Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 150
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochemistry ; 61(19): 2159-2164, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36126313

ABSTRACT

4-Formylaminooxyvinylglycine (FVG) is an herbicidal and antibacterial nonproteinogenic amino acid produced by several strains of the Pseudomonas fluorescens species complex. It contains a unique vinyl alkoxyamine moiety with an O-N bond, and its biosynthetic origin remains unknown. Here, we show that the gvg cluster from P. fluorescens WH6 is responsible for the biosynthesis of FVG and two additional O-N bond-containing oxyvinylglycines, guanidinooxyvinylglycine and aminooxyvinylglycine. Feeding studies in the producing bacteria indicate that these compounds originate from homoserine. We identify a formyltransferase gvgI that is required for the production of FVG and characterize the activity of this enzyme in vitro toward amino acids with a side chain amine. Sequence similarity network analysis reveals that GvgI and homologues make up a distinct group from the main classes of formyltransferases.


Subject(s)
Hydroxymethyl and Formyl Transferases , Pseudomonas fluorescens , Amines/metabolism , Amino Acids/metabolism , Anti-Bacterial Agents/metabolism , Glycine , Homoserine , Hydroxymethyl and Formyl Transferases/metabolism
2.
J Biol Chem ; 296: 100682, 2021.
Article in English | MEDLINE | ID: mdl-33894199

ABSTRACT

Methylofuran (MYFR) is a formyl-carrying coenzyme essential for the oxidation of formaldehyde in most methylotrophic bacteria. In Methylorubrum extorquens, MYFR contains a large and branched polyglutamate side chain of up to 24 glutamates. These glutamates play an essential role in interfacing the coenzyme with the formyltransferase/hydrolase complex, an enzyme that generates formate. To date, MYFR has not been identified in other methylotrophs, and it is unknown whether its structural features are conserved. Here, we examined nine bacterial strains for the presence and structure of MYFR using high-resolution liquid chromatography-mass spectrometry (LC-MS). Two of the strains produced MYFR as present in M. extorquens, while a modified MYFR containing tyramine instead of tyrosine in its core structure was detected in six strains. When M. extorquens was grown in the presence of tyramine, the compound was readily incorporated into MYFR, indicating that the biosynthetic enzymes are unable to discriminate tyrosine from tyramine. Using gene deletions in combination with LC-MS analyses, we identified three genes, orf5, orfY, and orf17 that are essential for MYFR biosynthesis. Notably, the orfY and orf5 mutants accumulated short MYFR intermediates with only one and two glutamates, respectively, suggesting that these enzymes catalyze glutamate addition. Upon homologous overexpression of orf5, a drastic increase in the number of glutamates in MYFR was observed (up to 40 glutamates), further corroborating the function of Orf5 as a glutamate ligase. We thus renamed OrfY and Orf5 to MyfA and MyfB to highlight that these enzymes are specifically involved in MYFR biosynthesis.


Subject(s)
Coenzymes/chemistry , Coenzymes/metabolism , Furans/chemistry , Furans/metabolism , Polyglutamic Acid/biosynthesis , Polyglutamic Acid/chemistry , Formaldehyde/metabolism , Glutamic Acid/metabolism , Hydrolases/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Methylobacterium extorquens/enzymology
3.
Nature ; 529(7585): 239-42, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26762462

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) are very large proteins that produce small peptide molecules with wide-ranging biological activities, including environmentally friendly chemicals and many widely used therapeutics. NRPSs are macromolecular machines, with modular assembly-line logic, a complex catalytic cycle, moving parts and many active sites. In addition to the core domains required to link the substrates, they often include specialized tailoring domains, which introduce chemical modifications and allow the product to access a large expanse of chemical space. It is still unknown how the NRPS tailoring domains are structurally accommodated into megaenzymes or how they have adapted to function in nonribosomal peptide synthesis. Here we present a series of crystal structures of the initiation module of an antibiotic-producing NRPS, linear gramicidin synthetase. This module includes the specialized tailoring formylation domain, and states are captured that represent every major step of the assembly-line synthesis in the initiation module. The transitions between conformations are large in scale, with both the peptidyl carrier protein domain and the adenylation subdomain undergoing huge movements to transport substrate between distal active sites. The structures highlight the great versatility of NRPSs, as small domains repurpose and recycle their limited interfaces to interact with their various binding partners. Understanding tailoring domains is important if NRPSs are to be utilized in the production of novel therapeutics.


Subject(s)
Biocatalysis , Brevibacillus/enzymology , Gramicidin/biosynthesis , Peptide Synthases/chemistry , Peptide Synthases/metabolism , Amino Acid Isomerases/chemistry , Amino Acid Isomerases/metabolism , Anti-Bacterial Agents/biosynthesis , Binding Sites , Carbohydrate Metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Catalytic Domain , Coenzymes/metabolism , Crystallography, X-Ray , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/metabolism , Models, Molecular , Multienzyme Complexes/chemistry , Multienzyme Complexes/metabolism , Pantetheine/analogs & derivatives , Pantetheine/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Transfer/chemistry , RNA, Transfer/metabolism
4.
Proc Natl Acad Sci U S A ; 116(12): 5487-5492, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30819897

ABSTRACT

The smallest histone deacetylase (HDAC) and the only class IV HDAC member, HDAC11, is reported to regulate immune activation and tumorigenesis, yet its biochemical function is largely unknown. Here we identify HDAC11 as an efficient lysine defatty-acylase that is >10,000-fold more efficient than its deacetylase activity. Through proteomics studies, we hypothesized and later biochemically validated SHMT2 as a defatty-acylation substrate of HDAC11. HDAC11-catalyzed defatty-acylation did not affect the enzymatic activity of SHMT2. Instead, it affects the ability of SHMT2 to regulate type I IFN receptor ubiquitination and cell surface level. Correspondingly, HDAC11 depletion increased type I IFN signaling in both cell culture and mice. This study not only demonstrates that HDAC11 has an activity that is much more efficient than the corresponding deacetylase activity, but also expands the physiological functions of HDAC11 and protein lysine fatty acylation, which opens up opportunities to develop HDAC11-specific inhibitors as therapeutics to modulate immune responses.


Subject(s)
Glycine Hydroxymethyltransferase/metabolism , Histone Deacetylases/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Interferon Type I/metabolism , Signal Transduction , Acylation , Animals , HEK293 Cells , Humans , Mice , Mice, Knockout , Signal Transduction/physiology
5.
Proc Natl Acad Sci U S A ; 116(51): 25583-25590, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31776258

ABSTRACT

Methylotrophy, the ability of microorganisms to grow on reduced one-carbon substrates such as methane or methanol, is a feature of various bacterial species. The prevailing oxidation pathway depends on tetrahydromethanopterin (H4MPT) and methylofuran (MYFR), an analog of methanofuran from methanogenic archaea. Formyltransferase/hydrolase complex (Fhc) generates formate from formyl-H4MPT in two consecutive reactions where MYFR acts as a carrier of one-carbon units. Recently, we chemically characterized MYFR from the model methylotroph Methylorubrum extorquens and identified an unusually long polyglutamate side chain of up to 24 glutamates. Here, we report on the crystal structure of Fhc to investigate the function of the polyglutamate side chain in MYFR and the relatedness of the enzyme complex with the orthologous enzymes in archaea. We identified MYFR as a prosthetic group that is tightly, but noncovalently, bound to Fhc. Surprisingly, the structure of Fhc together with MYFR revealed that the polyglutamate side chain of MYFR is branched and contains glutamates with amide bonds at both their α- and γ-carboxyl groups. This negatively charged and branched polyglutamate side chain interacts with a cluster of conserved positively charged residues of Fhc, allowing for strong interactions. The MYFR binding site is located equidistantly from the active site of the formyltransferase (FhcD) and metallo-hydrolase (FhcA). The polyglutamate serves therefore an additional function as a swinging linker to shuttle the one-carbon carrying amine between the two active sites, thereby likely increasing overall catalysis while decreasing the need for high intracellular MYFR concentrations.


Subject(s)
Bacterial Proteins , Furans , Hydroxymethyl and Formyl Transferases , Methane , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography , Formates/chemistry , Formates/metabolism , Furans/chemistry , Furans/metabolism , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/genetics , Hydroxymethyl and Formyl Transferases/metabolism , Methane/chemistry , Methane/metabolism , Methanol/chemistry , Methanol/metabolism , Methylobacterium extorquens/enzymology , Methylobacterium extorquens/genetics , Polyglutamic Acid/chemistry , Polyglutamic Acid/metabolism
6.
J Biol Chem ; 295(28): 9551-9566, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32439803

ABSTRACT

The purinosome is a dynamic metabolic complex composed of enzymes responsible for de novo purine biosynthesis, whose formation has been associated with elevated purine demand. However, the physiological conditions that govern purinosome formation in cells remain unknown. Here, we report that purinosome formation is up-regulated in cells in response to a low-oxygen microenvironment (hypoxia). We demonstrate that increased purinosome assembly in hypoxic human cells requires the activation of hypoxia inducible factor 1 (HIF-1) and not HIF-2. Hypoxia-driven purinosome assembly was inhibited in cells lacking 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/IMP cyclohydrolase (ATIC), a single enzyme in de novo purine biosynthesis, and in cells treated with a small molecule inhibitor of ATIC homodimerization. However, despite the increase in purinosome assembly in hypoxia, we observed no associated increase in de novo purine biosynthesis in cells. Our results indicate that this was likely due to a reduction in mitochondrial one-carbon metabolism, resulting in reduced mitochondrion-derived one-carbon units needed for de novo purine biosynthesis. The findings of our study further clarify and deepen our understanding of purinosome formation by revealing that this process does not solely depend on cellular purine demand.


Subject(s)
Hydroxymethyl and Formyl Transferases/metabolism , Hypoxia-Inducible Factor 1/metabolism , Multienzyme Complexes/metabolism , Nucleotide Deaminases/metabolism , Purines/biosynthesis , Cell Hypoxia , HeLa Cells , Humans , Hydroxymethyl and Formyl Transferases/genetics , Hypoxia-Inducible Factor 1/genetics , Multienzyme Complexes/genetics , Nucleotide Deaminases/genetics
7.
Int J Mol Sci ; 22(4)2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33672312

ABSTRACT

The problematic opportunistic pathogen Pseudomonas aeruginosa secretes a siderophore, pyoverdine. Pyoverdine scavenges iron needed by the bacteria for growth and for pathogenicity in a range of different infection models. PvdF, a hydroxyornithine transformylase enzyme, is essential for pyoverdine synthesis, catalysing synthesis of formylhydroxyornithine (fOHOrn) that forms part of the pyoverdine molecule and provides iron-chelating hydroxamate ligands. Using a mass spectrometry assay, we confirm that purified PvdF catalyses synthesis of fOHOrn from hydroxyornithine and formyltetrahydrofolate substrates. Site directed mutagenesis was carried out to investigate amino acid residues predicted to be required for enzymatic activity. Enzyme variants were assayed for activity in vitro and also in vivo, through measuring their ability to restore pyoverdine production to a pvdF mutant strain. Variants at two putative catalytic residues N168 and H170 greatly reduced enzymatic activity in vivo though did not abolish activity in vitro. Change of a third residue D229 abolished activity both in vivo and in vitro. A change predicted to block entry of N10-formyltetrahydrofolate (fTHF) to the active site also abolished activity both in vitro and in vivo. A co-purification assay showed that PvdF binds to an enzyme PvdA that catalyses synthesis of hydroxyornithine, with this interaction likely to increase the efficiency of fOHOrn synthesis. Our findings advance understanding of how P. aeruginosa synthesises pyoverdine, a key factor in host-pathogen interactions.


Subject(s)
Bacterial Proteins/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Mixed Function Oxygenases/metabolism , Siderophores/biosynthesis , Bacterial Proteins/genetics , Bacterial Proteins/isolation & purification , Catalytic Domain , Hydroxymethyl and Formyl Transferases/genetics , Hydroxymethyl and Formyl Transferases/isolation & purification , Mixed Function Oxygenases/genetics , Mutagenesis, Site-Directed , Oligopeptides/biosynthesis , Protein Interaction Maps , Protein Stability , Pseudomonas aeruginosa/metabolism
8.
J Am Chem Soc ; 142(46): 19754-19762, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33147013

ABSTRACT

The congested nature of quaternary carbons hinders their preparation, most notably when stereocontrol is required. Here we report a biocatalytic method for the creation of quaternary carbon centers with broad substrate scope, leading to different compound classes bearing this structural feature. The key step comprises the aldol addition of 3,3-disubstituted 2-oxoacids to aldehydes catalyzed by metal dependent 3-methyl-2-oxobutanoate hydroxymethyltransferase from E. coli (KPHMT) and variants thereof. The 3,3,3-trisubstituted 2-oxoacids thus produced were converted into 2-oxolactones and 3-hydroxy acids and directly to ulosonic acid derivatives, all bearing gem-dialkyl, gem-cycloalkyl, and spirocyclic quaternary centers. In addition, some of these reactions use a single enantiomer from racemic nucleophiles to afford stereopure quaternary carbons. The notable substrate tolerance and stereocontrol of these enzymes are indicative of their potential for the synthesis of structurally intricate molecules.


Subject(s)
Aldehydes/metabolism , Escherichia coli Proteins/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Keto Acids/metabolism , Aldehydes/chemistry , Binding Sites , Biocatalysis , Catalytic Domain , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/genetics , Keto Acids/chemistry , Mutagenesis, Site-Directed , Stereoisomerism , Substrate Specificity
9.
J Inherit Metab Dis ; 43(6): 1254-1264, 2020 11.
Article in English | MEDLINE | ID: mdl-32557644

ABSTRACT

5-Amino-4-imidazolecarboxamide-ribosiduria (AICA)-ribosiduria is an exceedingly rare autosomal recessive condition resulting from the disruption of the bifunctional purine biosynthesis protein PURH (ATIC), which catalyzes the last two steps of de novo purine synthesis. It is characterized biochemically by the accumulation of AICA-riboside in urine. AICA-ribosiduria had been reported in only one individual, 15 years ago. In this article, we report three novel cases of AICA-ribosiduria from two independent families, with two novel pathogenic variants in ATIC. We also provide a clinical update on the first patient. Based on the phenotypic features shared by these four patients, we define AICA-ribosiduria as the syndromic association of severe-to-profound global neurodevelopmental impairment, severe visual impairment due to chorioretinal atrophy, ante-postnatal growth impairment, and severe scoliosis. Dysmorphic features were observed in all four cases, especially neonatal/infancy coarse facies with upturned nose. Early-onset epilepsy is frequent and can be pharmacoresistant. Less frequently observed features are aortic coarctation, chronic hepatic cytolysis, minor genital malformations, and nephrocalcinosis. Alteration of the transformylase activity of ATIC might result in a more severe impairment than the alteration of the cyclohydrolase activity. Data from literature points toward a cytotoxic mechanism of the accumulated AICA-riboside.


Subject(s)
Congenital Abnormalities/genetics , Epilepsy/genetics , Hydroxymethyl and Formyl Transferases/deficiency , Intellectual Disability/genetics , Multienzyme Complexes/genetics , Nucleotide Deaminases/deficiency , Aminoimidazole Carboxamide/metabolism , Child , Child, Preschool , Female , Humans , Hydroxymethyl and Formyl Transferases/genetics , Hydroxymethyl and Formyl Transferases/metabolism , Infant , Infant, Newborn , Male , Multienzyme Complexes/metabolism , Mutation , Nucleotide Deaminases/genetics , Nucleotide Deaminases/metabolism , Phenotype , Ribonucleosides/metabolism
10.
J Biol Chem ; 293(13): 4845-4859, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29414769

ABSTRACT

The enzyme AICAR-transformylase/IMP cyclohydrolase (ATIC) catalyzes the last two steps of purine de novo synthesis. It metabolizes 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), which is an AMP analogue, leading to activation of AMP-activated kinase (AMPK). We investigated whether the AICAR-ATIC pathway plays a role in the high glucose (HG)-mediated DNA damage response and AICAR-mediated AMPK activation, explaining the detrimental effects of glucose on neuronal damage and shortening of the lifespan. HG up-regulated the expression and activity of the Caenorhabditis elegans homologue of ATIC, C55F2.1 (atic-1), and increased the levels of reactive oxygen species and methylglyoxal-derived advanced glycation end products. Overexpression of atic-1 decreased the lifespan and head motility and increased neuronal damage under both standard and HG conditions. Inhibition of atic-1 expression, by RNAi, under HG was associated with increased lifespan and head motility and reduced neuronal damage, reactive oxygen species, and methylglyoxal-derived advanced glycation end product accumulation. This effect was independent of an effect on DNA damage or antioxidant defense pathways, such as superoxide dismutase (sod-3) or glyoxalase-1 (glod-4), but was dependent on AMPK and accumulation of AICAR. Through AMPK, AICAR treatment also reduced the negative effects of HG. The mitochondrial inhibitor rotenone abolished the AICAR/AMPK-induced amelioration of HG effects, pointing to mitochondria as a prime target of the glucotoxic effects in C. elegans We conclude that atic-1 is involved in glucotoxic effects under HG conditions, either by blocked atic-1 expression or via AICAR and AMPK induction.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Glucose/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Multienzyme Complexes/metabolism , Nucleotide Deaminases/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Hydroxymethyl and Formyl Transferases/genetics , Multienzyme Complexes/genetics , Neurons/metabolism , Nucleotide Deaminases/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
11.
BMC Microbiol ; 19(1): 232, 2019 10 26.
Article in English | MEDLINE | ID: mdl-31655553

ABSTRACT

BACKGROUND: Co-translational processes in bacteria are attractive drug targets, but while some processes are essential, others are not. The essentiality of Peptide Deformylase (PDF, def) for vitality of mycobacteria was speculated, but never unequivocally proven. RESULTS: Here we show by targeted deletion experiments that def can only be deleted from M. smegmatis when an additional copy is present; that prior deletion of tRNAfMet-Formyl Transferase (FMT, encoded by fmt) renders def completely dispensable; and that re-introduction of fmt into a Δdef mutant is not possible - constituting a definitive proof for the essentiality of def in mycobacteria. CONCLUSIONS: Peptide deformylase is essential in M. smegmatis, but the fact that inactivation of fmt renders the gene completely dispensable, and thus any inhibitor of def useless, casts doubt on the usefulness of PDF as a drug-target in mycobacteria.


Subject(s)
Amidohydrolases/genetics , Amidohydrolases/metabolism , Methionine/chemistry , Mycobacterium smegmatis/growth & development , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Formates/chemistry , Gene Deletion , Genes, Essential , Hydroxymethyl and Formyl Transferases/metabolism , Microbial Viability , Mycobacterium smegmatis/enzymology , Mycobacterium smegmatis/genetics
12.
Arch Biochem Biophys ; 664: 40-50, 2019 03 30.
Article in English | MEDLINE | ID: mdl-30689984

ABSTRACT

The hydroxyornithine transformylase from Pseudomonas aeruginosa is known by the gene name pvdF, and has been hypothesized to use N10-formyltetrahydrofolate (N10-fTHF) as a co-substrate formyl donor to convert N5-hydroxyornithine (OHOrn) to N5-formyl- N5-hydroxyornithine (fOHOrn). PvdF is in the biosynthetic pathway for pyoverdin biosynthesis, a siderophore generated under iron-limiting conditions that has been linked to virulence, quorum sensing and biofilm formation. The structure of PvdF was determined by X-ray crystallography to 2.3 Å, revealing a formyltransferase fold consistent with N10-formyltetrahydrofolate dependent enzymes, such as the glycinamide ribonucleotide transformylases, N-sugar transformylases and methionyl-tRNA transformylases. Whereas the core structure, including the catalytic triad, is conserved, PvdF has three insertions of 18 or more amino acids, which we hypothesize are key to binding the OHOrn substrate. Steady state kinetics revealed a non-hyperbolic rate curve, promoting the hypothesis that PvdF uses a random-sequential mechanism, and favors folate binding over OHOrn.


Subject(s)
Formyltetrahydrofolates/metabolism , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/metabolism , Oligopeptides/biosynthesis , Folic Acid/metabolism , Models, Molecular , Protein Conformation , Pseudomonas aeruginosa/enzymology
13.
Biochim Biophys Acta Proteins Proteom ; 1866(2): 254-263, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29042184

ABSTRACT

Aminoimidazolecarboxamide ribonucleotide formyl transferase (AICARFT): Inosine monophosphate cyclohydrolase (IMPCH, collectively called ATIC) is a bifunctional enzyme that catalyses the penultimate and final steps in the purine de novo biosynthesis pathway. The bifunctional protein is dimeric and each monomer contains two different active sites both of which are capable of binding nucleotide substrates, this means to a potential total of four distinct binding events might be observed. Within this work we used a combination of site-directed and truncation mutants of ATIC to independently investigate the binding at these two sites using calorimetry. A single S10W mutation is sufficient to block the IMPCH active site allowing investigation of the effects of mutation on ligand binding in the AICARFT active site. The majority of nucleotide ligands bind selectively at one of the two active sites with the exception of xanthosine monophosphate, XMP, which, in addition to binding in both AICARFT and IMPCH active sites, shows evidence for cooperative binding with communication between symmetrically-related active sites in the two IMPCH domains. The AICARFT site is capable of independently binding both nucleotide and folate substrates with high affinity however no evidence for positive cooperativity in binding could be detected using the model ligands employed in this study.


Subject(s)
Hydroxymethyl and Formyl Transferases/chemistry , Models, Molecular , Multienzyme Complexes/chemistry , Nucleotide Deaminases/chemistry , Nucleotides/chemistry , Catalytic Domain , Humans , Hydroxymethyl and Formyl Transferases/genetics , Hydroxymethyl and Formyl Transferases/metabolism , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Nucleotide Deaminases/genetics , Nucleotide Deaminases/metabolism , Nucleotides/genetics , Nucleotides/metabolism , Protein Binding , Substrate Specificity/physiology
14.
Pharmacol Res ; 138: 37-42, 2018 12.
Article in English | MEDLINE | ID: mdl-30267763

ABSTRACT

Mitochondrial myopathy (MM) is characterised by muscle weakness, exercise intolerance and various histopathological changes. Recently, a subset of MM has also been associated with aberrant activation of mammalian target of rapamycin complex 1 (mTORC1) in skeletal muscle. This aberrant mTORC1 activation promotes increased de novo nucleotide synthesis, which contributes to abnormal expansion and imbalance of skeletal muscle deoxyribonucleoside triphosphates (dNTP) pools. However, the exact mechanism via which mTORC1-stimulated de novo nucleotide biosynthesis ultimately disturbs muscle dNTP pools remains unclear. In this article, it is proposed that mTORC1-stimulated de novo nucleotide synthesis in skeletal muscle cells with respiratory chain dysfunction promotes an asymmetric increase of purine nucleotides, probably due to NAD+ deficiency. This in turn could disrupt purine nucleotide-dependent allosteric feedback regulatory mechanisms, ultimately leading to dNTP pools aberration. Pharmacological down-modulation of aminoimidazole carboxamide ribonucleotide transformylase/inosine monophosphate cyclohydrolase (ATIC) activity is also proposed as a potential therapeutic strategy in MM exhibiting mTORC1-driven abnormal metabolic reprogramming, including aberrant dNTPs pools.


Subject(s)
Mitochondrial Myopathies/metabolism , Purine Nucleotides/metabolism , Animals , Humans , Hydroxymethyl and Formyl Transferases/antagonists & inhibitors , Hydroxymethyl and Formyl Transferases/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mitochondrial Myopathies/drug therapy , Multienzyme Complexes/antagonists & inhibitors , Multienzyme Complexes/metabolism , Nucleotide Deaminases/antagonists & inhibitors , Nucleotide Deaminases/metabolism
15.
Mol Biol Rep ; 45(6): 2707-2716, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30377949

ABSTRACT

De novo synthesis of purines has been suggested to be an important factor for the pathogenesis of uropathogenic E. coli (UPEC). We analyzed the role of the redundant purine biosynthesis genes purN and purT, responsible for the third step in the purine biosynthesis, during UPEC infection. Growth experiments in M9 (minimal media), MOPS (rich media), filtered urine, and human serum with E. coli UTI89 and ΔpurN, ΔpurT, and ΔpurN/T mutants revealed that UPEC relies on de novo purine synthesis for growth in minimal medium. Mutants in individual genes as well as the double mutant grew equally well as the wild type in urine, rich media, and serum. However, during competition for growth in urine, the wild type UTI89 strain significantly outcompeted the purine auxotrophic ΔpurN/T mutant from late exponential growth phase. Inactivation of purN and/or purT significantly affected UPEC invasion of human bladder cells, but not the intracellular survival. Cytotoxicity levels to bladder cells were also diminished when both purN and purT were deleted, while single gene mutants did not differ from the wild type. When infecting human macrophages, no differences were observed between UTI89 and mutants in uptake, survival or cytotoxicity. Finally, the lack of the pur-gene(s), whether analysed as single or double gene knock-out, did not affect recovery rates after in vivo infection in a mouse model of UTI. These findings suggest that de novo synthesis of purines might be required only when UPEC is fully deprived of nucleotides and when grown in competition with other microorganisms in urine.


Subject(s)
Escherichia coli Proteins/genetics , Hydroxymethyl and Formyl Transferases/genetics , Purines/biosynthesis , Uropathogenic Escherichia coli/genetics , Animals , Escherichia coli/metabolism , Escherichia coli Infections/genetics , Escherichia coli Infections/metabolism , Escherichia coli Proteins/metabolism , Female , Humans , Hydroxymethyl and Formyl Transferases/metabolism , Mice , Mice, Inbred C3H , Primary Cell Culture , Purines/metabolism , Urinary Bladder , Urinary Tract Infections/genetics , Urinary Tract Infections/metabolism , Urine/microbiology , Uropathogenic Escherichia coli/metabolism , Virulence , Virulence Factors
16.
Biochemistry ; 56(28): 3657-3668, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28636341

ABSTRACT

It has become increasingly apparent within the last several years that unusual N-formylated sugars are often found on the O-antigens of such Gram negative pathogenic organisms as Francisella tularensis, Campylobacter jejuni, and Providencia alcalifaciens, among others. Indeed, in some species of Brucella, for example, the O-antigen contains 1,2-linked 4-formamido-4,6-dideoxy-α-d-mannosyl groups. These sugars, often referred to as N-formylperosamine, are synthesized in pathways initiating with GDP-mannose. One of the enzymes required for the production of N-formylperosamine, namely, WbkC, was first identified in 2000 and was suggested to function as an N-formyltransferase. Its biochemical activity was never experimentally verified, however. Here we describe a combined structural and functional investigation of WbkC from Brucella melitensis. Four high resolution X-ray structures of WbkC were determined in various complexes to address its active site architecture. Unexpectedly, the quaternary structure of WbkC was shown to be different from that previously observed for other sugar N-formyltransferases. Additionally, the structures revealed a second binding site for a GDP molecule distinct from that required for GDP-perosamine positioning. In keeping with this additional binding site, kinetic data with the wild type enzyme revealed complex patterns. Removal of GDP binding by mutating Phe 142 to an alanine residue resulted in an enzyme variant displaying normal Michaelis-Menten kinetics. These data suggest that this nucleotide binding pocket plays a role in enzyme regulation. Finally, by using an alternative substrate, we demonstrate that WbkC can be utilized to produce a trideoxysugar not found in nature.


Subject(s)
Brucella melitensis/enzymology , Hydroxymethyl and Formyl Transferases/metabolism , Binding Sites , Brucella melitensis/chemistry , Brucellosis/microbiology , Catalytic Domain , Crystallography, X-Ray , Guanosine Diphosphate/metabolism , Hexosamines/metabolism , Humans , Hydroxymethyl and Formyl Transferases/chemistry , Kinetics , Models, Molecular , Protein Conformation , Substrate Specificity
17.
Biochemistry ; 56(29): 3818-3825, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28665588

ABSTRACT

The causative agent of tuberculosis, Mycobacterium tuberculosis, is a bacterium with a complex cell wall and a complicated life cycle. The genome of M. tuberculosis contains well over 4000 genes thought to encode proteins. One of these codes for a putative enzyme referred to as Rv3404c, which has attracted research attention as a potential virulence factor for over 12 years. Here we demonstrate that Rv3404c functions as a sugar N-formyltransferase that converts dTDP-4-amino-4,6-dideoxyglucose into dTDP-4-formamido-4,6-dideoxyglucose using N10-formyltetrahydrofolate as the carbon source. Kinetic analyses demonstrate that Rv3404c displays a significant catalytic efficiency of 1.1 × 104 M-1 s-1. In addition, we report the X-ray structure of a ternary complex of Rv3404c solved in the presence of N5-formyltetrahydrofolate and dTDP-4-amino-4,6-dideoxyglucose. The final model of Rv3404c was refined to an overall R-factor of 16.8% at 1.6 Å resolution. The results described herein are especially intriguing given that there have been no published reports of N-formylated sugars associated with M. tuberculosis. The data thus provide a new avenue of research into this fascinating, yet deadly, organism that apparently has been associated with human infection since ancient times.


Subject(s)
Bacterial Proteins/chemistry , Hydroxymethyl and Formyl Transferases/chemistry , Models, Molecular , Mycobacterium tuberculosis/enzymology , Virulence Factors/chemistry , Bacterial Proteins/metabolism , Catalysis , Crystallography, X-Ray , Deoxy Sugars/chemistry , Deoxy Sugars/metabolism , Formyltetrahydrofolates/chemistry , Formyltetrahydrofolates/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Kinetics , Mycobacterium tuberculosis/pathogenicity , Thymine Nucleotides/chemistry , Thymine Nucleotides/metabolism , Virulence Factors/metabolism
18.
J Struct Biol ; 200(3): 267-278, 2017 12.
Article in English | MEDLINE | ID: mdl-28263875

ABSTRACT

N-formylated sugars are found on the lipopolysaccharides of various pathogenic Gram negative bacteria including Campylobacter jejuni 81116, Francisella tularensis, Providencia alcalifaciens O30, and Providencia alcalifaciens O40. The last step in the biosynthetic pathways for these unusual sugars is catalyzed by N-formyltransferases that utilize N10-formyltetrahydrofolate as the carbon source. The substrates are dTDP-linked amino sugars with the functional groups installed at either the C-3' or C-4' positions of the pyranosyl rings. Here we describe a structural and enzymological investigation of the putative N-formyltransferase, FdtF, from Salmonella enterica O60. In keeping with its proposed role in the organism, the kinetic data reveal that the enzyme is more active with dTDP-3-amino-3,6-dideoxy-d-galactose than with dTDP-3-amino-3,6-dideoxy-d-glucose. The structural data demonstrate that the enzyme contains, in addition to the canonical N-formyltransferase fold, an ankyrin repeat moiety that houses a second dTDP-sugar binding pocket. This is only the second time an ankyrin repeat has been shown to be involved in small molecule binding. The research described herein represents the first structural analysis of a sugar N-formyltransferase that specifically functions on dTDP-3-amino-3,6-dideoxy-d-galactose in vivo and thus adds to our understanding of these intriguing enzymes.


Subject(s)
Bacterial Proteins/chemistry , Hydroxymethyl and Formyl Transferases/chemistry , Hydroxymethyl and Formyl Transferases/metabolism , Salmonella enterica/enzymology , Amino Sugars/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Catalytic Domain , Crystallography, X-Ray , Glucosamine/analogs & derivatives , Glucosamine/metabolism , Hydroxymethyl and Formyl Transferases/genetics , Kinetics , Models, Molecular , Protein Conformation
19.
BMC Struct Biol ; 17(1): 1, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28143508

ABSTRACT

BACKGROUND: The post-translational modification pathway referred to as pupylation marks proteins for proteasomal degradation in Mycobacterium tuberculosis and other actinobacteria by covalently attaching the small protein Pup (prokaryotic ubiquitin-like protein) to target lysine residues. In contrast to the functionally analogous eukaryotic ubiquitin, Pup is intrinsically disordered in its free form. Its unfolded state allows Pup to adopt different structures upon interaction with different binding partners like the Pup ligase PafA and the proteasomal ATPase Mpa. While the disordered behavior of free Pup has been well characterized, it remained unknown whether Pup adopts a distinct structure when attached to a substrate. RESULTS: Using a combination of NMR experiments and biochemical analysis we demonstrate that Pup remains unstructured when ligated to two well-established pupylation substrates targeted for proteasomal degradation in Mycobacterium tuberculosis, malonyl transacylase (FabD) and ketopantoyl hydroxylmethyltransferase (PanB). Isotopically labeled Pup was linked to FabD and PanB by in vitro pupylation to generate homogeneously pupylated substrates suitable for NMR analysis. The single target lysine of PanB was identified by a combination of mass spectroscopy and mutational analysis. Chemical shift comparison between Pup in its free form and ligated to substrate reveals intrinsic disorder of Pup in the conjugate. CONCLUSION: When linked to the proteasomal substrates FabD and PanB, Pup is unstructured and retains the ability to interact with its different binding partners. This suggests that it is not the conformation of Pup attached to these two substrates which determines their delivery to the proteasome, but the availability of the degradation complex and the depupylase.


Subject(s)
Acyl-Carrier Protein S-Malonyltransferase/chemistry , Bacterial Proteins/chemistry , Hydroxymethyl and Formyl Transferases/chemistry , Mycobacterium tuberculosis/physiology , Proteasome Endopeptidase Complex/metabolism , Ubiquitins/chemistry , Acyl-Carrier Protein S-Malonyltransferase/metabolism , Bacterial Proteins/metabolism , Hydroxymethyl and Formyl Transferases/metabolism , Models, Molecular , Protein Conformation , Proteolysis , Substrate Specificity , Ubiquitination , Ubiquitins/metabolism
20.
Cell Commun Signal ; 15(1): 52, 2017 Dec 16.
Article in English | MEDLINE | ID: mdl-29246230

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the cancer types with poor prognosis. To effectively treat HCC, new molecular targets and therapeutic approaches must be identified. 5-aminoimidazole-4-carboxamide ribonucleotide formyltransferase/inosine monophosphate (IMP) cyclohydrolase (ATIC), a bifunctional protein enzyme, catalyzes the last two steps of the de novo purine biosynthetic pathway. Whether ATIC contributes to cancer development remains unclear. METHODS: ATIC mRNA levels in different types of human HCC samples or normal tissues were determined from Gene Expression across Normal and Tumor tissue (GENT) database. The expression level of ATIC in human HCC samples or cell lines were examined by RT-PCR and western blot. Overall survival and disease-free survival of HCC patients in the ATIC low and ATIC high groups were determined by Kaplan-Meier analysis. Effects of ATIC knockdown by lentivirus infection were evaluated on cell-proliferation, cell-apoptosis, colony formation and migration. The mechanisms involved in HCC cells growth, apoptosis and migration were analyzed by western blot and Compound C (C-C) rescue assays. RESULTS: Here, we first demonstrated that expression of ATIC is aberrantly up-regulated in HCC tissues and high level of ATIC is correlated with poor survival in HCC patients. Knockdown of ATIC expression resulted in a dramatic decrease in proliferation, colony formation and migration of HCC cells. We also identified ATIC as a novel regulator of adenosine monophosphate-activated protein kinase (AMPK) and its downstream signaling mammalian target of rapamycin (mTOR). ATIC suppresses AMPK activation, thus activates mTOR-S6 K1-S6 signaling and supports growth and motility activity of HCC cells. CONCLUSION: Taken together, our results indicate that ATIC acts as an oncogenic gene that promotes survival, proliferation and migration by targeting AMPK-mTOR-S6 K1 signaling.


Subject(s)
Adenylate Kinase/metabolism , Carcinoma, Hepatocellular/pathology , Hydroxymethyl and Formyl Transferases/metabolism , Liver Neoplasms/pathology , Multienzyme Complexes/metabolism , Nucleotide Deaminases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Disease Progression , Disease-Free Survival , Enzyme Activation , Gene Knockdown Techniques , Humans , Hydroxymethyl and Formyl Transferases/deficiency , Hydroxymethyl and Formyl Transferases/genetics , Molecular Targeted Therapy , Multienzyme Complexes/deficiency , Multienzyme Complexes/genetics , Nucleotide Deaminases/deficiency , Nucleotide Deaminases/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL