective drug cocktails. Given the myriad possible cocktails and inspired by examples of search algorithms in diverse fields outside of medicine, we developed a novel, efficient search strategy called Medicinal Algorithmic Combinatorial Screen (MACS). Such algorithms work by enriching for the fitness of cocktails, as defined by specific attributes through successive generations. Because assessment of synergy was not feasible, we developed a novel alternative fitness function based on the level of inhibition and the number of drugs. Using a WST-1 assay on the A549 cell line, through MACS, we screened 72 combinations of arbitrary size formed from a 19-drug pool across four generations. Fenretinide, suberoylanilide hydroxamic acid, and bortezomib (FSB) was the fittest. FSB performed up to 4.18 SD above the mean of a random set of cocktails or "too well" to have been found by chance, supporting the utility of the MACS strategy. Validation studies showed FSB was inhibitory in all 7 other NSCLC cell lines tested. It was also synergistic in A549, the one cell line in which this was evaluated. These results suggest that when guided by MACS, screening larger drug combinations may be feasible as a first step in combination drug discovery in a relatively small number of experiments.


Asunto(s)
Algoritmos , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Neoplasias Pulmonares/patología , Peso Molecular , Profármacos/química , Profármacos/uso terapéutico , Células Tumorales Cultivadas
6.
Carcinogenesis ; 30(11): 1949-56, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19793800

RESUMEN

Recent studies have demonstrated that K-ras mutations in lung epithelial cells elicit inflammation that promotes carcinogenesis in mice (intrinsic inflammation). The finding that patients with chronic obstructive pulmonary disease (COPD), an inflammatory disease of the lung, have an increased risk of lung cancer after controlling for smoking suggests a further link between lung cancer and extrinsic inflammation. Besides exposure to cigarette smoke, it is thought that airway inflammation in COPD is caused by bacterial colonization, particularly with non-typeable Hemophilus influenzae (NTHi). Previously, we have shown that NTHi-induced COPD-like airway inflammation promotes lung cancer in an airway conditional K-ras-induced mouse model. To further test the role of inflammation in cancer promotion, we administered the natural anti-inflammatory agent, curcumin, 1% in diet before and during weekly NTHi exposure. This significantly reduced the number of visible lung tumors in the absence of NTHi exposure by 85% and in the presence of NTHi exposures by 53%. Mechanistically, curcumin markedly suppressed NTHi-induced increased levels of the neutrophil chemoattractant keratinocyte-derived chemokine by 80% and neutrophils by 87% in bronchoalveolar lavage fluid. In vitro studies of murine K-ras-induced lung adenocarcinoma cell lines (LKR-10 and LKR-13) indicated direct anti-tumoral effects of curcumin by reducing cell viability, colony formation and inducing apoptosis. We conclude that curcumin suppresses the progression of K-ras-induced lung cancer in mice by inhibiting intrinsic and extrinsic inflammation and by direct anti-tumoral effects. These findings suggest that curcumin could be used to protract the premalignant phase and inhibit lung cancer progression in high-risk COPD patients.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Antineoplásicos/farmacología , Curcumina/farmacología , Haemophilus influenzae , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/prevención & control , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocinas/metabolismo , Curcumina/uso terapéutico , Femenino , Genes ras , Infecciones por Haemophilus/complicaciones , Inflamación , Neoplasias Pulmonares/genética , Masculino , Ratones , Neutrófilos/metabolismo , Neutrófilos/patología , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología
7.
Clin Cancer Res ; 14(19): 6014-22, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829480

RESUMEN

PURPOSE: To identify the patterns of protein expression of basic fibroblast growth factor (bFGF) and FGF receptors 1 and 2 in non-small cell lung carcinoma (NSCLC) and their role in the early pathogenesis of squamous cell carcinoma (SCC) of the lung. EXPERIMENTAL DESIGN: Archived tissue from NSCLC (adenocarcinoma and SCC; n = 321) and adjacent bronchial epithelial specimens (n = 426) were analyzed for the immunohistochemical expression of bFGF, FGFR1, and FGFR2, and the findings were correlated with clinicopathologic features of the patients. RESULTS: High expression of bFGF, FGFR1, and FGFR2 was shown in most NSCLC tumors. The pattern of expression for all markers varied according to tumor histologic type and cellular localization. Cytoplasmic expression scores were significantly higher in tumors than in normal epithelia. Nuclear bFGF (P = 0.03) and FGFR1 (P = 0.02) levels were significantly higher in women than in men. Although cytoplasmic FGFR1 expression was significantly higher (P = 0.002) in ever smokers than in never smokers, nuclear FGFR1 (P = 0.0001) and FGFR2 (P = 0.003) expression was significantly higher in never smokers. Different prognostic patterns for the expression of these markers were detected for both NSCLC histologic types. Dysplastic changes showed significantly higher expression of all markers compared with squamous metaplasia. CONCLUSIONS: bFGF, FGFR1, and FGFR2 are frequently overexpressed in SCC and adenocarcinoma of the lung. bFGF signaling pathway activation may be an early phenomenon in the pathogenesis of SCC and thus an attractive novel target for lung cancer chemopreventive and therapeutic strategies.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Inmunohistoquímica/métodos , Neoplasias Pulmonares/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Células Escamosas/genética , Femenino , Humanos , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad
8.
Cancer Res ; 67(14): 6565-73, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638865

RESUMEN

Squamous cell carcinoma in the lung originates from bronchial epithelial cells that acquire increasingly abnormal phenotypes. Currently, no known biomarkers are clinically efficient for the early detection of premalignant lesions and lung cancer. We sought to identify secreted molecules produced from squamous bronchial epithelial cells cultured with organotypic culture methods. We analyzed protein expression patterns in the apical surface fluid (ASF) from aberrantly differentiated squamous metaplastic normal human tracheobronchial epithelial (NHTBE) and mucous NHTBE cells. Comparative two-dimensional PAGE analysis revealed 174 unique proteins in the ASF of squamous NHTBE cells compared with normal mucociliary differentiated NHTBE cells. Among them, 64 well-separated protein spots were identified by liquid chromatography-tandem mass spectrometry, revealing 22 different proteins in the ASF from squamous NHTBE cells. Expression of six of these proteins [SCC antigen 1 (SCCA1), SCC antigen 2 (SCCA2), S100A8, S100A9, Annexin I, and Annexin II] in the squamous NHTBE cells was further confirmed with immunoblot analysis. Notably, SCCA1 and SCCA2 were verified as being expressed in squamous metaplastic NHTBE cells but not in normal mucous NHTBE or normal bronchial epithelium. Moreover, SCCA1 and SCCA2 expression increased in in vitro lung carcinogenesis model cell lines with increasing malignancy. In summary, we identified proteins that are uniquely secreted from squamous metaplastic primary human bronchial epithelial cells cultured by the organotypic air-liquid interface method. These ASF proteins may be used to detect abnormal lesions in the lung without collecting invasive biopsy specimens.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Biología Molecular/métodos , Proteínas/química , Proteómica/métodos , Neoplasias de la Tráquea/metabolismo , Aire , Antígenos de Neoplasias/biosíntesis , Línea Celular Tumoral , Células Cultivadas , Humanos , Metástasis de la Neoplasia , Análisis por Matrices de Proteínas , Serpinas/biosíntesis , Propiedades de Superficie
9.
Brief Funct Genomic Proteomic ; 7(4): 322-6, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18467351

RESUMEN

High throughput genomic and proteomic methods are often used for comparisons between expression of genes and proteins, respectively in normal cells and malignant counterparts for the identification of potential tumor markers for diagnosis and prognosis. Some experiments use normal and malignant cells cultured in vitro as a source of the mRNA or proteins for analysis. The conditions used for cell culture can exert major effects on the expression of genes and proteins. The interpretation of results of some such studies can be erroneous if normal cells and cancer cells are cultured in serum-free medium (SFM) and serum-supplemented media, respectively as recommended for their optimal growth. The reason for potential complications in the data interpretation is that serum contains different factors that affect gene expression. Likewise, SFM is usually supplemented with specific growth factors as well as bovine pituitary extract. Experimental examples demonstrating the issue include the stimulatory effects of serum on the expression of retinoic acid-inducible genes (e.g. GPRC5A) leading to the potentially erroneous conclusion that such genes are overexpressed in cancer cells. Potential remedy for this problem is to grow the normal and malignant cells in the same medium (serum-free or serum-containing) before analysis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Regulación de la Expresión Génica , Genómica , Humanos , Proteómica , ARN Mensajero/genética , ARN Neoplásico/genética , Valores de Referencia
10.
Clin Cancer Res ; 13(16): 4817-24, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17699860

RESUMEN

PURPOSE: Retinoids inhibit proliferation and induce differentiation in melanoma cells. Retinoic acid receptors (RAR) and retinoid X receptors (RXR) mediate the various modulatory effects of retinoids in cells. We have studied the in situ expression of each RAR and RXR protein (alpha, beta, gamma) in a large series of melanocytic lesions and correlated the expression with clinicopathologic features and prognosis of the patients. EXPERIMENTAL DESIGN: Tissue microarray blocks of 226 melanocytic lesions were semiquantitatively evaluated by immunohistochemistry for the cytoplasmic and nuclear expression of RAR and RXR protein (alpha, beta, gamma). RESULTS: A significant decrease of RARbeta protein (P < 0.0001), nuclear expression of RARgamma (P < 0.0001), and RXRalpha (P < 0.0001) was found in primary and metastatic melanomas as compared with nevi. Loss of nuclear immunoreactivity for RARgamma (P = 0.048) and RXRalpha (P = 0.001) was observed in the lesions showing vertical growth pattern. In addition, in patients with concomitant loss of cytoplasmic staining for RARalpha and RXRalpha, the probability of overall survival (log-rank test, P = 0.002) and disease-specific survival (log-rank test, P = 0.014) was significantly lower. CONCLUSIONS: Aberrant expression of retinoid receptors seems to be a frequent event in melanoma and suggests an impairment of the retinoid pathway in this cancer. Our data indicate the loss of retinoid receptor expression with melanoma progression and suggest a possible prognostic significance of the analysis of retinoid receptors in melanoma.


Asunto(s)
Melanoma/patología , Receptores de Ácido Retinoico/análisis , Receptor alfa X Retinoide/análisis , Progresión de la Enfermedad , Humanos , Melanoma/química , Melanoma/mortalidad , Melanoma/secundario , Nevo/patología , Pronóstico , Modelos de Riesgos Proporcionales , Análisis de Matrices Tisulares
11.
Cancer Res ; 66(23): 11194-206, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145864

RESUMEN

The identification of proteins, which exhibit different levels in normal, premalignant, and malignant lung cells, could improve early diagnosis and intervention. We compared the levels of proteins in normal human bronchial epithelial (NHBE) and tumorigenic HBE cells (1170-I) by high-throughput immunoblotting (PowerBlot Western Array) using 800 monoclonal antibodies. This analysis revealed that 87 proteins increased by >2-fold, and 45 proteins decreased by >2-fold, in 1170-I compared with NHBE cells. These proteins are involved in DNA synthesis and repair, cell cycle regulation, RNA transcription and degradation, translation, differentiation, angiogenesis, apoptosis, cell adhesion, cytoskeleton and cell motility, and the phosphatidylinositol 3-kinase signaling pathway. Conventional Western blotting using lysates of normal, immortalized, transformed, and tumorigenic HBEs and non-small cell lung cancer cell lines confirmed some of these changes. The expression of several of these proteins has been then analyzed by immunohistochemistry in tissue microarrays containing 323 samples, including normal bronchial epithelium, hyperplasia, squamous metaplasia, dysplasias, squamous cell carcinomas, atypical adenomatous hyperplasia, and adenocarcinomas from 144 patients. The results of the immunohistochemical studies correlated with the Western blotting findings and showed gradual increases (caspase-8, signal transducers and activators of transcription 5, and p70s6K) or decrease (E-cadherin) in levels with tumor progression. These results indicate that the changes in proteins detected in this study may occur early in lung carcinogenesis and persist in lung cancer. In addition, some of the proteins detected by this approach may be novel biomarkers for early detection of lung cancer and novel targets for chemoprevention or therapy.


Asunto(s)
Neoplasias Pulmonares/metabolismo , Pulmón/metabolismo , Lesiones Precancerosas/metabolismo , Proteínas/análisis , Proteómica/métodos , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting/métodos , Moléculas de Adhesión Celular/análisis , Proteínas de Ciclo Celular/análisis , Línea Celular Tumoral , Células Cultivadas , Electroforesis en Gel de Poliacrilamida , Femenino , Humanos , Inmunohistoquímica , Pulmón/citología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/patología , Reproducibilidad de los Resultados
12.
Cancer Res ; 66(14): 7111-8, 2006 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-16849557

RESUMEN

The expression of retinoic acid receptor beta2 (RAR-beta2) is frequently lost in various cancers and their premalignant lesions. However, the restoration of RAR-beta2 expression inhibits tumor cell growth and suppresses cancer development. To understand the molecular mechanisms responsible for this RAR-beta2-mediated antitumor activity, we did restriction fragment differential display-PCR and cloned a novel retinoid receptor-induced gene 1 (RRIG1), which is differentially expressed in RAR-beta2-positive and RAR-beta2-negative tumor cells. RRIG1 cDNA contains 2,851 bp and encodes a protein with 276 amino acids; the gene is localized at chromosome 9q34. Expressed in a broad range of normal tissues, RRIG1 is also lost in various cancer specimens. RRIG1 mediates the effect of RAR-beta2 on cell growth and gene expression (e.g., extracellular signal-regulated kinase 1/2 and cyclooxygenase-2). The RRIG1 protein is expressed in the cell membrane and binds to and inhibits the activity of a small GTPase RhoA. Whereas induction of RRIG1 expression inhibits RhoA activation and f-actin formation and consequently reduces colony formation, invasion, and proliferation of esophageal cancer cells, antisense RRIG1 increases RhoA activity and f-actin formation and thus induces the colony formation, invasion, and proliferation of these cells. Our findings therefore show a novel molecular pathway involving RAR-beta2 regulation of RRIG1 expression and RRIG1-RhoA interaction. An understanding of this pathway may translate into better control of human cancer.


Asunto(s)
Neoplasias Esofágicas/metabolismo , Proteínas de la Membrana/metabolismo , Receptores de Ácido Retinoico/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Procesos de Crecimiento Celular , Línea Celular Tumoral , Chlorocebus aethiops , Clonación Molecular , ADN Complementario/genética , Neoplasias Esofágicas/genética , Humanos , Proteínas de la Membrana/genética , Datos de Secuencia Molecular , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Ácido Retinoico/biosíntesis , Receptores de Ácido Retinoico/genética , Proteínas Supresoras de Tumor/genética , Proteína de Unión al GTP rhoA/antagonistas & inhibidores
13.
Cancer Res ; 66(6): 3114-9, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540661

RESUMEN

Prostate cancer is one of the malignant tumors which exhibit resistance to anticancer drugs, at least in part due to enhanced antiapoptotic mechanisms. Therefore, the understanding of such mechanisms should improve the design of chemotherapy against prostate cancer. Galectin-3 (Gal-3), a multifunctional oncogenic protein involved in the regulation of tumor proliferation, angiogenesis, and apoptosis has shown antiapoptotic effects in certain cell types. Here, we show that the expression of exogenous Gal-3 in human prostate cancer LNCaP cells, which do not express Gal-3 constitutively, inhibits anticancer drug-induced apoptosis by stabilizing the mitochondria. Thus, Gal-3-negative cells showed 66.31% apoptosis after treatment with 50 micromol/L cis-diammine-dichloroplatinum for 48 hours, whereas two clones of Gal-3-expressing cells show only 2.92% and 1.42% apoptotic cells. Similarly, Gal-3-negative cells showed 43.8% apoptosis after treatment with 300 micromol/L etoposide for 48 hours, whereas only 15.38% and 14.51% of Gal-3-expressing LNCaP cells were apoptotic. The expression of Gal-3 stimulated the phosphorylation of Ser(112) of Bcl-2-associated death (Bad) protein and down-regulated Bad expression after treatment with cis-diammine-dichloroplatinum. Gal-3 also inhibited mitochondrial depolarization and damage after translocation from the nuclei to the cytoplasm, resulting in inhibition of cytochrome c release and caspase-3 activation. These findings indicate that Gal-3 inhibits anticancer drug-induced apoptosis through regulation of Bad protein and suppression of the mitochondrial apoptosis pathway. Therefore, targeting Gal-3 could improve the efficacy of anticancer drug chemotherapy in prostate cancer.


Asunto(s)
Apoptosis/fisiología , Galectina 3/fisiología , Mitocondrias/fisiología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Apoptosis/efectos de los fármacos , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cisplatino/farmacología , Citocromos c/antagonistas & inhibidores , Citocromos c/metabolismo , Activación Enzimática , Etopósido/farmacología , Galectina 3/biosíntesis , Galectina 3/genética , Galectina 3/metabolismo , Humanos , Masculino , Mitocondrias/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Transfección , Proteína Letal Asociada a bcl/biosíntesis , Proteína Letal Asociada a bcl/metabolismo
14.
Mol Cancer Ther ; 6(11): 2967-75, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18025281

RESUMEN

Alterations in histone acetylation status have been implicated in carcinogenesis. Histone deacetylase inhibitors, such as suberoylanilide hydroxamic acid (SAHA), can potentially reactivate aberrantly silenced genes by restoring histone acetylation and allowing gene transcription. However, the mechanisms underlying the effects of SAHA on cell growth, differentiation, and death remain unclear. In this study, we assessed the activity of SAHA in modulating cell growth and apoptosis in head and neck squamous cell carcinoma (HNSCC) cells compared with premalignant leukoplakia and normal oral cells. SAHA induced growth inhibition, cell cycle changes, and apoptosis in HNSCC cell lines but had limited effects on premalignant and normal cells. Although SAHA triggered the mitochondrial pathway of apoptosis, including cytochrome c release, caspase-3 and caspase-9 activation, and poly(ADP-ribose) polymerase cleavage in HNSCC cells, specific inhibition of caspase-9 only partially blocked the induction of apoptosis induction. SAHA also activated the extrinsic apoptosis pathway, including increased Fas and Fas ligand (FasL) expression, activation of caspase-8, and cleavage of Bid. Interfering with Fas signaling blocked apoptosis induction and blunted growth inhibition by SAHA. Our results show for the first time that SAHA induces apoptosis in HNSCC cells through activation of the Fas/FasL death pathway in addition to the intrinsic mitochondrial pathway although having comparatively little activity against precancerous and normal oral cells with intrinsic Fas and FasL expression.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Neoplasias de Cabeza y Cuello/patología , Inhibidores de Histona Desacetilasas , Ácidos Hidroxámicos/farmacología , Mitocondrias/metabolismo , Receptor fas/metabolismo , Acetilación/efectos de los fármacos , Carcinoma de Células Escamosas/enzimología , Carcinoma de Células Escamosas/genética , Caspasas/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Activación Enzimática/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Neoplasias de Cabeza y Cuello/enzimología , Neoplasias de Cabeza y Cuello/genética , Histonas/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Boca/citología , Boca/efectos de los fármacos , Lesiones Precancerosas , Receptores de Muerte Celular/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre , Vorinostat
15.
J Exp Ther Oncol ; 6(3): 183-92, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17552358

RESUMEN

The arachidonic acid-metabolizing enzymes cyclooxygenase-2 (COX-2) or 5-lipoxygenase (5-LOX) are overexpressed during lung carcinogenesis and their end products (e.g.; PGE2, 5-HETE, and LTB4) have been implicated in tumor development. Recently, COX-2 inhibitors (e.g.; celecoxib) and 5-LOX inhibitors (e.g.; MK886 and REV5901) used as single agents have shown promising activities in the treatment and chemoprevention of cancer. However, little is known about the effects of combinations of these inhibitors. We found that simultaneous treatment of premalignant and malignant human lung cell lines with celecoxib, MK886, and REV5901 is more potent in growth suppression and induction of cell death than single or dual combination of these agents. However, their sensitivity to the inhibitors was not directly associated with the expression of COX-2, 5-LOX, or 5-LOX-activating protein (FLAP), but correlated with the production of corresponding metabolites. Furthermore, partial protection of cell death was observed when PGE2 and/or 5-HETE was added to cell cultures treated with celecoxib, MK886, and REV5901 simultaneously. Our data indicate that a triple drug combination of distinct inhibitors of the eicosanoid metabolism at clinically feasible concentrations were more effective than each agent alone suggesting further investigations.


Asunto(s)
Inhibidores de la Ciclooxigenasa 2/farmacología , Indoles/farmacología , Inhibidores de la Lipooxigenasa/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Pirazoles/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Proteínas Activadoras de la 5-Lipooxigenasa , Apoptosis/efectos de los fármacos , Western Blotting , Proteínas Portadoras/antagonistas & inhibidores , Celecoxib , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Línea Celular Tumoral , Dinoprostona/farmacología , Humanos , Ácidos Hidroxieicosatetraenoicos/farmacología , Leucotrieno B4/farmacología , Neoplasias Pulmonares/patología , Proteínas de la Membrana/antagonistas & inhibidores , Espectrometría de Masa por Ionización de Electrospray
16.
Clin Cancer Res ; 12(24): 7353-8, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17189407

RESUMEN

PURPOSE: We investigated the methylation status and protein expression of four tumor suppressor genes to determine their role in salivary gland tumorigenesis. EXPERIMENTAL DESIGN: We performed methylation-specific PCR and protein analyses of 29 normal salivary glands, 23 benign, and 79 malignant salivary gland neoplasms to determine the pattern and potential diagnostic and/or biological role of the RASSF1, RARbeta2, DAPK, and MGMT tumor suppressor gene methylation in these tumors. RESULTS: No methylation was detected in the normal tissues. Methylation occurred in 9 of 23 (39.1%) benign tumors; 3 (25.0%) pleomorphic adenomas and 6 (66.7%) Warthin's tumors at the MGMT, DAPK, or RASSF1 genes. Methylation occurred in 33 of 79 (41.8%) malignant tumors; 8 (30.8%) adenoid cystic carcinomas, 6 (33.3%) mucoepidermoid carcinomas, 6 (42.9%) acinic cell carcinomas, and 13 (62.0%) salivary duct carcinomas. RASSF1 and RARbeta2 represented 75.8% of methylation events occurring most frequently in salivary duct and acinic cell carcinomas. Overall, we found no significant correlation between protein expression and methylation status of individual genes, but observed low or absent protein expression in several methylated tumors. Significant correlations were found between methylation and aggressive malignant phenotypes (P = 0.0004) and age (P = 0.05). CONCLUSIONS: (a) Benign and malignant salivary tumors differed in the frequency and pattern of gene methylation; (b) high-grade carcinomas were significantly methylated compared with low-grade phenotypes; (c) RASSF1 and RARbeta2 were highly methylated in malignant tumors and can be targeted for therapy; and (d) methylation pattern may serve as a diagnostic and biological marker in assessing these tumors.


Asunto(s)
Carcinoma/metabolismo , Metilación de ADN , Neoplasias de las Glándulas Salivales/genética , Neoplasias de las Glándulas Salivales/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Carcinoma de Células Acinares/metabolismo , Carcinoma Adenoide Quístico/metabolismo , Carcinoma Mucoepidermoide/metabolismo , Niño , Metilasas de Modificación del ADN , Enzimas Reparadoras del ADN , Proteínas Quinasas Asociadas a Muerte Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Receptores de Ácido Retinoico/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo
17.
Clin Cancer Res ; 12(10): 3109-14, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707609

RESUMEN

PURPOSE: To test the hypothesis that the retinamide N-(4-hydroxyphenyl)retinamide (fenretinide) would be clinically active potentially via receptor-independent apoptosis and receptor-dependent effects in natural retinoid-resistant oral leukoplakia patients--the first test of this hypothesis in any in vivo setting. EXPERIMENTAL DESIGN: A phase II trial of fenretinide (200 mg/d for 3 months) in oral leukoplakia patients who had not responded (de novo resistance) or who had responded and then relapsed (acquired resistance) to previous treatment with natural retinoids. We analyzed apoptosis via the terminal deoxynucleotidyl transferase-mediated nick end labeling in situ DNA fragmentation assay. RESULTS: We accrued 35 evaluable patients with retinoid-resistant oral leukoplakia, 12 (34.3%) had partial responses to fenretinide (95% confidence interval, 19.2-52.4%), and response was associated with acquired resistance to natural retinoids (P = 0.015, Fisher's exact test). Nine responders progressed within 9 months of stopping fenretinide. Toxicity was minimal and compliance was excellent. Mean apoptosis values (SE) increased from 0.35% (0.25%) at baseline to 1.18% (0.64%) at 3 months (P = 0.001, sign test); this increase did not correlate with clinical response. The increases in 3-month mean serum concentrations of fenretinide (0.23 micromol/L) and N-(4-methoxyphenyl)retinamide (0.57 micromol/L) correlated with decreased retinol concentrations [Spearman correlation coefficient of -0.57 (P = 0.001) and -0.43 (P = 0.01), respectively]. CONCLUSIONS: Low-dose fenretinide was clinically active and produced a small increase in apoptosis in retinoid-resistant oral leukoplakia.


Asunto(s)
Anticarcinógenos/uso terapéutico , Fenretinida/uso terapéutico , Leucoplasia Bucal/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Daño del ADN , ADN Nucleotidilexotransferasa , Resistencia a Medicamentos , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Masculino , Persona de Mediana Edad , Retinoides/farmacología , Resultado del Tratamiento
18.
Clin Cancer Res ; 12(12): 3661-97, 2006 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-16778094

RESUMEN

This article reviews progress in chemopreventive drug development, especially data and concepts that are new since the 2002 AACR report on treatment and prevention of intraepithelial neoplasia. Molecular biomarker expressions involved in mechanisms of carcinogenesis and genetic progression models of intraepithelial neoplasia are discussed and analyzed for how they can inform mechanism-based, molecularly targeted drug development as well as risk stratification, cohort selection, and end-point selection for clinical trials. We outline the concept of augmenting the risk, mechanistic, and disease data from histopathologic intraepithelial neoplasia assessments with molecular biomarker data. Updates of work in 10 clinical target organ sites include new data on molecular progression, significant completed trials, new agents of interest, and promising directions for future clinical studies. This overview concludes with strategies for accelerating chemopreventive drug development, such as integrating the best science into chemopreventive strategies and regulatory policy, providing incentives for industry to accelerate preventive drugs, fostering multisector cooperation in sharing clinical samples and data, and creating public-private partnerships to foster new regulatory policies and public education.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Glandulares y Epiteliales/prevención & control , Neoplasias de la Mama/prevención & control , Quimioprevención , Neoplasias Colorrectales/prevención & control , Progresión de la Enfermedad , Femenino , Humanos , Infecciones , Inflamación , Masculino , Monitoreo Fisiológico , Transducción de Señal
19.
Cancer Res ; 65(20): 9555-65, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16230421

RESUMEN

N-(4-hydroxyphenyl) retinamide [4-HPR], a synthetic retinoid, has been shown to inhibit tumor cell growth, invasion, and metastasis by a mechanism that is not fully understood. Because the nuclear factor-kappaB (NF-kappaB) has also been shown to regulate proliferation, invasion, and metastasis of tumor cells, we postulated that 4-HPR modulates the activity of NF-kappaB. To test this postulate, we examined the effect of this retinoid on NF-kappaB and NF-kappaB-regulated gene products. We found that 4-HPR potentiated the apoptosis induced by tumor necrosis factor (TNF) and chemotherapeutic agents, suppressed TNF-induced invasion, and inhibited RANKL-induced osteoclastogenesis, all of which are known to require NF-kappaB activation. We found that 4-HPR suppressed both inducible and constitutive NF-kappaB activation without interfering with the direct DNA binding of NF-kappaB. 4-HPR was found to be synergistic with Velcade, a proteasome inhibitor. Further studies showed that 4-HPR blocked the phosphorylation and degradation of IkappaBalpha through the inhibition of activation of IkappaBalpha kinase (IKK), and this led to suppression of the phosphorylation and nuclear translocation of p65. 4-HPR also inhibited TNF-induced Akt activation linked with IKK activation. NF-kappaB-dependent reporter gene expression was also suppressed by 4-HPR, as was NF-kappaB reporter activity induced by TNFR1, TRADD, TRAF2, NIK, and IKK but not that induced by p65 transfection. The expression of NF-kappaB-regulated gene products involved in antiapoptosis (IAP1, Bfl-1/A1, Bcl-2, cFLIP, and TRAF1), proliferation (cyclin D1 and c-Myc), and angiogenesis (vascular endothelial growth factor, cyclooxygenase-2, and matrix metalloproteinase-9) were also down-regulated by 4-HPR. This correlated with potentiation of apoptosis induced by TNF and chemotherapeutic agents.


Asunto(s)
Apoptosis/efectos de los fármacos , Fenretinida/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Osteoclastos/efectos de los fármacos , Antineoplásicos/farmacología , Ácidos Borónicos/farmacología , Bortezomib , Carcinoma de Células Pequeñas/tratamiento farmacológico , Carcinoma de Células Pequeñas/genética , Carcinoma de Células Pequeñas/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Procesos de Crecimiento Celular/efectos de los fármacos , Ciclina D1/biosíntesis , Ciclina D1/genética , Ciclooxigenasa 2/genética , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Genes myc/efectos de los fármacos , Humanos , Quinasa I-kappa B/biosíntesis , Quinasa I-kappa B/genética , Proteínas I-kappa B/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Glicoproteínas de Membrana/antagonistas & inhibidores , Inhibidor NF-kappaB alfa , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína Oncogénica v-akt/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Proteínas Proto-Oncogénicas c-myc/genética , Pirazinas/farmacología , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Factor A de Crecimiento Endotelial Vascular/genética
20.
Cancer Res ; 65(4): 1117-23, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15734991

RESUMEN

The 10th International Conference on Differentiation Therapy was held between April 29 and May 3, 2004, in Shanghai, China. In the tradition of previous conferences from this series, which have been held biannually since the first meeting organized 20 years ago by Samuel Waxman and Giovanni Rossi in Sardinia, the organizers of the 10th International Conference on Differentiation Therapy aimed to gather basic and clinical cancer investigators in a setting of plenary sessions, workshops, and poster presentations to maximize the effective exchange of information and foster the establishment of collaborative interactions. Approximately 300 scientists attended the meeting with a mission to discuss targeted approaches to cancer treatment, which stem from our understanding of basic biological processes and the mechanisms of their deregulation during tumorigenesis.


Asunto(s)
Neoplasias/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Ensayos Clínicos como Asunto , Humanos , Neoplasias/genética , Neoplasias/patología , Tretinoina/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA