Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
2.
Chemistry ; 26(45): 10336-10347, 2020 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-32368816

RESUMEN

In this work, the change of reactivity induced by the introduction of two para-ethynyl substituents (CCSi(iPr)3 or CCH) to the organic electron-donor 1,2,4,5-tetrakis(tetramethylguanidino)-benzene is evaluated. The redox-properties and redox-state dependent fluorescence are evaluated, and dinuclear CuI and CuII complexes synthesized. The Lewis-acidic B(C6 F5 )3 substitutes the proton of the ethynyl -CCH groups to give new anionic -CCB(C6 F5 )3 - substituents, leading eventually to a novel dianionic strong electron donor in its diprotonated form. Its two-electron oxidation with dioxygen in the presence of a copper catalyst yields the first redox-active guanidine that is neutral (instead of cationic) in its oxidized form.

3.
Chemistry ; 25(15): 3781-3785, 2019 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-30688382

RESUMEN

Proton-coupled electron transfer (PCET) reactions are of great importance in synthetic chemistry and in biology, but the acquisition of kinetic information for these reactions is often difficult. Herein, we report the synthesis of a new PCET reagent, showing redox-state dependent fluorescence, by merging the concept of cross-conjugated cruciform chromophores with the strategy of imposing redox activity and Brønsted basicity to aromatic compounds by substitution with guanidino groups. The compound is isolated and characterized in all stable states-reduced, twofold and fourfold protonated and twofold oxidized-and then applied in PCET reactions by using its redox-state dependent fluorescence signal for kinetic measurements.

4.
Angew Chem Int Ed Engl ; 55(40): 12455-9, 2016 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-27603008

RESUMEN

Manganese-catalyzed C-H bond activation chemistry is emerging as a powerful and complementary method for molecular functionalization. A highly reactive seven-membered Mn(I) intermediate is detected and characterized that is effective for H-transfer or reductive elimination to deliver alkenylated or pyridinium products, respectively. The two pathways are determined at Mn(I) by judicious choice of an electron-deficient 2-pyrone substrate containing a 2-pyridyl directing group, which undergoes regioselective C-H bond activation, serving as a valuable system for probing the mechanistic features of Mn C-H bond activation chemistry.

5.
Med Hypotheses ; 88: 63-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26880641

RESUMEN

Formaldehyde is extremely toxic reacting with proteins to crosslinks peptide chains. Formaldehyde is a metabolic product in many enzymatic reactions and the question of how these enzymes are protected from the formaldehyde that is generated has largely remained unanswered. Early experiments from our laboratory showed that two liver mitochondrial enzymes, dimethylglycine dehydrogenase (DMGDH) and sarcosine dehydrogenase (SDH) catalyze oxidative demethylation reactions (sarcosine is a common name for monomethylglycine). The enzymatic products of these enzymes were the demethylated substrates and formaldehyde, produced from the removed methyl group. Both DMGDH and SDH contain FAD and both have tightly bound tetrahydrofolate (THF), a folate coenzyme. THF binds reversibly with formaldehyde to form 5,10-methylene-THF. At that time we showed that purified DMGDH, with tightly bound THF, reacted with formaldehyde generated during the reaction to form 5,10-methylene-THF. This effectively scavenged the formaldehyde to protect the enzyme. Recently, post-translational modifications on histone tails have been shown to be responsible for epigenetic regulation of gene expression. One of these modifications is methylation of lysine residues. The first enzyme discovered to accomplish demethylation of these modified histones was histone lysine demethylase (LSD1). LSD1 specifically removes methyl groups from di- and mono-methylated lysines at position 4 of histone 3. This enzyme contained tightly bound FAD and the products of the reaction were the demethylated lysine residue and formaldehyde. The mechanism of LSD1 demethylation is analogous to the mechanism previously postulated for DMGDH, i.e. oxidation of the N-methyl bond to the methylene imine followed by hydrolysis to generate formaldehyde. This suggested that THF might also be involved in the LSD1 reaction to scavenge the formaldehyde produced. Our hypotheses are that THF is bound to native LSD1 by analogy to DMGDH and SDH and that the bound THF serves to protect the FAD class of histone demethylases from the destructive effects of formaldehyde generation by formation of 5,10-methylene-THF. We present pilot data showing that decreased folate in livers as a result of dietary folate deficiency is associated with increased levels of methylated lysine 4 of histone 3. This can be a result of decreased LSD1 activity resulting from the decreased folate available to scavenge the formaldehyde produced at the active site caused by the folate deficiency. Because LSD1 can regulate gene expression this suggests that folate may play a more important role than simply serving as a carrier of one-carbon units and be a factor in other diseases associated with low folate.


Asunto(s)
Metilación de ADN , Deficiencia de Ácido Fólico/inmunología , Histona Demetilasas/química , Histonas/química , Dominio Catalítico , Epigénesis Genética , Escherichia coli/metabolismo , Ácido Fólico/química , Humanos , Lisina/química , Espectrometría de Masas , Modelos Teóricos , Proyectos Piloto , Procesamiento Proteico-Postraduccional , Sarcosina-Deshidrogenasa/química , Tetrahidrofolatos/química
6.
J Hepatol ; 64(2): 409-418, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26394163

RESUMEN

BACKGROUND & AIMS: Glycine N-methyltransferase (GNMT) expression is decreased in some patients with severe non-alcoholic fatty liver disease. Gnmt deficiency in mice (Gnmt-KO) results in abnormally elevated serum levels of methionine and its metabolite S-adenosylmethionine (SAMe), and this leads to rapid liver steatosis development. Autophagy plays a critical role in lipid catabolism (lipophagy), and defects in autophagy have been related to liver steatosis development. Since methionine and its metabolite SAMe are well known inactivators of autophagy, we aimed to examine whether high levels of both metabolites could block autophagy-mediated lipid catabolism. METHODS: We examined methionine levels in a cohort of 358 serum samples from steatotic patients. We used hepatocytes cultured with methionine and SAMe, and hepatocytes and livers from Gnmt-KO mice. RESULTS: We detected a significant increase in serum methionine levels in steatotic patients. We observed that autophagy and lipophagy were impaired in hepatocytes cultured with high methionine and SAMe, and that Gnmt-KO livers were characterized by an impairment in autophagy functionality, likely caused by defects at the lysosomal level. Elevated levels of methionine and SAMe activated PP2A by methylation, while blocking PP2A activity restored autophagy flux in Gnmt-KO hepatocytes, and in hepatocytes treated with SAMe and methionine. Finally, normalization of methionine and SAMe levels in Gnmt-KO mice using a methionine deficient diet normalized the methylation capacity, PP2A methylation, autophagy, and ameliorated liver steatosis. CONCLUSIONS: These data suggest that elevated levels of methionine and SAMe can inhibit autophagic catabolism of lipids contributing to liver steatosis.


Asunto(s)
Autofagia/fisiología , Hígado Graso/metabolismo , Hepatocitos/metabolismo , Metionina/sangre , Proteína Fosfatasa 2/metabolismo , S-Adenosilmetionina/sangre , Animales , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Hígado Graso/patología , Humanos , Metilación , Ratones
7.
Orphanet J Rare Dis ; 10: 99, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-26289392

RESUMEN

BACKGROUND: This paper summarizes the results of a group effort to bring together the worldwide available data on patients who are either homozygotes or compound heterozygotes for mutations in MAT1A. MAT1A encodes the subunit that forms two methionine adenosyltransferase isoenzymes, tetrameric MAT I and dimeric MAT III, that catalyze the conversion of methionine and ATP to S-adenosylmethionine (AdoMet). Subnormal MAT I/III activity leads to hypermethioninemia. Individuals, with hypermethioninemia due to one of the MAT1A mutations that in heterozygotes cause relatively mild and clinically benign hypermethioninemia are currently often being flagged in screening programs measuring methionine elevation to identify newborns with defective cystathionine ß-synthase activity. Homozygotes or compound heterozygotes for MAT1A mutations are less frequent. Some but not all, such individuals have manifested demyelination or other CNS abnormalities. PURPOSE OF THE STUDY: The goals of the present effort have been to determine the frequency of such abnormalities, to find how best to predict whether they will occur, and to evaluate the outcomes of the variety of treatment regimens that have been used. Data have been gathered for 64 patients, of whom 32 have some evidence of CNS abnormalities (based mainly on MRI findings), and 32 do not have such evidence. RESULTS AND DISCUSSION: The results show that mean plasma methionine concentrations provide the best indication of the group into which a given patient will fall: those with means of 800 µM or higher usually have evidence of CNS abnormalities, whereas those with lower means usually do not. Data are reported for individual patients for MAT1A genotypes, plasma methionine, total homocysteine (tHcy), and AdoMet concentrations, liver function studies, results of 15 pregnancies, and the outcomes of dietary methionine restriction and/or AdoMet supplementation. Possible pathophysiological mechanisms that might contribute to CNS damage are discussed, and tentative suggestions are put forth as to optimal management.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/genética , Heterocigoto , Homocigoto , Metionina Adenosiltransferasa/genética , Adolescente , Adulto , Preescolar , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Encuestas y Cuestionarios , Adulto Joven
8.
Mol Genet Metab ; 116(1-2): 44-52, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26095522

RESUMEN

A child with severe S-adenosylhomocysteine hydrolase (AHCY) deficiency (AHCY c.428A>G, p.Tyr143Cys; c.982T>G, p.Tyr328Asp) presented at 8 months of age with growth failure, microcephaly, global developmental delay, myopathy, hepatopathy, and factor VII deficiency. Plasma methionine, S-adenosylmethionine (AdoMet), and S-adenosylhomocysteine (AdoHcy) were markedly elevated and the molar concentration ratio of AdoMet:AdoHcy, believed to regulate a myriad of methyltransferase reactions, was 15% of the control mean. Dietary therapy failed to normalize biochemical markers or alter the AdoMet to AdoHcy molar concentration ratio. At 40 months of age, the proband received a liver segment from a healthy, unrelated living donor. Mean AdoHcy decreased 96% and the AdoMet:AdoHcy concentration ratio improved from 0.52±0.19 to 1.48±0.79 mol:mol (control 4.10±2.11 mol:mol). Blood methionine and AdoMet were normal and stable during 6 months of follow-up on an unrestricted diet. Average calculated tissue methyltransferase activity increased from 43±26% to 60±22%, accompanied by signs of increased transmethylation in vivo. Factor VII activity increased from 12% to 100%. During 6 postoperative months, head growth accelerated 4-fold and the patient made promising gains in gross motor, language, and social skills.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/cirugía , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Glicina N-Metiltransferasa/deficiencia , Trasplante de Hígado , Errores Innatos del Metabolismo de los Aminoácidos/complicaciones , Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Preescolar , Discapacidades del Desarrollo/etiología , Dietoterapia , Femenino , Cabeza/crecimiento & desarrollo , Cabeza/patología , Humanos , Metionina/sangre , Microcefalia/etiología , Enfermedades Musculares/etiología , Polimorfismo de Nucleótido Simple , S-Adenosilhomocisteína/sangre , S-Adenosilmetionina/sangre
9.
10.
Am J Cancer Res ; 5(1): 458-65, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25628954

RESUMEN

Methionine metabolism is an important component of one-carbon metabolism. S-adenosylmethionine (SAM), the methyl donor for nearly all methylation reactions, is irreversibly converted to S-adenosylhomocysteine (SAH), an inhibitor of methyltransferases, some of which are key enzymes for methylation. Changes in DNA methylation are common in colorectal cancers. We evaluated plasma SAM and SAH with colorectal adenoma risk in a matched case-control study conducted among individuals undergoing routine colonoscopy. 216 cases were individually matched to polyp-free controls in a 1:1 ratio on age (± 5 years), sex, race (white/non-white), study site (academic medical center/VA hospital) and date of sample collection (± 60 days). Sex-specific quantiles were evaluated based on the control distribution due to vastly different metabolite levels by sex. Conditional logistic regression models were used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). Among males, both higher SAM (OR = 0.38, 95% CI: 0.18-0.77, p for trend = 0.007) and higher SAH (OR = 0.45, 95% CI: 0.22-0.91, p for trend = 0.02) were associated with statistically significantly decreased risks of colorectal adenoma in comparison to lowest plasma SAM or SAH tertile. Conversely, among females, both higher SAM and higher SAH were associated with increased risk of colorectal adenoma, which was statistically significant for SAH (OR = 5.18, 95% CI: 1.09-24.62, p for trend = 0.04). The difference in these associations between men and women was statistically significant (p < 0.05). The ratio of SAM/SAH was not associated with colorectal adenoma risk among males or females. These findings suggest SAM and SAH may be involved in the development of colorectal adenoma and the association may be modified by sex.

11.
Lab Invest ; 95(2): 223-36, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25531568

RESUMEN

Glycine-N-methyltransferase (GNMT) is essential to preserve liver homeostasis. Cirrhotic patients show low expression of GNMT that is absent in hepatocellular carcinoma (HCC) samples. Accordingly, GNMT deficiency in mice leads to steatohepatitis, fibrosis, cirrhosis, and HCC. Lack of GNMT triggers NK cell activation in GNMT(-/-) mice and depletion of TRAIL significantly attenuates acute liver injury and inflammation in these animals. Chronic inflammation leads to fibrogenesis, further contributing to the progression of chronic liver injury regardless of the etiology. The aim of our study is to elucidate the implication of TRAIL-producing NK cells in the progression of chronic liver injury and fibrogenesis. For this we generated double TRAIL(-/-)/GNMT(-/-) mice in which we found that TRAIL deficiency efficiently protected the liver against chronic liver injury and fibrogenesis in the context of GNMT deficiency. Next, to better delineate the implication of TRAIL-producing NK cells during fibrogenesis we performed bile duct ligation (BDL) to GNMT(-/-) and TRAIL(-/-)/GNMT(-/-) mice. In GNMT(-/-) mice, exacerbated fibrogenic response after BDL concurred with NK1.1(+) cell activation. Importantly, specific inhibition of TRAIL-producing NK cells efficiently protected GNMT(-/-) mice from BDL-induced liver injury and fibrogenesis. Finally, TRAIL(-/-)/GNMT(-/-) mice showed significantly less fibrosis after BDL than GNMT(-/-) mice further underlining the relevance of the TRAIL/DR5 axis in mediating liver injury and fibrogenesis in GNMT(-/-) mice. Finally, in vivo silencing of DR5 efficiently protected GNMT(-/-) mice from BDL-liver injury and fibrogenesis, overall underscoring the key role of the TRAIL/DR5 axis in promoting fibrogenesis in the context of absence of GNMT. Overall, our work demonstrates that TRAIL-producing NK cells actively contribute to liver injury and further fibrogenesis in the pathological context of GNMT deficiency, a molecular scenario characteristic of chronic human liver disease.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/inmunología , Enfermedad Hepática en Estado Terminal/etiología , Enfermedad Hepática en Estado Terminal/patología , Glicina N-Metiltransferasa/deficiencia , Células Asesinas Naturales/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Conductos Biliares/cirugía , Western Blotting , Citometría de Flujo , Glicina N-Metiltransferasa/inmunología , Humanos , Inmunohistoquímica , Ligadura , Ratones , Ratones Noqueados , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
12.
J Hepatol ; 62(3): 673-81, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25457203

RESUMEN

BACKGROUND & AIMS: Very-low-density lipoproteins (VLDLs) export lipids from the liver to peripheral tissues and are the precursors of low-density-lipoproteins. Low levels of hepatic S-adenosylmethionine (SAMe) decrease triglyceride (TG) secretion in VLDLs, contributing to hepatosteatosis in methionine adenosyltransferase 1A knockout mice but nothing is known about the effect of SAMe on the circulating VLDL metabolism. We wanted to investigate whether excess SAMe could disrupt VLDL plasma metabolism and unravel the mechanisms involved. METHODS: Glycine N-methyltransferase (GNMT) knockout (KO) mice, GNMT and perilipin-2 (PLIN2) double KO (GNMT-PLIN2-KO) and their respective wild type (WT) controls were used. A high fat diet (HFD) or a methionine deficient diet (MDD) was administrated to exacerbate or recover VLDL metabolism, respectively. Finally, 33 patients with non-alcoholic fatty-liver disease (NAFLD); 11 with hypertriglyceridemia and 22 with normal lipidemia were used in this study. RESULTS: We found that excess SAMe increases the turnover of hepatic TG stores for secretion in VLDL in GNMT-KO mice, a model of NAFLD with high SAMe levels. The disrupted VLDL assembly resulted in the secretion of enlarged, phosphatidylethanolamine-poor, TG- and apoE-enriched VLDL-particles; special features that lead to increased VLDL clearance and decreased serum TG levels. Re-establishing normal SAMe levels restored VLDL secretion, features and metabolism. In NAFLD patients, serum TG levels were lower when hepatic GNMT-protein expression was decreased. CONCLUSIONS: Excess hepatic SAMe levels disrupt VLDL assembly and features and increase circulating VLDL clearance, which will cause increased VLDL-lipid supply to tissues and might contribute to the extrahepatic complications of NAFLD.


Asunto(s)
Lipoproteínas VLDL/sangre , Enfermedad del Hígado Graso no Alcohólico/metabolismo , S-Adenosilmetionina/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Glicina N-Metiltransferasa/deficiencia , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/metabolismo , Humanos , Lipoproteínas VLDL/metabolismo , Hígado/metabolismo , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Modelos Biológicos , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/patología , Perilipina-2 , S-Adenosilmetionina/deficiencia , Triglicéridos/metabolismo , Adulto Joven
13.
Biochem Biophys Res Commun ; 449(4): 392-8, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24858690

RESUMEN

Dimethylglycine dehydrogenase (DMGDH) is a mammalian mitochondrial enzyme which plays an important role in the utilization of methyl groups derived from choline. DMGDH is a flavin containing enzyme which catalyzes the oxidative demethylation of dimethylglycine in vitro with the formation of sarcosine (N-methylglycine), hydrogen peroxide and formaldehyde. DMGDH binds tetrahydrofolate (THF) in vivo, which serves as an acceptor of formaldehyde and in the cell the product of the reaction is 5,10-methylenetetrahydrofolate instead of formaldehyde. To gain insight into the mechanism of the reaction we solved the crystal structures of the recombinant mature and precursor forms of rat DMGDH and DMGDH-THF complexes. Both forms of DMGDH reveal similar kinetic parameters and have the same tertiary structure fold with two domains formed by N- and C-terminal halves of the protein. The active center is located in the N-terminal domain while the THF binding site is located in the C-terminal domain about 40Å from the isoalloxazine ring of FAD. The folate binding site is connected with the enzyme active center via an intramolecular channel. This suggests the possible transfer of the intermediate imine of dimethylglycine from the active center to the bound THF where they could react producing a 5,10-methylenetetrahydrofolate. Based on the homology of the rat and human DMGDH the structural basis for the mechanism of inactivation of the human DMGDH by naturally occurring His109Arg mutation is proposed.


Asunto(s)
Dimetilglicina-Deshidrogenasa/química , Proteínas Mitocondriales/química , Tetrahidrofolatos/química , Secuencia de Aminoácidos , Animales , Sitios de Unión , Dominio Catalítico , Cristalización , Cristalografía por Rayos X , Dimetilglicina-Deshidrogenasa/metabolismo , Humanos , Cinética , Proteínas Mitocondriales/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Ratas , Sarcosina/análogos & derivados , Tetrahidrofolatos/metabolismo
14.
Protein Sci ; 23(7): 993-8, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24715612

RESUMEN

An important epigenetic modification is the methylation/demethylation of histone lysine residues. The first histone demethylase to be discovered was a lysine-specific demethylase 1, LSD1, a flavin containing enzyme which carries out the demethylation of di- and monomethyllysine 4 in histone H3. The removed methyl groups are oxidized to formaldehyde. This reaction is similar to those performed by dimethylglycine dehydrogenase and sarcosine dehydrogenase, in which protein-bound tetrahydrofolate (THF) was proposed to serve as an acceptor of the generated formaldehyde. We showed earlier that LSD1 binds THF with high affinity which suggests its possible participation in the histone demethylation reaction. In the cell, LSD1 interacts with co-repressor for repressor element 1 silencing transcription factor (CoREST). In order to elucidate the role of folate in the demethylating reaction we solved the crystal structure of the LSD1-CoREST-THF complex. In the complex, the folate-binding site is located in the active center in close proximity to flavin adenine dinucleotide. This position of the folate suggests that the bound THF accepts the formaldehyde generated in the course of histone demethylation to form 5,10-methylene-THF. We also show the formation of 5,10-methylene-THF during the course of the enzymatic reaction in the presence of THF by mass spectrometry. Production of this form of folate could act to prevent accumulation of potentially toxic formaldehyde in the cell. These studies suggest that folate may play a role in the epigenetic control of gene expression in addition to its traditional role in the transfer of one-carbon units in metabolism.


Asunto(s)
Proteínas Co-Represoras/química , Histona Demetilasas/química , Lisina/metabolismo , Tetrahidrofolatos/química , Sitios de Unión , Proteínas Co-Represoras/metabolismo , Cristalografía por Rayos X , Flavina-Adenina Dinucleótido/química , Histona Demetilasas/metabolismo , Humanos , Espectrometría de Masas , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato , Tetrahidrofolatos/metabolismo
15.
Int J Cancer ; 135(9): 2191-8, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24692023

RESUMEN

Previous epidemiological studies of circulating folate concentration and colorectal cancer have reported inconsistent results. We evaluated associations of prediagnostic plasma folate concentration with colorectal cancer risk in a case-control study nested within the Shanghai Men's Health Study (2002-2010). Included herein are 288 cases who were diagnosed with incident colorectal cancer and 575 controls who were individually matched to cases on baseline characteristics. Folate concentrations in plasma were measured by microbiological assay. Multivariate conditional logistic regression was used to assess associations of plasma folate concentrations with colorectal cancer risk. Plasma folate was nonsignificantly but positively associated with colorectal cancer risk. Odds ratios (OR) and 95% confidence intervals (CI) were 1.38 (0.95-2.02) for the middle tertile of plasma folate concentrations and 1.33 (0.90-1.98) for the highest compared to the lowest tertile. The positive association reached statistical significance for the highest tertile of folate concentrations for men with late-stage colorectal cancer (OR = 2.66; 95% CI = 1.03-6.86) and for the middle tertile for cases diagnosed within the first 4 years after blood collection (OR = 1.72; 95% CI = 1.02-2.92) and for men in the high BMI group (OR = 1.88; 95% CI = 1.14-3.11). In our study population, where folic acid fortification of the food supply and vitamin supplement use are uncommon, plasma folate concentration was positively associated with colorectal cancer risk among men who may have had preneoplastic lesions. These findings need to be confirmed in studies with specific assessment of preneoplastic lesions and repeated measurements of folate level over time.


Asunto(s)
Neoplasias Colorrectales/sangre , Neoplasias Colorrectales/etiología , Ácido Fólico/sangre , Adulto , Anciano , Estudios de Casos y Controles , China , Neoplasias Colorrectales/patología , Estudios de Seguimiento , Humanos , Masculino , Salud del Hombre , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Estudios Prospectivos , Factores de Riesgo
16.
Hippocampus ; 24(7): 840-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24687756

RESUMEN

The hippocampus is a brain area characterized by its high plasticity, observed at all levels of organization: molecular, synaptic, and cellular, the latter referring to the capacity of neural precursors within the hippocampus to give rise to new neurons throughout life. Recent findings suggest that promoter methylation is a plastic process subjected to regulation, and this plasticity seems to be particularly important for hippocampal neurogenesis. We have detected the enzyme GNMT (a liver metabolic enzyme) in the hippocampus. GNMT regulates intracellular levels of SAMe, which is a universal methyl donor implied in almost all methylation reactions and, thus, of prime importance for DNA methylation. In addition, we show that deficiency of this enzyme in mice (Gnmt-/-) results in high SAMe levels within the hippocampus, reduced neurogenic capacity, and spatial learning and memory impairment. In vitro, SAMe inhibited neural precursor cell division in a concentration-dependent manner, but only when proliferation signals were triggered by bFGF. Indeed, SAMe inhibited the bFGF-stimulated MAP kinase signaling cascade, resulting in decreased cyclin E expression. These results suggest that alterations in the concentration of SAMe impair neurogenesis and contribute to cognitive decline.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/psicología , Cognición/fisiología , Glicina N-Metiltransferasa/deficiencia , Hipocampo/enzimología , Proteínas del Tejido Nervioso/fisiología , Neurogénesis/fisiología , S-Adenosilmetionina/fisiología , Animales , Ciclina E/biosíntesis , Ciclina E/genética , Factor 2 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica , Glicina N-Metiltransferasa/genética , Glicina N-Metiltransferasa/fisiología , Hipocampo/fisiopatología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/etiología , Metionina/metabolismo , Metionina Adenosiltransferasa/deficiencia , Metionina Adenosiltransferasa/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/deficiencia , Proteínas del Tejido Nervioso/genética , Plasticidad Neuronal , Prueba de Desempeño de Rotación con Aceleración Constante , S-Adenosilmetionina/biosíntesis
17.
Neuron ; 81(5): 1024-1039, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24607226

RESUMEN

Axonal myelination is essential for rapid saltatory impulse conduction in the nervous system, and malformation or destruction of myelin sheaths leads to motor and sensory disabilities. DNA methylation is an essential epigenetic modification during mammalian development, yet its role in myelination remains obscure. Here, using high-resolution methylome maps, we show that DNA methylation could play a key gene regulatory role in peripheral nerve myelination and that S-adenosylmethionine (SAMe), the principal methyl donor in cytosine methylation, regulates the methylome dynamics during this process. Our studies also point to a possible role of SAMe in establishing the aberrant DNA methylation patterns in a mouse model of diabetic neuropathy, implicating SAMe in the pathogenesis of this disease. These critical observations establish a link between SAMe and DNA methylation status in a defined biological system, providing a mechanism that could direct methylation changes during cellular differentiation and in diverse pathological situations.


Asunto(s)
Metilación de ADN/genética , Vaina de Mielina/metabolismo , Nervios Periféricos/metabolismo , S-Adenosilmetionina/metabolismo , Células de Schwann/metabolismo , Animales , Diferenciación Celular/fisiología , División Celular/fisiología , Femenino , Genómica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Vaina de Mielina/fisiología , Nervios Periféricos/citología , Cultivo Primario de Células , Ratas , Células de Schwann/citología
18.
JIMD Rep ; 11: 149-63, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23733603

RESUMEN

We describe a family illustrating the diagnostic difficulties occurring when pyridoxine-responsive cystathionine beta-synthase (CBS) deficiency presents with thrombotic disease without associated ocular, skeletal, or CNS abnormalities, a situation increasingly recognized. This family had several thromboembolic episodes in two generations with apparently inconstant elevations of plasma total homocysteine (tHcy). When taking (sometimes even low amounts) of pyridoxine, the affected family members had low-normal tHcy and normal values for cystathionine, methionine, and cysteine. Withdrawal of vitamin therapy was necessary before lower cystathionine, elevated methionine, and decreased cysteine became apparent, a pattern suggestive of CBS deficiency, leading to the finding that the affected members were each compound heterozygotes for CBS p.G307S and p.P49L. To assist more accurate diagnosis of adults presenting with thrombophilia found to have elevated tHcy, the patterns of methionine-related metabolites in CBS-deficient patients are compared in this article to those in patients with homocysteine remethylation defects, including inborn errors of folate or cobalamin metabolism, and untreated severe cobalamin or folate deficiency. Usually serum cystathionine is low in subjects with CBS deficiency and elevated in those with remethylation defects. S-Adenosylmethionine and S-adenosylhomocysteine are often markedly elevated in CBS deficiency when tHcy is above 100 umol/L. We conclude that there are likely other undiagnosed, highly B6-responsive adult patients with CBS deficiency, and that additional testing of cystathionine, total cysteine, methionine, and S-adenosylmethionine will be helpful in diagnosing them correctly and distinguishing CBS deficiency from remethylation defects.

19.
Hepatology ; 58(4): 1296-305, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23505042

RESUMEN

UNLABELLED: Methionine adenosyltransferase 1A (MAT1A) and glycine N-methyltransferase (GNMT) are the primary genes involved in hepatic S-adenosylmethionine (SAMe) synthesis and degradation, respectively. Mat1a ablation in mice induces a decrease in hepatic SAMe, activation of lipogenesis, inhibition of triglyceride (TG) release, and steatosis. Gnmt-deficient mice, despite showing a large increase in hepatic SAMe, also develop steatosis. We hypothesized that as an adaptive response to hepatic SAMe accumulation, phosphatidylcholine (PC) synthesis by way of the phosphatidylethanolamine (PE) N-methyltransferase (PEMT) pathway is stimulated in Gnmt(-/-) mice. We also propose that the excess PC thus generated is catabolized, leading to TG synthesis and steatosis by way of diglyceride (DG) generation. We observed that Gnmt(-/-) mice present with normal hepatic lipogenesis and increased TG release. We also observed that the flux from PE to PC is stimulated in the liver of Gnmt(-/-) mice and that this results in a reduction in PE content and a marked increase in DG and TG. Conversely, reduction of hepatic SAMe following the administration of a methionine-deficient diet reverted the flux from PE to PC of Gnmt(-/-) mice to that of wildtype animals and normalized DG and TG content preventing the development of steatosis. Gnmt(-/-) mice with an additional deletion of perilipin2, the predominant lipid droplet protein, maintain high SAMe levels, with a concurrent increased flux from PE to PC, but do not develop liver steatosis. CONCLUSION: These findings indicate that excess SAMe reroutes PE towards PC and TG synthesis and lipid sequestration.


Asunto(s)
Hígado/metabolismo , Fosfatidilcolinas/metabolismo , Fosfatidiletanolaminas/metabolismo , S-Adenosilmetionina/metabolismo , Triglicéridos/metabolismo , Animales , Modelos Animales de Enfermedad , Hígado Graso/metabolismo , Hígado Graso/fisiopatología , Femenino , Glicina N-Metiltransferasa/deficiencia , Glicina N-Metiltransferasa/genética , Homeostasis/fisiología , Metabolismo de los Lípidos/fisiología , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Perilipina-2
20.
Gastroenterology ; 143(3): 787-798.e13, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22687285

RESUMEN

BACKGROUND & AIMS: Patients with cirrhosis are at high risk for developing hepatocellular carcinoma (HCC), and their liver tissues have abnormal levels of S-adenosylmethionine (SAMe). Glycine N-methyltransferase (GNMT) catabolizes SAMe, but its expression is down-regulated in HCC cells. Mice that lack GNMT develop fibrosis and hepatomas and have alterations in signaling pathways involved in carcinogenesis. We investigated the role of GNMT in human HCC cell lines and in liver carcinogenesis in mice. METHODS: We studied hepatoma cells from GNMT knockout mice and analyzed the roles of liver kinase B1 (LKB1, STK11) signaling via 5'-adenosine monophosphate-activated protein kinase (AMPK) and Ras in regulating proliferation and transformation. RESULTS: Hepatoma cells from GNMT mice had defects in LKB1 signaling to AMPK, making them resistant to induction of apoptosis by adenosine 3',5'-cyclic monophosphate activation of protein kinase A and calcium/calmodulin-dependent protein kinase kinase 2. Ras-mediated hyperactivation of LKB1 promoted proliferation of GNMT-deficient hepatoma cells and required mitogen-activated protein kinase 2 (ERK) and ribosomal protein S6 kinase polypeptide 2 (p90RSK). Ras activation of LKB1 required expression of RAS guanyl releasing protein 3 (RASGRP3). Reduced levels of GNMT and phosphorylation of AMPKα at Thr172 and increased levels of Ras, LKB1, and RASGRP3 in HCC samples from patients were associated with shorter survival times. CONCLUSIONS: Reduced expression of GNMT in mouse hepatoma cells and human HCC cells appears to increase activity of LKB1 and RAS; activation of RAS signaling to LKB1 and RASGRP3, via ERK and p90RSK, might be involved in liver carcinogenesis and be used as a prognostic marker. Reagents that disrupt this pathway might be developed to treat patients with HCC.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Glicina N-Metiltransferasa/deficiencia , Neoplasias Hepáticas/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Proteínas ras/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Azacitidina/farmacología , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Metilación de ADN , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Glicina N-Metiltransferasa/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Transfección , Carga Tumoral , Factores de Intercambio de Guanina Nucleótido ras
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...