Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Clin Infect Dis ; 60(2): 208-15, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25305284

RESUMEN

BACKGROUND: Enterotoxigenic Bacteroides fragilis (ETBF) produces the Bacteroides fragilis toxin, which has been associated with acute diarrheal disease, inflammatory bowel disease, and colorectal cancer (CRC). ETBF induces colon carcinogenesis in experimental models. Previous human studies have demonstrated frequent asymptomatic fecal colonization with ETBF, but no study has investigated mucosal colonization that is expected to impact colon carcinogenesis. METHODS: We compared the presence of the bft gene in mucosal samples from colorectal neoplasia patients (cases, n = 49) to a control group undergoing outpatient colonoscopy for CRC screening or diagnostic workup (controls, n = 49). Single bacterial colonies isolated anaerobically from mucosal colon tissue were tested for the bft gene with touch-down polymerase chain reaction. RESULTS: The mucosa of cases was significantly more often bft-positive on left (85.7%) and right (91.7%) tumor and/or paired normal tissues compared with left and right control biopsies (53.1%; P = .033 and 55.5%; P = .04, respectively). Detection of bft was concordant in most paired mucosal samples from individual cases or controls (75% cases; 67% controls). There was a trend toward increased bft positivity in mucosa from late- vs early-stage CRC patients (100% vs 72.7%, respectively; P = .093). In contrast to ETBF diarrheal disease where bft-1 detection dominates, bft-2 was the most frequent toxin isotype identified in both cases and controls, whereas multiple bft isotypes were detected more frequently in cases (P ≤ .02). CONCLUSIONS: The bft gene is associated with colorectal neoplasia, especially in late-stage CRC. Our results suggest that mucosal bft exposure is common and may be a risk factor for developing CRC.


Asunto(s)
Toxinas Bacterianas/genética , Bacteroides fragilis/genética , Colon/patología , Neoplasias Colorrectales/patología , ADN Bacteriano/análisis , Genes Bacterianos , Mucosa Intestinal/patología , Metaloendopeptidasas/genética , Anciano , Colon/microbiología , Neoplasias Colorrectales/epidemiología , Neoplasias Colorrectales/microbiología , ADN Bacteriano/genética , Femenino , Humanos , Mucosa Intestinal/microbiología , Masculino , Persona de Mediana Edad , Factores de Riesgo
2.
Amino Acids ; 42(2-3): 529-38, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21814790

RESUMEN

Ornithine decarboxylase antizyme 1 (AZ1) is a major regulatory protein responsible for the regulation and degradation of ornithine decarboxylase (ODC). To better understand the role of AZ1 in polyamine metabolism and in modulating the response to anticancer polyamine analogues, a small interfering RNA strategy was used to create a series of stable clones in human H157 non-small cell lung cancer cells that expressed less than 5-10% of basal AZ1 levels. Antizyme 1 knockdown clones accumulated greater amounts of the polyamine analogue N (1),N (11)-bis(ethyl)norspermine (BENSpm) and were more sensitive to analogue treatment. The possibility of a loss of polyamine uptake regulation in the knockdown clones was confirmed by polyamine uptake analysis. These results are consistent with the hypothesis that AZ1 knockdown leads to dysregulation of polyamine uptake, resulting in increased analogue accumulation and toxicity. Importantly, there appears to be little difference between AZ1 knockdown cells and cells with normal levels of AZ1 with respect to ODC regulation, suggesting that another regulatory protein, potentially AZ2, compensates for the loss of AZ1. The results of these studies are important for the understanding of both the regulation of polyamine homeostasis and in understanding the factors that regulate tumor cell sensitivity to the anti-tumor polyamine analogues.


Asunto(s)
Técnicas de Silenciamiento del Gen , Neoplasias Pulmonares/metabolismo , Proteínas/metabolismo , Espermina/análogos & derivados , Secuencia de Bases , Northern Blotting , Línea Celular Tumoral , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Datos de Secuencia Molecular , Poliaminas/metabolismo , Proteínas/genética , ARN Interferente Pequeño , Espermina/metabolismo , Espermina/toxicidad
3.
Proc Natl Acad Sci U S A ; 108(37): 15354-9, 2011 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-21876161

RESUMEN

It is estimated that the etiology of 20-30% of epithelial cancers is directly associated with inflammation, although the direct molecular events linking inflammation and carcinogenesis are poorly defined. In the context of gastrointestinal disease, the bacterium enterotoxigenic Bacteroides fragilis (ETBF) is a significant source of chronic inflammation and has been implicated as a risk factor for colorectal cancer. Spermine oxidase (SMO) is a polyamine catabolic enzyme that is highly inducible by inflammatory stimuli resulting in increased reactive oxygen species (ROS) and DNA damage. We now demonstrate that purified B. fragilis toxin (BFT) up-regulates SMO in HT29/c1 and T84 colonic epithelial cells, resulting in SMO-dependent generation of ROS and induction of γ-H2A.x, a marker of DNA damage. Further, ETBF-induced colitis in C57BL/6 mice is associated with increased SMO expression and treatment of mice with an inhibitor of polyamine catabolism, N(1),N(4)-bis(2,3-butandienyl)-1,4-butanediamine (MDL 72527), significantly reduces ETBF-induced chronic inflammation and proliferation. Most importantly, in the multiple intestinal neoplasia (Min) mouse model, treatment with MDL 72527 reduces ETBF-induced colon tumorigenesis by 69% (P < 0.001). The results of these studies indicate that SMO is a source of bacteria-induced ROS directly associated with tumorigenesis and could serve as a unique target for chemoprevention.


Asunto(s)
Bacteroides fragilis/fisiología , Neoplasias del Colon/microbiología , Poliaminas/metabolismo , Lesiones Precancerosas/microbiología , Acetiltransferasas/metabolismo , Animales , Toxinas Bacterianas/toxicidad , Bacteroides fragilis/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Colitis/patología , Neoplasias del Colon/complicaciones , Neoplasias del Colon/patología , Daño del ADN , Modelos Animales de Enfermedad , Inducción Enzimática/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/enzimología , Humanos , Inflamación/complicaciones , Inflamación/patología , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/biosíntesis , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Lesiones Precancerosas/patología , Putrescina/análogos & derivados , Putrescina/farmacología , Proteínas Recombinantes/toxicidad , Poliamino Oxidasa
4.
Methods Mol Biol ; 720: 173-81, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21318873

RESUMEN

Spermine oxidase (SMO), the most recently characterized polyamine metabolic enzyme, catalyzes the direct back-conversion of spermine to spermidine in an FAD-dependent reaction that also yields the byproducts hydrogen peroxide (H(2)O(2)) and 3-aminopropanal. These metabolites, particularly H(2)O(2), have been implicated in cytotoxic cellular responses to specific antitumor polyamine analogs, as well as in the inflammation-associated generation of DNA damage. This chapter describes a rapid, sensitive, and inexpensive method for the chemiluminescent measurement of SMO (or alternatively, N (1)-acetyl polyamine oxidase, APAO) enzyme activity in cultured cell lysates, without the need for radioactive reagents or the use of high performance liquid chromatography (HPLC). Specifically, H(2)O(2) production by SMO is coupled to chemiluminescence generated by the horseradish peroxidase-catalyzed oxidation of luminol. Detailed protocols for preparation of reagents, harvesting cell lysates, generation of a standard curve, assaying of samples, and calculation of SMO enzyme activity are presented.


Asunto(s)
Enfermedad , Pruebas de Enzimas/métodos , Mamíferos/metabolismo , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Poliaminas/metabolismo , Animales , Adhesión Celular , Extractos Celulares , Separación Celular , Peróxido de Hidrógeno/metabolismo , Poliaminas/química , Estándares de Referencia , Poliamino Oxidasa
5.
Microbiology (Reading) ; 154(Pt 8): 2231-2240, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18667556

RESUMEN

Adaptation to the acidic microenvironment, and adherence to mucosal epithelium, are essential for persistent colonization of the human stomach by Helicobacter pylori. The expression of SabA, an adhesin implicated in the ability of H. pylori to adhere to the host gastric epithelium, can be modulated by phase variation via slipped-strand mispairing in repetitive nucleotide tracts located in both the promoter region and the coding region. This study demonstrates the occurrence of phase variation at the sabA locus within individual strains of H. pylori, and among multiple isolates from a single patient. In addition, transcription of sabA is repressed by the acid-responsive ArsRS two-component signal transduction system in vitro. Our results demonstrate that isogenic inactivation of the arsS (jhp0151/HP0165) histidine kinase locus results in a 10-fold SabA-dependent increase in adherence to gastric epithelial cells in strain J99 (contains an in-frame sabA allele), but not in strain 26695 (out-of-frame sabA allele). The combination of transcriptional regulation of the sabA locus by the ArsRS two-component signal-transduction system and the generation of subpopulations harbouring alternate sabA alleles by slipped-strand mispairing during chromosomal replication could permit H. pylori to rapidly adapt to varying microenvironments or host immune responses. As a pathogen with a paucity of regulatory proteins, this dual regulation indicates that SabA expression is a tightly regulated process in H. pylori infection.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Infecciones por Helicobacter/microbiología , Helicobacter pylori/metabolismo , Proteínas Quinasas/metabolismo , Transducción de Señal , Adhesinas Bacterianas/genética , Adhesión Bacteriana , Proteínas Bacterianas/genética , Línea Celular Tumoral , Mucosa Gástrica/microbiología , Helicobacter pylori/genética , Histidina Quinasa , Humanos , Fenotipo , Proteínas Quinasas/genética
6.
Prostate ; 68(7): 766-72, 2008 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-18302221

RESUMEN

BACKGROUND: Inflammation has been strongly implicated in prostate carcinogenesis, but the precise molecular mechanisms linking inflammation and carcinogenic DNA damage are not known. Induction of the polyamine catabolic enzyme, spermine oxidase (SMO) has been linked to increased reactive oxygen species (ROS) and DNA damage in human gastric and lung epithelial cells and suggest direct mechanistic links between inflammation, SMO activity, ROS production, and epithelial carcinogenesis that are likely relevant in prostate cancer. METHODS: Tissue microarrays consisting of matched normal and diseased specimens from patients diagnosed with prostate cancer, prostatic intraepithelial neoplasia (PIN), or proliferative inflammatory atrophy (PIA), as well as unaffected individuals, were stained for SMO expression and analyzed using image analysis techniques and TMAJ software tools. RESULTS: Average SMO staining was significantly higher in prostate cancer and PIN tissues compared to patient-matched benign tissues. Benign tissues from prostate cancer, PIN, and PIA patients also exhibited significantly higher mean SMO expression versus tissues from prostate disease-free patients. CONCLUSIONS: Tissues from patients diagnosed with prostate cancer and PIN exhibit, on average, locally increased SMO expression in regions of prostatic disease and higher overall SMO expression in prostatic epithelial cells compared to healthy individuals. Further studies are warranted to directly examine the role of SMO-produced ROS in prostate carcinogenesis.


Asunto(s)
Adenocarcinoma/enzimología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Neoplasia Intraepitelial Prostática/enzimología , Neoplasias de la Próstata/enzimología , Adenocarcinoma/química , Adenocarcinoma/patología , Biomarcadores de Tumor/análisis , Técnica del Anticuerpo Fluorescente Directa , Humanos , Técnicas para Inmunoenzimas , Masculino , Neoplasia Intraepitelial Prostática/química , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/química , Neoplasias de la Próstata/patología , Análisis de Matrices Tisulares , Poliamino Oxidasa
7.
Proc Natl Acad Sci U S A ; 104(19): 8023-8, 2007 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-17463086

RESUMEN

Epigenetic chromatin modification is a major regulator of eukaryotic gene expression, and aberrant epigenetic silencing of gene expression contributes to tumorigenesis. Histone modifications include acetylation, phosphorylation, and methylation, resulting in a combination of histone marks known collectively as the histone code. The chromatin marks at a given promoter determine, in part, whether specific promoters are in an open/active conformation or closed/repressed conformation. Dimethyl-lysine 4 histone H3 (H3K4me2) is a transcription-activating chromatin mark at gene promoters, and demethylation of this mark by the lysine-specific demethylase 1 (LSD1), a homologue of polyamine oxidases, may broadly repress gene expression. We now report that novel biguanide and bisguanidine polyamine analogues are potent inhibitors of LSD1. These analogues inhibit LSD1 in human colon carcinoma cells and affect a reexpression of multiple, aberrantly silenced genes important in the development of colon cancer, including members of the secreted frizzle-related proteins (SFRPs) and the GATA family of transcription factors. Furthermore, we demonstrate by chromatin immunoprecipitation analysis that the reexpression is concurrent with increased H3K4me2 and acetyl-H3K9 marks, decreased H3K9me1 and H3K9me2 repressive marks. We thus define important new agents for reversing aberrant repression of gene transcription.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Silenciador del Gen/efectos de los fármacos , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , Poliaminas/farmacología , Biguanidas/farmacología , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Metilación de ADN , Factor de Transcripción GATA4/genética , Genes Supresores de Tumor , Histona Demetilasas , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA