Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(36): e2207190119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037354

ABSTRACT

Mercaptoethane sulfonate or coenzyme M (CoM) is the smallest known organic cofactor and is most commonly associated with the methane-forming step in all methanogenic archaea but is also associated with the anaerobic oxidation of methane to CO2 in anaerobic methanotrophic archaea and the oxidation of short-chain alkanes in Syntrophoarchaeum species. It has also been found in a small number of bacteria capable of the metabolism of small organics. Although many of the steps for CoM biosynthesis in methanogenic archaea have been elucidated, a complete pathway for the biosynthesis of CoM in archaea or bacteria has not been reported. Here, we present the complete CoM biosynthesis pathway in bacteria, revealing distinct chemical steps relative to CoM biosynthesis in methanogenic archaea. The existence of different pathways represents a profound instance of convergent evolution. The five-step pathway involves the addition of sulfite, the elimination of phosphate, decarboxylation, thiolation, and the reduction to affect the sequential conversion of phosphoenolpyruvate to CoM. The salient features of the pathway demonstrate reactivities for members of large aspartase/fumarase and pyridoxal 5'-phosphate-dependent enzyme families.


Subject(s)
Bacteria , Coenzymes , Euryarchaeota , Mesna , Anaerobiosis , Archaea/metabolism , Bacteria/metabolism , Coenzymes/biosynthesis , Euryarchaeota/metabolism , Mesna/metabolism , Methane/metabolism , Oxidation-Reduction , Phosphates/metabolism
2.
J Biol Chem ; 299(8): 104919, 2023 08.
Article in English | MEDLINE | ID: mdl-37315792

ABSTRACT

Coenzymes are important for all classes of enzymatic reactions and essential for cellular metabolism. Most coenzymes are synthesized from dedicated precursors, also referred to as vitamins, which prototrophic bacteria can either produce themselves from simpler substrates or take up from the environment. The extent to which prototrophs use supplied vitamins and whether externally available vitamins affect the size of intracellular coenzyme pools and control endogenous vitamin synthesis is currently largely unknown. Here, we studied coenzyme pool sizes and vitamin incorporation into coenzymes during growth on different carbon sources and vitamin supplementation regimes using metabolomics approaches. We found that the model bacterium Escherichia coli incorporated pyridoxal, niacin, and pantothenate into pyridoxal 5'-phosphate, NAD, and coenzyme A (CoA), respectively. In contrast, riboflavin was not taken up and was produced exclusively endogenously. Coenzyme pools were mostly homeostatic and not affected by externally supplied precursors. Remarkably, we found that pantothenate is not incorporated into CoA as such but is first degraded to pantoate and ß-alanine and then rebuilt. This pattern was conserved in various bacterial isolates, suggesting a preference for ß-alanine over pantothenate utilization in CoA synthesis. Finally, we found that the endogenous synthesis of coenzyme precursors remains active when vitamins are supplied, which is consistent with described expression data of genes for enzymes involved in coenzyme biosynthesis under these conditions. Continued production of endogenous coenzymes may ensure rapid synthesis of the mature coenzyme under changing environmental conditions, protect against coenzyme limitation, and explain vitamin availability in naturally oligotrophic environments.


Subject(s)
Coenzymes , Escherichia coli , beta-Alanine , beta-Alanine/metabolism , Coenzyme A/biosynthesis , Coenzymes/biosynthesis , Pyridoxal , Pyridoxal Phosphate/metabolism , Vitamins/metabolism , Escherichia coli/metabolism , NAD/metabolism , Culture Media/chemistry , Culture Media/metabolism
3.
Molecules ; 29(18)2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39339452

ABSTRACT

Molybdenum (Mo) is an essential micronutrient across all kingdoms of life, where it functions as a key component of the active centers of molybdenum-dependent enzymes. For these enzymes to gain catalytic activity, Mo must be complexed with a pterin scaffold to form the molybdenum cofactor (Moco). The final step of Moco biosynthesis is catalyzed by the enzyme Mo-insertase. This review focuses on eukaryotic Mo-insertases, with an emphasis on those found in plants and mammals, which have been instrumental in advancing the understanding of Mo biochemistry. Additionally, a historical perspective is provided, tracing the discovery of Mo-insertase from the early 1960s to the detailed characterization of its reaction mechanism in 2021. This review also highlights key milestones in the study of Mo-insertase, including mutant characterization, gene cloning, structural elucidation at the atomic level, functional domain assignment, and the spatial organization of the enzyme within cellular protein networks.


Subject(s)
Coenzymes , Metalloproteins , Molybdenum Cofactors , Pteridines , Pteridines/metabolism , Pteridines/chemistry , Coenzymes/metabolism , Coenzymes/biosynthesis , Coenzymes/chemistry , Metalloproteins/metabolism , Metalloproteins/biosynthesis , Metalloproteins/chemistry , Humans , Animals , Molybdenum/chemistry , Molybdenum/metabolism
4.
Nature ; 543(7643): 78-82, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28225763

ABSTRACT

Methane biogenesis in methanogens is mediated by methyl-coenzyme M reductase, an enzyme that is also responsible for the utilization of methane through anaerobic methane oxidation. The enzyme uses an ancillary factor called coenzyme F430, a nickel-containing modified tetrapyrrole that promotes catalysis through a methyl radical/Ni(ii)-thiolate intermediate. However, it is unclear how coenzyme F430 is synthesized from the common primogenitor uroporphyrinogen iii, incorporating 11 steric centres into the macrocycle, although the pathway must involve chelation, amidation, macrocyclic ring reduction, lactamization and carbocyclic ring formation. Here we identify the proteins that catalyse the biosynthesis of coenzyme F430 from sirohydrochlorin, termed CfbA-CfbE, and demonstrate their activity. The research completes our understanding of how the repertoire of tetrapyrrole-based pigments are constructed, permitting the development of recombinant systems to use these metalloprosthetic groups more widely.


Subject(s)
Biocatalysis , Biosynthetic Pathways , Coenzymes/biosynthesis , Metalloporphyrins/metabolism , Methane/biosynthesis , Methanosarcina barkeri/enzymology , Tetrapyrroles/biosynthesis , Amidohydrolases/genetics , Amidohydrolases/metabolism , Biosynthetic Pathways/genetics , Coenzymes/chemistry , Lyases/genetics , Lyases/metabolism , Metalloporphyrins/chemistry , Methane/analogs & derivatives , Methane/metabolism , Methanosarcina barkeri/genetics , Methanosarcina barkeri/metabolism , Multigene Family , Nickel/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism , Tetrapyrroles/chemistry , Uroporphyrins/chemistry , Uroporphyrins/metabolism
5.
FASEB J ; 34(8): 10871-10886, 2020 08.
Article in English | MEDLINE | ID: mdl-32649804

ABSTRACT

Human riboflavin kinase (HsRFK) catalyzes vitamin B2 (riboflavin) phosphorylation to flavin mononucleotide (FMN), obligatory step in flavin cofactor synthesis. HsRFK expression is related to protection from oxidative stress, amyloid-ß toxicity, and some malignant cancers progression. Its downregulation alters expression profiles of clock-controlled metabolic-genes and destroys flavins protection on stroke treatments, while its activity reduction links to protein-energy malnutrition and thyroid hormones decrease. We explored specific features of the mechanisms underlying the regulation of HsRFK activity, showing that both reaction products regulate it through competitive inhibition. Fast-kinetic studies show that despite HsRFK binds faster and preferably the reaction substrates, the complex holding both products is kinetically most stable. An intricate ligand binding landscape with all combinations of substrates/products competing with the catalytic complex and exhibiting moderate cooperativity is also presented. These data might contribute to better understanding the molecular bases of pathologies coursing with aberrant HsRFK availability, and envisage that interaction with its client-apoproteins might favor FMN release. Finally, HsRFK parameters differ from those of the so far evaluated bacterial counterparts, reinforcing the idea of species-specific mechanisms in RFK catalysis. These observations support HsRFK as potential therapeutic target because of its key functions, while also envisage bacterial RFK modules as potential antimicrobial targets.


Subject(s)
Coenzymes/biosynthesis , Coenzymes/metabolism , Flavin Mononucleotide/biosynthesis , Flavin Mononucleotide/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Catalysis , Humans , Kinetics , Riboflavin/metabolism , Species Specificity , Substrate Specificity
6.
Environ Microbiol ; 22(6): 2007-2026, 2020 06.
Article in English | MEDLINE | ID: mdl-32239579

ABSTRACT

The biosynthesis of the molybdenum cofactor (Moco) is highly conserved among all kingdoms of life. In all molybdoenzymes containing Moco, the molybdenum atom is coordinated to a dithiolene group present in the pterin-based 6-alkyl side chain of molybdopterin (MPT). In general, the biosynthesis of Moco can be divided into four steps in in bacteria: (i) the starting point is the formation of the cyclic pyranopterin monophosphate (cPMP) from 5'-GTP, (ii) in the second step the two sulfur atoms are inserted into cPMP leading to the formation of MPT, (iii) in the third step the molybdenum atom is inserted into MPT to form Moco and (iv) in the fourth step bis-Mo-MPT is formed and an additional modification of Moco is possible with the attachment of a nucleotide (CMP or GMP) to the phosphate group of MPT, forming the dinucleotide variants of Moco. This review presents an update on the well-characterized Moco biosynthesis in the model organism Escherichia coli including novel discoveries from the recent years.


Subject(s)
Coenzymes/biosynthesis , Escherichia coli/metabolism , Metalloproteins/biosynthesis , Coenzymes/chemistry , Metalloproteins/chemistry , Molybdenum/metabolism , Molybdenum Cofactors , Organophosphorus Compounds , Pteridines/chemistry , Pterins/chemistry
7.
Angew Chem Int Ed Engl ; 59(17): 6887-6893, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32022452

ABSTRACT

NifEN plays a crucial role in the biosynthesis of nitrogenase, catalyzing the final step of cofactor maturation prior to delivering the cofactor to NifDK, the catalytic component of nitrogenase. The difficulty in expressing NifEN, a complex, heteromultimeric metalloprotein sharing structural/functional homology with NifDK, is a major challenge in the heterologous expression of nitrogenase. Herein, we report the expression and engineering of Azotobacter vinelandii NifEN in Escherichia coli. Biochemical and spectroscopic analyses demonstrate the integrity of the heterologously expressed NifEN in composition and functionality and, additionally, the ability of an engineered NifEN variant to mimic NifDK in retaining the matured cofactor at an analogous cofactor-binding site. This is an important step toward piecing together a viable pathway for the heterologous expression of nitrogenase and identifying variants for the mechanistic investigation of this enzyme.


Subject(s)
Bacterial Proteins/genetics , Coenzymes/biosynthesis , Genetic Engineering , Nitrogenase/metabolism , Azotobacter vinelandii/genetics , Gene Expression
8.
J Bacteriol ; 201(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31235512

ABSTRACT

Molybdenum cofactor (Moco) biosynthesis is a complex process that involves the coordinated function of several proteins. In recent years it has become obvious that the availability of iron plays an important role in the biosynthesis of Moco. First, the MoaA protein binds two [4Fe-4S] clusters per monomer. Second, the expression of the moaABCDE and moeAB operons is regulated by FNR, which senses the availability of oxygen via a functional [4Fe-4S] cluster. Finally, the conversion of cyclic pyranopterin monophosphate to molybdopterin requires the availability of the l-cysteine desulfurase IscS, which is a shared protein with a main role in the assembly of Fe-S clusters. In this report, we investigated the transcriptional regulation of the moaABCDE operon by focusing on its dependence on cellular iron availability. While the abundance of selected molybdoenzymes is largely decreased under iron-limiting conditions, our data show that the regulation of the moaABCDE operon at the level of transcription is only marginally influenced by the availability of iron. Nevertheless, intracellular levels of Moco were decreased under iron-limiting conditions, likely based on an inactive MoaA protein in addition to lower levels of the l-cysteine desulfurase IscS, which simultaneously reduces the sulfur availability for Moco production.IMPORTANCE FNR is a very important transcriptional factor that represents the master switch for the expression of target genes in response to anaerobiosis. Among the FNR-regulated operons in Escherichia coli is the moaABCDE operon, involved in Moco biosynthesis. Molybdoenzymes have essential roles in eukaryotic and prokaryotic organisms. In bacteria, molybdoenzymes are crucial for anaerobic respiration using alternative electron acceptors. This work investigates the connection of iron availability to the biosynthesis of Moco and the production of active molybdoenzymes.


Subject(s)
Coenzymes/biosynthesis , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial/physiology , Iron/metabolism , Metalloproteins/biosynthesis , Escherichia coli Proteins/genetics , Molybdenum Cofactors , Proteomics , Pteridines
9.
PLoS Pathog ; 13(11): e1006752, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29176894

ABSTRACT

The unique ability of the tuberculosis (TB) bacillus, Mycobacterium tuberculosis, to persist for long periods of time in lung hypoxic lesions chiefly contributes to the global burden of latent TB. We and others previously reported that the M. tuberculosis ancestor underwent massive episodes of horizontal gene transfer (HGT), mostly from environmental species. Here, we sought to explore whether such ancient HGT played a part in M. tuberculosis evolution towards pathogenicity. We were interested by a HGT-acquired M. tuberculosis-specific gene set, namely moaA1-D1, which is involved in the biosynthesis of the molybdenum cofactor. Horizontal acquisition of this gene set was striking because homologues of these moa genes are present all across the Mycobacterium genus, including in M. tuberculosis. Here, we discovered that, unlike their paralogues, the moaA1-D1 genes are strongly induced under hypoxia. In vitro, a M. tuberculosis moaA1-D1-null mutant has an impaired ability to respire nitrate, to enter dormancy and to survive in oxygen-limiting conditions. Conversely, heterologous expression of moaA1-D1 in the phylogenetically closest non-TB mycobacterium, Mycobacterium kansasii, which lacks these genes, improves its capacity to respire nitrate and grants it with a marked ability to survive oxygen depletion. In vivo, the M. tuberculosis moaA1-D1-null mutant shows impaired survival in hypoxic granulomas in C3HeB/FeJ mice, but not in normoxic lesions in C57BL/6 animals. Collectively, our results identify a novel pathway required for M. tuberculosis resistance to host-imposed stress, namely hypoxia, and provide evidence that ancient HGT bolstered M. tuberculosis evolution from an environmental species towards a pervasive human-adapted pathogen.


Subject(s)
Coenzymes/biosynthesis , Gene Transfer, Horizontal , Metalloproteins/biosynthesis , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Oxygen/metabolism , Tuberculosis/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Gene Expression Regulation, Bacterial , Humans , Hypoxia/metabolism , Hypoxia/microbiology , Mice , Mice, Inbred C57BL , Molybdenum Cofactors , Mycobacterium/genetics , Mycobacterium/metabolism , Nitrates/metabolism , Pteridines , Tuberculosis/metabolism
10.
New Phytol ; 222(1): 275-285, 2019 04.
Article in English | MEDLINE | ID: mdl-30471121

ABSTRACT

In cereal crops, ABA deficiency during seed maturation phase causes pre-harvest sprouting (PHS), and molybdenum cofactor (MoCo) is required for ABA biosynthesis. Here, two rice PHS mutants F254 and F5-1 were characterized. In addition to the PHS, these mutants showed pleiotropic phenotypes such as twisting and slender leaves, and then died when the seedling developed to four or five leaves. Map-based cloning showed that OsCNX6 and OsCNX1 encoding homologs of MoaE and MoeA were responsible for F254 and F5-1 mutants, respectively. Genetic complementation indicated that OsCNX6 not only rescued the PHS and seedling lethal phenotype of the cnx6 mutant, but also recovered the MoCo-dependent enzyme activities such as xanthine dehydrogenase (XDH), aldehyde oxidase (AO), nitrate reductase (NR) and sulfite oxidase (SO). Expression pattern showed that OsCNX6 was richly expressed in seed during embryo maturation by quantitative reverse transcriptase PCR and RNA in situ hybridization. Furthermore, the OsCNX6 overexpression plants can significantly enhance the MoCo-dependent enzyme activities, and improved the osmotic and salt stress tolerance without unfavorable phenotypes. Collectively, these data indicated that OsCNX6 participated in MoCo biosynthesis, and is essential for rice development, especially for seed dormancy and germination, and OsCNX6 could be an effective target for improving abiotic stress tolerance in rice.


Subject(s)
Biosynthetic Pathways , Coenzymes/biosynthesis , Metalloproteins/biosynthesis , Mutation/genetics , Oryza/growth & development , Oryza/genetics , Adaptation, Physiological , Gene Expression Regulation, Plant , Molybdenum Cofactors , Oryza/physiology , Osmosis , Phenotype , Plant Proteins/genetics , Plant Proteins/metabolism , Pteridines , Salt Stress/genetics , Stress, Physiological/genetics
11.
Biochem J ; 475(2): 495-509, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29247140

ABSTRACT

Mitochondria play a key role in the biosynthesis of two metal cofactors, iron-sulfur (FeS) clusters and molybdenum cofactor (Moco). The two pathways intersect at several points, but a scarcity of mutants has hindered studies to better understand these links. We screened a collection of sirtinol-resistant Arabidopsis thaliana mutants for lines with decreased activities of cytosolic FeS enzymes and Moco enzymes. We identified a new mutant allele of ATM3 (ABC transporter of the mitochondria 3), encoding the ATP-binding cassette transporter of the mitochondria 3 (systematic name ABCB25), confirming the previously reported role of ATM3 in both FeS cluster and Moco biosynthesis. We also identified a mutant allele in CNX2, cofactor of nitrate reductase and xanthine dehydrogenase 2, encoding GTP 3',8-cyclase, the first step in Moco biosynthesis which is localized in the mitochondria. A single-nucleotide polymorphism in cnx2-2 leads to substitution of Arg88 with Gln in the N-terminal FeS cluster-binding motif. cnx2-2 plants are small and chlorotic, with severely decreased Moco enzyme activities, but they performed better than a cnx2-1 knockout mutant, which could only survive with ammonia as a nitrogen source. Measurement of cyclic pyranopterin monophosphate (cPMP) levels by LC-MS/MS showed that this Moco intermediate was below the limit of detection in both cnx2-1 and cnx2-2, and accumulated more than 10-fold in seedlings mutated in the downstream gene CNX5 Interestingly, atm3-1 mutants had less cPMP than wild type, correlating with previous reports of a similar decrease in nitrate reductase activity. Taken together, our data functionally characterize CNX2 and suggest that ATM3 is indirectly required for cPMP synthesis.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Arabidopsis Proteins/genetics , Arabidopsis/genetics , Gene Expression Regulation, Plant , Mitochondria/metabolism , Organophosphorus Compounds/metabolism , Pterins/metabolism , ATP-Binding Cassette Transporters/metabolism , Amino Acid Sequence , Ammonia/pharmacology , Arabidopsis/drug effects , Arabidopsis/growth & development , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Chromatography, Liquid , Coenzymes/biosynthesis , Gene Deletion , Metalloproteins/biosynthesis , Mitochondria/ultrastructure , Molybdenum Cofactors , Plant Cells/metabolism , Plant Cells/ultrastructure , Polymorphism, Single Nucleotide , Protein Isoforms/genetics , Protein Isoforms/metabolism , Pteridines , Seedlings/drug effects , Seedlings/genetics , Seedlings/growth & development , Seedlings/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Tandem Mass Spectrometry
12.
Z Naturforsch C J Biosci ; 74(3-4): 71-76, 2019 Feb 25.
Article in English | MEDLINE | ID: mdl-30685749

ABSTRACT

Multi-enzyme cascade reactions capture the essence of nature's efficiency by increasing the productivity of a process. Here we describe one such three-enzyme cascade for the synthesis of 6-hydroxyhexanoic acid. Whole cells of Escherichia coli co-expressing an alcohol dehydrogenase and a Baeyer-Villiger monooxygenase (CHMO) for internal cofactor regeneration were used without the supply of external NADPH or NADP+. The product inhibition caused by the ε-caprolactone formed by the CHMO was overcome by the use of lipase CAL-B for in situ conversion into 6-hydroxyhexanoic acid. A stirred tank reactor under fed-batch mode was chosen for efficient catalysis. By using this setup, a product titre of >20 g L-1 was achieved in a 500 mL scale with an isolated yield of 81% 6-hydroxyhexanoic acid.


Subject(s)
Alcohol Dehydrogenase/genetics , Caproates/chemical synthesis , Escherichia coli Proteins/genetics , Escherichia coli/enzymology , Fungal Proteins/chemistry , Hydroxy Acids/chemical synthesis , Lipase/chemistry , Mixed Function Oxygenases/genetics , Alcohol Dehydrogenase/metabolism , Batch Cell Culture Techniques , Biocatalysis , Bioreactors , Caproates/chemistry , Caproates/metabolism , Coenzymes/biosynthesis , Coenzymes/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , Fungal Proteins/metabolism , Gene Expression , Hydroxy Acids/metabolism , Kinetics , Lactones/chemistry , Lactones/metabolism , Lipase/metabolism , Mixed Function Oxygenases/metabolism , NADP/biosynthesis , NADP/chemistry
13.
Biochemistry ; 57(4): 390-402, 2018 01 30.
Article in English | MEDLINE | ID: mdl-29072833

ABSTRACT

The radical SAM (S-adenosyl-l-methionine) superfamily is one of the largest group of enzymes with >113000 annotated sequences [Landgraf, B. J., et al. (2016) Annu. Rev. Biochem. 85, 485-514]. Members of this superfamily catalyze the reductive cleavage of SAM using an oxygen sensitive 4Fe-4S cluster to transiently generate 5'-deoxyadenosyl radical that is subsequently used to initiate diverse free radical-mediated reactions. Because of the unique reactivity of free radicals, radical SAM enzymes frequently catalyze chemically challenging reactions critical for the biosynthesis of unique structures of cofactors and natural products. In this Perspective, I will discuss the impact of characterizing novel functions in radical SAM enzymes on our understanding of biosynthetic pathways and use two recent examples from my own group with a particular emphasis on two radical SAM enzymes that are responsible for carbon skeleton formation during the biosynthesis of a cofactor and natural products.


Subject(s)
Biological Products , Coenzymes/biosynthesis , Escherichia coli Proteins/metabolism , Isomerases/metabolism , Metalloproteins/biosynthesis , Nuclear Proteins/metabolism , Organophosphorus Compounds/metabolism , Pterins/metabolism , Carbon-Carbon Lyases , Crystallography, X-Ray , Guanosine Triphosphate/metabolism , Humans , Models, Molecular , Molecular Structure , Molybdenum Cofactors , Protein Conformation , Pteridines , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism
14.
Nat Prod Rep ; 35(7): 660-694, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29633774

ABSTRACT

Covering: up to the end of 2017 C-C bond formations are frequently the key steps in cofactor and natural product biosynthesis. Historically, C-C bond formations were thought to proceed by two electron mechanisms, represented by Claisen condensation in fatty acids and polyketide biosynthesis. These types of mechanisms require activated substrates to create a nucleophile and an electrophile. More recently, increasing number of C-C bond formations catalyzed by radical SAM enzymes are being identified. These free radical mediated reactions can proceed between almost any sp3 and sp2 carbon centers, allowing introduction of C-C bonds at unconventional positions in metabolites. Therefore, free radical mediated C-C bond formations are frequently found in the construction of structurally unique and complex metabolites. This review discusses our current understanding of the functions and mechanisms of C-C bond forming radical SAM enzymes and highlights their important roles in the biosynthesis of structurally complex, naturally occurring organic molecules. Mechanistic consideration of C-C bond formation by radical SAM enzymes identifies the significance of three key mechanistic factors: radical initiation, acceptor substrate activation and radical quenching. Understanding the functions and mechanisms of these characteristic enzymes will be important not only in promoting our understanding of radical SAM enzymes, but also for understanding natural product and cofactor biosynthesis.


Subject(s)
Biological Products/chemistry , Coenzymes/biosynthesis , Enzymes/chemistry , Enzymes/metabolism , S-Adenosylmethionine/metabolism , Adenine/analogs & derivatives , Adenine/biosynthesis , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteriochlorophylls/biosynthesis , Biological Products/metabolism , Carbon/chemistry , Coenzymes/chemistry , Endopeptidases/chemistry , Endopeptidases/metabolism , Hydrolases/chemistry , Hydrolases/metabolism , Molybdenum/chemistry , Molybdenum/metabolism , Tunicamycin/biosynthesis , Vitamin K 2/metabolism
15.
Environ Microbiol ; 20(8): 2809-2823, 2018 08.
Article in English | MEDLINE | ID: mdl-29659156

ABSTRACT

Aquatic environments contain large communities of microorganisms whose synergistic interactions mediate the cycling of major and trace nutrients, including vitamins. B-vitamins are essential coenzymes that many organisms cannot synthesize. Thus, their exchange among de novo synthesizers and auxotrophs is expected to play an important role in the microbial consortia and explain some of the temporal and spatial changes observed in diversity. In this study, we analyzed metatranscriptomes of a natural marine microbial community, diel sampled quarterly over one year to try to identify the potential major B-vitamin synthesizers and consumers. Transcriptomic data showed that the best-represented taxa dominated the expression of synthesis genes for some B-vitamins but lacked transcripts for others. For instance, Rhodobacterales dominated the expression of vitamin-B12 synthesis, but not of vitamin-B7 , whose synthesis transcripts were mainly represented by Flavobacteria. In contrast, bacterial groups that constituted less than 4% of the community (e.g., Verrucomicrobia) accounted for most of the vitamin-B1 synthesis transcripts. Furthermore, ambient vitamin-B1 concentrations were higher in samples collected during the day, and were positively correlated with chlorophyll-a concentrations. Our analysis supports the hypothesis that the mosaic of metabolic interdependencies through B-vitamin synthesis and exchange are key processes that contribute to shaping microbial communities in nature.


Subject(s)
Bacteria/metabolism , Microbial Consortia , Vitamin B Complex/metabolism , Alphaproteobacteria/genetics , Alphaproteobacteria/metabolism , Bacteria/genetics , Coenzymes/biosynthesis , Coenzymes/metabolism , Flavobacteriaceae/genetics , Flavobacteriaceae/metabolism , Transcriptome , Vitamin B Complex/biosynthesis
16.
Arch Biochem Biophys ; 654: 40-46, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30026025

ABSTRACT

Tryptophyquinone-bearing enzymes contain protein-derived cofactors formed by posttranslational modifications of Trp residues. Tryptophan tryptophylquinone (TTQ) is comprised of a di-oxygenated Trp residue, which is cross-linked to another Trp residue. Cysteine tryptophylquinone (CTQ) is comprised of a di-oxygenated Trp residue, which is cross-linked to a Cys residue. Despite the similarity of these cofactors, it has become evident in recent years that the overall structures of the enzymes that possess these cofactors vary, and that the gene clusters that encode the enzymes are quite diverse. While it had been long assumed that all tryptophylquinone enzymes were dehydrogenases, recently discovered classes of these enzymes are oxidases. A common feature of enzymes that have these cofactors is that the posttranslational modifications that form the mature cofactors are catalyzed by a modifying enzyme. However, it is now clear that modifying enzymes are different for different tryptophylquinone enzymes. For methylamine dehydrogenase a di-heme enzyme, MauG, is needed to catalyze TTQ biosynthesis. However, no gene similar to mauG is present in the gene clusters that encode the other enzymes, and the recently characterized family of CTQ-dependent oxidases, termed LodA-like proteins, require a flavoenzyme for cofactor biosynthesis.


Subject(s)
Coenzymes/biosynthesis , Coenzymes/chemistry , Indolequinones/metabolism , Tryptophan/analogs & derivatives , Catalysis , Protein Conformation , Protein Processing, Post-Translational , Tryptophan/metabolism
17.
Proc Natl Acad Sci U S A ; 112(20): 6347-52, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25941396

ABSTRACT

The molybdenum cofactor (Moco) is essential for all kingdoms of life, plays central roles in various biological processes, and must be biosynthesized de novo. During Moco biosynthesis, the characteristic pyranopterin ring is constructed by a complex rearrangement of guanosine 5'-triphosphate (GTP) into cyclic pyranopterin (cPMP) through the action of two enzymes, MoaA and MoaC (molybdenum cofactor biosynthesis protein A and C, respectively). Conventionally, MoaA was considered to catalyze the majority of this transformation, with MoaC playing little or no role in the pyranopterin formation. Recently, this view was challenged by the isolation of 3',8-cyclo-7,8-dihydro-guanosine 5'-triphosphate (3',8-cH2GTP) as the product of in vitro MoaA reactions. To elucidate the mechanism of formation of Moco pyranopterin backbone, we performed biochemical characterization of 3',8-cH2GTP and functional and X-ray crystallographic characterizations of MoaC. These studies revealed that 3',8-cH2GTP is the only product of MoaA that can be converted to cPMP by MoaC. Our structural studies captured the specific binding of 3',8-cH2GTP in the active site of MoaC. These observations provided strong evidence that the physiological function of MoaA is the conversion of GTP to 3',8-cH2GTP (GTP 3',8-cyclase), and that of MoaC is to catalyze the rearrangement of 3',8-cH2GTP into cPMP (cPMP synthase). Furthermore, our structure-guided studies suggest that MoaC catalysis involves the dynamic motions of enzyme active-site loops as a way to control the timing of interaction between the reaction intermediates and catalytically essential amino acid residues. Thus, these results reveal the previously unidentified mechanism behind Moco biosynthesis and provide mechanistic and structural insights into how enzymes catalyze complex rearrangement reactions.


Subject(s)
Coenzymes/biosynthesis , Escherichia coli Proteins/metabolism , Hydrolases/metabolism , Metalloproteins/biosynthesis , Models, Molecular , Pterins/chemistry , Carbon Isotopes , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Hydrolases/chemistry , Hydrolases/isolation & purification , Magnetic Resonance Spectroscopy , Molecular Structure , Molybdenum Cofactors , Mutagenesis, Site-Directed , Protein Conformation , Pteridines
18.
Molecules ; 23(12)2018 Dec 11.
Article in English | MEDLINE | ID: mdl-30545001

ABSTRACT

All eukaryotic molybdenum (Mo) enzymes contain in their active site a Mo Cofactor (Moco), which is formed by a tricyclic pyranopterin with a dithiolene chelating the Mo atom. Here, the eukaryotic Moco biosynthetic pathway and the eukaryotic Moco enzymes are overviewed, including nitrate reductase (NR), sulfite oxidase, xanthine oxidoreductase, aldehyde oxidase, and the last one discovered, the moonlighting enzyme mitochondrial Amidoxime Reducing Component (mARC). The mARC enzymes catalyze the reduction of hydroxylated compounds, mostly N-hydroxylated (NHC), but as well of nitrite to nitric oxide, a second messenger. mARC shows a broad spectrum of NHC as substrates, some are prodrugs containing an amidoxime structure, some are mutagens, such as 6-hydroxylaminepurine and some others, which most probably will be discovered soon. Interestingly, all known mARC need the reducing power supplied by different partners. For the NHC reduction, mARC uses cytochrome b5 and cytochrome b5 reductase, however for the nitrite reduction, plant mARC uses NR. Despite the functional importance of mARC enzymatic reactions, the structural mechanism of its Moco-mediated catalysis is starting to be revealed. We propose and compare the mARC catalytic mechanism of nitrite versus NHC reduction. By using the recently resolved structure of a prokaryotic MOSC enzyme, from the mARC protein family, we have modeled an in silico three-dimensional structure of a eukaryotic homologue.


Subject(s)
Coenzymes/metabolism , Enzymes/metabolism , Metalloproteins/metabolism , Pteridines/metabolism , Animals , Cardiac Myosins/metabolism , Coenzymes/biosynthesis , Enzymes/chemistry , Enzymes/genetics , Eukaryotic Cells/metabolism , Mammals , Metabolic Networks and Pathways , Metalloproteins/biosynthesis , Molybdenum/metabolism , Molybdenum Cofactors , Myosin Light Chains/metabolism , Nitrate Reductase/metabolism , Nitrites/metabolism , Oxidoreductases/genetics , Oxidoreductases/metabolism
19.
Biochemistry ; 56(34): 4592-4605, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28766335

ABSTRACT

The trafficking and delivery of sulfur to cofactors and nucleosides is a highly regulated and conserved process among all organisms. All sulfur transfer pathways generally have an l-cysteine desulfurase as an initial sulfur-mobilizing enzyme in common, which serves as a sulfur donor for the biosynthesis of sulfur-containing biomolecules like iron-sulfur (Fe-S) clusters, thiamine, biotin, lipoic acid, the molybdenum cofactor (Moco), and thiolated nucleosides in tRNA. The human l-cysteine desulfurase NFS1 and the Escherichia coli homologue IscS share a level of amino acid sequence identity of ∼60%. While E. coli IscS has a versatile role in the cell and was shown to have numerous interaction partners, NFS1 is mainly localized in mitochondria with a crucial role in the biosynthesis of Fe-S clusters. Additionally, NFS1 is also located in smaller amounts in the cytosol with a role in Moco biosynthesis and mcm5s2U34 thio modifications of nucleosides in tRNA. NFS1 and IscS were conclusively shown to have different interaction partners in their respective organisms. Here, we used functional complementation studies of an E. coli iscS deletion strain with human NFS1 to dissect their conserved roles in the transfer of sulfur to a specific target protein. Our results show that human NFS1 and E. coli IscS share conserved binding sites for proteins involved in Fe-S cluster assembly like IscU, but not with proteins for tRNA thio modifications or Moco biosynthesis. In addition, we show that human NFS1 was almost fully able to complement the role of IscS in Moco biosynthesis when its specific interaction partner protein MOCS3 from humans was also present.


Subject(s)
Carbon-Sulfur Lyases , Coenzymes , Escherichia coli , Genetic Complementation Test , Metalloproteins , Pteridines , Binding Sites , Carbon-Sulfur Lyases/genetics , Carbon-Sulfur Lyases/metabolism , Coenzymes/biosynthesis , Coenzymes/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Humans , Metalloproteins/biosynthesis , Metalloproteins/genetics , Molybdenum Cofactors , Nucleotidyltransferases/metabolism , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Transfer/metabolism , Sulfurtransferases/metabolism
20.
Biochemistry ; 56(14): 1987-2000, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28323419

ABSTRACT

In Escherichia coli, two different systems that are important for the coordinate formation of Fe-S clusters have been identified, namely, the ISC and SUF systems. The ISC system is the housekeeping Fe-S machinery, which provides Fe-S clusters for numerous cellular proteins. The IscS protein of this system was additionally revealed to be the primary sulfur donor for several sulfur-containing molecules with important biological functions, among which are the molybdenum cofactor (Moco) and thiolated nucleosides in tRNA. Here, we show that deletion of central components of the ISC system in addition to IscS leads to an overall decrease in Fe-S cluster enzyme and molybdoenzyme activity in addition to a decrease in the number of Fe-S-dependent thiomodifications of tRNA, based on the fact that some proteins involved in Moco biosynthesis and tRNA thiolation are Fe-S-dependent. Complementation of the ISC deficient strains with the suf operon restored the activity of Fe-S-containing proteins, including the MoaA protein, which is involved in the conversion of 5'GTP to cyclic pyranopterin monophosphate in the fist step of Moco biosynthesis. While both systems share a high degree of similarity, we show that the function of their respective l-cysteine desulfurase IscS or SufS is specific for each cellular pathway. It is revealed that SufS cannot play the role of IscS in sulfur transfer for the formation of 2-thiouridine, 4-thiouridine, or the dithiolene group of molybdopterin, being unable to interact with TusA or ThiI. The results demonstrate that the role of the SUF system is exclusively restricted to Fe-S cluster assembly in the cell.


Subject(s)
Carbon-Sulfur Lyases/metabolism , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Iron-Sulfur Proteins/metabolism , Lyases/metabolism , Carbon-Sulfur Lyases/genetics , Coenzymes/biosynthesis , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Iron-Sulfur Proteins/genetics , Isomerases/genetics , Isomerases/metabolism , Lyases/genetics , Metalloproteins/biosynthesis , Molybdenum Cofactors , Operon , Pteridines , RNA, Transfer/genetics , RNA, Transfer/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sulfurtransferases/genetics , Sulfurtransferases/metabolism , Thiouridine/analogs & derivatives , Thiouridine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL