Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 274
Filtrar
1.
Mol Biol (Mosk) ; 57(4): 717-725, 2023.
Artículo en Ruso | MEDLINE | ID: mdl-37528794

RESUMEN

Enzymatic methyltransferase reactions are of crucial importance for cell metabolism. S-Adenosyl-L-methionine (AdoMet) is a main donor of the methyl group. DNA, RNA, proteins, and low-molecular-weight compounds are substrates of methyltransferases. In mammals, DNA methyltransferase Dnmt3a de novo methylates the C5 position of cytosine residues in CpG sequences in DNA. The methylation pattern is one of the factors that determine the epigenetic regulation of gene expression. Here, interactions with the catalytic domain of Dnmt3a was for the first time studied for phosphonous and phosphonic analogs of AdoMet and S-adenosyl-L-homocysteine (AdoHcy), in which the carboxyl group was substituted for respective phosphorus-containing group. These AdoMet analogs were shown to be substrates of Dnmt3a, and the methylation efficiency was only halved as compared with that of natural AdoMet. Both phosphorus-containing analogs of AdoHcy, which is a natural methyltransferase inhibitor, showed similar inhibitory activities toward Dnmt3a and were approximately four times less active than AdoHcy. The finding that the phosphonous and phosphonic analogs are similar in activity was quite unexpected because the geometry and charge of their phosphorus-containing groups differ substantially. The phosphorus-containing analogs of AdoMet and AdoHcy are discussed as promising tools for investigation of methyltransferases.


Asunto(s)
S-Adenosilhomocisteína , S-Adenosilmetionina , Animales , S-Adenosilmetionina/metabolismo , S-Adenosilhomocisteína/metabolismo , S-Adenosilhomocisteína/farmacología , Epigénesis Genética , Metionina/metabolismo , Metiltransferasas/metabolismo , ADN/metabolismo , Mamíferos
2.
Bioorg Med Chem Lett ; 25(7): 1532-7, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25746813

RESUMEN

A series of novel enhancer of zeste homolog 2 (EZH2) inhibitors was designed based on the chemical structure of the histone methyltransferase (HMT) inhibitor SAH (S-adenosyl-l-homocysteine). These nucleoside-based EZH2 inhibitors blocked the methylation of nucleosomes at H3K27 in biochemical assays employing both WT PRC2 complex as well as a Y641N mutant PRC2 complex. The most potent compound, 27, displayed IC50's against both complexes of 270 nM and 70 nM, respectively. To our knowledge, compound 27 is the most potent SAH-derived inhibitor of the EZH2 PRC2 complex yet identified. This compound also displayed improved potency, lipophilic efficiency (LipE), and selectivity profile against other lysine methyltransferases compared with SAH.


Asunto(s)
Complejo Represivo Polycomb 2/antagonistas & inhibidores , S-Adenosilhomocisteína/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Modelos Moleculares , Estructura Molecular , S-Adenosilhomocisteína/síntesis química , S-Adenosilhomocisteína/química , Relación Estructura-Actividad
3.
Zygote ; 23(5): 758-70, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25314965

RESUMEN

Adult canine fibroblasts stably transfected with either cytomegalovirus (CMV) or POU5F1 promoter-driven enhanced green fluorescent protein (EGFP) were used to investigate if pre-treatment of these donor cells with two epigenetic drugs [trichostatin A (TSA), or S-adenosylhomocysteine (SAH)] can improve the efficiency of interspecies somatic cell nuclear transfer (iSCNT). Fluorescence-activated cell sorting (FACS), analyses revealed that TSA, but not SAH, treatment of both transgenic and non-transgenic fibroblasts significantly increased acetylation levels compared with untreated relatives. The expression levels of Bcl2 and P53 were significantly affected in TSA-treated cells compared with untreated cells, whereas SAH treatment had no significant effect on cell apoptosis. Irrespective of epigenetic modification, dog/bovine iSCNT embryos had overall similar rates of cleavage and development to 8-16-cell and morula stages in non-transgenic groups. For transgenic reconstructed embryos, however, TSA and SAH could significantly improve development to 8-16-cell and morula stages compared with control. Even though, irrespective of cell transgenesis and epigenetic modification, none of the iSCNT embryos developed to the blastocyst stage. The iSCNT embryos carrying CMV-EGFP expressed EGFP at all developmental stages (2-cell, 4-cell, 8-16-cell, and morula) without mosaicism, while no POU5F1-EGFP signal was observed in any stage of developing iSCNT embryos irrespective of TSA/SAH epigenetic modifications. These results indicated that bovine oocytes partially remodel canine fibroblasts and that TSA and SAH have marginal beneficial effects on this process.


Asunto(s)
Clonación de Organismos/métodos , Desarrollo Embrionario/efectos de los fármacos , Epigénesis Genética , Proteínas Fluorescentes Verdes/metabolismo , Ácidos Hidroxámicos/farmacología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , S-Adenosilhomocisteína/farmacología , Acetilación/efectos de los fármacos , Animales , Animales Modificados Genéticamente/embriología , Animales Modificados Genéticamente/crecimiento & desarrollo , Animales Modificados Genéticamente/metabolismo , Apoptosis , Blastocisto/citología , Blastocisto/efectos de los fármacos , Blastocisto/metabolismo , Bovinos/embriología , Bovinos/crecimiento & desarrollo , Bovinos/metabolismo , Células Cultivadas , Quimera/embriología , Quimera/crecimiento & desarrollo , Quimera/metabolismo , Clonación de Organismos/veterinaria , Perros/embriología , Perros/crecimiento & desarrollo , Perros/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Proteínas Fluorescentes Verdes/genética , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Técnicas de Transferencia Nuclear/veterinaria , Factor 3 de Transcripción de Unión a Octámeros/genética , Oocitos/citología , Oocitos/efectos de los fármacos , Oocitos/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
4.
Physiol Genomics ; 46(17): 617-23, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25180283

RESUMEN

S-adenosylmethionine (SAM), the unique methyl donor in DNA methylation, has been shown to lower lipopolysaccharide (LPS)-induced expression of the proinflammatory cytokine TNF-α and increase the expression of the anti-inflammatory cytokine IL-10 in macrophages. The aim of this study was to assess whether epigenetic mechanisms mediate the anti-inflammatory effects of SAM. Human monocytic THP1 cells were differentiated into macrophages and treated with 0, 500, or 1,000 µmol/l SAM for 24 h, followed by stimulation with LPS. TNFα and IL-10 expression levels were measured by real-time PCR, cellular concentrations of SAM and S-adenosylhomocysteine (SAH), a metabolite of SAM, were measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), and DNA methylation was measured with LC-MS/MS and microarrays. Relative to control (0 µmol/l SAM), treatment with 500 µmol/l SAM caused a significant decrease in TNF-α expression (-45%, P < 0.05) and increase in IL-10 expression (+77%, P < 0.05). Treatment with 1,000 µmol/l SAM yielded no significant additional benefits. Relative to control, 500 µmol/l SAM increased cellular SAM concentrations twofold without changes in SAH, and 1,000 µmol/l SAM increased cellular SAM sixfold and SAH fourfold. Global DNA methylation increased 7% with 500 µmol/l SAM compared with control. Following treatment with 500 µmol/l SAM, DNA methylation microarray analysis identified 765 differentially methylated regions associated with 918 genes. Pathway analysis of these genes identified a biological network associated with cardiovascular disease, including a subset of genes that were differentially hypomethylated and whose expression levels were altered by SAM. Our data indicate that SAM modulates the expression of inflammatory genes in association with changes in specific gene promoter DNA methylation.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Inflamación/patología , Macrófagos/metabolismo , S-Adenosilmetionina/farmacología , Enfermedades Cardiovasculares/genética , Línea Celular , Metilación de ADN/genética , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Humanos , Interleucina-10/metabolismo , Macrófagos/efectos de los fármacos , S-Adenosilhomocisteína/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
5.
Mol Reprod Dev ; 81(1): 87-97, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24347442

RESUMEN

The goal of this study was to improve the development of bovine somatic-cell nuclear transfer (SCNT) embryos by optimizing the combination of DNA methyltransferases inhibitor S-adenosylhomocysteine (SAH) and histone deacetylase inhibitor Scriptaid (SPD). A. 4 × 4-factor design of different drug combinations (0, 0.75, 1.0, and 1.5 mM SAH and 0, 5, 250, and 500 nM SPD) was used to identify an optimal combination of 0.75 mM SAH and 250 nM SPD that improved the developmental competence of bovine SCNT embryos. Further experiments using this combination revealed that methylation levels of CpG islands near exon 1 of the pluripotent gene SOX2; the epigenetic-related gene HDAC3 and DNMT3a; imprinted genes XIST and PEG3; as well as apoptosis-related genes BCL2 and BAX were returned to levels similar to those of in vitro fertilized (IVF) embryo after treatment, which also normalized transcript levels for these genes. This combination also returned global DNA methylation to a normal level, correcting H4K12ac levels while enhancing H3K9ac levels. Thus, the combined application of 0.75 mM SAH and 250 nM SPD can significantly improve the reprogramming of bovine SCNT embryos by stabilizing how embryos utilize their genomes.


Asunto(s)
Cruzamiento/métodos , Bovinos/embriología , Embrión de Mamíferos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Hidroxilaminas/farmacología , Quinolinas/farmacología , S-Adenosilhomocisteína/farmacología , Animales , Secuencia de Bases , Islas de CpG/genética , Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Cartilla de ADN/genética , Combinación de Medicamentos , Embrión de Mamíferos/embriología , Técnica del Anticuerpo Fluorescente , Técnicas Histológicas , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Datos de Secuencia Molecular , Técnicas de Transferencia Nuclear , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción SOXB1/genética , Análisis de Secuencia de ADN
6.
Bioorg Med Chem ; 22(24): 6961-4, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25456389

RESUMEN

The naturally occurring adenine based carbocyclic nucleosides aristeromycin and neplanocin A and their 3-deaza analogues have found a prominent place in the search for diverse antiviral activity agent scaffolds because of their ability to inhibit S-adenosylhomocysteine (AdoHcy) hydrolase. Following the lead of these compounds, their 3-deaza-3-fluoroaristeromycin analogues have been synthesized and their effect on S-adenosylhomocysteine hydrolase and RNA and DNA viruses determined.


Asunto(s)
Antivirales/síntesis química , S-Adenosilhomocisteína/análogos & derivados , Adenosilhomocisteinasa/antagonistas & inhibidores , Adenosilhomocisteinasa/metabolismo , Animales , Antivirales/química , Antivirales/farmacología , Línea Celular , Chlorocebus aethiops , Virus ADN/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Células HEK293 , Humanos , Virus ARN/efectos de los fármacos , S-Adenosilhomocisteína/síntesis química , S-Adenosilhomocisteína/farmacología , Células Vero
7.
Bioorg Med Chem ; 21(7): 1787-1794, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23433670

RESUMEN

Chemical inhibition of proteins involved in chromatin-mediated signaling is an emerging strategy to control chromatin compaction with the aim to reprogram expression networks to alter disease states. Protein methyltransferases constitute one of the protein families that participate in epigenetic control of gene expression, and represent a novel therapeutic target class. Recruitment of the protein lysine methyltransferase DOT1L at aberrant loci is a frequent mechanism driving acute lymphoid and myeloid leukemias, particularly in infants, and pharmacological inhibition of DOT1L extends survival in a mouse model of mixed lineage leukemia. A better understanding of the structural chemistry of DOT1L inhibition would accelerate the development of improved compounds. Here, we report that the addition of a single halogen atom at a critical position in the cofactor product S-adenosylhomocysteine (SAH, an inhibitor of SAM-dependent methyltransferases) results in an 8-fold increase in potency against DOT1L, and reduced activities against other protein and non-protein methyltransferases. We solved the crystal structure of DOT1L in complex with Bromo-deaza-SAH and rationalized the observed effects. This discovery reveals a simple strategy to engineer selectivity and potency towards DOT1L into the adenosine scaffold of the cofactor shared by all methyltransferases, and can be exploited towards the development of clinical candidates against mixed lineage leukemia.


Asunto(s)
Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , S-Adenosilhomocisteína/análogos & derivados , S-Adenosilhomocisteína/farmacología , Cristalografía por Rayos X , Halogenación , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia/tratamiento farmacológico , Leucemia/enzimología , Metiltransferasas/química , Modelos Moleculares
8.
J Med Chem ; 65(14): 9750-9788, 2022 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-35849534

RESUMEN

Selective manipulation of the epitranscriptome could be beneficial for the treatment of cancer and also broaden the understanding of epigenetic inheritance. Inhibitors of the tRNA methyltransferase DNMT2, the enzyme catalyzing the S-adenosylmethionine-dependent methylation of cytidine 38 to 5-methylcytidine, were designed, synthesized, and analyzed for their enzyme-binding and -inhibiting properties. For rapid screening of potential DNMT2 binders, a microscale thermophoresis assay was established. Besides the natural inhibitors S-adenosyl-l-homocysteine (SAH) and sinefungin (SFG), we identified new synthetic inhibitors based on the structure of N-adenosyl-2,4-diaminobutyric acid (Dab). Structure-activity relationship studies revealed the amino acid side chain and a Y-shaped substitution pattern at the 4-position of Dab as crucial for DNMT2 inhibition. The most potent inhibitors are alkyne-substituted derivatives, exhibiting similar binding and inhibitory potencies as the natural compounds SAH and SFG. CaCo-2 assays revealed that poor membrane permeabilities of the acids and rapid hydrolysis of an ethylester prodrug might be the reasons for the insufficient activity in cellulo.


Asunto(s)
Metiltransferasas , Neoplasias , Proteínas Arqueales , Células CACO-2 , ADN , Humanos , Neoplasias/tratamiento farmacológico , S-Adenosilhomocisteína/química , S-Adenosilhomocisteína/metabolismo , S-Adenosilhomocisteína/farmacología , S-Adenosilmetionina/metabolismo
9.
Atherosclerosis ; 353: 1-10, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35753115

RESUMEN

BACKGROUND AND AIMS: It has been established that endothelial senescence plays a critical role in the development of atherosclerosis. Elevated S-adenosylhomocysteine (SAH) level induced by inhibition of S-adenosylhomocysteine hydrolase (SAHH) is one of the risk factors of atherosclerosis; however, the interplay between endothelial senescence and inhibition of SAHH is largely unknown. METHODS: Human umbilical vein endothelial cells (HUVECs) after serial passage were used. SAHH-specific inhibitor adenosine dialdehyde (ADA) and SAHH siRNA treated HUVECs and SAHH+/-mice were used to investigate the effect of SAHH inhibition on endothelial senescence. RESULTS: HUVECs exhibited distinct senescence morphology as HUVECs were passaged, together with a decrease in intracellular SAHH expression and an increase in intracellular SAH levels. SAHH inhibition by ADA or SAHH siRNA elevated SA ß-gal activity, arrested proliferation, and increased the expression of p16, p21 and p53 in HUVECs and the aortas of mice. In addition, decreased expression of hTERT and reduced occupancy of H3K4me3 over the hTERT promoter region were observed following SAHH inhibition treatment. To further verify the role of hTERT in the endothelial senescence induced by SAHH inhibition, hTERT was overexpressed with a plasmid vector under CMV promoter. hTERT overexpression rescued the senescence phenotypes in endothelial cells induced by SAHH inhibition. CONCLUSIONS: SAHH inhibition induces endothelial senescence via downregulation of hTERT expression, which is associated with attenuated histone methylation over the hTERT promoter region.


Asunto(s)
Aterosclerosis , S-Adenosilhomocisteína , Telomerasa/metabolismo , Adenosilhomocisteinasa/genética , Adenosilhomocisteinasa/metabolismo , Animales , Aterosclerosis/metabolismo , Senescencia Celular , Regulación hacia Abajo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , ARN Interferente Pequeño , S-Adenosilhomocisteína/metabolismo , S-Adenosilhomocisteína/farmacología
10.
Stem Cell Res Ther ; 13(1): 418, 2022 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-35964109

RESUMEN

BACKGROUND: Autologous stem cell therapy is a promising strategy for cardiovascular diseases including diabetic cardiomyopathy (DCM), but conclusions from clinical trials were compromised. We assumed that diabetes might induce the dysfunction of stem cells and thus limit its therapeutic effect. This study aimed to compare the effect of diabetes and nondiabetes-derived bone marrow mesenchymal stem cells (BMSCs) transplantation on DCM and explored the potential mechanism. METHODS: Rats with diabetes were induced using high-fat diets and streptozotocin (STZ) injection. BMSCs harvested from diabetic and nondiabetic rats were infused into DCM rats, and the effects on the heart were identified by echocardiography and histopathology. The inhibition or overexpression of SAHH in nondiabetic and diabetic BMSCs was used to confirm its key role in stem cell activity and cardiac therapy. RESULTS: Compared with normal BMSCs, the therapeutic effects of diabetic rat-derived stem cells on improving cardiac function and adverse remodeling were significantly attenuated. In vitro, diabetic BMSCs had lower cell viability and paracrine function than nondiabetic BMSCs. It was further found that diabetic BMSCs had obvious mitochondrial oxidative stress damage and S-adenosylhomocysteine (SAH) accumulation due to S-adenosylhomocysteine hydrolase (SAHH) deficiency. SAHH inhibition by adenosine dialdehyde (ADA) or shSAHH plasmid in normal BMSCs significantly reduced the favorable effects on endothelial cell proliferation and tube-forming capacity. In contrast, SAHH overexpression in diabetic BMSCs significantly improved cellular activity and paracrine function. Transplantation of BMSCs with SAHH overexpression improved cardiac adverse remodeling and angiogenesis. Activation of the Nrf2 signaling pathway may be one of the key mechanisms of SAHH-mediated improvement of stem cell viability and cardiac repair. CONCLUSIONS: Diabetes leads to compromised bioactivity and repair capacity of BMSCs. Our study suggests that SAHH activation may improve the cardioprotective effect of autologous transplantation of diabetes-derived BMSCs on patients with DCM. Diabetes induced the inhibition of S-adenosylhomocysteine (SAH) expression and aging phenotype in BMSCs and thus decreased the cell viability and paracrine function. Compared with normal BMSCs, the therapeutic effects of diabetic rat-derived BMSCs on improving cardiac function and adverse remodeling were significantly attenuated. SAHH overexpression in diabetic BMSCs significantly rescued cellular function partly via activating Nrf2/HO-1 signal. Transplantation of diabetic BMSCs with SAHH overexpression improved angiogenesis and cardiac adverse remodeling in rats.


Asunto(s)
Diabetes Mellitus Experimental , Cardiomiopatías Diabéticas , Células Madre Mesenquimatosas , Adenosilhomocisteinasa/metabolismo , Adenosilhomocisteinasa/farmacología , Animales , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/metabolismo , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/terapia , Células Madre Mesenquimatosas/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Ratas , S-Adenosilhomocisteína/metabolismo , S-Adenosilhomocisteína/farmacología
11.
RNA ; 15(4): 666-74, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19218551

RESUMEN

A 2,2,7-trimethylguanosine (TMG) cap is a signature feature of eukaryal snRNAs, telomerase RNAs, and trans-spliced nematode mRNAs. TMG and 2,7-dimethylguanosine (DMG) caps are also present on mRNAs of two species of alphaviruses (positive strand RNA viruses of the Togaviridae family). It is presently not known how viral mRNAs might acquire a hypermethylated cap. Mimivirus, a giant DNA virus that infects amoeba, encodes many putative enzymes and proteins implicated in RNA transactions, including the synthesis and capping of viral mRNAs and the promotion of cap-dependent translation. Here we report the identification, purification, and characterization of a mimivirus cap-specific guanine-N2 methyltransferase (MimiTgs), a monomeric enzyme that catalyzes a single round of methyl transfer from AdoMet to an m(7)G cap substrate to form a DMG cap product. MimiTgs, is apparently unable to convert a DMG cap to a TMG cap, and is thereby distinguished from the structurally homologous yeast and human Tgs1 enzymes. Nonetheless, we show genetically that MimiTgs is a true ortholog of Saccharomyces cerevisiae Tgs1. Our results hint that DMG caps can satisfy many of the functions of TMG caps in vivo. We speculate that DMG capping of mimivirus mRNAs might favor viral protein synthesis in the infected host.


Asunto(s)
Virus ADN/enzimología , Metiltransferasas/química , Adenosina/análogos & derivados , Adenosina/farmacología , Secuencia de Aminoácidos , Animales , Inhibidores Enzimáticos/farmacología , Guanosina/análogos & derivados , Guanosina/metabolismo , Humanos , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , Datos de Secuencia Molecular , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , S-Adenosilhomocisteína/farmacología , Alineación de Secuencia
12.
Mol Reprod Dev ; 78(8): 576-84, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21721066

RESUMEN

In this study, fibroblast cells were stably transfected with mouse POU5F1 promoter-driven enhanced green fluorescent protein (EGFP) to investigate the effect of S-adenosylhomocysteine (SAH), the reversible non-toxic inhibitor of DNA-methyltransferases (DNMTs), at different intervals post-fusion on in vitro development of cloned bovine embryos. Treatment with SAH for 12 hr resulted in 54.6 ± 7.7% blastocyst production, which was significantly greater than in vitro fertilized embryos (IVF: 37.2 ± 2.7%), cloned embryos treated with SAH for 72 hr (31.0 ± 7.6%), and control cloned embryos (34.6 ± 3.6%). The fluorescence intensities of the EGFP-POU5F1 reporter gene at all intervals of SAH treatment, except of 72 hr, were significantly higher than control somatic cell nuclear transfers (SCNT) embryos. The intensity of DNA-methylation in cloned embryos treated with SAH for 48 hr was similar to that of IVF embryos, and was significantly lower than the other SCNT groups. The levels of H3K9 acetylation in all SCNT groups were significantly lower than IVF embryos. Real-time PCR analysis of gene expression revealed significantly higher expression of POU5F1 in cloned versus IVF blastocysts. Neither embryo production method (SCNT vs. IVF) nor the SAH treatment interval affected expression of the BCL2 gene. Cloned embryos at all intervals of SAH treatment, except for 24 hr, had significantly increased VEGF transcript compared to IVF and control SCNT embryos. It was suggested that the time interval of DNMT inhibition may have important consequences on different in vitro features of bovine SCNT, and the improving effects of DNMT inhibition on developmental competency of cloned embryos are restricted to a specific period of time preceding de novo methylation.


Asunto(s)
Blastocisto/efectos de los fármacos , Clonación de Organismos/métodos , Embrión de Mamíferos/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , S-Adenosilhomocisteína/farmacología , Acetilación/efectos de los fármacos , Animales , Blastocisto/metabolismo , Bovinos , Metilación de ADN/efectos de los fármacos , Embrión de Mamíferos/fisiología , Epigénesis Genética , Femenino , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Masculino , Ratones , Microscopía Fluorescente , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Transfección
13.
Mol Cell Biochem ; 358(1-2): 229-39, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21739151

RESUMEN

We previously found that homocysteine (Hcy) induced plasma membrane flip-flop, apoptosis, and necrosis in cardiomyocytes. Inactivation of flippase by Hcy induced membrane flip-flop, while apoptosis was induced via a NOX2-dependent mechanism. It has been suggested that S-adenosylhomocysteine (SAH) is the main causative factor in hyperhomocysteinemia (HHC)-induced pathogenesis of cardiovascular disease. Therefore, we evaluated whether the observed cytotoxic effect of Hcy in cardiomyocytes is SAH dependent. Rat cardiomyoblasts (H9c2 cells) were treated under different conditions: (1) non-treated control (1.5 nM intracellular SAH with 2.8 µM extracellular L -Hcy), (2) incubation with 50 µM adenosine-2,3-dialdehyde (ADA resulting in 83.5 nM intracellular SAH, and 1.6 µM extracellular L -Hcy), (3) incubation with 2.5 mM D, L -Hcy (resulting in 68 nM intracellular SAH and 1513 µM extracellular L -Hcy) with or without 10 µM reactive oxygen species (ROS)-inhibitor apocynin, and (4) incubation with 100 nM, 10 µM, and 100 µM SAH. We then determined the effect on annexin V/propodium iodide positivity, flippase activity, caspase-3 activity, intracellular NOX2 and p47(phox) expression and localization, and nuclear ROS production. In contrast to Hcy, ADA did not induce apoptosis, necrosis, or membrane flip-flop. Remarkably, both ADA and Hcy induced a significant increase in nuclear NOX2 expression. However, in contrast to ADA, Hcy additionally induced nuclear p47(phox) expression, increased nuclear ROS production, and inactivated flippase. Incubation with SAH did not have an effect on cell viability, nor on flippase activity, nor on nuclear NOX2-, p47phox expression or nuclear ROS production. HHC-induced membrane flip-flop and apoptosis in cardiomyocytes is due to increased Hcy levels and not primarily related to increased intracellular SAH, which plays a crucial role in nuclear p47(phox) translocation and subsequent ROS production.


Asunto(s)
Apoptosis/efectos de los fármacos , Membrana Celular/metabolismo , Núcleo Celular/enzimología , Homocisteína/farmacología , Miocitos Cardíacos/citología , NADPH Oxidasas/metabolismo , S-Adenosilhomocisteína/farmacología , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina Trifosfato/metabolismo , Animales , Caspasa 3/metabolismo , Membrana Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Homocisteína/metabolismo , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Glicoproteínas de Membrana/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/enzimología , NADPH Oxidasa 2 , Proteínas de Transferencia de Fosfolípidos/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , S-Adenosilmetionina/metabolismo , Tirosina/análogos & derivados , Tirosina/metabolismo
14.
Sci Rep ; 11(1): 14693, 2021 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-34282217

RESUMEN

It has been previously shown that chronic ethanol administration-induced increase in adipose tissue lipolysis and reduction in the secretion of protective adipokines collectively contribute to alcohol-associated liver disease (ALD) pathogenesis. Further studies have revealed that increased adipose S-adenosylhomocysteine (SAH) levels generate methylation defects that promote lipolysis. Here, we hypothesized that increased intracellular SAH alone causes additional related pathological changes in adipose tissue as seen with alcohol administration. To test this, we used 3-deazaadenosine (DZA), which selectively elevates intracellular SAH levels by blocking its hydrolysis. Fully differentiated 3T3-L1 adipocytes were treated in vitro for 48 h with DZA and analysed for lipolysis, adipokine release and differentiation status. DZA treatment enhanced adipocyte lipolysis, as judged by lower levels of intracellular triglycerides, reduced lipid droplet sizes and higher levels of glycerol and free fatty acids released into the culture medium. These findings coincided with activation of both adipose triglyceride lipase and hormone sensitive lipase. DZA treatment also significantly reduced adipocyte differentiation factors, impaired adiponectin and leptin secretion but increased release of pro-inflammatory cytokines, IL-6, TNF and MCP-1. Together, our results demonstrate that elevation of intracellular SAH alone by DZA treatment of 3T3-L1 adipocytes induces lipolysis and dysregulates adipokine secretion. Selective elevation of intracellular SAH by DZA treatment mimics ethanol's effects and induces adipose dysfunction. We conclude that alcohol-induced elevations in adipose SAH levels contribute to the pathogenesis and progression of ALD.


Asunto(s)
Adipocitos/efectos de los fármacos , Hepatopatías Alcohólicas/metabolismo , S-Adenosilhomocisteína/farmacología , Células 3T3-L1 , Adipocitos/metabolismo , Adipocitos/fisiología , Tejido Adiposo/metabolismo , Animales , Etanol/farmacología , Hígado Graso Alcohólico/metabolismo , Hígado Graso Alcohólico/patología , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Hepatopatías Alcohólicas/patología , Ratones , S-Adenosilhomocisteína/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
J Biol Chem ; 284(46): 31962-71, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19762471

RESUMEN

In plants the triple methylation of phosphoethanolamine to phosphocholine catalyzed by phosphoethanolamine N-methyltransferase (PEAMT) is considered a rate-limiting step in the de novo synthesis of phosphatidylcholine. Besides being a major membrane phospholipid, phosphatidylcholine can be hydrolyzed into choline and phosphatidic acid. Phosphatidic acid is widely recognized as a second messenger in stress signaling, and choline can be oxidized within the chloroplast to yield the putative osmoprotectant glycine betaine. Here we describe the cloning and biochemical characterization of a second wheat PEAMT isoform that has a four times higher specific activity than the previously described WPEAMT/TaPEAMT1 enzyme and is less sensitive to product inhibition by S-adenosyl homocysteine, but more sensitive to inhibition by phosphocholine. Both enzymes follow a sequential random Bi Bi mechanism and show mixed-type product inhibition patterns with partial inhibition for TaPEAMT1 and a strong non-competitive component for TaPEAMT2. An induction of TaPEAMT protein expression and activity is observed after cold exposure, ahead of an increase in gene expression. Our results demonstrate direct repression of in vitro enzymatic activities by phosphatidic acid for both enzymes, with TaPEAMT1 being more sensitive than TaPEAMT2 in the physiological concentration range. Other lipid ligands identified in protein-lipid overlays are phosphoinositide mono- as well as some di-phosphates and cardiolipin. These results provide new insights into the complex regulatory circuits of phospholipid biosynthesis in plants and underline the importance of head group biosynthesis in adaptive stress responses.


Asunto(s)
Ácidos Fosfatidicos/farmacología , Fosfatidiletanolamina N-Metiltransferasa/antagonistas & inhibidores , Fosfatidiletanolamina N-Metiltransferasa/metabolismo , S-Adenosilhomocisteína/farmacología , Triticum/enzimología , Aclimatación , Animales , Western Blotting , Clonación Molecular , Frío , Inmunoglobulina G/inmunología , Isoenzimas , Fosfatidiletanolamina N-Metiltransferasa/genética , Fosfatidilinositoles/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Conejos , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Biochem Biophys Res Commun ; 391(2): 1291-6, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20026058

RESUMEN

The proteasome is a multi-catalytic protein degradation enzyme that is regulated by ethanol-induced oxidative stress; such suppression is attributed to CYP2E1-generated metabolites. However, under certain conditions, it appears that in addition to oxidative stress, other mechanisms are also involved in proteasome regulation. This study investigated whether impaired protein methylation that occurs during exposure of liver cells to ethanol, may contribute to suppression of proteasome activity. We measured the chymotrypsin-like proteasome activity in Huh7CYP cells, hepatocytes, liver cytosols and nuclear extracts or purified 20S proteasome under conditions that maintain or prevent protein methylation. Reduction of proteasome activity of hepatoma cell and hepatocytes by ethanol or tubercidin was prevented by simultaneous treatment with S-adenosylmethionine (SAM). Moreover, the tubercidin-induced decline in proteasome activity occurred in both nuclear and cytosolic fractions. In vitro exposure of cell cytosolic fractions or highly purified 20S proteasome to low SAM:S-adenosylhomocysteine (SAH) ratios in the buffer also suppressed proteasome function, indicating that one or more methyltransferase(s) may be associated with proteasomal subunits. Immunoblotting a purified 20S rabbit red cell proteasome preparation using methyl lysine-specific antibodies revealed a 25kDa proteasome subunit that showed positive reactivity with anti-methyl lysine. This reactivity was modified when 20S proteasome was exposed to differential SAM:SAH ratios. We conclude that impaired methylation of proteasome subunits suppressed proteasome activity in liver cells indicating an additional, yet novel mechanism of proteasome activity regulation by ethanol.


Asunto(s)
Hígado/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Animales , Antimetabolitos Antineoplásicos/farmacología , Línea Celular Tumoral , Quimotripsina/metabolismo , Etanol/farmacología , Humanos , Hígado/efectos de los fármacos , Metilación , Ratones , Ratones Endogámicos C57BL , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Conejos , S-Adenosilhomocisteína/metabolismo , S-Adenosilhomocisteína/farmacología , S-Adenosilmetionina/metabolismo , S-Adenosilmetionina/farmacología , Tubercidina/farmacología
17.
DNA Cell Biol ; 39(1): 37-49, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31750745

RESUMEN

Cloning using somatic cell nuclear transfer (SCNT) has many potential applications such as in transgenic and genomic-edited animal production. Abnormal epigenetic reprogramming of somatic cell nuclei is probably the major cause of the low efficiency associated with SCNT. Strategies to alter DNA reprogramming in donor cell nuclei may help improve the cloning efficiency. In the present study, we aimed to characterize the effects of procaine and S-adenosyl-l-homocysteine (SAH) as demethylating agents during the cell culture of bovine skin fibroblasts. We characterized the effects of procaine and SAH on the expression of genes related to the epigenetic machinery, including the DNA methyltransferase 1 (DNMT1), DNA methyltransferase 3 alpha (DNMT3A), DNA methyltransferase 3 beta (DNMT3B), TET1, TET2, TET3, and OCT4 genes, and on DNA methylation levels of bovine skin fibroblasts. We found that DNA methylation levels of satellite I were reduced by SAH (p = 0.0495) and by the combination of SAH and procaine (p = 0.0479) compared with that in the control group. Global DNA methylation levels were lower in cells that were cultivated with both compounds than in control cells (procaine [p = 0.0116], SAH [p = 0.0408], and both [p = 0.0163]). Regarding gene expression, there was a decrease in the DNMT1 transcript levels in cells cultivated with SAH (p = 0.0151) and SAH/procaine (0.0001); a decrease in the DNMT3A transcript levels in cells cultivated with SAH/procaine (p = 0.016); and finally, a decrease in the DNMT3B transcript levels in cells cultivated with procaine (p = 0.0007), SAH (p = 0.0060), and SAH/procaine (p = 0.0021) was found. Higher levels of TET3 transcripts in cells cultivated with procaine (p = 0.0291), SAH (p = 0.0373), and procaine/SAH (p = 0.0013) compared with the control were also found. Regarding the OCT4 gene, no differences were found. Our results showed that the use of procaine and SAH during bovine cell culture was able to alter the epigenetic profile of the cells. This approach may be a useful alternative strategy to improve the efficiency of reprogramming the somatic nuclei after fusion, which in turn will improve the SCNT efficiency.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Procaína/farmacología , S-Adenosilhomocisteína/farmacología , Animales , Bovinos , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasas/genética , Dioxigenasas/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Proto-Oncogénicas/genética , Piel/citología
18.
SLAS Discov ; 25(4): 361-371, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31585521

RESUMEN

A variety of covalent modifications of RNA have been identified and demonstrated to affect RNA processing, stability, and translation. Methylation of adenosine at the N6 position (m6A) in messenger RNA (mRNA) is currently the most well-studied RNA modification and is catalyzed by the RNA methyltransferase complex METTL3/METTL14. Once generated, m6A can modulate mRNA splicing, export, localization, degradation, and translation. Although potent and selective inhibitors exist for several members of the Type I S-adenosylmethionine (SAM)-dependent methyltransferase family, no inhibitors have been reported for METTL3/METTL14 to date. To facilitate drug discovery efforts, a sensitive and robust mass spectrometry-based assay for METTL3/METTL14 using self-assembled monolayer desorption/ionization (SAMDI) technology has been developed. The assay uses an 11-nucleotide single-stranded RNA compared to a previously reported 27-nucleotide substrate. IC50 values of mechanism-based inhibitors S-adenosylhomocysteine (SAH) and sinefungin (SFG) are comparable between the SAMDI and radiometric assays that use the same substrate. This work demonstrates that SAMDI technology is amenable to RNA substrates and can be used for high-throughput screening and compound characterization for RNA-modifying enzymes.


Asunto(s)
Espectrometría de Masas/métodos , Metiltransferasas/genética , Procesamiento Postranscripcional del ARN/efectos de los fármacos , Adenosina/análogos & derivados , Adenosina/genética , Adenosina/farmacología , Descubrimiento de Drogas/tendencias , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Humanos , Metilación/efectos de los fármacos , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/genética , Procesamiento Postranscripcional del ARN/genética , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , ARN Mensajero/efectos de los fármacos , ARN Mensajero/genética , S-Adenosilhomocisteína/farmacología
19.
J Virol ; 82(24): 12280-90, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18829753

RESUMEN

There are many unique aspects of vesicular stomatitis virus (VSV) transcription. In addition to its unusual mRNA capping and methyltransferase mechanisms, the addition of S-adenosyl homocysteine (SAH), which is the by-product and competitive inhibitor of S-adenosyl methionine (SAM)-mediated methyltransferase reactions, leads to synthesis of poly(A) tails on the 3' end of VSV mRNAs that are 10- or 20-fold longer than normal. The mechanism by which this occurs is not understood, since it has been shown that productive transcription is not dependent on 5' cap methylation and full-length VSV mRNAs can be synthesized in the absence of SAM. To investigate this unusual phenotype, we assayed the effects of SAH on transcription using a panel of recombinant viruses that contained mutations in domain VI of the VSV L protein. The L proteins we investigated displayed a range of 5' cap methyltransferase activities. In the present study, we show that the ability of the VSV L protein to catalyze methyl transfer correlates with its sensitivity to SAH with respect to polyadenylation, thereby indicating an intriguing connection between 5' and 3' end mRNA modifications. We also identified an L protein mutant that hyperpolyadenylates mRNA irrespective of the presence or absence of exogenous SAH. Further, the data presented here show that the wild-type L protein hyperpolyadenylates a percentage of VSV mRNAs in infected cells as well as in vitro.


Asunto(s)
Adenosina/genética , Adenosina/metabolismo , Polímeros/metabolismo , Proteína Metiltransferasas/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , S-Adenosilhomocisteína/farmacología , Vesiculovirus/efectos de los fármacos , Vesiculovirus/metabolismo , Proteínas Virales/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Metilación , Modelos Moleculares , Estructura Terciaria de Proteína , ARN Mensajero/genética , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Especificidad por Sustrato , Transcripción Genética/genética , Vesiculovirus/genética , Proteínas Virales/química , Proteínas Virales/genética
20.
Science ; 225(4663): 745-6, 1984 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-6463652

RESUMEN

When isolated apical membrane vesicles prepared from cultured A6 epithelia were incubated in vitro with the methyl donor S-adenosylmethionine, the control rate of amiloride-inhibitable sodium transport was doubled. The methylation inhibitors 3-deazaadenosine and S-adenosyl homocysteine returned the S-adenosyl-methionine-stimulated sodium transport to control levels. Neither these agents nor adenosine affected sodium transport into control vesicles. In vesicles incubated with S-adenosyl-[3H-methyl]methionine, both membrane phospholipids and proteins were labeled, and this labeling was inhibited by deazaadenosine. In vesicles prepared from A6 cells treated with aldosterone, sodium transport was twice the control value and S-adenosylmethionine did not cause any further stimulation of transport. In those vesicles, both lipid and protein methylation were increased. These results suggest that methylation, which increases the rate of amiloride-sensitive sodium transport is involved in the action of aldosterone at the apical membrane level in epithelia.


Asunto(s)
Aldosterona/fisiología , Membrana Celular/metabolismo , Sodio/metabolismo , Amilorida/farmacología , Anfibios , Animales , Transporte Biológico Activo/efectos de los fármacos , Línea Celular , Membrana Celular/efectos de los fármacos , Riñón/metabolismo , Metilación , S-Adenosilhomocisteína/farmacología , S-Adenosilmetionina/farmacología , Tubercidina/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda