Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Trace Elem Med Biol ; 28(4): 436-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25175345

ABSTRACT

Early studies on dietary nickel deprivation found decreased reproduction rate in pigs and decreased insemination and conception rates in goats. Studies from our laboratory demonstrated that nickel deprivation impaired male reproductive function of rats. A physiological amount of nickel modulates the function of cyclic nucleotide-gated cation channels (CNG channels) in vitro. Thus, because CNG channels have important roles in spermatozoa function, it was speculated that the impairment of reproduction by nickel deprivation was through an effect on CNG channels. Because CNG channels are found in retinal photoreceptor, olfactory receptor, and taste receptor cells, we hypothesized that nickel deprivation would also alter light/dark preference, odor preference to female rat urine, and taste preference/aversion in rats. In the light/dark Y-maze, nickel deprivation significantly decreased time spent in the dark arm by rats. The number of sniffs to estrous female urine was significantly increased only in nickel-supplemented rats. The number of licks at the saccharin bottle was significantly decreased by dietary nickel deprivation. These findings suggest that nickel has a biological role in the special senses: vision, olfaction and taste.


Subject(s)
Nickel/deficiency , Smell/physiology , Taste/physiology , Vision, Ocular/physiology , Animals , Cyclic Nucleotide-Gated Cation Channels/metabolism , Dietary Supplements , Female , Male , Rats
2.
J Nutr ; 143(10): 1533-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23946348

ABSTRACT

Maternal low-protein diets result in lower birth weight followed by accelerated catch-up growth that is accompanied by the development of obesity and glucose intolerance in later life. Whether postnatal high-fat (HF) diets further contribute to the development of obesity and insulin resistance in offspring by affecting adipose tissue metabolism and DNA methylation is currently unknown. Obese-prone Sprague-Dawley rats were fed 8% low protein (LP) or 20% normal protein diets for 3 wk prior to conception and throughout pregnancy and lactation to investigate whether prenatal LP and postnatal HF diets affect the rate of adipose tissue growth, insulin-like growth factor 2 (Igf2) expression, and DNA methylation in male offspring. At weaning, the offspring were fed 10% normal fat or 45% HF diets for 12 wk. The adipose tissue growth rate was increased (up to 26-fold) by the LP prenatal and HF postnatal diets. Adipose tissue Igf2 mRNAs and DNA methylation were increased by the LP prenatal and HF postnatal diets. The LP prenatal and HF postnatal diet increased the number of small adipocytes in adipose tissue and decreased insulin sensitivity. These findings suggest that prenatal LP and postnatal HF intake result in adipose tissue catch-up growth through alterations in the expression of the Igf2 gene and DNA methylation within adipocytes. These alterations in adiposity are accompanied by an increased risk of development of type 2 diabetes.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Diet, Protein-Restricted/adverse effects , Insulin-Like Growth Factor II/metabolism , Obesity/etiology , Prenatal Exposure Delayed Effects , Prenatal Nutritional Physiological Phenomena , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/growth & development , Animals , DNA Methylation , Dietary Fats/adverse effects , Dietary Proteins/administration & dosage , Female , Insulin Resistance , Insulin-Like Growth Factor II/genetics , Lactation , Obesity/metabolism , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Weaning
4.
Free Radic Res ; 45(9): 1052-60, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21726174

ABSTRACT

Visceral obesity is linked to insulin resistance and cardiovascular disease. A recent genetic study indicated that the gene locus for the anti-oxidant defense enzyme methionine sulphoxide reductase A (MsrA) is positively associated with the development of visceral adiposity. This work tested the hypothesis that Msr activity is diminished in visceral fat as a result of obesity. It used two animal models of obesity, wild-type rats fed a high-fat (45% of calories from fat) diet and Zucker rats fed a 10% fat calorie diet. The data indicate that MsrA activity was selectively reduced by ∼ 25% in the visceral adipose, but not subcutaneous adipose or liver, of both rat models as compared to control, wild type rats receiving a 10% fat calorie diet. MsrB activity was similarly reduced only in visceral fat. The data indicate that Msr activity is reduced by obesity and may alter oxidative stress signalling of obesity.


Subject(s)
Intra-Abdominal Fat/enzymology , Obesity/enzymology , Oxidoreductases/metabolism , Animals , Liver/enzymology , Male , Oxidative Stress , Rats , Rats, Zucker , Subcutaneous Fat/enzymology
5.
Nutr J ; 10: 75, 2011 Jul 18.
Article in English | MEDLINE | ID: mdl-21767397

ABSTRACT

BACKGROUND: Selenium (Se) status in non-deficient subjects is typically assessed by the Se contents of plasma/serum. That pool comprises two functional, specific selenoprotein components and at least one non-functional, non-specific components which respond differently to changes in Se intake. A more informative means of characterizing Se status in non-deficient individuals is needed. METHODS: Multiple biomarkers of Se status (plasma Se, serum selenoprotein P [SEPP1], plasma glutathione peroxidase activity [GPX3], buccal cell Se, urinary Se) were evaluated in relation to selenoprotein genotypes (GPX1, GPX3, SEPP1, SEP15), dietary Se intake, and parameters of single-carbon metabolism in a cohort of healthy, non-Se-deficient men (n = 106) and women (n = 155). CONCLUSIONS: Plasma Se concentration was 142.0 ± 23.5 ng/ml, with GPX3 and serum-derived SEPP1 calculated to comprise 20% and 34%, respectively, of that total. The balance, comprised of non-specific components, accounted for virtually all of the interindividual variation in total plasma Se. Buccal cell Se was associated with age and plasma homocysteine (hCys), but not plasma Se. SEPP1 showed a quadratic relationship with body mass index, peaking at BMI 25-30. Urinary Se was greater in women than men, and was associated with metabolic body weight (kg0.75), plasma folate, vitamin B12 and hCys (negatively). One GPX1 genotype (679T/T) was associated with significantly lower plasma Se levels than other allelic variants. Selenium intake, estimated from food frequency questionnaires, did not predict Se status as indicated by any biomarker. These results show that genotype, methyl-group status and BMI contribute to variation in Se biomarkers in Se-adequate individuals.


Subject(s)
Diet , Selenium/blood , Selenium/urine , Adult , Aged , Biomarkers/blood , Body Mass Index , Body Weight , Cohort Studies , DNA/genetics , Female , Folic Acid/blood , Genotype , Glutathione Peroxidase/blood , Glutathione Peroxidase/genetics , Humans , Male , Middle Aged , Nutritional Status , Selenoprotein P/blood , Selenoprotein P/genetics , Vitamin B 12/blood
6.
J Nutr ; 141(8): 1464-8, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21653573

ABSTRACT

The regulation of site-specific DNA methylation of tumor suppressor genes has been considered as a leading mechanism by which certain nutrients exert their anticancer property. This study was to investigate whether selenium (Se) affects the methylation of globe genomic DNA and the exon-specific p53 gene. Three groups of rats (n = 6-7/group) were fed the AIN-93G basal diet supplemented with 0 [Se deficient (D)], 0.15 [Se adequate (A)], or 4 mg [Se supranutritional (S)] (Se as l-selenomethionine)/kg diet for 104 d, respectively. Rats fed the A or S diet had greater plasma and liver glutathione peroxidase activity, liver thioredoxin reductase activity, and plasma homocysteine concentration than those fed the D diet. However, compared with the A diet, rats fed the S diet did not further increase these Se-dependent enzyme activities or homocysteine concentration. In contrast, Se concentrations in kidney, liver, gastrocnemius muscle, and plasma were increased in a Se-dose-dependent manner. Interestingly, rats fed the S diet had significantly less global liver genomic DNA methylation than those fed the D diet. However, the S diet significantly increased the methylation of the p53 gene (exons 5-8) but not the ß-actin gene (exons 2-3) DNA in liver and colon mucosa compared with those fed the D diet. Taken together, long-term Se consumption not only affects selenoprotein enzyme activities, homocysteine, tissue Se concentrations, and global genomic DNA methylation but also increases exon-specific DNA methylation of the p53 gene in a Se-dose-dependent manner in rat liver and colon mucosa.


Subject(s)
Colon/metabolism , DNA Methylation , Exons , Genes, p53 , Liver/metabolism , Selenomethionine/administration & dosage , Animals , Glutathione Peroxidase/blood , Homocysteine/blood , Intestinal Mucosa/metabolism , Male , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley
7.
Anal Biochem ; 401(1): 68-73, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20167203

ABSTRACT

A capillary electrophoresis (CE) method for the determination of methionine sulfoxide reductase A and methionine sulfoxide reductase B activities in mouse liver is described. The method is based on detection of the 4-(dimethylamino)azobenzene-4'-sulfonyl derivative of l-methionine (dabsyl Met), the product of the enzymatic reactions when either dabsyl l-methionine S-sulfoxide or dabsyl l-methionine R-sulfoxide is used as a substrate. The method provides baseline resolution of the substrates and, therefore, can be used to easily determine the purity of the substrates. The method is rapid ( approximately 20min sample to sample), requires no column regeneration, and uses very small amounts of buffers. Separation was performed by using a 75-mum internal diameter polyimide-coated fused silica capillary (no inside coating) with 60cm total length (50cm to the detector window). Samples were separated at 22.5kV, and the separation buffer was 25mM KH(2)PO(4) (pH 8.0) containing 0.9ml of N-lauroylsarcosine (sodium salt, 30% [w/v] solution) per 100ml of buffer. Prior to use, the capillary was conditioned with the same buffer that also contained 25mM sodium dodecyl sulfate. The CE method is compared with high-performance liquid chromatography (HPLC) as determined by comparing results from measurements of hepatic enzyme activities in mice fed either deficient or adequate selenium.


Subject(s)
Electrophoresis, Capillary/methods , Methionine Sulfoxide Reductases/metabolism , Animals , Liver/enzymology , Methionine/analogs & derivatives , Methionine/metabolism , Methionine Sulfoxide Reductases/chemistry , Mice , Selenium/pharmacology
8.
Biol Trace Elem Res ; 131(1): 71-80, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19263001

ABSTRACT

Our previous studies have shown that selenium (Se) is protective against dimethylhydrazine (DMH)-induced preneoplastic colon cancer lesions, and protection against DNA damage has been hypothesized to be one mechanism for the anticancer effect of Se. The present study was designed to determine whether dietary selenite affects somatic mutation frequency in vivo. We used the Big Blue transgenic model to evaluate the in vivo mutation frequency of the cII gene in rats fed either a Se-deficient (0 microg Se/g diet) or Se-supplemented diet (0.2 or 2 microg Se/g diet; n = 3 rats/diet in experiment 1 and n = 5 rats/group in experiment 2) and injected with DMH (25 mg/kg body weight, i.p.). There were no significant differences in body weight between the Se-deficient and Se-supplemented (0.2 or 2 microg Se/g diet) rats, but the activities of liver glutathione peroxidase and thioredoxin reductase and concentration of liver Se were significantly lower (p < 0.0001) in Se-deficient rats compared to rats supplemented with Se. We found no effect of dietary Se on liver 8-hydroxy-2'-deoxyguanosine. Gene mutation frequency was significantly lower in liver (p < 0.001) than that of colon regardless of dietary Se. However, there were no differences in gene mutation frequency in DNA from colon mucosa or liver from rats fed the Se-deficient diet compared to those fed the Se-supplemented (0.2 or 2 microg Se/g diet) diet. Although gene mutations have been implicated in the etiology of cancer, our data suggest that decreasing gene mutation is not likely a key mechanism through which dietary selenite exerts its anticancer action against DMH-induced preneoplastic colon cancer lesions in a Big Blue transgenic rat model.


Subject(s)
Colon/drug effects , Colon/metabolism , Diet , Liver/drug effects , Liver/metabolism , Mutation/drug effects , Sodium Selenite/pharmacology , Animals , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/prevention & control , Dimethylhydrazines/toxicity , Glutathione Peroxidase/metabolism , Rats , Sodium Selenite/administration & dosage , Transcription Factors
9.
Biol Trace Elem Res ; 129(1-3): 213-20, 2009.
Article in English | MEDLINE | ID: mdl-19104759

ABSTRACT

Previously, we reported that both Se deficiency and Cu deficiency decreased plasma homocysteine (pHcys) and increased plasma glutathione (pGSH) in rats. We also showed that the catalytic subunit of glutamate-cysteine ligase (Gclc), which catalyzes the rate-limiting step in glutathione biosynthesis, was upregulated in Se and Cu deficiencies. We suggested that in both deficiencies, Hcys was being shunted through the trans-sulfuration pathway as a result of this up-regulation. Because both Se and Cu deficiencies have similar effects, we hypothesized that a combined deficiency would exacerbate the decrease in pHcys and the increase in pGSH by further up-regulating Gclc. In a 2 x 2 experiment, male weanling Sprague-Dawley rats (n = 8-20/group) were fed an amino-acid-based diet containing either 0 or 0.2 microg Se (as selenite)/g and <1 or 6 microg Cu (as Cu carbonate)/g for 5 weeks. Our findings show that a combined deficiency of both Se and Cu results in lower pHcys and significantly elevated pGSH. However, the up-regulation of liver Gclc alone cannot explain why rats fed with the doubly deficient diet have the lowest pHcys and the highest pGSH.


Subject(s)
Animal Feed , Copper/administration & dosage , Copper/pharmacology , S-Adenosylhomocysteine/metabolism , Selenium/administration & dosage , Selenium/pharmacology , Animals , Copper/deficiency , Drug Interactions , Glutathione/blood , Liver/chemistry , Liver/drug effects , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , S-Adenosylhomocysteine/chemistry , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Selenium/deficiency
10.
Biol Trace Elem Res ; 115(3): 265-76, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17625247

ABSTRACT

Reactive oxygen species-mediated oxidation of methionine residues in protein results in a racemic mixture of R and S forms of methionine sulfoxide (MetO). MetO is reduced back to methionine by the methionine sulfoxide reductases MsrA and MsrB. MsrA is specific toward the S form and MsrB is specific toward the R form of MetO. MsrB is a selenoprotein reported to contain zinc (Zn). To determine the effects of dietary selenium (Se) and Zn on Msr activity, CD-1 mice (N=16/group) were fed, in a 2 x 2 design, diets containing 0 or 0.2 microg Se/g and 3 or 15 microg Zn/g. As an oxidative stress, half of the mice received L-buthionine sulfoximine (BSO; ip; 2 mmol/kg, three times per week for the last 3 wk); the others received saline. After 9.5 wk, Msr (the combined specific activities of MsrA and MsrB) was measured in the brain, kidney, and liver. Se deficiency decreased (p<0.0001) Msr in all three tissues, but Zn had no direct effect. BSO treatment was expected to result in increased Msr activity; this was not seen. Additionally, we found that the ratio of MetO to methionine in liver protein was increased (indicative of oxidative damage) by Se deficiency. The results show that Se deficiency increases oxidation of methionyl residues in protein, that Se status affects Msr (most likely through effects on the selenoprotein MsrB), and that marginal Zn deficiency has little effect on Msr in liver and kidney. Finally, the results show that the oxidative effects of limited BSO treatment did not upregulate Msr activity.


Subject(s)
Oxidoreductases/metabolism , Selenium/pharmacology , Zinc/metabolism , Animals , Cysteine/blood , Diet , Glutathione/blood , Homocysteine/blood , Male , Methionine Sulfoxide Reductases , Mice , Models, Biological , Oxygen/metabolism , Reactive Oxygen Species , Selenium/analysis , Selenium/blood , Trace Elements/analysis , Up-Regulation , Zinc/analysis
11.
J Nutr ; 137(6): 1370-4, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17513393

ABSTRACT

The purpose of this study was to determine the effects of copper deficiency on key aspects of homocysteine metabolism that involve methionine recycling and transsulfuration. Male weanling Sprague-Dawley rats were fed AIN-93G-based diets containing <1 or approximately 6 mg Cu/kg. After 6 wk (Expt. 1) and 4 wk (Expt. 2) we found that plasma homocysteine was significantly decreased, and plasma glutathione significantly increased, in rats fed the low-Cu diet. Real-time RT-PCR was used to determine the expression of the subunits of glutamate-cysteine ligase (Gcl) in liver that catalyzes the rate-limiting step in glutathione biosynthesis. The expression of Gclc, the catalytic subunit of Gcl, was upregulated by Cu deficiency; Gclm, the modifier subunit, was not affected. Hepatic betaine-homocysteine methyltransferase (Bhmt), which catalyzes one of the two ways that homocysteine can be remethylated to methionine, was downregulated by Cu deficiency. Because Cu deficiency results in upregulation of Gclc and an increase in the biosynthesis of glutathione, it is plausible that the net flux of homocysteine through the transsulfuration pathway is increased. Furthermore, if Bhmt is downregulated, less homocysteine is available for remethylation (methionine recycling) and more is then available to irreversibly enter the transsulfuration pathway where it is lost. The net effect of increased Gclc and decreased Bhmt would be a decrease in homocysteine as a result of Cu deficiency.


Subject(s)
Copper/deficiency , Glutathione/blood , Homocysteine/blood , Liver/metabolism , Animals , Ceruloplasmin/drug effects , Ceruloplasmin/metabolism , Copper/pharmacology , Homocysteine/metabolism , Liver/drug effects , Liver/enzymology , Male , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , S-Adenosylmethionine/drug effects , S-Adenosylmethionine/metabolism
12.
J Nutr ; 137(5): 1132-6, 2007 May.
Article in English | MEDLINE | ID: mdl-17449570

ABSTRACT

In our previous work with rats, plasma and tissue homocysteine concentrations were decreased by selenium deprivation. The purpose of this study was to follow up and expand on that work by determining the effects of selenium status (deficient, adequate, and supranutritional) on several aspects of homocysteine metabolism involving methionine recycling and transsulfuration. A 2nd objective was to determine whether there are differences in how selenium status affects homocysteine metabolism in rats and mice. Male weanling Fischer-344 rats and male weanling CD-1 mice were fed diets containing 0, 0.2, or 2.0 microg selenium (as sodium selenite)/g for 72 d or 60 d, respectively. Plasma homocysteine and cysteine were significantly decreased by feeding rats or mice the selenium-deficient diet compared with feeding adequate or supranutritional selenium. On the other hand, plasma glutathione was increased by selenium deficiency only in rats. Also, the specific activities of liver betaine homocysteine methyltransferase and glycine N-methyltransferase were decreased by selenium deficiency in rats, but were unaffected by selenium status in mice. Real-time RT-PCR was used to determine the expression of the subunits of glutamate-cysteine ligase, which catalyzes the rate-limiting step in glutathione biosynthesis. The expression of Gclc, the catalytic subunit of glutamate-cysteine ligase, was upregulated by selenium deprivation in both rat and mouse liver. Gclm, the modifier subunit of glutamate-cysteine ligase, was downregulated in rats fed 2 microg Se/g compared with rats fed adequate or deficient selenium. Based on these findings, it is evident that selenium deficiency has different outcomes in mice and rats. These variables are all related to methionine/methyl metabolism. Although only one strain of rat was compared with one strain of mouse, this work suggests that differences between species may prove vital in determining which animal model is used in studies of selenium deficiency or in studies that are designed to ascertain chemopreventive mechanisms of selenium.


Subject(s)
Diet , Homocysteine/metabolism , Mice/metabolism , Rats/metabolism , Selenium/administration & dosage , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Cysteine/blood , Glutamate-Cysteine Ligase/metabolism , Glutathione/blood , Glycine N-Methyltransferase/metabolism , Isoenzymes/metabolism , Liver/metabolism , Male , Methionine/metabolism , Mice, Inbred Strains , Rats, Inbred F344 , Selenium/deficiency , Selenium/metabolism , Selenium/pharmacology , Species Specificity , Sulfur/metabolism
13.
Biol Trace Elem Res ; 109(3): 201-14, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16632891

ABSTRACT

A previous study compared the effects of folate on methyl metabolism in colon and liver of rats fed a selenium-deficient diet (< 3 microg Se/kg) to those of rats fed a diet containing supranutritional Se (2 mg selenite/kg). The purpose of this study was to investigate the effects of folate and adequate Se (0.2 mg/kg) on methyl metabolism in colon and liver. Weanling, Fischer-344 rats (n = 8/diet) were fed diets containing 0 or 0.2 mg selenium (as selenite)/kg and 0 or 2 mg folic acid/kg in a 2 x 2 design. After 70 d, plasma homocysteine was increased (p < 0.0001) by folate deficiency; this increase was markedly attenuated (p < 0.0001) in rats fed the selenium-deficient diet compared to those fed 0.2 mg Se/kg. The activity of hepatic glycine N-methyltransferase (GNMT), an enzyme involved in the regulation of tissue S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH), was increased by folate deficiency (p < 0.006) and decreased by selenium deprivation (p < 0.0003). Colon and liver SAH were highest (p < 0.006) in rats fed deficient folate and adequate selenium. Although folate deficiency decreased liver SAM (p < 0.001), it had no effect on colon SAM. Global DNA methylation was decreased (p<0.04) by selenium deficiency in colon but not liver; folate had no effect. Selenium deficiency did not affect DNA methyltransferase (Dnmt) activity in liver but tended to decrease (p < 0.06) the activity of the enzyme in the colon. Dietary folate did not affect liver or colon Dnmt. These results in rats fed adequate selenium are similar to previous results found in rats fed supranutritional selenium. This suggests that selenium deficiency appears to be a more important modifier of methyl metabolism than either adequate or supplemental selenium.


Subject(s)
Colon/metabolism , DNA Methylation , Folic Acid/pharmacology , Liver/metabolism , Selenium/pharmacology , Animals , Colon/cytology , Colon/drug effects , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Deficiency Diseases/metabolism , Deficiency Diseases/pathology , Folic Acid/administration & dosage , Folic Acid/metabolism , Glutathione/blood , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Glycine N-Methyltransferase/metabolism , Homocysteine/blood , Homocysteine/metabolism , Liver/cytology , Liver/drug effects , Male , Rats , Rats, Inbred F344 , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Selenium/administration & dosage , Selenium/metabolism
14.
Mech Ageing Dev ; 127(5): 444-50, 2006 May.
Article in English | MEDLINE | ID: mdl-16519922

ABSTRACT

Long-lived Ames dwarf mice lack growth hormone, prolactin, and thyroid stimulating hormone. Additionally the dwarf mice have enzyme activities and levels that combat oxidative stress more efficiently than those of normal mice. We have shown that methionine metabolism in Ames mice is markedly different than in their wild type littermates. In our previous work we hypothesized that the flux of methionine to the transsulfuration pathway is enhanced in the dwarf mice. The current study was designed to determine whether the flux of methionine to the transsulfuration pathway is increased. We did this by injecting either l-[methyl-(3)H]-methionine or l-[(35)S]-methionine into dwarf or normal mice and then determined retained label (in form of S-adenosylmethionine) 45 min later. The amount of retained hepatic (3)H and (35)S label was significantly reduced in the dwarf mice; at 45 min the specific radioactivity of SAM (pCi/nmol SAM) was 56% lower (p < 0.05) for (3)H-label and 64% lower (p < 0.005) for (35)S-label in dwarf than wild type mice. Retention of (35)S was significantly lower in the brain (37%, p < .04) and kidney (47%, p < 0.02) of the dwarf compared to wild type mice; there was no statistical difference in retained (3)H-label in either brain or kidney. This suggests that both the methyl-moiety and the carbon chain of methionine are lost much faster in the dwarf compared to the wild type mouse, implying that both transmethylation in the liver and transsulfuration in the liver, brain, and kidney are increased in the dwarf mice. As further support, we determined by real-time RT PCR the expression of methionine metabolism genes in livers of mice. Compared to wild type, the Ames dwarf had increased expression of methionine adenosyltransferase 1a (2.3-fold, p = 0.013), glycine N-methyltransferase (3.8-fold, p = 0.023), betaine homocysteine methyltransferase (5.5-fold, p = 0.0006), S-adenosylhomocysteine hydrolase (3.8-fold, p = 0.0005), and cystathionase (2.6-fold; tended to be increased, p = 0.055). Methionine synthase expression was significantly decreased in dwarf compared to wild type (0.48-fold, p = 0.023). These results confirm that the flux of methionine to transsulfuration is enhanced in the Ames dwarf. This, along with data from previous studies support the hypothesis that altered methionine metabolism plays a significant role in the oxidative defense of the dwarf mouse and that the mechanism for the enhanced oxidative defense may be through altered GSH metabolism as a result of the distinctive methionine metabolism.


Subject(s)
Longevity , Methionine/metabolism , Sulfur/metabolism , Animals , Antioxidants/metabolism , Disease Models, Animal , Dwarfism/genetics , Female , Glutathione/metabolism , Humans , Male , Mice , Models, Biological , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
15.
J Inorg Biochem ; 99(6): 1269-74, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15917080

ABSTRACT

Selenium is an essential trace element for humans and other animals, and there is mounting evidence for the efficacy of certain forms of selenium as cancer-chemopreventive compounds. However, over the years, numerous elements such as As, Cu, Zn, Cd, Hg, Sn, Pb, Ni, Co, Sb, Bi, Ag, Au, and Mo have been found to inhibit anti-carcinogenic effects of selenium, which may affect the anti-carcinogenic activity of selenium. The interaction between selenium and arsenic has been one of the most extensively studied. The proposed mechanisms of this interaction include the increase of biliary excretion and direct interaction/precipitation of selenium and arsenic, and their effects on zinc finger protein function, cellular signaling and methylation pathways. This article focuses on these proposed mechanisms and how anti-carcinogenic effects of selenium may be affected by arsenic.


Subject(s)
Anticarcinogenic Agents/pharmacology , Arsenic/toxicity , Selenium/pharmacology , Animals , Anticarcinogenic Agents/metabolism , Arsenic/metabolism , DNA Methylation/drug effects , Drug Interactions , Humans , Methylation , Selenium/metabolism , Signal Transduction/drug effects , Zinc Fingers/drug effects
16.
Biol Trace Elem Res ; 103(2): 133-45, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15772437

ABSTRACT

Cell culture studies have suggested that arsenic exposure results in decreased S-adenosylmethionine (SAM), causing DNA hypomethylation. Previously, we have shown that hepatic SAM is decreased and/or S-adenosylhomocysteine increased in arsenic-deprived rats; these rats tended to have hypomethylated DNA. To determine the effect of dietary arsenic on dimethylhydrazine (DMH)-induced aberrant crypt formation in the colon, Fisher 344 weanling male rats were fed diets containing 0, 0.5, or 50 microg As (as NaAsO2)/g. After 12 wk, dietary arsenic affected the number of aberrant crypts (p<0.02) and aberrant crypt foci (p<0.007) in the colon and the amount of global DNA methylation (p<0.04) and activity of DNA methyltransferase (DNMT) (p<0.003) in the liver. In each case, there were more aberrant crypts and aberrant crypt foci, a relative DNA hypomethylation, and increased activity of DNMT in the rats fed 50 microg As/g compared to those fed 0.5 microg As/g. The same phenomenon, an increased number of aberrant crypts and aberrant crypt foci, DNA hypomethylation, and increased DNMT tended to hold when comparing rats fed the diet containing no supplemental arsenic compared to rats fed 0.5 microg As/g. The data suggest that there is a threshold for As toxicity and that possibly too little dietary As could also be detrimental.


Subject(s)
Arsenic/toxicity , Colon/drug effects , DNA Methylation , DNA Modification Methylases/metabolism , Diet , Dimethylhydrazines/pharmacology , Liver/drug effects , Animals , Arsenic/administration & dosage , Carcinogens/toxicity , Colon/anatomy & histology , Humans , Liver/physiology , Male , Rats , Rats, Inbred F344
17.
Mech Ageing Dev ; 126(3): 389-98, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15664625

ABSTRACT

Reduced signaling of the growth hormone (GH)/insulin-like growth factor-1(IGF-1)/insulin pathway is associated with extended life span in several species. Ames dwarf mice are GH and IGF-1 deficient and live 50-64% longer than wild type littermates (males and females, respectively). Previously, we have shown that Ames mice exhibit elevated levels of antioxidative enzymes and lower oxidative damage. To further explore the relationship between GH and antioxidant expression, we administered GH or saline to dwarf mice and evaluated components of the methionine and glutathione (GSH) metabolic pathways. Treatment of dwarf mice with GH significantly suppressed methionine adenosyltransferase (40 and 38%) and glycine-N-methyltransferase (44 and 43%) activities (in 3- and 12-month-old mice, respectively). Growth hormone treatment elevated kidney gamma-glutamyl-cysteine synthetase protein levels in 3- and 12-month-old dwarf mice. In contrast, the activity of the GSH degradation enzyme, gamma-glutamyl transpeptidase, was suppressed by GH administration in heart and liver. The activity of glutathione-S-transferase, an enzyme involved in detoxification, was also affected by GH treatment. Taken together, the current results along with data from previous studies support a role for growth hormone in the regulation of antioxidative defense and ultimately, life span in organisms with altered GH or IGF-1 signaling.


Subject(s)
Glutathione/metabolism , Growth Hormone/administration & dosage , Longevity/drug effects , Methionine/metabolism , Signal Transduction/drug effects , Animals , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Liver/metabolism , Longevity/genetics , Mice , Oxidation-Reduction/drug effects , Swine
18.
Exp Biol Med (Maywood) ; 229(10): 988-95, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15522834

ABSTRACT

DNA methylation is an important epigenetic mechanism of transcriptional control. DNA methylation plays an essential role in maintaining cellular function, and changes in methylation patterns may contribute to the development of cancer. Aberrant methylation of DNA (global hypomethylation accompanied by region-specific hypermethylation) is frequently found in tumor cells. Global hypomethylation can result in chromosome instability, and hypermethylation has been associated with the inaction of tumor suppressor genes. Preclinical and clinical studies suggest that part of the cancer-protective effects associated with several bioactive food components may relate to DNA methylation patterns. Dietary factors that are involved in one-carbon metabolism provide the most compelling data for the interaction of nutrients and DNA methylation because they influence the supply of methyl groups, and therefore the biochemical pathways of methylation processes. These nutrients include folate, vitamin B(12), vitamin B(6), methionine, and choline. However, looking at individual nutrients may be too simplistic. Dietary methyl (folate, choline, and methionine) deficiency in combination causes decreased tissue S-adeno-sylmethionine, global DNA hypomethylation, hepatic steatosis, cirrhosis, and ultimately hepatic tumorigenesis in rodents in the absence of carcinogen treatment. Other dietary components such as vitamin B(12), alcohol, and selenium may modify the response to inadequate dietary folate.


Subject(s)
DNA Methylation , Diet , Neoplasms/etiology , Animals , Choline Deficiency/metabolism , Choline Deficiency/pathology , Deficiency Diseases/genetics , Deficiency Diseases/metabolism , Deficiency Diseases/pathology , Disease Susceptibility , Folic Acid/administration & dosage , Folic Acid Deficiency/metabolism , Folic Acid Deficiency/pathology , Humans , Methionine/deficiency , Methionine/metabolism , Neoplasms/genetics , Selenium/administration & dosage , Selenium/deficiency
19.
Ann N Y Acad Sci ; 1019: 317-20, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15247035

ABSTRACT

Reduced signaling of the growth hormone (GH)/insulin-like growth factor-1(IGF-1)/insulin pathway is associated with extended life span in several species. Ames dwarf mice are GH and IGF-1 deficient and live 50-64% longer than wild-type littermates (males and females, respectively). Previously, we have shown that Ames mice exhibit elevated levels of antioxidative enzymes and lower oxidative damage. To further explore the relationship between GH and antioxidant expression, we administered GH or saline to dwarf mice and evaluated components of the glutathione (GSH) synthesis and degradation system. Growth hormone treatment significantly elevated kidney gamma-glutamyl-cysteine synthetase protein levels in 3- and 12-month-old dwarf mice. In contrast, the activity of the GSH degradation enzyme, gamma-glutamyl transpeptidase, was suppressed by GH administration in brain (P <.05), kidney (P <.01), heart (P <.005), and liver (P <.06). Activity levels of the detoxification enzyme, glutathione-S-transferase, were also suppressed in kidney tissues at 3 and 12 months of age and in 12-month-old dwarf liver tissues (P <.05). Taken together, the current results along with data from previous studies support a role for growth hormone in the regulation of antioxidative defense and, ultimately, life span in organisms with altered GH or IGF-1 signaling.


Subject(s)
Glutathione/metabolism , Growth Hormone/physiology , Animals , Antioxidants/metabolism , Body Weight , Brain/metabolism , Female , Glutathione Transferase/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Kidney/metabolism , Male , Mice , Mice, Mutant Strains , Organ Size , Oxygen/metabolism , Signal Transduction , Swine
20.
J Nutr ; 133(9): 2907-14, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12949386

ABSTRACT

Several observations suggest a role for DNA methylation in cancer pathogenesis. Although both selenium and folate deficiency have been shown to cause global DNA hypomethylation and increased cancer susceptibility, the nutrients have different effects on one-carbon metabolism. Thus, the purpose of this study was to investigate the interactive effects of dietary selenium and folate. Weanling, Fischer-344 rats (n = 23/diet) were fed diets containing 0 or 2.0 mg selenium (as selenite)/kg and 0 or 2.0 mg folate/kg in a 2 x 2 factorial design. After 3 and 4 wk of a 12-wk experiment, 19 rats/diet were injected intraperitoneally with dimethylhydrazine (DMH, 25 mg/kg) and 4 rats/diet were administered saline. Selenium deficiency decreased (P < 0.05) colonic DNA methylation and the activities of liver DNA methyltransferase and betaine homocysteine methyltransferase and increased plasma glutathione concentrations. Folate deficiency increased (P < 0.05) the number of aberrant crypts per aberrant crypt foci, the concentration of colonic S-adenosylhomocysteine and the activity of liver cystathionine synthase. Selenium and folate interacted (P < 0.0001) to influence one-carbon metabolism and cancer susceptibility such that the number of aberrant crypts and the concentrations of plasma homocysteine and liver S-adenosylhomocysteine were the highest and the concentrations of plasma folate and liver S-adenosylmethionine and the activity of liver methionine synthase were the lowest in rats fed folate-deficient diets and supplemental selenium. These results suggest that selenium deprivation ameliorates some of the effects of folate deficiency, probably by shunting the buildup of homocysteine (as a result of folate deficiency) to glutathione.


Subject(s)
Antioxidants/administration & dosage , Carbon/metabolism , Carcinogens/administration & dosage , Colon/drug effects , Colon/pathology , Dimethylhydrazines/administration & dosage , Folic Acid/administration & dosage , Selenium/administration & dosage , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Animals , Colon/metabolism , Cystathionine beta-Synthase/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Deficiency Diseases/genetics , Deficiency Diseases/metabolism , Deficiency Diseases/pathology , Diet , Disease Susceptibility , Drug Interactions , Folic Acid/blood , Glutathione/blood , Homocysteine/blood , Injections, Intraperitoneal , Liver/enzymology , Liver/metabolism , Male , Rats , Rats, Inbred F344 , S-Adenosylhomocysteine/metabolism , Selenium/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...