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










Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 36(35): 5035-5044, 2017 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-28459462

RESUMO

Colorectal cancer (CRC) is characterized by genome-wide alterations to DNA methylation that influence gene expression and genomic stability. Less is known about the extent to which methylation is disrupted in the earliest stages of CRC development. In this study, we have combined laser-capture microdissection with reduced representation bisulfite sequencing to identify cancer-associated DNA methylation changes in human aberrant crypt foci (ACF), the earliest putative precursor to CRC. Using this approach, methylation profiles have been generated for 10 KRAS-mutant ACF and 10 CRCs harboring a KRAS mutation, as well as matched samples of normal mucosa. Of 811 differentially methylated regions (DMRs) identified in ACF, 537 (66%) were hypermethylated and 274 (34%) were hypomethylated. DMRs located within intergenic regions were heavily enriched for AP-1 transcription factor binding sites and were frequently hypomethylated. Furthermore, gene ontology analysis demonstrated that DMRs associated with promoters were enriched for genes involved in intestinal development, including homeobox genes and targets of the Polycomb repressive complex 2. Consistent with their role in the earliest stages of colonic neoplasia, 75% of the loci harboring methylation changes in ACF were also altered in CRC samples, though the magnitude of change at these sites was lesser in ACF. Although aberrant promoter methylation was associated with altered gene expression in CRC, this was not the case in ACF, suggesting the insufficiency of methylation changes to modulate gene expression in early colonic neoplasia. Altogether, these data demonstrate that DNA methylation changes, including significant hypermethylation, occur more frequently in early colonic neoplasia than previously believed, and identify epigenomic features of ACF that may provide new targets for cancer chemoprevention or lead to the development of new biomarkers for CRC risk.


Assuntos
Neoplasias do Colo/genética , Metilação de DNA , Lesões Pré-Cancerosas/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias do Colo/patologia , Estudo de Associação Genômica Ampla , Humanos , Microdissecção e Captura a Laser , Lesões Pré-Cancerosas/patologia
2.
Br J Cancer ; 98(3): 587-95, 2008 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-18212756

RESUMO

Recent studies suggest that secreted phospholipases A2 (sPLA2s) represent attractive potential tumour biomarkers and therapeutic targets for various cancers. As a first step to address this issue in human colorectal cancer, we examined the expression of the full set of sPLA2s in sporadic adenocarcinomas and normal matched mucosa from 21 patients by quantitative PCR and immunohistochemistry. In normal colon, PLA2G2A and PLA2G12A were expressed at high levels, PLA2G2D, PLA2G5, PLA2G10 and PLA2G12B at moderate levels, and PLA2G1B, PLA2G2F and PLA2G3 at low levels. In adenocarcinomas from left and right colon, the expression of PLA2G3 was increased by up to 40-fold, while that of PLA2G2D and PLA2G5 was decreased by up to 23- and 14-fold. The variations of expression for sPLA2-IID, sPLA2-III and sPLA2-V were confirmed at the protein level. The expression pattern of these sPLA2s appeared to be linked respectively to the overexpression of interleukin-8, defensin alpha6, survivin and matrilysin, and downregulation of SFRP-1 and RLPA-1, all these genes being associated to colon cancer. This original sPLA2 profile observed in adenocarcinomas highlights the potential role of certain sPLA2s in colon cancer and suggests that sPLA2-III might be a good candidate as a novel biomarker for both left and right colon cancers.


Assuntos
Adenocarcinoma/enzimologia , Neoplasias do Colo/enzimologia , Fosfolipases A2 do Grupo III/biossíntese , Fosfolipases A2 Secretórias/metabolismo , Adenocarcinoma/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/análise , Colite/enzimologia , Colo/enzimologia , Neoplasias do Colo/genética , Feminino , Expressão Gênica , Fosfolipases A2 do Grupo III/metabolismo , Humanos , Imuno-Histoquímica , Mucosa Intestinal/enzimologia , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Regulação para Cima
3.
Vet Pathol ; 42(6): 735-52, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16301570

RESUMO

Transcriptional profiling of entire tumors has yielded considerable insight into the molecular mechanisms of heterogeneous cell populations within different types of neoplasms. The data thus acquired can be further refined by microdissection methods that enable the analyses of subpopulations of neoplastic cells. Separation of the various components of a neoplasm (i.e., stromal cells, inflammatory infiltrates, and blood vessels) has been problematic, primarily because of a paucity of tools for accurate microdissection. The advent of laser capture microdissection combined with powerful tools of linear amplification of RNA and high-throughput microarray-based assays have allowed the transcriptional mapping of intricate and highly complex networks within pure populations of neoplastic cells. With this approach, specific "molecular signatures" can be assigned to tumors of distinct or even similar histomorphology, thereby aiding the desired objective of pattern recognition, tumor classification, and prognostication. This review highlights the potential benefits of global gene expression profiling of tumor cells as a complement to conventional histopathologic analyses.


Assuntos
Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/veterinária , Microdissecção/veterinária , Neoplasias/metabolismo , Neoplasias/veterinária , Medicina Veterinária/métodos , Animais , Processamento de Imagem Assistida por Computador/métodos , Lasers , Análise em Microsséries/métodos , Análise em Microsséries/veterinária , Microdissecção/métodos , Neoplasias/genética , Especificidade da Espécie
4.
Mol Carcinog ; 31(4): 204-13, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11536370

RESUMO

Alterations in the transforming growth factor-beta (TGF-beta) pathway are implicated in the pathogenesis of colorectal cancer. We hypothesize that alterations in the TGF-beta pathway contribute to differential sensitivity of mice to the colon carcinogen azoxymethane (AOM). A/J (sensitive) and AKR/J (resistant) mice were injected intraperitoneally with AOM (10 mg/kg of body weight once a week for 6 wk). Twenty-four weeks after AOM exposure, mutational analysis of TGF-beta type II receptor (TbetaR-II) from normal colons and from tumors showed no AOM-induced alterations. A significant decrease (1.5-fold, P < 0.05) in TbetaR-II mRNA levels, however, was found in A/J tumors with the RNase protection assay. Immunofluorescence of TbetaR-II showed marked loss of staining in A/J tumors. The RNase protection assay and sequence analysis of the downstream signaling molecule Smad3 revealed no carcinogen-induced alterations in either strain. To gain further insight into the functionality of the pathway, expression of TGF-beta, TGF-beta type I receptor (TbetaR-I), and several downstream targets of TGF-beta signaling, including Smad7, c-myc, and p15, was examined. Although no alterations in TGF-beta, TbetaR-I, or Smad7 were found in tumors, a significant increase in c-myc expression (2.5-fold, P < 0.05 ) and a significant decrease in p15 expression (4.5-fold, P < 0.05 ) were noted. Concomitant repression of TbetaR-II and overexpression of c-myc may render epithelial cells insensitive to TGF-beta-mediated growth arrest, a possibility that also is suggested by this model. The significant decrease in p15 expression in tumors provides additional evidence that TGF-beta signaling may be markedly attenuated during colon tumorigenesis.


Assuntos
Azoximetano , Carcinógenos , Proteínas de Ciclo Celular , Neoplasias do Colo/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/fisiologia , Proteínas Supressoras de Tumor , Adenoma/induzido quimicamente , Adenoma/genética , Animais , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Inibidor de Quinase Dependente de Ciclina p15 , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos AKR , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/genética , Proteína Smad3 , Proteína Smad7 , Transativadores/biossíntese , Transativadores/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 279(6): G1282-91, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11093952

RESUMO

The transcription factor nuclear factor (NF)-kappaB regulates the expression of genes that can influence cell proliferation and death. Here we analyze the contribution of NF-kappaB to the regulation of epithelial cell turnover in the colon. Immunohistochemical, immunoblot, and DNA binding analyses indicate that NF-kappaB complexes change as colonocytes mature: p65-p50 complexes predominate in proliferating epithelial cells of the colon, whereas the p50-p50 dimer is prevalent in mature epithelial cells. NF-kappaB1 (p50) knockout mice were used to study the role of NF-kappaB in regulating epithelial cell turnover. Knockout animals lacked detectable NF-kappaB DNA binding activity in isolated epithelial cells and had significantly longer crypts with a more extensive proliferative zone than their wild-type counterparts (as determined by proliferating cell nuclear antigen staining and in vivo bromodeoxyuridine labeling). Gene expression profiling reveals that the NF-kappaB1 knockout mice express the potentially growth-enhancing tumor necrosis factor (TNF)-alpha and nerve growth factor-alpha genes at elevated levels, with in situ hybridization localizing some of the TNF-alpha expression to epithelial cells. TNF-alpha is NF-kappaB regulated, and its upregulation in NF-kappaB1 knockouts may result from an alleviation of p50-p50 repression. NF-kappaB complexes may therefore influence cell proliferation in the colon through their ability to selectively activate and/or repress gene expression.


Assuntos
Colo/anatomia & histologia , Células Epiteliais/fisiologia , Mucosa Intestinal/fisiologia , NF-kappa B/fisiologia , Fosfatase Alcalina/metabolismo , Animais , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Knockout , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
6.
Mol Carcinog ; 29(1): 25-36, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11020244

RESUMO

The transcription factor nuclear factor-kappaB (NF-kappaB) regulates genes that can influence cell proliferation, apoptosis, and inflammatory responses. Since these events can contribute to carcinogenesis, we examined the expression of NF-kappaB inhibitory proteins (IkappaBs) in normal and transformed colonic epithelial cells. Immunohistochemical analysis of the mouse colon revealed a high level of IkappaBbeta expression in epithelial cells relative to the rest of the tissue, whereas IkappaBalpha was found primarily in cells of the lamina propria. Mouse colon tumors showed a similar cell-specific staining pattern. Immunoblot analysis of IkappaBbeta from mouse colonocytes and the human HT-29 colon cancer cell line indicated that most of the IkappaBbeta in these cells was similar to the C-terminal-truncated IkappaBbeta2 isoform. Cell fractionation studies were consistent with IkappaBbeta being a major regulator of p65-p50 NF-kappaB complexes in HT-29 cells. Interestingly, two larger proteins specifically recognized by IkappaBbeta antibodies (p106 and p112) were found in HT-29 cells and in colon tissue of carcinogen-exposed mice. The p106 and p112 proteins bound to NF-kappaB, and their levels changed during the transient interleukin-1beta activation of NF-kappaB in HT-29 cells. Evidence was obtained indicating that p106 and p112 are stably ubiquitinated forms of IkappaBbeta. We propose that deficiencies in the proteasomal degradation of IkappaBbeta lead to p106 and p112 accumulation, which in turn alter NF-kappaB regulation in colon cancer cells.


Assuntos
Linhagem Celular Transformada , Colo/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas I-kappa B , Animais , Células Epiteliais/metabolismo , Células HT29 , Humanos , Camundongos
7.
Mol Carcinog ; 28(3): 139-47, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10942530

RESUMO

Alterations in the p16(INK4a) gene have been implicated in the pathogenesis of different human cancers and animal tumors. We postulated that alterations in the p16(INK4a) gene may also be involved in mouse colon tumorigenesis induced by the chemical carcinogen azoxymethane (AOM). In the present study, p16(INK4a) deletion status and its expression were examined in an AOM-induced mouse colon tumor model. Polymerase chain reaction-based deletion analysis of p16(INK4a) exon 2 showed no deletions in the colon tumors. The expression and localization of p16(INK4a) and its gene product were examined by reverse transcription-polymerase chain reaction and immunohistochemical analyses, respectively. The p16(INK4a) mRNA levels were low, and in some cases undetectable, in control colon tissue. However, colon tumors exhibited an eightfold increase in p16(INK4a) mRNA level when compared with control colon tissue (P < 0.01). Whereas control colon epithelium was uniformly negative for p16(INK4a) immunoreactivity, p16(INK4a)-immunoreactive cells were markedly increased in preneoplastic lesions and adenomas isolated from AOM-treated mice. To further examine the p16(INK4a) regulatory pathway, the retinoblastoma tumor-suppressor protein (Rb) was also examined immunohistochemically in these tissues. A heterogeneous Rb immunostaining was observed in preneoplastic lesions and adenomas. Immunohistochemical analysis also showed a reciprocal relationship between p16(INK4a) and Rb protein expression. These findings suggest that alterations in the p16(INK4a)/Rb pathway may play an important role in AOM-induced mouse colon tumorigenesis. Mol. Carcinog. 28:139-147, 2000.


Assuntos
Adenocarcinoma/genética , Azoximetano/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/genética , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Deleção de Genes , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes p16/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Adenocarcinoma/química , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenoma/induzido quimicamente , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Azoximetano/farmacologia , Carcinógenos/farmacologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Genes do Retinoblastoma/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas de Neoplasias/genética , Reação em Cadeia da Polimerase , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Proteína do Retinoblastoma/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
Int J Oncol ; 17(3): 551-8, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10938397

RESUMO

Alterations in transforming growth factor beta1 (TGF-beta1) and its type II receptor (TbetaR-II) have been implicated in the pathogenesis of a variety of human cancers and animal tumor models. We postulated that TGF-beta1 and TbetaR-II alterations may also be involved in mouse colon tumorigenesis induced by the chemical carcinogen, azoxymethane (AOM). In the present study, normal colon tissues and AOM-induced colon tumors from SWR/J mice were analyzed for mutational changes in the TbetaR-II gene, and the expression and localization of TGF-beta1 and TbetaR-II were examined by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemisty. Direct DNA sequencing of the coding sequence of the TbetaR-II gene revealed no mutational changes in tumors induced by AOM when compared with the sequence from normal colon tissue. However, TGF-beta1 and TbetaR-II mRNA levels in tumor samples were increased 1.8-fold (p<0.01) and 1.3-fold (p<0.01), respectively, when compared with control mouse colon tissue. The results of immunohistochemical analysis of TGF-beta1 and TbetaR-II were correlated with mRNA expression data. An increase in staining intensity of both TGF-beta and TbetaR-II were observed in colon tumors. These findings suggest that alterations in the expression of TGF-beta1 and TbetaR-II may be involved in the pathogenesis of colon tumors induced by AOM in mice.


Assuntos
Adenocarcinoma/metabolismo , Azoximetano/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/biossíntese , Receptores de Fatores de Crescimento Transformadores beta/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Adenocarcinoma/induzido quimicamente , Adenocarcinoma/genética , Substituição de Aminoácidos , Animais , Códon/genética , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Análise Mutacional de DNA , DNA de Neoplasias/genética , Masculino , Camundongos , Proteínas de Neoplasias/genética , Mutação Puntual , Proteínas Serina-Treonina Quinases , RNA Mensageiro/biossíntese , RNA Neoplásico/biossíntese , Receptor do Fator de Crescimento Transformador beta Tipo II , Receptores de Fatores de Crescimento Transformadores beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta1
9.
Carcinogenesis ; 21(8): 1567-72, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10910960

RESUMO

Aberrant crypt foci (ACF), putative preneoplastic lesions, are early morphological changes induced by the colon carcinogen azoxymethane (AOM). Although inbred mice differ markedly in their susceptibility to AOM carcinogenesis, we have previously shown that ACF develop in both resistant and sensitive mouse strains after AOM treatment. The purpose of this study was to examine the sequential development and identify the morphological characteristics of ACF induced by AOM in the distal colon of sensitive and resistant mice. A/J (highly susceptible), SWR/J (relatively susceptible) and AKR/J (resistant) mice were treated with 10 mg/kg AOM or saline i.p. once a week for 6 weeks and were killed at 1, 2, 4, 6, 9 and 24 weeks after the last injection. The distal colons were stained with methylene blue and the numbers of ACF and tumors determined. Tumors were present as early as 4 weeks after AOM exposure in SWR/J and A/J mice and increased in frequency throughout the study in both strains. No tumors developed in the AKR/J mice. ACF, however, formed in all strains of mice. The greatest difference between susceptible and resistant strains was in the number of large ACF that developed at later time points. Furthermore, morphometric analysis revealed that A/J mice had the highest percentage of dysplastic ACF, followed by SWR/J mice. These data indicate that the difference in cancer risk from AOM may be due to the lack of progression of smaller ACF in the resistant mice and to the development of dysplasia in a higher percentage of ACF from susceptible strains.


Assuntos
Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/patologia , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/patologia , Animais , Suscetibilidade a Doenças , Resistência a Medicamentos , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos AKR , Especificidade da Espécie
10.
Chem Biol Interact ; 124(3): 149-60, 2000 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-10728775

RESUMO

Azoxymethane (AOM) is an indirect-acting colon carcinogen that produces a high incidence of precancerous lesions, referred to as aberrant crypt foci (ACF), in rats. This study was undertaken to determine whether high dose gavage administration of the cytochrome P-450 2E1 (CYP2E1) inhibitor and chemopreventive agent, diallyl sulfide, would reduce the incidence and severity of ACF formation in the distal colons of AOM-treated Fischer 344 rats. Seven-week-old male rats received 150 or 50 mg/kg diallyl sulfide by gavage 24 and 2 h prior to two weekly i.p. injections of AOM (20 mg/kg). Ten weeks after the last injection of AOM the rats were sacrificed and the colons removed and stained with 0.2% methylene blue. ACF were visualized using stereomicroscopy. Rats pretreated with diallyl sulfide exhibited a significant increase in the number of ACF/cm in the distal colon compared with rats receiving AOM alone. This increase in ACF number was seen in ACF of all sizes. To examine the effects of diallyl sulfide on the initiation stage of AOM-induced carcinogenesis, mutations in the K-ras proto-oncogene were also investigated. ACF and normal appearing colonic mucosa (0.2-0.5 mm3) were microdissected for subsequent PCR-RFLP analysis of a codon 12 (GGT-GGA) activating mutation in the K-ras gene. Greater than 90% of ACF from AOM-treated animals, regardless of diallyl sulfide treatment, exhibited activating K-ras mutations. K-ras mutations were also detected in normal appearing mucosa of AOM-treated animals, although at a lesser frequency (15-35%). These studies demonstrate that diallyl sulfide given in large gavage doses enhances AOM-induced preneoplasia in rats and suggests that diallyl sulfide may alter the disposition of AOM intermediates and/or enhance colonic promotional activity in the rat.


Assuntos
Compostos Alílicos/toxicidade , Anticarcinógenos/toxicidade , Compostos Azo/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/induzido quimicamente , Lesões Pré-Cancerosas/induzido quimicamente , Sulfetos/toxicidade , Compostos Alílicos/uso terapêutico , Animais , Anticarcinógenos/uso terapêutico , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Neoplasias do Colo/prevenção & controle , Inibidores do Citocromo P-450 CYP2E1 , Sinergismo Farmacológico , Inibidores Enzimáticos/toxicidade , Genes ras/efeitos dos fármacos , Genes ras/genética , Masculino , Mutação , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Lesões Pré-Cancerosas/enzimologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/prevenção & controle , Ratos , Ratos Endogâmicos F344 , Sulfetos/uso terapêutico
11.
Gastroenterology ; 118(4): 724-34, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10734024

RESUMO

BACKGROUND & AIMS: The transcription factor nuclear factor-kappaB (NF-kappaB) plays a central role in regulating immune and inflammatory responses. Because butyrate deficiency has been associated with inflammatory bowel disease, we examined the effect of butyrate on NF-kappaB activity in the human HT-29 colonic cell line. METHODS: The influence of butyrate (4 mmol/L) on NF-kappaB activity was determined using the gel mobility shift assay. The effect of butyrate on the expression of NF-kappaB subunits and inhibitory proteins was determined by immunoblotting. NF-kappaB-regulated gene expression was assayed by primer extension of intercellular adhesion molecule 1 and Mn superoxide dismutase messenger RNA, and by analysis of a transfected luciferase reporter. RESULTS: Exposure of HT-29 cells to butyrate eliminated their constitutive NF-kappaB, p50 dimer activity. This inhibition corresponded with a reduction in p50 nuclear localization, without a reduction in expression. Butyrate also selectively modulated activation of NF-kappaB, suppressing its activation by tumor necrosis factor alpha and phorbol ester more than 10-fold, without affecting the activity induced by interleukin (IL)-1beta. Butyrate did, however, enhance formation of the stronger p65-p50 transcriptional activator in IL-1beta-stimulated cells. The changes in NF-kappaB activation did not correlate with changes in IkappaBalpha levels. Gene expression reflected DNA binding. The influence of butyrate on NF-kappaB may result in part from its ability to inhibit deacetylases because the specific deacetylase inhibitor trichostatin A has a similar effect. CONCLUSIONS: These findings suggest that the influences of butyrate on colonic inflammatory responses may result in part from its influence on NF-kappaB activation. This activity of butyrate apparently involves its ability to inhibit deacetylases.


Assuntos
Butiratos/farmacologia , Colo/metabolismo , Mucosa Intestinal/metabolismo , NF-kappa B/metabolismo , Acetilação , Linhagem Celular , Colo/citologia , Citocinas/fisiologia , Dimerização , Regulação da Expressão Gênica/fisiologia , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/efeitos dos fármacos
12.
Mol Carcinog ; 27(3): 210-8, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10708483

RESUMO

A differential susceptibility phenotype to the organotropic colon carcinogen azoxymethane (AOM) has been described in mice. The following studies were undertaken to test the hypothesis that intraspecific susceptibility can be accounted for by the specific complement of genetic alterations acquired by precancerous colon lesions referred to as aberrant crypt foci (ACF). As an initial approach to this question, mutations in codons 12 and 13 of the Ki-ras proto-oncogene were assessed in ACF, normal-appearing AOM-treated colonic epithelium, and tumors from A/J and SWR/J (susceptible) as well as AKR/J (resistant) mice. Four-week-old male mice were injected intraperitonealy, with AOM once a week for a total of 6 wk and killed 4 and 24 wk after the last injection. DNA was isolated from microdissected tissue, and polymerase chain reaction (PCR)-amplified products of Ki-ras exon 1 (codons 12 and 13) were directly sequenced from microdissected tissues. At 4 wk after AOM exposure, there was no significant difference in the frequency of Ki-ras activation (20-33%) between the three strains. Ki-ras mRNA expression was also evaluated by reverse transcription (RT)-PCR analysis and was comparably reduced (40-50%) in all three strains at the 4 wk time point. However, Ki-ras expression returned to normal by 24 wk after treatment. Finally, to gain further insight into the molecular pathogenesis underlying this experimental tumor model, analysis of the adenomatous polyposis coli (APC) protein within the colonic epithelium was undertaken by using an immunohistochemical approach. Although the APC protein was lost to a varying extent in tumors from A/J and SWR/J mice, the full-length form of the protein was still present in precancerous ACF isolated from each of the three strains, regardless of the degree of dysplasia of the lesion. A further molecular genetic analyses of ACF will be required to gain a more complete understanding of the molecular basis of tumor susceptibility phenotype in this murine model.


Assuntos
Azoximetano/farmacologia , Colo/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Genes ras , Animais , Sequência de Bases , Colo/metabolismo , Primers do DNA , Éxons , Imuno-Histoquímica , Masculino , Camundongos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Carcinogenesis ; 21(2): 133-8, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10657948

RESUMO

The murine non-pancreatic secretory phospholipase A(2) (sPLA(2)) has been proposed as a tumor modifier of multiple intestinal neoplasia (Min). A genetic polymorphism in the mouse gene that causes a disruption in exon 3 results in loss of functional protein. Mouse strains with a disrupted sPLA(2) gene are susceptible to the Min phenotype and develop numerous intestinal polyps, whereas mice with normal sPLA(2) develop only a limited number of polyps. The following study was undertaken to test the hypothesis that sPLA(2) plays an equivalent role in murine susceptibility to the colon carcinogen azoxymethane (AOM). sPLA(2) status was confirmed by sequencing in mice that are highly susceptible (A/J), susceptible (SWR/J) and resistant (AKR/J) to AOM-induced tumorigenesis. Constitutive expression of sPLA(2) mRNA was compared in small intestine and colon of untreated mice using semi-quantitative RT-PCR. Whereas mRNA expression was nearly absent in A/J mice, AKR/J mice exhibited extensive expression throughout the intestine. Despite the wild-type sPLA(2) gene, colonic mRNA expression in SWR/J mice was significantly lower relative to AKR/J. Immunohistochemical analysis of sPLA(2) protein confirmed the mRNA data. The effect of AOM on colonic sPLA(2) expression was also examined. Twenty-four weeks after the last of six weekly injections of AOM (10 mg/kg i.p.), RT-PCR analysis of distal colons revealed a significant increase in mRNA in normal-appearing epithelium and tumor tissue from AOM-treated mice relative to controls. However, there was no corresponding increase in protein expression in A/J mice. The absence of sPLA(2) expression within control colons of tumor-susceptible A/J mice together with low expression in SWR/J colons is consistent with its potential role as an intestinal tumor modifier, but the carcinogen-induced increase in expression raises doubts as to the significance of sPLA(2) in inhibiting carcinogenesis.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/genética , Pólipos do Colo/genética , Intestinos/enzimologia , Fosfolipases A/biossíntese , Animais , Colo/enzimologia , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/enzimologia , Pólipos do Colo/induzido quimicamente , Pólipos do Colo/enzimologia , Éxons/genética , Predisposição Genética para Doença , Fosfolipases A2 do Grupo II , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos AKR , Camundongos Endogâmicos , Fenótipo , Fosfolipases A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Carcinogenesis ; 20(4): 691-7, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10223201

RESUMO

Inbred mice exhibit differential susceptibility to colon carcinogens. The following study addresses the possibility that differences are intrinsic to colonic mucosa (cell autonomous) or are mediated by extracolonic systemic factors (e.g. liver activation of carcinogens). Our approach was to construct mouse aggregation chimeras, mice whose tissues are a mosaic of cells derived from two parental genotypes, from a susceptible (SWR) and a resistant (DBA/2) strain. Forty-five embryo aggregations yielded 11 viable pups, four of which were chimeric by coat color. Six-week-old SWR<-->BA/2 chimeras were injected i.p. with azoxymethane (AOM) once a week for 8 weeks (5 and 7.5 mg/kg body wt for 2 weeks followed by 10 mg/kg for 6 weeks) and tumor incidence in distal colon was evaluated 15 weeks after the last injection. Additional groups of parental mice received the same treatment. In the parental SWR treatment group, 1.7 +/- 0.82 tumors/colon were found. No tumors were observed in AOM-treated DBA/2 mice. In SWR<-->DBA/2 chimeras exposed to AOM, 2.8 +/- 2.1 tumors/colon were found. Tumor lineage was examined in paraffin sections stained with Dolichos biflorus agglutinin-peroxidase, a cell surface specific marker that stains intestinal endothelial cells of SWR and epithelial cells of DBA/2. Cellular lineage of tumors was further evaluated by microsatellite analysis of DNA isolated by microdissection. There was no significant difference in tumor incidence between SWR parental and chimera treatment groups. Histochemical analysis of tumor tissue in chimeras suggested that most tumors were derived from SWR. However, subsequent genetic analysis of tumors indicated mixed parental composition. These preliminary studies suggest that DBA/2 resistance mechanisms are not sufficient to protect adjacent SWR-derived epithelium from the tumorigenic effects of AOM.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Colo/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Mucosa Intestinal/efeitos dos fármacos , Camundongos Endogâmicos/genética , Animais , Azoximetano/administração & dosagem , Biomarcadores , Carcinógenos/administração & dosagem , Linhagem da Célula , Quimera , Células Clonais/patologia , Colo/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , DNA de Neoplasias/genética , Resistência a Medicamentos/genética , Predisposição Genética para Doença , Genótipo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos DBA , Repetições de Microssatélites
15.
Exp Mol Pathol ; 65(3): 141-9, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10234360

RESUMO

Heritable differences in tumor susceptibility are observed in mice after repetitive exposures to the organotropic colon carcinogen azoxymethane (AOM). The following study was undertaken to determine whether early morphological alterations within the colonic epithelium correlate with subsequent cancer risk. A/J and SWR/J (susceptible) and AKR/J (resistant) mice were injected once a week with AOM at a dose of 10 mg/kg, i.p., for a total of 6 weeks. Four weeks after the last injection, methylene blue-stained whole-mount colons were examined for the presence of colonic epithelial lesions referred to as aberrant crypt foci (ACF). Putative lesions identified under low magnification were further characterized by H&E staining of corresponding sections. AOM produced a treatment-related increase in ACFs in each of the mouse lines examined. The tumor-susceptible SWR/J and A/J mice developed on average between three- and sixfold more ACFs in the distal colon (32 and 15/cm of colon, respectively) than the resistant AKR/J mice (5/cm colon). The size distribution of ACFs was further analyzed in each of the strains. In SWR/J and A/J, 20-35% of lesions were classified as large ACFs, consisting of 5 or more aberrant crypts per focus. This is in striking contrast to the size distribution of lesions identified in the AKR/J colons, where fewer than 5% of grossly identified lesions were classified as large. In fact, the majority (> 80%) of ACFs in AKR/J mice consisted of only 1-2 aberrant crypts@focus. In addition, there was no evidence of dysplasia in any of the AKR/J lesions examined, whereas the lesions in susceptible mice were dysplastic (adenomas). Our data indicate that tumorigenic response is associated with the extent and multiplicity of ACFs that form within the colonic epithelium at an early time point after carcinogen exposure. These studies further support the use of this morphological biomarker as a short-term endpoint of colon tumorigenesis.


Assuntos
Azoximetano , Carcinógenos , Colo/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Animais , Colo/patologia , Neoplasias do Colo/patologia , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos , Lesões Pré-Cancerosas/induzido quimicamente
16.
Carcinogenesis ; 19(11): 2001-6, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9855016

RESUMO

Alterations in the expression of the cell cycle regulators, cyclin D1 and cyclin-dependent kinase 4 (Cdk4), have been implicated in malignancies of both humans and experimental animal models. We hypothesize that altered expression of cyclin D1 and Cdk4 may also be involved in mouse colon tumorigenesis induced by the chemical carcinogen, azoxymethane (AOM). In the present study, SWR/J mice were given AOM by i.p. injection at a dose of 10 mg/kg once a week for 8 weeks, and colonic tissue and tumors were isolated 18 weeks later. The expression and localization of cyclin D1 and Cdk4 were examined by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemical analyses. Cyclin D1 and Cdk4 mRNA levels in tumor samples were increased 1.3-fold (P < 0.01) and 1.2-fold (P < 0.01), respectively, when compared with control mouse colon tissue. Control colon epithelium was uniformly negative for cyclin D1 immunoreactivity, whereas minimal Cdk4 nuclear staining was confined to the lower portion of the crypts within the control tissue. Both cyclin D1 and Cdk4 immunoreactive cells were markedly increased in preneoplastic lesions and in adenomas isolated from AOM-treated mice. Furthermore, some morphologically normal colon crypts from AOM-treated mice showed positive cyclin D1 immunoreactivity. These findings suggest that overexpression of cyclin D1 and Cdk4 occurs early in the AOM-induced mouse colon tumorigenesis and may contribute to tumor progression in this model.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Neoplasias do Colo/induzido quimicamente , Ciclina D1/análise , Quinases Ciclina-Dependentes/análise , Proteínas Proto-Oncogênicas , Animais , Colo/efeitos dos fármacos , Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Ciclina D1/genética , Quinase 4 Dependente de Ciclina , Quinases Ciclina-Dependentes/genética , Genes ras , Imuno-Histoquímica , Masculino , Camundongos , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
17.
Toxicol Sci ; 45(1): 66-71, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9848112

RESUMO

The aim of this study was to determine whether the cytosolic enzyme alcohol dehydrogenase (ADH) activates methylazoxymethanol (MAM) in the mouse colon and whether differential tumor susceptibility in the mouse is dependent, in part, on strain-related differences in MAM metabolism by ADH. Liver and colon cytosols were isolated from 7-week-old male tumor-susceptible (SWR/J) and -resistant (AKR/J) mice. Minimal reduction of NAD+ was found in colon cytosols from AKR/J mice at the highest concentration (2 mM) of MAM tested. In liver cytosols, only SWR was capable of sustaining NAD+ reduction with MAM, although at very low levels. Despite minimal reactivity with MAM, however, mouse cytosols did effectively reduce NAD+ in the presence of the common ADH subrates ethanol and benzyl alcohol. NAD(+)-coupled oxidation of benzyl alcohol was significantly higher (two- to three-fold, p < 0.05) in mouse colon cytosols compared to activity present within corresponding rat tissues. Incubation of colon and liver cytosols with the ADH-3 inhibitor 4-methylpyrazole markedly (95-100% of controls) reduced ethanol oxidation in both strains. However, 4-methylpyrazole was a less effective inhibitor of benzyl alcohol oxidation in AKR/J colons, suggesting a different ADH isoform complement. An opposite inhibition pattern of benzyl alcohol oxidation was seen in the liver, where 4-methylpyrazole produced a greater inhibition in SWR/J mice. These studies suggest that the metabolism of the proximate mutagen MAM occurs by processes in the mouse that are independent of ADH.


Assuntos
Álcool Desidrogenase/metabolismo , Carcinógenos/metabolismo , Colo/metabolismo , Fígado/metabolismo , Acetato de Metilazoximetanol/análogos & derivados , Álcool Desidrogenase/antagonistas & inibidores , Álcoois/metabolismo , Animais , Fomepizol , Masculino , Acetato de Metilazoximetanol/metabolismo , Camundongos , Camundongos Endogâmicos AKR , Pirazóis/farmacologia , Ratos
18.
Cancer Lett ; 130(1-2): 29-34, 1998 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-9751253

RESUMO

Azoxymethane (AOM) is an organotropic colon carcinogen that is commonly used to induce colon tumors in rodents. Unlike its parent compound, 1,2-dimethylhydrazine (DMH), a tumor susceptibility phenotype in inbred mice with respect to AOM has not been established. Thus, this study was undertaken to determine whether genetic susceptibility extends to this carcinogen. SWR/J, A/J (both susceptible to DMH carcinogenesis) and AKR/J (resistant) mice were treated with 10 mg/kg AOM i.p. once a week for 8 weeks. Twenty-five weeks after the initial injection, tumor yield was determined. With a single exception, only SWR/J and A/J mice developed tumors, with a distribution that was limited to the distal colon (16.3+/-1.1 and 36.4+/-2.4. respectively). The formation of aberrant crypt foci (ACF), putative preneoplastic lesions, was also assessed in whole-mount colons using Methylene Blue staining. Consistent with tumor multiplicity, the total number of ACF was highest in A/J mice, followed by SWR/J mice. In addition, A/J mice had a significantly greater number of large ACF (five or more crypts per foci) than the other strains. Despite the absence of colon tumors, however, AKR/J mice did develop a significant number of ACF. This finding suggests that ACF in resistant mice are persistent but do not progress to tumors.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Colo/efeitos dos fármacos , Neoplasias do Colo/induzido quimicamente , Camundongos Endogâmicos , Lesões Pré-Cancerosas/induzido quimicamente , Animais , Colo/patologia , Suscetibilidade a Doenças , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos AKR , Fenótipo , Especificidade da Espécie
19.
Toxicol Appl Pharmacol ; 150(1): 196-203, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9630469

RESUMO

Inbred mice vary in susceptibility to colon carcinogens such as 1,2-dimethylhydrazine (DMH). Differential susceptibility may depend, in part, on formation of promutagenic DNA methyl adducts within target colonic mucosa. The present study was undertaken to evaluate the extent of DNA adduct formation in susceptible (SWR) and resistant (AKR) mice acutely exposed to the colon carcinogen azoxymethane (AOM), a direct metabolite of DMH. In the first experiment, 8-week-old SWR and AKR mice were treated i.p. with 20 mg/kg AOM and sacrificed 6 h later. DNA was isolated from distal colon and liver, and O6-methylguanine (O6-MeGua) adduct levels were assessed by immunoslot blot (ISB) analysis, using a monospecific antibody raised against O6-methyldeoxyguanosine. HPLC-fluorescence detection was also used to quantitate 06-MeGua and 7-methylguanine (7-MeGua), and to generate standard curves. At 6 h, both O6-MeGua and 7-MeGua were significantly higher (2- to 3-fold, p < 0.05) in AKR colon, while an opposite pattern was found in liver. In Experiment 2, mice were injected with AOM (20 mg/kg) and euthanized 12 and 48 h later. At 12 h, O6-MeGua levels were higher in colons (1.4-fold) of SWR mice. Forty-eight hours after treatment, however, adduct levels in colon were markedly (5-fold) reduced in SWR but were unchanged from 12 h in AKR. To further compare activation of AOM in both strains, colon microsomes were incubated with AOM and calf thymus DNA. Comparable levels of O6-MeGua were detected by ISB, demonstrating equivalent metabolic capacity in both SWR and AKR mice. These studies suggest that differential susceptibility to AOM-induced colon carcinogenesis is not based on initial target tissue DNA alkylation and unlikely to depend on differential metabolic capacity.


Assuntos
Azoximetano/toxicidade , Carcinógenos/toxicidade , Adutos de DNA/metabolismo , Neoplasias Experimentais/induzido quimicamente , Animais , Biotransformação , Carcinógenos/farmacologia , Bovinos , Cromatografia Líquida de Alta Pressão , DNA/análise , DNA/efeitos dos fármacos , DNA/isolamento & purificação , Adutos de DNA/efeitos dos fármacos , Guanina/análogos & derivados , Guanina/metabolismo , Immunoblotting , Masculino , Metilnitrosoureia/farmacologia , Camundongos , Camundongos Endogâmicos AKR , Camundongos Endogâmicos , Valor Preditivo dos Testes
20.
Drug Metab Dispos ; 24(4): 408-13, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8801055

RESUMO

1,2-Dimethylhydrazine (DMH) is an organotropic colon carcinogen that undergoes metabolic activation to DNA-reactive metabolites. Twenty hours after parenteral treatment of AKR/J (colon tumor resistant) and SWR/J (susceptible) mice with DMH.2HCl (70 mg/kg), functional levels of Cyp1a1 and Cyp2e1 were examined by measuring O-deethylation of ethoxyresorufin (EROD) and hydroxylation of p-nitrophenol, respectively. In control animals, SWR/J mice exhibited higher hepatic EROD activity (1.4-fold) when compared with AKR/J mice. In carcinogen-treated animals, EROD activity was decreased 20-30% in both mouse lines. Hepatic p-nitrophenol hydroxylase activity, similar in control animals of both strains, was reduced comparably (45-50% of control) after DMH administration. In liver, a decrease in immunoreactive Cyp2e1 protein paralleled the decline in enzyme activity, whereas in the colon, no significant treatment-related differences were detected in either strain. In liver and colon cytosols, alcohol dehydrogenase activity was not significantly different in either mouse line, both in control and DMH-treated animals. Glutathione levels were elevated (1.7-fold) in livers of AKR/J mice after DMH administration. Total glutathione-S-transferase (GST) activity was significantly increased (1.8-fold) in the colons of SWR/J mice and in the livers (1.4-fold) of AKR/J mice. Furthermore, the GST isoform, GST-Yp, was reduced 40% in the SWR/J colon. These data demonstrate the importance of metabolic capacity as a factor in conferring differential tumor susceptibility in a murine cancer model to the indirect-acting colon carcinogen, DMH.


Assuntos
Carcinógenos/administração & dosagem , Colo/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Dimetilidrazinas/administração & dosagem , Isoenzimas/metabolismo , Fígado/efeitos dos fármacos , 1,2-Dimetilidrazina , Álcool Desidrogenase/genética , Álcool Desidrogenase/metabolismo , Animais , Biotransformação , Colo/enzimologia , Citocromo P-450 CYP2E1 , Sistema Enzimático do Citocromo P-450/genética , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Isoenzimas/genética , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos AKR , Oxirredutases N-Desmetilantes/genética , Oxirredutases N-Desmetilantes/metabolismo , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...