Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
J Am Chem Soc ; 145(49): 26947-26961, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38050996

ABSTRACT

The evolutionarily conserved bacterial proteins MnmE and MnmG (and their homologues in Eukarya) install a 5-carboxymethylaminomethyl (cmnm5) or a 5-taurinomethyl (τm5) group onto wobble uridines of several tRNA species. The Escherichia coli MnmE binds guanosine-5'-triphosphate (GTP) and methylenetetrahydrofolate (CH2THF), while MnmG binds flavin adenine dinucleotide (FAD) and a reduced nicotinamide adenine dinucleotide (NADH). Together with glycine, MnmEG catalyzes the installation of cmnm5 in a reaction that also requires hydrolysis of GTP. In this letter, we investigated key steps of the MnmEG reaction using a combination of biochemical techniques. We show multiple lines of evidence supporting flavin-iminium FADH[N5═CH2]+ as a central intermediate in the MnmEG reaction. Using a synthetic FADH[N5═CD2]+ analogue, the intermediacy of the FAD in the transfer of the methylene group from CH2THF to the C5 position of U34 was unambiguously demonstrated. Further, MnmEG reactions containing the deuterated flavin-iminium intermediate and alternate nucleophiles such as taurine and ammonia also led to the formation of the anticipated U34-modified tRNAs, showing FAD[N5═CH2]+ as the universal intermediate for all MnmEG homologues. Additionally, an RNA-protein complex stable to urea-denaturing polyacrylamide gel electrophoresis was identified. Studies involving a series of nuclease (RNase T1) and protease (trypsin) digestions along with reverse transcription experiments suggest that the complex may be noncovalent. While the conserved MnmG cysteine C47 and C277 mutant variants were shown to reduce FAD, they were unable to promote the modified tRNA formation. Overall, this study provides critical insights into the biochemical mechanism underlying tRNA modification by the MnmEG.


Subject(s)
Escherichia coli Proteins , Escherichia coli Proteins/chemistry , Uridine/metabolism , GTP Phosphohydrolases/metabolism , Flavin-Adenine Dinucleotide/metabolism , Escherichia coli/metabolism , Guanosine Triphosphate/metabolism , RNA, Transfer/chemistry , One-Carbon Group Transferases/chemistry , One-Carbon Group Transferases/metabolism
2.
J Biol Chem ; 298(11): 102548, 2022 11.
Article in English | MEDLINE | ID: mdl-36181794

ABSTRACT

The evolutionarily conserved bacterial proteins MnmE and MnmG collectively install a carboxymethylaminomethyl (cmnm) group at the fifth position of wobble uridines of several tRNA species. While the reaction catalyzed by MnmEG is one of the central steps in the biosynthesis of the methylaminomethyl (mnm) posttranscriptional tRNA modification, details of the reaction remain elusive. Glycine is known to be the source of the carboxy methylamino moiety of cmnm, and a tetrahydrofolate (THF) analog is thought to supply the one carbon that is appended to the fifth position of U. However, the nature of the folate analog remains unknown. This article reports the in vitro biochemical reconstitution of the MnmEG reaction. Using isotopically labeled methyl and methylene THF analogs, we demonstrate that methylene THF is the true substrate. We also show that reduced FAD is required for the reaction and that DTT can replace the NADH in its role as a reductant. We discuss the implications of these methylene-THF and reductant requirements on the mechanism of this key tRNA modification catalyzed by MnmEG.


Subject(s)
Escherichia coli Proteins , One-Carbon Group Transferases , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , Uridine , Escherichia coli Proteins/metabolism , Reducing Agents , RNA, Transfer/metabolism
3.
Biomolecules ; 11(10)2021 10 19.
Article in English | MEDLINE | ID: mdl-34680179

ABSTRACT

Disturbances in the one-carbon metabolism are often indicated by altered levels of the endogenous amino acid homocysteine (HCys), which is additionally discussed to causally contribute to diverse pathologies. In the first part of the present review, we profoundly and critically discuss the metabolic role and pathomechanisms of HCys, as well as its potential impact on different human disorders. The use of adequate animal models can aid in unravelling the complex pathological processes underlying the role of hyperhomocysteinemia (HHCys). Therefore, in the second part, we systematically searched PubMed/Medline for animal studies regarding HHCys and focused on the potential impact on cognitive performance and decline. The majority of reviewed studies reported a significant effect of HHCys on the investigated behavioral outcomes. Despite of persistent controversial discussions about equivocal findings, especially in clinical studies, the present evaluation of preclinical evidence indicates a causal link between HHCys and cognition-related- especially dementia-like disorders, and points out the further urge for large-scale, well-designed clinical studies in order to elucidate the normalization of HCys levels as a potential preventative or therapeutic approach in human pathologies.


Subject(s)
Cognitive Dysfunction/physiopathology , Homocysteine/metabolism , Hyperhomocysteinemia/physiopathology , One-Carbon Group Transferases/metabolism , Animals , Cognition/physiology , Cognitive Dysfunction/complications , Cognitive Dysfunction/epidemiology , Humans , Hyperhomocysteinemia/complications , Hyperhomocysteinemia/epidemiology , One-Carbon Group Transferases/genetics
4.
Clin Epigenetics ; 13(1): 182, 2021 09 26.
Article in English | MEDLINE | ID: mdl-34565452

ABSTRACT

BACKGROUND: Stunting is a condition in which a child does not reach their full growth potential due to chronic undernutrition. It arises during the first 2 years of a child's life and is associated with developmental deficiencies and life-long health problems. Current interventions provide some benefit, but new approaches to prevention and treatment grounded in a molecular understanding of stunting are needed. Epigenetic analyses are critical as they can provide insight into how signals from a poor environment lead to changes in cell function. RESULTS: Here we profiled histone H3 acetylation on lysine 27 (H3K27ac) in peripheral blood mononuclear cells (PBMCs) of 18-week-old (n = 14) and 1-year-old children (n = 22) living in an urban slum in Dhaka, Bangladesh. We show that 18-week-old children destined to become stunted have elevated levels of H3K27ac overall, functional analysis of which indicates activation of the immune system and stress response pathways as a primary response to a poor environment with high pathogen load. Conversely, overt stunting at 1-year-of age is associated with globally reduced H3K27ac that is indicative of metabolic rewiring and downregulation of the immune system and DNA repair pathways that are likely secondary responses to chronic exposure to a poor environment with limited nutrients. Among processes altered in 1-year-old children, we identified one-carbon metabolism, the significance of which is supported by integrative analysis with results from histone H3 trimethylation on lysine 4 (H3K4me3). Together, these results suggest altered one-carbon metabolism in this population of stunted children. CONCLUSIONS: The epigenomes of stunted children undergo two global changes in H3K27ac within their first year of life, which are associated with probable initial hyperactive immune responses followed by reduced metabolic capacity. Limitation of one-carbon metabolites may play a key role in the development of stunting. Trial registration ClinicalTrials.gov NCT01375647. Registered 17 June 2011, retrospectively registered, https://clinicaltrials.gov/ct2/show/NCT01375647 .


Subject(s)
Histones/analysis , Lysine/analysis , Malnutrition/blood , Acetylation/drug effects , Child, Preschool , Female , Histones/metabolism , Humans , Infant , Lysine/metabolism , Male , Malnutrition/physiopathology , One-Carbon Group Transferases/metabolism
5.
Rocz Panstw Zakl Hig ; 72(2): 123-136, 2021.
Article in English | MEDLINE | ID: mdl-34114759

ABSTRACT

DNA methylation is a reversible epigenetic modification that plays a crucial role in transcriptional gene silencing. Both excessive (hypermethylation) and reduced DNA methylation (hypomethylation) can contribute to the disturbance of the proper course of many important processes in the human body. The aim of the study was to discuss the relationship between methyl nutrients and the DNA methylation process in the course of selected diseases in adults. Methyl nutrients include folates (vitamin B9), riboflavin (vitamin B2), cobalamin (vitamin B12), pyridoxine (vitamin B6) and choline (vitamin B4), as well as methionine and betaine. These substances play the role of both substrates and cofactors in transformations related to one-carbon metabolism. The deficiency of methyl nutrients in the body can lead to disturbances in SAM synthesis, which is the primary donor of methyl groups in the DNA methylation process. However, the mechanism explaining the discussed relationship has not been fully explained so far. Both the concentration in the body and the intake of folate and vitamin B12 in the diet can, to some extent, have an effect on the level of DNA methylation in healthy people. In comparison, data on the effect of excessive intake of vitamin B12 in the diet on the risk of cancer development are inconsistent. An adequate betaine and choline intake in the diet might not only affect the overall improvement of the DNA methylation profile, but, to some extent, also reduce the risk of cancer, the effect of which can depend on the content of folic acid in the body. Research results on the effect of supplementation of methyl nutrients on the DNA methylation process are inconclusive. It is therefore necessary to conduct further research in this area to draw clear conclusions.


Subject(s)
Carbon/metabolism , DNA Methylation , Folic Acid/metabolism , One-Carbon Group Transferases/metabolism , Vitamin B 12/metabolism , Vitamin B 6/metabolism , Adult , Diet , Epigenesis, Genetic , Humans , Nutrients
6.
J Nutr ; 150(11): 2874-2881, 2020 11 19.
Article in English | MEDLINE | ID: mdl-32939549

ABSTRACT

BACKGROUND: Choline plays an integral role in one-carbon metabolism in the body, but it is unclear whether genetic polymorphisms are associated with variations in plasma choline and its metabolites. OBJECTIVES: This study aimed to evaluate the association of genetic variants in choline and one-carbon metabolism with plasma choline and its metabolites. METHODS: We analyzed data from 1423 postmenopausal women in a case-control study nested within the Women's Health Initiative Observational Study. Plasma concentrations of choline, betaine, dimethylglycine (DMG), and trimethylamine N-oxide were determined in 12-h fasting blood samples collected at baseline (1993-1998). Candidate and tagging single-nucleotide polymorphisms (SNPs) were genotyped in betaine-homocysteine S-methyltransferase (BHMT), BHMT2, 5,10-methylenetetrahydrofolate reductase (MTHFR), methylenetetrahydrofolate dehydrogenase (NADP+ dependent 1) (MTHFD1), 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR), and 5-methyltetrahydrofolate-homocysteine methyltransferase reductase (MTRR). Linear regression was used to derive percentage difference in plasma concentrations per variant allele, adjusting for confounders, including B-vitamin biomarkers. Potential effect modification by plasma vitamin B-12, vitamin B-6, and folate concentrations and folic-acid fortification periods was examined. RESULTS: The candidate SNP BHMT R239Q (rs3733890) was associated with lower concentrations of plasma betaine and DMG concentrations (-4.00% and -6.75% per variant allele, respectively; both nominal P < 0.05). Another candidate SNP, BHMT2 rs626105 A>G, was associated with higher plasma DMG concentration (13.0%; P < 0.0001). Several tagSNPs in these 2 genes were associated with plasma concentrations after correction for multiple comparisons. Vitamin B-12 status was a significant effect modifier of the association between the genetic variant BHMT2 rs626105 A>G and plasma DMG concentration. CONCLUSIONS: Genetic variations in metabolic enzymes were associated with plasma concentrations of choline and its metabolites. Our findings contribute to the knowledge on the variation in blood nutrient concentrations in postmenopausal women.


Subject(s)
Choline/metabolism , Gene Expression Regulation, Enzymologic/physiology , One-Carbon Group Transferases/metabolism , Oxidoreductases/metabolism , Polymorphism, Single Nucleotide , Postmenopause , Aged , Biomarkers , Case-Control Studies , Choline/blood , Colorectal Neoplasms , Female , Genetic Variation , Humans , Middle Aged , One-Carbon Group Transferases/genetics , Oxidoreductases/genetics , Risk Factors
7.
Nutrients ; 12(9)2020 Sep 19.
Article in English | MEDLINE | ID: mdl-32961717

ABSTRACT

Vitamins B9 (folate) and B12 are essential water-soluble vitamins that play a crucial role in the maintenance of one-carbon metabolism: a set of interconnected biochemical pathways driven by folate and methionine to generate methyl groups for use in DNA synthesis, amino acid homeostasis, antioxidant generation, and epigenetic regulation. Dietary deficiencies in B9 and B12, or genetic polymorphisms that influence the activity of enzymes involved in the folate or methionine cycles, are known to cause developmental defects, impair cognitive function, or block normal blood production. Nutritional deficiencies have historically been treated with dietary supplementation or high-dose parenteral administration that can reverse symptoms in the majority of cases. Elevated levels of these vitamins have more recently been shown to correlate with immune dysfunction, cancer, and increased mortality. Therapies that specifically target one-carbon metabolism are therefore currently being explored for the treatment of immune disorders and cancer. In this review, we will highlight recent studies aimed at elucidating the role of folate, B12, and methionine in one-carbon metabolism during normal cellular processes and in the context of disease progression.


Subject(s)
Folic Acid Deficiency/prevention & control , Folic Acid/pharmacology , One-Carbon Group Transferases/metabolism , Vitamin B 12 Deficiency/prevention & control , Vitamin B 12/pharmacology , Folic Acid Deficiency/genetics , Humans , Polymorphism, Genetic , Vitamin B 12 Deficiency/genetics
8.
Nutr Res Rev ; 32(2): 218-230, 2019 12.
Article in English | MEDLINE | ID: mdl-31303188

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder. It is characterised by the progressive degeneration of dopaminergic (DA) neurons. The cause of degeneration is not well understood; however, both genetics and environmental factors, such as nutrition, have been implicated in the disease process. Deficiencies in one-carbon metabolism in particular have been associated with increased risk for PD onset and progression, though the precise relationship is unclear. The aim of the present review is to determine the role of one-carbon metabolism and elevated levels of homocysteine in PD onset and pathology and to identify potential mechanisms involved. A search of PubMed, Google Scholar and Web of Science was undertaken to identify relevant human and animal studies. Case-control, prospective cohort studies, meta-analyses and non-randomised trials were included in the present review. The results from human studies indicate that polymorphisms in one-carbon metabolism may increase risk for PD development. There is an unclear role for dietary B-vitamin intake on PD onset and progression. However, dietary supplementation with B-vitamins may be beneficial for PD-affected individuals, particularly those on l-DOPA (levodopa or l-3,4-dihydroxyphenylalanine) treatment. Additionally, one-carbon metabolism generates methyl groups, and methylation capacity in PD-affected individuals is reduced. This reduced capacity has an impact on expression of disease-specific genes that may be involved in PD progression. During B-vitamin deficiency, animal studies report increased vulnerability of DA cells through increased oxidative stress and altered methylation. Nutrition, especially folates and related B-vitamins, may contribute to the onset and progression of PD by making the brain more vulnerable to damage; however, further investigation is required.


Subject(s)
Homocysteine/metabolism , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , Parkinson Disease/etiology , Animals , Diet , Folic Acid/metabolism , Genetic Predisposition to Disease , Humans , Levodopa/therapeutic use , Methylation , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Nutritional Status , Parkinson Disease/genetics , Parkinson Disease/therapy , Polymorphism, Genetic , Vitamin B Complex/administration & dosage
9.
RNA Biol ; 15(9): 1167-1173, 2018.
Article in English | MEDLINE | ID: mdl-30249152

ABSTRACT

The MnmE-MnmG complex of Escherichia coli uses either ammonium or glycine as a substrate to incorporate the 5-aminomethyl or 5-carboxymethylaminomethyl group into the wobble uridine of certain tRNAs. Both modifications can be converted into a 5-methylaminomethyl group by the independent oxidoreductase and methyltransferase activities of MnmC, which respectively reside in the MnmC(o) and MnmC(m) domains of this bifunctional enzyme. MnmE and MnmG, but not MnmC, are evolutionarily conserved. Bacillus subtilis lacks genes encoding MnmC(o) and/or MnmC(m) homologs. The glycine pathway has been considered predominant in this typical gram-positive species because only the 5-carboxymethylaminomethyl group has been detected in tRNALysUUU and bulk tRNA to date. Here, we show that the 5-methylaminomethyl modification is prevalent in B. subtilis tRNAGlnUUG and tRNAGluUUC. Our data indicate that B. subtilis has evolved MnmC(o)- and MnmC(m)-like activities that reside in non MnmC homologous protein(s), which suggests that both activities provide some sort of biological advantage.


Subject(s)
RNA, Transfer, Gln/metabolism , RNA, Transfer, Glu/metabolism , Uridine/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Escherichia coli Proteins/metabolism , Multienzyme Complexes/metabolism , Mutation , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , RNA Processing, Post-Transcriptional
10.
J Mol Biol ; 430(17): 2822-2842, 2018 08 17.
Article in English | MEDLINE | ID: mdl-29870725

ABSTRACT

The Escherichia coli homodimeric proteins MnmE and MnmG form a functional complex, MnmEG, that modifies tRNAs using GTP, methylene-tetrahydrofolate, FAD, and glycine or ammonium. MnmE is a tetrahydrofolate- and GTP-binding protein, whereas MnmG is a FAD-binding protein with each protomer composed of the FAD-binding domain, two insertion domains, and the helical C-terminal domain. The detailed mechanism of the MnmEG-mediated reaction remains unclear partially due to incomplete structural information on the free- and substrate-bound forms of the complex. In this study, we show that MnmG can adopt in solution a dimer arrangement (form I) different from that currently considered as the only biologically active (form II). Normal mode analysis indicates that form I can oscillate in a range of open and closed conformations. Using isothermal titration calorimetry and native red electrophoresis, we show that a form-I open conformation, which can be stabilized in vitro by the formation of an interprotomer disulfide bond between the catalytic C277 residues, appears to be involved in the assembly of the MnmEG catalytic center. We also show that residues R196, D253, R436, R554 and E585 are important for the stabilization of form I and the tRNA modification function. We propose that the form I dynamics regulates the alternative access of MnmE and tRNA to the MnmG FAD active site. Finally, we show that the C-terminal region of MnmG contains a sterile alpha motif domain responsible for tRNA-protein and protein-protein interactions.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , One-Carbon Group Transferases/chemistry , One-Carbon Group Transferases/metabolism , Protein Multimerization , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Catalytic Domain , GTP Phosphohydrolases/chemistry , GTP Phosphohydrolases/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs
11.
J Am Coll Cardiol ; 71(6): 654-667, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29420962

ABSTRACT

BACKGROUND: In response to pressure overload, the heart develops ventricular hypertrophy that progressively decompensates and leads to heart failure. This pathological hypertrophy is mediated, among others, by the phosphatase calcineurin and is characterized by metabolic changes that impair energy production by mitochondria. OBJECTIVES: The authors aimed to determine the role of the calcineurin splicing variant CnAß1 in the context of cardiac hypertrophy and its mechanism of action. METHODS: Transgenic mice overexpressing CnAß1 specifically in cardiomyocytes and mice lacking the unique C-terminal domain in CnAß1 (CnAß1Δi12 mice) were used. Pressure overload hypertrophy was induced by transaortic constriction. Cardiac function was measured by echocardiography. Mice were characterized using various molecular analyses. RESULTS: In contrast to other calcineurin isoforms, the authors show here that cardiac-specific overexpression of CnAß1 in transgenic mice reduces cardiac hypertrophy and improves cardiac function. This effect is mediated by activation of serine and one-carbon metabolism, and the production of antioxidant mediators that prevent mitochondrial protein oxidation and preserve ATP production. The induction of enzymes involved in this metabolic pathway by CnAß1 is dependent on mTOR activity. Inhibition of serine and one-carbon metabolism blocks the beneficial effects of CnAß1. CnAß1Δi12 mice show increased cardiac hypertrophy and declined contractility. CONCLUSIONS: The metabolic reprogramming induced by CnAß1 redefines the role of calcineurin in the heart and shows for the first time that activation of the serine and one-carbon pathway has beneficial effects on cardiac hypertrophy and function, paving the way for new therapeutic approaches.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/metabolism , One-Carbon Group Transferases/metabolism , Serine/metabolism , Ventricular Function/drug effects , Animals , Calcineurin/pharmacology , Calcineurin/therapeutic use , Cardiomegaly/drug therapy , Humans , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Ventricular Function/physiology
12.
Mol Psychiatry ; 23(2): 282-294, 2018 02.
Article in English | MEDLINE | ID: mdl-28809400

ABSTRACT

The methionine-folate cycle-dependent one-carbon metabolism is implicated in the pathophysiology of schizophrenia. Since schizophrenia is a developmental disorder, we examined the effects that perturbation of the one-carbon metabolism during gestation has on mice progeny. Pregnant mice were administered methionine equivalent to double their daily intake during the last week of gestation. Their progeny (MET mice) exhibited schizophrenia-like social deficits, cognitive impairments and elevated stereotypy, decreased neurogenesis and synaptic plasticity, and abnormally reduced local excitatory synaptic connections in CA1 neurons. Neural transcript expression of only one gene, encoding the Npas4 transcription factor, was >twofold altered (downregulated) in MET mice; strikingly, similar Npas4 downregulation occurred in the prefrontal cortex of human patients with schizophrenia. Finally, therapeutic actions of typical (haloperidol) and atypical (clozapine) antipsychotics in MET mice mimicked effects in human schizophrenia patients. Our data support the validity of MET mice as a model for schizophrenia, and uncover methionine metabolism as a potential preventive and/or therapeutic target.


Subject(s)
Methionine/metabolism , Schizophrenia/metabolism , Animals , Antipsychotic Agents/therapeutic use , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , CA1 Region, Hippocampal/drug effects , Clozapine/therapeutic use , Developmental Disabilities/physiopathology , Disease Models, Animal , Female , Folic Acid/metabolism , Haloperidol/therapeutic use , Humans , Male , Mice , Neurogenesis , Neuronal Plasticity , One-Carbon Group Transferases/metabolism , Prefrontal Cortex/embryology , Pregnancy , Prenatal Exposure Delayed Effects , Stereotyped Behavior/drug effects , Tetrahydrofolates
13.
Alcohol Clin Exp Res ; 41(6): 1105-1111, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28369960

ABSTRACT

BACKGROUND: Epigenetic mechanisms such as DNA methylation play an important role in regulating the pathophysiology of alcoholism. Chronic alcohol exposure leads to behavioral changes as well as decreased expression of genes associated with synaptic plasticity. In the liver, it has been documented that chronic alcohol exposure impairs methionine synthase (Ms) activity leading to a decrease in S-adenosyl methionine/S-adenosyl homocysteine (SAM/SAH) ratio which results in DNA hypomethylation; however, it is not known whether similar alterations of SAM and SAH levels are also produced in brain. METHODS: Male adult Sprague Dawley rats were fed chronically with Lieber-DeCarli ethanol (EtOH) (9% v/v) or control diet. The EtOH-diet-fed rats were withdrawn for 0 and 24 hours. The cerebellum and liver tissues were dissected and used to investigate changes in one-carbon metabolism, SAM, and SAH levels. RESULTS: We found that chronic EtOH exposure decreased SAM levels, SAM/SAH ratio, Ms, methylene tetrahydrofolate reductase, and betaine homocysteine methyltransferase (Bhmt) expression and increased methionine adenosyltransferase-2b (Mat2b) but not Mat2a expression in the liver. In contrast, chronic EtOH exposure decreased SAH levels, increased SAM/SAH ratio and the expression of Mat2a and S-adenosyl homocysteine hydrolase, while the levels of SAM or Bhmt expression in cerebellum remained unaltered. However, in both liver and cerebellum, chronic EtOH exposure decreased the expression of Ms and increased Mat2b expression. All chronic EtOH-induced changes of one-carbon metabolism in cerebellum, but not liver, returned to near-normal levels during EtOH withdrawal. CONCLUSIONS: These results indicate a decreased "methylation index" in liver and an increased "methylation index" in cerebellum. The opposing changes of the "methylation index" suggest altered DNA methylation in liver and cerebellum, thus implicating one-carbon metabolism in the pathophysiology of alcoholism.


Subject(s)
Alcohol Drinking/metabolism , Brain/metabolism , Carbon/metabolism , DNA Methylation/physiology , Ethanol/administration & dosage , Liver/metabolism , Animals , Brain/drug effects , DNA Methylation/drug effects , Liver/drug effects , Male , One-Carbon Group Transferases/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism
14.
Cell Death Differ ; 24(2): 193-194, 2017 02.
Article in English | MEDLINE | ID: mdl-28060379
15.
Methods ; 118-119: 146-162, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27939506

ABSTRACT

The diverse functional cellular roles played by ribonucleic acids (RNA) have emphasized the need to develop rapid and accurate methodologies to elucidate the relationship between the structure and function of RNA. Structural biology tools such as X-ray crystallography and Nuclear Magnetic Resonance are highly useful methods to obtain atomic-level resolution models of macromolecules. However, both methods have sample, time, and technical limitations that prevent their application to a number of macromolecules of interest. An emerging alternative to high-resolution structural techniques is to employ a hybrid approach that combines low-resolution shape information about macromolecules and their complexes from experimental hydrodynamic (e.g. analytical ultracentrifugation) and solution scattering measurements (e.g., solution X-ray or neutron scattering), with computational modeling to obtain atomic-level models. While promising, scattering methods rely on aggregation-free, monodispersed preparations and therefore the careful development of a quality control pipeline is fundamental to an unbiased and reliable structural determination. This review article describes hydrodynamic techniques that are highly valuable for homogeneity studies, scattering techniques useful to study the low-resolution shape, and strategies for computational modeling to obtain high-resolution 3D structural models of RNAs, proteins, and RNA-protein complexes.


Subject(s)
Chromatography, Gel/methods , Neutron Diffraction/methods , RNA, Transfer, Lys/chemistry , RNA-Binding Proteins/chemistry , Ultracentrifugation/methods , X-Ray Diffraction/methods , 2',5'-Oligoadenylate Synthetase/chemistry , 2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , Computer Simulation , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Hydrodynamics , Models, Molecular , One-Carbon Group Transferases/chemistry , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , RNA, Transfer, Lys/genetics , RNA, Transfer, Lys/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Scattering, Small Angle , Software , West Nile virus/genetics , Zea mays/genetics , Zea mays/metabolism
16.
Microb Pathog ; 89: 100-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26427881

ABSTRACT

Ribonucleic acid (RNA) molecules consist of numerous chemically modified nucleosides that are highly conserved in eukarya, archeae, and bacteria, while others are unique to each domain of life. In bacteria, hundreds of RNA modification enzymes have been identified and implicated in biological pathways associated with many cell processes. The glucose-inhibited division (gid) operon encodes genes for two RNA modification enzymes named GidA and GidB. Studies have shown GidA is essential for the proper biosynthesis of 5-methylaminomethyl-2-thiouridine (mnm(5)s(2)U) of bacterial transfer RNA (tRNA) with GidB responsible for the methylation of the 16S ribosomal RNA (rRNA). Furthermore, deletion of gidA and gidB has shown to alter numerous bacterial properties like virulence, stress response, morphology, growth, antibiotic susceptibility, and others. In this review, we discuss the present knowledge of the RNA modification enzymes GidA and GidB, and their potential role in the biology and virulence of bacteria.


Subject(s)
Bacteria/enzymology , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , Operon , RNA Processing, Post-Transcriptional , RNA, Bacterial/metabolism , Bacteria/genetics , Gene Deletion
17.
Cell Metab ; 22(5): 861-73, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26411344

ABSTRACT

S-adenosylmethionine (SAM) and S-adenosylhomocysteine (SAH) link one-carbon metabolism to methylation status. However, it is unknown whether regulation of SAM and SAH by nutrient availability can be directly sensed to alter the kinetics of key histone methylation marks. We provide evidence that the status of methionine metabolism is sufficient to determine levels of histone methylation by modulating SAM and SAH. This dynamic interaction led to rapid changes in H3K4me3, altered gene transcription, provided feedback regulation to one-carbon metabolism, and could be fully recovered upon restoration of methionine. Modulation of methionine in diet led to changes in metabolism and histone methylation in the liver. In humans, methionine variability in fasting serum was commensurate with concentrations needed for these dynamics and could be partly explained by diet. Together these findings demonstrate that flux through methionine metabolism and the sensing of methionine availability may allow direct communication to the chromatin state in cells.


Subject(s)
Carbon/metabolism , Epigenesis, Genetic/genetics , Histones/metabolism , Methionine/metabolism , Animals , Chromatin/genetics , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Humans , Liver/metabolism , Methylation , Mice , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism
18.
BMC Med ; 13: 18, 2015 Jan 29.
Article in English | MEDLINE | ID: mdl-25630355

ABSTRACT

BACKGROUND: Maternal smoking is one of the most important modifiable risk factors for low birthweight, which is strongly associated with increased cardiometabolic disease risk in adulthood. Maternal smoking reduces the levels of the methyl donor vitamin B12 and is associated with altered DNA methylation at birth. Altered DNA methylation may be an important mechanism underlying increased disease susceptibility; however, the extent to which this can be induced in the developing fetus is unknown. METHODS: In this retrospective study, we measured concentrations of cobalt, vitamin B12, and mRNA transcripts encoding key enzymes in the 1-carbon cycle in 55 fetal human livers obtained from 11 to 21 weeks of gestation elective terminations and matched for gestation and maternal smoking. DNA methylation was measured at critical regions known to be susceptible to the in utero environment. Homocysteine concentrations were analyzed in plasma from 60 fetuses. RESULTS: In addition to identifying baseline sex differences, we found that maternal smoking was associated with sex-specific alterations of fetal liver vitamin B12, plasma homocysteine and expression of enzymes in the 1-carbon cycle in fetal liver. In the majority of the measured parameters which showed a sex difference, maternal smoking reduced the magnitude of that difference. Maternal smoking also altered DNA methylation at the imprinted gene IGF2 and the glucocorticoid receptor (GR/NR3C1). CONCLUSIONS: Our unique data strengthen studies linking in utero exposures to altered DNA methylation by showing, for the first time, that such changes are present in fetal life and in a key metabolic target tissue, human fetal liver. Furthermore, these data propose a novel mechanism by which such changes are induced, namely through alterations in methyl donor availability and changes in 1-carbon metabolism.


Subject(s)
Carbon/metabolism , DNA Methylation/drug effects , Fetus/metabolism , Liver/metabolism , One-Carbon Group Transferases/metabolism , Smoking/adverse effects , Adult , Body Weight , Cobalt/analysis , Female , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , Liver/chemistry , Male , One-Carbon Group Transferases/genetics , Pregnancy , RNA, Messenger/analysis , Receptors, Glucocorticoid/metabolism , Retrospective Studies , Sex Factors , Vitamin B 12/analysis
19.
Nanoscale ; 6(14): 8265-73, 2014 Jul 21.
Article in English | MEDLINE | ID: mdl-24931221

ABSTRACT

Gold nanorods (GNRs) are commonly used nanomaterials with potential harmful effects on male reproduction. However, the mechanism by which GNRs affect male reproduction remains largely undetermined. In this study, the metabolic changes in spermatocyte-derived cells GC-2 and Sertoli cell line TM-4 were analyzed after GNR treatment for 24 h. Metabolomic analysis revealed that glycine was highly decreased in TM-4 cells after GNR-10 nM treatment while there was no significant change in GC-2 cells. RT-PCR showed that the mRNA levels of glycine synthases in the mitochondrial pathway decreased after GNR treatment, while there was no significant difference in mRNA levels of glycine synthases in the cytoplasmic pathway. High content screening (HCS) showed that GNRs decreased membrane permeability and mitochondrial membrane potential of TM-4 cells, which was also confirmed by JC-1 staining. In addition, RT-PCR and Western blot indicated that the mRNA and protein levels of blood-testis barrier (BTB) factors (ZO-1, occludin, claudin-5, and connexin-43) in TM-4 cells were also disrupted by GNRs. After glycine was added into the medium, the GNR-induced harmful effects on mitochondria and BTB factors were recovered in TM-4 cells. Our results showed that even low doses of GNRs could induce significant toxic effects on mitochondria and BTB factors in TM-4 cells. Furthermore, we revealed that glycine was a potentially important metabolic intermediary for the changes of membrane permeability, mitochondrial membrane potential and BTB factors after GNR treatment in TM-4 cells.


Subject(s)
Blood-Testis Barrier/drug effects , Glycine/metabolism , Gold/chemistry , Metabolome/drug effects , Mitochondria/drug effects , Nanotubes/toxicity , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line , Gas Chromatography-Mass Spectrometry , Glycine/chemistry , Humans , Male , Mitochondria/metabolism , Nanotubes/chemistry , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism
20.
RNA Biol ; 11(12): 1495-507, 2014.
Article in English | MEDLINE | ID: mdl-25607529

ABSTRACT

Posttranscriptional modification of the uridine located at the wobble position (U34) of tRNAs is crucial for optimization of translation. Defects in the U34 modification of mitochondrial-tRNAs are associated with a group of rare diseases collectively characterized by the impairment of the oxidative phosphorylation system. Retrograde signaling pathways from mitochondria to nucleus are involved in the pathophysiology of these diseases. These pathways may be triggered by not only the disturbance of the mitochondrial (mt) translation caused by hypomodification of tRNAs, but also as a result of nonconventional roles of mt-tRNAs and mt-tRNA-modifying enzymes. The evolutionary conservation of these enzymes supports their importance for cell and organismal functions. Interestingly, bacterial and eukaryotic cells respond to stress by altering the expression or activity of these tRNA-modifying enzymes, which leads to changes in the modification status of tRNAs. This review summarizes recent findings about these enzymes and sets them within the previous data context.


Subject(s)
Escherichia coli/metabolism , RNA Processing, Post-Transcriptional , RNA/metabolism , Uridine/analogs & derivatives , Uridine/metabolism , Anticodon/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Codon/metabolism , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , One-Carbon Group Transferases/genetics , One-Carbon Group Transferases/metabolism , Oxidative Phosphorylation , RNA/genetics , RNA, Mitochondrial , RNA, Transfer, Amino Acid-Specific/genetics , RNA, Transfer, Amino Acid-Specific/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...