Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(14): e2315568121, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38530900

RESUMO

Methanogenic archaea inhabiting anaerobic environments play a crucial role in the global biogeochemical material cycle. The most universal electrogenic reaction of their methane-producing energy metabolism is catalyzed by N    5-methyl-tetrahydromethanopterin: coenzyme M methyltransferase (MtrABCDEFGH), which couples the vectorial Na+ transport with a methyl transfer between the one-carbon carriers tetrahydromethanopterin and coenzyme M via a vitamin B12 derivative (cobamide) as prosthetic group. We present the 2.08 Šcryo-EM structure of Mtr(ABCDEFG)3 composed of the central Mtr(ABFG)3 stalk symmetrically flanked by three membrane-spanning MtrCDE globes. Tetraether glycolipids visible in the map fill gaps inside the multisubunit complex. Putative coenzyme M and Na+ were identified inside or in a side-pocket of a cytoplasmic cavity formed within MtrCDE. Its bottom marks the gate of the transmembrane pore occluded in the cryo-EM map. By integrating Alphafold2 information, functionally competent MtrA-MtrH and MtrA-MtrCDE subcomplexes could be modeled and thus the methyl-tetrahydromethanopterin demethylation and coenzyme M methylation half-reactions structurally described. Methyl-transfer-driven Na+ transport is proposed to be based on a strong and weak complex between MtrCDE and MtrA carrying vitamin B12, the latter being placed at the entrance of the cytoplasmic MtrCDE cavity. Hypothetically, strongly attached methyl-cob(III)amide (His-on) carrying MtrA induces an inward-facing conformation, Na+ flux into the membrane protein center and finally coenzyme M methylation while the generated loosely attached (or detached) MtrA carrying cob(I)amide (His-off) induces an outward-facing conformation and an extracellular Na+ outflux. Methyl-cob(III)amide (His-on) is regenerated in the distant active site of the methyl-tetrahydromethanopterin binding MtrH implicating a large-scale shuttling movement of the vitamin B12-carrying domain.


Assuntos
Mesna , Metiltransferases , Mesna/metabolismo , Metiltransferases/metabolismo , Metilação , Vitamina B 12/metabolismo , Metano/metabolismo , Amidas , Vitaminas
2.
Chembiochem ; 25(1): e202300595, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-37815851

RESUMO

Methanogenic and methanotrophic archaea play important roles in the global carbon cycle by interconverting CO2 and methane. To conserve energy from these metabolic pathways that happen close to the thermodynamic equilibrium, specific electron carriers have evolved to balance the redox potentials between key steps. Reduced ferredoxins required to activate CO2 are provided by energetical coupling to the reduction of the high-potential heterodisulfide (HDS) of coenzyme M (2-mercaptoethanesulfonate) and coenzyme B (7-mercaptoheptanoylthreonine phosphate). While the standard redox potential of this important HDS has been determined previously to be -143 mV (Tietze et al. 2003 DOI: 10.1002/cbic.200390053), we have measured thiol disulfide exchange kinetics and reassessed this value by equilibrating thiol-disulfide mixtures of coenzyme M, coenzyme B, and mercaptoethanol. We determined the redox potential of the HDS of coenzyme M and coenzyme B to be -16.4±1.7 mV relative to the reference thiol mercaptoethanol (E0 '=-264 mV). The resulting E0 ' values are -281 mV for the HDS, -271 mV for the homodisulfide of coenzyme M, and -270 mV for the homodisulfide of coenzyme B. We discuss the importance of these updated values for the physiology of methanogenic and methanotrophic archaea and their implications in terms of energy conservation.


Assuntos
Archaea , Mesna , Mesna/metabolismo , Archaea/metabolismo , Compostos de Sulfidrila , Mercaptoetanol , Dissulfetos/metabolismo , Dióxido de Carbono/metabolismo , Elétrons , Transporte de Elétrons , Metano/metabolismo , Oxirredução
3.
Environ Toxicol Pharmacol ; 100: 104137, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37127110

RESUMO

Human lymphoblast cells were treated with the marine algal toxin, brevetoxin-2 (PbTx-2), and its effects on the proteome were assessed by redox proteomics using cysteine reactive tandem mass tags (TMT). Additionally, cells were simultaneously treated with PbTx-2 and the antioxidant and acrolein scavenger sodium 2-mercaptoethylsulfonate (MESNA) to determine if MESNA could prevent the proteomic effects of brevetoxin-2. A massive shift in the redox state of the proteome of brevetoxin-2 treated cells was observed. The main pathway affected was genetic information processing. Significantly oxidized proteins included Trx-1, peroxyredoxins (Prxs), ribosomal proteins, and the eukaryotic initiation factor 2 ß subunit (eIF2ß). Proteins that were overexpressed in brevetoxin-treated cells included four folding chaperones. These effects were diminished in the presence of MESNA indicating that MESNA may act through its antioxidant properties or as a brevetoxin scavenger. These studies provide novel insights into new prophylactics for brevetoxicosis in humans and wildlife.


Assuntos
Dinoflagelados , Proteoma , Animais , Humanos , Proteoma/metabolismo , Acroleína , Mesna/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Proteômica , Peixes/metabolismo , Oxirredução , Resposta a Proteínas não Dobradas , Dinoflagelados/metabolismo
4.
Commun Biol ; 5(1): 1113, 2022 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-36266535

RESUMO

Methanogens and anaerobic methane-oxidizing archaea (ANME) are important players in the global carbon cycle. Methyl-coenzyme M reductase (MCR) is a key enzyme in methane metabolism, catalyzing the last step in methanogenesis and the first step in anaerobic methane oxidation. Divergent mcr and mcr-like genes have recently been identified in uncultured archaeal lineages. However, the assembly and biochemistry of MCRs from uncultured archaea remain largely unknown. Here we present an approach to study MCRs from uncultured archaea by heterologous expression in a methanogen, Methanococcus maripaludis. Promoter, operon structure, and temperature were important determinants for MCR production. Both recombinant methanococcal and ANME-2 MCR assembled with the host MCR forming hybrid complexes, whereas tested ANME-1 MCR and ethyl-coenzyme M reductase only formed homogenous complexes. Together with structural modeling, this suggests that ANME-2 and methanogen MCRs are structurally similar and their reaction directions are likely regulated by thermodynamics rather than intrinsic structural differences.


Assuntos
Archaea , Mesna , Archaea/genética , Archaea/metabolismo , Mesna/metabolismo , Oxirredutases/metabolismo , Metano/metabolismo
5.
Proc Natl Acad Sci U S A ; 119(36): e2207190119, 2022 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-36037354

RESUMO

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.


Assuntos
Bactérias , Coenzimas , Euryarchaeota , Mesna , Anaerobiose , Archaea/metabolismo , Bactérias/metabolismo , Coenzimas/biossíntese , Euryarchaeota/metabolismo , Mesna/metabolismo , Metano/metabolismo , Oxirredução , Fosfatos/metabolismo
6.
Biochemistry ; 61(10): 805-821, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35500274

RESUMO

Microbial anaerobic oxidation of alkanes intrigues the scientific community by way of its impact on the global carbon cycle, and its biotechnological applications. Archaea are proposed to degrade short- and long-chain alkanes to CO2 by reversing methanogenesis, a theoretically reversible process. The pathway would start with alkane activation, an endergonic step catalyzed by methyl-coenzyme M reductase (MCR) homologues that would generate alkyl-thiols carried by coenzyme M. While the methane-generating MCR found in methanogens has been well characterized, the enzymatic activity of the putative alkane-fixing counterparts has not been validated so far. Such an absence of biochemical investigations contrasts with the current explosion of metagenomics data, which draws new potential alkane-oxidizing pathways in various archaeal phyla. Therefore, validating the physiological function of these putative alkane-fixing machines and investigating how their structures, catalytic mechanisms, and cofactors vary depending on the targeted alkane have become urgent needs. The first structural insights into the methane- and ethane-capturing MCRs highlighted unsuspected differences and proposed some explanations for their substrate specificity. This Perspective reviews the current physiological, biochemical, and structural knowledge of alkyl-CoM reductases and offers fresh ideas about the expected mechanistic and chemical differences among members of this broad family. We conclude with the challenges of the investigation of these particular enzymes, which might one day generate biofuels for our modern society.


Assuntos
Alcanos , Archaea , Alcanos/metabolismo , Anaerobiose , Archaea/química , Catálise , Mesna/metabolismo , Metano/metabolismo , Oxirredução , Oxirredutases/metabolismo , Filogenia
7.
J Biol Chem ; 298(5): 101884, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35367206

RESUMO

2-Ketopropyl-coenzyme M oxidoreductase/carboxylase (2-KPCC) is a member of the flavin and cysteine disulfide containing oxidoreductase family (DSOR) that catalyzes the unique reaction between atmospheric CO2 and a ketone/enolate nucleophile to generate acetoacetate. However, the mechanism of this reaction is not well understood. Here, we present evidence that 2-KPCC, in contrast to the well-characterized DSOR enzyme glutathione reductase, undergoes conformational changes during catalysis. Using a suite of biophysical techniques including limited proteolysis, differential scanning fluorimetry, and native mass spectrometry in the presence of substrates and inhibitors, we observed conformational differences between different ligand-bound 2-KPCC species within the catalytic cycle. Analysis of site-specific amino acid variants indicated that 2-KPCC-defining residues, Phe501-His506, within the active site are important for transducing these ligand induced conformational changes. We propose that these conformational changes promote substrate discrimination between H+ and CO2 to favor the metabolically preferred carboxylation product, acetoacetate.


Assuntos
Carboxiliases , Mesna , Acetoacetatos/metabolismo , Dióxido de Carbono/metabolismo , Carboxiliases/metabolismo , Catálise , Ligantes , Mesna/metabolismo , Oxirredutases/metabolismo , Xanthobacter/metabolismo
8.
Dig Dis Sci ; 65(12): 3583-3591, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32088797

RESUMO

BACKGROUND: Acute pancreatitis (AP) is a sudden inflammation of the pancreas that may be life-threatening disease with high mortality rates, particularly in the presence of systemic inflammatory response and multiple organ failure. Oxidative stress has been shown to be involved in the pathophysiology of acute pancreatitis. AIM: This study is designed to investigate the possible effect of mesna on an experimental model of cerulein-induced acute pancreatitis. METHODS: Animals were divided into five groups: Group 1 served as a control group given the saline; group II (mesna group) received mesna at a dose of (100 mg/kg per dose, i.p.) four times; group III (acute pancreatitis group) received cerulein at a dose of (20 µg/kg/dose, s.c.) four times with 1-h intervals; group VI, cerulein + mesna, was treated with mesna at a dose of (100 mg/kg, i.p.) 15 min before each cerulein injection. RESULTS: Animals with acute pancreatitis showed elevated serum amylase and lipase levels. Biochemical parameters showed increased pancreatic tumor necrosis factors-α (TNF-α) and interleukin-1ß (IL-1ß) levels. A disturbance in oxidative stress markers was evident by elevated pancreatic lipid peroxides (TBARS) and decline in pancreatic antioxidants' concentrations including reduced glutathione (GSH); superoxide dismutase (SOD); and glutathione peroxidase (GSH-Px). Histological examination confirmed pancreatic injury. Pre-treatment with mesna was able to abolish the changes in pancreatic enzymes, oxidative stress markers (TBARS, SOD, GSH and GSH-Px), pancreatic inflammatory markers (TNF-α, IL-1ß) as well as histological changes. CONCLUSIONS: Mesna mitigates AP by alleviating pancreatic oxidative stress damage and inhibiting inflammation.


Assuntos
Ceruletídeo/farmacologia , Mesna , Estresse Oxidativo/efeitos dos fármacos , Pâncreas , Pancreatite , Animais , Antioxidantes/análise , Colagogos e Coleréticos/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Interleucina-1beta/sangue , Mesna/metabolismo , Mesna/farmacologia , Pâncreas/efeitos dos fármacos , Pâncreas/enzimologia , Pâncreas/patologia , Pancreatite/induzido quimicamente , Pancreatite/metabolismo , Pancreatite/prevenção & controle , Substâncias Protetoras/metabolismo , Substâncias Protetoras/farmacologia , Ratos , Resultado do Tratamento , Fator de Necrose Tumoral alfa/sangue
9.
Environ Int ; 131: 105006, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31330362

RESUMO

Electron allocation through the suppression or the stimulation of methanogenesis is critical for microbial electrolysis cells (MECs) to produce the desired target product (e.g., CH4 or H2). In this study, selective methanogenesis control using the coenzyme M (CoM) and 2-bromoethanesulfonate (2-BES) was investigated in a two-chambered MEC to evaluate the effect of CoM and 2-BES on the production of different electrobiofuels, net energy conversion efficiency and microbial community structure. Because the CoM is a crucial methyl-group carrier in the final process of methanogenesis, it was postulated that CoM would stimulate methanogenic activity at the anode, while a structural analog of the CoM (i.e., 2-BES) was expected to improve cathodic H2 yield using electrons conserved because of methanogen inhibition (electron equivalence: 8 mol e- = 1 mol CH4 = 4 mol H2). CoM injection in MECs significantly enhanced their CH4 production rate, purity, and yield by 4.5-fold, 14.5%, and 76.1%, respectively, compared to the control. Moreover, microbial community analysis indicated that Methanosaeta, the major acetoclastic methanogen, continued to dominate the microbial community but steadily decreased in relative abundance after the CoM injection. On the other hand, drastic increases in hydrogenotrophic methanogens, such as Methanoculleus and Methanolinea, were observed along with potential syntrophic acetate-oxidizing bacteria. In contrast, CH4 production in the 2-BES injected trials was significantly inhibited by 79.5%, resulting in a corresponding increase of H2 production by 145.5% compared to the control. Unlike the CoM, the microbial community did not noticeably change when 2-BES was injected, although the population size gradually decreased over time. Also, a single injection of CoM and 2-BES, even at low concentrations (500 µM), enabled the desired allocation of electrons as characterized by a high sensitivity, fast response, and negligible interference. In terms of energy conversion efficiency, methanogenesis stimulation approach resulted in higher net energy production than inhibition approach, whereas the remained electrons were not fully converted to hydrogen in case of the inhibition trial, thus producing less energy.


Assuntos
Ácidos Alcanossulfônicos/metabolismo , Biocombustíveis , Mesna/metabolismo , Metano/biossíntese , Acetatos/metabolismo , Bactérias/metabolismo , Eletrólise
10.
Biochemistry ; 58(15): 1958-1962, 2019 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-30932481

RESUMO

Coenzyme M is an essential coenzyme for the biochemical production of methane. This Communication reports on the identification of an enzyme catalyzing the last step in the biosynthesis of coenzyme M in methanogens. Data presented here show that the enzyme, derived from mj1681, catalyzes the conversion of the aldehyde functional group of sulfoacetaldehyde into the thiol group of 2-mercaptoethanesulfonic acid. Thus, a putative coenzyme M synthase (comF) has similarities in sequence with both MJ0100 and MJ0099 proteins previously shown to be involved in the biosynthesis of homocysteine [Allen, K. D., et al. (2015) Biochemistry 54, 3129-3132], and both reactions likely proceed by the same mechanism. In the MJ0100-catalyzed reaction, Rauch has proposed [Rauch, B. L. (2017) Biochemistry 56, 1051-1061] that MJ1526 and its homologues in other methanogens likely supply the sulfane sulfur required for the reaction.


Assuntos
Acetaldeído/análogos & derivados , Mesna/metabolismo , Metano/metabolismo , Mathanococcus/metabolismo , Acetaldeído/metabolismo , Biocatálise , Carbono-Oxigênio Liases/metabolismo , Homocisteína/metabolismo , Compostos de Sulfidrila/metabolismo , Enxofre/metabolismo
11.
J Bacteriol ; 201(12)2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30936368

RESUMO

Methanogenesis from methylated substrates is initiated by substrate-specific methyltransferases that generate the central metabolic intermediate methyl-coenzyme M. This reaction involves a methyl-corrinoid protein intermediate and one or two cognate methyltransferases. Based on genetic data, the Methanosarcina acetivorans MtpC (corrinoid protein) and MtpA (methyltransferase) proteins were suggested to catalyze the methylmercaptopropionate (MMPA):coenzyme M (CoM) methyl transfer reaction without a second methyltransferase. To test this, MtpA was purified after overexpression in its native host and characterized biochemically. MtpA catalyzes a robust methyl transfer reaction using free methylcob(III)alamin as the donor and mercaptopropionate (MPA) as the acceptor, with kcat of 0.315 s-1 and apparent Km for MPA of 12 µM. CoM did not serve as a methyl acceptor; thus, a second unidentified methyltransferase is required to catalyze the full MMPA:CoM methyl transfer reaction. The physiologically relevant methylation of cob(I)alamin with MMPA, which is thermodynamically unfavorable, was also demonstrated, but only at high substrate concentrations. Methylation of cob(I)alamin with methanol, dimethylsulfide, dimethylamine, and methyl-CoM was not observed, even at high substrate concentrations. Although the corrinoid protein MtpC was poorly expressed alone, a stable MtpA/MtpC complex was obtained when both proteins were coexpressed. Biochemical characterization of this complex was not feasible, because the corrinoid cofactor of this complex was in the inactive Co(II) state and was not reactivated by incubation with strong reductants. The MtsF protein, composed of both corrinoid and methyltransferase domains, copurifies with the MtpA/MtpC, suggesting that it may be involved in MMPA metabolism.IMPORTANCE Methylmercaptopropionate (MMPA) is an environmentally significant molecule produced by degradation of the abundant marine metabolite dimethylsulfoniopropionate, which plays a significant role in the biogeochemical cycles of both carbon and sulfur, with ramifications for ecosystem productivity and climate homeostasis. Detailed knowledge of the mechanisms for MMPA production and consumption is key to understanding steady-state levels of this compound in the biosphere. Unfortunately, the biochemistry required for MMPA catabolism under anoxic conditions is poorly characterized. The data reported here validate the suggestion that the MtpA protein catalyzes the first step in the methanogenic catabolism of MMPA. However, the enzyme does not catalyze a proposed second step required to produce the key intermediate, methyl coenzyme M. Therefore, the additional enzymes required for methanogenic MMPA catabolism await discovery.


Assuntos
Mercaptopurina/análogos & derivados , Methanosarcina/enzimologia , Metiltransferases/metabolismo , Catálise , Mercaptopurina/metabolismo , Mesna/análogos & derivados , Mesna/metabolismo , Methanosarcina/genética , Metilação , Metiltransferases/genética , Vitamina B 12/metabolismo
12.
J Hazard Mater ; 374: 258-266, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31005708

RESUMO

Methanogenesis is commonly mass-produced under anaerobic conditions and serves as a major terminal electron accepting process driving the degradation of organic biomass. In this study, a cofactor of methanogenesis (coenzyme M, CoM) and a classic methanogensis inhibitor (2-bromoethanesulfonate, BES) were added at different concentrations to investigate how methanogenesis would affect PCP degradation in flooded soil. Strikingly, the processes of methanogenesis and PCP degradation were simultaneously promoted with CoM, or inhibited with BES, significantly (p < 0.05). High-throughput sequencing for soil bacterial and archaeal community structures revealed that members of Desulfitobacterium, Dethiobacter, Sedimentibacter, Bacillus and Methanosarcina might act as the core functional groups jointly perform PCP degradation in flooded soil, possibly through assisting microbial mediated dechlorination in direct organohalide-respiration, and/or indirect co-metabolization in complex anaerobic soil conditions. This study implied an underlying synergistic coupling between methanogenesis and dechlorination, and provided insights into a novel consideration with respect to coordinating methanogenesis while promoting anaerobic degradation of PCP for complex polluted soil environment, which is necessary for the improved all-win remediation.


Assuntos
Anaerobiose , Biodegradação Ambiental , Cloro/análise , Metano/química , Pentaclorofenol/análise , Poluentes do Solo/análise , Solo/química , Ácidos Alcanossulfônicos/metabolismo , Archaea/metabolismo , Bacillus , Clostridiales , Desulfitobacterium , Firmicutes , Inundações , Concentração de Íons de Hidrogênio , Mesna/metabolismo , Methanosarcina , Microbiologia do Solo , Fatores de Tempo
13.
J Biol Chem ; 293(14): 5236-5246, 2018 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-29414784

RESUMO

For nearly 30 years, coenzyme M (CoM) was assumed to be present solely in methanogenic archaea. In the late 1990s, CoM was reported to play a role in bacterial propene metabolism, but no biosynthetic pathway for CoM has yet been identified in bacteria. Here, using bioinformatics and proteomic approaches in the metabolically versatile bacterium Xanthobacter autotrophicus Py2, we identified four putative CoM biosynthetic enzymes encoded by the xcbB1, C1, D1, and E1 genes. Only XcbB1 was homologous to a known CoM biosynthetic enzyme (ComA), indicating that CoM biosynthesis in bacteria involves enzymes different from those in archaea. We verified that the ComA homolog produces phosphosulfolactate from phosphoenolpyruvate (PEP), demonstrating that bacterial CoM biosynthesis is initiated similarly as the phosphoenolpyruvate-dependent methanogenic archaeal pathway. The bioinformatics analysis revealed that XcbC1 and D1 are members of the aspartase/fumarase superfamily (AFS) and that XcbE1 is a pyridoxal 5'-phosphate-containing enzyme with homology to d-cysteine desulfhydrases. Known AFS members catalyze ß-elimination reactions of succinyl-containing substrates, yielding fumarate as the common unsaturated elimination product. Unexpectedly, we found that XcbC1 catalyzes ß-elimination on phosphosulfolactate, yielding inorganic phosphate and a novel metabolite, sulfoacrylic acid. Phosphate-releasing ß-elimination reactions are unprecedented among the AFS, indicating that XcbC1 is an unusual phosphatase. Direct demonstration of phosphosulfolactate synthase activity for XcbB1 and phosphate ß-elimination activity for XcbC1 strengthened their hypothetical assignment to a CoM biosynthetic pathway and suggested functions also for XcbD1 and E1. Our results represent a critical first step toward elucidating the CoM pathway in bacteria.


Assuntos
Mesna/metabolismo , Fosfatos/metabolismo , Xanthobacter/metabolismo , Aspartato Amônia-Liase/metabolismo , Bactérias/metabolismo , Biologia Computacional/métodos , Cristalografia por Raios X , Fumarato Hidratase/metabolismo , Fumaratos , Fosfoenolpiruvato/metabolismo , Ácidos Fosfóricos , Monoéster Fosfórico Hidrolases , Proteômica , Fosfato de Piridoxal
14.
Archaea ; 2017: 5793620, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29234237

RESUMO

(R)-Sulfolactate dehydrogenase (EC 1.1.1.337), termed ComC, is a member of an NADH/NADPH-dependent oxidoreductase family of enzymes that catalyze the interconversion of 2-hydroxyacids into their corresponding 2-oxoacids. The ComC reaction is reversible and in the biosynthetic direction causes the conversion of (R)-sulfolactate to sulfopyruvate in the production of coenzyme M (2-mercaptoethanesulfonic acid). Coenzyme M is an essential cofactor required for the production of methane by the methyl-coenzyme M reductase complex. ComC catalyzes the third step in the first established biosynthetic pathway of coenzyme M and is also involved in methanopterin biosynthesis. In this study, ComC from Methanobrevibacter millerae SM9 was cloned and expressed in Escherichia coli and biochemically characterized. Sulfopyruvate was the preferred substrate using the reduction reaction, with 31% activity seen for oxaloacetate and 0.2% seen for α-ketoglutarate. Optimal activity was observed at pH 6.5. The apparent KM for coenzyme (NADH) was 55.1 µM, and for sulfopyruvate, it was 196 µM (for sulfopyruvate the Vmax was 93.9 µmol min-1 mg-1 and kcat was 62.8 s-1). The critical role of ComC in two separate cofactor pathways makes this enzyme a potential means of developing methanogen-specific inhibitors for controlling ruminant methane emissions which are increasingly being recognized as contributing to climate change.


Assuntos
Lactatos/metabolismo , Methanobrevibacter/enzimologia , Oxirredutases/biossíntese , Oxirredutases/isolamento & purificação , Piruvatos/metabolismo , Vias Biossintéticas , Clonagem Molecular , Estabilidade Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Concentração de Íons de Hidrogênio , Cinética , Mesna/metabolismo , Oxirredutases/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
15.
Biochemistry ; 56(46): 6137-6144, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29064676

RESUMO

Here I report on the identification of 1-mercaptoethanesulfonic acid (1-MES), an analogue of 2-mercaptoethanesulfonic acid (coenzyme M, HSCoM). 1-MES and HSCoM were both present in the growth media of eight different methanogens at concentrations ranging from ∼1 to 100 µM. In an effort to determine a chemical origin of 1-MES, several plausible chemical routes were examined each assuming that HSCoM was the precursor. In all examined routes, no 1-MES was formed. However, 1-MES was formed when a solution of vinylsulfonic acid and sulfide were exposed to ultraviolet light. On the basis of these results, I conclude 1-MES is formed enzymatically. This was confirmed by growing a culture of Methanococcus maripaludis S2 in the presence of [1,1',2,2'-2H4]HSCoM and measuring the incorporation of deuterium into 1-MES. 1-MES incorporated three of the four deuteriums from the fed HSCoM. This result is consistent with the abstraction of a C-2 deuterium of the HSCoM, likely by a 5'-dAdoCH2• radical, followed by a radical rearrangement in which the sulfonic acid moves to position C-1, followed by abstraction of a H• likely from 5'-dAdoCH2D. At present, the reason for the production of 1-MES is not clear. This is the first report of the occurrence of 1-MES in Nature.


Assuntos
Vias Biossintéticas , Mesna/análogos & derivados , Mesna/metabolismo , Mathanococcus/metabolismo , Etilenos/metabolismo , Mathanococcus/enzimologia , Mathanococcus/crescimento & desenvolvimento , Sulfetos/metabolismo , Ácidos Sulfônicos/metabolismo
16.
Biotechnol Prog ; 33(5): 1243-1249, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28556629

RESUMO

Methanol:coenzyme M methyltransferase is an enzyme complex composed of three subunits, MtaA, MtaB, and MtaC, found in methanogenic archaea and is needed for their growth on methanol ultimately producing methane. MtaABC catalyzes the energetically favorable methyl transfer from methanol to coenzyme M to form methyl coenzyme M. Here we demonstrate that this important reaction for possible production of methanol from the anaerobic oxidation of methane can be reversed in vitro. To this effect, we have expressed and purified the Methanosarcina barkeri MtaABC enzyme, and developed an in vitro functional assay that demonstrates MtaABC can catalyze the energetically unfavorable (ΔG° = 27 kJ/mol) reverse reaction starting from methyl coenzyme M and generating methanol as a product. Demonstration of an in vitro ability of MtaABC to produce methanol may ultimately enable the anaerobic oxidation of methane to produce methanol and from methanol alternative fuel or fuel-precursor molecules. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 33:1243-1249, 2017.


Assuntos
Mesna/análogos & derivados , Metanol/metabolismo , Methanosarcina barkeri/enzimologia , Methanosarcina barkeri/genética , Reatores Biológicos/microbiologia , Escherichia coli/genética , Escherichia coli/metabolismo , Mesna/metabolismo , Metano/metabolismo , Modelos Moleculares , Oxirredução , Vitamina B 12/análogos & derivados , Vitamina B 12/metabolismo
17.
Nature ; 539(7629): 396-401, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27749816

RESUMO

The anaerobic formation and oxidation of methane involve unique enzymatic mechanisms and cofactors, all of which are believed to be specific for C1-compounds. Here we show that an anaerobic thermophilic enrichment culture composed of dense consortia of archaea and bacteria apparently uses partly similar pathways to oxidize the C4 hydrocarbon butane. The archaea, proposed genus 'Candidatus Syntrophoarchaeum', show the characteristic autofluorescence of methanogens, and contain highly expressed genes encoding enzymes similar to methyl-coenzyme M reductase. We detect butyl-coenzyme M, indicating archaeal butane activation analogous to the first step in anaerobic methane oxidation. In addition, Ca. Syntrophoarchaeum expresses the genes encoding ß-oxidation enzymes, carbon monoxide dehydrogenase and reversible C1 methanogenesis enzymes. This allows for the complete oxidation of butane. Reducing equivalents are seemingly channelled to HotSeep-1, a thermophilic sulfate-reducing partner bacterium known from the anaerobic oxidation of methane. Genes encoding 16S rRNA and methyl-coenzyme M reductase similar to those identifying Ca. Syntrophoarchaeum were repeatedly retrieved from marine subsurface sediments, suggesting that the presented activation mechanism is naturally widespread in the anaerobic oxidation of short-chain hydrocarbons.


Assuntos
Archaea/metabolismo , Butanos/metabolismo , Mesna/química , Mesna/metabolismo , Alquilação , Anaerobiose , Archaea/genética , Proteínas Arqueais/química , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Biocatálise , Evolução Molecular , Oxirredução , Sulfatos/metabolismo , Temperatura
18.
Angew Chem Int Ed Engl ; 55(44): 13648-13649, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27571920

RESUMO

Find your path: Methyl-coenzyme M reductase (MCR, turquoise) reversibly catalyzes the reduction of methyl-coenzyme M (methyl-S-CoM) with coenzyme B (CoB-SH) to form methane and the heterodisulfide. Recently, spectroscopic methods were used to detect trapped intermediates in a stopped-flow system, and CoM-S-NiII was identified after half a turnover of the MCR reaction (F430 =nickel porphinoid). This finding supports a methyl-radical catalytic mechanism.


Assuntos
Mesna/análogos & derivados , Metano/metabolismo , Fosfotreonina/análogos & derivados , Espectroscopia de Ressonância de Spin Eletrônica , Mesna/metabolismo , Metano/química , Modelos Moleculares , Estrutura Molecular , Fosfotreonina/metabolismo
19.
Biochemistry ; 55(2): 313-21, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26684934

RESUMO

Glutaredoxins (GRXs) are thiol-disulfide oxidoreductases abundant in prokaryotes, although little is understood of these enzymes from the domain Archaea. The numerous characterized GRXs from the domain Bacteria utilize a diversity of low-molecular-weight thiols in addition to glutathione as reductants. We report here the biochemical and structural properties of a GRX-like protein named methanoredoxin (MRX) from Methanosarcina acetivorans of the domain Archaea. MRX utilizes coenzyme M (CoMSH) as reductant for insulin disulfide reductase activity, which adds to the diversity of thiol protectants in prokaryotes. Cell-free extracts of M. acetivorans displayed CoMS-SCoM reductase activity that complements the CoMSH-dependent activity of MRX. The crystal structure exhibits a classic thioredoxin-glutaredoxin fold comprising three α-helices surrounding four antiparallel ß-sheets. A pocket on the surface contains a CVWC motif, identifying the active site with architecture similar to GRXs. Although it is a monomer in solution, the crystal lattice has four monomers in a dimer of dimers arrangement. A cadmium ion is found within the active site of each monomer. Two such ions stabilize the N-terminal tails and dimer interfaces. Our modeling studies indicate that CoMSH and glutathione (GSH) bind to the active site of MRX similar to the binding of GSH in GRXs, although there are differences in the amino acid composition of the binding motifs. The results, combined with our bioinformatic analyses, show that MRX represents a class of GRX-like enzymes present in a diversity of methane-producing Archaea.


Assuntos
Proteínas Arqueais/metabolismo , Glutarredoxinas/metabolismo , Mesna/metabolismo , Methanosarcina/metabolismo , Proteínas Arqueais/química , Glutarredoxinas/química , Proteína Dissulfeto Redutase (Glutationa)/química , Proteína Dissulfeto Redutase (Glutationa)/metabolismo , Estrutura Secundária de Proteína
20.
PLoS One ; 10(11): e0142569, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26562416

RESUMO

2-bromoethanesulfonate (BES) is a structural analogue of coenzyme M (Co-M) and potent inhibitor of methanogenesis. Several studies confirmed, BES can inhibit CH4 prodcution in rice soil, but the suppressing effectiveness of BES application on CH4 emission under rice cultivation has not been studied. In this pot experiment, different levels of BES (0, 20, 40 and 80 mg kg-1) were applied to study its effect on CH4 emission and plant growth during rice cultivation. Application of BES effectively suppressed CH4 emission when compared with control soil during rice cultivation. The CH4 emission rates were significantly (P<0.001) decreased by BES application possibly due to significant (P<0.001) reduction of methnaogenic biomarkers like Co-M concentration and mcrA gene copy number (i.e. methanogenic abunadance). BES significantly (P<0.001) reduced methanogen activity, while it did not affect soil dehydrogenase activity during rice cultivation. A rice plant growth and yield parameters were not affected by BES application. The maximum CH4 reduction (49% reduction over control) was found at 80 mg kg-1 BES application during rice cultivation. It is, therefore, concluded that BES could be a suitable soil amendment for reducing CH4 emission without affecting rice plant growth and productivity during rice cultivation.


Assuntos
Ácidos Alcanossulfônicos/farmacologia , Metano/antagonistas & inibidores , Oryza/efeitos dos fármacos , Agricultura/métodos , Biomassa , Relação Dose-Resposta a Droga , Mesna/metabolismo , Metano/metabolismo , Oryza/crescimento & desenvolvimento , Oryza/metabolismo , Oxirredutases/genética , Oxirredutases/metabolismo , Reação em Cadeia da Polimerase , Solo/química , Microbiologia do Solo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...