Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 645
Filter
1.
Life Sci Alliance ; 7(11)2024 Nov.
Article in English | MEDLINE | ID: mdl-39159974

ABSTRACT

Regeneration of insulin-producing ß-cells is an alternative avenue to manage diabetes, and it is crucial to unravel this process in vivo during physiological responses to the lack of ß-cells. Here, we aimed to characterize how hepatocytes can contribute to ß-cell regeneration, either directly or indirectly via secreted proteins or metabolites, in a zebrafish model of ß-cell loss. Using lineage tracing, we show that hepatocytes do not directly convert into ß-cells even under extreme ß-cell ablation conditions. A transcriptomic analysis of isolated hepatocytes after ß-cell ablation displayed altered lipid- and glucose-related processes. Based on the transcriptomics, we performed a genetic screen that uncovers a potential role of the molybdenum cofactor (Moco) biosynthetic pathway in ß-cell regeneration and glucose metabolism in zebrafish. Consistently, molybdenum cofactor synthesis 2 (Mocs2) haploinsufficiency in mice indicated dysregulated glucose metabolism and liver function. Together, our study sheds light on the liver-pancreas crosstalk and suggests that the molybdenum cofactor biosynthesis pathway should be further studied in relation to glucose metabolism and diabetes.


Subject(s)
Coenzymes , Glucose , Hepatocytes , Insulin-Secreting Cells , Liver , Metalloproteins , Molybdenum Cofactors , Pteridines , Zebrafish , Animals , Insulin-Secreting Cells/metabolism , Pteridines/metabolism , Coenzymes/metabolism , Mice , Liver/metabolism , Liver/cytology , Metalloproteins/metabolism , Metalloproteins/genetics , Hepatocytes/metabolism , Glucose/metabolism , Regeneration/genetics , Pancreas/metabolism , Pancreas/cytology , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
2.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119794, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39033933

ABSTRACT

Sulfur-containing biomolecules such as [FeS] clusters, thiamin, biotin, molybdenum cofactor, and sulfur-containing tRNA nucleosides are essential for various biochemical reactions. The amino acid l-cysteine serves as the major sulfur source for the biosynthetic pathways of these sulfur-containing cofactors in prokaryotic and eukaryotic systems. The first reaction in the sulfur mobilization involves a class of pyridoxal-5'-phosphate (PLP) dependent enzymes catalyzing a Cys:sulfur acceptor sulfurtransferase reaction. The first half of the catalytic reaction involves a PLP-dependent CS bond cleavage, resulting in a persulfide enzyme intermediate. The second half of the reaction involves the subsequent transfer of the thiol group to a specific acceptor molecule, which is responsible for the physiological role of the enzyme. Structural and biochemical analysis of these Cys sulfurtransferase enzymes shows that specific protein-protein interactions with sulfur acceptors modulate their catalytic reactivity and restrict their biochemical functions.


Subject(s)
Cysteine , Pyridoxal Phosphate , Sulfur , Sulfurtransferases , Sulfur/metabolism , Sulfur/chemistry , Cysteine/metabolism , Cysteine/chemistry , Sulfurtransferases/metabolism , Sulfurtransferases/chemistry , Pyridoxal Phosphate/metabolism , Humans , Molybdenum Cofactors , Carbon-Sulfur Lyases/metabolism , Carbon-Sulfur Lyases/chemistry
3.
Neural Dev ; 19(1): 14, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39068495

ABSTRACT

Gephyrin is thought to play a critical role in clustering glycine receptors at synapses within the central nervous system (CNS). The main in vivo evidence for this comes from Gephyrin (Gphn)-null mice, where glycine receptors are depleted from synaptic regions. However, these mice die at birth, possibly due to impaired molybdenum cofactor (MoCo) synthesis, an essential role Gephyrin assumes throughout an animal. This complicates the interpretation of synaptic phenotypes in Gphn-null mice and raises the question whether the synaptic and enzymatic functions of Gephyrin can be investigated separately. Here, we generated a gephyrinb zebrafish mutant, vo84, that almost entirely lacks Gephyrin staining in the spinal cord. gephyrinbvo84 mutants exhibit normal gross morphology at both larval and adult stages. In contrast to Gphn-null mice, gephyrinbvo84 mutants exhibit normal motor activity and MoCo-dependent enzyme activity. Instead, gephyrinbvo84 mutants display impaired rheotaxis and increased mortality in late development. To investigate what may mediate these defects in gephyrinbvo84 mutants, we examined the cell density of neurons and myelin in the spinal cord and found no obvious changes. Surprisingly, in gephyrinbvo84 mutants, glycine receptors are still present in the synaptic regions. However, their abundance is reduced, potentially contributing to the observed defects. These findings challenge the notion that Gephyrin is absolutely required to cluster glycine receptors at synapses and reveals a new role of Gephyrin in regulating glycine receptor abundance and rheotaxis. They also establish a powerful new model for studying the mechanisms underlying synaptic, rather than enzymatic, functions of Gephyrin.


Subject(s)
Carrier Proteins , Membrane Proteins , Mutation , Spinal Cord , Synapses , Zebrafish , Animals , Synapses/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Spinal Cord/metabolism , Mutation/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Receptors, Glycine/metabolism , Receptors, Glycine/genetics , Molybdenum Cofactors , Pteridines , Neurons/metabolism , Myelin Sheath/metabolism , Motor Activity/physiology , Motor Activity/genetics , Animals, Genetically Modified
4.
Proc Natl Acad Sci U S A ; 121(25): e2319903121, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38870058

ABSTRACT

Biofilm formation and surface attachment in multiple Alphaproteobacteria is driven by unipolar polysaccharide (UPP) adhesins. The pathogen Agrobacterium tumefaciens produces a UPP adhesin, which is regulated by the intracellular second messenger cyclic diguanylate monophosphate (c-di-GMP). Prior studies revealed that DcpA, a diguanylate cyclase-phosphodiesterase, is crucial in control of UPP production and surface attachment. DcpA is regulated by PruR, a protein with distant similarity to enzymatic domains known to coordinate the molybdopterin cofactor (MoCo). Pterins are bicyclic nitrogen-rich compounds, several of which are produced via a nonessential branch of the folate biosynthesis pathway, distinct from MoCo. The pterin-binding protein PruR controls DcpA activity, fostering c-di-GMP breakdown and dampening its synthesis. Pterins are excreted, and we report here that PruR associates with these metabolites in the periplasm, promoting interaction with the DcpA periplasmic domain. The pteridine reductase PruA, which reduces specific dihydro-pterin molecules to their tetrahydro forms, imparts control over DcpA activity through PruR. Tetrahydromonapterin preferentially associates with PruR relative to other related pterins, and the PruR-DcpA interaction is decreased in a pruA mutant. PruR and DcpA are encoded in an operon with wide conservation among diverse Proteobacteria including mammalian pathogens. Crystal structures reveal that PruR and several orthologs adopt a conserved fold, with a pterin-specific binding cleft that coordinates the bicyclic pterin ring. These findings define a pterin-responsive regulatory mechanism that controls biofilm formation and related c-di-GMP-dependent phenotypes in A. tumefaciens and potentially acts more widely in multiple proteobacterial lineages.


Subject(s)
Agrobacterium tumefaciens , Bacterial Proteins , Biofilms , Cyclic GMP , Pterins , Biofilms/growth & development , Agrobacterium tumefaciens/metabolism , Agrobacterium tumefaciens/genetics , Pterins/metabolism , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Proteobacteria/metabolism , Proteobacteria/genetics , Molybdenum Cofactors , Periplasm/metabolism , Periplasmic Proteins/metabolism , Periplasmic Proteins/genetics , Periplasmic Binding Proteins/metabolism , Periplasmic Binding Proteins/genetics , Gene Expression Regulation, Bacterial
5.
Microbiol Spectr ; 12(8): e0055624, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38916309

ABSTRACT

All sulfur transfer pathways have generally a l-cysteine desulfurase as an initial sulfur-mobilizing enzyme in common, which serves as a sulfur donor for the biosynthesis of numerous sulfur-containing biomolecules in the cell. In Escherichia coli, the housekeeping l-cysteine desulfurase IscS has several interaction partners, which bind at different sites of the protein. So far, the interaction sites of IscU, Fdx, CyaY, and IscX involved in iron-sulfur (Fe-S) cluster assembly have been mapped, in addition to TusA, which is required for molybdenum cofactor biosynthesis and mnm5s2U34 tRNA modifications, and ThiI, which is involved in thiamine biosynthesis and s4U8 tRNA modifications. Previous studies predicted that the sulfur acceptor proteins bind to IscS one at a time. E. coli TusA has, however, been suggested to be involved in Fe-S cluster assembly, as fewer Fe-S clusters were detected in a ∆tusA mutant. The basis for this reduction in Fe-S cluster content is unknown. In this work, we investigated the role of TusA in iron-sulfur cluster assembly and iron homeostasis. We show that the absence of TusA reduces the translation of fur, thereby leading to pleiotropic cellular effects, which we dissect in detail in this study.IMPORTANCEIron-sulfur clusters are evolutionarily ancient prosthetic groups. The ferric uptake regulator plays a major role in controlling the expression of iron homeostasis genes in bacteria. We show that a ∆tusA mutant is impaired in the assembly of Fe-S clusters and accumulates iron. TusA, therefore, reduces fur mRNA translation leading to pleiotropic cellular effects.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Homeostasis , Iron-Sulfur Proteins , Iron , Repressor Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/genetics , Iron/metabolism , Iron-Sulfur Proteins/metabolism , Iron-Sulfur Proteins/genetics , Repressor Proteins/metabolism , Repressor Proteins/genetics , Carbon-Sulfur Lyases/metabolism , Carbon-Sulfur Lyases/genetics , Gene Expression Regulation, Bacterial , Sulfur/metabolism , Protein Biosynthesis , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Pteridines/metabolism , Molybdenum Cofactors
6.
J Inherit Metab Dis ; 47(4): 598-623, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38627985

ABSTRACT

Sulfite intoxication is the hallmark of four ultrarare disorders that are caused by impaired sulfite oxidase activity due to genetic defects in the synthesis of the molybdenum cofactor or of the apoenzyme sulfite oxidase. Delays on the diagnosis of these disorders are common and have been caused by their unspecific presentation of acute neonatal encephalopathy with high early mortality, followed by the evolution of dystonic cerebral palsy and also by the lack of easily available and reliable diagnostic tests. There is significant variation in survival and in the quality of symptomatic management of affected children. One of the four disorders, molybdenum cofactor deficiency type A (MoCD-A) has recently become amenable to causal treatment with synthetic cPMP (fosdenopterin). The evidence base for the rational use of cPMP is very limited. This prompted the formulation of these clinical guidelines to facilitate diagnosis and support the management of patients. The guidelines were developed by experts in diagnosis and treatment of sulfite intoxication disorders. It reflects expert consensus opinion and evidence from a systematic literature search.


Subject(s)
Metal Metabolism, Inborn Errors , Sulfite Oxidase , Humans , Infant, Newborn , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/genetics , Coenzymes/deficiency , Consensus , Metal Metabolism, Inborn Errors/diagnosis , Metal Metabolism, Inborn Errors/therapy , Metalloproteins/deficiency , Molybdenum Cofactors , Pteridines , Sulfite Oxidase/deficiency , Sulfite Oxidase/genetics
7.
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
8.
Insect Mol Biol ; 33(3): 246-258, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38323672

ABSTRACT

Molybdenum cofactor sulfurase (MoCoS) is a key gene involved in the uric acid metabolic pathway that activates xanthine dehydrogenase to synthesise uric acid. Uric acid is harmful to mammals but plays crucial roles in insects, one of which is the immune responses. However, the function of Bombyx mori MoCoS in response to BmNPV remains unclear. In this study, BmMoCoS was found to be relatively highly expressed in embryonic development, gonads and the Malpighian tubules. In addition, the expression levels of BmMoCoS were significantly upregulated in three silkworm strains with different levels of resistance after virus infection, suggesting a close link between them. Furthermore, RNAi and overexpression studies showed that BmMoCoS was involved in resistance to BmNPV infection, and its antivirus effects were found to be related to the regulation of uric acid metabolism, which was uncovered by inosine- and febuxostat-coupled RNAi and overexpression. Finally, the BmMoCoS-mediated uric acid pathway was preliminarily confirmed to be a potential target to protect silkworms from BmNPV infection. Overall, this study provides new evidence for elucidating the molecular mechanism of silkworms in response to BmNPV infection and new strategies for the prevention of viral infections in sericulture.


Subject(s)
Bombyx , Insect Proteins , Nucleopolyhedroviruses , Animals , Bombyx/enzymology , Bombyx/genetics , Bombyx/virology , Insect Proteins/metabolism , Insect Proteins/genetics , Larva/metabolism , Larva/growth & development , Larva/virology , Metalloproteins/metabolism , Metalloproteins/genetics , Molybdenum Cofactors , Nucleopolyhedroviruses/physiology , RNA Interference , Uric Acid/metabolism
9.
Microbiol Spectr ; 12(2): e0348023, 2024 Feb 06.
Article in English | MEDLINE | ID: mdl-38193660

ABSTRACT

The expression of most molybdoenzymes in Escherichia coli has so far been revealed to be regulated by anaerobiosis and requires the presence of iron, based on the necessity of the transcription factor FNR to bind one [4Fe-4S] cluster. One exception is trimethylamine-N-oxide reductase encoded by the torCAD operon, which has been described to be expressed independently from FNR. In contrast to other alternative anaerobic respiratory systems, the expression of the torCAD operon was shown not to be completely repressed by the presence of dioxygen. To date, the basis for the O2-dependent expression of the torCAD operon has been related to the abundance of the transcriptional regulator IscR, which represses the transcription of torS and torT, and is more abundant under aerobic conditions than under anaerobic conditions. In this study, we reinvestigated the regulation of the torCAD operon and its dependence on the presence of iron and identified a novel regulation that depends on the presence of the bis-molybdopterin guanine dinucleotide (bis-MGD) molybdenum cofactor . We confirmed that the torCAD operon is directly regulated by the heme-containing protein TorC and is indirectly regulated by ArcA and by the availability of iron via active FNR and Fur, both regulatory proteins that influence the synthesis of the molybdenum cofactor. Furthermore, we identified a novel regulation mode of torCAD expression that is dependent on cellular levels of bis-MGD and is not used by other bis-MGD-containing enzymes like nitrate reductase.IMPORTANCEIn bacteria, molybdoenzymes are crucial for anaerobic respiration using alternative electron acceptors. FNR is a very important transcription factor that represents the master switch for the expression of target genes in response to anaerobiosis. Only Escherichia coli trimethylamine-N-oxide (TMAO) reductase escapes this regulation by FNR. We identified that the expression of TMAO reductase is regulated by the amount of bis-molybdopterin guanine dinucleotide (bis-MGD) cofactor synthesized by the cell itself, representing a novel regulation pathway for the expression of an operon coding for a molybdoenzyme. Furthermore, TMAO reductase gene expression is indirectly regulated by the presence of iron, which is required for the production of the bis-MGD cofactor in the cell.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Methylamines , Escherichia coli/genetics , Iron/metabolism , Operon , Escherichia coli Proteins/genetics , Transcription Factors/metabolism , Oxidoreductases/genetics , Molybdenum Cofactors , Oxides/metabolism , Anaerobiosis , Bacterial Proteins/genetics , Gene Expression Regulation, Bacterial
10.
Molecules ; 28(19)2023 Oct 09.
Article in English | MEDLINE | ID: mdl-37836841

ABSTRACT

Sulfite oxidase is one of five molybdenum-containing enzymes known in eukaryotes where it catalyzes the oxidation of sulfite to sulfate. This review covers the history of sulfite oxidase research starting out with the early years of its discovery as a hepatic mitochondrial enzyme in vertebrates, leading to basic biochemical and structural properties that have inspired research for decades. A personal view on sulfite oxidase in plants, that sulfates are assimilated for their de novo synthesis of cysteine, is presented by Ralf Mendel with numerous unexpected findings and unique properties of this single-cofactor sulfite oxidase localized to peroxisomes. Guenter Schwarz connects his research to sulfite oxidase via its deficiency in humans, demonstrating its unique role amongst all molybdenum enzymes in humans. In essence, in both the plant and animal kingdoms, sulfite oxidase represents an important player in redox regulation, signaling and metabolism, thereby connecting sulfur and nitrogen metabolism in multiple ways.


Subject(s)
Sulfite Oxidase , Animals , Humans , Sulfite Oxidase/metabolism , Molybdenum/chemistry , Sulfites , Plants/metabolism , Molybdenum Cofactors , Sulfates/metabolism
11.
Cell Mol Neurobiol ; 43(6): 2895-2907, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36862242

ABSTRACT

Isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies are genetic diseases biochemically characterized by the toxic accumulation of sulfite in the tissues of patients, including the brain. Neurological dysfunction and brain abnormalities are commonly observed soon after birth, and some patients also have neuropathological alterations in the prenatal period (in utero). Thus, we investigated the effects of sulfite on redox and mitochondrial homeostasis, as well as signaling proteins in the cerebral cortex of rat pups. One-day-old Wistar rats received an intracerebroventricular administration of sulfite (0.5 µmol/g) or vehicle and were euthanized 30 min after injection. Sulfite administration decreased glutathione levels and glutathione S-transferase activity, and increased heme oxygenase-1 content in vivo in the cerebral cortex. Sulfite also reduced the activities of succinate dehydrogenase, creatine kinase, and respiratory chain complexes II and II-III. Furthermore, sulfite increased the cortical content of ERK1/2 and p38. These findings suggest that redox imbalance and bioenergetic impairment induced by sulfite in the brain are pathomechanisms that may contribute to the neuropathology of newborns with ISOD and MoCD. Sulfite disturbs antioxidant defenses, bioenergetics, and signaling pathways in the cerebral cortex of neonatal rats. CII: complex II; CII-III: complex II-III; CK: creatine kinase; GST: glutathione S-transferase; HO-1: heme oxygenase-1; SDH: succinate dehydrogenase; SO32-: sulfite.


Subject(s)
Cerebral Cortex , Energy Metabolism , Molybdenum Cofactors , Sulfite Oxidase , Sulfites , Animals , Rats , Animals, Newborn , Oxidation-Reduction , Sulfites/adverse effects , Sulfite Oxidase/metabolism , Molybdenum Cofactors/metabolism , Rats, Wistar , Homeostasis , Mitochondria/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Antioxidants/metabolism
12.
Biomolecules ; 13(1)2023 01 10.
Article in English | MEDLINE | ID: mdl-36671528

ABSTRACT

Sulfur is an important element that is incorporated into many biomolecules in humans. The incorporation and transfer of sulfur into biomolecules is, however, facilitated by a series of different sulfurtransferases. Among these sulfurtransferases is the human mercaptopyruvate sulfurtransferase (MPST) also designated as tRNA thiouridine modification protein (TUM1). The role of the human TUM1 protein has been suggested in a wide range of physiological processes in the cell among which are but not limited to involvement in Molybdenum cofactor (Moco) biosynthesis, cytosolic tRNA thiolation and generation of H2S as signaling molecule both in mitochondria and the cytosol. Previous interaction studies showed that TUM1 interacts with the L-cysteine desulfurase NFS1 and the Molybdenum cofactor biosynthesis protein 3 (MOCS3). Here, we show the roles of TUM1 in human cells using CRISPR/Cas9 genetically modified Human Embryonic Kidney cells. Here, we show that TUM1 is involved in the sulfur transfer for Molybdenum cofactor synthesis and tRNA thiomodification by spectrophotometric measurement of the activity of sulfite oxidase and liquid chromatography quantification of the level of sulfur-modified tRNA. Further, we show that TUM1 has a role in hydrogen sulfide production and cellular bioenergetics.


Subject(s)
Molybdenum Cofactors , Sulfurtransferases , Humans , Cytosol/metabolism , Sulfurtransferases/metabolism , Energy Metabolism , Sulfur/metabolism , RNA, Transfer/metabolism , Kidney/metabolism , Carbon-Sulfur Lyases/metabolism
13.
J Biol Chem ; 299(1): 102736, 2023 01.
Article in English | MEDLINE | ID: mdl-36423681

ABSTRACT

Molybdenum cofactor (Moco) is a prosthetic group necessary for the activity of four unique enzymes, including the essential sulfite oxidase (SUOX-1). Moco is required for life; humans with inactivating mutations in the genes encoding Moco-biosynthetic enzymes display Moco deficiency, a rare and lethal inborn error of metabolism. Despite its importance to human health, little is known about how Moco moves among and between cells, tissues, and organisms. The prevailing view is that cells that require Moco must synthesize Moco de novo. Although, the nematode Caenorhabditis elegans appears to be an exception to this rule and has emerged as a valuable system for understanding fundamental Moco biology. C. elegans has the seemingly unique capacity to both synthesize its own Moco as well as acquire Moco from its microbial diet. However, the relative contribution of Moco from the diet or endogenous synthesis has not been rigorously evaluated or quantified biochemically. We genetically removed dietary or endogenous Moco sources in C. elegans and biochemically determined their impact on animal Moco content and SUOX-1 activity. We demonstrate that dietary Moco deficiency dramatically reduces both animal Moco content and SUOX-1 activity. Furthermore, these biochemical deficiencies have physiological consequences; we show that dietary Moco deficiency alone causes sensitivity to sulfite, the toxic substrate of SUOX-1. Altogether, this work establishes the biochemical consequences of depleting dietary Moco or endogenous Moco synthesis in C. elegans and quantifies the surprising contribution of the diet to maintaining Moco homeostasis in C. elegans.


Subject(s)
Metalloproteins , Molybdenum Cofactors , Sulfite Oxidase , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Diet , Metalloproteins/genetics , Metalloproteins/metabolism , Molybdenum/metabolism , Molybdenum Cofactors/metabolism , Pteridines/metabolism , Sulfite Oxidase/genetics , Sulfite Oxidase/metabolism
14.
Molecules ; 27(20)2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36296488

ABSTRACT

Molybdenum cofactor (Moco) deficiency (MoCD) is characterized by neonatal-onset myoclonic epileptic encephalopathy and dystonia with cerebral MRI changes similar to hypoxic-ischemic lesions. The molecular cause of the disease is the loss of sulfite oxidase (SOX) activity, one of four Moco-dependent enzymes in men. Accumulating toxic sulfite causes a secondary increase of metabolites such as S-sulfocysteine and thiosulfate as well as a decrease in cysteine and its oxidized form, cystine. Moco is synthesized by a three-step biosynthetic pathway that involves the gene products of MOCS1, MOCS2, MOCS3, and GPHN. Depending on which synthetic step is impaired, MoCD is classified as type A, B, or C. This distinction is relevant for patient management because the metabolic block in MoCD type A can be circumvented by administering cyclic pyranopterin monophosphate (cPMP). Substitution therapy with cPMP is highly effective in reducing sulfite toxicity and restoring biochemical homeostasis, while the clinical outcome critically depends on the degree of brain injury prior to the start of treatment. In the absence of a specific treatment for MoCD type B/C and SOX deficiency, we summarize recent progress in our understanding of the underlying metabolic changes in cysteine homeostasis and propose novel therapeutic interventions to circumvent those pathological changes.


Subject(s)
Brain Diseases , Metalloproteins , Sulfite Oxidase , Male , Infant, Newborn , Humans , Cysteine , Thiosulfates , Cystine , Coenzymes/metabolism , Metalloproteins/metabolism , Sulfite Oxidase/genetics , Sulfites , Molybdenum Cofactors , Molybdenum
15.
Molecules ; 27(19)2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36235107

ABSTRACT

The molybdenum cofactor (Moco) is the active site prosthetic group found in numerous vitally important enzymes (Mo-enzymes), which predominantly catalyze 2 electron transfer reactions. Moco is synthesized by an evolutionary old and highly conserved multi-step pathway, whereby the metal insertion reaction is the ultimate reaction step here. Moco and its intermediates are highly sensitive towards oxidative damage and considering this, they are believed to be permanently protein bound during synthesis and also after Moco maturation. In plants, a cellular Moco transfer and storage system was identified, which comprises proteins that are capable of Moco binding and release but do not possess a Moco-dependent enzymatic activity. The first protein described that exhibited these properties was the Moco carrier protein (MCP) from the green alga Chlamydomonas reinhardtii. However, MCPs and similar proteins have meanwhile been described in various plant species. This review will summarize the current knowledge of the cellular Moco distribution system.


Subject(s)
Chlamydomonas reinhardtii , Metalloproteins , Carrier Proteins/metabolism , Catalytic Domain , Chlamydomonas reinhardtii/metabolism , Coenzymes/chemistry , Metalloproteins/chemistry , Molybdenum/metabolism , Molybdenum Cofactors , Plants/metabolism
16.
Molecules ; 27(17)2022 Aug 23.
Article in English | MEDLINE | ID: mdl-36080140

ABSTRACT

For most organisms molybdenum is essential for life as it is found in the active site of various vitally important molybdenum dependent enzymes (Mo-enzymes). Here, molybdenum is bound to a pterin derivative called molybdopterin (MPT), thus forming the molybdenum cofactor (Moco). Synthesis of Moco involves the consecutive action of numerous enzymatic reaction steps, whereby molybdenum insertases (Mo-insertases) catalyze the final maturation step, i.e., the metal insertion reaction yielding Moco. This final maturation step is subdivided into two partial reactions, each catalyzed by a distinctive Mo-insertase domain. Initially, MPT is adenylylated by the Mo-insertase G-domain, yielding MPT-AMP which is used as substrate by the E-domain. This domain catalyzes the insertion of molybdate into the MPT dithiolene moiety, leading to the formation of Moco-AMP. Finally, the Moco-AMP phosphoanhydride bond is cleaved by the E-domain to liberate Moco from its synthesizing enzyme. Thus formed, Moco is physiologically active and may be incorporated into the different Mo-enzymes or bind to carrier proteins instead.


Subject(s)
Metalloproteins , Molybdenum , Adenosine Monophosphate , Catalytic Domain , Coenzymes/chemistry , Metalloproteins/chemistry , Molybdenum/metabolism , Molybdenum Cofactors , Pterins
17.
Molecules ; 27(15)2022 Aug 03.
Article in English | MEDLINE | ID: mdl-35956883

ABSTRACT

The transition element molybdenum (Mo) is an essential micronutrient for plants, animals, and microorganisms, where it forms part of the active center of Mo enzymes. To gain biological activity in the cell, Mo has to be complexed by a pterin scaffold to form the molybdenum cofactor (Moco). Mo enzymes and Moco are found in all kingdoms of life, where they perform vital transformations in the metabolism of nitrogen, sulfur, and carbon compounds. In this review, I recall the history of Moco in a personal view, starting with the genetics of Moco in the 1960s and 1970s, followed by Moco biochemistry and the description of its chemical structure in the 1980s. When I review the elucidation of Moco biosynthesis in the 1990s and the early 2000s, I do it mainly for eukaryotes, as I worked with plants, human cells, and filamentous fungi. Finally, I briefly touch upon human Moco deficiency and whether there is life without Moco.


Subject(s)
Metalloproteins , Molybdenum Cofactors , Animals , Coenzymes/chemistry , Eukaryota/metabolism , Humans , Metalloproteins/metabolism , Molybdenum/metabolism , Molybdenum Cofactors/genetics , Molybdenum Cofactors/metabolism , Plants/metabolism , Pterins
18.
Molecules ; 27(12)2022 Jun 10.
Article in English | MEDLINE | ID: mdl-35744859

ABSTRACT

Molybdenum cofactor (Moco) biosynthesis requires iron, copper, and ATP. The Moco-containing enzyme sulfite oxidase catalyzes terminal oxidation in oxidative cysteine catabolism, and another Moco-containing enzyme, xanthine dehydrogenase, functions in purine catabolism. Thus, molybdenum enzymes participate in metabolic pathways that are essential for cellular detoxication and energy dynamics. Studies of the Moco biosynthetic enzymes MoaE (in the Ada2a-containing (ATAC) histone acetyltransferase complex) and MOCS2 have revealed that Moco biosynthesis and molybdenum enzymes align to regulate signaling and metabolism via control of transcription and translation. Disruption of these functions is involved in the onset of dementia and neurodegenerative disease. This review provides an overview of the roles of MoaE and MOCS2 in normal cellular processes and neurodegenerative disease, as well as directions for future research.


Subject(s)
Metalloproteins , Neurodegenerative Diseases , Sulfite Oxidase , Coenzymes/metabolism , Humans , Molybdenum/metabolism , Molybdenum Cofactors , Sulfite Oxidase/metabolism , Sulfurtransferases , Xanthine Dehydrogenase/metabolism
19.
Chemistry ; 28(50): e202201660, 2022 Sep 06.
Article in English | MEDLINE | ID: mdl-35670547

ABSTRACT

In this study we propose to coin the term Wolfium bond (WfB) to refer to a net attractive force (noncovalent interaction) between any element of group 6 and electron donor atoms (neutral molecules or anions) and to differentiate it from a coordination bond (metal-ligand interaction). We provide evidence of the existence of this interaction by inspecting the X-ray crystal structure of proteins containing Molybdopterin and Tungstopterin cofactors from the Protein Data Bank (PDB). The plausible biological role of the interaction as well as its physical nature (antibonding Wf-Ligand orbital involved) are also analyzed by means of ab initio calculations (RI-MP2/def2-TZVP level of theory), Atoms in Molecules (AIM), Natural Bond Orbital (NBO) and Noncovalent Interactions plot (NCIplot) analyses.


Subject(s)
Metalloproteins , Quantum Theory , Ligands , Molybdenum Cofactors , Thermodynamics
20.
Clin Pharmacol Ther ; 112(4): 808-816, 2022 10.
Article in English | MEDLINE | ID: mdl-35538648

ABSTRACT

Therapy of molybdenum cofactor (Moco) deficiency has received US Food and Drug Administration (FDA) approval in 2021. Whereas urothione, the urinary excreted catabolite of Moco, is used as diagnostic biomarker for Moco-deficiency, its catabolic pathway remains unknown. Here, we identified the urothione-synthesizing methyltransferase using mouse liver tissue by anion exchange/size exclusion chromatography and peptide mass fingerprinting. We show that the catabolic Moco S-methylating enzyme corresponds to thiopurine S-methyltransferase (TPMT), a highly polymorphic drug-metabolizing enzyme associated with drug-related hematotoxicity but unknown physiological role. Urothione synthesis was investigated in vitro using recombinantly expressed human TPMT protein, liver lysates from Tpmt wild-type and knock-out (Tpmt-/- ) mice as well as human liver cytosol. Urothione levels were quantified by liquid-chromatography tandem mass spectrometry in the kidneys and urine of mice. TPMT-genotype/phenotype and excretion levels of urothione were investigated in human samples and validated in an independent population-based study. As Moco provides a physiological substrate (thiopterin) of TPMT, thiopterin-methylating activity was associated with TPMT activity determined with its drug substrate (6-thioguanin) in mice and humans. Urothione concentration was extremely low in the kidneys and urine of Tpmt-/- mice. Urinary urothione concentration in TPMT-deficient patients depends on common TPMT polymorphisms, with extremely low levels in homozygous variant carriers (TPMT*3A/*3A) but normal levels in compound heterozygous carriers (TPMT*3A/*3C) as validated in the population-based study. Our work newly identified an endogenous substrate for TPMT and shows an unprecedented link between Moco catabolism and drug metabolism. Moreover, the TPMT example indicates that phenotypic consequences of genetic polymorphisms may differ between drug- and endogenous substrates.


Subject(s)
Methyltransferases , Molybdenum Cofactors , Animals , Genotype , Humans , Methyltransferases/physiology , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL