Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
1.
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
2.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119731, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38631442

ABSTRACT

Molybdenum cofactor (Moco) biosynthesis is a complex process that involves the coordinated function of several proteins. In the recent years it has become evident that the availability of Fe-S clusters play an important role for 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 an enzyme involved in the transfer of sulfur to various acceptor proteins with a main role in the assembly of Fe-S clusters. In this review, we dissect the dependence of the production of active molybdoenzymes in detail, starting from the regulation of gene expression and further explaining sulfur delivery and Fe-S cluster insertion into target enzymes. Further, Fe-S cluster assembly is also linked to iron availability. While the abundance of selected molybdoenzymes is largely decreased under iron-limiting conditions, we explain that the expression of the genes is dependent on an active FNR protein. FNR is a very important transcription factor that represents the master-switch for the expression of target genes in response to anaerobiosis. Moco biosynthesis is further directly dependent on the presence of ArcA and also on an active Fur protein.


Subject(s)
Coenzymes , Iron-Sulfur Proteins , Metalloproteins , Molybdenum Cofactors , Pteridines , Metalloproteins/metabolism , Metalloproteins/genetics , Metalloproteins/biosynthesis , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Coenzymes/metabolism , Coenzymes/biosynthesis , Coenzymes/genetics , Pteridines/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Iron/metabolism , Sulfur/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Carbon-Sulfur Lyases/metabolism , Carbon-Sulfur Lyases/genetics , Gene Expression Regulation, Bacterial , Operon , Isomerases
3.
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
4.
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
5.
Nat Commun ; 12(1): 2116, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837188

ABSTRACT

Nicotinamide adenine dinucleotide (NAD) and its reduced form are indispensable cofactors in life. Diverse NAD mimics have been developed for applications in chemical and biological sciences. Nicotinamide cytosine dinucleotide (NCD) has emerged as a non-natural cofactor to mediate redox transformations, while cells are fed with chemically synthesized NCD. Here, we create NCD synthetase (NcdS) by reprograming the substrate binding pockets of nicotinic acid mononucleotide (NaMN) adenylyltransferase to favor cytidine triphosphate and nicotinamide mononucleotide over their regular substrates ATP and NaMN, respectively. Overexpression of NcdS alone in the model host Escherichia coli facilitated intracellular production of NCD, and higher NCD levels up to 5.0 mM were achieved upon further pathway regulation. Finally, the non-natural cofactor self-sufficiency was confirmed by mediating an NCD-linked metabolic circuit to convert L-malate into D-lactate. NcdS together with NCD-linked enzymes offer unique tools and opportunities for intriguing studies in chemical biology and synthetic biology.


Subject(s)
Coenzymes/biosynthesis , Escherichia coli Proteins/genetics , Niacinamide/biosynthesis , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Protein Engineering , Coenzymes/chemistry , Cytidine Triphosphate/metabolism , Enzyme Assays , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/metabolism , High-Throughput Screening Assays , Lactic Acid/metabolism , Malates/metabolism , Niacinamide/chemistry , Nicotinamide Mononucleotide/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Oxidation-Reduction , Substrate Specificity/genetics , Synthetic Biology/methods
6.
Biochimie ; 184: 143-157, 2021 May.
Article in English | MEDLINE | ID: mdl-33675854

ABSTRACT

There are multiple lines of evidence for an impaired sulfur amino acid (SAA) metabolism in autism spectrum disorder (ASD). For instance, the concentrations of methionine, cysteine and S-adenosylmethionine (SAM) in body fluids of individuals with ASD is significantly lower while the concentration of S-adenosylhomocysteine (SAH) is significantly higher as compared to healthy individuals. Reduced methionine and SAM may reflect impaired remethylation pathway whereas increased SAH may reflect reduced S-adenosylhomocysteine hydrolase activity in the catabolic direction. Reduced SAM/SAH ratio reflects an impaired methylation capacity. We hypothesize multiple mechanisms to explain how the interplay of oxidative stress, neuroinflammation, mercury exposure, maternal use of valproate, altered gut microbiome and certain genetic variants may lead to these SAA metabotypes. Furthermore, we also propose a number of mechanisms to explain the metabolic consequences of abnormal SAA metabotypes. For instance in the brain, reduced SAM/SAH ratio will result in melatonin deficiency and hypomethylation of a number of biomolecules such as DNA, RNA and histones. In addition to previously proposed mechanisms, we propose that impaired activity of "radical SAM" enzymes will result in reduced endogenous lipoic acid synthesis, reduced molybdenum cofactor synthesis and impaired porphyrin metabolism leading to mitochondrial dysfunction, porphyrinuria and impaired sulfation capacity. Furthermore depletion of SAM may also lead to the disturbed mTOR signaling pathway in a subgroup of ASD. The proposed "SAM-depletion hypothesis" is an inclusive model to explain the relationship between heterogeneous risk factors and metabotypes observed in a subset of children with ASD.


Subject(s)
Autism Spectrum Disorder/metabolism , Coenzymes/biosynthesis , Cysteine/metabolism , Metalloproteins/biosynthesis , Oxidative Stress , S-Adenosylmethionine/metabolism , Brain , Humans , Molybdenum Cofactors , Pteridines
7.
Biochim Biophys Acta Mol Cell Res ; 1868(1): 118883, 2021 01.
Article in English | MEDLINE | ID: mdl-33017596

ABSTRACT

The molybdenum cofactor (Moco) represents an ancient metal­sulfur cofactor, which participates as catalyst in carbon, nitrogen and sulfur cycles, both on individual and global scale. Given the diversity of biological processes dependent on Moco and their evolutionary age, Moco is traced back to the last universal common ancestor (LUCA), while Moco biosynthetic genes underwent significant changes through evolution and acquired additional functions. In this review, focused on eukaryotic Moco biology, we elucidate the benefits of gene fusions on Moco biosynthesis and beyond. While originally the gene fusions were driven by biosynthetic advantages such as coordinated expression of functionally related proteins and product/substrate channeling, they also served as origin for the development of novel functions. Today, Moco biosynthetic genes are involved in a multitude of cellular processes and loss of the according gene products result in severe disorders, both related to Moco biosynthesis and secondary enzyme functions.


Subject(s)
Coenzymes/genetics , Eukaryota/genetics , Metalloproteins/genetics , Molybdenum/metabolism , Coenzymes/biosynthesis , Coenzymes/classification , Gene Fusion/genetics , Humans , Metalloproteins/biosynthesis , Metalloproteins/classification , Molybdenum Cofactors , Pteridines/classification , Substrate Specificity
8.
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
9.
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
10.
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
11.
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
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.
Mol Brain ; 12(1): 1, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30606245

ABSTRACT

Natural aggressiveness is commonly observed in all animal species, and is displayed frequently when animals compete for food, territory and mating. Aggression is an innate behaviour, and is influenced by both environmental and genetic factors. However, the genetics of aggression remains largely unclear. In this study, we identify the peacefulness (pfs) gene as a novel player in the control of male-male aggression in Drosophila. Mutations in pfs decreased intermale aggressiveness, but did not affect locomotor activity, olfactory avoidance response and sexual behaviours. pfs encodes for the evolutionarily conserved molybdenum cofactor (MoCo) synthesis 1 protein (Mocs1), which catalyzes the first step in the MoCo biosynthesis pathway. Neuronal-specific knockdown of pfs decreased aggressiveness. By contrast, overexpression of pfs greatly increased aggressiveness. Knocking down Cinnamon (Cin) catalyzing the final step in the MoCo synthesis pathway, caused a pfs-like aggression phenotype. In humans, inhibition of MoCo-dependent enzymes displays anti-aggressive effects. Thus, the control of aggression by Pfs-dependent MoCo pathways may be conserved throughout evolution.


Subject(s)
Aggression/physiology , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Genes, Insect , Nuclear Proteins/genetics , Animals , Avoidance Learning , Brain/metabolism , Carbon-Carbon Lyases , Coenzymes/biosynthesis , Drosophila Proteins/metabolism , Gene Knockdown Techniques , Male , Metalloproteins/biosynthesis , Molybdenum Cofactors , Motor Activity , Mutagenesis, Insertional/genetics , Neurons/metabolism , Nuclear Proteins/metabolism , Pteridines , Sexual Behavior, Animal , Smell/physiology
14.
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
15.
Microb Biotechnol ; 12(2): 346-359, 2019 03.
Article in English | MEDLINE | ID: mdl-30549216

ABSTRACT

Cobamides (Cbas) are essential cofactors of reductive dehalogenases (RDases) in organohalide-respiring bacteria (OHRB). Changes in the Cba structure can influence RDase function. Here, we report on the cofactor versatility or selectivity of Desulfitobacterium RDases produced either in the native organism or heterologously. The susceptibility of Desulfitobacterium hafniense strain DCB-2 to guided Cba biosynthesis (i.e. incorporation of exogenous Cba lower ligand base precursors) was analysed. Exogenous benzimidazoles, azabenzimidazoles and 4,5-dimethylimidazole were incorporated by the organism into Cbas. When the type of Cba changed, no effect on the turnover rate of the 3-chloro-4-hydroxy-phenylacetate-converting enzyme RdhA6 and the 3,5-dichlorophenol-dehalogenating enzyme RdhA3 was observed. The impact of the amendment of Cba lower ligand precursors on RDase function was also investigated in Shimwellia blattae, the Cba producer used for the heterologous production of Desulfitobacterium RDases. The recombinant tetrachloroethene RDase (PceAY51 ) appeared to be non-selective towards different Cbas. However, the functional production of the 1,2-dichloroethane-dihaloeliminating enzyme (DcaA) of Desulfitobacterium dichloroeliminans was completely prevented in cells producing 5,6-dimethylbenzimidazolyl-Cba, but substantially enhanced in cells that incorporated 5-methoxybenzimidazole into the Cba cofactor. The results of the study indicate the utilization of a range of different Cbas by Desulfitobacterium RDases with selected representatives apparently preferring distinct Cbas.


Subject(s)
Cobamides/biosynthesis , Coenzymes/biosynthesis , Desulfitobacterium/enzymology , Enterobacteriaceae/enzymology , Hydrolases/metabolism , Vitamin B Complex/biosynthesis
16.
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
17.
Methods Enzymol ; 606: 485-522, 2018.
Article in English | MEDLINE | ID: mdl-30097104

ABSTRACT

MoaA is one of the founding members of the radical S-adenosyl-L-methionine (SAM) superfamily, and together with the second enzyme, MoaC, catalyzes the construction of the pyranopterin backbone structure of the molybdenum cofactor (Moco). However, the exact functions of both MoaA and MoaC had remained ambiguous for more than 2 decades. Recently, their functions were finally elucidated through successful characterization of the MoaA product as 3',8-cyclo-7,8-dihydro-GTP (3',8-cH2GTP), which was shown to be converted to cyclic pyranopterin monophosphate (cPMP) by MoaC. 3',8-cH2GTP was produced in a small quantity and was highly oxygen sensitive, which explains why this compound had previously eluded characterization. This chapter describes the methodologies for the characterization of MoaA, MoaC, and 3',8-cH2GTP, which together significantly altered the view of the mechanism of the pyranopterin backbone construction during the Moco biosynthesis. Through this chapter, we hope to share not only the protocols to study the first step of Moco biosynthesis but also the lessons we learned from the characterization of the chemically labile biosynthetic intermediate, which would be informative for the study of many other metabolic pathways and enzymes.


Subject(s)
Coenzymes/biosynthesis , Enzyme Assays/methods , Escherichia coli Proteins/metabolism , Hydrolases/metabolism , Metalloproteins/biosynthesis , Escherichia coli Proteins/isolation & purification , Hydrolases/isolation & purification , Metabolic Networks and Pathways , Molybdenum Cofactors , Organophosphorus Compounds/metabolism , Pteridines , Pterins/metabolism , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
18.
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
19.
Insect Biochem Mol Biol ; 99: 11-16, 2018 08.
Article in English | MEDLINE | ID: mdl-29803701

ABSTRACT

Translucency of the larval integument in Bombyx mori is caused by a lack of uric acid in the epidermis. Hime'nichi translucent (ohi) is a unique mutation causing intermediate translucency of the larval integument and male-specific flaccid paralysis. To determine the gene associated with the ohi mutation, the ohi locus was mapped to a 400-kb region containing 29 predicted genes. Among the genes in this region, we focused on Bombyx homolog of mammalian Gephyrin (BmGphn), which regulates molybdenum cofactor (MoCo) biosynthesis, because MoCo is indispensable for the activity of xanthine dehydrogenase (XDH), a key enzyme in uric acid biosynthesis. The translucent integument of ohi larvae turned opaque after injection of bovine xanthine oxidase, which is a mammalian equivalent to XDH, indicating that XDH activity is defective in ohi larvae. RT-PCR and sequencing analysis showed that (i) in ohi larvae, expression of the BmGphn gene was repressed in the fat body where uric acid is synthesized, and (ii) there was no amino acid substitution in the ohi mutant allele. Finally, we obtained BmGphn knockout alleles (hereafter denoted as BmGphnΔ) by using CRISPR/Cas9. The resulting ohi/BmGphnΔ larvae had translucent integuments, demonstrating that BmGphn is the gene responsible for the ohi phenotype. Our results show that repressed expression of BmGphn is a causative factor for the defective MoCo biosynthesis and XDH activity observed in ohi larvae. Interestingly, all male BmGphnΔ homozygotes died before pupation and showed a flaccid paralysis phenotype. The genetic and physiological mechanisms underlying this flaccid paralysis phenotype are also discussed.


Subject(s)
Bombyx , Coenzymes , Gene Editing , Insect Proteins , Metalloproteins , Pteridines , Animals , Bombyx/genetics , Bombyx/metabolism , Coenzymes/biosynthesis , Coenzymes/genetics , Insect Proteins/genetics , Insect Proteins/metabolism , Larva , Metalloproteins/biosynthesis , Metalloproteins/genetics , Molybdenum Cofactors
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL