Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Amino Acids ; 53(10): 1589-1595, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34550462

ABSTRACT

One-carbon units, critical intermediates for cell growth, may be produced by a variety of means, one of which is via the production of formate. Excessive formate accumulation, known as formate overflow and a characteristic of oxidative cancer, has been observed in cancer cells. However, the basis for this high rate of formate production is unknown. We examined the effect of elevated expression of oncogenic Ras (RasV12), on formate production in NIH-3T3 cells (mouse fibroblasts) cultured with either labelled 13C-serine or 13C-glycine. Formate accumulation by the fibroblasts transformed by RasV12 was increased two-threefold over those by vector control (Babe) cells. The production of formate exceeded the rate of utilization in both cell types. 13C-formate was produced almost exclusively from the #3 carbon of 13C-serine. Virtually no labelled formate was produced from either the #2 carbon of serine or the #2 carbon of glycine. The increased formate production by RasV12 cells was associated with increased mRNA abundances for enzymes of formate production in both the mitochondria and the cytosol. Thus, we find the oncogenic RasV12 significantly increases formate overflow and may be one way for tumor cells to produce one-carbon units required for enhanced proliferation of these cells and/or for other processes which have not been identified.


Subject(s)
Formates/metabolism , Genes, ras , ras Proteins , Aminohydrolases/genetics , Aminohydrolases/metabolism , Animals , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation , Glycine/metabolism , Glycine Hydroxymethyltransferase/genetics , Glycine Hydroxymethyltransferase/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mice , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , NIH 3T3 Cells , RNA, Messenger/genetics , Serine/metabolism , ras Proteins/genetics , ras Proteins/metabolism
2.
J Biosci ; 462021.
Article in English | MEDLINE | ID: mdl-34168100

ABSTRACT

Obesity has been followed with interest as a risk factor for COVID-19, with triglycerides as one of four common criteria used to define obesity, which have been used to study the mechanism of obesity. In this study, we showed that angiotensin-converting enzyme-2 (ACE2) is widely expressed in the mouse body, including the kidney, spleen, brain, heart, lung, liver, and testis, and that ACE2 levels increased after a high-fat diet. The ACE2 levels were recorded at 0 days, 3 days, 7 days, and 14 days after a high-fat diet, and they increased at 14 days after high-fat diet initiation. In addition, triglyceride levels were also significantly increased at 14 days after high-fat diet initiation, but body weight was not changed. Furthermore, we examined the ACE2 levels in Calu3 cells (a lung cancer cell line) after triglyceride treatment, and the results indicated that ACE2 levels were increased at 25 µM and reached their peak at 200 µM. Finally, we found that the mRNA level of mthfd1 was significantly increased in the high-fat diet group. Given these findings, we hypothesize that triglycerides can regulate the expression of ACE2 and Mthfd1.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Diet, High-Fat/adverse effects , Formate-Tetrahydrofolate Ligase/metabolism , Gene Expression Regulation/drug effects , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multifunctional Enzymes/metabolism , Triglycerides/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Animals , Biomarkers/blood , COVID-19/etiology , COVID-19/virology , Cell Line , Cell Survival , Formate-Tetrahydrofolate Ligase/genetics , Humans , Male , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Inbred C57BL , Multifunctional Enzymes/genetics , Obesity/complications , Obesity/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Risk Factors , SARS-CoV-2 , Triglycerides/administration & dosage
3.
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
4.
J Nutr ; 148(4): 501-509, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29659962

ABSTRACT

Background: Suboptimal folate intake, a risk factor for birth defects, is common even in areas with folate fortification. A polymorphism in methylenetetrahydrofolate dehydrogenase 1 (MTHFD1), R653Q (MTHFD1 c.1958 G > A), has also been associated with increased birth defect risk, likely through reduced purine synthesis. Objective: We aimed to determine if the interaction of MTHFD1 synthetase deficiency and low folate intake increases developmental abnormalities in a mouse model for MTHFD1 R653Q. Methods: Female Mthfd1S+/+ and Mthfd1S+/- mice were fed control or low-folate diets (2 and 0.3 mg folic acid/kg diet, respectively) before mating and during pregnancy. Embryos and placentas were examined for anomalies at embryonic day 10.5. Maternal 1-carbon metabolites were measured in plasma and liver. Results: Delays and defects doubled in litters of Mthfd1S+/- females fed low-folate diets compared to wild-type females fed either diet, or Mthfd1S+/- females fed control diets [P values (defects): diet 0.003, maternal genotype 0.012, diet × maternal genotype 0.014]. These adverse outcomes were associated with placental dysmorphology. Intrauterine growth restriction was increased by embryonic Mthfd1S+/- genotype, folate deficiency, and interaction of maternal Mthfd1S+/- genotype with folate deficiency (P values: embryonic genotype 0.045, diet 0.0081, diet × maternal genotype 0.0019). Despite a 50% increase in methylenetetrahydrofolate reductase expression in low-folate maternal liver (P diet = 0.0007), methyltetrahydrofolate concentration decreased 70% (P diet <0.0001) and homocysteine concentration doubled in plasma (P diet = 0.0001); S-adenosylmethionine decreased 40% and S-adenosylhomocysteine increased 20% in low-folate maternal liver (P diet = 0.002 and 0.0002, respectively). Conclusions: MTHFD1 synthetase-deficient mice are more sensitive to low folate intake than wild-type mice during pregnancy. Reduced purine synthesis due to synthetase deficiency and altered methylation potential due to low folate may increase pregnancy complications. Further studies and individualized intake recommendations may be required for women homozygous for the MTHFD1 R653Q variant.


Subject(s)
Congenital Abnormalities/etiology , Folic Acid Deficiency/complications , Folic Acid/administration & dosage , Formate-Tetrahydrofolate Ligase/deficiency , Genotype , Methenyltetrahydrofolate Cyclohydrolase/deficiency , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Multifunctional Enzymes/deficiency , Polymorphism, Genetic , Pregnancy Complications/etiology , Animals , DNA Methylation , Diet , Disease Models, Animal , Female , Fetal Development , Fetal Growth Retardation/etiology , Folic Acid/blood , Folic Acid Deficiency/blood , Folic Acid Deficiency/genetics , Folic Acid Deficiency/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Ligases , Liver/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Mice , Multifunctional Enzymes/genetics , Multifunctional Enzymes/metabolism , Placenta , Pregnancy , Pregnancy Complications/blood , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy, Animal , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Tetrahydrofolates/blood
5.
Reumatol. clín. (Barc.) ; 13(6): 318-325, nov.-dic. 2017. tab, ilus
Article in Spanish | IBECS | ID: ibc-167205

ABSTRACT

Objetivo. Analizar el efecto de polimorfismos de nucleótido único (SNPs) de la metilen-tetrahidrofolatorreductasa (MTHFR; rs1801131 y rs1801133), el transportador de membrana que une ATP B1 (ABCB1; rs1045642), la aicartransformilasa/IMP ciclohidrolasa (ATIC; rs2372536) y la folilpoliglutamatosintetasa (FPGS; rs1544105) en la toxicidad hepática y medular de metotrexato (MTX). Pacientes y métodos. Se analizaron 1.415 visitas (732 con MTX, 683 sin MTX) de 350 pacientes del Princesa Early Arthritis Register Longytudinal study. El genotipo de los diferentes SNP se determinó mediante sondas TaqMan (Applied Biosystems). Se realizaron análisis multivariables mediante modelos lineales generalizados en los que las variables dependientes fueron los niveles séricos de transaminasa glutámico-pirúvica (toxicidad hepática), leucocitos, plaquetas o hemoglobina (toxicidad hematológica) y se ajustaron por variables clínicas (actividad de la enfermedad, etc.), analíticas (función renal, etc.), sociodemográficas (edad, sexo, etc.) y las variantes genéticas de MTHFR, ABCB1, ATIC y FPGS. También se analizaron las variables que influyeron en las dosis de MTX administradas a lo largo del seguimiento. Resultados. Cuando recibían MTX los portadores del genotipo CC del SNP rs1045642 de ABCB1 presentaron niveles significativamente mayores de GPT (7,1±2,0U/l; p<0,001). Los portadores de al menos un alelo G de rs1544105 en FPGS presentaron niveles significativamente menores de leucocitos (−0,67±0,32; 0,038), hemoglobina (−0,34±0,11g/dl; p=0,002) y de plaquetas (−11,8±4,7; p=0,012). La presencia del alelo G de rs1544105 (FPGS) y T de rs1801133 (MTHFR) se asoció, de forma aditiva y significativa, al uso de menores dosis de MTX. Discusión. Nuestros datos sugieren que variantes genéticas de las enzimas FGPS y MTHFR, y del transportador ABCB1, podrían ayudar a detectar pacientes con mayor riesgo de toxicidad por MTX (AU)


Objective. To analyze the effect of single nucleotide polymorphisms (SNPs) with well-known functional impact of methylenetetrahydrofolatereductase (MTHFR; rs1801131 and rs1801133), the membrane transporter ABCB1 (rs1045642), the AICAR transformylase/IMP cyclohydrolase (ATIC; rs2372536) and folyl-polyglutamatesynthetase (FPGS; rs1544105), on liver and bone marrow toxicity of methotrexate (MTX). Patients and methods. We analyzed 1415 visits from 350 patients of the PEARL (Princesa Early Arthritis Register Longitudinal) study: (732 with MTX, 683 without MTX). The different SNPs were genotyped using specific TaqMan probes (Applied Biosystems). Multivariate analyzes were performed using generalized linear models in which the dependent variables were the levels of serum alanine aminotransferase (liver toxicity), leukocytes, platelets or hemoglobin (hematologic toxicity) and adjusted for clinical variables (disease activity, etc.), analytical (renal function, etc.), sociodemographic (age, sex, etc.) and genetic variants of MTHFR, ABCB1, ATIC and FPGS. The effect of these variables on the MTX doses prescribed throughout follow-up was also analyzed through multivariate analysis nested by visit and patient. Results. When taking MTX, those patients carrying the CC genotype of rs1045642 in ABCB1 showed significantly higher GPT levels (7.1±2.0 U/L; P<.001). Carrying at least one G allele of rs1544105 in FPGS was associated with lower leukocyte (−0.67±0.32; 0.038), hemoglobin (−0.34±0.11g/dL; P=.002), and platelet (−11.8±4.7; P=.012) levels. The presence of the G allele of rs1544105 in FPGS, and the T allele of rs1801133 in MTHFR, was significantly associated with the use of lower doses of MTX. Discussion. Our data suggest that genotyping functional variants in FGPS and MTHFR enzymes and the transporter ABCB1 could help to identify patients with increased risk of MTX toxicity (AU)


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , ATP Binding Cassette Transporter 1/genetics , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methotrexate/administration & dosage , Polymorphism, Single Nucleotide , Polymorphism, Single Nucleotide/genetics , Multivariate Analysis , Linear Models , 28599
6.
Behav Brain Res ; 332: 71-74, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28559181

ABSTRACT

The MTHFD1 gene encodes for methylenetetrahydrofolate dehydrogenase 1, an enzyme that has an important role in folate-mediated one-carbon metabolism. In people, a single nucleotide polymorphism of this gene (1958G>A; rs2236225) is associated with increased risk for bipolar disorder and schizophrenia, neural tube and other birth defects. Mice homozygous for a loss of Mthfd1 via a gene-trap mutation are not viable, and heterozygotes, though they appear healthy, have metabolic imbalances in the folate- and choline-mediated 1-carbon metabolic pathways. In this study, we evaluated cognitive function in Mthfd1gt/+ male and female mice using a behavioral battery composed of eight different tests. We found that these mice display impaired cue-conditioned learning, while other behaviors remain intact.


Subject(s)
Formate-Tetrahydrofolate Ligase/deficiency , Learning Disabilities/enzymology , Methenyltetrahydrofolate Cyclohydrolase/deficiency , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Multifunctional Enzymes/deficiency , Animals , Body Weight , Cognition/physiology , Female , Formate-Tetrahydrofolate Ligase/genetics , Male , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice, Transgenic , Multifunctional Enzymes/genetics , Neuropsychological Tests , Phenotype
7.
Nat Commun ; 7: 11640, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27291711

ABSTRACT

DNA methylation is an epigenetic mechanism that has important functions in transcriptional silencing and is associated with repressive histone methylation (H3K9me). To further investigate silencing mechanisms, we screened a mutagenized Arabidopsis thaliana population for expression of SDCpro-GFP, redundantly controlled by DNA methyltransferases DRM2 and CMT3. Here, we identify the hypomorphic mutant mthfd1-1, carrying a mutation (R175Q) in the cytoplasmic bifunctional methylenetetrahydrofolate dehydrogenase/methenyltetrahydrofolate cyclohydrolase (MTHFD1). Decreased levels of oxidized tetrahydrofolates in mthfd1-1 and lethality of loss-of-function demonstrate the essential enzymatic role of MTHFD1 in Arabidopsis. Accumulation of homocysteine and S-adenosylhomocysteine, genome-wide DNA hypomethylation, loss of H3K9me and transposon derepression indicate that S-adenosylmethionine-dependent transmethylation is inhibited in mthfd1-1. Comparative analysis of DNA methylation revealed that the CMT3 and CMT2 pathways involving positive feedback with H3K9me are mostly affected. Our work highlights the sensitivity of epigenetic networks to one-carbon metabolism due to their common S-adenosylmethionine-dependent transmethylation and has implications for human MTHFD1-associated diseases.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/genetics , DNA Methylation/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Arabidopsis Proteins/genetics , Cytoplasm/drug effects , Cytoplasm/metabolism , DNA Demethylation , Epigenesis, Genetic , Folic Acid/metabolism , Gene Expression Regulation, Plant/drug effects , Gene Silencing , Green Fluorescent Proteins/metabolism , Histones/metabolism , Homeostasis/drug effects , Lysine/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methionine/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Models, Biological , Mutation/genetics , Protein Transport/drug effects , S-Adenosylmethionine/metabolism , Tetrahydrofolates/pharmacology
8.
J Exp Med ; 213(7): 1285-306, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27325891

ABSTRACT

Drugs targeting metabolism have formed the backbone of therapy for some cancers. We sought to identify new such targets in acute myeloid leukemia (AML). The one-carbon folate pathway, specifically methylenetetrahydrofolate dehydrogenase-cyclohydrolase 2 (MTHFD2), emerged as a top candidate in our analyses. MTHFD2 is the most differentially expressed metabolic enzyme in cancer versus normal cells. Knockdown of MTHFD2 in AML cells decreased growth, induced differentiation, and impaired colony formation in primary AML blasts. In human xenograft and MLL-AF9 mouse leukemia models, MTHFD2 suppression decreased leukemia burden and prolonged survival. Based upon primary patient AML data and functional genomic screening, we determined that FLT3-ITD is a biomarker of response to MTHFD2 suppression. Mechanistically, MYC regulates the expression of MTHFD2, and MTHFD2 knockdown suppresses the TCA cycle. This study supports the therapeutic targeting of MTHFD2 in AML.


Subject(s)
Citric Acid Cycle , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Leukemic , Leukemia, Myeloid, Acute/epidemiology , Methenyltetrahydrofolate Cyclohydrolase/biosynthesis , Proto-Oncogene Proteins c-myc/metabolism , Animals , Gene Knockdown Techniques , HL-60 Cells , Heterografts , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Methenyltetrahydrofolate Cyclohydrolase/genetics , Mice , Neoplasm Transplantation , Proto-Oncogene Proteins c-myc/genetics , U937 Cells
9.
Science ; 351(6274): 728-733, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26912861

ABSTRACT

In response to growth signals, mechanistic target of rapamycin complex 1 (mTORC1) stimulates anabolic processes underlying cell growth. We found that mTORC1 increases metabolic flux through the de novo purine synthesis pathway in various mouse and human cells, thereby influencing the nucleotide pool available for nucleic acid synthesis. mTORC1 had transcriptional effects on multiple enzymes contributing to purine synthesis, with expression of the mitochondrial tetrahydrofolate (mTHF) cycle enzyme methylenetetrahydrofolate dehydrogenase 2 (MTHFD2) being closely associated with mTORC1 signaling in both normal and cancer cells. MTHFD2 expression and purine synthesis were stimulated by activating transcription factor 4 (ATF4), which was activated by mTORC1 independent of its canonical induction downstream of eukaryotic initiation factor 2α eIF2α phosphorylation. Thus, mTORC1 stimulates the mTHF cycle, which contributes one-carbon units to enhance production of purine nucleotides in response to growth signals.


Subject(s)
Mitochondria/metabolism , Multiprotein Complexes/metabolism , Purines/biosynthesis , TOR Serine-Threonine Kinases/metabolism , Tetrahydrofolates/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Eukaryotic Initiation Factor-2/metabolism , HEK293 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Multiprotein Complexes/genetics , Phosphorylation , Protein Biosynthesis , TOR Serine-Threonine Kinases/genetics , Transcription, Genetic
10.
Microbiology (Reading) ; 162(1): 145-155, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26531681

ABSTRACT

Most organisms possess bifunctional FolD [5,10-methylenetetrahydrofolate (5,10-CH2-THF) dehydrogenase-cyclohydrolase] to generate NADPH and 10-formyltetrahdrofolate (10-CHO-THF) required in various metabolic steps. In addition, some organisms including Clostridium perfringens possess another protein, Fhs (formyltetrahydrofolate synthetase), to synthesize 10-CHO-THF. Here, we show that unlike the bifunctional FolD of Escherichia coli (EcoFolD), and contrary to its annotated bifunctional nature, C. perfringens FolD (CpeFolD) is a monofunctional 5,10-CH2-THF dehydrogenase. The dehydrogenase activity of CpeFolD is about five times more efficient than that of EcoFolD. The 5,10-methenyltetrahydrofolate (5,10-CH+-THF) cyclohydrolase activity in C. perfringens is provided by another protein, FchA (5,10-CH+-THF cyclohydrolase), whose cyclohydrolase activity is ∼ 10 times more efficient than that of EcoFolD. Kinetic parameters for CpeFhs were also determined for utilization of all of its substrates. Both CpeFolD and CpeFchA are required to substitute for the single bifunctional FolD in E. coli. The simultaneous presence of CpeFolD and CpeFchA is also necessary to rescue an E. coli folD deletion strain (harbouring CpeFhs support) for its formate and glycine auxotrophies, and to alleviate its susceptibility to trimethoprim (an antifolate drug) or UV light. The presence of the three clostridial proteins (FolD, FchA and Fhs) is required to maintain folate homeostasis in the cell.


Subject(s)
Clostridium perfringens/enzymology , Escherichia coli/enzymology , Formate-Tetrahydrofolate Ligase/metabolism , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Amino Acid Sequence , Clostridium perfringens/chemistry , Clostridium perfringens/genetics , Escherichia coli/chemistry , Escherichia coli/genetics , Formate-Tetrahydrofolate Ligase/chemistry , Formate-Tetrahydrofolate Ligase/genetics , Kinetics , Methenyltetrahydrofolate Cyclohydrolase/chemistry , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Molecular Sequence Data , Sequence Alignment
11.
Mol Biochem Parasitol ; 181(2): 178-85, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22108435

ABSTRACT

Three enzyme activities in the protozoan Leishmania major, namely N(5),N(10)-methylenetetrahydrofolate dehydrogenase/N(5),N(10)-methenyltetrahydrofolate cyclohydrolase (DHCH) and N(10)-formyltetrahydrofolate ligase (FTL) produce the essential intermediate N(10)-formyltetrahydrofolate. Although trypanosomatids possess at least one functional DHCH, the same is not true for FTL, which is absent in Trypanosoma brucei. Here, we present the 2.7 Å resolution crystal structure of the bifunctional apo-DHCH from L. major, which is a potential drug target. Sequence alignments show that the cytosolic enzymes found in trypanosomatids share a high level of identity of approximately 60%. Additionally, residues that interact and participate in catalysis in the human homologue are conserved amongst trypanosomatid sequences and this may complicate attempts to derive potent, parasite specific DHCH inhibitors.


Subject(s)
Leishmania major/enzymology , Methenyltetrahydrofolate Cyclohydrolase/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Protozoan Proteins/chemistry , Amino Acid Sequence , Antiprotozoal Agents/pharmacology , Enzyme Activation/drug effects , Humans , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Protozoan Proteins/genetics , Sequence Homology , Sequence Homology, Amino Acid
12.
Biochem Biophys Res Commun ; 406(3): 459-63, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21333632

ABSTRACT

Folate co-enzymes play a pivotal role in one-carbon transfer cellular processes. Many eukaryotes encode the tri-functional tetrahydrofolate dehydrogenase/cyclohydrolase/synthetase (deh/cyc/syn) enzyme, which consists of a N-terminal bifunctional domain (deh/cyc) and a C-terminal monofunctional domain (syn). Here, we report the first analogous archeal enzyme structures, for the bifunctional methylenetetrahydrofolate dehydrogenase/cyclohydrolase from Thermoplasma acidophilum (TaMTHFDC) as the native protein and also as its NADP complex. The TaMTHFDC structure is a dimer with a polar interface, as well as a NADP binding site that shows minor conformational change. The orientations of the residues in the NADP binding site do not change on ligand binding, incorporating three water molecules which are hydrogen bonded with phosphate groups of NADP in the structure of the complex. Our structural information will contribute to an improved understanding of the basis of THF and one-carbon metabolism.


Subject(s)
Methenyltetrahydrofolate Cyclohydrolase/chemistry , Thermoplasma/enzymology , Amino Acid Sequence , Binding Sites , Conserved Sequence , Crystallization , Crystallography, X-Ray , Methenyltetrahydrofolate Cyclohydrolase/genetics , Molecular Sequence Data , NADP/chemistry , Protein Multimerization , Protein Structure, Secondary , Tetrahydrofolates/chemistry
13.
Carcinogenesis ; 32(3): 427-33, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21156972

ABSTRACT

The causal metabolic pathways underlying associations between folate and risk for colorectal cancer (CRC) have yet to be established. Folate-mediated one-carbon metabolism is required for the de novo synthesis of purines, thymidylate and methionine. Methionine is converted to S-adenosylmethionine (AdoMet), the major one-carbon donor for cellular methylation reactions. Impairments in folate metabolism can modify DNA synthesis, genomic stability and gene expression, characteristics associated with tumorigenesis. The Mthfd1 gene product, C1-tetrahydrofolate synthase, is a trifunctional enzyme that generates one-carbon substituted tetrahydrofolate cofactors for one-carbon metabolism. In this study, we use Mthfd1(gt/+) mice, which demonstrate a 50% reduction in C1-tetrahydrofolate synthase, to determine its influence on tumor development in two mouse models of intestinal cancer, crosses between Mthfd1(gt/+) and Apc(min)(/+) mice and azoxymethane (AOM)-induced colon cancer in Mthfd1(gt/+) mice. Mthfd1 hemizygosity did not affect colon tumor incidence, number or load in Apc(min/+) mice. However, Mthfd1 deficiency increased tumor incidence 2.5-fold, tumor number 3.5-fold and tumor load 2-fold in AOM-treated mice. DNA uracil content in the colon was lower in Mthfd1(gt/+) mice, indicating that thymidylate biosynthesis capacity does not play a significant role in AOM-induced colon tumorigenesis. Mthfd1 deficiency-modified cellular methylation potential, as indicated by the AdoMet: S-adenosylhomocysteine ratio and gene expression profiles, suggesting that changes in the transcriptome and/or decreased de novo purine biosynthesis and associated mutability cause cellular transformation in the AOM CRC model. This study emphasizes the impact and complexity of gene-nutrient interactions with respect to the relationships among folate metabolism and colon cancer initiation and progression.


Subject(s)
Aminohydrolases/physiology , Colonic Neoplasms/genetics , DNA, Neoplasm/metabolism , Formate-Tetrahydrofolate Ligase/physiology , Methenyltetrahydrofolate Cyclohydrolase/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/physiology , Multienzyme Complexes/physiology , Multifunctional Enzymes/physiology , S-Adenosylhomocysteine/metabolism , S-Adenosylmethionine/metabolism , Aminohydrolases/genetics , Animals , Apoptosis , Azoxymethane/toxicity , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinogens/toxicity , Cell Proliferation , Colonic Neoplasms/chemically induced , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Female , Formate-Tetrahydrofolate Ligase/genetics , Gene Expression Profiling , Immunoenzyme Techniques , Male , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes/genetics , Multifunctional Enzymes/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Uracil/metabolism
14.
Mol Microbiol ; 71(6): 1386-401, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19183277

ABSTRACT

10-Formyl tetrahydrofolate (10-CHO-THF) is a key metabolite in C1 carbon metabolism, arising through the action of formate-tetrahydrofolate ligase (FTL) and/or 5,10-methenyltetrahydrofolate cyclohydrolase/5,10-methylene tetrahydrofolate dehydrogenase (DHCH). Leishmania major possesses single DHCH1 and FTL genes encoding exclusively cytosolic proteins, unlike other organisms where isoforms occur in the mitochondrion as well. Recombinant DHCH1 showed typical NADP(+)-dependent methylene tetrahydrofolate DH and 5,10-methenyltetrahydrofolate CH activities, and the DH activity was potently inhibited by a substrate analogue 5,10-CO-THF (K(i) 105 nM), as was Leishmania growth (EC(50) 1.1 microM). Previous studies showed null ftl(-) mutants were normal, raising the possibility that loss of the purine synthetic pathway had rendered 10-CHO-THF dispensable in evolution. We were unable to generate dhch1(-) null mutants by gene replacement, despite using a wide spectrum of nutritional supplements expected to bypass DHCH function. We applied an improved method for testing essential genes in Leishmania, based on segregational loss of episomal complementing genes rather than transfection; analysis of approximately 1400 events without successful loss of DHCH1 again established its requirement. Lastly, we employed 'genetic metabolite complementation' using ectopically expressed FTL as an alternative source of 10-CHO-THF; now dhch1(-) null parasites were readily obtained. These data establish a requirement for 10-CHO-THF metabolism in L. major, and provide genetic and pharmacological validation of DHCH as a target for chemotherapy, in this and potentially other protozoan parasites.


Subject(s)
Leishmania major/enzymology , Leucovorin/analogs & derivatives , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Animals , Cloning, Molecular , Folic Acid Antagonists/pharmacology , Gene Knockout Techniques , Genes, Essential , Genes, Protozoan , Leishmania major/drug effects , Leishmania major/genetics , Leucovorin/metabolism , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mutation , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
15.
Antimicrob Agents Chemother ; 53(4): 1673-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19171795

ABSTRACT

Bacillus subtilis mutants with resistance against peptide deformylase inhibitors were isolated. All showed a bypass of the pathway through mutations in three genes required for formylation of Met-tRNA(fMet), fmt, folD, and glyA. glyA corresponds to a yet uncharacterized locus inducing resistance. The bypass of formylation caused robust fitness reduction but was not accompanied by alterations of the transcription profile. A subtle adaptation of the enzymes of the intermediary metabolism was observed.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bacillus subtilis/drug effects , Enzyme Inhibitors/pharmacology , Mutation , Bacillus subtilis/genetics , Drug Resistance, Bacterial , Glycine Hydroxymethyltransferase/genetics , Hydroxamic Acids/pharmacology , Methenyltetrahydrofolate Cyclohydrolase/genetics , Polymerase Chain Reaction , RNA, Transfer, Met/genetics
16.
Hum Mutat ; 30(2): 212-20, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18767138

ABSTRACT

Methylenetetrahydrofolate dehydrogenase)methenyltetrahydrofolate cyclohydrolase)formyltetrahydrofolate synthetase (MTHFD1) is a trifunctional enzyme that interconverts tetrahydrofolate (THF) derivatives for nucleotide synthesis. A common variant in MTHFD1, p.Arg653Gln (c.1958G>A), may increase the risk for neural tube defects (NTD). To examine the biological impact of this variant on MTHFD1 function, we measured enzyme activity and stability in vitro and assessed substrate flux in transfected mammalian cells. The purified Arg653Gln enzyme has normal substrate affinity but a 36% reduction in half)life at 42 degrees C. Thermolability is reduced by magnesium adenosine triphosphate and eliminated by the substrate analog folate pentaglutamate, suggesting that folate status may modulate impact of the variant. The mutation reduces the metabolic activity of MTHFD1 within cells: formate incorporation into DNA in murine Mthfd1 knockout cells transfected with Arg653Gln is reduced by 26%+/-7.7% (P<0.05), compared to cells transfected with wild)type protein, indicating a disruption of de novo purine synthesis. We assessed the impact of the variant on risk for congenital heart defects (CHD) in a cohort of Quebec children (158 cases, 110 controls) and mothers of children with heart defects (199 cases, 105 controls). The 653QQ genotype in children is associated with increased risk for heart defects (odds ratio [OR], 2.11; 95% confidence interval [CI], 1.01-4.42), particularly Tetralogy of Fallot (OR, 3.60; 95% CI, 1.38-9.42) and aortic stenosis (OR, 3.13; 95% CI, 1.13-8.66). There was no effect of maternal genotype. Our results indicate that the Arg653Gln polymorphism decreases enzyme stability and increases risk for CHD. Further evaluation of this polymorphism in folate)related disorders and its potential interaction with folate status is warranted.


Subject(s)
Amino Acid Substitution , Genetic Predisposition to Disease , Heart Defects, Congenital/enzymology , Heart Defects, Congenital/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mutation/genetics , Adolescent , Animals , Case-Control Studies , Coenzymes , Enzyme Inhibitors , Enzyme Stability , Female , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/isolation & purification , Formate-Tetrahydrofolate Ligase/metabolism , Gene Frequency , Humans , Kinetics , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methenyltetrahydrofolate Cyclohydrolase/isolation & purification , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/isolation & purification , Mice , Minor Histocompatibility Antigens , Polymorphism, Genetic , Structural Homology, Protein , Substrate Specificity , Temperature
18.
Brain Res Mol Brain Res ; 139(1): 163-8, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-15953655

ABSTRACT

There is growing evidence that folate metabolism is involved in migraine pathophysiology, mainly in migraine with aura. Even though folate metabolism is regulated by a number of enzymes, only two functional polymorphisms have been tested in association studies with migraine. Here, we have explored the possible role in migraine of other folate-metabolizing enzymes which are in close interdependency with 5',10'-methylenetetrahydrofolate reductase analyzing functional polymorphisms of these enzymes in a case-control study. Individually, thymidylate synthase (TS), methenyltetrahydrofolate cyclohydrolase formyltetrahydrofolate synthase (MTHFD1), or methionine synthase (MS) polymorphisms did not modify the general risk for suffering migraine. Nevertheless, we observed a strong interaction between TS and MTHFR mutated genotypes, which increased over 8-fold the risk for experiencing aura among migraineurs; MTHFD1 and MTHFR mutated genotypes also increased together the risk for migraine in general (OR = 3.08; 95% CI = 1.3-7.4). We conclude that the pathogenetic role of the MTHFR T677 allele in migraine is modulated by functional polymorphisms of TS and MTHFD1.


Subject(s)
Genetic Predisposition to Disease , Migraine Disorders/genetics , Polymorphism, Genetic , Promoter Regions, Genetic , Tandem Repeat Sequences , Thymidylate Synthase/genetics , Adult , Case-Control Studies , Female , Folic Acid/metabolism , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/metabolism , Humans , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Middle Aged , Migraine Disorders/physiopathology , Risk Factors , Thymidylate Synthase/metabolism
19.
Microbiology (Reading) ; 150(Pt 1): 9-19, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14702393

ABSTRACT

Over the past few years, the genetic 'toolkit' available for use with Methylobacterium extorquens AM1 has expanded significantly. Here a further advance is presented and demonstrated, an insertional expression system that allows expression of genes from a stable, unmarked chromosomal locus. This system has been used to better understand the role of the tetrahydrofolate (H4F) pathway in methylotrophy. Previously, it has not been possible to generate null mutants lacking either mtdA (encoding an NADP-dependent methylene-H4F/methylene-tetrahydromethanopterin dehydrogenase) or fch (encoding methenyl-H4F cyclohydrolase). An unmarked strain was generated that expressed the analogous folD gene (encoding a bifunctional NADP-dependent methylene-H4F dehydrogenase/methenyl-H4F cyclohydrolase) from Methylobacterium chloromethanicum CM4T. In this strain, null mutants could be obtained that grew normally on multicarbon substrates but were defective for growth on C1 substrates. Additionally, null mutants of mtdA and/or fch could also be generated in the wild-type by supplementing the succinate medium with formate. These strains were unable to grow on C1 compounds but were not methanol-sensitive. These approaches have demonstrated that the apparent essentiality of mtdA and fch is due to the need for formyl-H4F for biosynthesis of purines and other compounds, and have provided clear genetic evidence that the H4F pathway is required for methylotrophy.


Subject(s)
Methylobacterium extorquens/genetics , Formates/metabolism , Genes, Bacterial , Genetic Vectors , Methenyltetrahydrofolate Cyclohydrolase/genetics , Methylobacterium/enzymology , Methylobacterium/genetics , Methylobacterium extorquens/growth & development , Methylobacterium extorquens/metabolism , Molecular Sequence Data , Mutagenesis, Insertional , Mutation , Oxidoreductases Acting on CH-NH Group Donors/genetics , Phenotype , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...