Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Immunol ; 12: 745784, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34616406

RESUMEN

Antiretroviral drugs effectively halt HIV-1 replication and disease progression, however, due to the presence of a stable viral latent reservoir, the infection cannot be cured by antiretroviral drugs alone. Elucidating the molecular mechanisms underlying HIV-1 latent infection remains a critical hurdle that precludes the development of novel therapeutic strategies aiming for a potential functional cure. Cellular metabolism has been reported to affect HIV-1 replication in CD4+ T cells, but it remains largely unclear whether it is involved in the regulation of HIV-1 latency. Here, we performed a sub-pooled CRISPR library knockout screen targeting 1773 metabolic-related genes in a cell model of HIV-1 latent infection and found that Methionine Adenosyltransferase 2A (MAT2A) contributes to HIV-1 latency. MAT2A knockout enhanced the reactivation of latent HIV-1 while MAT2A overexpression did the opposite. Mechanistically, MAT2A modulates HIV-1 latency through S-Adenosylmethionine (SAM)-mediated one-carbon flux. MAT2A knockout resulted in a significant downregulation of DNA and histone methylation at the HIV-1 5'-LTR. Importantly, we found that the plasma level of SAM is positively correlated with HIV-1 DNA in PBMCs from ART-treated infected individuals, suggesting SAM could serve as a potential biomarker for the latent viral reservoir. Overall, this study reveals an important role of MAT2A-mediated one-carbon metabolism in regulating HIV-1 latency and provides a promising target for the development of new strategies for a functional cure of HIV-1.


Asunto(s)
Linfocitos T CD4-Positivos/enzimología , Infecciones por VIH/inmunología , VIH-1/fisiología , Infección Latente/inmunología , Metionina Adenosiltransferasa/fisiología , S-Adenosilmetionina/sangre , Adulto , Fármacos Anti-VIH/uso terapéutico , Linfocitos T CD4-Positivos/inmunología , Sistemas CRISPR-Cas , Carbono/metabolismo , ADN Viral/sangre , Técnicas de Inactivación de Genes , Biblioteca de Genes , Células HEK293 , Infecciones por VIH/sangre , Infecciones por VIH/tratamiento farmacológico , Duplicado del Terminal Largo de VIH , Código de Histonas , Humanos , Células Jurkat , Infección Latente/sangre , Interferencia de ARN , ARN Interferente Pequeño/genética , Activación Viral
2.
Hepatology ; 70(6): 2018-2034, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31077594

RESUMEN

Methionine adenosyltransferase α1 (MATα1, encoded by MAT1A) is responsible for hepatic biosynthesis of S-adenosyl methionine, the principal methyl donor. MATα1 also act as a transcriptional cofactor by interacting and influencing the activity of several transcription factors. Mat1a knockout (KO) mice have increased levels of cytochrome P450 2E1 (CYP2E1), but the underlying mechanisms are unknown. The aims of the current study were to identify binding partners of MATα1 and elucidate how MATα1 regulates CYP2E1 expression. We identified binding partners of MATα1 by coimmunoprecipitation (co-IP) and mass spectrometry. Interacting proteins were confirmed using co-IP using recombinant proteins, liver lysates, and mitochondria. Alcoholic liver disease (ALD) samples were used to confirm relevance of our findings. We found that MATα1 negatively regulates CYP2E1 at mRNA and protein levels, with the latter being the dominant mechanism. MATα1 interacts with many proteins but with a predominance of mitochondrial proteins including CYP2E1. We found that MATα1 is present in the mitochondrial matrix of hepatocytes using immunogold electron microscopy. Mat1a KO hepatocytes had reduced mitochondrial membrane potential and higher mitochondrial reactive oxygen species, both of which were normalized when MAT1A was overexpressed. In addition, KO hepatocytes were sensitized to ethanol and tumor necrosis factor α-induced mitochondrial dysfunction. Interaction of MATα1 with CYP2E1 was direct, and this facilitated CYP2E1 methylation at R379, leading to its degradation through the proteasomal pathway. Mat1a KO livers have a reduced methylated/total CYP2E1 ratio. MATα1's influence on mitochondrial function is largely mediated by its effect on CYP2E1 expression. Patients with ALD have reduced MATα1 levels and a decrease in methylated/total CYP2E1 ratio. Conclusion: Our findings highlight a critical role of MATα1 in regulating mitochondrial function by suppressing CYP2E1 expression at multiple levels.


Asunto(s)
Citocromo P-450 CYP2E1/genética , Metionina Adenosiltransferasa/fisiología , Mitocondrias Hepáticas/fisiología , Animales , Femenino , Proteínas HSP70 de Choque Térmico/fisiología , Humanos , Hepatopatías Alcohólicas/metabolismo , Masculino , Potencial de la Membrana Mitocondrial , Metilación , Ratones , Proteínas Mitocondriales/fisiología , Especies Reactivas de Oxígeno/metabolismo
3.
Brain Dev ; 41(4): 382-388, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30389272

RESUMEN

Methionine adenosyltransferase I/III (MAT I/III) deficiency is characterized by persistent hypermethioninemia. The clinical manifestations in cases with MAT I/III deficiency vary from a complete lack of symptoms to neurological problems associated with brain demyelination. We experienced a neonatal case with MAT I/III deficiency, in which severe hypermethioninemia was detected during the newborn screening test. The patient gradually showed hyperreflexia, foot clonus, and irritability from the age of 1 month onwards, and his brain magnetic resonance imaging scans showed abnormal signal intensity in the bilateral central tegmental tracts. His neurological manifestations improved after the S-adenosylmethionine (SAMe) treatment, deteriorated after discontinuation of SAMe, and re-improved owing to re-administration of SAMe. He achieved normal neurodevelopment through SAMe and methionine restriction therapy. Lack of SAMe as well as severe hypermethioninemia were thought to contribute towards the clinical psychophysical state. Moreover, impaired MAT I/III activity contributed to the development of neurological disorder from the early neonatal period.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/fisiopatología , Glicina N-Metiltransferasa/deficiencia , Metionina Adenosiltransferasa/deficiencia , Metionina Adenosiltransferasa/fisiología , Encéfalo/metabolismo , Enfermedades Desmielinizantes/tratamiento farmacológico , Humanos , Lactante , Recién Nacido , Masculino , Metionina/metabolismo , Tamizaje Neonatal , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Tegmento Pontino/fisiopatología , S-Adenosilmetionina/uso terapéutico
4.
Gastroenterology ; 145(5): 1076-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23916847

RESUMEN

BACKGROUND & AIMS: Cirrhosis and liver cancer are potential outcomes of advanced nonalcoholic fatty liver disease (NAFLD). It is not clear what factors determine whether patients will develop advanced or mild NAFLD, limiting noninvasive diagnosis and treatment before clinical sequelae emerge. We investigated whether DNA methylation profiles can distinguish patients with mild disease from those with advanced NAFLD, and how these patterns are functionally related to hepatic gene expression. METHODS: We collected frozen liver biopsies and clinical data from patients with biopsy-proven NAFLD (56 in the discovery cohort and 34 in the replication cohort). Samples were divided into groups based on histologic severity of fibrosis: F0-1 (mild) and F3-4 (advanced). DNA methylation profiles were determined and coupled with gene expression data from the same biopsies; differential methylation was validated in subsets of the discovery and replication cohorts. We then analyzed interactions between the methylome and transcriptome. RESULTS: Clinical features did not differ between patients known to have mild or advanced fibrosis based on biopsy analysis. There were 69,247 differentially methylated CpG sites (76% hypomethylated, 24% hypermethylated) in patients with advanced vs mild NAFLD (P < .05). Methylation at fibroblast growth factor receptor 2, methionine adenosyl methyltransferase 1A, and caspase 1 was validated by bisulfite pyrosequencing and the findings were reproduced in the replication cohort. Methylation correlated with gene transcript levels for 7% of differentially methylated CpG sites, indicating that differential methylation contributes to differences in expression. In samples with advanced NAFLD, many tissue repair genes were hypomethylated and overexpressed, and genes in certain metabolic pathways, including 1-carbon metabolism, were hypermethylated and underexpressed. CONCLUSIONS: Functionally relevant differences in methylation can distinguish patients with advanced vs mild NAFLD. Altered methylation of genes that regulate processes such as steatohepatitis, fibrosis, and carcinogenesis indicate the role of DNA methylation in progression of NAFLD.


Asunto(s)
Metilación de ADN/fisiología , Progresión de la Enfermedad , Hígado Graso/fisiopatología , Índice de Severidad de la Enfermedad , Transcriptoma/fisiología , Adulto , Biopsia , Caspasa 1/genética , Caspasa 1/fisiología , Estudios de Cohortes , Islas de CpG/genética , Islas de CpG/fisiología , Metilación de ADN/genética , Diagnóstico Diferencial , Hígado Graso/diagnóstico , Hígado Graso/genética , Femenino , Regulación de la Expresión Génica/fisiología , Humanos , Hígado/metabolismo , Hígado/patología , Masculino , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/fisiología , Persona de Mediana Edad , Enfermedad del Hígado Graso no Alcohólico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/fisiología , Transcriptoma/genética
5.
Biochem J ; 452(1): 27-36, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23425511

RESUMEN

MAT (methionine adenosyltransferase) utilizes L-methionine and ATP to form SAM (S-adenosylmethionine), the principal methyl donor in biological methylation. Mammals encode a liver-specific isoenzyme, MAT1A, that is genetically linked with an inborn metabolic disorder of hypermethioninaemia, as well as a ubiquitously expressed isoenzyme, MAT2A, whose enzymatic activity is regulated by an associated subunit MAT2B. To understand the molecular mechanism of MAT functions and interactions, we have crystallized the ligand-bound complexes of human MAT1A, MAT2A and MAT2B. The structures of MAT1A and MAT2A in binary complexes with their product SAM allow for a comparison with the Escherichia coli and rat structures. This facilitates the understanding of the different substrate or product conformations, mediated by the neighbouring gating loop, which can be accommodated by the compact active site during catalysis. The structure of MAT2B reveals an SDR (short-chain dehydrogenase/reductase) core with specificity for the NADP/H cofactor, and harbours the SDR catalytic triad (YxxxKS). Extended from the MAT2B core is a second domain with homology with an SDR sub-family that binds nucleotide-sugar substrates, although the equivalent region in MAT2B presents a more open and extended surface which may endow a different ligand/protein-binding capability. Together, the results of the present study provide a framework to assign structural features to the functional and catalytic properties of the human MAT proteins, and facilitate future studies to probe new catalytic and binding functions.


Asunto(s)
Dominio Catalítico , Cristalización , Metionina Adenosiltransferasa/química , S-Adenosilmetionina/biosíntesis , Animales , Dominio Catalítico/fisiología , Cristalización/métodos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/fisiología , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/fisiología , Hígado/enzimología , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/metabolismo , Metionina Adenosiltransferasa/fisiología , Mapeo de Interacción de Proteínas/métodos , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/fisiología , Ratas , S-Adenosilmetionina/química , Especificidad por Sustrato/fisiología
6.
Hepatology ; 55(6): 1942-53, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22271545

RESUMEN

UNLABELLED: Methionine adenosyltransferases (MATs) are critical enzymes that catalyze the formation of the methyl donor S-adenosyl methionine (SAM). The MAT2A gene, which encodes the catalytic subunit α2, is induced in dedifferentiated liver. We previously demonstrated that MAT2A expression is enhanced in activated hepatic stellate cells (HSCs) and that silencing this gene reduces HSC activation. In this study, we examined the molecular mechanisms responsible for the transcriptional regulation of the MAT2A gene in HSCs. We identified peroxisome proliferator-activated receptor (PPAR) response elements (PPREs) in the rat MAT2A promoter. The PPARγ agonist rosiglitazone (RSG) promoted quiescence in the activated rat HSC cell line (BSC) or culture-activated primary rat HSCs, decreased MAT2A expression and promoter activity, and enhanced PPARγ binding to MAT2A PPREs. In vivo HSC activation in bile duct-ligated rats lowered PPARγ interaction with MAT2A PPREs. Silencing PPARγ increased MAT2A transcription, whereas overexpressing it had the opposite effect, demonstrating that PPARγ negatively controls this gene. Site-directed mutagenesis of PPREs abolished PPARγ recruitment to the MAT2A promoter and its inhibitory effect on MAT2A transcription in quiescent HSCs. PPRE mutations decreased the basal promoter activity of MAT2A in activated HSCs independent of PPARγ, indicating that other factors might be involved in PPRE interaction. We identified PPARß binding to wild-type but not to mutated PPREs in activated cells. Furthermore, silencing PPARß inhibited MAT2A expression and promoter activity. Forced expression of MAT2A in RSG-treated HSCs lowered PPARγ and enhanced PPARß expression, thereby promoting an activated phenotype. CONCLUSION: We identified PPARγ as a negative regulator of MAT2A in quiescent HSCs. A switch from quiescence to activation abolishes this control and allows PPARß to up-regulate MAT2A transcription.


Asunto(s)
Células Estrelladas Hepáticas/metabolismo , Metionina Adenosiltransferasa/genética , PPAR gamma/fisiología , PPAR-beta/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/fisiología , Masculino , Metionina Adenosiltransferasa/fisiología , Regiones Promotoras Genéticas , Ratas , Ratas Wistar , Elementos de Respuesta/fisiología , Rosiglitazona , Tiazolidinedionas/farmacología , Transcripción Genética
8.
Gastroenterology ; 134(1): 281-91, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18045590

RESUMEN

BACKGROUND & AIMS: Methionine adenosyltransferase (MAT) catalyzes S-adenosylmethionine biosynthesis. Two genes (MAT1A and MAT2A) encode for the catalytic subunit of MAT, while a third gene (MAT2beta) encodes for a regulatory subunit that modulates the activity of MAT2A-encoded isoenzyme. We uncovered multiple splicing variants while characterizing its 5'-flanking region. The aims of our current study are to examine the expression pattern, regulation, and functions of the 2 major variants: V1 and V2. METHODS: Studies were conducted using RNA from normal human tissues, resected hepatocellular carcinoma specimens, and cell lines. Gene expression, promoter and nuclear binding activities, growth, and apoptosis were measured by routine assays. RESULTS: MAT2beta is expressed in most but not all tissues, and the 2 variants are differentially expressed. The messenger RNA levels of both variants are markedly increased in hepatocellular carcinoma. Tumor necrosis factor (TNF)-alpha, which induces MAT2A in HepG2 cells, also induced V1 (but not V2) expression. TNF-alpha induced the promoter activity of MAT2beta V1, likely via nuclear factor kappaB and activator protein 1. Both variants regulate growth, but only V1 regulates apoptosis. Reduced expression of V1 led to c-Jun-N-terminal kinase (JNK) activation, apoptosis, and sensitized HepG2 cells to TNF-alpha-induced apoptosis, while overexpression of V1 was protective. However, blocking JNK1 or JNK2 activation did not prevent apoptosis induced by V1 knockdown. V1 (but not V2) knockdown also leads to apoptosis in a colon cancer cell line, suggesting these variants play similar roles in many cell types. CONCLUSIONS: Different variants of MAT2beta regulate growth and death, which broadens their importance in biology.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metionina Adenosiltransferasa/fisiología , Apoptosis/fisiología , Estudios de Casos y Controles , Técnicas de Cultivo de Célula , Línea Celular , Perfilación de la Expresión Génica , Humanos , Empalme del ARN/genética , Factor de Necrosis Tumoral alfa/fisiología
9.
Biochim Biophys Acta ; 1760(1): 10-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16280200

RESUMEN

Methionine adenosyltransferase (MAT: EC 2.5.1.6) catalyzes the synthesis of S-adenosylmethionine (AdoMet) in two sequential steps, AdoMet formation and subsequent tripolyphosphate (PPPi) cleavage, induced by AdoMet. In pursuit of a better understanding of the biological function of the enzyme, the MAT gene was cloned into vector PX63NEO to induce episomal overexpression in leishmania parasites. Neomycin-selected clones originated a strain of such overexpressing parasites that accumulated more than 3-fold AdoMet than mock-transfected cells and showed over ten times the wild type MAT activity, concurring with a significant accumulation of the MAT protein during the early logarithmic phase and MAT transcripts throughout the growth cycle. The rate of AdoMet efflux, practically nil in the control promastigotes, was exceptionally high in the MAT-overexpressing parasites, whilst growth in this strain was comparable to development in control cells, i.e., it was not affected by deleterious hypermethylation. Moreover, the modified strain was 10-fold more resistant to sinefungin, a S-adenosylmethionine-like antibiotic, than control cells. The effects of overexpression on polyamine metabolism and transport were likewise studied.


Asunto(s)
Leishmania donovani/enzimología , Metionina Adenosiltransferasa/genética , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Clonación Molecular , Resistencia a Medicamentos , Leishmania donovani/genética , Metionina Adenosiltransferasa/fisiología , Neomicina , Poliaminas/metabolismo , S-Adenosilmetionina/biosíntesis , Trypanosomatina/enzimología , Trypanosomatina/genética
10.
Alcohol ; 35(3): 227-34, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16054984

RESUMEN

Two genes (MAT1A and MAT2A) encode for the essential enzyme methionine adenosyltransferase (MAT), which catalyzes the biosynthesis of S-adenosylmethionine (SAMe), the principal methyl donor and, in the liver, a precursor of glutathione. MAT1A is expressed mostly in the liver, whereas MAT2A is widely distributed. MAT2A is induced in the liver during periods of rapid growth and dedifferentiation. In human hepatocellular carcinoma (HCC) MAT1A is replaced by MAT2A. This is important pathogenetically because MAT2A expression is associated with lower SAMe levels and faster growth, whereas exogenous SAMe treatment inhibits growth. Rats fed ethanol intragastrically for 9 weeks also exhibit a relative switch in hepatic MAT expression, decreased SAMe levels, hypomethylation of c-myc, increased c-myc expression, and increased DNA strand break accumulation. Patients with alcoholic liver disease have decreased hepatic MAT activity owing to both decreased MAT1A expression and inactivation of the MAT1A-encoded isoenzymes, culminating in decreased SAMe biosynthesis. Consequences of chronic hepatic SAMe depletion have been examined in the MAT1A knockout mouse model. In this model, the liver is more susceptible to injury. In addition, spontaneous steatohepatitis develops by 8 months, and HCC develops by 18 months. Accumulating evidence shows that, in addition to being a methyl donor, SAMe controls hepatocyte growth response and death response. Whereas transient SAMe depletion is necessary for the liver to regenerate, chronic hepatic SAMe depletion may lead to malignant transformation. It is interesting that SAMe is antiapoptotic in normal hepatocytes, but proapoptotic in liver cancer cells. This should make SAMe an attractive agent for both chemoprevention and treatment of HCC.


Asunto(s)
Depresores del Sistema Nervioso Central/efectos adversos , Etanol/efectos adversos , Neoplasias Hepáticas/inducido químicamente , Metionina Adenosiltransferasa/fisiología , S-Adenosilmetionina/fisiología , Animales , Muerte Celular/efectos de los fármacos , Femenino , Hepatocitos/efectos de los fármacos , Hepatocitos/fisiología , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Hígado/citología , Hígado/metabolismo , Hepatopatías Alcohólicas/enzimología , Hepatopatías Alcohólicas/metabolismo , Metionina/metabolismo , Metionina Adenosiltransferasa/biosíntesis , Metionina Adenosiltransferasa/genética , S-Adenosilmetionina/metabolismo
11.
J Biol Chem ; 279(32): 33273-80, 2004 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-15190060

RESUMEN

Leishmania is a trypanosomatid parasite causing serious disease and displaying resistance to various drugs. Here, we present comparative proteomic analyses of Leishmania major parasites that have been either shocked with or selected in vitro for high level resistance to the model antifolate drug methotrexate. Numerous differentially expressed proteins were identified by these experiments. Some were associated with the stress response, whereas others were found to be overexpressed due to genetic linkage to primary resistance mediators present on DNA amplicons. Several proteins not previously associated with resistance were also identified. The role of one of these, methionine adenosyltransferase, was confirmed by gene transfection and metabolite analysis. After a single exposure to low levels of methotrexate, L. major methionine adenosyltransferase transfectants could grow at high concentrations of the drug. Methotrexate resistance was also correlated to increased cellular S-adenosylmethionine levels. The folate and S-adenosylmethionine regeneration pathways are intimately connected, which may provide a basis for this novel resistance phenotype. This thorough comparative proteomic analysis highlights the variety of responses required for drug resistance to be achieved.


Asunto(s)
Resistencia a Medicamentos/genética , Leishmania major/metabolismo , Metionina Adenosiltransferasa/fisiología , Metotrexato , Proteínas Protozoarias/genética , S-Adenosilmetionina/fisiología , Animales , Northern Blotting , Southern Blotting , Electroforesis en Gel Bidimensional , Expresión Génica , Concentración de Iones de Hidrógeno , Hidroxiprostaglandina Deshidrogenasas/genética , Leishmania major/efectos de los fármacos , Leishmania major/genética , Metionina Adenosiltransferasa/genética , Metotrexato/farmacología , Mutación , Fosfopiruvato Hidratasa/genética , Reacción en Cadena de la Polimerasa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
FASEB J ; 18(7): 914-6, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15033934

RESUMEN

Methionine adenosyltransferase (MAT) is an essential enzyme because it catalyzes the formation of S-adenosylmethionine (SAMe), the principal biological methyl donor. Of the two genes that encode MAT, MAT1A is mainly expressed in adult liver and MAT2A is expressed in all extrahepatic tissues. Mice lacking MAT1A have reduced hepatic SAMe content and spontaneously develop hepatocellular carcinoma. The current study examined the influence of chronic hepatic SAMe deficiency on liver regeneration. Despite having higher baseline hepatic staining for proliferating cell nuclear antigen, MAT1A knockout mice had impaired liver regeneration after partial hepatectomy (PH) as determined by bromodeoxyuridine incorporation. This can be explained by an inability to up-regulate cyclin D1 after PH in the knockout mice. Upstream signaling pathways involved in cyclin D1 activation include nuclear factor kappaB (NFkappaB), the c-Jun-N-terminal kinase (JNK), extracellular signal-regulated kinases (ERKs), and signal transducer and activator of transcription-3 (STAT-3). At baseline, JNK and ERK are more activated in the knockouts whereas NFkappaB and STAT-3 are similar to wild-type mice. Following PH, early activation of these pathways occurred, but although they remained increased in wild-type mice, c-jun and ERK phosphorylation fell progressively in the knockouts. Hepatic SAMe levels fell progressively following PH in wild-type mice but remained unchanged in the knockouts. In culture, MAT1A knockout hepatocytes have higher baseline DNA synthesis but failed to respond to the mitogenic effect of hepatocyte growth factor. Taken together, our findings define a critical role for SAMe in ERK signaling and cyclin D1 regulation during regeneration and suggest chronic hepatic SAMe depletion results in loss of responsiveness to mitogenic signals.


Asunto(s)
Hepatocitos/enzimología , Regeneración Hepática/fisiología , Hígado/enzimología , Metionina Adenosiltransferasa/fisiología , S-Adenosilmetionina/fisiología , Adenosina Trifosfato/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Ciclina D1/fisiología , Replicación del ADN , Proteínas de Unión al ADN/fisiología , Perfilación de la Expresión Génica , Hepatectomía/métodos , Factor de Crecimiento de Hepatocito/farmacología , Hepatocitos/metabolismo , Interleucina-6/fisiología , Proteínas Quinasas JNK Activadas por Mitógenos , Hígado/metabolismo , Regeneración Hepática/genética , Sistema de Señalización de MAP Quinasas , Masculino , Metionina Adenosiltransferasa/deficiencia , Metionina Adenosiltransferasa/genética , Ratones , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/fisiología , Mitosis/efectos de los fármacos , FN-kappa B/fisiología , Óxido Nítrico/fisiología , Especificidad de Órganos , ARN Mensajero/biosíntesis , S-Adenosilmetionina/deficiencia , Factor de Transcripción STAT3 , Transducción de Señal , Transactivadores/fisiología , Factor de Necrosis Tumoral alfa/fisiología
13.
FASEB J ; 16(10): 1292-4, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12060674

RESUMEN

In mammals, methionine metabolism occurs mainly in the liver via methionine adenosyltransferase-catalyzed conversion to S-adenosylmethionine. Of the two genes that encode methionine adenosyltransferase(MAT1Aand MAT2A), MAT1A is mainly expressed in adult liver whereas MAT2A is expressed in all extrahepatic tissues. Mice lacking MAT1A have reduced hepatic S-adenosylmethionine content and hyperplasia and spontaneously develop nonalcoholic steatohepatitis. In this study, we examined whether chronic hepatic S-adenosylmethionine deficiency generates oxidative stress and predisposes to injury and malignant transformation. Differential gene expression in MAT1A knockout mice was analyzed following the criteria of the Gene Ontology Consortium. Susceptibility of MAT1A knockout mice to CCl4-induced hepatotoxicity and malignant transformation was determined in 3- and 18-month-old mice, respectively. Analysis of gene expression profiles revealed an abnormal expression of genes involved in the metabolism of lipids and carbohydrates in MAT1A knockout mice, a situation that is reminiscent of that found in diabetes, obesity, and other conditions associated with nonalcoholic steatohepatitis. This aberrant expression of metabolic genes in the knockout mice was associated with hyperglycemia, increased hepatic CYP2E1 and UCP2 expression and triglyceride levels, and reduced hepatic glutathione content. The knockout animals have increased lipid peroxidation and enhanced sensitivity to CCl4-induced liver damage, which was largely due to increased CYP2E1 expression because diallyl sulfide, an inhibitor of CYP2E1, prevented CCl4-induced liver injury. Hepatocellular carcinoma developed in more than half of the knockout mice by 18 months of age. Taken together, our findings define a critical role for S-adenosylmethionine in maintaining normal hepatic function and tumorigenesis of the liver.


Asunto(s)
Neoplasias Hepáticas Experimentales/etiología , Proteínas de Transporte de Membrana , Metionina Adenosiltransferasa/fisiología , Proteínas Mitocondriales , Estrés Oxidativo , Animales , Tetracloruro de Carbono , Enfermedad Hepática Inducida por Sustancias y Drogas , Citocromo P-450 CYP2E1/genética , Citocromo P-450 CYP2E1/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Susceptibilidad a Enfermedades , Perfilación de la Expresión Génica , Hepatitis Animal/etiología , Hepatitis Animal/genética , Hepatitis Animal/metabolismo , Canales Iónicos , Hígado/metabolismo , Hepatopatías/enzimología , Neoplasias Hepáticas Experimentales/patología , Metionina Adenosiltransferasa/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Obesidad/genética , Obesidad/metabolismo , Biosíntesis de Proteínas , Proteínas/genética , ARN Mensajero/biosíntesis , S-Adenosilmetionina/deficiencia , Proteína Desacopladora 2
14.
Proc Natl Acad Sci U S A ; 98(10): 5560-5, 2001 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-11320206

RESUMEN

Liver-specific and nonliver-specific methionine adenosyltransferases (MATs) are products of two genes, MAT1A and MAT2A, respectively, that catalyze the formation of S-adenosylmethionine (AdoMet), the principal biological methyl donor. Mature liver expresses MAT1A, whereas MAT2A is expressed in extrahepatic tissues and is induced during liver growth and dedifferentiation. To examine the influence of MAT1A on hepatic growth, we studied the effects of a targeted disruption of the murine MAT1A gene. MAT1A mRNA and protein levels were absent in homozygous knockout mice. At 3 months, plasma methionine level increased 776% in knockouts. Hepatic AdoMet and glutathione levels were reduced by 74 and 40%, respectively, whereas S-adenosylhomocysteine, methylthioadenosine, and global DNA methylation were unchanged. The body weight of 3-month-old knockout mice was unchanged from wild-type littermates, but the liver weight was increased 40%. The Affymetrix genechip system and Northern and Western blot analyses were used to analyze differential expression of genes. The expression of many acute phase-response and inflammatory markers, including orosomucoid, amyloid, metallothionein, Fas antigen, and growth-related genes, including early growth response 1 and proliferating cell nuclear antigen, is increased in the knockout animal. At 3 months, knockout mice are more susceptible to choline-deficient diet-induced fatty liver. At 8 months, knockout mice developed spontaneous macrovesicular steatosis and predominantly periportal mononuclear cell infiltration. Thus, absence of MAT1A resulted in a liver that is more susceptible to injury, expresses markers of an acute phase response, and displays increased proliferation.


Asunto(s)
División Celular/genética , Regulación Enzimológica de la Expresión Génica , Predisposición Genética a la Enfermedad , Cirrosis Hepática Experimental/genética , Metionina Adenosiltransferasa/fisiología , Animales , Metilación de ADN , Modelos Animales de Enfermedad , Hígado/metabolismo , Cirrosis Hepática Experimental/metabolismo , Metionina/sangre , Metionina/metabolismo , Metionina Adenosiltransferasa/genética , Ratones , Ratones Noqueados , Fenotipo
15.
Pharmacol Ther ; 85(1): 1-9, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10674710

RESUMEN

Hepatic methionine adenosyltransferase (MAT) deficiency is caused by mutations in the human MAT1A gene that abolish or reduce hepatic MAT activity that catalyzes the synthesis of S-adenosylmethionine from methionine and ATP. This genetic disorder is characterized by isolated persistent hypermethioninemia in the absence of cystathionine beta-synthase deficiency, tyrosinemia, or liver disease. Depending on the nature of the genetic defect, hepatic MAT deficiency can be transmitted either as an autosomal recessive or dominant trait. Genetic analyses have revealed that mutations identified in the MAT1A gene only partially inactivate enzymatic activity, which is consistent with the fact that most hepatic MAT-deficient individuals are clinically well. Two hypermethioninemic individuals with null MAT1A mutations have developed neurological problems, including brain demyelination, although this correlation is by no means absolute. Presently, it is recommended that a DNA-based diagnosis should be performed for isolated hypermethioninemic individuals with unusually high plasma methionine levels to assess if therapy aimed at the prevention of neurological manifestations is warranted.


Asunto(s)
Isoenzimas/genética , Hígado/enzimología , Metionina Adenosiltransferasa/deficiencia , Mutación/genética , Adenosina Trifosfato/metabolismo , Humanos , Metionina/metabolismo , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/fisiología , S-Adenosilmetionina/biosíntesis
16.
Cancer Res ; 58(7): 1444-50, 1998 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-9537246

RESUMEN

Methionine adenosyltransferase (MAT) catalyzes the formation of S-adenosylmethionine (SAM), the principal methyl donor, and is essential to normal cell function. The two forms of MAT, liver specific and non-liver specific, are products of two genes, MAT1A and MAT2A, respectively. We have reported a switch from MAT1A to MAT2A gene expression in human liver cancer cells. In the current work, we examined whether the type of MAT expressed by the cell influences cell growth. HuH-7 cells were stably transfected with MAT1A and were subsequently treated with antisense oligonucleotides directed against MAT2A. MAT2A antisense treatment reduced the amount of MAT2A mRNA by 99% but had no effect on MAT1A mRNA. Cell growth and DNA synthesis rates were reduced by approximately 20-25% after transfection with MAT1A and by an additional 30-40% after MAT2A antisense treatment. SAM level and SAM:S-adenosylhomocysteine (SAH) ratio increased by 50-75% after MAT1A transfection and by an additional 60-80% after MAT2A antisense treatment. DNA methylation changed in parallel to changes in SAM level and SAM:SAH ratio. Supplementing untransfected HuH-7 cells with SAM in the culture medium increased SAM level, SAM:SAH ratio, and DNA methylation and decreased cell growth and DNA synthesis. In conclusion, cell growth is influenced by the type of MAT expressed. The mechanism likely involves changes in SAM:SAH ratio and DNA methylation.


Asunto(s)
Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Metionina Adenosiltransferasa/biosíntesis , División Celular/fisiología , ADN de Neoplasias/biosíntesis , Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metionina Adenosiltransferasa/genética , Metionina Adenosiltransferasa/fisiología , Células Tumorales Cultivadas
19.
J Neurochem ; 36(6): 2092-3, 1981 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7241150

RESUMEN

The circadian rhythm of methionine S-adenosyltransferase, which catalyzes the formation of S-adenosylmethionine, a cosubstrate for melatonin in the pineal gland, follows the pattern of hydroxyindole-O-methyltransferase. Around the middle of the dark period, methionine S-adenosyltransferase and hydroxyindole-O-methyltransferase appear to be elevated by 2.5- and 1.5-fold, respectively, and tend to fall back during the light period.


Asunto(s)
Ritmo Circadiano , Metionina Adenosiltransferasa/fisiología , Glándula Pineal/enzimología , Transferasas/fisiología , Acetilserotonina O-Metiltransferasa/fisiología , Animales , Masculino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...