Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
PLoS One ; 9(1): e84583, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24392144

RESUMO

High mobility group nucleosome-binding protein 5 (HMGN5) is a chromatin architectural protein that binds specifically to nucleosomes and reduces the compaction of the chromatin fiber. The protein is present in most vertebrate tissues however the physiological function of this protein is unknown. To examine the function of HMGN5 in vivo, mice lacking the nucleosome-binding domain of HMGN5 were generated and characterized. Serological analysis revealed that compared to wild-type littermates (Hmgn5(+/Y)), mice with a targeted mutation in the HMGN5 gene (Hmgn5(tm1/Y)), had elevated serum albumin, non-HDL cholesterol, triglycerides, and alanine transaminase, suggesting mild hepatic abnormalities. Metabolomics analysis of liver extracts and urine revealed clear differences in metabolites between Hmgn5(tm1/Y) and their Hmgn5(+/Y) littermates. Hmgn5(tm1/Y) mice had a significant increase in hepatic glutathione levels and decreased urinary concentrations of betaine, phenylacetylglycine, and creatine, all of which are metabolically related to the glutathione precursor glycine. Microarray and qPCR analysis revealed that expression of two genes affecting glutathione metabolism, glutathione peroxidase 6 (Gpx6) and hexokinase 1 (Hk1), was significantly decreased in Hmgn5(tm1/Y) mouse liver tissue. Analysis of chromatin structure by DNase I digestion revealed alterations in the chromatin structure of these genes in the livers of Hmgn5(tm1/Y) mice. Thus, functional loss of HMGN5 leads to changes in transcription of Gpx6 and Hk1 that alter glutathione metabolism.


Assuntos
Glutationa/metabolismo , Proteínas HMGN/metabolismo , Metabolômica , Animais , Cromatina/metabolismo , Feminino , Regulação da Expressão Gênica , Ordem dos Genes , Marcação de Genes , Glutationa Peroxidase/genética , Glutationa Peroxidase/metabolismo , Hexoquinase/genética , Hexoquinase/metabolismo , Fígado/metabolismo , Testes de Função Hepática , Masculino , Metaboloma , Metabolômica/métodos , Camundongos , Camundongos Knockout , Ligação Proteica
2.
J Nutr Biochem ; 23(10): 1207-13, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22209007

RESUMO

Inflammation associated with obesity may play a role in colorectal carcinogenesis, but the underlying mechanism remains unclear. This study investigated whether the Wnt pathway, an intracellular signaling cascade that plays a critical role in colorectal carcinogenesis, is activated by obesity-induced elevation of the inflammatory cytokine tumor necrosis factor-alpha (TNF-α). Animal studies were conducted on C57BL/6 mice, and obesity was induced by utilizing a high-fat diet (60% kcal). An inflammation-specific microarray was performed, and results were confirmed with real-time polymerase chain reaction. The array revealed that diet-induced obesity increased the expression of TNF-α in the colon by 72% (P=.004) and that of interleukin-18 by 41% (P=.023). The concentration of colonic TNF-α protein, determined by ex vivo culture assay, was nearly doubled in the obese animals (P=.002). The phosphorylation of glycogen synthase kinase 3 beta (GSK3ß), an important intermediary inhibitor of Wnt signaling and a potential target of TNF-α, was quantitated by immunohistochemistry. The inactivated (phosphorylated) form of GSK3ß was elevated in the colonic mucosa of obese mice (P<.02). Moreover, ß-catenin, the key effector of canonical Wnt signaling, was elevated in the colons of obese mice (P<.05), as was the expression of a downstream target gene, c-myc (P<.05). These data demonstrate that diet-induced obesity produces an elevation in colonic TNF-α and instigates a number of alterations of key components within the Wnt signaling pathway that are protransformational in nature. Thus, these observations offer evidence for a biologically plausible avenue, the Wnt pathway, by which obesity increases the risk of colorectal cancer.


Assuntos
Neoplasias Colorretais/genética , Dieta Hiperlipídica/efeitos adversos , Obesidade/genética , Fator de Necrose Tumoral alfa/metabolismo , Via de Sinalização Wnt , Animais , Proliferação de Células , Colo/metabolismo , Neoplasias Colorretais/complicações , Neoplasias Colorretais/patologia , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Inflamação/complicações , Inflamação/patologia , Interleucina-18/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Modelos Lineares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Análise em Microsséries , Obesidade/complicações , Obesidade/patologia , Fosforilação , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Risco , Fator de Necrose Tumoral alfa/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
3.
Nutr Rev ; 69(10): 561-71, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21967157

RESUMO

Dietary intake of one-carbon nutrients, particularly folate, vitamin B(2) (riboflavin), vitamin B(6) , vitamin B(12) , and choline have been linked to the risk of cancers of the colon and breast in both human and animal studies. More recently, experimental and epidemiological data have emerged to suggest that maternal intake of these nutrients during gestation may also have an impact on the risk of cancer in offspring later in life. Given the plasticity of DNA methylation in the developing embryo and the established role of one-carbon metabolism in supporting biological methylation reactions, it is plausible that alterations in maternal one-carbon nutrient availability might induce subtle epigenetic changes in the developing embryo and fetus that persist into later life, altering the risk of tumorigenesis throughout the lifespan. This review summarizes the current literature on maternal one-carbon nutrient intake and offspring cancer risk, with an emphasis on cancers of the colon and breast, and discusses specific epigenetic modifications that may play a role in their pathogenesis.


Assuntos
Neoplasias da Mama/epidemiologia , Neoplasias Colorretais/epidemiologia , Fenômenos Fisiológicos da Nutrição Materna , Complexo Vitamínico B/administração & dosagem , Animais , Neoplasias da Mama/prevenção & controle , Neoplasias Colorretais/prevenção & controle , Metilação de DNA , Feminino , Humanos , Masculino , Metilação , Gravidez , Fatores de Risco , Complexo Vitamínico B/metabolismo , Deficiência de Vitaminas do Complexo B/fisiopatologia
4.
FASEB J ; 25(9): 3136-45, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21646397

RESUMO

The Wnt pathway is a pivotal signaling cascade in colorectal carcinogenesis. The purpose of this work is to determine whether depletion of folate and other metabolically related B vitamins induces in vivo activation of intestinal Wnt signaling and whether this occurs in parallel with increased tumorigenesis. A hybrid mouse was created by crossing a Wnt-reporter animal (BAT-LacZ) with a model of colorectal cancer (Apc1638N). A mild depletion of folate and vitamins B2, B6, and B12 was induced over 16 wk, and the control animals in each instance were pair fed a diet containing the basal requirement of these nutrients. The multiplicity of macroscopic tumors and aberrant crypt foci both increased by ~50% in the hybrid mice fed the depletion diet (P<0.05). A 4-fold elevation in Wnt signaling was produced by the depletion diet (P<0.05) and was accompanied by significant changes in the expression of a number of Wnt-related genes in a pattern consistent with its activation. Proliferation and apoptosis of the colonic mucosa both changed in a protransformational direction (P<0.05). In summary, mild depletion of multiple B vitamins produces in vivo activation of colonic Wnt signaling, implicating it as a key pathway by which B-vitamin inadequacies enhance intestinal tumorigenesis.


Assuntos
Neoplasias Colorretais/etiologia , Óperon Lac/fisiologia , Transdução de Sinais/fisiologia , Deficiência de Vitaminas do Complexo B/complicações , Proteínas Wnt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular , Proliferação de Células , Colo/citologia , Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Dieta , Células Epiteliais , Regulação da Expressão Gênica/fisiologia , Genes Reporter , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Óperon Lac/genética , Camundongos , Deficiência de Vitaminas do Complexo B/sangue , Deficiência de Vitaminas do Complexo B/metabolismo
5.
Gut ; 60(12): 1695-702, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21659408

RESUMO

OBJECTIVE: Variations in the intake of folate are capable of modulating colorectal tumorigenesis; however, the outcome appears to be dependent on timing. This study sought to determine the effect of altering folate (and related B vitamin) availability during in-utero development and the suckling period on intestinal tumorigenesis. DESIGN: Female wildtype mice were fed diets either mildly deficient, replete or supplemented with vitamins B(2), B(6), B(12) and folate for 4 weeks before mating to Apc(1638N) males. Females remained on their diet throughout pregnancy and until weaning. After weaning, all Apc(1638N) offspring were maintained on replete diets for 29 weeks. RESULTS: At 8 months of age tumour incidence was markedly lower among offspring of supplemented mothers (21%) compared with those of replete (59%) and deficient (55%) mothers (p=0.03). Furthermore, tumours in pups born to deficient dams were most likely to be invasive (p=0.03). The expression of Apc, Sfrp1, Wif1 and Wnt5a--all of which are negative regulatory elements of the Wnt signalling cascade--in the normal small intestinal mucosa of pups decreased with decreasing maternal B vitamin intake, and for Sfrp1 this was inversely related to promoter methylation. ß-Catenin protein was elevated in offspring of deficient dams. CONCLUSIONS: These changes indicate a de-repression of the Wnt pathway in pups of deficient dams and form a plausible mechanism by which maternal B vitamin intake modulates tumorigenesis in offspring. These data indicate that maternal B vitamin supplementation suppresses, while deficiency promotes, intestinal tumorigenesis in Apc(1638N) offspring.


Assuntos
Neoplasias Colorretais/prevenção & controle , Suplementos Nutricionais , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Complexo Vitamínico B/farmacologia , Deficiência de Vitamina D/complicações , Animais , Animais Recém-Nascidos , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/genética , Modelos Animais de Doenças , Feminino , Ácido Fólico/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Gravidez , Riboflavina/farmacologia , Vitamina B 12/farmacologia , Vitamina B 6/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
6.
Am J Clin Nutr ; 89(6): 1927-36, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19403629

RESUMO

BACKGROUND: Five genes--UNG, SMUG1, MBD4, TDG, and DUT--are involved in the repair or prevention of uracil misincorporation into DNA, an anomaly that can cause mutagenic events that lead to cancer. Little is known about the determinants of uracil misincorporation, including the effects of single nucleotide polymorphisms (SNPs) in the abovementioned genes. Because of their metabolic function, folate and other one-carbon micronutrients may be important factors in the control of uracil misincorporation. OBJECTIVES: We sought to identify polymorphisms in uracil-processing genes that are determinants of DNA uracil concentration and to establish whether one-carbon nutrient status can further modify their effects. DESIGN: We examined the relations between 23 selected variants in the 5 uracil-processing genes, uracil concentrations in whole-blood DNA, and one-carbon nutrient (folate, vitamins B-6 and B-12, and riboflavin) status in 431 participants of the Boston Puerto Rican Health Study. RESULTS: Four SNPs in DUT, UNG, and SMUG1 showed a significant association with DNA uracil concentration. The SNPs in SMUG1 (rs2029166 and rs7296239) and UNG (rs34259) were associated with increased uracil concentrations in the variant genotypes (P = 0.011, 0.022, and 0.045, respectively), whereas the DUT SNP (rs4775748) was associated with a decrease (P = 0.023). In this population, one-carbon nutrient status was not associated with DNA uracil concentration, and it did not modify the effect of these 4 identified SNPs. CONCLUSION: Because elevated uracil misincorporation may induce mutagenic lesions, possibly leading to cancer, we propose that the 4 characterized SNPs in DUT, UNG, and SMUG1 may influence cancer risk and therefore deserve further investigation.


Assuntos
Dano ao DNA , Reparo do DNA , DNA/química , Predisposição Genética para Doença , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Uracila/metabolismo , Complexo Vitamínico B/sangue , Idoso , Feminino , Frequência do Gene , Genes , Genótipo , Hispânico ou Latino , Humanos , Masculino , Pessoa de Meia-Idade , Fatores Sexuais , Uracila/análise , Uracila/sangue
7.
Am J Clin Nutr ; 88(4): 1149-58, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18842806

RESUMO

BACKGROUND: Folate is an essential nutrient that supports nucleotide synthesis and biological methylation reactions. Diminished folate status results in chromosome breakage and is associated with several diseases, including colorectal cancer. Folate status is also inversely related to plasma homocysteine concentrations -- a risk factor for cardiovascular disease. OBJECTIVE: We sought to gain further understanding of the genetic determinants of plasma folate and homocysteine concentrations. Because folate is required for the synthesis of thymidine from uracil, the latter accumulating and being misincorporated into DNA during folate depletion, the DNA uracil content was also measured. DESIGN: Thirteen single nucleotide polymorphisms (SNPs) in genes involved in folate uptake and metabolism, including folate hydrolase (FOLH1), folate polyglutamate synthase (FPGS), gamma-glutamyl hydrolase (GGH), methylene tetrahydrofolate reductase (MTHFR), methionine synthase (MTR), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC1), were studied in a cohort of 991 individuals. RESULTS: The MTHFR 677TT genotype was associated with increased plasma homocysteine and decreased plasma folate. MTHFR 1298A>C and RFC1 intron 5A>G polymorphisms were associated with significantly altered plasma homocysteine concentrations. The FOLH1 1561C>T SNP was associated with altered plasma folate concentrations. The MTHFR 677TT genotype was associated with a approximately 34% lower DNA uracil content (P = 0.045), whereas the G allele of the GGH -124T>G SNP was associated with a stepwise increase in DNA uracil content (P = 0.022). CONCLUSION: Because the accumulation of uracil in DNA induces chromosome breaks, mutagenic lesions, we suggest that, as for MTHFR C677T, the GGH -124 T>G SNP may modulate the risk of carcinogenesis and therefore warrants further attention.


Assuntos
Ácido Fólico/metabolismo , Regulação Enzimológica da Expressão Gênica , Homocisteína/sangue , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Polimorfismo de Nucleotídeo Único , Uracila/metabolismo , Idoso , Aberrações Cromossômicas , Estudos de Coortes , Dano ao DNA/genética , Metilação de DNA , Feminino , Ácido Fólico/sangue , Humanos , Masculino , Proteínas de Membrana Transportadoras/genética , Pessoa de Meia-Idade , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA