Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Gen Comp Endocrinol ; 269: 88-95, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30144443

RESUMO

Follicular cells from various species secrete steroids and prostaglandins, which are crucial for reproduction, in response to gonadotropins. Here, we examined prostaglandin E2 (PGE2) secretion from immortalized rat granulosa cells derived from preovulaotry follicles expressing the rat follicle stimulating hormone receptor (denoted as FSHR cells) that produce progesterone in response to gonadotropins. The cells were stimulated with a) pregnant mare's serum gonadotropin (PMSG; a rat FSH receptor agonist), b) activators of the protein kinase A (PKA) pathway (forskolin and a cell permeable cAMP analog Dibutyryl-cAMP (DB-cAMP)) and c) protein kinase C (PKC) (12-O-tetradecanoylphorbol 13-acetate; TPA), alone and in combination for 24 h. Thereafter, PGE2 and progesterone levels in the culture media were determined. In accordance with previous studies, while PMSG and the PKA pathway activators induced progesterone accumulation in the media, TPA did not. In contrast, our data indicate that TPA, but neither PMSG, forskolin and DB-cAMP evoked PGE2 accumulation in the media. Western Blot analysis of cell lysate showed a drastic TPA induced increase of COX-2 levels, which was not seen with neither PMSG nor forskolin treatment. This association between the COX-2 and PGE2 levels suggests that the enzyme activity is the likely factor that determines the synthesis and levels of the prostaglandin in the culture media of the granulosa-derived cells. The addition of the PKA inhibitor H-89 to the FSHR cultures suppressed the gonadotropin and forskolin induction of progesterone secretion. Incubation in the presence of GF109203X (a PKC inhibitor) attenuated the TPA induced PGE2 accumulation in the culture media of the cells (a dose dependent reduction of 40-70%). In addition, while TPA inhibited the PMSG and forskolin induced-accumulation of progesterone in the media, the gonadotropin and forskolin inhibited the elevation of PGE2 levels evoked by TPA (a dose dependent decrease of 35-55%). These data suggest that cAMP/PKA and PKC signaling have opposite effects on PGE2 and progesterone synthesis in FSHR cells. We propose that this PKA and PKC interplay on progesterone and PGE2 may be advantageous for the coordination of these key mediators for successful ovulation and luteinization.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Células da Granulosa/metabolismo , Progesterona/metabolismo , Proteína Quinase C/metabolismo , Transdução de Sinais , Animais , Técnicas de Cultura de Células , Linhagem Celular Transformada , Colforsina/farmacologia , Feminino , Gonadotropinas/farmacologia , Células da Granulosa/efeitos dos fármacos , Cavalos , Ratos , Receptores do FSH/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esteroides/metabolismo , Acetato de Tetradecanoilforbol/farmacologia
2.
Anticancer Res ; 38(6): 3333-3339, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29848681

RESUMO

BACKGROUND/AIM: Familial adenomatous polyposis (FAP) was found to be completely reversed in a patient treated with mycophenolate mofetil (MMF) and tacrolimus following kidney transplantation. In this preliminary study, we assessed whether MMF and tacrolimus alone or in combination interfere with the cell cycle and proliferation in a human colonic adenocarcinoma cell line and in the colonic polyps of the patient with FAP. MATERIALS AND METHODS: Human colonic adenocarcinoma HT-29 cells were treated with tacrolimus and MMF alone and in combination at different concentrations. Cell viability and proliferation were assessed using the MTT assay. Cell-cycle distribution was analyzed by flow cytometry. Expression of Ki-67, a marker of mitotic activity, was evaluated in the patient's colonic polyps before and under drug treatment. RESULTS: MMF in combination with tacrolimus induced S-phase cell-cycle arrest and markedly inhibited HT-29 cell proliferation. Ki-67 expression in the patient's colonic polyps was significantly reduced following combined tacrolimus and MMF treatment. CONCLUSION: MMF and tacrolimus synergistically inhibited proliferation of a human colonic adenocarcinoma cell line and interfered with the expansion of colonic crypt proliferation in the polyp from a patient with FAP. The results confirm our clinical observation and indicate the possibility of novel approach to therapy of colorectal neoplasia.


Assuntos
Carcinogênese/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colo/efeitos dos fármacos , Ácido Micofenólico/farmacologia , Tacrolimo/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Polipose Adenomatosa do Colo/tratamento farmacológico , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Colo/patologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Sinergismo Farmacológico , Células HT29 , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/farmacologia , Antígeno Ki-67/metabolismo , Ácido Micofenólico/administração & dosagem , Tacrolimo/administração & dosagem
3.
Cancers (Basel) ; 10(3)2018 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-29495500

RESUMO

It is well recognized that cancer cells subvert the phenotype of stromal naïve fibroblasts and instruct the neighboring cells to sustain their growth agenda. The mechanisms underpinning the switch of fibroblasts to cancer-associated fibroblasts (CAFs) are the focus of intense investigation. One of the most significant hallmarks of the biological identity of CAFs is that their tumor-promoting phenotype is stably maintained during in vitro and ex vivo propagation without the continual interaction with the adjacent cancer cells. In this review, we discuss robust evidence showing that the master cytokine Transforming Growth Factor-ß1 (TGFß-1) is a prime mover in reshaping, via epigenetic switches, the phenotype of stromal fibroblasts to a durable state. We also examine, in detail, the pervasive involvement of TGFß-1 signaling from both cancer cells and CAFs in fostering cancer development, taking colorectal cancer (CRC) as a paradigm of human neoplasia. Finally, we review the stroma-centric anticancer therapeutic approach focused on CAFs-the most abundant cell population of the tumor microenvironment (TME)-as target cells.

4.
Anticancer Res ; 36(12): 6225-6234, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27919940

RESUMO

The multifaceted involvement of the active vitamin D metabolite 1,25-dihydroxyvitamin D3 (henceforth referred to by the synonyms 1,25(OH)2D3, calcitriol or vitamin D) in blunting the growth of cancer cells is amply recognized. In this review we focused our attention on the cross-talk between 1,25 (OH)2D3 and the tumor microenvironment (TME), signaling out stromal cancer-associated fibroblasts (CAFs), the most abundant TME population, as a target for calcitriol anticancer action. In view of the commonality of the phenotypic signature in myofibroblasts, resident in the cancer stroma and in non-neoplastic fibrotic loci, we examined modes of action of vitamin D in non-neoplastic chronic diseases and in cancer to assess mechanistic similarities and divergences. A constant observation was that 1,25(OH)2D3 or synthetic ligands via the active vitamin D receptor (VDR) impede transforming growth factor (TGF)-ß/mothers against decapentaplegic homologs (SMADs) signaling in myofibroblasts regardless of the initiating insult. The translational impact of 1,25(OH)2D3 in targetting stromal CAFs is discussed.


Assuntos
Neoplasias Colorretais/metabolismo , Miofibroblastos/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Vitamina D/análogos & derivados , Neoplasias Colorretais/patologia , Fibrose , Humanos , Vitamina D/metabolismo
5.
Anticancer Res ; 36(9): 4503-7, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27630288

RESUMO

BACKGROUND/AIM: Cancer-associated fibroblasts (CAFs) play an important role in tumor development and progression. The prevailing consensus favors the view that a specific epigenetic signature underpins the stable CAF phenotype. The aim of the present study was to assess global DNA methylation in CAFs during the adenoma-carcinoma sequence in non-familial sporadic human colorectal cancer (CRC). PATIENTS AND METHODS: Immunohistochemical staining of nuclear 5-methylcytosine (5'-meCyt) was performed in matched samples of colonic tumor tissue and normal colonic mucosa excised from six patients with adenomas and four with adenocarcinomas. The staining intensity was expressed semi-quantitatively as the immunohistochemical staining score (ISS). RESULTS: ISS values of human colonic CAFs and adenomatous samples were 14.00±2.2 and 14.08±1.8, respectively, showing no statistically significant difference. In contrast, a marked trend was found towards global DNA hypomethylation in CAFs from adenocarcinomatous specimens compared to matched normal mucosa: ISS: 9.25±2.44 (range=6-11) vs. 16.17±0.75, respectively, p<0.03. CONCLUSION: Final stages of cancer development in CRC are associated with global DNA hypomethylation in stromal CAFs.


Assuntos
Adenocarcinoma/metabolismo , Adenoma/embriologia , Fibroblastos Associados a Câncer/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Metilação de DNA , Biomarcadores Tumorais/metabolismo , Biópsia , Linhagem Celular Tumoral , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Estadiamento de Neoplasias , Fenótipo
7.
Eur J Immunol ; 46(2): 400-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26548451

RESUMO

Colitis, an inflammation of the colon, is a well-characterized massive tissue injury. Cytosolic phospholipase A2 α (cPLA2 α) upregulation plays an important role in the development of several inflammatory diseases. The aim of the present study was to define the role of cPLA2 α upregulation in the development of colitis. We used a mouse model of dextran sulfate sodium induced colitis. Immunoblotting analysis showed that cPLA2 α and NF-κB were upregulated and activated in the colon from day 2 of colitis induction. This molecular event preceded the development of the disease, as determined by Disease Activity Index score, body weight, colon length, and the expression of colonic inflammatory markers, including neutrophil infiltration detected by myeloperoxidase and by NIMP-R14, ICAM-1, COX-2, iNOS upregulation and LTB4 and TNF-α secretion. Prevention of cPLA2 α upregulation and activity in the colon by i.v. administration of specific antisense oligonucleotides against cPLA2 α 1 day prior and every day of exposure to dextran sulfate sodium significantly impeded the development of the disease and prevented NF-κB activation, neutrophils infiltration into the colonic mucosa, and expression of proinflammatory proteins in the colon. Our results demonstrate a critical role of cPLA2 α upregulation in inflammation and development of murine colitis.


Assuntos
Colite/imunologia , Fosfolipases A2 do Grupo IV/metabolismo , NF-kappa B/metabolismo , Neutrófilos/imunologia , Animais , Biomarcadores/metabolismo , Movimento Celular/genética , Células Cultivadas , Colite/induzido quimicamente , Colite/prevenção & controle , Sulfato de Dextrana/administração & dosagem , Fosfolipases A2 do Grupo IV/genética , Humanos , Mucosa Intestinal/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , NF-kappa B/genética , Oligonucleotídeos Antissenso/administração & dosagem , Regulação para Cima/genética
8.
Carcinogenesis ; 36(8): 811-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26116624

RESUMO

A prominent model of tumor progression posits that normal self-renewing and multipotent stem cells(SCs) are the initial target of transformation. This view has been robustly challenged by the recurring observation that transit-amplifying cells and differentiated progenitors can initiate neoplasia outside the SC zone thus qualifying as cancer cells-of-origin. The emerging concept is that a cancer SC and a cancer cell-of-origin are not necessarily the same cell. Importantly, progenitor cells were shown to possess remarkable plasticity and to revert, on demand, to a SC-like state. The present review revisits our early hypothesis that colonic progenitors acquiring a mutant adenomatous polyposis coli gene after exiting the stem zone may serve as genuine cancer cells-of-origin. New findings consonant with this view are examined, and tenable molecular and cellular mechanisms underpinning the plasticity of progenitor cells in the gastrointestinal tract and in other tissues are discussed. The translational impact of cell plasticity is addressed, and recommendations for future research are advanced.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Neoplasias Gastrointestinais/patologia , Células-Tronco Neoplásicas/patologia , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/patologia , Trato Gastrointestinal/patologia , Humanos , Mutação
9.
Anticancer Res ; 35(4): 1881-916, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25862842

RESUMO

AIM: to identify biological interactions between proliferating fibroblasts and HeLa cells in vitro. MATERIALS AND METHODS: Fibroblasts were isolated from both normal and tumour human tissues. Coverslip co-cultures of HeLa and fibroblasts in various ratios with medium replacement every 48 h were studied using fixed cell staining with dyes such as Giemsa and silver staining, with immunochemistry for Ki-67 and E-cadherin, with dihydrofolate reductase (DHFR) enzyme reaction, as well as live cell staining for non-specific esterases and lipids. Other techniques included carmine cell labeling, autoradiography and apoptosis assessment. RESULTS: Under conditions of feeding and cell: cell ratios allowing parallel growth of human fibroblasts and HeLa cells, co-cultured for up to 20 days, a series of phenomena occur consecutively: profound affinity between the two cell types and exchange of small molecules; encircling of the HeLa colonies by the fibroblasts and enhanced growth of both cell types at their contact areas; expression of carbonic anhydrase in both cell types and high expression of non-specific esterases and cytoplasmic argyrophilia in the surrounding fibroblasts; intense production and secretion of lipid droplets by the surrounding fibroblasts; development of a complex net of argyrophilic projections of the fibroblasts; E-cadherin expression in the HeLa cells; from the 10th day onwards, an increasing detachment of batches of HeLa cells at the peripheries of colonies and appearance of areas with many multi-nucleated and apoptotic HeLa cells, and small HeLa fragments; from the 17th day, appearance of fibroblasts blocked at the G2-M phase. Co-cultures at approximately 17-20 days display a cell-cell fight with foci of (a) sparse growth of both cell types, (b) overgrowth of the fibroblasts and (c) regrowth of HeLa in small colonies. These results indicate that during their interaction with HeLa cells in vitro, proliferating fibroblasts can be activated against HeLa. This type of activation is not observed if fibroblast proliferation is blocked by contact inhibition of growth at confluency, or by omitting replacement of the nutrient medium. CONCLUSION: The present observations show that: (a) interaction between proliferating fibroblasts and HeLa cells in vitro drastically influences each other's protein expression, growth pattern, chromatin features and survival; (b) these functions depend on the fibroblast/HeLa ratio, cell topology (cell-cell contact and the architectural pattern developed during co-culture) and frequent medium change, as prerequisites for fibroblast proliferation; (c) this co-culture model is useful in the study of the complex processes within the tumour microenvironment, as well as the in vitro reproduction and display of several phenomena conventionally seen in tumour cytological sections, such as desmoplasia, apoptosis, nuclear abnormalities; and (d) overgrown fibroblasts adhering to the boundaries of HeLa colonies produce and secrete lipid droplets.


Assuntos
Proliferação de Células/genética , Técnicas In Vitro , Microambiente Tumoral/genética , Comunicação Celular/genética , Sobrevivência Celular/genética , Cromatina/genética , Técnicas de Cocultura , Fibroblastos/metabolismo , Fibroblastos/patologia , Células HeLa , Humanos , Células Estromais/patologia
10.
J Nutr Biochem ; 22(4): 360-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20655721

RESUMO

There is great interest in the role of polyunsaturated fatty acids (PUFAs) in promoting (n-6 class) or inhibiting (n-3 class) inflammation. Mammalian cells are devoid of desaturase that converts n-6 to n-3 PUFAs. Consequently, essential n-3 fatty acids must be supplied with the diet. We have studied the effect of endogenously produced n-3 PUFAs on colitis development in fat-1 transgenic mice carrying the Caenorhabditis elegans fat-1 gene encoding n-3 desaturase. Colonic cell lipid profile was measured by capillary gas chromatography in fat-1 and wild-type (WT) littermates fed standard diet supplemented with 10% (w/w) safflower oil rich (76%) in n-6 polyunsaturated linoleic acid (LA). Experimental colitis was induced by administrating 3% dextran sodium sulphate (DSS). Colitis was scored by histopatological analysis. Cyclooxygenase-2 (Cox-2) expression was evaluated by real time polymerase chain reaction. Prostaglandin E(2) (PGE(2)) levels and cytokine production were determined by enzyme and microsphere-based immunoassays, respectively. The n-6/n-3 PUFA ratios in colonic cells of fat-1 mice were markedly lower (9.83±2.62) compared to WT (54.5±9.24, P<.001). Results also showed an attenuation of colonic acute and chronic inflammation in fat-1 mice with significant decreases in PGE(2) production (P<.01) and Cox-2 expression (P<.01). High levels of colitis-induced proinflammatory cytokines, interleukin (IL)-18, IL-1α, IL-1ß, IL-6, monocytes chemotactic proteins 1, 2 and 3 (MCP 1,2,3), matrix metalloproteinase 9 and tumor necrosis factor α (TNF-α) were down-regulated in DSS acutely and chronically treated fat-1 mice. The expression of fat-1 gene in the colon was associated with endogenous n-3 PUFAs production, decreased Cox-2 expression, increased PGE(2) and cytokine production.


Assuntos
Colite/metabolismo , Ciclo-Oxigenase 2/biossíntese , Citocinas/biossíntese , Dinoprostona/biossíntese , Ácidos Graxos Ômega-3/biossíntese , Animais , Proteínas de Caenorhabditis elegans/genética , Colite/induzido quimicamente , Colite/patologia , Colo/metabolismo , Colo/patologia , Sulfato de Dextrana , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Insaturados/farmacologia , Camundongos , Camundongos Transgênicos
11.
Carcinogenesis ; 30(11): 1923-6, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19755659

RESUMO

We have previously reported that sulindac, a non-steroidal anti-inflammatory drug, inhibited tumor formation in the small intestine but increased tumors in the colon of Apc(Min/+) mice, a model of human familial adenomatous polyposis. To further explore intestinal regional responses, we studied effects of sulindac on additional gene-targeted mouse models of human intestinal tumorigenesis; these were (i) Apc(1638N/+) mouse (chain termination mutation in exon 15 of the Apc gene); (ii) Mlh1(+/-) mouse (DNA mismatch repair deficiency, a mouse model of human hereditary non-polyposis colorectal cancer) and (iii) double-heterozygous Mlh1(+/-)Apc(1638N/+) mutant mouse. Mice were fed AIN-76A control diet with or without 0.02% sulindac for 6 months. Intestinal regional tumor incidence, multiplicity, volume and degree of inflammation were used as end points. The results showed the following: (i) sulindac inhibited tumor development in the small intestine of Apc(1638N/+) mice; (ii) in contrast, sulindac increased tumors in the small intestine of Mlh1 mutant mice, a neoplastic effect which persisted in heterozygous compound Mlh1(+/-)Apc(1638N/+) mutant mice; (iii) sulindac increased tumors in the cecum of all mice regardless of genetic background; (iv) sulindac decreased inflammation in the small intestine of Apc(1638N/+) mice, but it increased inflammation in the small intestine of Mlh1(+/-) mice and Mlh1(+/-)Apc(1638N/+) mice and (v) sulindac enhanced inflammation in the cecum of all mutant mice. Findings indicate that the effects of sulindac in the intestine of these mutant mouse models are probably related to genetic background and appear to be associated with its inflammatory-inducing response.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Proteína da Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/farmacologia , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Neoplasias Colorretais Hereditárias sem Polipose , Mutação , Proteínas Nucleares/genética , Sulindaco/efeitos adversos , Sulindaco/farmacologia , Polipose Adenomatosa do Colo/induzido quimicamente , Polipose Adenomatosa do Colo/tratamento farmacológico , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Antineoplásicos/uso terapêutico , Ceco/efeitos dos fármacos , Ceco/patologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Neoplasias Colorretais Hereditárias sem Polipose/induzido quimicamente , Neoplasias Colorretais Hereditárias sem Polipose/tratamento farmacológico , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/patologia , Modelos Animais de Doenças , Humanos , Inflamação/induzido quimicamente , Inflamação/genética , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/patologia , Camundongos , Proteína 1 Homóloga a MutL , Sulindaco/uso terapêutico
12.
J Nutr ; 138(9): 1658-63, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18716166

RESUMO

Both epidemiological and experimental findings have indicated that components of Western diets influence colonic tumorigenesis. Among dietary constituents, calcium and cholecalciferol have emerged as promising chemopreventive agents. We have demonstrated that a Western-style diet (WD) with low levels of calcium and cholecalciferol and high levels of (n-6) PUFA, increased the incidence of neoplasia in mouse intestine compared with a standard AIN-76A diet; models included wild-type mice and mice with targeted mutations. In the present study, adenomatous polyposis coli (Apc)(1638N/+) mice carrying a heterozygous Apc mutation were fed either an AIN-76A diet, a WD, or a WD supplemented with calcium and cholecalciferol (WD/Ca/VitD3). Diets were fed for 24 wk and effects on cellular and molecular events were assessed by performing immunohistochemistry in colonic epithelium along the crypt-to-surface continuum. Feeding WD to Apc(1638N/+) mice not only enhanced cyclin D1 expression in colonic epithelium compared with AIN-76A treatment as previously reported but also significantly increased the expression of the antiapoptotic protein B-cell lymphoma 2 (Bcl-2) concomitantly with a decrease in the proapoptotic Bcl2-associated X protein and the number of apoptotic epithelial cells. WD treatment enhanced mutant Apc-driven small intestinal carcinogenesis and also resulted in the formation of a small number of colonic adenomas (0.16 +/- 0.09; P < 0.05). By contrast, the WD/Ca/VitD3 diet reversed WD-induced growth, promoting changes in colonic epithelium. Importantly, Apc(1638N/+) mice fed the WD/Ca/VitD3 diet did not develop colonic tumors, further indicating that dietary calcium and cholecalciferol have a key role in the chemoprevention of colorectal neoplasia in this mouse model of human colon cancer.


Assuntos
Polipose Adenomatosa do Colo/prevenção & controle , Apoptose/efeitos dos fármacos , Cálcio da Dieta/farmacologia , Colecalciferol/farmacologia , Ciclina D1/metabolismo , Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Peso Corporal , Cálcio da Dieta/uso terapêutico , Testes de Carcinogenicidade , Colecalciferol/uso terapêutico , Colo/patologia , Ciclina D1/genética , Dieta , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Mutação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Distribuição Aleatória , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
14.
Int J Cancer ; 116(4): 495-9, 2005 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-15818612

RESUMO

The role of the nuclear peroxisome proliferator-activated receptor-gamma (PPAR-gamma) in colon tumorigenesis remains controversial. Notwithstanding evidence that PPAR-gamma ligands impede murine colorectal carcinogenesis, PPAR-gamma agonists have been shown to enhance in vivo tumor formation in mouse models of human colon cancer. Our study was designed to determine whether troglitazone (TGZ) induces colonic tumor formation in normal C57BL/6J mice and enhances colorectal carcinogenesis in double mutant Apc1638N/+ Mlh1+/- mice fed a standard AIN-76A diet. We report herein that not only does TGZ enhance carcinogenesis in the large intestine of mutant mice predisposed to intestinal carcinogenesis but TGZ also induces colonic tumors in normal mice without gene targeting or carcinogen administration. This observation indicates that preexisting mutational events are not necessary for induction of colonic tumors by activated PPAR-gamma in vivo.


Assuntos
Antineoplásicos/farmacologia , Cromanos/farmacologia , Neoplasias do Colo/induzido quimicamente , Tiazolidinedionas/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Carcinógenos/administração & dosagem , Proteínas de Transporte , Transformação Celular Neoplásica , Neoplasias do Colo/fisiopatologia , Neoplasias do Colo/veterinária , Dieta , Genes APC , Camundongos , Camundongos Endogâmicos C57BL , Proteína 1 Homóloga a MutL , Mutação , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , PPAR gama , Troglitazona
15.
Nutr Rev ; 61(7): 255-8, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12918879

RESUMO

Mouse models have been generated to help identify dietary components that are protective against gastrointestinal tumorigenesis. Some of the models are produced by gene-targeting procedures, whereas others use the normal mouse as an experimental animal. These preclinical mouse models have provided valuable information on the chemopreventive efficacy of specific nutrients greatly enhancing our understanding of the molecular mechanisms involved in gastrointestinal tumorigenesis.


Assuntos
Polipose Adenomatosa do Colo/genética , Neoplasias do Colo/prevenção & controle , Dieta , Modelos Biológicos , Animais , Neoplasias do Colo/genética , Ácido Fólico/uso terapêutico , Camundongos
16.
Nat Rev Cancer ; 3(8): 601-14, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12894248

RESUMO

Recent findings have indicated that dietary calcium, vitamin D and folate can modulate and inhibit colon carcinogenesis. Supporting evidence has been obtained from a wide variety of preclinical experimental studies, epidemiological findings and a few human clinical trials. Important molecular events and cellular actions of these micronutrients that contribute to their tumour-modulating effects are discussed. They include a complex series of signalling events that affect the structural and functional organization of colon cells.


Assuntos
Anticarcinógenos/farmacologia , Cálcio da Dieta/farmacologia , Neoplasias do Colo/prevenção & controle , Ácido Fólico/farmacologia , Vitamina D/farmacologia , Animais , Anticarcinógenos/uso terapêutico , Cálcio da Dieta/uso terapêutico , Neoplasias do Colo/etiologia , Ácido Fólico/uso terapêutico , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Biológicos , Transdução de Sinais , Vitamina D/uso terapêutico
17.
Carcinogenesis ; 23(11): 1777-80, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12419824

RESUMO

It is widely believed that stem cells are the relevant target cells for colonic cell transformation. Evidence is presented that a proliferative transit daughter cell acquiring a mutant adenomatous polyposis coli gene during upward migration from the crypt base can develop retention abnormalities and permanence in the crypt, thus qualifying as a transformed clone which is retained in the colonic epithelium.


Assuntos
Adenocarcinoma/patologia , Adenoma/patologia , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/patologia , Mucosa Intestinal/citologia , Adenocarcinoma/genética , Adenoma/genética , Proteína da Polipose Adenomatosa do Colo/fisiologia , Animais , Movimento Celular , Neoplasias do Colo/genética , Células Epiteliais/citologia , Genes APC , Humanos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA