Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Enzyme Inhib Med Chem ; 38(1): 2163242, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36629431

ABSTRACT

Androgen deprivation therapy (ADT) is a common treatment for recurrent prostate cancer (PC). However, after a certain period of responsiveness, ADT resistance occurs virtually in all patients and the disease progresses to lethal metastatic castration-resistant prostate cancer (mCRPC). Aberrant expression and function of the epigenetic modifiers EZH2 and BET over activates c-myc, an oncogenic transcription factor critically contributing to mCRPC. In the present work, we tested, for the first time, the combination of an EZH2 inhibitor with a BET inhibitor in metastatic PC cells. The combination outperformed single drugs in inhibiting cell viability, cell proliferation and clonogenic ability, and concomitantly reduced both c-myc and NF-kB expression. Although these promising results will warrant further in vivo validation, they represent the first step to establishing the rationale that the proposed combination might be suitable for mCRPC treatment, by exploiting molecular targets different from androgen receptor.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms, Castration-Resistant , Humans , Male , Androgen Antagonists/pharmacology , Androgen Antagonists/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/pathology , Transcription Factors , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/metabolism
2.
PLoS One ; 16(5): e0250486, 2021.
Article in English | MEDLINE | ID: mdl-33975330

ABSTRACT

Research into the epigenome is of growing importance as a loss of epigenetic control has been implicated in the development of neurodegenerative diseases. Previous studies have implicated aberrant DNA and histone methylation in multiple sclerosis (MS) disease pathogenesis. We have previously reported that the methyl donor betaine is depleted in MS and is linked to changes in histone H3 trimethylation (H3K4me3) in neurons. We have also shown that betaine increases histone methyltransferase activity by activating chromatin bound betaine homocysteine S-methyltransferase (BHMT). Here, we investigated the role of the BHMT-betaine methylation pathway in oligodendrocytes. Immunocytochemistry in the human MO3.13 cell line, primary rat oligodendrocytes, and tissue from MS postmortem brain confirmed the presence of the BHMT enzyme in the nucleus in oligodendrocytes. BHMT expression is increased 2-fold following oxidative insult, and qRT-PCR demonstrated that betaine can promote an increase in expression of oligodendrocyte maturation genes SOX10 and NKX-2.2 under oxidative conditions. Chromatin fractionation provided evidence of a direct interaction of BHMT on chromatin and co-IP analysis indicates an interaction between BHMT and DNMT3a. Our data show that both histone and DNA methyltransferase activity are increased following betaine administration. Betaine effects were shown to be dependent on BHMT expression following siRNA knockdown of BHMT. This is the first report of BHMT expression in oligodendrocytes and suggests that betaine acts through BHMT to modulate histone and DNA methyltransferase activity on chromatin. These data suggest that methyl donor availability can impact epigenetic changes and maturation in oligodendrocytes.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Betaine/metabolism , Multiple Sclerosis/pathology , Oligodendroglia/drug effects , Animals , Betaine/pharmacology , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/genetics , Brain/metabolism , Brain/pathology , Cells, Cultured , Chromatin/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Epigenesis, Genetic , Gene Expression/drug effects , Histones/metabolism , Humans , Methionine/metabolism , Methylation , Multiple Sclerosis/genetics , Nitroprusside/pharmacology , Oligodendroglia/cytology , Oligodendroglia/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , SOXE Transcription Factors/metabolism
3.
FASEB J ; 29(3): 1069-79, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25466894

ABSTRACT

The embryonic pattern of global DNA methylation is first established in the inner cell mass (ICM) of the mouse blastocyst. The methyl donor S-adenosylmethionine (SAM) is produced in most cells through the folate cycle, but only a few cell types generate SAM from betaine (N,N,N-trimethylglycine) via betaine-homocysteine methyltransferase (BHMT), which is expressed in the mouse ICM. Here, mean ICM cell numbers decreased from 18-19 in controls to 11-13 when the folate cycle was inhibited by the antifolate methotrexate and to 12-14 when BHMT expression was knocked down by antisense morpholinos. Inhibiting both pathways, however, much more severely affected ICM development (7-8 cells). Total SAM levels in mouse blastocysts decreased significantly only when both pathways were inhibited (from 3.1 to 1.6 pmol/100 blastocysts). DNA methylation, detected as 5-methylcytosine (5-MeC) immunofluorescence in isolated ICMs, was minimally affected by inhibition of either pathway alone but decreased by at least 45-55% when both BHMT and the folate cycle were inhibited simultaneously. Effects on cell numbers and 5-MeC levels in the ICM were completely rescued by methionine (immediate SAM precursor) or SAM. Both the folate cycle and betaine/BHMT appear to contribute to a methyl pool required for normal ICM development and establishing initial embryonic DNA methylation.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Blastocyst/metabolism , DNA Methylation , Embryo, Mammalian/metabolism , Folic Acid/metabolism , Gene Expression Regulation, Enzymologic , S-Adenosylmethionine/metabolism , 5-Methylcytosine/analysis , Animals , Antimetabolites, Antineoplastic/pharmacology , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Blastocyst/cytology , Blastocyst/drug effects , Cell Lineage , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/drug effects , Female , Fluorescent Antibody Technique , Liver/cytology , Liver/drug effects , Liver/metabolism , Methotrexate/pharmacology , Mice , snRNP Core Proteins/metabolism
4.
Atherosclerosis ; 236(1): 91-100, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25016363

ABSTRACT

Despite the benefit of statins in reducing cardiovascular risk, a sizable proportion of patients still remain at risk. Since HDL reduces CVD risk through a process that involves formation of pre-beta particles that facilitates the removal of cholesterol from the lipid-laden macrophages in the arteries, inducing pre-beta particles, may reduce the risk of CVD. A novel BET bromodomain antagonist, RVX-208, was reported to raise apoA-I and increase preß-HDL particles in non-human primates and humans. In the present study, we investigated the effect of RVX-208 on aortic lesion formation in hyperlipidemic apoE(-/-) mice. Oral treatments of apoE(-/-) mice with 150 mg/kg b.i.d RVX-208 for 12 weeks significantly reduced aortic lesion formation, accompanied by 2-fold increases in the levels of circulating HDL-C, and ∼50% decreases in LDL-C, although no significant changes in plasma apoA-I were observed. Circulating adhesion molecules as well as cytokines also showed significant reduction. Haptoglobin, a proinflammatory protein, known to bind with HDL/apoA-I, decreased >2.5-fold in the RVX-208 treated group. With a therapeutic dosing regimen in which mice were fed Western diet for 10 weeks to develop lesions followed by switching to a low fat diet and concurrent treatment with RVX-208 for 14 weeks, RVX-208 similarly reduced lesion formation by 39% in the whole aorta without significant changes in the plasma lipid parameters. RVX-208 significantly reduced the proinflammatory cytokines IP-10, MIP1(®) and MDC. These results show that the antiatherogenic activity of BET inhibitor, RVX-208, occurs via a combination of lipid changes and anti-inflammatory activities.


Subject(s)
Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Hyperlipidemias/drug therapy , Quinazolines/therapeutic use , Animals , Aorta/drug effects , Aorta/metabolism , Aorta/pathology , Aortic Diseases/blood , Aortic Diseases/etiology , Aortic Diseases/pathology , Apolipoprotein A-I/blood , Apolipoproteins E/deficiency , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/pathology , Cell Line , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Cytokines/blood , Diet, Fat-Restricted , Diet, Western/adverse effects , Drug Evaluation, Preclinical , Endothelial Cells , Gene Expression Profiling , Humans , Hyperlipidemias/blood , Hyperlipidemias/complications , Hyperlipidemias/diet therapy , Hyperlipidemias/genetics , Inflammation/blood , Inflammation/prevention & control , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinazolines/pharmacology , Quinazolinones , RNA, Messenger/analysis , U937 Cells
5.
Nutrients ; 5(9): 3481-95, 2013 Sep 09.
Article in English | MEDLINE | ID: mdl-24022817

ABSTRACT

Methyl groups are important for numerous cellular functions such as DNA methylation, phosphatidylcholine synthesis, and protein synthesis. The methyl group can directly be delivered by dietary methyl donors, including methionine, folate, betaine, and choline. The liver and the muscles appear to be the major organs for methyl group metabolism. Choline can be synthesized from phosphatidylcholine via the cytidine-diphosphate (CDP) pathway. Low dietary choline loweres methionine formation and causes a marked increase in S-adenosylmethionine utilization in the liver. The link between choline, betaine, and energy metabolism in humans indicates novel functions for these nutrients. This function appears to goes beyond the role of the nutrients in gene methylation and epigenetic control. Studies that simulated methyl-deficient diets reported disturbances in energy metabolism and protein synthesis in the liver, fatty liver, or muscle disorders. Changes in plasma concentrations of total homocysteine (tHcy) reflect one aspect of the metabolic consequences of methyl group deficiency or nutrient supplementations. Folic acid supplementation spares betaine as a methyl donor. Betaine is a significant determinant of plasma tHcy, particularly in case of folate deficiency, methionine load, or alcohol consumption. Betaine supplementation has a lowering effect on post-methionine load tHcy. Hypomethylation and tHcy elevation can be attenuated when choline or betaine is available.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/metabolism , Metabolic Networks and Pathways , S-Adenosylmethionine/deficiency , Animals , Betaine/administration & dosage , Betaine/blood , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Choline/administration & dosage , Choline/blood , DNA Methylation , Dietary Supplements , Disease Models, Animal , Fasting , Fatty Liver/etiology , Fatty Liver/pathology , Folic Acid/administration & dosage , Folic Acid/blood , Homocysteine/blood , Humans , Liver/drug effects , Liver/metabolism , Methionine/metabolism , Muscular Diseases/etiology , Muscular Diseases/pathology , S-Adenosylmethionine/metabolism
6.
Eur J Med Chem ; 65: 256-75, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23727536

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) is an important zinc-dependent methyltransferase that uses betaine as the methyl donor for the remethylation of homocysteine to form methionine. In the liver, BHMT performs to half of the homocysteine remethylation. In this study, we systematically investigated the tolerance of the enzyme for modifications at the "homocysteine" part of the previously reported potent inhibitor (R,S)-5-(3-amino-3-carboxy-propylsulfanyl)-pentanoic acid (1). In the new compounds, which are S-alkylated homocysteine derivatives, we replaced the carboxylic group in the "homocysteine" part of inhibitor 1 with different isosteric moieties (tetrazole and oxadiazolone); we suppressed the carboxylic negative charge by amidations; we enhanced acidity by replacing the carboxylate with phosphonic or phosphinic acids; and we introduced pyrrolidine steric constraints. Some of these compounds display high affinity toward human BHMT and may be useful for further pharmacological studies of this enzyme. Although none of the new compounds were more potent inhibitors than the reference inhibitor 1, this study helped to completely define the structural requirements of the active site of BHMT and revealed the remarkable selectivity of the enzyme for homocysteine.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Pentanoic Acids/pharmacology , Sulfides/pharmacology , Betaine-Homocysteine S-Methyltransferase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pentanoic Acids/chemical synthesis , Pentanoic Acids/chemistry , Structure-Activity Relationship , Sulfides/chemical synthesis , Sulfides/chemistry
7.
Biochim Biophys Acta ; 1834(8): 1596-606, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23689031

ABSTRACT

Both cardiovascular disease and liver injury are major public health issues. Hyperhomocysteinemia has been linked to cardiovascular diseases, and defects in methyl group metabolism, often resulting in hyperhomocysteinemia, are among the key molecular events postulated to play a role in liver injury. We employed proteomics and metabolomics analyses of human hepatocytes in primary cell culture to explore the spectrum of proteins and associated metabolites affected by the disruption of methyl group metabolism. We treated the hepatocytes with homocysteine (Hcy, 0.1mM and 2mM) to follow the impact of hyperhomocysteinemia, and in parallel, we used a specific inhibitor of betaine-homocysteine S-methyltransferase (BHMT) to extend our understanding of the physiological functions of the enzyme. The major effect of BHMT inhibition was a 50% decrease in S-adenosylmethionine levels. The treatments with Hcy resulted in multiple changes in the metabolite levels depending on the treatment modality. The BHMT inhibition and 0.1mM Hcy treatment induced only moderate changes in the hepatocyte proteome and secretome, while the changes induced by the 2mM Hcy treatment were extensive. Phosphatidylethanolamine carboxykinase and ornithine aminotransferase were up-regulated about two fold indicating an intervention into metabolism. Cellular proliferation was suspended, secretome composition was changed and signs of apoptosis were discernible. We have detected fibrinogen gamma dimers, which might have a role as a potentially new biomarker of early liver injury. Finally, we have demonstrated the failed maturation of apolipoprotein A1, which might be a new mechanism of disruption of cholesterol efflux from tissues.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Colorectal Neoplasms/pathology , Hepatocytes/metabolism , Hyperhomocysteinemia/metabolism , Liver Neoplasms/pathology , Metabolomics , Proteome/analysis , Apolipoprotein A-I/metabolism , Apoptosis , Betaine-Homocysteine S-Methyltransferase/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Electrophoresis, Gel, Two-Dimensional , Fibrinogen/metabolism , Hepatocytes/drug effects , Homocysteine/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Male , Middle Aged , Protein Multimerization , Proteome/metabolism , S-Adenosylmethionine/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
8.
J Med Chem ; 55(15): 6822-31, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22775318

ABSTRACT

Betaine-homocysteine S-methyltransferase 2 (BHMT-2) catalyzes the transfer of a methyl group from S-methylmethionine to l-homocysteine, yielding two molecules of l-methionine. It is one of three homocysteine methyltransferases in mammals, but its overall contribution to homocysteine remethylation and sulfur amino acid homeostasis is not known. Moreover, recombinant BHMT-2 is highly unstable, which has slowed research on its structural and catalytic properties. In this study, we have prepared the first series of BHMT-2 inhibitors to be described, and we have tested them with human recombinant BHMT-2 that has been stabilized by copurification with human recombinant BHMT. Among the compounds synthesized, (2S,8RS,11RS)-5-thia-2,11-diamino-8-methyldodecanedioic acid (11) was the most potent (K(i)(app) ∼77 nM) and selective inhibitor of BHMT-2. Compound 11 only weakly inhibited human BHMT (IC(50) about 77 µM). This compound (11) may be useful in future in vivo studies to probe the physiological significance of BHMT-2 in sulfur amino acid metabolism.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Homocysteine/analogs & derivatives , Sulfides/chemical synthesis , Betaine-Homocysteine S-Methyltransferase/chemistry , Enzyme Assays , Homocysteine/chemical synthesis , Homocysteine/chemistry , Humans , Kinetics , Recombinant Proteins/antagonists & inhibitors , Stereoisomerism , Structure-Activity Relationship , Sulfides/chemistry
9.
J Nutr Sci Vitaminol (Tokyo) ; 58(2): 69-77, 2012.
Article in English | MEDLINE | ID: mdl-22790564

ABSTRACT

The effect of betaine status on folate deficiency-induced hyperhomocysteinemia was investigated to determine whether folate deficiency impairs homocysteine removal not only by the methionine synthase (MS) pathway but also by the betaine-homocysteine S-methyltransferase (BHMT) pathway. For this purpose, we investigated the effect of dietary supplementation with betaine at a high level (1%) in rats fed a folate-deprived 10% casein diet (10C) and 20% casein diet (20C). We also investigated the effect of choline deprivation on folate deficiency-induced hyperhomocysteinemia in rats fed 20C. Supplementation of folate-deprived 10C and 20C with 1% betaine significantly suppressed folate deprivation-induced hyperhomocysteinemia, but the extent of suppression was partial or limited, especially in rats fed 10C, the suppression of plasma homocysteine increment being 48.5% in rats fed 10C and 69.7% in rats fed 20C. Although betaine supplementation greatly increased hepatic betaine concentration and BHMT activity, these increases did not fully explain why the effect of betaine supplementation was partial or limited. Folate deprivation markedly increased the hepatic concentration of N,N-dimethylglycine (DMG), a known inhibitor of BHMT, and there was a significant positive correlation between hepatic DMG concentration and plasma homocysteine concentration, suggesting that folate deficiency increases hepatic DMG concentration and thereby depresses BHMT reaction, leading to interference with the effect of betaine supplementation. Choline deprivation did not increase plasma homocysteine concentration in rats fed 20C, but it markedly enhanced plasma homocysteine concentration when rats were fed folate-deprived 20C. This indicates that choline deprivation reinforced folate deprivation-induced hyperhomocysteinemia. Increased hepatic DMG concentration was also associated with such an effect. These results support the concept that folate deficiency impairs homocysteine metabolism not only by the MS pathway but also by the BHMT pathway.


Subject(s)
Betaine/administration & dosage , Choline Deficiency/blood , Folic Acid Deficiency/complications , Hyperhomocysteinemia/drug therapy , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Animals , Betaine/analysis , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/metabolism , Dietary Supplements , Enzyme Inhibitors/analysis , Folic Acid/administration & dosage , Folic Acid Deficiency/blood , Homocysteine/blood , Hyperhomocysteinemia/blood , Liver/chemistry , Liver/enzymology , Male , Rats , Rats, Wistar , Sarcosine/analogs & derivatives , Sarcosine/analysis
10.
Nutr Res ; 31(7): 563-71, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21840473

ABSTRACT

Methylation of homocysteine (Hcy) by betaine-Hcy S-methyltransferase (BHMT) produces methionine, which is required for S-adenosylmethionine (SAM) synthesis. We have recently shown that short-term dietary intake of S-(Δ-carboxybutyl)-dl-Hcy (D,L-CBHcy), a potent and specific inhibitor of BHMT, significantly decreases liver BHMT activity and SAM concentrations but does not have an adverse affect on liver histopathology, plasma markers of liver damage, or DNA methylation in rats. The present study was designed to investigate the hypothesis that BHMT is required to maintain normal liver and plasma amino acid and glutathione profiles, and liver SAM and lipid accumulation. Rats were fed an adequate (4.5 g/kg methionine and 3.7 g/kg cystine), cysteine-devoid (4.5 g/kg methionine and 0 g/kg cystine), or methionine-deficient (1.5 g/kg methionine and 3.7 g/kg cystine) diet either with or without L-CBHcy for 3 or 14 days. All rats fed L-CBHcy had increased total plasma Hcy (2- to 5-fold) and reduced liver BHMT activity (>90%) and SAM concentrations (>40%). S-(Δ-carboxybutyl)-l-Hcy treatment slightly reduced liver glutathione levels in rats fed the adequate or cysteine-devoid diet for 14 days. Rats fed the methionine-deficient diet with L-CBHcy developed fatty liver. Liver cystathionine ß-synthase activity was reduced in all L-CBHcy-treated animals, and the effect was exacerbated as time on the L-CBHcy diet increased. Our data indicate that BHMT activity is required to maintain adequate levels of liver SAM and low levels of total plasma Hcy and might be critical for liver glutathione and triglyceride homeostasis under some dietary conditions.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Cystathionine beta-Synthase/metabolism , Diet , Enzyme Inhibitors/pharmacology , Homocysteine/blood , Hyperhomocysteinemia/etiology , Liver/metabolism , S-Adenosylmethionine/metabolism , Animals , Cysteine/administration & dosage , Fatty Liver/etiology , Fatty Liver/metabolism , Glutathione/metabolism , Homeostasis , Homocysteine/analogs & derivatives , Homocysteine/pharmacology , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/metabolism , Male , Methionine/administration & dosage , Methionine/metabolism , Methylation , Rats , Rats, Inbred Strains
11.
Nutr Res ; 30(7): 492-500, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20797482

ABSTRACT

Betaine homocysteine S-methyltransferase (BHMT) catalyzes the transfer of a methyl group from betaine to homocysteine (Hcy), forming dimethylglycine and methionine. We previously showed that inhibiting BHMT in mice by intraperitoneal injection of S-(alpha-carboxybutyl)-DL-homocysteine (CBHcy) results in hyperhomocysteinemia. In the present study, CBHcy was fed to rats to determine whether it could be absorbed and cause hyperhomocysteinemia as observed in the intraperitoneal administration of the compound in mice. We hypothesized that dietary administered CBHcy will be absorbed and will result in the inhibition of BHMT and cause hyperhomocysteinemia. Rats were meal-fed every 8 hours an L-amino acid-defined diet either containing or devoid of CBHcy (5 mg per meal) for 3 days. The treatment decreased liver BHMT activity by 90% and had no effect on methionine synthase, methylenetetrahydrofolate reductase, phosphatidylethanolamine N-methyltransferase, and CTP:phosphocholine cytidylyltransferase activities. In contrast, cystathionine beta-synthase activity and immunodetectable protein decreased (56% and 26%, respectively) and glycine N-methyltransferase activity increased (52%) in CBHcy-treated rats. Liver S-adenosylmethionine levels decreased by 25% in CBHcy-treated rats, and S-adenosylhomocysteine levels did not change. Furthermore, plasma choline decreased (22%) and plasma betaine increased (15-fold) in CBHcy-treated rats. The treatment had no effect on global DNA and CpG island methylation, liver histology, and plasma markers of liver damage. We conclude that CBHcy-mediated BHMT inhibition causes an elevation in total plasma Hcy that is not normalized by the folate-dependent conversion of Hcy to methionine. Furthermore, metabolic changes caused by BHMT inhibition affect cystathionine beta-synthase and glycine N-methyltransferase activities, which further deteriorate plasma Hcy levels.


Subject(s)
Diet , Homocysteine/analogs & derivatives , Hyperhomocysteinemia/chemically induced , Amino Acids/analysis , Amino Acids/blood , Animals , Betaine/blood , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Choline/blood , Cystathionine beta-Synthase/analysis , Cystathionine beta-Synthase/metabolism , Enzyme Inhibitors/administration & dosage , Glycine N-Methyltransferase/metabolism , Homocysteine/administration & dosage , Homocysteine/pharmacokinetics , Hyperhomocysteinemia/pathology , Liver/chemistry , Liver/enzymology , Liver/pathology , Male , Rats , Rats, Inbred F344 , S-Adenosylhomocysteine/analysis , S-Adenosylmethionine/analysis
12.
J Med Chem ; 52(12): 3652-65, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19534555

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) catalyzes the transfer of a methyl group from betaine to l-homocysteine, yielding dimethylglycine and l-methionine. In this study, we prepared a new series of BHMT inhibitors. The inhibitors were designed to mimic the hypothetical transition state of BHMT substrates and consisted of analogues with NH, N(CH(3)), or N(CH(3))(2) groups separated from the homocysteine sulfur atom by a methylene, ethylene, or a propylene spacer. Only the inhibitor with the N(CH(3)) moiety and ethylene spacer gave moderate inhibition. This result led us to prepare two inhibitors lacking a nitrogen atom in the S-linked alkyl chain: (RS,RS)-5-(3-amino-3-carboxypropylthio)-3-methylpentanoic acid and (RS)-5-(3-amino-3-carboxypropylthio)-3,3-dimethylpentanoic acid. Both of these compounds were highly potent inhibitors of BHMT. The finding that BHMT does not tolerate a true betaine mimic within these inhibitors, especially the nitrogen atom, is surprising and evokes questions about putative conformational changes of BHMT upon the binding of the substrates/products and inhibitors.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Homocysteine/analogs & derivatives , Pentanoic Acids/pharmacology , Drug Design , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Homocysteine/chemical synthesis , Homocysteine/chemistry , Homocysteine/pharmacology , Humans , Molecular Structure , Pentanoic Acids/chemical synthesis , Pentanoic Acids/chemistry , Stereoisomerism , Structure-Activity Relationship
13.
Arterioscler Thromb Vasc Biol ; 28(9): 1596-605, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18556571

ABSTRACT

Hyperhomocysteinemia is an established risk factor for arterial as well as venous thromboembolism. Individuals with severe hyperhomocysteinemia caused by inherited genetic defects in homocysteine metabolism have an extremely high incidence of vascular thrombosis unless they are treated aggressively with homocysteine-lowering therapy. The clinical value of homocysteine-lowering therapy in individuals with moderate hyperhomocysteinemia, which is very common in populations at risk for vascular disease, is more controversial. Considerable progress in our understanding of the molecular mechanisms underlying the association between hyperhomocysteinemia and vascular thrombotic events has been provided by the development of a variety of murine models. Because levels of homocysteine are regulated by both the methionine and folate cycles, hyperhomocysteinemia can be induced in mice through both genetic and dietary manipulations. Mice deficient in the cystathionine beta-synthase (CBS) gene have been exploited widely in many studies investigating the vascular pathophysiology of hyperhomocysteinemia. In this article, we review the established murine models, including the CBS-deficient mouse as well as several newer murine models available for the study of hyperhomocysteinemia. We also summarize the major vascular phenotypes observed in these murine models.


Subject(s)
Endothelium, Vascular/physiopathology , Homocysteine/metabolism , Hyperhomocysteinemia/etiology , Thrombosis/etiology , Animals , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/metabolism , Diet/adverse effects , Disease Models, Animal , Endothelium, Vascular/metabolism , Enzyme Inhibitors/adverse effects , Genetic Predisposition to Disease , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/physiopathology , Mice , Phenotype , Thrombosis/metabolism , Thrombosis/physiopathology
14.
J Biol Chem ; 283(14): 8939-45, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18230605

ABSTRACT

We demonstrate that purified recombinant human betainehomocysteine methyltransferase-2 (BHMT-2) is a zinc metalloenzyme that uses S-methylmethionine (SMM) as a methyl donor for the methylation of homocysteine. Unlike the highly homologous betaine-homocysteine methyltransferase (BHMT), BHMT-2 cannot use betaine. The K(m) of BHMT-2 for SMM was determined to be 0.94 mm, and it has a turnover number similar to BHMT. Several compounds were tested as inhibitors of recombinant human BHMT and BHMT-2. The SMM-specific methyltransferase activity of BHMT-2 is not inhibited by dimethylglycine and betaine, whereas the former is a potent inhibitor of BHMT. Methionine is a stronger inhibitor of BHMT-2 than BHMT, and S-adenosylmethionine does not inhibit BHMT but is a weak inhibitor of BHMT-2. BHMT can use SMM as a methyl donor with a k(cat)/K(m) that is 5-fold lower than the k(cat)/K(m) for betaine. However, SMM does not inhibit BHMT activity when it is presented to the enzyme at concentrations that are 10-fold greater than the subsaturating amounts of betaine used in the assay. Based on these data, it is our current hypothesis that in vivo most if not all of the SMM-dependent methylation of homocysteine occurs via BHMT-2.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/chemistry , Homocysteine S-Methyltransferase/chemistry , Metalloproteins/chemistry , Zinc/chemistry , Betaine/chemistry , Betaine/metabolism , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/genetics , Betaine-Homocysteine S-Methyltransferase/metabolism , Homocysteine/chemistry , Homocysteine/metabolism , Homocysteine S-Methyltransferase/antagonists & inhibitors , Homocysteine S-Methyltransferase/genetics , Homocysteine S-Methyltransferase/metabolism , Humans , Metalloproteins/antagonists & inhibitors , Metalloproteins/genetics , Metalloproteins/metabolism , Methylation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Sarcosine/chemistry , Sarcosine/metabolism , Substrate Specificity/physiology , Vitamin U/chemistry , Vitamin U/metabolism , Zinc/metabolism
15.
Biochem J ; 401(1): 87-96, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16953798

ABSTRACT

BHMT (betaine-homocysteine methyltransferase) remethylates homocysteine to form methionine. SAM (S-adenosylmethionine) inhibits BHMT activity, but whether SAM modulates BHMT gene expression is unknown. Transcriptional regulation of the human BHMT is also unknown. The present study examined regulation of the human BHMT gene by SAM and its metabolite, MTA (5'-methylthioadenosine). To facilitate these studies, we cloned the 2.7 kb 5'-flanking region of the human BHMT gene (GenBank accession number AY325901). Both SAM and MTA treatment of HepG2 cells resulted in a dose- and time-dependent decrease in BHMT mRNA levels, which paralleled their effects on the BHMT promoter activity. Maximal suppression was observed with the BHMT promoter construct -347/+33, which contains a number of NF-kappaB (nuclear factor kappaB) binding sites. SAM and MTA treatment increased NF-kappaB nuclear binding and NF-kappaB-driven luciferase activities, and increased nuclear binding activity of multiple histone deacetylase co-repressors to the NF-kappaB sites. Overexpression of p50 and p65 decreased BHMT promoter activity, while blocking NF-kappaB activation increased BHMT expression and promoter activity, and prevented SAM but not MTA's ability to inhibit BHMT expression. The NF-kappaB binding site at -301 is responsible, at least in part, for this effect. Lower BHMT expression can impair homocysteine metabolism, which can induce ER (endoplasmic reticulum) stress. Indeed, MTA treatment resulted in increased expression ER stress markers. In conclusion, SAM and MTA down-regulate BHMT expression in HepG2 cells in part by inducing NF-kappaB, which acts as a repressor for the human BHMT gene. While SAM's mechanism is NF-kappaB-dependent, MTA has both NF-kappaB-dependent and -independent mechanisms.


Subject(s)
Adenosine/analogs & derivatives , Betaine-Homocysteine S-Methyltransferase/genetics , Gene Expression Regulation, Enzymologic/drug effects , S-Adenosylmethionine/pharmacology , Thionucleosides/pharmacology , Adenosine/pharmacology , Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Cell Line , Cell Line, Tumor , Cloning, Molecular , DNA Primers , Genetic Vectors , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , NF-kappa B/pharmacology , Plasmids , Recombinant Proteins/antagonists & inhibitors , Transcription, Genetic
16.
J Med Chem ; 49(13): 3982-9, 2006 Jun 29.
Article in English | MEDLINE | ID: mdl-16789755

ABSTRACT

A series of S-alkylated derivatives of homocysteine were synthesized and characterized as inhibitors of human recombinant betaine-homocysteine S-methyltransferase (BHMT). Some of these compounds inhibit BHMT with IC50 values in the nanomolar range. BHMT is very sensitive to the structure of substituents on the sulfur atom of homocysteine. The S-carboxybutyl and S-carboxypentyl derivatives make the most potent inhibitors, and an additional sulfur atom in the alkyl chain is well tolerated. The respective (R,S)-5-(3-amino-3-carboxy-propylsulfanyl)-pentanoic, (R,S)-6-(3-amino-3-carboxy-propylsulfanyl)-hexanoic, and (R,S)-2-amino-4-(2-carboxymethylsulfanyl-ethylsulfanyl)-butyric acids are very potent inhibitors and are the strongest ever reported. We determined that (R,S)-5-(3-amino-3-carboxy-propylsulfanyl)-pentanoic acid displays competitive inhibition with respect to betaine binding with a Kappi of 12 nM. Some of these compounds are currently being tested in mice to study the influence of BHMT on the metabolism of sulfur amino acids in vivo.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Butyrates/chemical synthesis , Caproates/chemical synthesis , Homocysteine/analogs & derivatives , Homocysteine/chemical synthesis , Pentanoic Acids/chemical synthesis , Sulfides/chemical synthesis , Betaine-Homocysteine S-Methyltransferase/chemistry , Butyrates/chemistry , Caproates/chemistry , Homocysteine/chemistry , Humans , Pentanoic Acids/chemistry , Stereoisomerism , Structure-Activity Relationship , Sulfides/chemistry
17.
J Nutr ; 136(6): 1493-7, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16702310

ABSTRACT

Inhibitors and methyl donor substrates for betaine-homocysteine S-methyltransferase (BHMT) were used to study the role of this enzyme in the regulation of plasma total homocysteine (tHcy). Mice were administered an i.p. injection of S-(delta-carboxybutyl)-dl-homocysteine (CBHcy; 1 mg), a specific and potent inhibitor of BHMT, and tHcy and hepatic BHMT protein and activity levels were monitored over a 24-h period. Compared with saline-injected control mice, at 2 h postinjection, the CBHcy-treated mice had 87% lower BHMT activity and a 2.7-fold increase (11.1 vs. 3.0 micromol/L) in tHcy, effects that lasted nearly 8 h but returned to normal by 24 h. The level of BHMT protein remained constant over the 24-h period. After 6 CBHcy (1 mg) injections (one every 12 h), the mice had 7-fold higher tHcy, a 65% reduction in the liver S-adenosylmethionine:S-adenosylhomocysteine ratio, and a marked upregulation of BHMT protein expression. At 2 h after injection of the sulfoxide derivative of CBHcy (10 mg) into mice, there was a modest reduction in BHMT activity and a 90% increase in tHcy. When given an injection of Met (3 mg) or Met plus CBHcy (1 mg), post-Met load tHcy levels were 2.2-fold higher (128 vs. 40 micromol/L) at 2 h postinjection in the mice given CBHcy. Like betaine, dimethylsulfoniopropionate was an effective tHcy-lowering agent when given with a Met load. These studies are the first to show that transient inhibition of BHMT in vivo causes transient hyperhomocysteinemia, and that dimethylsulfoniopropionate can reduce a post-Met load rise in tHcy.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Hyperhomocysteinemia/etiology , Sterol Regulatory Element Binding Proteins/pharmacology , Animals , Betaine-Homocysteine S-Methyltransferase/metabolism , Liver/drug effects , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred BALB C
18.
Biochem J ; 392(Pt 3): 443-8, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16038618

ABSTRACT

Betaine-homocysteine S-methyltransferase (BHMT) transfers a methyl group from betaine to Hcy to form DMG (dimethylglycine) and Met. The reaction is ordered Bi Bi; Hcy is the first substrate to bind and Met is the last product off. Using intrinsic tryptophan fluorescence [Castro, Gratson, Evans, Jiracek, Collinsova, Ludwig and Garrow (2004) Biochemistry 43, 5341-5351], it was shown that BHMT exists in three steady-state conformations: enzyme alone, enzyme plus occupancy at the first substrate-binding site (Hcy or Met), or enzyme plus occupancy at both substrate-binding sites (Hcy plus betaine, or Hcy plus DMG). Betaine or DMG alone do not bind to the enzyme, indicating that the conformational change associated with Hcy binding creates the betaine-binding site. CBHcy [S-(d-carboxybutyl)-D,L-homocysteine] is a bisubstrate analogue that causes BHMT to adopt the same conformation as the ternary complexes. We report that BHMT is susceptible to conformation-dependent oxidative inactivation. Two oxidants, MMTS (methyl methanethiosulphonate) and hydrogen peroxide (H2O2), cause a loss of the enzyme's catalytic Zn (Zn2+ ion) and a correlative loss of activity. Addition of 2-mercaptoethanol and exogenous Zn after MMTS treatment restores activity, but oxidation due to H2O2 is irreversible. CD and glutaraldehyde cross-linking indicate that H2O2 treatment causes small perturbations in secondary structure but no change in quaternary structure. Oxidation is attenuated when both binding sites are occupied by CBHcy, but Met alone has no effect. Partial digestion of ligand-free BHMT with trypsin produces two large peptides, excising a seven-residue peptide within loop L2. CBHcy but not Met binding slows down proteolysis by trypsin. These findings suggest that L2 is involved in the conformational change associated with occupancy at the betaine-binding site and that this conformational change and/or occupancy at both ligand-binding sites protect the enzyme from oxidative inactivation.


Subject(s)
Betaine-Homocysteine S-Methyltransferase/antagonists & inhibitors , Betaine-Homocysteine S-Methyltransferase/chemistry , Hydrogen Peroxide/pharmacology , Betaine-Homocysteine S-Methyltransferase/metabolism , Binding Sites , Circular Dichroism , Enzyme Activation/drug effects , Glutaral/metabolism , Homocysteine/analogs & derivatives , Homocysteine/metabolism , Humans , Mercaptoethanol/chemistry , Methyl Methanesulfonate/analogs & derivatives , Methyl Methanesulfonate/metabolism , Models, Molecular , Oxidation-Reduction , Protein Binding , Protein Conformation/drug effects , Zinc/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...