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










Base de dados
Intervalo de ano de publicação
1.
Nutr Cancer ; 69(2): 276-288, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28045548

RESUMO

Inflammatory bowel disease (IBD) patients are at increased risk of developing colorectal cancer (CRC). Vitamin D (vD) induces NOD2 gene expression, enhancing immunity, while deficiency impairs intestinal epithelial integrity, increasing inflammation. This study investigated the effect of vD on CRC in colitis, and if preventive benefits are mediated via NOD2. Inflammation-associated CRC was induced by treating C57BL/6J and Nod2-/- mice with azoxymethane (AOM) and dextran sodium sulfate (DSS) cycles (×3). vD-deficient mice displayed more severe colitis compared to vD-supplemented mice, with greater weight loss, higher colitis activity index, increased colonic weight/length ratios, and lower survival rates. Increased histological inflammation score and increased IL-6 were observed in the mucosa of vD-deficient mice. Overall incidence of colonic tumors was not significantly different between vD-deficient and vD-supplemented mice. Higher tumor multiplicity was observed in vD-deficient vs vD-supplemented groups (both mouse strains). After AOM/DSS treatment, decreased plasma 25(OH)D3 levels and downregulation of vD target genes Cyp24 and Vdr were observed in both mice strains (vD-deficient or vD-supplemented diet), compared to saline-treated controls on the vD-deficient diet. In conclusion, vD supplementation reduced colitis severity and decreased the number of inflammation-associated colorectal tumors in both C57BL/6J and Nod2-/- mice, independent of NOD2.


Assuntos
Colite/tratamento farmacológico , Neoplasias Colorretais/prevenção & controle , Vitamina D/farmacologia , Animais , Peso Corporal , Calcifediol/sangue , Colite/induzido quimicamente , Colite/complicações , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Família 24 do Citocromo P450/genética , Sulfato de Dextrana/toxicidade , Regulação da Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Adaptadora de Sinalização NOD2/genética , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptores de Calcitriol/genética
2.
Int J Oncol ; 47(2): 755-63, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26043725

RESUMO

Butyrate is a potent anticarcinogenic compound against colon cancer cells in vitro. However, its rapid metabolism is hypothesized to limit its anticancer benefits in colonic epithelial cells. Carnitine, a potent antioxidant, is essential to fatty acid oxidation. The aims of this study were to identify a colon cancer cell line capable of transporting carnitine. We evaluated the effect of carnitine and acetylcarnitine (ALCAR) on the response of colon carcinoma cells to butyrate. We explored the mechanisms underlying the anticarcinogenic benefit. SW480 cells were incubated with butyrate ± carnitine or ALCAR. Carnitine uptake was assessed using [3H]-carnitine. Apoptosis and cell viability were assessed using an ELISA kit and flow cytometry, respectively. Modulation of proteins implicated in carnitine transport, cell death and proliferation were assessed by western blotting. SW480 cells were found to transport carnitine primarily via the OCTN2 transporter. Butyrate induced SW480 cell death occurred at concentrations of 2 mM and higher. Cells treated with the combination of butyrate (3 mM) with ALCAR exhibited increased mortality. The addition of carnitine or ALCAR also increased butyrate-induced apoptosis. Butyrate increased levels of cyclin D1, p21 and PARP p86, but decreased Bcl-XL and survivin levels. Butyrate also downregulated dephospho-ß-catenin and increased acetylated histone H4 levels. Butyrate and carnitine decreased survivin levels by ≥25%. ALCAR independently induced a 20% decrease in p21. These results demonstrate that butyrate and ALCAR are potentially beneficial anticarcinogenic nutrients that inhibit colon cancer cell survival in vitro. The combination of both agents may have superior anticarcinogenic properties than butyrate alone.


Assuntos
Acetilcarnitina/farmacologia , Antineoplásicos/farmacologia , Butiratos/farmacologia , Neoplasias do Colo/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto
3.
Nutr Cancer ; 64(8): 1279-87, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23163856

RESUMO

Carnitine is known for its essential role in intermediary metabolism. In vitro studies suggest that its antioxidant and anti-inflammatory properties are potentially beneficial toward cancer prevention. This study tested effects of carnitine on the development of colon cancer in vivo using 2 murine models: azoxymethane (AOM) treatment as a model of carcinogen-induced colon cancer and a genetically induced model using Apc (Min/+) mice. AOM and Apc (Min/+) mice divided into dietary groups varying in lipid content, with or without carnitine supplementation (0.08%). AOM-exposed mice on a high butterfat diet had significantly increased aberrant crypts (ACF) (9.3 ± 0.88 vs. 6.3 ± 0.65), and macroscopic tumors (3.8 ± 0.95 vs. 2.0 ± 0.25) compared to mice on a control diet. In AOM mice fed the high butterfat diet, carnitine supplementation inhibited ACF (4.9 ± 0.7 vs. 9.3 ± 0.88, P < 0.001), crypt multiciplicity (1.6 ± 0.08 vs. 1.92 ± 0.1, P < 0.01) and tumors (1.5 ± 0.38 vs. 3.8 ± 0.95, P < 0.001). Carnitine supplementation resulted in significantly increased tissue carnitine and acylcarnitine levels. Carnitine inhibited the development of precancerous lesions and macroscopic colonic tumors in AOM-treated mice. However, carnitine did not exert protective effects on intestinal tumors in Apc (Min/+) mice.


Assuntos
Anticarcinógenos/farmacologia , Carnitina/farmacologia , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias do Colo/prevenção & controle , Animais , Azoximetano , Carnitina/análise , Carnitina O-Acetiltransferase/análise , Carnitina O-Acetiltransferase/metabolismo , Neoplasias do Colo/etiologia , Neoplasias do Colo/patologia , Dieta , Modelos Animais de Doenças , Genes APC , Intestinos/química , Intestinos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação
4.
J Crohns Colitis ; 6(2): 189-97, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22325173

RESUMO

BACKGROUND: The IBD5 locus is a genetic risk factor for IBD, particularly Crohn's Disease, coding for the organic cation/carnitine transporters (OCTN1 and 2). Two variants of OCTN are associated with susceptibility to Crohn's Disease. Modified transport of carnitine in vitro has been reported for a polymorphism of OCTN1. The aim was to investigate the function of intestinal OCTNs in IBD in relation to genetic polymorphisms. METHODS: Intestinal tissue was obtained from endoscopic biopsies and surgical resections from IBD patients (n=33 and 14, resp.) and controls (n=22 and 14, resp.). OCTN protein levels were measured in intestinal biopsies and carnitine transport was quantified in intestinal resections. RESULTS: OCTN1 protein levels were significantly higher in ileal versus colonic tissue (2.95% ± 0.4 vs 0.66% ± 0.2, resp.; p<0.0002). OCTN1 expression was higher in Crohn's disease patients with mutant homozygous or heterozygous genotypes (0.6% ± 0.1 vs 3% ± 0.8, resp., p<0.02). Carnitine transport was very rapid and Na+ dependent (10s). It was not different comparing Crohn's Disease and control groups (0.45 ± 0.12 vs 0.51 ± 0.12 nM carnitine/mg prot/min, resp.). Carnitine transport tended to be higher in subjects with mutant homozygous and heterozygous OCTN1 and OCTN2 genotypes (0.19 vs 0.59 and 0.25 vs 0.6, respectively). CONCLUSIONS: The present data reveal that OCTN protein levels appear to be similar in intestinal tissue from Crohn's Disease patients and controls. Overall, ileal carnitine transport appears to as well equal in Crohn's Disease and control groups. However, there was a trend towards higher carnitine transport in subjects with OCTN1 and OCTN2 mutations.


Assuntos
Carnitina/farmacocinética , Doença de Crohn/genética , Doença de Crohn/metabolismo , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Adolescente , Adulto , Idoso , Análise de Variância , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Criança , Colite Ulcerativa/genética , Colite Ulcerativa/metabolismo , Colo/metabolismo , Feminino , Genótipo , Heterozigoto , Homozigoto , Humanos , Íleo/metabolismo , Masculino , Microvilosidades/metabolismo , Pessoa de Meia-Idade , Mutação , Proteínas de Transporte de Cátions Orgânicos/genética , Polimorfismo Genético , Sódio/farmacologia , Membro 5 da Família 22 de Carreadores de Soluto , Simportadores , Adulto Jovem
5.
Am J Physiol Cell Physiol ; 287(2): C263-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15238359

RESUMO

Maternofetal transport of l-carnitine, a molecule that shuttles long-chain fatty acids to the mitochondria for oxidation, is thought to be important in preparing the fetus for its lipid-rich postnatal milk diet. Using brush-border membrane (BBM) vesicles from human term placentas, we showed that l-carnitine uptake was sodium and temperature dependent, showed high affinity for carnitine (apparent K(m) = 11.09 +/- 1.32 microM; V(max) = 41.75 +/- 0.94 pmol.mg protein(-1).min(-1)), and was unchanged over the pH range from 5.5 to 8.5. l-Carnitine uptake was inhibited in BBM vesicles by valproate, verapamil, tetraethylammonium, and pyrilamine and by structural analogs of l-carnitine, including d-carnitine, acetyl-d,l-carnitine, and propionyl-, butyryl-, octanoyl-, isovaleryl-, and palmitoyl-l-carnitine. Western blot analysis revealed that OCTN2, a high-affinity, Na(+)-dependent carnitine transporter, was present in placental BBM but not in isolated basal plasma membrane vesicles. The reported properties of OCTN2 resemble those observed for l-carnitine uptake in placental BBM vesicles, suggesting that OCTN2 may mediate most maternofetal carnitine transport in humans.


Assuntos
Carnitina/farmacocinética , Troca Materno-Fetal/fisiologia , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Placenta/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/fisiologia , Feminino , Humanos , Concentração de Íons de Hidrogênio , Técnicas In Vitro , Cinética , Microvilosidades/metabolismo , Gravidez , Sódio/farmacologia , Membro 5 da Família 22 de Carreadores de Soluto , Temperatura , Trítio , Xenobióticos/farmacocinética
6.
Am J Physiol Gastrointest Liver Physiol ; 284(5): G863-71, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12684216

RESUMO

l-Carnitine is derived both from dietary sources and biosynthesis. Dietary carnitine is absorbed in the small intestine and then distributed to other organs. Previous studies using Caco-2 cells demonstrated that the transport of l-carnitine in the intestine involves a carrier-mediated system. The purpose of this study was to determine whether the uptake of l-carnitine in Caco-2 cells is mediated by the recently identified organic cation/carnitine transporter (OCTN2). Kinetics of l-[(3)H]carnitine uptake were investigated with or without specific inhibitors. l-Carnitine uptake in mature cells was sodium dependent and linear with time. K(m) and V(max) values for saturable uptake were 14.07 +/- 1.70 micro M and 26.3 +/- 0.80 pmol. mg protein(-1). 6 min(-1), respectively. l-carnitine uptake was inhibited (P < 0.05-0.01) by valproate and other organic cations. Anti-OCTN2 antibodies recognized a protein in the brush-border membrane (BBM) of Caco-2 cells with an apparent molecular mass of 60 kDa. The OCTN2 expression was confirmed by double immunostaining. Our results demonstrate that l-carnitine uptake in differentiated Caco-2 cells is primarily mediated by OCTN2, located on the BBM.


Assuntos
Carnitina/metabolismo , Proteínas de Transporte/metabolismo , Perfilação da Expressão Gênica , Proteínas de Membrana/metabolismo , Proteínas de Transporte de Cátions Orgânicos , Transporte Biológico Ativo/efeitos dos fármacos , Western Blotting , Células CACO-2 , Proteínas de Transporte/análise , Cátions/farmacologia , Diferenciação Celular , Imunofluorescência , Humanos , Proteínas de Membrana/análise , Microvilosidades/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto
7.
Mol Genet Metab ; 76(1): 76-80, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12175785

RESUMO

The juvenile visceral steatosis (jvs) mouse, having a mutation in the carnitine transporter gene Octn2, is a model of primary systemic carnitine deficiency in humans (SCD, OMIM 212140). Like humans with SCD, homozygous jvs -/- mice have hepatic and cardiac steatoses, reduced plasma and tissue carnitines, and increased urinary carnitine clearance. Because symptomatic heterozygotes have been reported for some fatty acid oxidation disorders, including SCD, we compared the jvs heterozygotes to normal control mice. We measured the free and esterified carnitine, total cholesterol, and triglycerides in adult liver samples, myocardium, and skeletal muscle. Our results indicate significant differences between the livers of nonfasting adult normal (n = 8) vs jvs heterozygotes (n = 8) (means +/- SEM, p < 0.01) for the following parameters: free carnitine, 2.28 +/- 0.36 nmol/mg protein vs 0.41 +/- 0.13; total carnitine, 3.48 +/- 0.36 vs 1.27 +/- 0.25; triglycerides, 0.14 +/- 0.04 vs 0.39 +/- 0.02; and total cholesterol, 0.21 +/- 0.02 vs 0.39 +/- 0.04, but not for esterified carnitine, 1.18 +/- 0.17 vs 0.90 +/- 0.17 (p > 0.05). There is also a negative correlation between hepatic free carnitine and triglycerides from jvs heterozygotes (p < 0.05). Similar results were obtained with myocardium and skeletal muscle. We conclude that free and total carnitine levels are significantly lower in the heterozygote mouse liver and heart while triglyceride and total cholesterol levels are significantly higher. We speculate that in situations of lipolytic stress, some SCD heterozygotes might develop clinical symptoms of carnitine deficiency.


Assuntos
Carnitina/deficiência , Proteínas de Transporte/genética , Fígado/metabolismo , Proteínas de Membrana/genética , Proteínas de Transporte de Cátions Orgânicos , Animais , Peso Corporal/genética , Colesterol/metabolismo , Modelos Animais de Doenças , Heterozigoto , Camundongos , Músculo Esquelético/metabolismo , Mutação , Miocárdio/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto , Triglicerídeos/sangue , Triglicerídeos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...