Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Med Chem ; 67(1): 543-554, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38166392

RESUMEN

Small molecules capable of modulating methionine adenosyltransferase 2A (MAT2A) are of significant interest in precise cancer therapeutics. Herein, we raised the hole-electron Coulombic attraction as a reliable molecular descriptor for predicting the reactive oxygen generation capacity of MAT2A inhibitors, based on which we discovered compound H3 as a sonically activated degrader of MAT2A. Upon sonication, H3 can generate reactive oxygen species to specifically degrade cellular MAT2A via rapid oxidative reactions. Combination of H3 and sonication induced 87% MAT2A depletion in human colon cancer cells, thus elevating its antiproliferation effects by 8-folds. In vivo, H3 had a favorable pharmacokinetic profile (bioavailability = 77%) and ADME properties. Owing to the MAT2A degradation merits, H3 at a dosage of 10 mg/kg induced 31% tumor regression in xenograft colon tumor models. The significantly boosted antitumor potency can potentially alleviate the toxicity of high-dose MAT2A inhibitors to normal cells and tissues, especially to the liver.


Asunto(s)
Neoplasias Hepáticas , Metionina Adenosiltransferasa , Humanos , Metionina Adenosiltransferasa/metabolismo , Electrones , Neoplasias Hepáticas/metabolismo , S-Adenosilmetionina/metabolismo , Metionina
2.
Appl Microbiol Biotechnol ; 107(9): 2843-2854, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36941436

RESUMEN

Seleno-methylselenocysteine (SeMCys) is an effective component for selenium supplementation with anti-carcinogenic potential and can ameliorate neuropathology and cognitive deficits. In this study, we aimed to engineer Bacillus subtilis 168 for the microbial production of SeMCys. First, the accumulation of intracellular selenocysteine (SeCys) as the precursor of SeMCys was enhanced through overexpression of serine O-acetyltransferase, which was desensitized against feedback inhibition by cysteine. Next, the S-adenosylmethionine (SAM) synthetic pathway was optimized to improve methyl donor availability through expression of S-adenosylmethionine synthetase. Further, SeMCys was successfully produced through expression of the selenocysteine methyltransferase in SeCys and SAM-producing strain. The increased expression level of selenocysteine methyltransferase benefited the SeMCys production. Finally, all the heterologous genes were integrated into the genome of B. subtilis, and the strain produced SeMCys at a titer of 18.4 µg/L in fed-batch culture. This is the first report on the metabolic engineering of B. subtilis for microbial production of SeMCys and provides a good starting point for future pathway engineering to achieve the industrial-grade production of SeMCys. KEY POINTS: • Expression of the feedback-insensitive serine O-acetyltransferase provided B. subtilis the ability of accumulating SeCys. • SAM production was enhanced through expressing S-adenosylmethionine synthetase in B. subtilis. • Expression of selenocysteine methyltransferase in SeCys and SAM-accumulating strain facilitated SeMCys production.


Asunto(s)
Bacillus subtilis , Selenocisteína , Selenocisteína/genética , Selenocisteína/metabolismo , Bacillus subtilis/genética , Bacillus subtilis/metabolismo , Serina O-Acetiltransferasa/metabolismo , Metionina Adenosiltransferasa/metabolismo , Ingeniería Metabólica , S-Adenosilmetionina/metabolismo
3.
Development ; 150(20)2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-36975381

RESUMEN

Methionine is important for intestinal development and homeostasis in various organisms. However, the underlying mechanisms are poorly understood. Here, we demonstrate that the methionine adenosyltransferase gene Mat2a is essential for intestinal development and that the metabolite S-adenosyl-L-methionine (SAM) plays an important role in intestinal homeostasis. Intestinal epithelial cell (IEC)-specific knockout of Mat2a exhibits impaired intestinal development and neonatal lethality. Mat2a deletion in the adult intestine reduces cell proliferation and triggers IEC apoptosis, leading to severe intestinal epithelial atrophy and intestinal inflammation. Mechanistically, we reveal that SAM maintains the integrity of differentiated epithelium and protects IECs from apoptosis by suppressing the expression of caspases 3 and 8 and their activation. SAM supplementation improves the defective intestinal epithelium and reduces inflammatory infiltration sequentially. In conclusion, our study demonstrates that methionine metabolism and its intermediate metabolite SAM play essential roles in intestinal development and homeostasis in mice.


Asunto(s)
Metionina Adenosiltransferasa , S-Adenosilmetionina , Ratones , Animales , S-Adenosilmetionina/metabolismo , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Mucosa Intestinal/metabolismo , Metionina , Suplementos Dietéticos
4.
Nat Commun ; 13(1): 5696, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36171419

RESUMEN

Fatty liver is a highly heterogenous condition driven by various pathogenic factors in addition to the severity of steatosis. Protein insufficiency has been causally linked to fatty liver with incompletely defined mechanisms. Here we report that fatty liver is a sulfur amino acid insufficient state that promotes metabolic inflexibility via limiting coenzyme A availability. We demonstrate that the nutrient-sensing transcriptional factor EB synergistically stimulates lysosome proteolysis and methionine adenosyltransferase to increase cysteine pool that drives the production of coenzyme A and glutathione, which support metabolic adaptation and antioxidant defense during increased lipid influx. Intriguingly, mice consuming an isocaloric protein-deficient Western diet exhibit selective hepatic cysteine, coenzyme A and glutathione deficiency and acylcarnitine accumulation, which are reversed by cystine supplementation without normalizing dietary protein intake. These findings support a pathogenic link of dysregulated sulfur amino acid metabolism to metabolic inflexibility that underlies both overnutrition and protein malnutrition-associated fatty liver development.


Asunto(s)
Aminoácidos Sulfúricos , Hígado Graso , Aminoácidos Sulfúricos/metabolismo , Animales , Antioxidantes/metabolismo , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Coenzima A/metabolismo , Cisteína/metabolismo , Cistina/metabolismo , Proteínas en la Dieta/metabolismo , Hígado Graso/metabolismo , Glutatión/metabolismo , Homeostasis , Lípidos , Hígado/metabolismo , Metionina/metabolismo , Metionina Adenosiltransferasa/metabolismo , Ratones , Oxidación-Reducción
5.
Signal Transduct Target Ther ; 7(1): 192, 2022 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-35729157

RESUMEN

Folic acid, served as dietary supplement, is closely linked to one-carbon metabolism and methionine metabolism. Previous clinical evidence indicated that folic acid supplementation displays dual effect on cancer development, promoting or suppressing tumor formation and progression. However, the underlying mechanism remains to be uncovered. Here, we report that high-folate diet significantly promotes cancer development in mice with hepatocellular carcinoma (HCC) induced by DEN/high-fat diet (HFD), simultaneously with increased expression of methionine adenosyltransferase 2A (gene name, MAT2A; protein name, MATIIα), the key enzyme in methionine metabolism, and acceleration of methionine cycle in cancer tissues. In contrast, folate-free diet reduces MATIIα expression and impedes HFD-induced HCC development. Notably, methionine metabolism is dynamically reprogrammed with valosin-containing protein p97/p47 complex-interacting protein (VCIP135) which functions as a deubiquitylating enzyme to bind and stabilize MATIIα in response to folic acid signal. Consistently, upregulation of MATIIα expression is positively correlated with increased VCIP135 protein level in human HCC tissues compared to adjacent tissues. Furthermore, liver-specific knockout of Mat2a remarkably abolishes the advocating effect of folic acid on HFD-induced HCC, demonstrating that the effect of high or free folate-diet on HFD-induced HCC relies on Mat2a. Moreover, folate and multiple intermediate metabolites in one-carbon metabolism are significantly decreased in vivo and in vitro upon Mat2a deletion. Together, folate promotes the integration of methionine and one-carbon metabolism, contributing to HCC development via hijacking MATIIα metabolic pathway. This study provides insight into folate-promoted cancer development, strongly recommending the tailor-made folate supplement guideline for both sub-healthy populations and patients with cancer expressing high level of MATIIα expression.


Asunto(s)
Ácido Fólico , Metionina Adenosiltransferasa , Animales , Dieta , Ácido Fólico/farmacología , Neoplasias Hepáticas Experimentales/genética , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/patología , Metionina/metabolismo , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Ratones
6.
J Food Biochem ; 46(10): e14292, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35762419

RESUMEN

We studied the epigenetic regulation of how black carrot extract (BCE) protects against ethanol-induced hepatic damage. We have shown that the butanol-extracted fraction of BCE (BCE-BuOH) increased intracellular cyclic adenosine monophosphate (cAMP) levels by suppressing the expression of phosphodiesterase 4b (PDE4b); however, the detailed mechanism remains to be elucidated. We focused on changes in histone modifications involved in the suppression of pde4 expression. The methylation level of histone H3 lysine 9 (H3K9), which regulates gene expression of PDE4b, decreased after treatment with 100 mM ethanol but was significantly increased by treatment with 400 µg/ml BCE-BuOH. In contrast, ethanol induced an increase in H3K9 acetylation. However, treatment with BCE-BuOH inhibited the increase in acetylation through an increase in Sirtuin 1 (Sirt1), a histone deacetylase. Furthermore, BCE-BuOH treatment increased the level of methionine adenosyltransferase (MAT) 2a mRNA and increased intracellular S-adenosylmethionine. The present results indicate that BCE-BuOH is useful for protection against alcohol-induced hepatic injury. PRACTICAL APPLICATIONS: We have reported that black carrot extract (BCE) suppressed liver steatosis and liver fibrosis on a rat alcoholic liver disease model. The results from this study have shown that BCE regulated the alcoholic-induced hepatic injury at the level of epigenetic modifications. These results suggested that BCE is useful for protection against alcoholic-induced hepatic injury.


Asunto(s)
Daucus carota , Epigénesis Genética , Adenosina Monofosfato , Animales , Butanoles , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4/metabolismo , Daucus carota/genética , Etanol , Histonas/metabolismo , Lisina/metabolismo , Metionina Adenosiltransferasa/metabolismo , Extractos Vegetales , ARN Mensajero , Ratas , S-Adenosilmetionina/metabolismo , Sirtuina 1/genética , Sirtuina 1/metabolismo
7.
J Nutr ; 150(Suppl 1): 2506S-2517S, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33000152

RESUMEN

The metabolism of sulfur-containing amino acids (SAAs) requires an orchestrated interplay among several dozen enzymes and transporters, and an adequate dietary intake of methionine (Met), cysteine (Cys), and B vitamins. Known human genetic disorders are due to defects in Met demethylation, homocysteine (Hcy) remethylation, or cobalamin and folate metabolism, in Hcy transsulfuration, and Cys and hydrogen sulfide (H2S) catabolism. These disorders may manifest between the newborn period and late adulthood by a combination of neuropsychiatric abnormalities, thromboembolism, megaloblastic anemia, hepatopathy, myopathy, and bone and connective tissue abnormalities. Biochemical features include metabolite deficiencies (e.g. Met, S-adenosylmethionine (AdoMet), intermediates in 1-carbon metabolism, Cys, or glutathione) and/or their accumulation (e.g. S-adenosylhomocysteine, Hcy, H2S, or sulfite). Treatment should be started as early as possible and may include a low-protein/low-Met diet with Cys-enriched amino acid supplements, pharmacological doses of B vitamins, betaine to stimulate Hcy remethylation, the provision of N-acetylcysteine or AdoMet, or experimental approaches such as liver transplantation or enzyme replacement therapy. In several disorders, patients are exposed to long-term markedly elevated Met concentrations. Although these conditions may inform on Met toxicity, interpretation is difficult due to the presence of additional metabolic changes. Two disorders seem to exhibit Met-associated toxicity in the brain. An increased risk of demyelination in patients with Met adenosyltransferase I/III (MATI/III) deficiency due to biallelic mutations in the MATIA gene has been attributed to very high blood Met concentrations (typically >800 µmol/L) and possibly also to decreased liver AdoMet synthesis. An excessively high Met concentration in some patients with cystathionine ß-synthase deficiency has been associated with encephalopathy and brain edema, and direct toxicity of Met has been postulated. In summary, studies in patients with various disorders of SAA metabolism showed complex metabolic changes with distant cellular consequences, most of which are not attributable to direct Met toxicity.


Asunto(s)
Aminoácidos Sulfúricos/metabolismo , Cisteína/metabolismo , Homocisteína/metabolismo , Enfermedades Metabólicas/genética , Metionina/metabolismo , Compuestos de Azufre/metabolismo , Azufre/metabolismo , Animales , Encefalopatías/etiología , Encefalopatías/metabolismo , Glutatión/metabolismo , Homocistinuria/etiología , Homocistinuria/metabolismo , Humanos , Sulfuro de Hidrógeno/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Enfermedades Metabólicas/patología , Enfermedades Metabólicas/terapia , Errores Innatos del Metabolismo/patología , Errores Innatos del Metabolismo/terapia , Metionina Adenosiltransferasa/metabolismo , Metilación , S-Adenosilmetionina/metabolismo , Sulfitos/metabolismo
8.
J Ind Microbiol Biotechnol ; 45(5): 345-355, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29574602

RESUMEN

Streptomyces lincolnensis is generally utilized for the production of lincomycin A (Lin-A), a clinically useful antibiotic to treat Gram-positive bacterial infections. Three methylation steps, catalyzed by three different S-adenosylmethionine (SAM)-dependent methyltransferases, are required in the biosynthesis of Lin-A, and thus highlight the significance of methyl group supply in lincomycin production. In this study, we demonstrate that externally supplemented SAM cannot be taken in by cells and therefore does not enhance Lin-A production. Furthermore, bioinformatics and in vitro enzymatic assays revealed there exist two SAM synthetase homologs, MetK1 (SLCG_1651) and MetK2 (SLCG_3830) in S. lincolnensis that could convert L-methionine into SAM in the presence of ATP. Even though we attempted to inactivate metK1 and metK2, only metK2 was deleted in S. lincolnensis LCGL, named as ΔmetK2. Following a reduction of the intracellular SAM concentration, ΔmetK2 mutant exhibited a significant decrease of Lin-A in comparison to its parental strain. Individual overexpression of metK1 or metK2 in S. lincolnensis LCGL either elevated the amount of intracellular SAM, concomitant with 15% and 22% increase in Lin-A production, respectively. qRT-PCR assays showed that overexpression of either metK1 or metK2 increased the transcription of lincomycin biosynthetic genes lmbA and lmbR, and regulatory gene lmbU, indicating SAM may also function as a transcriptional activator. When metK1 and metK2 were co-expressed, Lin-A production was increased by 27% in LCGL, while by 17% in a high-yield strain LA219X.


Asunto(s)
Antibacterianos/metabolismo , Lincomicina/metabolismo , Metionina Adenosiltransferasa/metabolismo , Streptomyces/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , S-Adenosilmetionina , Metabolismo Secundario , Streptomyces/genética , Factores de Transcripción
9.
J Biosci Bioeng ; 125(1): 67-75, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28867618

RESUMEN

The leucine responsive regulatory protein (Lrp) is a global transcription factor that regulates the expression of genes involved in amino acid metabolism. To identify metabolic pathways and related genes under the control of Lrp in the acetic acid bacterium Komagataeibacter europaeus, the Kelrp null mutant (KGMA7110), which requires supplementation of all 20 amino acids for normal growth, was cultivated in minimal media containing or lacking particular amino acids. The results confirmed that KGMA7110 was auxotrophic for methionine and its catabolites S-adenosylmethionine (SAM) and spermidine (SPD). Quantitative reverse-transcription PCR analysis revealed lower metK (SAM synthetase) and mdtI (SPD efflux pump) expression in KGMA7110 than in wild-type KGMA0119. By contrast, these genes were significantly up-regulated in the Kelrp mutant lacking the putative C-terminal ligand-sensing domain (KGMA7203), indicating abnormal regulation of target genes by the KeLrp variant in KGMA7203. KGMA7110 (0.69±0.27 µM) and KGMA7203 (4.90±0.61 µM) excreted lower and higher quantities of SPD, respectively, than KGMA0119 (2.28±0.26 µM). This was attributed to imbalanced carbon flow caused by Kelrp disruption that respectively attenuated and stimulated metK and mdtI expression. These findings indicate that KeLrp plays a key role in SAM biosynthesis and intracellular polyamine homeostasis in K. europaeus.


Asunto(s)
Ácido Acético/metabolismo , Gluconacetobacter/metabolismo , Homeostasis , Proteína Reguladora de Respuesta a la Leucina/metabolismo , Metionina/metabolismo , Poliaminas/metabolismo , Eliminación de Gen , Gluconacetobacter/genética , Proteína Reguladora de Respuesta a la Leucina/deficiencia , Proteína Reguladora de Respuesta a la Leucina/genética , Metionina Adenosiltransferasa/metabolismo , S-Adenosilmetionina/metabolismo , Espermidina/metabolismo
10.
EMBO J ; 36(21): 3175-3193, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-29021282

RESUMEN

Methionine metabolism is critical for epigenetic maintenance, redox homeostasis, and animal development. However, the regulation of methionine metabolism remains unclear. Here, we provide evidence that SIRT1, the most conserved mammalian NAD+-dependent protein deacetylase, is critically involved in modulating methionine metabolism, thereby impacting maintenance of mouse embryonic stem cells (mESCs) and subsequent embryogenesis. We demonstrate that SIRT1-deficient mESCs are hypersensitive to methionine restriction/depletion-induced differentiation and apoptosis, primarily due to a reduced conversion of methionine to S-adenosylmethionine. This reduction markedly decreases methylation levels of histones, resulting in dramatic alterations in gene expression profiles. Mechanistically, we discover that the enzyme converting methionine to S-adenosylmethionine in mESCs, methionine adenosyltransferase 2a (MAT2a), is under control of Myc and SIRT1. Consistently, SIRT1 KO embryos display reduced Mat2a expression and histone methylation and are sensitive to maternal methionine restriction-induced lethality, whereas maternal methionine supplementation increases the survival of SIRT1 KO newborn mice. Our findings uncover a novel regulatory mechanism for methionine metabolism and highlight the importance of methionine metabolism in SIRT1-mediated mESC maintenance and embryonic development.


Asunto(s)
Desarrollo Embrionario/genética , Epigénesis Genética , Metionina Adenosiltransferasa/genética , Metionina/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Sirtuina 1/genética , Acetilación , Animales , Apoptosis , Diferenciación Celular , Embrión de Mamíferos , Histonas/genética , Histonas/metabolismo , Metabolómica , Metionina/administración & dosificación , Metionina Adenosiltransferasa/metabolismo , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Células Madre Embrionarias de Ratones/citología , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , S-Adenosilmetionina/metabolismo , Sirtuina 1/deficiencia
11.
Mol Nutr Food Res ; 61(11)2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28759161

RESUMEN

SCOPE: Serine lies at the central node linking biosynthetic flux from glycolysis to glutathione synthesis and one-carbon metabolic cycle which are closely related to antioxidant capacity. The present study was conducted to determine the effects of serine supplementation on oxidative stress and its relative mechanisms. METHODS AND RESULTS: Diquat treatment was performed to induce oxidative stress in mice and primary hepatocytes. The results showed that hepatic glutathione anti-oxidant systems were impaired and reactive oxygen species and homocysteine were increased in diquat-induced mice and hepatocytes, while such disadvantageous changes were diminished by serine supplementation both in vivo and in vitro. However, when cystathionine ß-synthase expression was inhibited by interference RNA in hepatocytes, the effects of serine supplementation on the improvement of glutathione synthesis and the alleviation of oxidative stress were diminished. Moreover, when hepatocytes were treated with cycloleucine, an inhibitor of methionine adenosyltransferase, the effects of serine supplementation on the improvement of methionine cycle and the alleviation of DNA hypomethylation and oxidative stress were also diminished. CONCLUSION: Our results indicated that serine supplementation alleviated oxidative stress via supporting glutathione synthesis and methionine cycle, mostly by condensing with homocysteine to synthesize cysteine and providing one-carbon units for homocysteine remethylation.


Asunto(s)
Antioxidantes/uso terapéutico , Suplementos Dietéticos , Glutatión/metabolismo , Hepatocitos/metabolismo , Metionina/metabolismo , Estrés Oxidativo , Serina/uso terapéutico , Animales , Antioxidantes/química , Antioxidantes/metabolismo , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Cicloleucina/farmacología , Cistationina betasintasa/antagonistas & inhibidores , Cistationina betasintasa/genética , Cistationina betasintasa/metabolismo , Metilación de ADN/efectos de los fármacos , Defoliantes Químicos/antagonistas & inhibidores , Defoliantes Químicos/toxicidad , Diquat/antagonistas & inhibidores , Diquat/toxicidad , Inhibidores Enzimáticos/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Homocisteína/metabolismo , Masculino , Metionina Adenosiltransferasa/antagonistas & inhibidores , Metionina Adenosiltransferasa/metabolismo , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Interferencia de ARN , Distribución Aleatoria , Serina/antagonistas & inhibidores , Serina/metabolismo , Organismos Libres de Patógenos Específicos
12.
Plant Physiol ; 172(1): 244-53, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27482079

RESUMEN

S-Adenosylmethionine is widely used in a variety of biological reactions and participates in the methionine (Met) metabolic pathway. In Arabidopsis (Arabidopsis thaliana), one of the four S-adenosylmethionine synthetase genes, METHIONINE ADENOSYLTRANSFERASE3 (MAT3), is highly expressed in pollen. Here, we show that mat3 mutants have impaired pollen tube growth and reduced seed set. Metabolomics analyses confirmed that mat3 pollen and pollen tubes overaccumulate Met and that mat3 pollen has several metabolite profiles, such as those of polyamine biosynthesis, which are different from those of the wild type. Additionally, we show that disruption of Met metabolism in mat3 pollen affected transfer RNA and histone methylation levels. Thus, our results suggest a connection between metabolism and epigenetics.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimología , Metionina Adenosiltransferasa/metabolismo , Tubo Polínico/enzimología , Arabidopsis/genética , Arabidopsis/crecimiento & desarrollo , Proteínas de Arabidopsis/genética , Regulación del Desarrollo de la Expresión Génica , Regulación de la Expresión Génica de las Plantas , Histonas/metabolismo , Metabolómica/métodos , Metionina/metabolismo , Metionina Adenosiltransferasa/genética , Metilación , Microscopía Fluorescente , Mutación , Plantas Modificadas Genéticamente , Polen/enzimología , Polen/genética , Polen/crecimiento & desarrollo , Tubo Polínico/genética , Tubo Polínico/crecimiento & desarrollo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , S-Adenosilmetionina/metabolismo , Semillas/genética , Semillas/crecimiento & desarrollo , Semillas/metabolismo
13.
Redox Biol ; 8: 18-23, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26722840

RESUMEN

BACKGROUND: The oxidation of the methionine adenosyltransferase (MAT) by the combined impact of peroxides contaminating parenteral nutrition (PN) and oxidized redox potential of glutathione is suspected to explain its inhibition observed in animals. A modification of MAT activity is suspected to be at origin of the PN-associated liver disease as observed in newborns. We hypothesized that the correction of redox potential of glutathione by adding glutathione in PN protects the MAT activity. AIM: To investigate whether the addition of glutathione to PN can reverse the inhibition of MAT observed in animal on PN. METHODS: Three days old guinea pigs received through a jugular vein catheter 2 series of solutions. First with methionine supplement, (1) Sham (no infusion); (2) PN: amino acids, dextrose, lipids and vitamins; (3) PN-GSSG: PN+10µM GSSG. Second without methionine, (4) D: dextrose; (5) D+180µM ascorbylperoxide; (6) D+350µM H2O2. Four days later, liver was sampled for determination of redox potential of glutathione and MAT activity in the presence or absence of 1mM DTT. Data were compared by ANOVA, p<0.05. RESULTS: MAT activity was 45±4% lower in animal infused with PN and 23±7% with peroxides generated in PN. The inhibition by peroxides was associated with oxidized redox potential and was reversible by DTT. Correction of redox potential (PN+GSSG) or DTT was without effect on the inhibition of MAT by PN. The slope of the linear relation between MAT activity and redox potential was two fold lower in animal infused with PN than in others groups. CONCLUSION: The present study suggests that prevention of peroxide generation in PN and/or correction of the redox potential by adding glutathione in PN are not sufficient, at least in newborn guinea pigs, to restore normal MAT activity.


Asunto(s)
Suplementos Dietéticos , Glutatión/metabolismo , Hígado/metabolismo , Metionina Adenosiltransferasa/metabolismo , Nutrición Parenteral , Animales , Biomarcadores , Activación Enzimática/efectos de los fármacos , Disulfuro de Glutatión/metabolismo , Cobayas , Hígado/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Soluciones para Nutrición Parenteral/farmacología , Peróxidos/metabolismo
14.
PLoS One ; 9(10): e108709, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25285660

RESUMEN

S-Adenosylmethionine synthetase (SAMS) catalyzes the synthesis of S-adenosylmethionine (SAM), a precursor for ethylene and polyamine biosynthesis. Here, we report the isolation of the 1498 bp full-length cDNA sequence encoding tetraploid black locust (Robinia pseudoacacia L.) SAMS (TrbSAMS), which contains an open reading frame of 1179 bp encoding 392 amino acids. The amino acid sequence of TrbSAMS has more than 94% sequence identity to SAMSs from other plants, with a closer phylogenetic relationship to SAMSs from legumes than to SAMS from other plants. The TrbSAMS monomer consists of N-terminal, central, and C-terminal domains. Subcellular localization analysis revealed that the TrbSAMS protein localizes mainly to in the cell membrane and cytoplasm of onion epidermal cells and Arabidopsis mesophyll cell protoplasts. Indole-3-butyric acid (IBA)-treated cuttings showed higher levels of TrbSAMS transcript than untreated control cuttings during root primordium and adventitious root formation. TrbSAMS and its downstream genes showed differential expression in shoots, leaves, bark, and roots, with the highest expression observed in bark. IBA-treated cuttings also showed higher SAMS activity than control cuttings during root primordium and adventitious root formation. These results indicate that TrbSAMS might play an important role in the regulation of IBA-induced adventitious root development in tetraploid black locust cuttings.


Asunto(s)
Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Genes de Plantas , Indoles/farmacología , Metionina Adenosiltransferasa/genética , Raíces de Plantas/crecimiento & desarrollo , Robinia/genética , Tetraploidía , Secuencia de Aminoácidos , Clonación Molecular , Etilenos/metabolismo , Metionina Adenosiltransferasa/química , Metionina Adenosiltransferasa/aislamiento & purificación , Metionina Adenosiltransferasa/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Raíces de Plantas/efectos de los fármacos , Raíces de Plantas/genética , Poliaminas/metabolismo , Estructura Secundaria de Proteína , Robinia/anatomía & histología , Robinia/efectos de los fármacos , Robinia/enzimología , Alineación de Secuencia , Análisis de Secuencia de Proteína , Fracciones Subcelulares/enzimología
15.
PLoS One ; 8(9): e75628, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24098708

RESUMEN

Methionine adenosyltransferase (MAT) is the cellular enzyme that catalyzes the synthesis of S-adenosylmethionine (SAM), the principal biological methyl donor and a key regulator of hepatocyte proliferation, death and differentiation. Two genes, MAT1A and MAT2A, encode 2 distinct catalytic MAT isoforms. A third gene, MAT2B, encodes a MAT2A regulatory subunit. In hepatocellular carcinoma (HCC), MAT1A downregulation and MAT2A upregulation occur, known as the MAT1A:MAT2A switch. The switch is accompanied with an increasing expression of MAT2B, which results in decreased SAM levels and facilitates cancer cell growth. Berberine, an isoquinoline alkaloid isolated from many medicinal herbs such as Coptis chinensis, has a wide range of pharmacological effects including anti-cancer effects. Because drug-induced microRNAs have recently emerged as key regulators in guiding their pharmacological effects, we examined whether microRNA expression is differentially altered by berberine treatment in HCC. In this study, we used microRNA microarrays to find that the expression level of miR-21-3p (previously named miR-21*) increased after berberine treatment in the HepG2 human hepatoma cell line. To predict the putative targets of miR-21-3p, we integrated the gene expression profiles of HepG2 cells after berberine treatment by comparing with a gene list generated from sequence-based microRNA target prediction software. We then confirmed these predictions through transfection of microRNA mimics and a 3' UTR reporter assay. Our findings provide the first evidence that miR-21-3p directly reduces the expression of MAT2A and MAT2B by targeting their 3' UTRs. In addition, an overexpression of miR-21-3p increased intracellular SAM contents, which have been proven to be a growth disadvantage for hepatoma cells. The overexpression of miR-21-3p suppresses growth and induces apoptosis in HepG2 cells. Overall, our results demonstrate that miR-21-3p functions as a tumor suppressor by directly targeting both MAT2A and MAT2B, indicating its therapeutic potential in HCC.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/fisiopatología , Metionina Adenosiltransferasa/metabolismo , MicroARNs/farmacología , Berberina/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cartilla de ADN/genética , Citometría de Flujo , Células HEK293 , Humanos , Luciferasas , MicroARNs/genética , MicroARNs/metabolismo , Análisis por Micromatrices , Mutagénesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Planta Med ; 79(12): 997-1002, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23807810

RESUMEN

It has been known that silymarin exhibits protective activity against oxidative liver injury induced by various hepatotoxicants, but the underlying mechanism of its beneficial action remains unclear. We determined the alterations in sulfur-containing amino acid metabolism induced by silymarin in association with its effects on the antioxidant capacity of liver. Male mice were treated with silymarin (100 or 200 mg/kg, p. o.) every 12 h for a total of 3 doses, and sacrificed 6 h after the final dosing. The hepatic methionine level was increased, but the activity and protein expression of methionine adenosyltransferase were decreased by silymarin in a dose-dependent manner. S-Adenosylmethionine or homocysteine concentration was not changed, whereas the sulfur-containing metabolites generated from homocysteine in the transsulfuration pathway including cystathionine, cysteine, and glutathione were increased significantly. Cystathionine ß-synthase was induced, but cysteine dioxygenase was downregulated, both of which would contribute to the elevation of cysteine and its product, glutathione, in liver. Oxygen radical scavenging capacity of liver cytosol against peroxyl radical and peroxynitrite was increased, and also hepatic lipid peroxidation was diminished in the silymarin-treated mice. Taken together, the results demonstrate that silymarin enhances hepatic glutathione generation by elevating cysteine availability via an increment in cysteine synthesis and an inhibition of its catabolism to taurine, which may subsequently contribute to the antioxidant defense of liver.


Asunto(s)
Aminoácidos Sulfúricos/metabolismo , Antioxidantes/metabolismo , Metionina Adenosiltransferasa/metabolismo , Silybum marianum/química , Silimarina/farmacología , Azufre/metabolismo , Aminoácidos Sulfúricos/análisis , Aminoácidos Sulfúricos/efectos de los fármacos , Animales , Antioxidantes/análisis , Glutatión/análisis , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Peroxidación de Lípido , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Taurina/análisis , Taurina/metabolismo
17.
Exp Cell Res ; 319(12): 1902-1911, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23588207

RESUMEN

Methionine adenosyltransferase (MAT) is an essential enzyme that is responsible for the biosynthesis of S-adenosylmethionine (SAMe), the principal methyl donor and precursor of polyamines. MAT1A is expressed in normal liver and MAT2A is expressed in all extrahepatic tissues. MAT2A expression is increased in human colon cancer and in colon cancer cells treated with mitogens, whereas silencing MAT2A resulted in apoptosis. The aim of the current work was to examine the mechanism responsible for MAT2A-dependent growth and apoptosis. We found that in RKO (human adenocarcinoma cell line) cells, MAT2A siRNA treatment lowered cellular SAMe and putrescine levels by 70-75%, increased apoptosis and inhibited growth. Putrescine supplementation blunted significantly MAT2A siRNA-induced apoptosis and growth suppression. Putrescine treatment (100pmol/L) raised MAT2A mRNA level to 4.3-fold of control, increased the expression of c-Jun and c-Fos and binding to an AP-1 site in the human MAT2A promoter and the promoter activity. In human colon cancer specimens, the expression levels of MAT2A, ornithine decarboxylase (ODC), c-Jun and c-Fos are all elevated as compared to adjacent non-tumorous tissues. Overexpression of ODC in RKO cells also raised MAT2A mRNA level and MAT2A promoter activity. ODC and MAT2A are also overexpressed in liver cancer and consistently, similar MAT2A-ODC-putrescine interactions and effects on growth and apoptosis were observed in HepG2 cells. In conclusion, there is a crosstalk between polyamines and MAT2A. Increased MAT2A expression provides more SAMe for polyamines biosynthesis; increased polyamine (putrescine in this case) can activate MAT2A at the transcriptional level. This along with increased ODC expression in cancer all feed forward to further enhance the proliferative capacity of the cancer cell.


Asunto(s)
Neoplasias del Colon/metabolismo , Neoplasias Hepáticas/metabolismo , Metionina Adenosiltransferasa/metabolismo , Putrescina/metabolismo , Anciano , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/enzimología , Femenino , Humanos , Neoplasias Hepáticas/enzimología , Masculino , Metionina Adenosiltransferasa/genética , Persona de Mediana Edad , Ornitina Descarboxilasa/genética , Ornitina Descarboxilasa/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Interferente Pequeño , S-Adenosilmetionina/metabolismo , Factor de Transcripción AP-1/metabolismo , Transcripción Genética
18.
Mol Genet Metab ; 107(3): 253-6, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22951388

RESUMEN

Methionine adenosyltransferase I/III (MAT I/III) deficiency, caused by mutations in the MAT1A gene, is an inherited metabolic disorder characterized by persistent hypermethioninemia, usually detected by newborn mass screening. There is a wide range of clinical manifestations, from completely asymptomatic to neurological problems associated with brain demyelination. Physiological role of S-adenosylmethionine (SAM), the metabolic product of methionine catalyzed by MAT, in the central nervous system has been investigated in vivo and in vitro, and case reports demonstrated an effectiveness of supplementary treatment of SAM in the improvement of neurological development and myelination. Methionine restriction can be an additional therapeutic strategy because hypermethioninemia alone may be neurotoxic; however, lowering methionine carries a risk to decrease the synthesis of SAM.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Encéfalo/metabolismo , Enfermedades Desmielinizantes/metabolismo , Metionina Adenosiltransferasa/metabolismo , Metionina/metabolismo , S-Adenosilmetionina/metabolismo , Alelos , Errores Innatos del Metabolismo de los Aminoácidos/dietoterapia , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/patología , Encéfalo/patología , Enfermedades Desmielinizantes/dietoterapia , Enfermedades Desmielinizantes/genética , Enfermedades Desmielinizantes/patología , Dieta , Pruebas Genéticas , Glicina N-Metiltransferasa/deficiencia , Humanos , Recién Nacido , Isoenzimas/genética , Isoenzimas/metabolismo , Metionina Adenosiltransferasa/deficiencia , Metionina Adenosiltransferasa/genética , Mutación , S-Adenosilmetionina/uso terapéutico , Índice de Severidad de la Enfermedad
19.
Mol Med ; 18: 423-32, 2012 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-22193356

RESUMEN

Methionine S-adenosyltransferase (MAT) catalyzes the only reaction that produces the major methyl donor in mammals. Low-dose methotrexate is the most commonly used disease-modifying antirheumatic drug in human rheumatic conditions. The present study was conducted to test the hypothesis that methotrexate inhibits MAT expression and activity in vitro and in vivo. HepG2 cells were cultured under folate restriction or in low-dose methotrexate with and without folate or methionine supplementation. Male C57BL/6J mice received methotrexate regimens that reflected low-dose clinical use in humans. S-adenosylmethionine and MAT genes, proteins and enzyme activity levels were determined. We found that methionine or folate supplementation greatly improved S-adenosylmethionine in folate-depleted cells but not in cells preexposed to methotrexate. Methotrexate but not folate depletion suppressed MAT genes, proteins and activity in vitro. Low-dose methotrexate inhibited MAT1A and MAT2A genes, MATI/II/III proteins and MAT enzyme activities in mouse tissues. Concurrent folinate supplementation with methotrexate ameliorated MAT2A reduction and restored S-adenosylmethionine in HepG2 cells. However, posttreatment folinate rescue failed to restore MAT2A reduction or S-adenosylmethionine level in cells preexposed to methotrexate. Our results provide both in vitro and in vivo evidence that low-dose methotrexate inhibits MAT genes, proteins, and enzyme activity independent of folate depletion. Because polyglutamated methotrexate stays in the hepatocytes, if methotrexate inhibits MAT in the liver, then the efficacy of clinical folinate rescue with respect to maintaining hepatic S-adenosylmethionine synthesis and normalizing the methylation reactions would be limited. These findings raise concerns on perturbed methylation reactions in humans on low-dose methotrexate. Future studies on the clinical physiological consequences of MAT inhibition by methotrexate and the potential benefits of S-adenosylmethionine supplementation on methyl group homeostasis in clinical methotrexate therapies are warranted.


Asunto(s)
Antirreumáticos/farmacología , Metionina Adenosiltransferasa/antagonistas & inhibidores , Metotrexato/farmacología , Animales , Dactinomicina , Dexametasona , Inhibidores Enzimáticos/farmacología , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Masculino , Metionina/farmacología , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , S-Adenosilmetionina/metabolismo
20.
Biochem Pharmacol ; 82(11): 1549-61, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21843511

RESUMEN

DHEA is known to have chemopreventive and antiproliferative activities, and was initially thought to be mediated by inhibition of G6PD. Our previous study has shown that DHEA may act through interference with energy metabolism. To study the effect of pharmacological dose of DHEA on cellular metabolism, and to further delineate the mechanism underlying its antiproliferative effect, we applied a metabolomic approach to globally profile the changes in metabolites in SK-Hep1 cells underexpressing G6PD (Sk-Gi) and control cells (Sk-Sc) after DHEA treatment. RRLC-TOF-MS was used to identify metabolites, and tandem mass spectrometry was used to confirm their identity. DHEA induced changes in glutathione metabolism, lipid metabolism, s-adenosylmethionine (SAM) metabolism, as well as lysine metabolism. Elevation in level of glutathione disulfide, together with a concomitant decrease in level of reduced glutathione, was indicative of increased oxidative stress. Depletion of carnitine and its acyl derivatives reflected decline in fatty acid catabolism. These changes were associated with mitochondrial malfunction and reduction in cellular ATP content. Cardiolipin (CL) and phosphatidylcholine (PC) levels decreased significantly, suggesting that alterations in lipid composition are causally related to decline in mitochondrial function after DHEA treatment. The decline in cellular SAM content was accompanied by decreased expression of methionine adenosyltransferase genes MAT2A and MAT2B. SAM supplementation partially rescued cells from DHEA-induced growth stagnation. Our findings suggest that DHEA causes perturbation of multiple pathways in cellular metabolism. Decreased SAM production, and cardiolipin depletion and the resulting mitochondrial dysfunction underlie the antiproliferative effect of DHEA.


Asunto(s)
Anticarcinógenos/farmacología , Antineoplásicos/farmacología , Deshidroepiandrosterona/farmacología , Metaboloma , Carcinoma Hepatocelular , Proliferación Celular/efectos de los fármacos , Cromatografía Liquida , Glucosafosfato Deshidrogenasa/metabolismo , Humanos , Neoplasias Hepáticas , Metionina Adenosiltransferasa/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo/efectos de los fármacos , Fosfatidilcolinas/metabolismo , Fosforilcolina/metabolismo , S-Adenosilmetionina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA