当前位置: 首页 > 期刊 > 《糖尿病学杂志》 > 2005年第11期 > 正文
编号:11274631
Homocysteine Metabolism in ZDF (Type 2) Diabetic Rats
http://www.100md.com 《糖尿病学杂志》
     1 Department of Biochemistry, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada

    2 Department of Nutrition, University of North Carolina, Chapel Hill, North Carolina

    Key Words: BHMT, betaine:homocysteine methyltransferase CBS, cystathionine -synthase CGL, cystathionine -lyase GNMT, glycine N-methyltransferase Hcy, homocysteine MAT, methionine adenosyltransferase MTHFR, methylenetetrahydrofolate reductase SAH, S-adenosylhomocysteine SAM, S-adenosylmethionine tHcy, total homocysteine

    ABSTRACT

    Mild hyperhomocysteinemia is a risk factor for many diseases, including cardiovascular disease. We determined the effects of insulin resistance and of type 2 diabetes on homocysteine (Hcy) metabolism using Zucker diabetic fatty rats (ZDF/Gmi fa/fa and ZDF/Gmi fa/). Plasma total Hcy was reduced in ZDF fa/fa rats by 24% in the pre-diabetic insulin-resistant stage, while in the frank diabetic stage there was a 59% reduction. Hepatic activities of several enzymes that play a role in the removal of Hcy:cystathionine -synthase (CBS), cystathionine -lyase, and betaine:Hcy methyltransferase (BHMT) were increased as was methionine adenosyltransferase. CBS and BHMT mRNA levels and the hepatic level of S-adenosylmethionine were also increased in the ZDF fa/fa rats. Studies with primary hepatocytes showed that Hcy export and the transsulfuration flux in cells from ZDF fa/fa rats were particularly sensitive to betaine. Interestingly, liver betaine concentration was found to be significantly lower in the ZDf fa/fa rats at both 5 and 11 weeks. These results emphasize the importance of betaine metabolism in determining plasma Hcy levels in type 2 diabetes.

    Patients with coronary, cerebrovascular, or peripheral arterial disease have mean plasma total homocysteine (tHcy) levels significantly higher than control subjects (1). Over the last decade, these findings led to the identification of hyperhomocysteinemia as an independent risk factor for vascular disease (1). In addition, hyperhomocysteinemia has also been identified as a risk factor for Alzheimer’s disease (2) and osteoporotic fractures (3).

    Homocysteine (Hcy) is formed after the donation of a methyl group from S-adenosylmethionine (SAM). It can then be metabolized by one of three enzymes. Methionine synthase and betaine:Hcy methyltransferase (BHMT) catalyze the remethylation of Hcy to methionine, and cystathionine -synthase (CBS) forms part of the transsulfuration pathway where Hcy condenses with serine to form cystathionine. Figure 1 shows the pathway of methionine metabolism.

    Diabetes, whether type 1 or type 2, is associated with an increased risk of cardiovascular mortality, with the prevalence of atherosclerosis being two- to sixfold higher in diabetic patients than in people without diabetes (4). Insulin resistance, which immediately precedes the development of type 2 diabetes, is also associated with an increased risk of coronary artery disease (5). Hyperhomocysteinemia has been shown to be a stronger risk factor for cardiovascular disease and for mortality in patients with type 2 diabetes than in subjects without diabetes (6).

    Plasma tHcy in diabetic patients is known to be dependent on the presence or absence of nephropathy. Both type 1 and type 2 diabetic patients with nephropathy have elevated levels of tHcy (6,7). However, type 1 diabetic patients with no renal complications have plasma tHcy levels lower than controls (8). This decrease was also shown in an animal model of type 1 diabetes (9).

    We, therefore, examined the effect of insulin resistance and type 2 diabetes on plasma tHcy and its metabolism in the liver. We used the leptin receptoreCdefective Zucker diabetic fatty rat (ZDF), an excellent model for type 2 diabetes (10). Before developing frank diabetes, they go through a phase of insulin resistance, thereby giving us an opportunity to study the effects of both insulin resistance and of type 2 diabetes.

    RESEARCH DESIGN AND METHODS

    Animals and tissue sampling.

    All procedures were approved by Memorial University’s Institutional Animal Care Committee. Male ZDF rats (ZDF/Gmi fa/fa) aged 5 and 11 weeks and male lean rats (ZDF/Gmi fa/) of the same ages were obtained from Charles River Laboratories, Indianapolis, Indiana. They were fed Purina 5008 chow ad libitum and had free access to water. On the day of the experiment, animals were anesthetized with an intraperitoneal injection of sodium pentobarbital (65 mg/kg), and blood was collected from the abdominal aorta into heparinized syringes. Before removal of the syringe, a piece of the liver was rapidly removed and freeze-clamped with aluminum tongs precooled in liquid nitrogen. Another portion was removed for immediate enzyme assays. The freeze-clamped liver was stored at eC70°C until further use. The blood was centrifuged at 3,700g for 15 min, and the plasma was kept at eC20°C until further use.

    Analytical procedures

    Plasma metabolites.

    Plasma tHcy was measured as described (11). Plasma glucose was determined enzymatically (12). Plasma creatinine was measured using a kit (catalog no. 555A) from Sigma Diagnostics (St. Louis, MO). Plasma insulin was measured with a rat insulin enzyme-linked immunosorbent assay kit (catalog no. INSKR020) from Crystal Chem (Chicago, IL) using anti-rat/mouse insulin antibody and a rat insulin standard.

    Enzyme assays.

    Fresh liver samples were diluted 1:5 with 50 mmol/l K phosphate buffer (pH 7.0) and homogenized with a Polytron (Brinkman Instruments, Toronto, Canada) for 20 s at 50% output. The homogenates were centrifuged at 20,000g for 30 min at 4°C, and the supernatant was removed and used immediately to measure the activities of five of the enzymes of methionine metabolism. Methionine adenosyltransferase (MAT) (13,14), CBS (13,15), cystathionine -lyase (CGL) (16), methionine synthase (17), and BHMT (18) were assayed using previously described methods. Methylenetetrahydrofolate reductase (MTHFR) (19) and glycine N-methyltransferase (GNMT) (20) were measured using previously frozen liver tissue. Choline dehydrogenase was measured using liver homogenates (21,22). Protein concentrations were determined by the Biuret method using BSA as a standard. All enzyme assays were determined to be linear with time and with protein.

    Liver metabolites.

    Choline and its metabolites were extracted from tissue and plasma by the method of Bligh and Dyer (23). Aqueous and organic compounds were separated, analyzed, and quantified directly by high-performance liquid chromatographyeCmass spectrometry (liquid chromatographyeCelectrospray ionizationeCisotope dilution mass spectrometry) after the addition of internal standards labeled with stable isotopes that were used to correct for recovery (24). The levels of SAM and S-adenosylhomocysteine (SAH) were measured in freeze-clamped liver tissue (25).

    mRNA abundance.

    Total RNA was isolated from the fresh liver samples as previously described (26). Two micrograms of RNA were reverse transcribed using a one-step reverse transcription kit (Qiagen, Mississauga, Canada) and was amplified by 27 cycles. An upstream primer (5'-GCCTTCAGGACATCCAGTGT-3') and a downstream primer (5'-TCTTTCCGGGTCTGCTCACGGGC-3') corresponding to the type 3 rat CBS cDNA (27) were used to amplify a 1,644-bp PCR fragment. For BHMT, an upstream primer (5'-ACCGATTGCCGGCAAGAA-3') and a downstream primer (5'-CTGTGCGGATTTGAATTTTTG-3') corresponding to the rat BHMT cDNA (NM-030850) were used to amplify a 1,216-bp PCR fragment. A 768-bp fragment of the rat -actin was coamplified using amplimer set primers (CLONTECH, Palo Alto, CA). Once amplified, the PCR products were separated on 0.8% agarose. The bands were stained with ethidium bromide and were visualized under UV illumination.

    Homocysteine metabolism in isolated hepatocytes.

    Hepatocytes were isolated from ZDF fa/fa and ZDF fa/ rats (28). Approximately 5 mg of hepatocytes (dry weight) were preincubated for 20 min at 37°C with Krebs-Henseleit saline with or without 0.3 mmol/l betaine. At the end of the preincubation L-[1-14C] methionine was added to a final concentration of 0.1 mmol/l and the incubation continued for a further 30 min. The total transsulfuration flux was determined as the sum of the 14C present in -ketobutyrate and in CO2 as previously described (29). In a separate series of experiments, hepatocytes were incubated with 0.1 mmol/l methionine ± 0.3 mmol/l betaine to measure Hcy export (29).

    Statistical analysis.

    Data are presented as means ± SD. Means were compared using Student’s unpaired t test, paired t test, or one-way ANOVA as appropriate. P < 0.05 was taken to indicate a significant difference.

    RESULTS

    The body weight, hepatosomatic index, plasma insulin, creatinine, and glucose concentrations of these rats have been reported (30). At 5 weeks, the concentrations of plasma glucose (12.1 ± 1.7 vs. 8.4 ± 1.3 mmol/l) and insulin (1.5 ± 0.5 vs. 0.14 ± 0.07 nmol/l) in the ZDF fa/fa and ZDF fa/ rats, respectively, showed that the ZDF fa/fa rats had become insulin resistant. At 11 weeks, the concentrations of plasma glucose (30.3 ± 1.6 vs. 10.9 ± 0.8 mmol/l) and insulin (0.61 ± 0.14 vs. 0.68 ± 0.1 nmol/l) in the ZDF fa/fa and the ZDF fa/ rats, respectively, showed that the ZDF fa/fa rats had become diabetic. Creatinine levels of ZDF fa/fa and control rats at 5 weeks were similar (32.4 ± 3.8 vs. 28.9 ± 3.1 e蘭ol/l), but at 11 weeks were lower in the ZDF fa/fa rats (18.9 ± 2.7 vs. 29.7 ± 6.1 e蘭ol/l)

    Plasma Hcy and hepatic enzymes of Hcy metabolism.

    At 5 weeks, plasma tHcy was about 25% lower in the ZDF fa/fa rats, while at 11 weeks it was reduced by about 60% (Table 1). The plasma tHcy level in the ZDF fa/fa rats at 11 weeks was further reduced from its value at 5 weeks, whereas the ZDF fa/ rats showed an increase in the plasma tHcy level with age. The reduced plasma tHcy at 5 weeks in the ZDF fa/fa rats was accompanied by changes in several enzymes that metabolize methionine (Table 1). Of the enzymes responsible for producing Hcy, MAT and GNMT showed slight but significant increases. CBS, CGL, BHMT, and MTHFR were increased, respectively, by about 79, 35, 80, and 22%. Methionine synthase showed a slight but significant decrease in the ZDF fa/fa rats. At 11 weeks, MAT, GNMT, CBS, CGL, and BHMT all showed significant increases in activity as did choline dehydrogenase.

    In addition to the differences in enzyme activity between the control and the experimental animals at each age-group, MAT, CBS, and GNMT all showed significant increases in their activity with age in the ZDF fa/ rats.

    mRNA levels.

    At 5 weeks, the increased activity of CBS in the ZDF fa/fa rats was accompanied by a comparable increase in CBS mRNA level (Fig. 2A). The increased BHMT activity observed in the ZDF fa/fa rats at 5 and 11 weeks was also accompanied by comparable increases in mRNA levels (Fig. 2B).

    SAM, SAH, and SAM-to-SAH ratio.

    The hepatic levels of SAM and SAH and the SAM-to-SAH ratio are shown in Table 2. At both 5 and 11 weeks, SAM was significantly increased in the ZDF fa/fa rats. The SAM-to-SAH ratio was significantly higher in the ZDF fa/fa rats at 5 weeks. Both SAM and the SAM-to-SAH ratio showed increases with age in ZDF fa/ rats, consistent with the age-related increase in MAT activity.

    Transsulfuration flux.

    In a preliminary study, we investigated the effect of betaine on the transsulfuration flux, as measured by the conversion of L-[1-14C] methionine to 14CO2 and 14C--ketobutyrate by isolated hepatocytes from Sprague Dawley rats. All concentrations of betaine used (0.1, 0.3, or 1 mmol/l) significantly reduced the catabolism of methionine, compared with incubations of hepatocytes with methionine alone. We then conducted studies with hepatocytes from ZDF rats using 0.1 mmol/l L-[1-14C] methionine and 0.3 mmol/l betaine. Incubation with 0.3 mmol/l betaine significantly decreased the transsulfuration flux in hepatocytes of ZDF fa/fa rats at both ages, but the reduction seen in the hepatocytes of ZDF fa/ rats was significant only at 11 weeks of age (Table 3).

    Hcy export.

    Hepatocytes from ZDF fa/fa and ZDF fa/ rats, at both ages, showed a significant reduction in Hcy export when incubated with 0.3 mmol/l betaine (Table 4). However, at both ages the decrease observed in the ZDF fa/fa rats was about twofold greater than that of the ZDF fa/ rats.

    Choline and related metabolites.

    Table 5 shows the liver concentrations of choline and related metabolites. The liver betaine concentration was lower in the ZDF fa/fa rats at both 5 and 11 weeks, while there was no difference in the choline concentration. One of the most remarkable findings was the high level of betaine found in the liver of the young animals and the reduction that occurs with age. Hepatic glycerophosphorylcholine was significantly higher in the ZDF fa/fa rats at 5 weeks, though at 11 weeks it was significantly lower. Phosphorylcholine was significantly higher in the ZDF fa/fa rats at both ages. Both glycerophosphorylcholine and phosphorylcholine levels increased with age in the ZDF fa/ rats. Phosphatidylcholine was significantly decreased in ZDF fa/fa rats at 11 weeks, while sphingomyelin was significantly reduced at both 5 and 11 weeks.

    Table 6 shows choline and related metabolite concentrations in the plasma. The only significant difference in plasma metabolites between the different rats was an increased phosphatidylcholine level in the ZDF fa/fa rats at 11 weeks. However, the plasma betaine concentration decreased with age in both groups of rats while the concentration of phosphatidylcholine increased in the ZDF fa/fa rats from 5 to 11 weeks.

    DISCUSSION

    This study describes the altered metabolism of methionine and Hcy that occurs in insulin resistance and in early type 2 diabetes. It is clear that the hormonal changes that are prevalent in these two disease states act at several sites in the pathway of methionine metabolism. Hyperphagia in the diabetic rats may also contribute to these metabolic changes. Indeed, we observed a twofold increase in food intake per 100 g body weight at the diabetic stage. However, there was no difference at 5 weeks. The increased hepatic CBS and BHMT activity at 5 weeks cannot, therefore, be attributed to increased protein intake. In addition, we have shown a direct effect of insulin in repressing CBS expression in both human and rat cultured hepatocytes (42). Our finding of lowered plasma tHcy in the 5-week ZDF fa/fa rats shows that insulin resistance alone can decrease plasma tHcy levels.

    The most striking finding in the present study concerns the role of betaine and BHMT in determining plasma tHcy. Not only was BHMT activity and mRNA levels elevated but the hepatic betaine concentration was remarkably reduced. Furthermore, this increased BHMT was clearly effective in decreasing hepatic Hcy output. This is evident from the data showing that addition of betaine to hepatocytes from ZDF fa/fa rats virtually eliminated Hcy export from cells compared with a lesser effect in hepatocytes from ZDF fa/ rats (Table 4). Increased remethylation of Hcy by BHMT also reduces the Hcy available for flux through the transsulfuration pathway, which decreased in these cells (Table 3). Although the importance of the BHMT-catalyzed remethylation reaction in determining Hcy levels has not received as much attention as the folate-dependent remethylation pathway, it should be recalled that Schwahn et al. (31) found a highly significant negative correlation between plasma betaine and plasma tHcy concentrations in humans. Supplementation with betaine was shown to lower plasma tHcy in humans (32) and in MTHFR +/+, +/eC, and eC/eC mice (31). Because of the elevated BHMT activity, betaine may be particularly efficacious in reducing plasma tHcy in the later stages of type 2 diabetes when renal complications bring about hyperhomocysteinemia.

    Our data suggest that there is enhanced utilization of betaine in the ZDF fa/fa rat. The observed decrease in hepatic phosphatidylcholine concentration (Table 5) is consistent with enhanced utilization of choline to form betaine in the livers of these rats. Sphingomyelin, which is derived from phosphatidylcholine, is also reduced in fa/fa liver. The increase in phosphorylcholine concentrations may reflect increased activity of the pathway for phosphatidylcholine biosynthesis that occurs when tissue phosphatidylcholine concentrations fall. The significant increase in plasma phosphatidylcholine concentrations in ZDF fa/fa rats at 11 weeks (Table 6) probably reflects increased secretion of lipoproteins from liver.

    Another remarkable feature of betaine metabolism is the extraordinarily high hepatic betaine concentration found in young rats. In the control animals (ZDF fa/), the hepatic betaine level was 17.6 e蘭ol/g at 5 weeks; this decreased to 5.3 e蘭ol/g at 11 weeks. Assuming that hepatic betaine is evenly distributed in intracellular water and that rat liver contains 0.45 ml of cell water per gram (35), we can calculate a betaine concentration in the 5-week-old control animals of 39 mmol/l. This should be compared with total osmolarity of about 305eC310 mOs mmol/l in mammalian tissues (except for the renal medulla).

    Davies et al. (36) have reported a very large urinary excretion of betaine in normal neonates and in young rats after weaning. High betaine concentrations are found in mammalian kidneys, particularly in the renal medulla. Renal betaine has been characterized as a compatible osmolyte (together with other trimethylamines and polyols) where it can offset the high extracellular osmolarity that arises as a result of the urinary concentrating mechanisms. In addition, these osmolytes may protect these cells from adverse effects of urea, which is present at high concentrations (37).

    Wettstein et al. (38) have showed that betaine plays a role in cell volume homeostasis in perfused livers of adult rats. Whether this is also the function of the high betaine concentrations in neonatal liver is a focus of our current work. We must also comment on the ratio of hepatocellular to plasma betaine. Plasma betaine was fairly constant, in the 90eC170 e蘭ol/l range, in all of the rats (Table 6). The liver-to-plasma concentration ratios for betaine were about 230, 130, 100, and 60, respectively, for ZDF fa/ and ZDF fa/fa rats at 5 and 11 weeks of age. How such gradients are produced and maintained are important issues for future work.

    Diabetes is characterized not only by the lack of insulin or the resistance to the action of insulin, but also by increases in the counter-regulatory hormones glucocorticoid and glucagon (39,40). Hepatic MAT activity has been shown to be increased by glucocorticoid administration (41). Finkelstein et al. (33) examined the effects of administration of a number of hormones on BHMT activity. The principal findings were that hydrocortisone increased BHMT activity, whereas thyroxin decreased it. This group also demonstrated increased activity in alloxan diabetic rats. This latter finding has been confirmed by Nieman et al. (34) in streptozotocin-induced diabetic rats. We are currently exploring the possibility that insulin may regulate BHMT expression. Insulin treatment of streptozotocin-induced diabetic rats was shown to restore the increased activity of CBS to normal levels (9) by acting at the level of CBS gene transcription to repress CBS promoter activity (42). Cyclic AMP, the intracellular messenger of glucagon, and glucocorticoids increase CBS expression in rat hepatoma cells (43). The increased activity of CBS, brought about both by allosteric activation by the increased hepatic SAM levels (44) and by increased expression, does not imply an increased transsulfuration flux. This flux is determined by the rate at which methionine enters into hepatic metabolism, which will be largely determined by dietary methionine consumption. It seems that the major effect of increased CBS activity is to decrease the steady-state Hcy concentration at which transsulfuration occurs (45). In this way it contributes to the decreased plasma tHcy concentration. Similarly, the increased activity of BHMT does not necessarily imply an increased remethylation flux in vivo but, rather, it decreases the steady-state Hcy concentration at which remethylation via BHMT occurs.

    It is also possible that alterations in renal function may contribute, somewhat, to the decreased plasma tHcy, at least when the rats are diabetic. The kidney is a major site for the removal and subsequent metabolism of Hcy (46). Hyperfiltration is a characteristic sign of renal dysfunction in early diabetes (47). Such hyperfiltration has been reported to occur in ZDF fa/fa rats from about 7 weeks of age and to continue until they are about 3 months old (48). Such an occurrence probably accounts for the decreased plasma creatinine seen at 11 weeks. The delivery of an increased quantity of Hcy to the kidney via filtration could result in increased renal catabolism of Hcy and contribute to its reduced plasma concentration. However, this cannot contribute to the decreased Hcy at the insulin-resistant stage where creatinine levels were unchanged.

    It is now clear that plasma tHcy levels decrease in both type 1 and type 2 diabetes when there is no renal damage. The present work showed that this is true even at the prediabetic, insulin-resistant stage. However, literature reports on the phenomenon show a variety of responses of Hcy to insulin resistance (5,49,50). This variability is likely to result from different degrees of insulin resistance as well as other factors such as impaired renal function. Our results, which at 5 weeks were obtained in an animal model in which there was no evidence of impaired renal function as well as a consistent degree of insulin resistance, agree remarkably well with Rosolova’s study of the relationship between Hcy and insulin resistance in healthy human subjects (5).

    In summary, this study shows a decreased plasma tHcy level in both insulin-resistant and type 2 diabetic rats. We show increased activities of BHMT and CBS in these states, as well as increased mRNA levels for these enzymes. We also show increased hepatic SAM levels, which will activate CBS. We report experiments in hepatocytes that directly demonstrate the role of betaine metabolism in enhanced Hcy removal. These results emphasize the importance of BHMT in regulating Hcy metabolism.

    ACKNOWLEDGMENTS

    This work was supported by a grant from the Canadian Diabetes Association in honor of the late Neil M. Miller (M.E.B. and J.T.B.) and by a grant from the National Institutes of Health (DK55865 to S.H.Z.). E.P.W. received a graduate fellowship from the School of Graduate Studies, Memorial University of Newfoundland. J.T.B. is a Senior Investigator of the Canadian Institutes of Health Research.

    REFERENCES

    Kang SS, Wong PWK, Malinow MR: Hyperhomocyst(e)inemia as a risk factor for occlusive vascular disease. Annu Rev Nutr12 :279 eC298,1992

    Seshadri S, Beiser A, Selhub J, Jacques PF, Rosenberg IH, D’Agostino RB, Wilson PWF, Wolf PA: Plasma homocysteine as a risk factor for dementia and Alzheimer’s disease. N Engl J Med346 :476 eC483,2002

    van Meurs JBJ, Dhonukshe-Rutten RAM, Pluijm SMF, van der Klift M, de Jonge R, Lindemans J, de Groot LCPGM, Hofman A, Witteman JCM, van Leeuwen JPTM, Breteler MMB, Lips P, Pols HAP, Uitterlinden AG: Homocysteine levels and the risk of osteoporotic fracture. N Engl J Med350 :2033 eC2041,2004

    Brand FN, Abbott RD, Kannel WB: Diabetes, intermittent claudication, and risk of cardiovascular events: the Framingham study. Diabetes38 :504 eC509,1989

    Rosolova H, Simon J, Mayer O, Racek J, Dierze T, Jacobson DW: Unexpected inverse relationship between insulin resistance and serum homocysteine in healthy subjects. Physiol Res51 :93 eC98,2002

    Hoogeveen EK, Kostense PJ, Beks PJ, Mackaay AJ, Jacobs C, Bouter LM, Heine RJ, Stehouwer DA: Hyperhomocysteinemia is associated with an increased risk of cardiovascular disease, especially in non-insulin-dependent diabetes mellitus. Arterioscler Thromb Vasc Biol18 :133 eC138,1998

    Hultberg B, Agargh E, Andersson A, Brattstrom L, Isaksson A, Israelsson B, Agardh, CD: Increased levels of plasma homocysteine are associated with nephropathy, but not severe retinopathy in type 1 diabetes mellitus. Scand J Clin Lab Invest51 :277 eC282,1991

    Robillon JF, Canivet B, Candito M, Sadoul JL, Jullien D, Morand P, Chambon P, Freychet P: Type 1 diabetes mellitus and homocyst(e)ine. Diabetes Metab20 :494 eC496,1994

    Jacobs RL, House JD, Brosnan ME, Brosnan JT: Effects of streptozotocin-induced diabetes and of insulin treatment on homocysteine metabolism in the rat. Diabetes47 :1967 eC1970,1998

    Peterson RG, Shaw WN, Noel M-A, Little LA, Eichberg J: Zucker Diabetic Fatty rat as a model for non-insulin dependent diabetes mellitus. Ilar News32 :16 eC19,1990

    Vester B, Rasmussen K: High performance liquid chromatography method for rapid and accurate determination of homocysteine in plasma and serum. Eur J Clin Chem Clin Biochem29 :549 eC554,1991

    Bergmeyer HU, Bernt E, Schmidt F, Stork H: D-glucose: determination with hexokinase and glucose-6-phosphate dehydrogenase. In Methods of Enzymatic Analysis, Vol. 3, 2nd ed. Bergmeyer HU, Ed. Deerfield Beach, Florida, Verleg Chemie,1974 , p.1196 eC1201

    Mudd SH, Finkelstein JD, Irreverre F, Laster L: Transsulfuration in mammals: microassays and tissue distribution of three enzymes of the pathway. J Biol Chem240 :4382 eC4392,1965

    Duce AM, Ortiz P, Cabrero C, Mato JM: S-adenosyl-L-methionine synthetase and phospholipid methyltransferases are inhibited in human cirrhosis. Hepatology8 :65 eC68,1988

    Taoka S, Ohja S, Shan X, Kruger WD, Banerjee R: Evidence for heme-mediated redox regulation of human cystathionine beta-synthase activity. J Biol Chem273 :25179 eC25184,1998

    Stipanuk MH: Effect of excess dietary methionine on the catabolism of cysteine in rats. J Nutr109 :2126 eC2139,1979

    Koblin DD, Watson JE, Deady JE, Stokstad ELR, Eger EI 2nd: Inactivation of methionine synthetase by nitrous oxide in mice. Anesthesiology54 :318 eC324,1981

    Garrow TA: Purification, kinetic properties, and cDNA cloning of mammalian betaine:homocysteine methyltransferase. J Biol Chem271 :22831 eC22838,1996

    Engberson AMT, Franken DG, Boers GHJ, Stevens EMB, Trijbels FJM, Blom HJ: Thermolabile 5,10-methylenetetrahydrofolate reductase as a cause of mild hyperhomocysteinemia. Am J Hum Genet56 :142 eC150,1995

    Rowling MJ, McMullen MH, Chipman DC, Schalinske KL: Hepatic glycine N-methyltransferase is up-regulated by excess dietary methionine in rats. J Nutr132 :2545 eC2550,2002

    Grossman EB, Herbert SC: Renal inner medullary choline dehydrogenase activity: characterization and modulation. Am J Physiol25 :F107 eCF112,1989

    Haubrich DR, Gerber NH: Choline dehydrogenase: assay, properties and inhibitors. Biochem Pharmacol30 :2993 eC3000,1981

    Bligh EG, Dyer WJ: A rapid method of total lipid extraction and purification. Can J Med Sci37 :911 eC917,1959

    Koc H, Mar MH, Ranasinghe A, Swenberg JA, Zeisel SH: Quantitation of choline and its metabolites in tissues and foods by liquid chromatography/electrospray ionization-isotope dilution mass spectrometry. Anal Chem74 :4734 eC4740,2002

    Jacobs RL, Stead LM, Brosnan ME, Brosnan JT: Hyperglucagonemia in rats results in decreased plasma homocysteine and increased flux through the transsulfuration pathway in liver. J Biol Chem276 :43740 eC43747,2001

    Chomczynski P, Sacchi N: Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction. Anal Chem162 :156 eC159,1987

    Kraus JP, Oliveriusova J, Sokolova J, Kraus E, Vlcek C, Franchis R, Maclean KN, Bao L, Bukovska G, Patterson D, Paces V, Ansorge W, Kozich, V: The human cystathionine synthase (CBS) gene: complete sequence, alternate splicing, and polymorphism. Genomics52 :312 eC324,1998

    Berry MN, Edwards AM, Barritt GJ: High yield preparation of isolated hepatocytes from rat liver. In Laboratory Techniques in Biochemistry and Molecular Biology. Vol. 21. Burdon RH, Van Knippenburg PH, Eds. Oxford, U.K., Elsevier,1991 , p.44 eC57

    Stead LM, Brosnan ME, Brosnan JT: Characterization of homocysteine metabolism in the rat liver. Biochem J350 :685 eC692,2000

    Wijekoon EP, Skinner C, Brosnan ME, Brosnan JT: Amino acid metabolism in the Zucker Diabetic Fatty rat: effects of insulin resistance and of type 2 diabetes. Can J Physiol Pharmacol82 :506 eC514,2004

    Schwahn BC, Chen Z, Laryea MD, Wendl U, Lussier-Cacan S, Genest J Jr, Mar M-H, Zeisel SH, Castro C, Garrow T, Rozen R: Homocysteine-betaine interactions in a murine model of 5,10-methylenetetrahydrofolate reductase deficiency. FASEB J17 :512 eC514,2003

    Steenge GR, Verhoef P, Katan MB: Betaine supplementation lowers plasma homocysteine in healthy men and women. J Nutr133 :1291 eC1295,2003

    Finkelstein JD, Kyle WE, Harris BJ: Methionine metabolism in mammals: regulation of homocysteine methyltransferases in rat tissue. Arch Biochem Biophys146 :84 eC92,1971

    Nieman KM, Rowling MJ, Garrow TA, Schalinske KL: Modulation of methyl group metabolism by streptozotocin-induced diabetes and all-trans-retinoic acid. J Biol Chem279 :45708 eC45712,2004

    Qian D, Brosnan JT: Administration of Escherichia coli endotoxin to rat increases liver mass and hepatocyte volume in vivo. Biochem J313 :479 eC486,1996

    Davies SE, Chalmers RA, Randall EW, Iles RA: Betaine metabolism in human neonates and developing rats. Clin Chim Acta178 :241 eC249,1988

    Beck FX, Burger-Kentischer A, Muller E: Cellular response to osmotic stress in the renal medulla. Pflugers Arch436 :814 eC827,1998

    Wettstein M, Peters-Regehr T, Kubitz R, Fischer R, Holneicher C, Monnighoff I, Haussinger D: Release of osmolytes induced by phagocytosis and hormones in rat liver. Am J Physiol Gastrointest Liver Physiol278 :G227 eCG233,2000

    Stubbs M, York DA: Central glucocorticoid regulation of parasympathetic drive to pancreatic -cells in the obese fa/farat. Int J Obes 15 :547 eC553,1991

    Consoli A, Nurjhan N, Reilly JJ Jr, Bier DM, Gerich JE: Mechanism of increased gluconeogenesis in noninsulin-dependent diabetes mellitus: role of alterations in systemic, hepatic, and muscle lactate and alanine metabolism. J Clin Invest86 :2038 eC2045,1990

    Gil B, Pajares MA, Mato JM, Alvarez L: Glucocorticoid regulation of hepatic S-adenosylmethionine synthetase gene expression. Endocrinology138 :1251 eC1258,1997

    Ratnam S, Maclean KN, Jacobs RL, Brosnan ME, Kraus JP, Brosnan JT: Hormonal regulation of cystathionine synthase expression in liver. J Biol Chem277 :42912 eC42918,2002

    Goss SJ: Characterization of cystathionine synthase as a selectable, liver specific trait in rat hepatomas. J Cell Sci82 :309 eC320,1986

    Finkelstein JD, Kyle WE, Martin JL, Pick AM: Activation of cystathionine synthase by adenosylmethionine and adenosylethionine. Biochem Biophys Res Commun66 :81 eC87,1975

    Reed MC, Nijhout HF, Sparks R, Ulrich CM: A mathematical model of the methionine cycle. J Theor Biol226 :33 eC43,2004

    Bostom A, Brosnan JT, Hall B, Nadeau MR, Selhub J: Net uptake of plasma homocysteine by the rat kidney in vivo. Atherosclerosis116 :59 eC62,1995

    Thomson SC, Vallon V, Blantz RC: Kidney function in early diabetes: the tubular hypothesis of glomerular filtration. Am J Physiol Renal Physiol286 :F8 eCF15,2004

    Hoshi S, Shu Y, Yoshida F, Inagaki T, Sonoda J, Watanage T, Nomoto K-I, Nagata M: Podocyte injury promotes progressive nephropathy in Zucker diabetic fatty rats. Lab Invest82 :25 eC35,2002

    Meigs JB, Jacques PF, Selhub J, Singer DE, Nathan DM, Rifai N, D’Agostino RB, Wilson PWF: Fasting plasma homocysteine levels in the insulin resistance syndrome: the Framingham Offspring Study. Diabetes Care24 :1403 eC1410,2001

    Abbasi F, Facchini F, Humphreys MH, Reaven GM: Plasma homocysteine concentrations in healthy volunteers are not related to differences in insulin-mediated glucose disposal. Atherosclerosis146 :175 eC178,1999(Enoka P. Wijekoon, Beatri)