Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Anal Bioanal Chem ; 413(13): 3573-3582, 2021 May.
Article in English | MEDLINE | ID: mdl-33829277

ABSTRACT

Mammalian folate-dependent one-carbon (1C) metabolism provides the building blocks essential during development via amino acid interconversion, methyl-donor production, regeneration of redox factors, and de novo purine and thymidylate synthesis. Folate supplementation prevents many neural tube defects (NTDs) that occur during the embryonic process of neurulation. The mechanism by which folate functions during neurulation is not well understood, and not all NTDs are preventable by folate supplementation. Mthfd1l is a mitochondrial 1C metabolism enzyme that produces formate, a 1C donor that fuels biosynthesis and the methyl cycle in the cytoplasm. Homozygous deletion of the Mthfd1l gene in mice (Mthfd1lz/z) causes embryonic lethality, developmental delay, and folate-resistant NTDs. These mice also have defects in cranial mesenchyme formation. In this work, mass spectrometry imaging was used to obtain ion maps of the cranial mesenchyme that identified the spatial distribution and relative abundance of metabolites in wild-type and Mthfd1lz/z embryos. The relative abundances of purine and thymidylate derivatives, as well as amino acids, were diminished in the cranial mesenchyme of Mthfd1lz/z embryos. Loss of Mthfd1l activity in this region also led to abnormal levels of methionine and dysregulated energy metabolism. These alterations in metabolism suggest possible approaches to preventing NTDs in humans.


Subject(s)
Aminohydrolases/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Mice , Mice, Knockout
2.
Birth Defects Res ; 111(19): 1520-1534, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31518072

ABSTRACT

BACKGROUND: Periconceptional intake of supplemental folic acid can reduce the incidence of neural tube defects by as much as 70%, but the mechanisms by which folic acid supports cellular processes during neural tube closure are unknown. The mitochondrial 10-formyl-tetrahydrofolate synthetase MTHFD1L catalyzes production of formate, thus generating one-carbon units for cytoplasmic processes. Deletion of Mthfd1l causes embryonic lethality, developmental delay, and neural tube defects in mice. METHODS: To investigate the role of mitochondrial one-carbon metabolism during cranial neural tube closure, we have analyzed cellular morphology and function in neural tissues in Mthfd1l knockout embryos. RESULTS: The head mesenchyme showed significantly lower cellular density in Mthfd1l nullizygous embryos compared to wildtype embryos during the process of neural tube closure. Apoptosis and neural crest cell specification were not affected by deletion of Mthfd1l. Sections from the cranial region of Mthfd1l knockout embryos exhibited decreased cellular proliferation, but only after completion of neural tube closure. Supplementation of pregnant dams with formate improved mesenchymal density and corrected cell proliferation in the nullizygous embryos. CONCLUSIONS: Deletion of Mthfd1l causes decreased density in the cranial mesenchyme and this defect is improved with formate supplementation. This study reveals a mechanistic link between folate-dependent mitochondrially produced formate, head mesenchyme formation and neural tube defects.


Subject(s)
Formate-Tetrahydrofolate Ligase/genetics , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multifunctional Enzymes/genetics , Neural Tube Defects/genetics , Animals , Embryo, Mammalian/metabolism , Female , Folic Acid/genetics , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Formates/metabolism , Male , Mesoderm/metabolism , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism , Multifunctional Enzymes/metabolism , Neural Crest/metabolism , Neural Tube Defects/metabolism , Neurulation , Sequence Deletion
3.
J Biol Chem ; 293(16): 5821-5833, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29483189

ABSTRACT

One-carbon (1C) metabolism is a universal folate-dependent pathway essential for de novo purine and thymidylate synthesis, amino acid interconversion, universal methyl-donor production, and regeneration of redox cofactors. Homozygous deletion of the 1C pathway gene Mthfd1l encoding methylenetetrahydrofolate dehydrogenase (NADP+-dependent) 1-like, which catalyzes mitochondrial formate production from 10-formyltetrahydrofolate, results in 100% penetrant embryonic neural tube defects (NTDs), underscoring the central role of mitochondrially derived formate in embryonic development and providing a mechanistic link between folate and NTDs. However, the specific metabolic processes that are perturbed by Mthfd1l deletion are not known. Here, we performed untargeted metabolomics on whole Mthfd1l-null and wildtype mouse embryos in combination with isotope tracer analysis in mouse embryonic fibroblast (MEF) cell lines to identify Mthfd1l deletion-induced disruptions in 1C metabolism, glycolysis, and the TCA cycle. We found that maternal formate supplementation largely corrects these disruptions in Mthfd1l-null embryos. Serine tracer experiments revealed that Mthfd1l-null MEFs have altered methionine synthesis, indicating that Mthfd1l deletion impairs the methyl cycle. Supplementation of Mthfd1l-null MEFs with formate, hypoxanthine, or combined hypoxanthine and thymidine restored their growth to wildtype levels. Thymidine addition alone was ineffective, suggesting a purine synthesis defect in Mthfd1l-null MEFs. Tracer experiments also revealed lower proportions of labeled hypoxanthine and inosine monophosphate in Mthfd1l-null than in wildtype MEFs, suggesting that Mthfd1l deletion results in increased reliance on the purine salvage pathway. These results indicate that disruptions of mitochondrial 1C metabolism have wide-ranging consequences for many metabolic processes, including those that may not directly interact with 1C metabolism.


Subject(s)
Aminohydrolases/genetics , Energy Metabolism , Formate-Tetrahydrofolate Ligase/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Metabolic Networks and Pathways , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mitochondria/metabolism , Multienzyme Complexes/genetics , Neural Tube Defects/genetics , Aminohydrolases/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Folic Acid/genetics , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Formates/metabolism , Glycolysis , Metabolome , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/pathology , Multienzyme Complexes/metabolism , Neural Tube Defects/metabolism , Neural Tube Defects/pathology
4.
Cancer Metab ; 5: 11, 2017.
Article in English | MEDLINE | ID: mdl-29225823

ABSTRACT

BACKGROUND: Folate-dependent one-carbon metabolism provides one-carbon units for several biological processes. This pathway is highly compartmentalized in eukaryotes, with the mitochondrial pathway producing formate for use in cytoplasmic processes. The mitochondrial enzyme MTHFD2 has been reported to use NAD+ as a cofactor while the isozyme MTHFD2L utilizes NAD+ or NADP+ at physiologically relevant conditions. Because MTHFD2 is highly expressed in many cancer types, we sought to determine the cofactor preference of this enzyme. RESULTS: Kinetic analysis shows that purified human MTHFD2 exhibits dual redox cofactor specificity, utilizing either NADP+ or NAD+ with the more physiologically relevant pentaglutamate folate substrate. CONCLUSION: These results show that the mitochondrial folate pathway isozymes MTHFD2 and MTHFD2L both exhibit dual redox cofactor specificity. Our kinetic analysis clearly supports a role for MTHFD2 in mitochondrial NADPH production, indicating that this enzyme is likely responsible for mitochondrial production of both NADH and NADPH in rapidly proliferating cells.

6.
Birth Defects Res A Clin Mol Teratol ; 100(8): 576-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24985542

ABSTRACT

BACKGROUND: Neural tube defects (NTDs) are one of the most common birth defects in humans. Maternal intake of folic acid was linked to prevention of NTDs in the 1970s. This realization led to the establishment of mandatory and/or voluntary food folic acid fortification programs in many countries that have reduced the incidence of NTDs by up to 70% in humans. Despite 40 years of intensive research, the biochemical mechanisms underlying the protective effects of folic acid remain unknown. RESULTS: Recent research reveals a role for mitochondrial folate-dependent one-carbon metabolism in neural tube closure. CONCLUSION: In this article, we review the evidence linking NTDs to aberrant mitochondrial one-carbon metabolism in humans and mouse models. The potential of formate, a product of mitochondrial one-carbon metabolism, to prevent NTDs is also discussed.


Subject(s)
Folic Acid/therapeutic use , Mitochondria/enzymology , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Neural Tube/embryology , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Dietary Supplements , Folic Acid/blood , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Formates/pharmacology , Humans , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Minor Histocompatibility Antigens , Mitochondria/metabolism , Models, Animal , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Neural Tube/enzymology
7.
J Biol Chem ; 289(22): 15507-17, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24733394

ABSTRACT

Mammalian mitochondria are able to produce formate from one-carbon donors such as serine, glycine, and sarcosine. This pathway relies on the mitochondrial pool of tetrahydrofolate (THF) and several folate-interconverting enzymes in the mitochondrial matrix. We recently identified MTHFD2L as the enzyme that catalyzes the oxidation of 5,10-methylenetetrahydrofolate (CH2-THF) in adult mammalian mitochondria. We show here that the MTHFD2L enzyme is bifunctional, possessing both CH2-THF dehydrogenase and 5,10-methenyl-THF cyclohydrolase activities. The dehydrogenase activity can use either NAD(+) or NADP(+) but requires both phosphate and Mg(2+) when using NAD(+). The NADP(+)-dependent dehydrogenase activity is inhibited by inorganic phosphate. MTHFD2L uses the mono- and polyglutamylated forms of CH2-THF with similar catalytic efficiencies. Expression of the MTHFD2L transcript is low in early mouse embryos but begins to increase at embryonic day 10.5 and remains elevated through birth. In adults, MTHFD2L is expressed in all tissues examined, with the highest levels observed in brain and lung.


Subject(s)
Aminohydrolases/metabolism , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/enzymology , Multienzyme Complexes/metabolism , Neural Tube/enzymology , Age Factors , Alternative Splicing/physiology , Aminohydrolases/genetics , Animals , Female , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Inbred C57BL , Multienzyme Complexes/genetics , NAD/metabolism , NADP/metabolism , Neural Tube/embryology , Oxidation-Reduction , Pregnancy , Rats , Substrate Specificity
8.
Proc Natl Acad Sci U S A ; 110(2): 549-54, 2013 Jan 08.
Article in English | MEDLINE | ID: mdl-23267094

ABSTRACT

Maternal supplementation with folic acid is known to reduce the incidence of neural tube defects (NTDs) by as much as 70%. Despite the strong clinical link between folate and NTDs, the biochemical mechanisms through which folic acid acts during neural tube development remain undefined. The Mthfd1l gene encodes a mitochondrial monofunctional 10-formyl-tetrahydrofolate synthetase, termed MTHFD1L. This gene is expressed in adults and at all stages of mammalian embryogenesis with localized regions of higher expression along the neural tube, developing brain, craniofacial structures, limb buds, and tail bud. In both embryos and adults, MTHFD1L catalyzes the last step in the flow of one-carbon units from mitochondria to cytoplasm, producing formate from 10-formyl-THF. To investigate the role of mitochondrial formate production during embryonic development, we have analyzed Mthfd1l knockout mice. All embryos lacking Mthfd1l exhibit aberrant neural tube closure including craniorachischisis and exencephaly and/or a wavy neural tube. This fully penetrant folate-pathway mouse model does not require feeding a folate-deficient diet to cause this phenotype. Maternal supplementation with sodium formate decreases the incidence of NTDs and partially rescues the growth defect in embryos lacking Mthfd1l. These results reveal the critical role of mitochondrially derived formate in mammalian development, providing a mechanistic link between folic acid and NTDs. In light of previous studies linking a common splice variant in the human MTHFD1L gene with increased risk for NTDs, this mouse model provides a powerful system to help elucidate the specific metabolic mechanisms that underlie folate-associated birth defects, including NTDs.


Subject(s)
Abnormalities, Multiple/genetics , Aminohydrolases/genetics , Craniofacial Abnormalities/genetics , Embryonic Development/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Multienzyme Complexes/genetics , Neural Tube Defects/genetics , Aminohydrolases/deficiency , Animals , DNA Primers/genetics , Embryonic Development/drug effects , Formate-Tetrahydrofolate Ligase/deficiency , Formates/administration & dosage , Formates/pharmacology , Gene Deletion , Genotype , Immunoblotting , Metabolic Networks and Pathways/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Mice , Mice, Knockout , Multienzyme Complexes/deficiency , Reverse Transcriptase Polymerase Chain Reaction
9.
J Biol Chem ; 286(7): 5166-74, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21163947

ABSTRACT

Previous studies in our laboratory showed that isolated, intact adult rat liver mitochondria are able to oxidize the 3-carbon of serine and the N-methyl carbon of sarcosine to formate without the addition of any other cofactors or substrates. Conversion of these 1-carbon units to formate requires several folate-interconverting enzymes in mitochondria. The enzyme(s) responsible for conversion of 5,10-methylene-tetrahydrofolate (CH(2)-THF) to 10-formyl-THF in adult mammalian mitochondria are currently unknown. A new mitochondrial CH(2)-THF dehydrogenase isozyme, encoded by the MTHFD2L gene, has now been identified. The recombinant protein exhibits robust NADP(+)-dependent CH(2)-THF dehydrogenase activity when expressed in yeast. The enzyme is localized to mitochondria when expressed in CHO cells and behaves as a peripheral membrane protein, tightly associated with the matrix side of the mitochondrial inner membrane. The MTHFD2L gene is subject to alternative splicing and is expressed in adult tissues in humans and rodents. This CH(2)-THF dehydrogenase isozyme thus fills the remaining gap in the pathway from CH(2)-THF to formate in adult mammalian mitochondria.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/biosynthesis , Mitochondria/enzymology , Mitochondrial Proteins/biosynthesis , Alternative Splicing/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Humans , Isoenzymes/biosynthesis , Isoenzymes/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Organ Specificity/physiology , Rats , Rats, Sprague-Dawley , Tetrahydrofolates/metabolism
11.
Annu Rev Nutr ; 30: 57-81, 2010 Aug 21.
Article in English | MEDLINE | ID: mdl-20645850

ABSTRACT

The recognition that mitochondria participate in folate-mediated one-carbon metabolism grew out of pioneering work beginning in the 1950s from the laboratories of D.M. Greenberg, C.G. Mackenzie, and G. Kikuchi. These studies revealed mitochondria as the site of oxidation of one-carbon donors such as serine, glycine, sarcosine, and dimethylglycine. Subsequent work from these laboratories and others demonstrated the participation of folate coenzymes and folate-dependent enzymes in these mitochondrial processes. Biochemical and molecular genetic approaches in the 1980s and 1990s identified many of the enzymes involved and revealed an interdependence of cytoplasmic and mitochondrial one-carbon metabolism. These studies led to the development of a model of eukaryotic one-carbon metabolism that comprises parallel cytosolic and mitochondrial pathways, connected by one-carbon donors such as serine, glycine, and formate. Sequencing of the human and other mammalian genomes has facilitated identification of the enzymes that participate in this intercompartmental one-carbon metabolism, and animal models are beginning to clarify the roles of the cytoplasmic and mitochondrial isozymes of these enzymes. Identifying the mitochondrial transporters for the one-carbon donors and elucidating how flux through these pathways is controlled are two areas ripe for exploration.


Subject(s)
Folic Acid/metabolism , Mammals/metabolism , Mitochondria/enzymology , One-Carbon Group Transferases/metabolism , Animals , Cell Nucleus/enzymology , Cytoplasm/enzymology , Humans
12.
J Biol Chem ; 285(7): 4612-20, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-19948730

ABSTRACT

Mitochondrial folate-dependent one-carbon (1-C) metabolism converts 1-C donors such as serine and glycine to formate, which is exported and incorporated into the cytoplasmic tetrahydrofolate (THF) 1-C pool. Developing embryos depend on this mitochondrial pathway to provide 1-C units for cytoplasmic process such as de novo purine biosynthesis and the methyl cycle. This pathway is composed of sequential methylene-THF dehydrogenase, methenyl-THF cyclohydrolase, and 10-formyl-THF synthetase activities. In embryonic mitochondria, the bifunctional MTHFD2 enzyme catalyzes the dehydrogenase and cyclohydrolase reactions, but the enzyme responsible for the mitochondrial synthetase reaction has not been identified in embryos. A monofunctional 10-formyl-THF synthetase (MTHFD1L gene product) functions in adult mitochondria and is a likely candidate for the embryonic activity. Here we show that the MTHFD1L enzyme is present in mitochondria from normal embryonic tissues and embryonic fibroblast cell lines, and embryonic mitochondria possess the ability to synthesize formate from glycine. The MTHFD1L transcript was detected at all stages of mouse embryogenesis examined. In situ hybridizations showed that MTHFD1L was expressed ubiquitously throughout the embryo but with localized regions of higher expression. The spatial pattern of MTHFD1L expression was virtually indistinguishable from that of MTHFD2 and MTHFD1 (cytoplasmic C(1)-THF synthase) in embryonic day 9.5 mouse embryos, suggesting coordinated regulation. Finally, we show using stable isotope labeling that in an embryonic mouse cell line, greater than 75% of 1-C units entering the cytoplasmic methyl cycle are mitochondrially derived. Thus, a complete pathway of enzymes for supplying 1-C units from the mitochondria to the methyl cycle in embryonic tissues is established.


Subject(s)
Aminohydrolases/metabolism , Carbon/metabolism , Embryo, Mammalian/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/metabolism , Multienzyme Complexes/metabolism , Aminohydrolases/genetics , Animals , Blotting, Northern , Cell Line , Chromatography, Gas , Deuterium/metabolism , Electrophoresis, Polyacrylamide Gel , Female , Formate-Tetrahydrofolate Ligase/genetics , Immunoblotting , In Situ Hybridization , Liver/metabolism , Mass Spectrometry , Methionine/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Models, Biological , Multienzyme Complexes/genetics , Pregnancy
13.
J Biol Chem ; 284(49): 34116-25, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19843529

ABSTRACT

Initiation of protein synthesis in mitochondria and chloroplasts normally uses a formylated initiator methionyl-tRNA (fMet-tRNA(f)(Met)). However, mitochondrial protein synthesis in Saccharomyces cerevisiae can initiate with nonformylated Met-tRNA(f)(Met), as demonstrated in yeast mutants in which the nuclear gene encoding mitochondrial methionyl-tRNA formyltransferase (FMT1) has been deleted. The role of formylation of the initiator tRNA is not known, but in vitro formylation increases binding of Met-tRNA(f)(Met) to translation initiation factor 2 (IF2). We hypothesize the existence of an accessory factor that assists mitochondrial IF2 (mIF2) in utilizing unformylated Met-tRNA(f)(Met). This accessory factor might be unnecessary when formylated Met-tRNA(f)(Met) is present but becomes essential when only the unformylated species are available. Using a synthetic petite genetic screen in yeast, we identified a mutation in the AEP3 gene that caused a synthetic respiratory-defective phenotype together with Delta fmt1. The same aep3 mutation also caused a synthetic respiratory defect in cells lacking formylated Met-tRNA(f)(Met) due to loss of the MIS1 gene that encodes the mitochondrial C(1)-tetrahydrofolate synthase. The AEP3 gene encodes a peripheral mitochondrial inner membrane protein that stabilizes mitochondrially encoded ATP6/8 mRNA. Here we show that the AEP3 protein (Aep3p) physically interacts with yeast mIF2 both in vitro and in vivo and promotes the binding of unformylated initiator tRNA to yeast mIF2. We propose that Aep3p functions as an accessory initiation factor in mitochondrial protein synthesis.


Subject(s)
Gene Expression Regulation, Fungal , Hydroxymethyl and Formyl Transferases/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , RNA, Transfer, Met/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Epitopes/chemistry , Membrane Proteins/chemistry , Models, Genetic , Mutation , Phenotype , Plasmids/metabolism , Point Mutation , Protein Binding , Protein Biosynthesis , RNA, Transfer/chemistry , Recombinant Fusion Proteins/chemistry , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/chemistry
14.
Arch Biochem Biophys ; 481(1): 86-93, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18996079

ABSTRACT

Mammalian mitochondrial C(1)-tetrahydrofolate (THF) synthase (MTHFDIL gene product) is a monofunctional 10-formyl-THF synthetase, lacking the 5,10-methylene-THF dehydrogenase and 5,10-methenyl-THF cyclohydrolase activities typically found in the trifunctional cytoplasmic proteins. Here, we report the submitochondrial localization of epitope-tagged human mitochondrial C(1)-THF synthase expressed in Chinese hamster ovary cells. Mitochondrial fractionation experiments show that human mitochondrial C(1)-THF synthase behaves as a peripheral membrane protein, tightly associated with the matrix side of the mitochondrial inner membrane. Inner mitochondrial membrane association was also observed for the endogenous mitochondrial C(1)-THF synthase in adult rat spleen. We also purified and characterized the recombinant protein product (short isoform) of the alternatively spliced short transcript of the mitochondrial isozyme. Methylene-THF dehydrogenase assays confirmed that the short isoform is not enzymatically active. The purified short isoform was used in the production of polyclonal antibodies specific for the mitochondrial isozyme. These antibodies detected endogenous full-length mitochondrial C(1)-THF synthase in mitochondria from adult rat spleen and human placenta, confirming the expression of the mitochondrial isozyme in adult mammalian tissues.


Subject(s)
Aminohydrolases/metabolism , Formate-Tetrahydrofolate Ligase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondrial Proteins/metabolism , Multienzyme Complexes/metabolism , Alternative Splicing , Animals , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mitochondrial Membranes/enzymology , Mitochondrial Proteins/genetics , Placenta/enzymology , Pregnancy , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spleen/enzymology
15.
Arch Biochem Biophys ; 467(2): 218-26, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17935688

ABSTRACT

Methionine synthase catalyzes the transfer of a methyl group from tetrahydrofolate to homocysteine to produce methionine. Although mammalian enzymes are cobalamin-dependent, fungal methionine synthases are cobalamin-independent. The opportunistic pathogen Candida albicans is a diploid and carries two copies of the methionine synthase gene, MET6. Homologous recombination was used to disrupt a single MET6 gene. MET6/met6 knock-outs, deleted with either the URA3 or ARG4 cassette, grew as well as the wild-type strain. However, we were unable to obtain a viable met6/met6 deletion strain, even on media supplemented with exogenous methionine. This suggests that methionine synthase is essential to C. albicans. To explore this further, a C. albicans strain was constructed in which one MET6 locus was deleted and the second placed under a regulatable promoter. The conditional mutant grew well under inducing conditions, even in the absence of methionine. It would not grow under repressing conditions in the absence of methionine, but would grow when the media was supplemented with exogenous methionine. A Western blot showed that a small amount of enzyme was expressed under repressing conditions. Taken together, these data reveal that methionine is necessary for growth of C. albicans, but not sufficient-a minimal level of methionine synthase expression is required, perhaps to limit homocysteine toxicity. Furthermore, these results suggest that cobalamin-independent methionine synthase is a plausible target for the design of antifungal agents.


Subject(s)
Candida albicans/enzymology , Candida albicans/growth & development , Drug Delivery Systems/methods , Gene Expression Regulation, Fungal/physiology , Methyltransferases/genetics , Methyltransferases/metabolism , Antifungal Agents/administration & dosage , Candida albicans/drug effects , Candida albicans/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Silencing
16.
J Biol Chem ; 282(38): 27744-53, 2007 Sep 21.
Article in English | MEDLINE | ID: mdl-17652090

ABSTRACT

The TRM5 gene encodes a tRNA (guanine-N1-)-methyltransferase (Trm5p) that methylates guanosine at position 37 (m(1)G37) in cytoplasmic tRNAs in Saccharomyces cerevisiae. Here we show that Trm5p is also responsible for m(1)G37 methylation of mitochondrial tRNAs. The TRM5 open reading frame encodes 499 amino acids containing four potential initiator codons within the first 48 codons. Full-length Trm5p, purified as a fusion protein with maltose-binding protein, exhibited robust methyltransferase activity with tRNA isolated from a Delta trm5 mutant strain, as well as with a synthetic mitochondrial initiator tRNA (tRNA(Met)(f)). Primer extension demonstrated that the site of methylation was guanosine 37 in both mitochondrial tRNA(Met)(f) and tRNA(Phe). High pressure liquid chromatography analysis showed the methylated product to be m(1)G. Subcellular fractionation and immunoblotting of a strain expressing a green fluorescent protein-tagged version of the TRM5 gene revealed that the enzyme was localized to both cytoplasm and mitochondria. The slightly larger mitochondrial form was protected from protease digestion, indicating a matrix localization. Analysis of N-terminal truncation mutants revealed that a Trm5p active in the cytoplasm could be obtained with a construct lacking amino acids 1-33 (Delta1-33), whereas production of a Trm5p active in the mitochondria required these first 33 amino acids. Yeast expressing the Delta1-33 construct exhibited a significantly lower rate of oxygen consumption, indicating that efficiency or accuracy of mitochondrial protein synthesis is decreased in cells lacking m(1)G37 methylation of mitochondrial tRNAs. These data suggest that this tRNA modification plays an important role in reading frame maintenance in mitochondrial protein synthesis.


Subject(s)
Gene Expression Regulation, Fungal , Guanosine/chemistry , RNA, Transfer/chemistry , RNA/chemistry , Saccharomyces cerevisiae Proteins/physiology , tRNA Methyltransferases/metabolism , Chromatography, High Pressure Liquid , Cloning, Molecular , Cytoplasm/metabolism , Mitochondria/metabolism , Mutation , Oxygen Consumption , Protein Structure, Tertiary , RNA, Mitochondrial , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Subcellular Fractions/metabolism , tRNA Methyltransferases/genetics , tRNA Methyltransferases/physiology
17.
Metab Eng ; 9(1): 8-20, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17049899

ABSTRACT

Metabolic enzymes control cellular metabolite concentrations dynamically in response to changing environmental and intracellular conditions. Such real-time feedback regulation suggests the global metabolome may sample distinct dynamic steady states, forming "basins of stability" in the energy landscape of possible metabolite concentrations and enzymatic activities. Using metabolite, protein and transcriptional profiling, we characterize three dynamic steady states of the yeast metabolome that form by perturbing synthesis of the universal methyl donor S-adenosylmethionine (AdoMet). Conversion between these states is driven by replacement of serine with glycine+formate in the media, loss of feedback inhibition control by the metabolic enzyme Met13, or both. The latter causes hyperaccumulation of methionine and AdoMet, and dramatic global compensatory changes in the metabolome, including differences in amino acid and sugar metabolism, and possibly in the global nitrogen balance, ultimately leading to a G1/S phase cell cycle delay. Global metabolic changes are not necessarily accompanied by global transcriptional changes, and metabolite-controlled post-transcriptional regulation of metabolic enzymes is clearly evident.


Subject(s)
Saccharomyces cerevisiae/metabolism , Feedback, Physiological , G1 Phase , Glutathione/metabolism , Magnetic Resonance Spectroscopy , Proteome , S Phase , S-Adenosylmethionine/metabolism , Transcription, Genetic
18.
Biochemistry ; 44(39): 13163-71, 2005 Oct 04.
Article in English | MEDLINE | ID: mdl-16185084

ABSTRACT

5,10-Methylenetetrahydrofolate dehydrogenase (MTD) catalyzes the reversible oxidation of 5,10-methylenetetrahydrofolate to 5,10-methenyltetrahydrofolate. This reaction is critical for the supply of one-carbon units at the required oxidation states for the synthesis of purines and dTMP. For most MTDs, dehydrogenase activity is co-located with a methenyl-THF cyclohydrolase activity as part of bifunctional or trifunctional enzyme. The yeast Saccharomyces cerevisiae contains a monofunctional NAD(+)-dependent 5,10-methylenetetrahydrofolate dehydrogenase (yMTD). Kinetic, crystallographic, and mutagenesis studies were conducted to identify critical residues in order to gain further insight into the reaction mechanism of this enzyme and its apparent lack of cyclohydrolase activity. Hydride transfer was found to be rate-limiting for the oxidation of methylenetetrahydrofolate by kinetic isotope experiments (V(H)/V(D) = 3.3), and the facial selectivity of the hydride transfer to NAD(+) was determined to be Pro-R (A-specific). Model building based on the previously solved structure of yMTD with bound NAD cofactor suggested a possible role for three conserved amino acids in substrate binding or catalysis: Glu121, Cys150, and Thr151. Steady-state kinetic measurements of mutant enzymes demonstrated that Glu121 and Cys150 were essential for dehydrogenase activity, whereas Thr151 allowed some substitution. Our results are consistent with a key role for Glu121 in correctly binding the folate substrate; however, the exact role of C150 is unclear. Single mutants Thr57Lys and Tyr98Gln and double mutant T57K/Y98Q were prepared to test the hypothesis that the lack of cyclohydrolase activity in yMTD was due to the substitution of a conserved Lys/Gln pair found in bifunctional MTDs. Each mutant retained dehydrogenase activity, but no cyclohydrolase activity was detected.


Subject(s)
Methylenetetrahydrofolate Dehydrogenase (NAD+)/metabolism , Binding Sites/genetics , Folic Acid/metabolism , Kinetics , Methylenetetrahydrofolate Dehydrogenase (NAD+)/chemistry , Methylenetetrahydrofolate Dehydrogenase (NAD+)/genetics , Molecular Structure , Mutation , Protein Conformation , Saccharomyces cerevisiae Proteins , Substrate Specificity
19.
Arch Biochem Biophys ; 442(2): 196-205, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16171773

ABSTRACT

A human mitochondrial isozyme of C1-tetrahydrofolate (THF) synthase was previously identified by its similarity to the human cytoplasmic C1-THF synthase. All C1-THF synthases characterized to date, from yeast to human, are trifunctional, containing the activities of 5,10-methylene-THF dehydrogenase, 5,10-methenyl-THF cyclohydrolase, and 10-formyl-THF synthetase. Here we report on the enzymatic characterization of the recombinant human mitochondrial isozyme. Enzyme assays of purified human mitochondrial C1-THF synthase protein revealed only the presence of 10-formyl-THF synthetase activity. Gel filtration and crosslinking studies indicated that human mitochondrial C1-THF synthase exists as a homodimer in solution. Steady-state kinetic characterization of the 10-formyl-THF synthetase activity was performed using (6R,S)-H4-PteGlu1, (6R,S)-H4-PteGlu3, and (6R,S)-H4-PteGlu5 substrates. The (6R,S)-H4-PteGlun Km dropped from greater than 500 microM for the monoglutamate to 15 microM and 3.6 microM for the tri- and pentaglutamates, respectively. The Km values for formate and ATP also are lowered when THF polyglutamates are used. The formate Km dropped 79-fold and the ATP Km dropped more than 5-fold when (6R,S)-H4-PteGlu5 was used as the substrate in place of (6R,S)-H4-PteGlu1.


Subject(s)
Aminohydrolases/chemistry , Formate-Tetrahydrofolate Ligase/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Mitochondria/enzymology , Multienzyme Complexes/chemistry , Aminohydrolases/genetics , Aminohydrolases/metabolism , Biological Assay/methods , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Glutamates/chemistry , Glutamates/metabolism , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
20.
Arch Biochem Biophys ; 441(1): 56-63, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16083849

ABSTRACT

In this study, we investigated methionine synthase from Candida albicans (CaMET 6p) and Saccharomyces cerevisiae (ScMET 6p). We describe the cloning of CaMet 6 and ScMet 6, and the expression of both the enzymes in S. cerevisiae. CaMET 6p is able to complement the disruption of met 6 in S. cerevisiae. Following the purification of ScMET 6p and CaMET 6p, kinetic assays were performed to determine substrate specificity. The Michaelis constants for ScMET 6p with CH(3)-H(4)PteGlu(2), CH(3)-H(4)PteGlu(3), CH(3)-H(4)PteGlu(4), and l-homocysteine are 108, 84, 95, and 13 microM, respectively. The Michaelis constants for CaMET 6p with CH(3)-H(4)PteGlu(2), CH(3)-H(4)PteGlu(3), CH(3)-H(4)PteGlu(4), and l-homocysteine are 113, 129, 120, and 14 microM, respectively. Neither enzyme showed activity with CH(3)-H(4)PteGlu(1) as a substrate. We conclude that ScMET 6p and CaMET 6p require a minimum of two glutamates on the methyltetrahydrofolate substrate, similar to the bacterial metE homologs. The cloning, purification, and characterization of these enzymes lay the groundwork for inhibitor-design studies on the cobalamin-independent fungal methionine synthases.


Subject(s)
Candida albicans/enzymology , Methyltransferases/chemistry , Methyltransferases/isolation & purification , Saccharomyces cerevisiae/metabolism , Tetrahydrofolates/chemistry , Amino Acid Sequence , Candida albicans/genetics , Enzyme Activation , Kinetics , Methyltransferases/genetics , Molecular Sequence Data , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Saccharomyces cerevisiae/genetics , Sequence Homology, Amino Acid , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...