Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Asian Pac J Cancer Prev ; 22(3): 861-869, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33773551

RESUMO

OBJECTIVE: The aim of the study was to develop a model for predicting cancer risk in colorectal polyps' patients (CPPs), as well as to reveal additional prognosis factors for Stage III colorectal cancer based on differences in subpopulations of tetraspanins, tetraspanin-associated and tetraspanin-non-associated proteases in blood plasma exosomes of CPPs and colorectal cancer patients (CRCPs). METHODS: The subpopulations of CD151- and Tspan8-positive exosomes, the subpopulations of metalloproteinase at the surface of СD9-positive exosomes and the level of 20S proteasomes in plasma exosomes in 15 CPPs (tubulovillous adenomas) and 60 CRCPs were evaluated using flow cytometry and Western blotting. Logistic regression analysis was performed to predict cancer risk of CPPs. RESULTS: The levels of 20S proteasomes in exosomes, MMP9+, MMP9+/MMP2+/EMMPRIN+ in CD9-positive blood plasma exosomes are associated with the risk of malignant transformation of colorectal tubulovillous adenomas.  In patients with Stage III CRC, the levels of 20S proteasomes (less than 2 units) and MMP9+ subpopulations (more than 61%) in plasma exosomes are unfavorable prognostic factors for overall survival. The levels of 20S proteasomes and ADAM10+/ADAM17- subpopulations in CD9-positive blood plasma exosomes are the most significant values for predicting relapse-free survival. CONCLUSION: Protease cargo in CD9-positive blood plasma exosomes is prognostic biomarker for colorectal polyps and colorectal cancer.


Assuntos
Adenoma/enzimologia , Carcinoma/enzimologia , Pólipos do Colo/enzimologia , Neoplasias Colorretais/enzimologia , Exossomos/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Adenoma/metabolismo , Adenoma/patologia , Adenoma Viloso/enzimologia , Adenoma Viloso/metabolismo , Adenoma Viloso/patologia , Basigina/metabolismo , Carcinoma/metabolismo , Carcinoma/patologia , Pólipos do Colo/metabolismo , Pólipos do Colo/patologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Exossomos/metabolismo , Feminino , Humanos , Pólipos Intestinais/enzimologia , Pólipos Intestinais/metabolismo , Pólipos Intestinais/patologia , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Peptídeo Hidrolases/metabolismo , Prognóstico , Tetraspanina 24/metabolismo , Tetraspaninas/metabolismo
2.
Int Immunopharmacol ; 90: 107128, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33191180

RESUMO

Sunitinib is a tyrosine kinase inhibitor for many tumors. Inflammation is one of the most important factors in the development of intestinal tumors. Many inflammation-related factors are regulated by tyrosine kinase receptors. It is reasonable to hypothesize that sunitinib can regulate the development of intestinal tumors by regulating the expression and/or activity of inflammation-related factors. Here, ApcMin/+ male mouse model was used to investigate the effect and mechanism of sunitinib malate against intestinal cancer. Results show that compared to vehicle, after sunitinib malate treatment, overall survival of ApcMin/+ mice was lengthened up to 25 days, with a gain of body weight, reduction of spleen/body weight index, and RBC, WBC and HGC regulated to normal levels of wild type mice, and a number of polyps no less than 1 mm significantly reduced. Meanwhile, in the intestines, the nuclear ß-Catenin protein and c-Myc mRNA were both down-regulated, and Bcl-6 was significantly reduced with Caspase-3 up regulated. Furthermore, inflammation-related factors including IL-6, TNF-α, IL-1α, IL-1ß and IFN-γ were down-regulated at mRNA levels in the intestines. These results suggest that sunitinib malate can significantly improve the survival status and inhibit intestinal tumor development in male ApcMin/+ mice, through inhibiting inflammation-related factors, while suppressing ß-cateinin/c-Myc pathway and re-balancing protein levels of Bcl-6 and Caspase-3.


Assuntos
Anti-Inflamatórios/farmacologia , Anticarcinógenos/farmacologia , Caspase 3/metabolismo , Colo/efeitos dos fármacos , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Neoplasias Intestinais/prevenção & controle , Pólipos Intestinais/prevenção & controle , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Sunitinibe/farmacologia , beta Catenina/metabolismo , Animais , Colo/enzimologia , Colo/patologia , Citocinas/genética , Regulação da Expressão Gênica , Genes APC , Neoplasias Intestinais/enzimologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Pólipos Intestinais/enzimologia , Pólipos Intestinais/genética , Pólipos Intestinais/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais
3.
Curr Cancer Drug Targets ; 18(9): 905-911, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-28786349

RESUMO

BACKGROUND: Recent epidemiological and experimental studies have shown that obesity is a major risk factor for Colorectal Cancer (CRC). Regular intake of high fat-containing diet can promote obesity and metabolic syndrome by increasing the insulin resistance and inflammatory response which contribute to carcinogenesis. Previously, we have shown that inhibition of polyol pathway enzyme aldose reductase (AR) prevents carcinogens- and inflammatory growth factorsinduced CRC. However, the effect of AR inhibition on a high-fat diet (HFD)-induced formation of intestinal polyps in Apc-deficient Min (multiple intestinal neoplasia; ApcMin/+) mice is not known. METHODS: We examined the effect of AR inhibitor, fidarestat on the HFD-induced formation of preneoplastic intestinal polyps in ApcMin/+ mice which is an excellent model of colon cancer. RESULTS: APCMin/+ mice fed for 12 weeks of HFD caused a significant increase in the formation of polyps in the small and large intestines and fidarestat given along with the HFD prevented the number of intestinal polyps. Fidarestat also decreased the size of the polyps in the intestines of HFDtreated APC Min mice. Further, the expression levels of beta-catenin, PCNA, PKC-ß2, P-AKT, Pp65, COX-2, and iNOS in the small and large intestines of HFD-treated mice significantly increased, and AR inhibitor prevented it. CONCLUSION: Our results thus suggest that fidarestat could be used as a potential chemopreventive drug for intestinal cancers due to APC gene mutations.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Aldeído Redutase/antagonistas & inibidores , Neoplasias do Colo/prevenção & controle , Dieta Hiperlipídica/efeitos adversos , Inibidores Enzimáticos/farmacologia , Imidazolidinas/farmacologia , Pólipos Intestinais/prevenção & controle , Animais , Neoplasias do Colo/enzimologia , Neoplasias do Colo/etiologia , Feminino , Pólipos Intestinais/enzimologia , Pólipos Intestinais/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
4.
World J Gastroenterol ; 22(11): 3261-7, 2016 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-27004004

RESUMO

AIM: To investigate multiple polyps in a Chinese Peutz-Jeghers syndrome (PJS) infant. METHODS: A nine-month-old PJS infant was admitted to our hospital for recurrent prolapsed rectal polyps for one month. The clinical characteristics, a colonoscopic image, the pathological characteristics of the polyps and X-ray images of the intestinal perforation were obtained. Serine threonine-protein kinase 11 (STK11) gene analysis was also performed using a DNA sample from this infant. RESULTS: Here we describe the youngest known Chinese infant with PJS. Five polyps, including a giant polyp of approximately 4 cm × 2 cm in size, were removed from the infant's intestine. Laparotomy was performed to repair a perforation caused by pneumoperitoneum. The pathological results showed that this child had PJS. Molecular analysis of the STK11 gene further revealed a novel frameshift mutation (c.64_65het_delAT) in exon 1 in this PJS infant. CONCLUSION: The appropriate treatment method for multiple polyps in an infant must be carefully considered. Our results also show that the STK11 gene mutation is the primary cause of PJS.


Assuntos
Colonoscopia , Mutação da Fase de Leitura , Pólipos Intestinais/genética , Pólipos Intestinais/cirurgia , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/cirurgia , Proteínas Serina-Treonina Quinases/genética , Neoplasias Retais/genética , Neoplasias Retais/cirurgia , Quinases Proteína-Quinases Ativadas por AMP , Biópsia , Análise Mutacional de DNA , Feminino , Marcadores Genéticos , Predisposição Genética para Doença , Humanos , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Síndrome de Peutz-Jeghers/enzimologia , Síndrome de Peutz-Jeghers/patologia , Fenótipo , Neoplasias Retais/enzimologia , Neoplasias Retais/patologia , Resultado do Tratamento , Carga Tumoral
5.
Cancer Sci ; 106(6): 692-699, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25855137

RESUMO

Extracellular signal-regulated kinase is an MAPK that is most closely associated with cell proliferation, and the MEK/ERK signaling pathway is implicated in various human cancers. Although epidermal growth factor receptor, KRAS, and BRAF are considered major targets for colon cancer treatment, the precise roles of the MEK/ERK pathway, one of their major downstream effectors, during colon cancer development remain to be determined. Using Apc(Δ716) mice, a mouse model of familial adenomatous polyposis and early-stage sporadic colon cancer formation, we show that MEK/ERK signaling is activated not only in adenoma epithelial cells, but also in tumor stromal cells including fibroblasts and vascular endothelial cells. Eight-week treatment of Apc(Δ716) mice with trametinib, a small-molecule MEK inhibitor, significantly reduced the number of polyps in the large size class, accompanied by reduced angiogenesis and tumor cell proliferation. Trametinib treatment reduced the COX-2 level in Apc(Δ716) tumors in vivo and in primary culture of intestinal fibroblasts in vitro. Antibody array analysis revealed that trametinib and the COX-2 inhibitor rofecoxib both reduced the level of CCL2, a chemokine known to be essential for the growth of Apc mutant polyps, in intestinal fibroblasts in vitro. Consistently, trametinib treatment reduced the Ccl2 mRNA level in Apc(Δ716) tumors in vivo. These results suggest that MEK/ERK signaling plays key roles in intestinal adenoma formation in Apc(Δ716) mice, at least in part, through COX-2 induction in tumor stromal cells.


Assuntos
Antineoplásicos/farmacologia , Ciclo-Oxigenase 2/análise , Genes APC/fisiologia , Pólipos Intestinais/tratamento farmacológico , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridonas/farmacologia , Pirimidinonas/farmacologia , Animais , Quimiocina CCL2/análise , Quimiocina CCL2/genética , Modelos Animais de Doenças , Feminino , Pólipos Intestinais/enzimologia , Masculino , Camundongos
6.
PLoS One ; 7(3): e33243, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22432006

RESUMO

BACKGROUND: Mutation of tumor suppressor gene, adenomatous polyposis coli (APC), is the primary molecular event in the development of most intestinal carcinomas. Animal model with APC gene mutation is an effective tool for study of preventive approaches against intestinal carcinomas. We aimed to evaluate the effect of Riccardin D, a macrocyclic bisbibenzyl compound, as a chemopreventive agent against intestinal adenoma formation in APC(Min/+) mice. METHODS: APC(Min/+) mice were given Riccardin D by p.o. gavage for 7 weeks. Mice were sacrificed, and the number, size and histopathology of intestinal polyps were examined under a microscope. We performed immunohistochemical staining, western blotting, reverse transcriptase-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) in intestinal polyps to investigate the mechanism of chemopreventive effect of Riccardin D. RESULTS: Riccardin D treatment resulted in a significant inhibition of intestinal adenoma formation, showing a reduction of polyp number by 41.7%, 31.1% and 44.4%, respectively, in proximal, middle and distal portions of small intestine. The activity of Riccardin D against polyp formation was more profound in colon, wherein Riccardin D decreased polyp number by 79.3%. Size distribution analysis revealed a significant reduction in large-size polyps (2-3 mm) by 40.0%, 42.5% and 33.3%, respectively, in proximal, middle and distal portions of small intestine, and 77.8% in colon. Histopathological analysis of the intestinal polyps revealed mostly hyperplastic morphology without obvious dysplasia in Riccardin D-treated mice. Molecular analyses of the polyps suggested that the inhibitory effect of Riccardin D on intestinal adenoma formation was associated with its abilities of reduction in cell proliferation, induction of apoptosis, antiangiogenesis, inhibition of the Wnt signaling pathway and suppression of inflammatory mediators in polyps. CONCLUSIONS: Our results suggested that Riccardin D exerts its chemopreventive effect against intestinal adenoma formation through multiple mechanisms including anti-proliferative, apoptotic, anti-angiogenic and anti-inflammatory activity.


Assuntos
Adenoma/prevenção & controle , Proteína da Polipose Adenomatosa do Colo/metabolismo , Produtos Biológicos/farmacologia , Hepatófitas/química , Neoplasias Intestinais/prevenção & controle , Éteres Fenílicos/farmacologia , Lesões Pré-Cancerosas/prevenção & controle , Estilbenos/farmacologia , Adenoma/irrigação sanguínea , Adenoma/patologia , Animais , Apoptose/efeitos dos fármacos , Produtos Biológicos/química , Caspases/metabolismo , Proliferação de Células/efeitos dos fármacos , Colo/efeitos dos fármacos , Colo/patologia , Ciclina D1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Inflamação/patologia , Neoplasias Intestinais/irrigação sanguínea , Neoplasias Intestinais/patologia , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Pólipos Intestinais/prevenção & controle , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Camundongos , NF-kappa B/metabolismo , Neovascularização Patológica/tratamento farmacológico , Éteres Fenílicos/química , Éteres Fenílicos/uso terapêutico , Lesões Pré-Cancerosas/patologia , Transdução de Sinais/efeitos dos fármacos , Estilbenos/química , Estilbenos/uso terapêutico , beta Catenina/metabolismo
7.
J Gastrointestin Liver Dis ; 19(2): 155-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20593048

RESUMO

BACKGROUND AND AIMS: Colorectal carcinoma is the second leading cause of death by cancer in Europe as its incidence increases with life span. Continuing research to detect new highly sensitive and specific noninvasive biomarkers is essential. The aim of this study was to compare 9 mismatching repair (MMR) genes activation levels in normal, polyp and malignant tissues in order to detect a MMR gene expression pattern in sporadic colorectal malignant pathology. METHODS: MMR mRNA levels were evaluated in tumor-normal tissue paired samples and polyps collected from 29 patients undergoing standard surgical procedures with curative intention. Real-Time quantitative Reverse Transcription PCR (qRT PCR) with TaqMan probes specific to ANKRD17, EXO1, MLH1, MLH3, MSH2, MSH3, MSH4, MSH5, MSH6 gene transcripts were used. RESULTS: The general tendency observed was a lower mRNA level of MMR genes in tumor samples compared with the normal tissue, with the exception of EXO1 gene. The number of patients that showed a higher expression of MMR genes in normal tissue was significantly greater than the number of patients that showed a higher expression inside the tumor (p=0.0024). ANKRD17 mRNA levels were higher in normal tissue than in tumor for 16 cases, by contrast with only 6 cases of higher mRNA levels in tumor. CONCLUSIONS: ANKRD17 mRNA appears to be the most sensitive target and may have a potential value as an additional marker for the existing multitarget assay panel for colorectal cancer detection.


Assuntos
Adenocarcinoma/genética , Neoplasias Colorretais/genética , Reparo de Erro de Pareamento de DNA , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Pólipos Intestinais/genética , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise por Conglomerados , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Perfilação da Expressão Gênica/métodos , Humanos , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Pessoa de Meia-Idade , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Romênia
9.
Pharmacogenet Genomics ; 19(2): 113-20, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19077918

RESUMO

INTRODUCTION: C-reactive protein (CRP) is a nonspecific marker of inflammation linked to cardiovascular disease and possibly colon cancer. Polymorphisms in CRP have been associated with differential CRP concentrations among healthy adults, with some evidence for functional effects on CRP expression. METHODS: A linkage disequilibrium-based tag single nucleotide polymorphism (SNP)-selection algorithm identified six tagSNPs for Europeans (-821A>G, -390C>T/A, 90A>T, 838G>C, 2043G>A, and 4363C>A), defining six haplotypes with more than 1% frequency. In a case-control study of adenomatous (n=491) or hyperplastic (n=184) polyps versus polyp-free controls (n=583) we investigated these SNPs in relation to colorectal polyp risk. RESULTS: Individuals with 838 GC or CC genotypes had a modestly, although not statistically significantly, increased risk of adenomas (odds ratio: 1.4 95% confidence interval: 0.9-2.1) and a nearly 2-fold increased risk of concurrent adenomas and hyperplastic polyps (odds ratio: 2.0 95% confidence interval: 1.1-3.6). Increased risk for concurrent adenomas and hyperplastic polyps was also observed for haplotype ACACAC. No other main associations were detected. Risk of adenomas associated with 2043G>A differed with nonsteroidal anti-inflammatory drug (NSAID) use. Among NSAID nonusers, there was a suggestion that the GA or AA genotypes were associated with decreased risk of adenomas; this was not seen among NSAID users (P interaction=0.03). We also observed interactions between UGT1A1 [TA](7) promoter repeat polymorphism and CRP tagSNPs -390C>T/A and 90A>T, in which only the homozygous variant CRP genotype was associated with increased risk of adenoma among those with the UGT1A1 6rpt/6rpt genotype (P interaction=0.02 and 0.04 for -390C>T/A and 90A>T, respectively). CONCLUSION: These results provide limited support for associations between genetic variation in CRP and colorectal polyp risk. The observed interactions should be evaluated further.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Proteína C-Reativa/genética , Neoplasias Colorretais/genética , Haplótipos , Pólipos Intestinais/genética , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Anti-Inflamatórios não Esteroides/uso terapêutico , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Proteína C-Reativa/metabolismo , Estudos de Casos e Controles , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/patologia , Citocromo P-450 CYP2C9 , Feminino , Variação Genética , Genótipo , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Humanos , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Masculino , Pessoa de Meia-Idade
10.
Mol Cancer Ther ; 7(9): 2779-87, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18790758

RESUMO

A large body of studies has suggested that peroxisome proliferator-activated receptor gamma (PPARgamma) ligands, such as thiazolidinedione, are potent candidates for chemopreventive agents. MCC-555 is a PPARgamma/alpha dual agonist and has been shown previously to induce apoptosis in vitro; however, the molecular mechanisms by which MCC-555 affects antitumorigenesis in vivo are poorly understood. In this study, we explored the antitumorigenic effects of MCC-555 both in cell culture and in Apc-deficient mice, an animal model for human familial adenomatous polyposis. MCC-555 increased MUC2 expression in colorectal and lung cancer cells, and treatment with the PPARgamma antagonist GW9662 revealed that MUC2 induction by MCC-555 was mediated in a PPARgamma-dependent manner. Moreover, MCC-555 increased transcriptional activity of human and mouse MUC2 promoters. Subsequently, treatment with MCC-555 (30 mg/kg/d) for 4 weeks reduced the number of small intestinal polyps to 54.8% of that in control mice. In agreement with in vitro studies, enhanced Muc2 expression was observed in the small intestinal tumors of Min mice treated with MCC-555, suggesting that MUC2 expression may be associated at least in part with the antitumorigenic action of MCC-555. In addition, highly phosphorylated extracellular signal-regulated kinase (ERK) was found in the intestinal tumors of MCC-555-treated Min mice, and inhibition of the ERK pathway by a specific inhibitor markedly suppressed MCC-555-induced Muc2 expression in vitro. Overall, these results indicate that MCC-555 has a potent tumor suppressor activity in intestinal tumorigenesis, likely involving MUC2 up-regulation by ERK and PPARgamma pathways.


Assuntos
Proteína da Polipose Adenomatosa do Colo/deficiência , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Tiazolidinedionas/farmacologia , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ligantes , Neoplasias Pulmonares/genética , Camundongos , Camundongos Endogâmicos C57BL , Mucina-2 , Mucinas/genética , Mucinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tiazolidinedionas/química , Transcrição Gênica/efeitos dos fármacos
11.
Cancer Res ; 67(19): 9380-8, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17909047

RESUMO

Clinical and animal studies indicate a role for cyclooxygenase-2 (COX-2) and the epidermal growth factor receptor (EGFR) in the development and progression of intestinal polyps and cancers. Although this combination of enzyme inhibition has shown synergy in intestinal polyp and tumor models, the exact mechanism for these effects remains undefined. Therefore, we sought to define the molecular mechanisms through which this process occurs. We observed a significant reduction in the number and size of small intestinal polyps in APC(min+/-) mice treated with either celecoxib (a selective COX-2 inhibitor) or erlotinib (Tarceva, an EGFR inhibitor). However, in combination, there was an overall prevention in the formation of polyps by over 96%. Furthermore, we observed a 70% reduction of colorectal xenograft tumors in mice treated with the combination and microarray analysis revealed genes involved in cell cycle progression were negatively regulated. Although we did not observe significant changes in mRNAs of genes with known apoptotic function, there was a significant increase of apoptosis in tumors from animals treated with the combination. The inhibition of EGFR also induced the down-regulation of COX-2 and further inhibited prostaglandin E2 formation. We observed similar effects on the prevention of intestinal adenomas and reduction of xenograft tumor volume when nonselective COX inhibitors were used in combination with erlotinib. Together, these findings suggest that the inhibition of both COX-2 and EGFR may provide a better therapeutic strategy than either single agent through a combination of decreased cellular proliferation and prostaglandin signaling as well as increased apoptosis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Inibidores de Ciclo-Oxigenase 2/farmacologia , Receptores ErbB/antagonistas & inibidores , Pirazóis/farmacologia , Quinazolinas/farmacologia , Sulfonamidas/farmacologia , Animais , Anti-Inflamatórios não Esteroides/administração & dosagem , Celecoxib , Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/prevenção & controle , Cloridrato de Erlotinib , Humanos , Imuno-Histoquímica , Pólipos Intestinais/tratamento farmacológico , Pólipos Intestinais/enzimologia , Pólipos Intestinais/metabolismo , Intestino Delgado/enzimologia , Intestino Delgado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Análise de Sequência com Séries de Oligonucleotídeos , Prostaglandinas/biossíntese , Prostaglandinas/metabolismo , Pirazóis/administração & dosagem , Quinazolinas/administração & dosagem , Sulfonamidas/administração & dosagem , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Am J Gastroenterol ; 102(5): 1070-6, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17378912

RESUMO

BACKGROUND: Detection of fecal DNA is a promising approach to colorectal cancer screening. However, the sensitivity of current fecal DNA tests for colorectal polyps is low. We evaluated the feasibility of detecting aberrantly methylated DNA or cyclooxygenase-2 (COX-2) mRNA in feces of patients with colorectal cancer or polyps. METHODS: Fecal samples were collected prior to colonoscopy from 20 patients with colorectal cancer, 30 patients with colorectal polyps, and 30 subjects with normal colonic examination. Presence of hypermethylated DNA in 7 tumor-related genes (APC, ATM, hMLH1, sFRP2, HLTF, MGMT, and GSTP1) in stool was analyzed by methylation-specific PCR. COX-2 mRNA in fecal samples was detected by RT-PCR. RESULTS: With the use of this panel of methylation markers, the sensitivity of detecting colorectal cancer and adenoma was 75% (95% CI 50.9-91.3%) and 68% (95% CI 46.5-85.1%), respectively. Three normal subjects also had methylated DNA detected in stool, which gives a specificity of 90% (95% CI 73.5-97.9%). The mean number of genes methylated in DNA from the stool of patients with colorectal cancer and adenoma was 1.4 and 0.9, respectively. In contrast, COX-2 mRNA was detected in the stool samples of 10 (50%) cancer patients and one (4%) patient with advanced adenoma only. Two (6.7%) stool samples from normal subjects also had COX-2 mRNA detected. CONCLUSION: Detection of aberrantly methylated DNA in fecal samples is more sensitive than COX-2 mRNA for detection of colorectal cancer and adenoma.


Assuntos
Neoplasias Colorretais/diagnóstico , Ciclo-Oxigenase 2/análise , DNA de Neoplasias/análise , Pólipos Intestinais/diagnóstico , Idoso , Neoplasias Colorretais/enzimologia , Metilação de DNA , Fezes , Humanos , Pólipos Intestinais/enzimologia , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade
13.
Cancer Res ; 66(14): 6955-63, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16849539

RESUMO

Members of the protein kinase C (PKC) family of serine/threonine kinases play key regulatory roles in numerous cellular processes, including differentiation and proliferation. Of the 11 mammalian PKC isoforms known, several have been implicated in tumor development and progression. However, in most cases, isotype specificity is poorly defined, and even contrary functions for a single PKC have been reported mostly because appropriate molecular and genetic tools were missing to specifically assess the contribution of single PKC isoforms in vivo. In this report, we therefore used PKC genetic targeting to study the role of PKCalpha and PKCzeta in colorectal cancer. Both isoforms were found to be strongly down-regulated in intestinal tumors of ApcMin/+ mice. A deletion of PKCzeta did not affect tumorigenesis in this animal model. In contrast, PKCalpha-deficient ApcMin/+ mice developed more aggressive tumors and died significantly earlier than their PKCalpha-proficient littermates. Even without an additional Apc mutation, PKCalpha knockout mice showed an elevated tendency to develop spontaneous intestinal tumors. Transcriptional profiling revealed a role for this kinase in regulating epidermal growth factor receptor (EGFR) signaling and proposed a synergistic mechanism for EGFR/activator protein and WNT/APC pathways in mediating intestinal tumor development.


Assuntos
Neoplasias Colorretais/enzimologia , Proteína Quinase C-alfa/fisiologia , Proteína Quinase C/fisiologia , Animais , Betacelulina , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Regulação para Baixo , Fator de Crescimento Epidérmico/biossíntese , Fator de Crescimento Epidérmico/genética , Receptores ErbB/biossíntese , Receptores ErbB/genética , Perfilação da Expressão Gênica , Genes jun , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase C/deficiência , Proteína Quinase C/genética , Proteína Quinase C-alfa/deficiência , Proteína Quinase C-alfa/genética , Transdução de Sinais , Transcrição Gênica
14.
Genes Dev ; 20(12): 1569-74, 2006 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-16778075

RESUMO

The tumor suppressor PTEN is frequently inactivated in human cancers. A major downstream effector of PTEN is Akt, which is hyperactivated via PTEN inactivation. It is not known, however, whether diminished Akt activity is sufficient to inhibit tumorigenesis initiated by Pten deficiency. Here we showed that the deficiency of Akt1 is sufficient to dramatically inhibit tumor development in Pten+/- mice. Akt1 deficiency had a profound effect on endometrium and prostate neoplasia, two types of human cancer, in which PTEN is frequently mutated, and also affected thyroid and adrenal medulla tumors and intestinal polyps. Even haplodeficiency of Akt1 was sufficient to markedly attenuate the development of high-grade prostate intraepithelial neoplasia (PIN) and endometrial carcinoma. These results have significant implications for cancer therapy.


Assuntos
Neoplasias/enzimologia , Neoplasias/patologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/deficiência , Neoplasias das Glândulas Suprarrenais/enzimologia , Neoplasias das Glândulas Suprarrenais/genética , Neoplasias das Glândulas Suprarrenais/patologia , Animais , Neoplasias do Endométrio/enzimologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Heterozigoto , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Masculino , Camundongos , Neoplasias/genética , Neoplasia Prostática Intraepitelial/enzimologia , Neoplasia Prostática Intraepitelial/genética , Neoplasia Prostática Intraepitelial/patologia , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Pseudolinfoma/enzimologia , Pseudolinfoma/genética , Pseudolinfoma/patologia , Neoplasias da Glândula Tireoide/enzimologia , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia
15.
Clin Cancer Res ; 10(17): 5870-9, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15355919

RESUMO

PURPOSE: Recent data support the hypothesis that the inducible isoform of cyclooxygenase (COX-2) plays a role in the early stages of colonic carcinogenesis and that nonsteroidal anti-inflammatory drugs (NSAIDs) retard the development of colon cancer by modulating COX-2. However, the cell types responsible for producing COX-2 in colorectal adenomas remain a subject of controversy. EXPERIMENTAL DESIGN: COX-2 expression in normal colonic mucosa (n = 50), hyperplastic polyps (n = 43), sporadic adenomas (n = 67), and invasive colonic adenocarcinoma (n = 39) was studied in formalin-fixed and paraffin-embedded tissue sections from endoscopy biopsy and colonic resection specimens. Immunohistochemistry (avidin-biotin complex technique with double immunolabeling) was used to identify the phenotypes of COX-2-producing cells. RESULTS: In colorectal adenomas, increased expression of COX-2 was detected and localized to alpha smooth muscle actin ( proportional, variant SMA)-positive subepithelial stromal cells (myofibroblasts) in the periluminal region of the lamina propria in 63 (94%) of 67 cases. In contrast, in normal colonic mucosa and in hyperplastic polyps with intact epithelium, COX-2 expression was found only in macrophages and endothelial cells. In areas in which the surface epithelium was ulcerated in normal mucosa as well as hyperplastic or neoplastic polyps, COX-2 expression was increased in granulation tissue (and present in macrophages, endothelium, and myofibroblasts). In invasive carcinoma, COX-2 expression in myofibroblasts was limited to the adenomatous portion of the tumor and was detected in 62% of cases (n = 39). In addition, focal expression of COX-2 by malignant epithelial cells was observed in 23% of invasive adenocarcinoma. CONCLUSIONS: These results show that increased COX-2 expression in sporadic adenoma of the colon is common and is localized specifically to subepithelial intestinal myofibroblasts. These findings further support the hypothesis that myofibroblasts are important target cells for NSAID-mediated chemoprevention of colorectal cancer.


Assuntos
Adenoma/enzimologia , Neoplasias Colorretais/enzimologia , Epitélio/enzimologia , Fibroblastos/enzimologia , Isoenzimas/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Actinas/metabolismo , Adenocarcinoma/enzimologia , Adenocarcinoma/patologia , Adenoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Colo/enzimologia , Neoplasias Colorretais/patologia , Ciclo-Oxigenase 2 , Feminino , Humanos , Hiperplasia/enzimologia , Técnicas Imunoenzimáticas , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Masculino , Proteínas de Membrana , Pessoa de Meia-Idade , Músculo Liso/enzimologia , Invasividade Neoplásica , Células Estromais/enzimologia
16.
Int J Cancer ; 109(4): 576-80, 2004 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-14991580

RESUMO

As with cyclooxygenase (COX)-2, genetic disruption of COX-1 gene or pharmacologic inhibition of its activity has been shown to decrease the number of intestinal polyps in Apc gene-deficient mice. The present study was designed to investigate the combined effects of COX-1 and COX-2 selective inhibitors on spontaneous polyp formation in APC1309 female mice. The animals were treated with 300 or 600 ppm mofezolac (a COX-1 selective inhibitor) alone, 200 or 400 ppm nimesulide (a COX-2 selective inhibitor) alone, 300 ppm mofezolac plus 200 ppm nimesulide, 600 ppm mofezolac plus 400 ppm nimesulide, or 10 ppm indomethacin (a dual-COX inhibitor) in the diet from 7 weeks of age for 4 weeks. Percentage inhibition of polyp area in the intestine was 17% with 600 ppm mofezolac alone and 25% with 400 ppm nimesulide alone, their sum of 42% being almost equal to the 37% observed for the combination treatment. Administration of 300 ppm mofezolac plus 200 ppm nimesulide also significantly decreased polyp area in the intestine by 30%. Moreover, the numbers of polyps more than 2.5 mm in diameter were markedly decreased by combined treatment of both COX inhibitors. With 10 ppm indomethacin, the dual inhibitor, polyp area was also clearly reduced by 46%. Our results indicate that COX-1 and COX-2 may to some extent contribute to polyp formation independently and inhibitor combination treatment thus has particular potential for chemoprevention of colon carcinogenesis.


Assuntos
Inibidores de Ciclo-Oxigenase/uso terapêutico , Genes APC/fisiologia , Neoplasias Intestinais/genética , Neoplasias Intestinais/prevenção & controle , Isoenzimas/antagonistas & inibidores , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Indometacina/administração & dosagem , Neoplasias Intestinais/enzimologia , Pólipos Intestinais/enzimologia , Pólipos Intestinais/patologia , Pólipos Intestinais/prevenção & controle , Isoxazóis/administração & dosagem , Proteínas de Membrana , Camundongos , Camundongos Knockout , Neoplasias Experimentais/enzimologia , Neoplasias Experimentais/patologia , Neoplasias Experimentais/prevenção & controle , Prostaglandina-Endoperóxido Sintases , Sulfonamidas/administração & dosagem
17.
Cancer Res ; 63(16): 4872-7, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12941808

RESUMO

Membrane arachidonic acid is converted by cyclooxygenase (COX) into prostaglandin (PG) G(2) and then to PGH(2) which is subsequently metabolized to PGE(2) by PGE synthase (PGES). Both COX-1 and COX-2 play critical roles in intestinal polyp formation, whereas COX-2 is also expressed in cancers of a variety of organs. Likewise, inducible microsomal PGES (mPGES-1) is expressed in several types of cancer, although its role in benign polyp formation has not been investigated. We demonstrated recently that most COX-2-expressing cells in the polyps are stromal fibroblasts. Here we show colocalization of COX-1, COX-2 and mPGES in the intestinal polyp stromal fibroblasts of Apc(Delta 716) mice, a model for familial adenomatous polyposis. Contrary to COX-2 that was induced only in polyps >1 mm in diameter, COX-1 was found in polyps of any size. In polyps >1 mm, not only COX-2 but also mPGES was induced in the stromal fibroblasts where COX-1 had already been expressed. Although polyp number and size were markedly reduced in COX-1 (-/-) or COX-2 (-/-) compound mutant Apc mice, both COX-2 and mPGES were induced in the COX-1 (-/-) polyps, whereas COX-1 was expressed in the COX-2 (-/-) polyps. We found also in human familial adenomatous polyposis polyps that COX-2 and mPGES were induced in the COX-1-expressing fibroblasts. On the basis of these results, we propose that COX-1 expression in the stromal cells secures the basal level of PGE(2) that can support polyp growth to approximately 1 mm, and that simultaneous inductions of COX-2 and mPGES support the polyp expansion beyond approximately 1 mm by boosting the stromal PGE(2) production.


Assuntos
Pólipos Intestinais/enzimologia , Isoenzimas/fisiologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Animais , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Indução Enzimática , Oxirredutases Intramoleculares/biossíntese , Proteínas de Membrana , Camundongos , Prostaglandina-E Sintases , Células Estromais/enzimologia
18.
Am J Gastroenterol ; 98(3): 671-8, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12650805

RESUMO

OBJECTIVE: Peutz-Jeghers syndrome (PJS) is an autosomal-dominant hamartomatous polyposis syndrome. Affected individuals are at risk for intestinal and extraintestinal malignancies. Prostaglandins and polyamines are small molecules believed to be important in tumor formation and growth. Cyclooxygenase (COX) and ornithine decarboxylase (ODC) are key enzymes in the prostaglandin and polyamine biosynthetic pathways, respectively. The aim of this study was to measure and compare COX-1 and COX-2 expression in normal and hamartomatous tissue of PJS patients. METHODS: We measured COX-1 and COX-2 protein expression in normal and hamartomatous GI tissues from affected PJS individuals and compared it with that in normal controls. COX-2 RNA in these tissues was also measured and compared by reverse transcription polymerase chain reaction (PCR). In addition, COX-2 expression was detected in tissue slides by immunostaining. ODC activity was measured between normal and hamartomatous tissues of PJS compared with control tissues. RESULTS: COX-1 expression was similar in normal and control GI tissues. In contrast, COX-2 overexpression was noted in hamartomatous polyp tissue from PJS patients compared with normal control and PJS tissue. COX-2 expression by reverse transcription PCR was 10-fold greater in a hamartoma compared with other tissues. COX-2 expression was noted in the epithelial cells of hamartomatous polyps, and also coursing throughout the stromal tissue of the lamina propria, including muscle cells. ODC activity was similar in the tissues studied. CONCLUSIONS: Selective COX-2 overexpression was noted in hamartomatous polyp tissue from PJS individuals. The results of the study provide an avenue for possible effective chemoprevention of polyp formation and growth in PJS.


Assuntos
Isoenzimas/análise , Síndrome de Peutz-Jeghers/enzimologia , Prostaglandina-Endoperóxido Sintases/análise , Western Blotting , Ciclo-Oxigenase 1 , Ciclo-Oxigenase 2 , Receptores ErbB/metabolismo , Regulação Enzimológica da Expressão Gênica , Hamartoma/enzimologia , Humanos , Imuno-Histoquímica , Pólipos Intestinais/enzimologia , Isoenzimas/genética , Proteínas de Membrana , Síndrome de Peutz-Jeghers/metabolismo , Prostaglandina-Endoperóxido Sintases/genética , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
19.
Cancer Res ; 62(23): 6846-9, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12460897

RESUMO

Cyclooxygenase-2 (COX-2), the inducible COX isozyme, plays a key role in intestinal tumorigenesis. We have demonstrated recently that COX-2 protein is induced in the polyp stroma near the intestinal luminal surface in the Apc(Delta716) mouse, a model for human familial adenomatous polyposis, and stimulate tumor angiogenesis. However, the precise cell types that express COX-2 are still to be determined. By immunohistochemical analysis, here we show that the majority of COX-2-expressing cells in the intestinal polyps of Apc(Delta716) mice are fibroblasts and endothelial cells. Furthermore, the COX-2-expressing cells in human familial adenomatous polyposis polyps are also fibroblasts and endothelial cells. In contrast, bone marrow-derived cells such as macrophages and leukocytes express little COX-2 protein in the intestinal polyps. These results clearly indicate that fibroblasts and endothelial cells play important roles in polyp expansion by expressing COX-2, resulting in tumor angiogenesis.


Assuntos
Pólipos Intestinais/enzimologia , Isoenzimas/biossíntese , Prostaglandina-Endoperóxido Sintases/biossíntese , Actinas/biossíntese , Polipose Adenomatosa do Colo/enzimologia , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Animais , Antígenos CD34/biossíntese , Antígenos de Diferenciação/biossíntese , Ciclo-Oxigenase 2 , Endotélio Vascular/citologia , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Humanos , Imuno-Histoquímica , Pólipos Intestinais/patologia , Macrófagos/enzimologia , Macrófagos/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Knockout , Células Estromais/enzimologia , Células Estromais/metabolismo , Vimentina/biossíntese
20.
Carcinogenesis ; 23(8): 1351-9, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12151354

RESUMO

The expression of COX-2 participates strongly in polyp formation of adenomatous polyposis coli (APC)-mutated mice. However, the mechanism of growth inhibition by COX-2 inhibition remains unclear. The aims of this study were to assess the role of COX-2 during the process of polyp formation in APC(Delta474) knockout mice. Starting at 4 weeks of age, the treated group (T group) were given a diet containing JTE-522, a selective COX-2 inhibitor, and the control group (C group) were given a control diet. At 12 weeks of age, mice were killed and polyps located in a proximal 10 cm of the small intestine were classified into two morphological stages: large adenomas (>300 microm in diameter) which lacked normal villous structure, and small adenomas (

Assuntos
Genes APC , Pólipos Intestinais/patologia , Isoenzimas/fisiologia , Prostaglandina-Endoperóxido Sintases/fisiologia , Animais , Apoptose/fisiologia , Divisão Celular/fisiologia , Ciclo-Oxigenase 2 , Pólipos Intestinais/enzimologia , Masculino , Camundongos , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA