Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Inflamm Res ; 64(5): 343-61, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25795230

RESUMEN

INTRODUCTION: Chronic pulmonary inflammation has been consistently shown to increase the risk of lung cancer. Therefore, assessing the molecular links between the two diseases and identification of chemopreventive agents that inhibit inflammation-driven lung tumorigenesis is indispensable. MATERIALS AND METHODS: Female A/J mice were treated with the tobacco smoke carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) and lipopolysaccharide (LPS), a potent inflammatory agent and constituent of tobacco smoke, and maintained on control diet or diet supplemented with the chemopreventive agents indole-3-carbinol (I3C) and/or silibinin (Sil). At the end of the study, mice were sacrificed and tumors on the surface of the lung were counted and gene expression levels in lung tissues were determined by RNA sequencing. RESULTS: The mean number of lung tumors induced by NNK and NNK + LPS was 5 and 15 tumors/mouse, respectively. Dietary supplementation with the combination of I3C and Sil significantly reduced the size and multiplicity (by 50 %) of NNK + LPS-induced lung tumors. Also, we found that 330, 2957, and 1143 genes were differentially regulated in mice treated with NNK, LPS, and NNK + LPS, respectively. The inflammatory response of lung tumors to LPS, as determined by the number of proinflammatory genes with altered gene expression or the level of alteration, was markedly less than that of normal lungs. Among 1143 genes differentially regulated in the NNK + LPS group, the expression of 162 genes and associated signaling pathways was significantly modulated by I3C and/or Sil + I3C. These genes include cytokines, chemokines, putative oncogenes and tumor suppressor genes and Ros1, AREG, EREG, Cyp1a1, Arntl, and Npas2. CONCLUSION: To our knowledge, this is the first report that provides insight into genes that are differentially expressed during inflammation-driven lung tumorigenesis and the modulation of these genes by chemopreventive agents.


Asunto(s)
Anticarcinógenos/farmacología , Regulación Neoplásica de la Expresión Génica/genética , Inflamación/complicaciones , Inflamación/genética , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/genética , Animales , Carcinógenos/farmacología , Femenino , Indoles/farmacología , Lipopolisacáridos/farmacología , Pulmón/patología , Ratones , Ratones Endogámicos A , ARN/biosíntesis , ARN/genética , Análisis de Secuencia de ARN , Silibina , Silimarina/farmacología
2.
Cancer Prev Res (Phila) ; 8(1): 77-85, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25403850

RESUMEN

Inflammatory conditions of the lung such as chronic obstructive pulmonary disease (COPD) are known to increase lung cancer risk, particularly lung squamous cell carcinoma (LSCC). In the present study, we developed a mouse model of inflammation-driven LSCC that was induced by N-nitroso-trischloroethylurea (NTCU) and enhanced by lipopolysaccharide (LPS), a potent proinflammatory agent contained in tobacco and tobacco smoke, and determined the chemopreventive effects of BioResponse diindolylmethane (DIM) in the same model. Compared with mice treated with NTCU alone, mice treated with the combination of NTCU and LPS had a 9-fold increase in the number of bronchioles with LSCC. Also, compared with mice treated with LPS alone, mice treated with NTCU plus LPS showed significantly increased expression of the inflammatory cytokines IL1α, IL6, and TNFα (all three increased about 7-fold). Parallel to the increased cytokine gene expression, the NTCU plus LPS-treated group exhibited significantly enhanced activation of NF-κB, STAT3, ERK, p-38, and Akt, expression of p53, COX-2, and Mcl-1, and NF-κB- and STAT3-DNA binding in the lung. Dietary administration of DIM (10 µmol/g diet or 2,460 ppm) to mice treated with NTCU plus LPS reduced the incidence of LSCC by 2-fold, suppressed activation/expression of proinflammatory and procarcinogenic proteins and NF-κB- and STAT3-DNA binding, but not the expression of cytokines and p53. This study highlights the potential significance of our mouse model to identify promising drugs or dietary agents for the chemoprevention of human LSCC and that DIM is a very good candidate for clinical lung cancer chemoprevention trials.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Indoles/administración & dosificación , Inflamación/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Carmustina/análogos & derivados , Carmustina/química , Citocinas/metabolismo , Dieta , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Indoles/química , Lipopolisacáridos/química , Pulmón/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , FN-kappa B/metabolismo
3.
Int J Pharm ; 477(1-2): 96-101, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25311179

RESUMEN

Indole-3-carbinol (I3C), a constituent of commonly consumed Brassica vegetables, has been shown to have anticancer effects in a variety of preclinical models of lung cancer. However, it has shown only limited efficacy in clinical trials, likely due to its poor oral bioavailability. Intranasal administration of I3C has the potential to enhance the pulmonary accumulation of the drug, thereby improving its availability at the target site of action. In this study, we developed a liposomal formulation of I3C and evaluated its lung delivery and chemopreventive potential in tobacco smoke carcinogen [4-(methylnitro-samino)-1-(3-pyridyl)-1-butanone (NNK)]-treated mice. Intranasal administration of I3C liposomes led to a ∼100-fold higher lung exposure of I3C than the oral route of administration. Further, intranasal delivery of liposomal I3C led to a significant reduction (37%; p<0.05) in the levels of the DNA adduct formation induced by NNK treatment. Liposomal I3C also significantly increased (by 10-fold) the expression of CYP1A1, a cytochrome P450 enzyme known to increase the detoxification of chemical carcinogens by enhancing their metabolism. Overall, our findings demonstrate that intranasal administration of liposomal I3C has the potential to significantly improve the efficacy of I3C for lung cancer chemoprevention.


Asunto(s)
Anticarcinógenos/administración & dosificación , Sistemas de Liberación de Medicamentos , Indoles/administración & dosificación , Neoplasias Pulmonares/prevención & control , Administración Intranasal , Administración Oral , Animales , Anticarcinógenos/farmacocinética , Disponibilidad Biológica , Carcinógenos/toxicidad , Citocromo P-450 CYP1A1/genética , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Indoles/farmacocinética , Liposomas , Pulmón/metabolismo , Ratones , Nitrosaminas/toxicidad , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...