Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 22(3): 1464-8, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22177783

ABSTRACT

Novel deazaxanthine-based DPP-4 inhibitors have been identified that are potent (IC(50) <10nM) and highly selective versus other dipeptidyl peptidases. Their synthesis and SAR are reported, along with initial efforts to improve the PK profile through decoration of the deazaxanthine core. Optimisation of compound 3a resulted in the identification of compound (S)-4i, which displayed an improved in vitro and ADME profile. Further enhancements to the PK profile were possible by changing from the deazahypoxanthine to the deazaxanthine template, culminating in compound 12g, which displayed good ex vivo DPP-4 inhibition and a superior PK profile in rat, suggestive of once daily dosing in man.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/chemistry , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Animals , Caco-2 Cells , Crystallography, X-Ray , Dipeptidyl-Peptidase IV Inhibitors/chemical synthesis , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Enzyme Activation/drug effects , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Humans , Inhibitory Concentration 50 , Male , Models, Molecular , Molecular Structure , Rats , Structure-Activity Relationship
2.
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
3.
Vitam Horm ; 79: 393-410, 2008.
Article in English | MEDLINE | ID: mdl-18804703

ABSTRACT

Folate-mediated metabolism involves enzyme-catalyzed reactions that occur in the cytoplasmic, mitochondrial, and nuclear compartments in mammalian cells. Which of the folate-dependent enzymes are expressed in these compartments depends on the stage of development, cell type, cell cycle, and whether or not the cell is transformed. Mitochondria become formate-generating organelles in cells and tissues expressing the MTHFD2 and MTHFD1L genes. The products of these nuclear genes were derived from trifunctional precursor proteins, expressing methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase, and formyltetrahydrofolate synthetase activities. The MTHFD2 protein is a bifunctional protein with dehydrogenase and cyclohydrolase activities that arose from a trifunctional precursor through the loss of the synthetase domain and a novel adaptation to NAD rather than NADP specificity for the dehydrogenase. The MTHFD1L protein retains the size of its trifunctional precursor, but through the mutation of critical residues, both the dehydrogenase and cyclohydrolase activities have been silenced. MTHFD1L is thus a monofunctional formyltetrahydrofolate synthetase. This review discusses the properties and functions of these mitochondrial proteins and their role in supporting cytosolic purine synthesis during embryonic development and in cells undergoing rapid growth.


Subject(s)
Formate-Tetrahydrofolate Ligase/metabolism , Methenyltetrahydrofolate Cyclohydrolase/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/enzymology , Animals , Mammals , Saccharomyces cerevisiae/metabolism
4.
Bioessays ; 28(6): 595-605, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16700064

ABSTRACT

In eukaryotes, folate metabolism is compartmentalized between the cytoplasm and organelles. The folate pathways of mitochondria are adapted to serve the metabolism of the organism. In yeast, mitochondria support cytoplasmic purine synthesis through the generation of formate. This pathway is important but not essential for survival, consistent with the flexibility of yeast metabolism. In plants, the mitochondrial pathways support photorespiration by generating serine from glycine. This pathway is essential under photosynthetic conditions and the enzyme expression varies with photosynthetic activity. In mammals, the expression of the mitochondrial enzymes varies in tissues and during development. In embryos, mitochondria supply formate and glycine for purine synthesis, a process essential for survival; in adult tissues, flux through mitochondria can favor serine production. The differences in the folate pathways of mitochondria depending on species, tissues and developmental stages, profoundly alter the nature of their metabolic contribution.


Subject(s)
Carbon/metabolism , Mitochondria/metabolism , Animals , Carbon/chemistry , Humans , Mammals , Plants , Saccharomyces cerevisiae
5.
Arch Biochem Biophys ; 442(1): 133-9, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16150419

ABSTRACT

Mouse fibroblasts in which the mthfd2 gene encoding mitochondrial NAD-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase (NMDMC) was previously inactivated were infected with retroviral expression constructs of dehydrogenase/cyclohydrolase cDNA. Cellular fractionation confirmed that the expressed proteins were properly targeted to the mitochondria. Expression of the NAD-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase enzyme in mitochondria corrected the glycine auxotrophy of the null mutant cells. A construct in which the cyclohydrolase activity of NMDMC was inactivated by point mutation also rescued the glycine auxotrophy, although poorly. This suggests that the cyclohydrolase activity is also required to ensure optimal production of 10-formyltetrahydrofolate. The expression of the NADP-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase-synthetase in the mitochondria also reversed the glycine requirement of the null cells demonstrating that the use of the NAD cofactor is not absolutely essential to maintain the flux of one-carbon metabolites. All rescued cells demonstrated a decrease in the ratio of incorporation of exogenous formate to serine in standardized radiolabeling studies. This ratio, which is approximately 2.5 for nmdmc(-/-) cells and 0.3 for the wild type cells under the conditions used, is a qualitative indicator of the ability of the mitochondria of the cells to generate formate.


Subject(s)
Fibroblasts/enzymology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/enzymology , NADP/metabolism , NAD/metabolism , Aminohydrolases/metabolism , Animals , Blotting, Western , Carbon Radioisotopes , Cell Line , DNA, Complementary/metabolism , Embryonic Development , Glycine/metabolism , Kinetics , Leucovorin/analogs & derivatives , Leucovorin/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mutation
6.
J Biol Chem ; 280(40): 34316-23, 2005 Oct 07.
Article in English | MEDLINE | ID: mdl-16100107

ABSTRACT

The mitochondrial NAD-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase (NMDMC) is believed to have evolved from a trifunctional NADP-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase-synthetase. It is unique in its absolute requirement for inorganic phosphate and magnesium ions to support dehydrogenase activity. To enable us to investigate the roles of these ions, a homology model of human NMDMC was constructed based on the structures of three homologous proteins. The model supports the hypothesis that the absolutely required Pi can bind in close proximity to the 2'-hydroxyl of NAD through interactions with Arg166 and Arg198. The characterization of mutants of Arg166, Asp190, and Arg198 show that Arg166 is primarily responsible for Pi binding, while Arg198 plays a secondary role, assisting in binding and properly orienting the ion in the cofactor binding site. Asp190 helps to properly position Arg166. Mutants of Asp133 suggest that the magnesium ion interacts with both Pi and the aspartate side chain and plays a role in positioning Pi and NAD. NMDMC uses Pi and magnesium to adapt an NADP binding site for NAD binding. This adaptation represents a novel variation of the classic Rossmann fold.


Subject(s)
Aminohydrolases/chemistry , Aminohydrolases/metabolism , Magnesium/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , NAD/metabolism , Phosphates/physiology , Amino Acid Sequence , Arginine , Binding Sites , Coenzymes , Escherichia coli/enzymology , Humans , Ions , Magnesium/chemistry , Models, Molecular , Molecular Sequence Data , Phosphates/chemistry , Protein Folding , Protein Structure, Tertiary , Saccharomyces cerevisiae/enzymology , Sequence Homology
7.
J Biol Chem ; 280(9): 7597-602, 2005 Mar 04.
Article in English | MEDLINE | ID: mdl-15611115

ABSTRACT

The Mthfd1 gene encoding the cytoplasmic methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase-formyltetrahydrofolate synthetase enzyme (DCS) was inactivated in embryonic stem cells. The null embryonic stem cells were used to generate spontaneously immortalized fibroblast cell lines that exhibit the expected purine auxotrophy. Elimination of these cytoplasmic activities allowed for the accurate assessment of similar activities encoded by other genes in these cells. A low level of 10-formyltetrahydrofolate synthetase was detected and was shown to be localized to mitochondria. However, NADP-dependent methylenetetrahydrofolate dehydrogenase activity was not detected. Northern blot analysis suggests that a recently identified mitochondrial DCS (Prasannan, P., Pike, S., Peng, K., Shane, B., and Appling, D. R. (2003) J. Biol. Chem. 278, 43178-43187) is responsible for the synthetase activity. The lack of NADP-dependent dehydrogenase activity suggests that this RNA may encode a monofunctional synthetase. Moreover, examination of the primary structure of this novel protein revealed mutations in key residues required for dehydrogenase and cyclohydrolase activities. This monofunctional synthetase completes the pathway for the production of formate from formyltetrahydrofolate in the mitochondria in our model of mammalian one-carbon folate metabolism in embryonic and transformed cells.


Subject(s)
Embryo, Mammalian/cytology , Fibroblasts/cytology , Formate-Tetrahydrofolate Ligase/genetics , Formate-Tetrahydrofolate Ligase/physiology , Methylenetetrahydrofolate Dehydrogenase (NADP)/physiology , Mitochondria/enzymology , Stem Cells/cytology , Amino Acid Sequence , Animals , Blotting, Northern , Cell Line , Cell Line, Transformed , DNA, Complementary/metabolism , Databases as Topic , Exons , Fibroblasts/metabolism , Genotype , Heterozygote , Homozygote , Humans , Methenyltetrahydrofolate Cyclohydrolase/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Models, Biological , Models, Genetic , Molecular Sequence Data , Mutation , RNA/chemistry , Sequence Homology, Amino Acid , Software , Time Factors
8.
Biochim Biophys Acta ; 1674(1): 78-84, 2004 Sep 06.
Article in English | MEDLINE | ID: mdl-15342116

ABSTRACT

Deletion of the gene encoding NAD-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase (NMDMC) in mice was demonstrated previously to result in failure to establish definitive erythropoiesis in the developing liver. We examined the expression pattern of nmdmc to look for evidence that would support a tissue specific role for this activity. However, whole mount in situ hybridization revealed ubiquitous expression of nmdmc in the tissues of E9.5 and E10.5 embryos suggesting a broader role. Analysis of chimeras demonstrated that nmdmc-/- cells can survive in liver and other tissues of chimeras establishing that the null defect can be rescued by metabolites supplied by surrounding normal cells. Both the expression pattern and metabolite rescue support the proposal that mitochondrial NMDMC provides one-carbon units for purine synthesis during embryogenesis. The elevated expression of NMDMC in tumour cells, but not in surrounding normal cells, is predicted to result in significant differences in folate-mediated support for purine synthesis in the two cell types.


Subject(s)
Aminohydrolases/metabolism , Cell Division/physiology , Embryo, Mammalian/enzymology , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Mitochondria/enzymology , Multienzyme Complexes/metabolism , Aminohydrolases/genetics , Animals , Chimera , Embryo, Mammalian/physiology , Female , In Situ Hybridization , Liver/cytology , Liver/metabolism , Male , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Multienzyme Complexes/genetics , Pregnancy , Purines/metabolism , Tissue Distribution
9.
Hum Mutat ; 22(1): 67-73, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12815595

ABSTRACT

Glutamate formiminotransferase deficiency, an autosomal recessive disorder and the second most common inborn error of folate metabolism, is presumed to be due to defects in the bifunctional enzyme glutamate formiminotransferase-cyclodeaminase (FTCD). Features of a severe phenotype, first identified in patients of Japanese descent, include elevated levels of formiminoglutamate (FIGLU) in the urine in response to histidine administration, megaloblastic anemia, and mental retardation. Features of a mild phenotype include high urinary excretion of FIGLU in the absence of histidine administration, mild developmental delay, and no hematological abnormalities. We found mutations in the human FTCD gene in three patients with putative glutamate formiminotransferase deficiency. Two siblings were heterozygous for missense mutations, c.457C>T (R135C) and c.940G>C (R299P). Mutagenesis of porcine FTCD and expression in E. coli showed that the R135C mutation reduced formiminotransferase activity to 61% of wild-type, whereas the R299P mutation reduced this activity to 57% of wild-type. The third patient was hemizygous for c.1033insG, with quantitative PCR indicating that the other allele contained a deletion. These mutations are the first identified in glutamate formiminotransferase deficiency and demonstrate that mutations in FTCD represent the molecular basis for the mild phenotype of this disease.


Subject(s)
Ammonia-Lyases/deficiency , Ammonia-Lyases/genetics , Hydroxymethyl and Formyl Transferases/deficiency , Hydroxymethyl and Formyl Transferases/genetics , Animals , Cell Line , Fibroblasts/chemistry , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Enzymologic/genetics , Glutamate Formimidoyltransferase , Humans , Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Multienzyme Complexes , Multifunctional Enzymes , Mutagenesis, Site-Directed/genetics , Mutation, Missense , Polymerase Chain Reaction/methods , Sequence Analysis, DNA/methods , Swine
10.
Biochem J ; 374(Pt 3): 773-8, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-12793858

ABSTRACT

The 5,10-methenyltetrahydrofolate (5,10-CH=H4folate) synthetase catalyses the physiologically irreversible formation of 5,10-CH=H4folate from 5-formyltetrahydrofolate (5-HCO-H4folate) and ATP. It is not clear how (or if) 5-HCO-H4folate is formed in vivo. Using a spectrophotometric assay for 5-HCO-H4folate, human recombinant 5,10-CH=H4folate cyclohydrolase, which catalyses the hydrolysis of 5,10-CH=H4folate to 10-HCO-H4folate, was previously shown to catalyse inefficiently the formation of 5-HCO-H4folate at pH 7.3 [Pelletier and MacKenzie (1996) Bioorg. Chem. 24, 220-228]. In the present study, we report that (i) the human cyclohydrolase enzyme catalyses the conversion of 10-HCO-/5,10-CH=H4folate into 5-HCO-H4folate (it is also chemically formed) at pH 4.0-7.0; (ii) rat liver has a very low capacity to catalyse the formation of 5-HCO-H4folate when compared with the traditional activity of 5,10-CH=H4folate cyclohydrolase and the activity of the 5,10-CH=H4folate synthetase; and (iii) a substantial amount of 5-HCO-H4folate reported to be present in rat liver is chemically formed during analytical procedures. We conclude that (i) the cyclohydrolase represents some of the capacity of rat liver to catalyse the formation of 5-HCO-H4folate; (ii) the amount of 5-HCO-H4folate reported to be present in rat liver is overestimated (liver 5-HCO-H4folate content may be negligible); and (iii) there is little evidence that 5-HCO-H4folate inhibits one-carbon metabolism in mammals.


Subject(s)
Aminohydrolases/chemistry , Leucovorin/biosynthesis , Liver/enzymology , Animals , Carbon-Nitrogen Ligases/chemistry , Catalysis , Humans , Leucovorin/metabolism , Methenyltetrahydrofolate Cyclohydrolase , Rats , Recombinant Proteins/chemistry
11.
J Biol Chem ; 278(21): 19436-41, 2003 May 23.
Article in English | MEDLINE | ID: mdl-12646567

ABSTRACT

Primary fibroblasts established from embryos of NAD-dependent mitochondrial methylenetetrahydrofolate dehydrogenase-cyclohydrolase (NMDMC) knockout mice were spontaneously immortalized or transformed with SV40 Large T antigen. Mitotracker Red CMXRos staining of the cells indicates the presence of intact mitochondria with a membrane potential. The nmdmc(-/-) cells are auxotrophic for glycine, demonstrating that NMDMC is the only methylenetetrahydrofolate dehydrogenase normally expressed in the mitochondria of these cell lines. Growth of null mutant but not wild type cells on complete medium with dialyzed serum is stimulated about 2-fold by added formate or hypoxanthine. Radiolabeling experiments demonstrated a 3-10 x enhanced incorporation of radioactivity into DNA from formate relative to serine by nmdmc(-/-) cells. The generation of one-carbon units by mitochondria in nmdmc(-/-) cells is completely blocked, and the cytoplasmic folate pathways alone are insufficient for optimal purine synthesis. The results demonstrate a metabolic role for NMDMC in supporting purine biosynthesis. Despite the recognition of these metabolic defects in the mutant cell lines, the phenotype of nmdmc(-/-) embryos that begin to die at E13.5 is not improved when pregnant dams are given a glycine-rich diet or daily injections of sodium formate.


Subject(s)
Aminohydrolases/deficiency , Cell Division , Fibroblasts/enzymology , Fibroblasts/ultrastructure , Glycine/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/deficiency , Mitochondria/enzymology , Multienzyme Complexes/deficiency , Aminohydrolases/genetics , Aminohydrolases/physiology , Animals , Blotting, Northern , Blotting, Southern , Carbon Radioisotopes , Cell Line, Transformed , Culture Media , DNA/metabolism , Diet , Dietary Supplements , Embryo, Mammalian , Female , Fibroblasts/metabolism , Formates/administration & dosage , Formates/pharmacology , Genotype , Glycine/administration & dosage , Hypoxanthine/pharmacology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/physiology , Mice , Mice, Knockout , Multienzyme Complexes/genetics , Multienzyme Complexes/physiology , Pregnancy , RNA, Messenger/analysis
12.
Arch Biochem Biophys ; 403(1): 145-8, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12061812

ABSTRACT

We have isolated the cDNA and the gene encoding the murine cytoplasmic methylenetetrahydrofolate dehydrogenase-methenyltetrahydrofolate cyclohydrolase-formyltetrahydrofolate synthetase (DCS). Comparison of these sequences with the 3'-untranslated region of the mitochondrial NAD(+)-dependent methylenetetrahydrofolate dehydrogenase-cyclohydrolase (mt-DC) revealed areas of significant homology. Both exon and intron sequences of the synthetase domain of DCS are homologous to sequences in the untranslated region of mt-DC. A similar comparison between the mt-DC and the DCS sequences of humans as well as Drosophila supports the conclusion that in higher eukaryotes the bifunctional mt-DC replaced a trifunctional precursor through inactivation of the synthetase domain. The mt-DC should be considered in models of one-carbon folate fluxes in mammals.


Subject(s)
Aminohydrolases/chemistry , Aminohydrolases/genetics , Formate-Tetrahydrofolate Ligase/genetics , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , Mitochondria/enzymology , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , 3' Untranslated Regions , Animals , Base Sequence , Blotting, Southern , DNA, Complementary , Exons , Folic Acid/metabolism , Introns , Mice , Models, Genetic , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Homology, Nucleic Acid
13.
J Biol Chem ; 277(21): 18703-9, 2002 May 24.
Article in English | MEDLINE | ID: mdl-11904299

ABSTRACT

The human bifunctional dehydrogenase-cyclohydrolase domain catalyzes the interconversion of 5,10-methylene-H(4)folate and 10-formyl-H(4)folate. Although previous structure and mutagenesis studies indicated the importance of lysine 56 in cyclohydrolase catalysis, the role of several surrounding residues had not been explored. In addition to further defining the role of lysine 56, the work presented in this study explores the functions of glutamine 100 and aspartate 125 through the use of site-directed mutagenesis and chemical modification. Mutants at position 100 are inactive with respect to cyclohydrolase activity while preserving significant dehydrogenase levels. We succeeded in producing a K56Q/Q100K double mutant, which has no cyclohydrolase yet retains more than two-thirds of wild type dehydrogenase activity. Neither activity is detectable in aspartate 125 mutants with the exception of D125E. The results indicate that the function of glutamine 100 is to activate lysine 56 for cyclohydrolase catalysis and that aspartate 125 is involved in the binding of the H(4)folate substrates. In highlighting the importance of these residues, catalytic mechanisms are proposed for both activities as well as an explanation for the differences in channeling efficiency in the forward and reverse directions.


Subject(s)
Aspartic Acid/metabolism , Glutamine/metabolism , Methylenetetrahydrofolate Dehydrogenase (NADP)/metabolism , Catalysis , Methylenetetrahydrofolate Dehydrogenase (NADP)/chemistry , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...