Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Am Chem Soc ; 146(6): 3926-3942, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38291562

ABSTRACT

(E)-4-Hydroxy-3-methylbut-2-enyl diphosphate reductase, or IspH (formerly known as LytB), catalyzes the terminal step of the bacterial methylerythritol phosphate (MEP) pathway for isoprene synthesis. This step converts (E)-4-hydroxy-3-methylbut-2-enyl diphosphate (HMBPP) into one of two possible isomeric products, either isopentenyl diphosphate (IPP) or dimethylallyl diphosphate (DMAPP). This reaction involves the removal of the C4 hydroxyl group of HMBPP and addition of two electrons. IspH contains a [4Fe-4S] cluster in its active site, and multiple cluster-based paramagnetic species of uncertain redox and ligation states can be detected after incubation with reductant, addition of a ligand, or during catalysis. To characterize the clusters in these species, 57Fe-labeled samples of IspH were prepared and studied by electron paramagnetic resonance (EPR), 57Fe electron-nuclear double resonance (ENDOR), and Mössbauer spectroscopies. Notably, this ENDOR study provides a rarely reported, complete determination of the 57Fe hyperfine tensors for all four Fe ions in a [4Fe-4S] cluster. The resting state of the enzyme (Ox) has a diamagnetic [4Fe-4S]2+ cluster. Reduction generates [4Fe-4S]+ (Red) with both S = 1/2 and S = 3/2 spin ground states. When the reduced enzyme is incubated with substrate, a transient paramagnetic reaction intermediate is detected (Int) which is thought to contain a cluster-bound substrate-derived species. The EPR properties of Int are indicative of a 3+ iron-sulfur cluster oxidation state, and the Mössbauer spectra presented here confirm this. Incubation of reduced enzyme with the product IPP induced yet another paramagnetic [4Fe-4S]+ species (Red+P) with S = 1/2. However, the g-tensor of this state is commonly associated with a 3+ oxidation state, while Mössbauer parameters show features typical for 2+ clusters. Implications of these complicated results are discussed.


Subject(s)
Hemiterpenes , Iron-Sulfur Proteins , Organophosphorus Compounds , Catalytic Domain , Ligands , Oxidation-Reduction , Electron Spin Resonance Spectroscopy , Catalysis , Iron-Sulfur Proteins/chemistry
2.
J Bacteriol ; 202(3)2020 01 15.
Article in English | MEDLINE | ID: mdl-31740491

ABSTRACT

Catalyzing the key step for anaerobic production and/or oxidation of methane and likely other short-chain alkanes, methyl coenzyme M reductase (Mcr) and its homologs play a key role in the global carbon cycle. The McrA subunit possesses up to five conserved posttranslational modifications (PTMs) at its active site. It was previously suggested that methanogenesis marker protein 10 (Mmp10) could play an important role in methanogenesis. To systematically examine its physiological role, mmpX (locus tag MMP1554), the gene encoding Mmp10 in Methanococcus maripaludis, was deleted with a new genetic tool, resulting in the complete loss of the 5-C-(S)-methylarginine PTM of residue 275 in the McrA subunit. When the ΔmmpX mutant was complemented with the wild-type gene expressed by either a strong or a weak promoter, methylation was fully restored. Compared to the parental strain, maximal rates of methane formation by whole cells were reduced by 40 to 60% in the ΔmmpX mutant. The reduction in activity was fully reversed by the complement with the strong promoter. Site-directed mutagenesis of mmpX resulted in a differential loss of arginine methylation among the mutants in vivo, suggesting that activities of Mmp10 directly modulated methylation. R275 was present in a highly conserved PXRR275(A/S)R(G/A) signature sequence in McrAs. The only other protein in M. maripaludis containing a similar sequence was not methylated, suggesting that Mmp10 is specific for McrA. In conclusion, Mmp10 modulates the methyl-Arg PTM on McrA in a highly specific manner, which has a profound impact on Mcr activity.IMPORTANCE Mcr is the key enzyme in methanogenesis and a promising candidate for bioengineering the conversion of methane to liquid fuel. Our knowledge of Mcr is still limited. In terms of complexity, uniqueness, and environmental importance, Mcr is more comparable to photosynthetic reaction centers than conventional enzymes. PTMs have long been hypothesized to play key roles in modulating Mcr activity. Here, we directly link the mmpX gene to the arginine PTM of Mcr, demonstrate its association with methanogenesis activity, and offer insights into its substrate specificity and putative cofactor binding sites. This is also the first time that a PTM of McrA has been shown to have a substantial impact on both methanogenesis and growth in the absence of additional stressors.


Subject(s)
Arginine/metabolism , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Binding Sites , Blotting, Western , Catalytic Domain , Computational Biology , Mass Spectrometry , Methanococcus/pathogenicity , Methylation , Oxidoreductases/genetics , Protein Processing, Post-Translational , Substrate Specificity
3.
BMC Microbiol ; 20(1): 323, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33096982

ABSTRACT

BACKGROUND: The production of methane by methanogens is dependent on numerous iron-sulfur (Fe-S) cluster proteins; yet, the machinery involved in Fe-S cluster biogenesis in methanogens remains largely unknown. Methanogen genomes encode uncharacterized homologs of the core components of the ISC (IscS and IscU) and SUF (SufBC) Fe-S cluster biogenesis systems found in bacteria and eukaryotes. Methanosarcina acetivorans contains three iscSU and two sufCB gene clusters. Here, we report genetic and biochemical characterization of M. acetivorans iscSU2. RESULTS: Purified IscS2 exhibited pyridoxal 5'- phosphate-dependent release of sulfur from L-cysteine. Incubation of purified IscU2 with IscS2, cysteine, and iron (Fe2+) resulted in the formation of [4Fe-4S] clusters in IscU2. IscU2 transferred a [4Fe-4S] cluster to purified M. acetivorans apo-aconitase. IscU2 also restored the aconitase activity in air-exposed M. acetivorans cell lysate. These biochemical results demonstrate that IscS2 is a cysteine desulfurase and that IscU2 is a Fe-S cluster scaffold. M. acetivorans strain DJL60 deleted of iscSU2 was generated to ascertain the in vivo importance of IscSU2. Strain DJL60 had Fe-S cluster content and growth similar to the parent strain but lower cysteine desulfurase activity. Strain DJL60 also had lower intracellular persulfide content compared to the parent strain when cysteine was an exogenous sulfur source, linking IscSU2 to sulfur metabolism. CONCLUSIONS: This study establishes that M. acetivorans contains functional IscS and IscU, the core components of the ISC Fe-S cluster biogenesis system and provides the first evidence that ISC operates in methanogens.


Subject(s)
Carbon-Sulfur Lyases/metabolism , Escherichia coli Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Iron-Sulfur Proteins/metabolism , Methanosarcina/genetics , Carbon-Sulfur Lyases/genetics , Cysteine/metabolism , Enzyme Activation , Escherichia coli Proteins/genetics , Iron/metabolism , Iron-Sulfur Proteins/genetics , Methanosarcina/enzymology , Sulfur/metabolism
4.
Proc Natl Acad Sci U S A ; 113(22): 6172-7, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27140643

ABSTRACT

Ruminants, such as cows, sheep, and goats, predominantly ferment in their rumen plant material to acetate, propionate, butyrate, CO2, and methane. Whereas the short fatty acids are absorbed and metabolized by the animals, the greenhouse gas methane escapes via eructation and breathing of the animals into the atmosphere. Along with the methane, up to 12% of the gross energy content of the feedstock is lost. Therefore, our recent report has raised interest in 3-nitrooxypropanol (3-NOP), which when added to the feed of ruminants in milligram amounts persistently reduces enteric methane emissions from livestock without apparent negative side effects [Hristov AN, et al. (2015) Proc Natl Acad Sci USA 112(34):10663-10668]. We now show with the aid of in silico, in vitro, and in vivo experiments that 3-NOP specifically targets methyl-coenzyme M reductase (MCR). The nickel enzyme, which is only active when its Ni ion is in the +1 oxidation state, catalyzes the methane-forming step in the rumen fermentation. Molecular docking suggested that 3-NOP preferably binds into the active site of MCR in a pose that places its reducible nitrate group in electron transfer distance to Ni(I). With purified MCR, we found that 3-NOP indeed inactivates MCR at micromolar concentrations by oxidation of its active site Ni(I). Concomitantly, the nitrate ester is reduced to nitrite, which also inactivates MCR at micromolar concentrations by oxidation of Ni(I). Using pure cultures, 3-NOP is demonstrated to inhibit growth of methanogenic archaea at concentrations that do not affect the growth of nonmethanogenic bacteria in the rumen.


Subject(s)
Methane/chemistry , Molecular Docking Simulation , Animals , Euryarchaeota/metabolism , Oxidoreductases/chemistry , Rumen/metabolism , Ruminants/metabolism
5.
J Bacteriol ; 200(7)2018 04 01.
Article in English | MEDLINE | ID: mdl-29339414

ABSTRACT

Methyl coenzyme M reductase (MCR) is a complex enzyme that catalyzes the final step in biological methanogenesis. To better understand its assembly, the recombinant MCR from the thermophile Methanothermococcus okinawensis (rMCRok) was expressed in the mesophile Methanococcus maripaludis The rMCRok was posttranslationally modified correctly and contained McrD and the unique nickel tetrapyrrole coenzyme F430 Subunits of the native M. maripaludis (MCRmar) were largely absent, suggesting that the recombinant enzyme was formed by an assembly of cotranscribed subunits. Strong support for this hypothesis was obtained by expressing a chimeric operon comprising the His-tagged mcrA from M. maripaludis and the mcrBDCG from M. okinawensis in M. maripaludis The His-tagged purified rMCR then contained the M. maripaludis McrA and the M. okinawensis McrBDG. The present study prompted us to form a working model for MCR assembly, which can be further tested by the heterologous expression system established here.IMPORTANCE Approximately 1.6% of the net primary production of plants, algae, and cyanobacteria are processed by biological methane production in anoxic environments. This accounts for about 74% of the total global methane production, up to 25% of which is consumed by anaerobic oxidation of methane (AOM). Methyl coenzyme M reductase (MCR) is the key enzyme in both methanogenesis and AOM. MCR is assembled as a dimer of two heterotrimers, where posttranslational modifications and F430 cofactors are embedded in the active sites. However, this complex assembly process remains unknown. Here, we established a heterologous expression system for MCR to learn how MCR is assembled.


Subject(s)
Methane/metabolism , Methanococcus/enzymology , Oxidoreductases/genetics , Protein Processing, Post-Translational/genetics , Binding Sites , Catalysis , Metalloporphyrins/chemistry , Methanococcus/metabolism , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/metabolism
6.
J Phys Chem A ; 122(36): 7118-7130, 2018 Sep 13.
Article in English | MEDLINE | ID: mdl-30160119

ABSTRACT

Exposure of sulfonated poly(ether etherketone), SPEEK, in aqueous solutions to 350 nm photons induced reduction of CHCl3 to CH2Cl2 and chloride ions in the presence of HCO2H/HCO2- buffers or poly(vinyl alcohol), PVA. The kinetics of the SPEEK-sensitized photoreaction was characterized by quantum yields of halide ion formation, ϕ(Cl-), evaluated from in situ determinations of [Cl-]. Particularly efficient reductions took place when formate buffers served as H atom donors in the absence of air and with excess CHCl3. The dependence of ϕ(Cl-) on the inverse square root of the light intensity together with postirradiation formation of Cl- in the dark indicated that the CHCl3 photoreduction occurred via a chain process. EPR determinations identified the α-hydroxy radical of SPEEK and •CHCl2 as chain carriers. Most of the kinetic findings were rationalized in terms of a free radical mechanism where dimerizations of the radicals acted as termination steps. Photoreduction of CHCl3 was also detected in the presence of air albeit with lower quantum efficiencies. Observations made during postirradiation experiments indicated that a chain process was also operative under such conditions.

7.
J Am Chem Soc ; 137(13): 4567-80, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25790339

ABSTRACT

Reduced forms of the C56S and C60S variants of the thioredoxin-like Clostridium pasteurianum [Fe2S2] ferredoxin (CpFd) provide the only known examples of valence-delocalized [Fe2S2](+) clusters, which constitute a fundamental building block of all higher nuclearity Fe-S clusters. In this work, we have revisited earlier work on the CpFd variants and carried out redox and spectroscopic studies on the [Fe2S2](2+,+) centers in wild-type and equivalent variants of the highly homologous and structurally characterized Aquifex aeolicus ferredoxin 4 (AaeFd4) using EPR, UV-visible-NIR absorption, CD and variable-temperature MCD, and protein-film electrochemistry. The results indicate that the [Fe2S2](+) centers in the equivalent AaeFd4 and CpFd variants reversibly interconvert between similar valence-localized S = 1/2 and valence-delocalized S = 9/2 forms as a function of pH, with pKa values in the range 8.3-9.0, because of protonation of the coordinated serinate residue. However, freezing high-pH samples results in partial or full conversion from valence-delocalized S = 9/2 to valence-localized S = 1/2 [Fe2S2](+) clusters. MCD saturation magnetization data for valence-delocalized S = 9/2 [Fe2S2](+) centers facilitated determination of transition polarizations and thereby assignments of low-energy MCD bands associated with the Fe-Fe interaction. The assignments provide experimental assessment of the double exchange parameter, B, for valence-delocalized [Fe2S2](+) centers and demonstrate that variable-temperature MCD spectroscopy provides a means of detecting and investigating the properties of valence-delocalized S = 9/2 [Fe2S2](+) fragments in higher nuclearity Fe-S clusters. The origin of valence delocalization in thioredoxin-like ferredoxin Cys-to-Ser variants and Fe-S clusters in general is discussed in light of these results.


Subject(s)
Ferredoxins/chemistry , Iron/chemistry , Sulfur/chemistry , Thioredoxins/chemistry , Aquifoliaceae , Clostridium , Ferredoxins/metabolism , Iron/metabolism , Oxidation-Reduction , Spectrum Analysis , Sulfur/metabolism
8.
J Bacteriol ; 196(13): 2491-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769699

ABSTRACT

Methyl-coenzyme M reductase (MCR) catalyzes the reversible reduction of methyl-coenzyme M (CH3-S-CoM) and coenzyme B (HS-CoB) to methane and heterodisulfide CoM-S-S-CoB (HDS). MCR contains the hydroporphinoid nickel complex coenzyme F430 in its active site, and the Ni center has to be in its Ni(I) valence state for the enzyme to be active. Until now, no in vitro method that fully converted the inactive MCRsilent-Ni(II) form to the active MCRred1-Ni(I) form has been described. With the potential use of recombinant MCR in the production of biofuels and the need to better understand this enzyme and its activation process, we studied its activation under nonturnover conditions and achieved full MCR activation in the presence of dithiothreitol and protein components A2, an ATP carrier, and A3a. It was found that the presence of HDS promotes the inactivation of MCRred1, which makes it essential that the activation process is isolated from the methane formation assay, which tends to result in minimal activation rates. Component A3a is a multienzyme complex that includes the mcrC gene product, an Fe-protein homolog, an iron-sulfur flavoprotein, and protein components involved in electron bifurcation. A hypothetical model for the cellular activation process of MCR is presented.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Methanobacteriaceae/enzymology , Oxidoreductases/metabolism , Amino Acid Sequence , Cloning, Molecular , Dithiothreitol , Enzyme Activation/physiology , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Methanobacteriaceae/genetics , Methanobacteriaceae/metabolism , Molecular Sequence Data , Oxidoreductases/genetics
9.
FEBS Open Bio ; 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877345

ABSTRACT

Methyl-coenzyme M reductase (MCR) is a multi-subunit (α2ß2γ2) enzyme responsible for methane formation via its unique F430 cofactor. The genes responsible for producing MCR (mcrA, mcrB and mcrG) are typically colocated with two other highly conserved genes mcrC and mcrD. We present here the high-resolution crystal structure for McrD from a human gut methanogen Methanomassiliicoccus luminyensis strain B10. The structure reveals that McrD comprises a ferredoxin-like domain assembled into an α + ß barrel-like dimer with conformational flexibility exhibited by a functional loop. The description of the M. luminyensis McrD crystal structure contributes to our understanding of this key conserved methanogen protein typically responsible for promoting MCR activity and the production of methane, a greenhouse gas.

10.
Chemosphere ; 353: 141562, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417493

ABSTRACT

The UV/sulfite-based advanced reduction process (ARP) emerges as an effective strategy to combat per- and polyfluoroalkyl substances (PFAS) pollution in water. Yet, the UV/sulfite-ARP typically operates at highly alkaline conditions (e.g., pH > 9 or even higher) since the generated reductive radicals for PFAS degradation can be quickly sequestered by protons (H+). To overcome the associated challenges, we prototyped a biochar-surfactant-system (BSS) to synergistically enhance PFAS sorption and degradation by UV/sulfite-ARP. The degradation and defluorination efficiencies of perfluorooctanoic acid (PFOA) depended on solution pH, and concentrations of surfactant (cetyltrimethylammonium bromide; CTAB), sulfite, and biochar. At high pH (8-10), adding biochar and BSS showed no or even small inhibitory effect on PFOA degradation, since the degradation efficiencies were already high enough that cannot be differentiated. However, at acidic and neutral pH (6-7), an evident enhancement of PFOA degradation and defluorination efficiencies occurred. This is due to the synergies between biochar and CTAB that create favorable microenvironments for enhanced PFOA sorption and deeper destruction by prolonging the longevity of reductive radicals (e.g., SO3•-), which is less affected by ambient pH conditions. The performance of UV/sulfite/BSS was further optimized and used for the degradation of four PFAS. At the optimal experimental condition, the UV/sulfite/BSS system can completely degrade PFOA with >30% defluorination efficiency for up to five continuous cycles (n = 5). Overall, our BSS provides a cost-effective and sustainable technique to effectively degrade PFAS in water under environmentally relevant pH conditions. The BSS-enabled ARP technique can be easily tied into PFAS treatment train technology (e.g., advanced oxidation process) for more efficient and deeper defluorination of various PFAS in water.


Subject(s)
Caprylates , Charcoal , Fluorocarbons , Pulmonary Surfactants , Water Pollutants, Chemical , Surface-Active Agents , Cetrimonium , Water , Hydrogen-Ion Concentration , Fluorocarbons/analysis , Sulfites , Water Pollutants, Chemical/analysis
11.
Biochemistry ; 52(10): 1705-16, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23394607

ABSTRACT

Acetyl-CoA synthase (ACS) catalyzes the reversible condensation of CO and CH3 units at a unique Ni-Fe cluster, the A cluster, to form an acetyl-Ni intermediate that subsequently reacts with CoA to produce acetyl-CoA. ACS is a component of the multienzyme complex acetyl-CoA decarbonylase/synthase (ACDS) in Archaea and CO dehydrogenase/ACS (CODH/ACS) in bacteria; in both systems, intraprotein CO channeling takes place between the CODH and ACS active sites. Previous studies indicated that protein conformational changes control the chemical reactivity of the A cluster and suggested the involvement of a conserved Phe residue that moves concomitantly into and out of the coordination environment of Ni. Herein, steady-state rate measurements in which both CO and CH3-corrinoid are varied, and rapid methylation reactions of the ACDS ß subunit, measured by stopped-flow methods, provide a kinetic model for acetyl-CoA synthesis that includes a description of the inhibitory effects of CO explained by competition of CO and CH3 for the same form of the enzyme. Electron paramagnetic resonance titrations revealed that the formation of a paramagnetic Ni(+)-CO species does not match the kinetics of CO interaction as a substrate but instead correlates well with an inhibited state of the enzyme, which requires revision of previous models that postulate that this species is an intermediate. Characterization of the ß subunit F195A variant showed markedly increased substrate reactivity with CO, which provides biochemical functional evidence of steric shielding of the CO substrate interaction site by the phenyl group side chain. The phenyl group also likely enhances the nucleophilicity of the Ni center to facilitate CH3 group transfer. A model was developed for how the catalytic properties of the A cluster are optimized by linking conformational changes to a repositionable aromatic shield able to modulate the nucleophilicity of Ni, sterically select the most productive order of substrate addition, and overcome intrinsic inhibition by CO.


Subject(s)
Carbon Monoxide/metabolism , Coenzyme A Ligases/chemistry , Coenzyme A Ligases/metabolism , Aldehyde Oxidoreductases/chemistry , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Archaeal Proteins/chemistry , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coenzyme A Ligases/genetics , Conserved Sequence , Electron Spin Resonance Spectroscopy , Kinetics , Methanosarcina/enzymology , Methanosarcina/genetics , Models, Molecular , Moorella/enzymology , Moorella/genetics , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , Mutagenesis, Site-Directed , Nickel/metabolism , Phenylalanine/chemistry , Protein Conformation , Protein Subunits
12.
J Inorg Biochem ; 240: 112098, 2023 03.
Article in English | MEDLINE | ID: mdl-36580832

ABSTRACT

Acetyl-CoA synthase (ACS) is a central enzyme in the carbon and energy metabolism of certain anaerobic species of bacteria and archaea that catalyzes the direct synthesis and cleavage of the acetyl CC bond of acetyl-CoA by an unusual enzymatic mechanism of special interest for its use of organonickel intermediates. An Fe4S4 cluster associated with a proximal, reactive Nip and distal spectator Nid comprise the active site metal complex, known as the A cluster. Experimental and theoretical methods have uncovered much about the ACS mechanism, but have also opened new unanswered questions about the structure and reactivity of the A cluster in various intermediate forms. Here we report a method for large scale isolation of ACS with its A cluster in the acetylated state. Isolated acetyl-ACS and the two-electron reduced ACS, produced by acetyl-ACS reaction with CoA, were characterized by UV-visible and EPR spectroscopy. Reactivity with electron acceptors provided an assessment of the apparent Em for two-electron reduction of the A cluster. The results help to distinguish between alternative electronic states of the reduced cluster, provide evidence for a role of the Fe/S cluster in catalysis, and offer an explanation of why one-electron reductive activation is observed for a reaction cycle involving 2-electron chemistry.


Subject(s)
Bacteria , Electrons , Acetyl Coenzyme A , Bacteria/metabolism , Electron Spin Resonance Spectroscopy , Archaea , Nitric Oxide Synthase , Carbon Monoxide/chemistry
13.
Biochemistry ; 51(24): 4835-49, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22646150

ABSTRACT

(E)-4-Hydroxy-3-methylbut-2-enyl diphosphate reductase (IspH or LytB) catalyzes the terminal step of the MEP/DOXP pathway where it converts (E)-4-hydroxy-3-methylbut-2-enyl diphosphate (HMBPP) into the two products, isopentenyl diphosphate and dimethylallyl diphosphate. The reaction involves the reductive elimination of the C4 hydroxyl group, using a total of two electrons. Here we show that the active form of IspH contains a [4Fe-4S] cluster and not the [3Fe-4S] form. Our studies show that the cluster is the direct electron source for the reaction and that a reaction intermediate is bound directly to the cluster. This active form has been trapped in a state, dubbed FeS(A), that was detected by electron paramagnetic resonance (EPR) spectroscopy when one-electron-reduced IspH was incubated with HMBPP. In addition, three mutants of IspH have been prepared and studied, His42, His124, and Glu126 (Aquifex aeolicus numbering), with particular attention paid to the effects on the cluster properties and possible reaction intermediates. None of the mutants significantly affected the properties of the [4Fe-4S](+) cluster, but different effects were observed when one-electron-reduced forms were incubated with HMBPP. Replacing His42 led to an increased K(M) value and a much lower catalytic efficiency, confirming the role of this residue in substrate binding. Replacing the His124 also resulted in a lower catalytic efficiency. In this case, however, the enzyme showed the loss of the [4Fe-4S](+) EPR signal upon addition of HMBPP without the subsequent formation of the FeS(A) signal. Instead, a radical-type signal was observed in some of the samples, indicating that this residue plays a role in the correct positioning of the substrate. The incorrect orientation in the mutant leads to the formation of substrate-based radicals instead of the cluster-bound intermediate complex FeS(A). Replacing the Glu126 also resulted in a lower catalytic efficiency, with yet a third type of EPR signal being detected upon incubation with HMBPP. (31)P and (2)H ENDOR measurements of the FeS(A) species incubated with regular and (2)H-C4-labeled HMBPP reveal that the substrate binds to the enzyme in the proximity of the active-site cluster with C4 adjacent to the site of linkage between the FeS cluster and HMBPP. Comparison of the spectroscopic properties of this intermediate to those of intermediates detected in (E)-4-hydroxy-3-methylbut-2-enyl diphosphate synthase and ferredoxin:thioredoxin reductase suggests that HMBPP binds to the FeS cluster via its hydroxyl group instead of a side-on binding as previously proposed for the species detected in the inactive Glu126 variant. Consequences for the IspH reaction mechanism are discussed.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutation , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Spectrum Analysis , Binding Sites , Escherichia coli/enzymology , Escherichia coli Proteins/genetics , Iron/metabolism , Mutant Proteins/genetics , Organophosphorus Compounds/metabolism , Oxidoreductases/genetics , Sulfur/metabolism
14.
Commun Biol ; 5(1): 1113, 2022 10 20.
Article in English | MEDLINE | ID: mdl-36266535

ABSTRACT

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.


Subject(s)
Archaea , Mesna , Archaea/genetics , Archaea/metabolism , Mesna/metabolism , Oxidoreductases/metabolism , Methane/metabolism
15.
J Biol Chem ; 285(20): 15450-15463, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20202935

ABSTRACT

Direct synthesis and cleavage of acetyl-CoA are carried out by the bifunctional CO dehydrogenase/acetyl-CoA synthase enzyme in anaerobic bacteria and by the acetyl-CoA decarbonylase/synthase (ACDS) multienzyme complex in Archaea. In both systems, a nickel- and Fe/S-containing active site metal center, the A cluster, catalyzes acetyl C-C bond formation/breakdown. Carbonyl group exchange of [1-(14)C]acetyl-CoA with unlabeled CO, a hallmark of CODH/ACS, is weakly active in ACDS, and exchange with CO(2) was up to 350 times faster, indicating tight coupling of CO release at the A cluster to CO oxidation to CO(2) at the C cluster in CO dehydrogenase. The basis for tight coupling was investigated by analysis of three recombinant A cluster proteins, ACDS beta subunit from Methanosarcina thermophila, acetyl-CoA synthase of Carboxydothermus hydrogenoformans (ACS(Ch)), and truncated ACS(Ch) lacking its 317-amino acid N-terminal domain. A comparison of acetyl-CoA synthesis kinetics, CO exchange, acetyltransferase, and A cluster Ni(+)-CO EPR characteristics demonstrated a direct role of the ACS N-terminal domain in promoting acetyl C-C bond fragmentation. Protein conformational changes, related to "open/closed" states previously identified crystallographically, were indicated to have direct effects on the coordination geometry and stability of the A cluster Ni(2+)-acetyl intermediate, controlling Ni(2+)-acetyl fragmentation and Ni(2+)(CO)(CH(3)) condensation. EPR spectral changes likely reflect variations in the Ni(+)-CO equatorial coordination environment in closed buried hydrophobic and open solvent-exposed states. The involvement of subunit-subunit interactions in ACDS, versus interdomain contacts in ACS, ensures that CO is not released from the ACDS beta subunit in the absence of appropriate interactions with the alpha(2)epsilon(2) CO dehydrogenase component. The resultant high efficiency CO transfer explains the low rate of CO exchange relative to CO(2).


Subject(s)
Aldehyde Oxidoreductases/metabolism , Methanosarcina/enzymology , Multienzyme Complexes/metabolism , Aldehyde Oxidoreductases/chemistry , Carbon Monoxide/metabolism , Crystallography, X-Ray , Electron Spin Resonance Spectroscopy , Models, Molecular , Multienzyme Complexes/chemistry , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
16.
Anal Chem ; 83(9): 3358-64, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21456571

ABSTRACT

We present a new methodology for generating a stepwise concentration gradient in a series of microdroplets by using monolithic micro valves that act as "faucets" in micrometer-scale. A distinct concentration gradient of a substrate was generated for the determination of the kinetic parameters of two different enzymes using only 10 picoliter-scale droplets. With a single experiment on a chip, we obtained K(M) and k(cat) values of matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9), and compared the catalytic competence of the two enzymes. The present system and method are highly suitable for applications where the reagents or samples are limited and precious.


Subject(s)
Enzyme Assays/methods , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Biocatalysis , Calibration , Kinetics , Microtechnology
17.
Anal Chem ; 83(16): 6148-53, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21696192

ABSTRACT

We demonstrate the accommodation of log-scale concentration gradients of inhibitors on a single microfluidic chip with a semidirect dilution capability of reagents for the determination of the half-inhibitory concentration or IC(50). The chip provides a unique tool for hosting a wide-range of concentration gradient for studies that require an equal distribution of measuring points on a logarithmic scale. Using Matrix metalloproteinase IX and three of its inhibitors, marimastat, batimastat, and CP471474, we evaluated the IC(50) of each inhibitor with a single experiment. The present work could be applied to the systematic study of biochemical binding and inhibition processes particularly in the field of mechanistic enzymology and the pharmaceutical industry.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Lab-On-A-Chip Devices , Matrix Metalloproteinase Inhibitors , Microchip Analytical Procedures/methods , Neoplasms/enzymology , Dose-Response Relationship, Drug , Humans , Hydroxamic Acids/pharmacology , Inhibitory Concentration 50 , Kinetics , Matrix Metalloproteinase 9/metabolism , Neoplasms/drug therapy , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Sensitivity and Specificity , Spectrometry, Fluorescence , Thiophenes/pharmacology
18.
Inorg Chem ; 50(18): 8869-78, 2011 Sep 19.
Article in English | MEDLINE | ID: mdl-21842834

ABSTRACT

A dormant macromolecular catalyst was prepared by polymerization of an aqueous styrene-butyl acrylate miniemulsion in the presence of a new polymerizable pentadentate ligand. The catalyst was activated by binding Cu(II) ions to the ligand site and then explored for its ability to hydrolyze glycosidic bonds in alkaline solution. The performance was correlated to the catalytic activity shown by low molecular weight analogs. A turnover rate of up to 43 × 10(-4) min(-1) was previously observed for cleavage of the glycosidic bond in selected p-nitrophenylglycosides with a binuclear, low molecular weight catalyst; by contrast, the same reaction is more than 1 order of magnitude faster and has a turnover rate of up to 380 × 10(-4) min(-1) when using the prepared macromolecular catalyst. The catalyzed hydrolysis is about 10(5)-fold accelerated over the uncatalyzed background reaction under the provided conditions, while a significant discrimination of the α- and ß-glycosidic bond or of the galacto- and gluco-configuration in the sugar moiety in the glycoside substrates is not observed.

19.
Phys Chem Chem Phys ; 13(6): 2179-87, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21132188

ABSTRACT

Fe- and Mn-promoted H(2)S sorbents Fe(x)-Mn(y)-Zn(1-x-y)O/SiO(2) (x, y = 0, 0.025) for desulfurization of model fuel reformates at room temperature were prepared, tested and characterized. Sulfur uptake capacity at 25 °C significantly exceeds that of both commercial unsupported ZnO sorbents and un-promoted supported ZnO/SiO(2) sorbents. Sulfur capacity and breakthrough characteristics remain satisfactory after multiple (∼10) cycles of adsorption/regeneration, with regeneration performed by a simple and robust heating in air. XRD shows that both "calcined" and "spent" sorbents contain nano-dispersed ZnO, and XPS confirms conversion of ZnO to ZnS. "Calcined" sorbent contains Fe(3+) and Mn(3+) that are reduced to Mn(2+) upon reaction with H(2)S, but not with H(2). Operando ESR is used for the first time to study dynamics of reduction of Mn(3+) promoter sites simultaneously with measuring sulfidation dynamics of the Fe(x)-Mn(y)-Zn(1-x-y)O/SiO(2) sorbent. Fe cations are believed to occupy the surface of supported ZnO nanocrystallites, while Mn cations are distributed within ZnO.

20.
Dalton Trans ; 50(3): 926-935, 2021 Jan 27.
Article in English | MEDLINE | ID: mdl-33350418

ABSTRACT

Two NNN pincer complexes of Cu(ii) and Ni(ii) with BPIMe- [BPIMe- = 1,3-bis((6-methylpyridin-2-yl)imino)isoindolin-2-ide] have been prepared and characterized structurally, spectroscopically, and electrochemically. The single crystal structures of the two complexes confirmed their distorted trigonal bipyramidal geometry attained by three equatorial N-atoms from the ligand and two axially positioned water molecules to give [Cu(BPIMe)(H2O)2]ClO4 and [Ni(BPIMe)(H2O)2]ClO4. Electrochemical studies of Cu(ii) and Ni(ii) complexes have been performed in acetonitrile to identify metal-based and ligand-based redox activity. When subjected to a saturated CO2 atmosphere, both complexes displayed catalytic activity for the reduction of CO2 with the Cu(ii) complex displaying higher activity than the Ni(ii) analogue. However, both complexes were shown to decompose into catalytically active heterogeneous materials on the electrode surface over extended reductive electrolysis periods. Surface analysis of these materials using energy dispersive spectroscopy as well as their physical appearance suggests the reductive deposition of copper and nickel metal on the electrode surface. Electrocatalysis and decomposition are proposed to be triggered by ligand reduction, where complex stability is believed to be tied to fluxional ligand coordination in the reduced state.

SELECTION OF CITATIONS
SEARCH DETAIL