Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 484
Filtrar
1.
Annu Rev Biochem ; 85: 455-83, 2016 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26844394

RESUMO

Nitrogenase is a versatile metalloenzyme that is capable of catalyzing two important reactions under ambient conditions: the reduction of nitrogen (N2) to ammonia (NH3), a key step in the global nitrogen cycle; and the reduction of carbon monoxide (CO) and carbon dioxide (CO2) to hydrocarbons, two reactions useful for recycling carbon waste into carbon fuel. The molybdenum (Mo)- and vanadium (V)-nitrogenases are two homologous members of this enzyme family. Each of them contains a P-cluster and a cofactor, two high-nuclearity metalloclusters that have crucial roles in catalysis. This review summarizes the progress that has been made in elucidating the biosynthetic mechanisms of the P-cluster and cofactor species of nitrogenase, focusing on what is known about the assembly mechanisms of the two metalloclusters in Mo-nitrogenase and giving a brief account of the possible assembly schemes of their counterparts in V-nitrogenase, which are derived from the homology between the two nitrogenases.


Assuntos
Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Coenzimas/metabolismo , Molibdênio/metabolismo , Nitrogenase/metabolismo , Subunidades Proteicas/metabolismo , Sequência de Aminoácidos , Amônia/química , Amônia/metabolismo , Azotobacter vinelandii/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Dióxido de Carbono/química , Dióxido de Carbono/metabolismo , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Coenzimas/química , Ferro/química , Ferro/metabolismo , Molibdênio/química , Nitrogênio/química , Nitrogênio/metabolismo , Nitrogenase/química , Nitrogenase/genética , Oxirredução , Subunidades Proteicas/química , Subunidades Proteicas/genética , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Vanádio/química , Vanádio/metabolismo
2.
Nat Chem Biol ; 20(8): 1078-1085, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38890433

RESUMO

Biological nitrogen fixation requires substantial metabolic energy in form of ATP as well as low-potential electrons that must derive from central metabolism. During aerobic growth, the free-living soil diazotroph Azotobacter vinelandii transfers electrons from the key metabolite NADH to the low-potential ferredoxin FdxA that serves as a direct electron donor to the dinitrogenase reductases. This process is mediated by the RNF complex that exploits the proton motive force over the cytoplasmic membrane to lower the midpoint potential of the transferred electron. Here we report the cryogenic electron microscopy structure of the nitrogenase-associated RNF complex of A. vinelandii, a seven-subunit membrane protein assembly that contains four flavin cofactors and six iron-sulfur centers. Its function requires the strict coupling of electron and proton transfer but also involves major conformational changes within the assembly that can be traced with a combination of electron microscopy and modeling.


Assuntos
Azotobacter vinelandii , Microscopia Crioeletrônica , Fixação de Nitrogênio , Azotobacter vinelandii/metabolismo , Azotobacter vinelandii/enzimologia , Modelos Moleculares , Conformação Proteica , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Oxirredutases/metabolismo , Oxirredutases/química
3.
Biochem Biophys Res Commun ; 728: 150345, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-38971001

RESUMO

The transfer of nitrogen fixation (nif) genes from diazotrophs to non-diazotrophic hosts is of increasing interest for engineering biological nitrogen fixation. A recombinant Escherichia coli strain expressing Azotobacter vinelandii 18 nif genes (nifHDKBUSVQENXYWZMF, nifiscA, and nafU) were previously constructed and showed nitrogenase activity. In the present study, we constructed several E. coli strain derivatives in which all or some of the 18 nif genes were additionally integrated into the fliK locus of the chromosome in various combinations. E. coli derivatives with the chromosomal integration of nifiscA, nifU, and nifS, which are involved in the biosynthesis of the [4Fe-4S] cluster of dinitrogenase reductase, exhibited enhanced nitrogenase activity. We also revealed that overexpression of E. coli fldA and ydbK, which encode flavodoxin and flavodoxin-reducing enzyme, respectively, enhanced nitrogenase activity, likely by facilitating electron transfer to dinitrogenase reductase. The additional expression of nifM, putatively involved in maturation of dinitrogenase reductase, further enhanced nitrogenase activity and the amount of soluble NifH. By combining these factors, we successfully improved nitrogenase activity 10-fold.


Assuntos
Azotobacter vinelandii , Escherichia coli , Fixação de Nitrogênio , Nitrogenase , Azotobacter vinelandii/genética , Azotobacter vinelandii/enzimologia , Azotobacter vinelandii/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Nitrogenase/metabolismo , Nitrogenase/genética , Fixação de Nitrogênio/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo
4.
Curr Microbiol ; 81(10): 311, 2024 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-39153035

RESUMO

The two-component system GacS/A and the posttranscriptional control system Rsm constitute a genetic regulation pathway in Gammaproteobacteria; in some species of Pseudomonas, this pathway is part of a multikinase network (MKN) that regulates the activity of the Rsm system. In this network, the activity of GacS is controlled by other kinases. One of the most studied MKNs is the MKN-GacS of Pseudomonas aeruginosa, where GacS is controlled by the kinases RetS and LadS; RetS decreases the kinase activity of GacS, whereas LadS stimulates the activity of the central kinase GacS. Outside of the Pseudomonas genus, the network has been studied only in Azotobacter vinelandii. In this work, we report the study of the RetS kinase of A. vinelandii; as expected, the phenotypes affected in gacS mutants, such as production of alginates, polyhydroxybutyrate, and alkylresorcinols and swimming motility, were also affected in retS mutants. Interestingly, our data indicated that RetS in A. vinelandii acts as a positive regulator of GacA activity. Consistent with this finding, mutation in retS also negatively affected the expression of small regulatory RNAs belonging to the Rsm family. We also confirmed the interaction of RetS with GacS, as well as with the phosphotransfer protein HptB.


Assuntos
Alginatos , Azotobacter vinelandii , Proteínas de Bactérias , Regulação Bacteriana da Expressão Gênica , Azotobacter vinelandii/genética , Azotobacter vinelandii/enzimologia , Azotobacter vinelandii/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Alginatos/metabolismo , Resorcinóis/metabolismo , Histidina Quinase/genética , Histidina Quinase/metabolismo , Poliésteres/metabolismo , Hidroxibutiratos/metabolismo
5.
PLoS Genet ; 17(6): e1009617, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34111137

RESUMO

The energetic requirements for biological nitrogen fixation necessitate stringent regulation of this process in response to diverse environmental constraints. To ensure that the nitrogen fixation machinery is expressed only under appropriate physiological conditions, the dedicated NifL-NifA regulatory system, prevalent in Proteobacteria, plays a crucial role in integrating signals of the oxygen, carbon and nitrogen status to control transcription of nitrogen fixation (nif) genes. Greater understanding of the intricate molecular mechanisms driving transcriptional control of nif genes may provide a blueprint for engineering diazotrophs that associate with cereals. In this study, we investigated the properties of a single amino acid substitution in NifA, (NifA-E356K) which disrupts the hierarchy of nif regulation in response to carbon and nitrogen status in Azotobacter vinelandii. The NifA-E356K substitution enabled overexpression of nitrogenase in the presence of excess fixed nitrogen and release of ammonia outside the cell. However, both of these properties were conditional upon the nature of the carbon source. Our studies reveal that the uncoupling of nitrogen fixation from its assimilation is likely to result from feedback regulation of glutamine synthetase, allowing surplus fixed nitrogen to be excreted. Reciprocal substitutions in NifA from other Proteobacteria yielded similar properties to the A. vinelandii counterpart, suggesting that this variant protein may facilitate engineering of carbon source-dependent ammonia excretion amongst diverse members of this family.


Assuntos
Amônia/metabolismo , Azotobacter vinelandii/genética , Proteínas de Bactérias/genética , Glutamato-Amônia Ligase/genética , Nitrogênio/metabolismo , Nitrogenase/genética , Fatores de Transcrição/genética , Substituição de Aminoácidos , Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Carbono/metabolismo , Regulação Bacteriana da Expressão Gênica , Teste de Complementação Genética , Glutamato-Amônia Ligase/metabolismo , Mutação , Fixação de Nitrogênio , Nitrogenase/metabolismo , Oxigênio/metabolismo , Solo/química , Microbiologia do Solo , Fatores de Transcrição/metabolismo , Transcrição Gênica
6.
Angew Chem Int Ed Engl ; 63(21): e202400273, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38527309

RESUMO

Nitrogenase reduces N2 to NH3 at its active-site cofactor. Previous studies of an N2-bound Mo-nitrogenase from Azotobacter vinelandii suggest binding of three N2 species via asymmetric belt-sulfur displacements in the two cofactors of its catalytic component (designated Av1*), leading to the proposal of stepwise N2 reduction involving all cofactor belt-sulfur sites; yet, the evidence for the existence of multiple N2 species on Av1* remains elusive. Here we report a study of ATP-independent, EuII/SO3 2--driven turnover of Av1* using GC-MS and frequency-selective pulse NMR techniques. Our data demonstrate incorporation of D2-derived D by Av1* into the products of C2H2- and H+-reduction, and decreased formation of NH3 by Av1* concomitant with the release of N2 under H2; moreover, they reveal a strict dependence of these activities on SO3 2-. These observations point to the presence of distinct N2 species on Av1*, thereby providing strong support for our proposed mechanism of stepwise reduction of N2 via belt-sulfur mobilization.


Assuntos
Azotobacter vinelandii , Nitrogênio , Nitrogenase , Nitrogenase/metabolismo , Nitrogenase/química , Azotobacter vinelandii/metabolismo , Azotobacter vinelandii/enzimologia , Nitrogênio/química , Nitrogênio/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/química
7.
J Biol Chem ; 296: 100492, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33662397

RESUMO

Thiol dioxygenases are a subset of nonheme iron oxygenases that catalyze the formation of sulfinic acids from sulfhydryl-containing substrates and dioxygen. Among this class, cysteine dioxygenases (CDOs) and 3-mercaptopropionic acid dioxygenases (3MDOs) are the best characterized, and the mode of substrate binding for CDOs is well understood. However, the manner in which 3-mercaptopropionic acid (3MPA) coordinates to the nonheme iron site in 3MDO remains a matter of debate. A model for bidentate 3MPA coordination at the 3MDO Fe-site has been proposed on the basis of computational docking, whereas steady-state kinetics and EPR spectroscopic measurements suggest a thiolate-only coordination of the substrate. To address this gap in knowledge, we determined the structure of Azobacter vinelandii 3MDO (Av3MDO) in complex with the substrate analog and competitive inhibitor, 3-hydroxypropionic acid (3HPA). The structure together with DFT computational modeling demonstrates that 3HPA and 3MPA associate with iron as chelate complexes with the substrate-carboxylate group forming an additional interaction with Arg168 and the thiol bound at the same position as in CDO. A chloride ligand was bound to iron in the coordination site assigned as the O2-binding site. Supporting HYSCORE spectroscopic experiments were performed on the (3MPA/NO)-bound Av3MDO iron nitrosyl (S = 3/2) site. In combination with spectroscopic simulations and optimized DFT models, this work provides an experimentally verified model of the Av3MDO enzyme-substrate complex, effectively resolving a debate in the literature regarding the preferred substrate-binding denticity. These results elegantly explain the observed 3MDO substrate specificity, but leave unanswered questions regarding the mechanism of substrate-gated reactivity with dioxygen.


Assuntos
Ácido 3-Mercaptopropiônico/metabolismo , Azotobacter vinelandii/enzimologia , Dioxigenases/química , Dioxigenases/metabolismo , Ferro/química , Ferro/metabolismo , Ácido 3-Mercaptopropiônico/química , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X/métodos , Cinética , Modelos Moleculares , Especificidade por Substrato
8.
PLoS Comput Biol ; 17(3): e1008719, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33661889

RESUMO

The enzyme nitrogenase reduces dinitrogen to ammonia utilizing electrons, protons, and energy obtained from the hydrolysis of ATP. Mo-dependent nitrogenase is a symmetric dimer, with each half comprising an ATP-dependent reductase, termed the Fe Protein, and a catalytic protein, known as the MoFe protein, which hosts the electron transfer P-cluster and the active-site metal cofactor (FeMo-co). A series of synchronized events for the electron transfer have been characterized experimentally, in which electron delivery is coupled to nucleotide hydrolysis and regulated by an intricate allosteric network. We report a graph theory analysis of the mechanical coupling in the nitrogenase complex as a key step to understanding the dynamics of allosteric regulation of nitrogen reduction. This analysis shows that regions near the active sites undergo large-scale, large-amplitude correlated motions that enable communications within each half and between the two halves of the complex. Computational predictions of mechanically regions were validated against an analysis of the solution phase dynamics of the nitrogenase complex via hydrogen-deuterium exchange. These regions include the P-loops and the switch regions in the Fe proteins, the loop containing the residue ß-188Ser adjacent to the P-cluster in the MoFe protein, and the residues near the protein-protein interface. In particular, it is found that: (i) within each Fe protein, the switch regions I and II are coupled to the [4Fe-4S] cluster; (ii) within each half of the complex, the switch regions I and II are coupled to the loop containing ß-188Ser; (iii) between the two halves of the complex, the regions near the nucleotide binding pockets of the two Fe proteins (in particular the P-loops, located over 130 Å apart) are also mechanically coupled. Notably, we found that residues next to the P-cluster (in particular the loop containing ß-188Ser) are important for communication between the two halves.


Assuntos
Molibdoferredoxina/química , Molibdoferredoxina/metabolismo , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Azotobacter vinelandii/enzimologia , Sítios de Ligação , Medição da Troca de Deutério , Transporte de Elétrons , Modelos Moleculares , Ligação Proteica
9.
Proc Natl Acad Sci U S A ; 116(50): 25078-25086, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31767756

RESUMO

The radical S-adenosylmethionine (SAM) enzyme NifB occupies a central and essential position in nitrogenase biogenesis. NifB catalyzes the formation of an [8Fe-9S-C] cluster, called NifB-co, which constitutes the core of the active-site cofactors for all 3 nitrogenase types. Here, we produce functional NifB in aerobically cultured Saccharomyces cerevisiae Combinatorial pathway design was employed to construct 62 strains in which transcription units driving different expression levels of mitochondria-targeted nif genes (nifUSXB and fdxN) were integrated into the chromosome. Two combinatorial libraries totaling 0.7 Mb were constructed: An expression library of 6 partial clusters, including nifUSX and fdxN, and a library consisting of 28 different nifB genes mined from the Structure-Function Linkage Database and expressed at different levels according to a factorial design. We show that coexpression in yeast of the nitrogenase maturation proteins NifU, NifS, and FdxN from Azotobacter vinelandii with NifB from the archaea Methanocaldococcus infernus or Methanothermobacter thermautotrophicus yields NifB proteins equipped with [Fe-S] clusters that, as purified, support in vitro formation of NifB-co. Proof of in vivo NifB-co formation was additionally obtained. NifX as purified from aerobically cultured S. cerevisiae coexpressing M. thermautotrophicus NifB with A. vinelandii NifU, NifS, and FdxN, and engineered yeast SAM synthase supported FeMo-co synthesis, indicative of NifX carrying in vivo-formed NifB-co. This study defines the minimal genetic determinants for the formation of the key precursor in the nitrogenase cofactor biosynthetic pathway in a eukaryotic organism.


Assuntos
Proteínas de Bactérias/metabolismo , Compostos de Ferro/metabolismo , Saccharomyces cerevisiae/metabolismo , Azotobacter vinelandii/enzimologia , Azotobacter vinelandii/genética , Proteínas de Bactérias/genética , Redes e Vias Metabólicas , Methanocaldococcus , Mitocôndrias/metabolismo , Fixação de Nitrogênio/fisiologia , Nitrogenase/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Biologia Sintética
10.
J Am Chem Soc ; 143(24): 9183-9190, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34110795

RESUMO

Mo-dependent nitrogenase is a major contributor to global biological N2 reduction, which sustains life on Earth. Its multi-metallic active-site FeMo-cofactor (Fe7MoS9C-homocitrate) contains a carbide (C4-) centered within a trigonal prismatic CFe6 core resembling the structural motif of the iron carbide, cementite. The role of the carbide in FeMo-cofactor binding and activation of substrates and inhibitors is unknown. To explore this role, the carbide has been in effect selectively enriched with 13C, which enables its detailed examination by ENDOR/ESEEM spectroscopies. 13C-carbide ENDOR of the S = 3/2 resting state (E0) is remarkable, with an extremely small isotropic hyperfine coupling constant, Ca = +0.86 MHz. Turnover under high CO partial pressure generates the S = 1/2 hi-CO state, with two CO molecules bound to FeMo-cofactor. This conversion surprisingly leaves the small magnitude of the 13C carbide isotropic hyperfine-coupling constant essentially unchanged, Ca = -1.30 MHz. This indicates that both the E0 and hi-CO states exhibit an exchange-coupling scheme with nearly cancelling contributions to Ca from three spin-up and three spin-down carbide-bound Fe ions. In contrast, the anisotropic hyperfine coupling constant undergoes a symmetry change upon conversion of E0 to hi-CO that may be associated with bonding and coordination changes at Fe ions. In combination with the negligible difference between CFe6 core structures of E0 and hi-CO, these results suggest that in CO-inhibited hi-CO the dominant role of the FeMo-cofactor carbide is to maintain the core structure, rather than to facilitate inhibitor binding through changes in Fe-carbide covalency or stretching/breaking of carbide-Fe bonds.


Assuntos
Molibdoferredoxina/metabolismo , Nitrogenase/metabolismo , Azotobacter vinelandii/enzimologia , Isótopos de Carbono , Domínio Catalítico , Espectroscopia de Ressonância de Spin Eletrônica , Marcação por Isótopo , Conformação Molecular , Molibdoferredoxina/química , Nitrogenase/química , Nitrogenase/isolamento & purificação
11.
Chembiochem ; 22(1): 151-155, 2021 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-32918851

RESUMO

Nitrogenase converts N2 to NH3 , and CO to hydrocarbons, at its cofactor site. Herein, we report a biochemical and spectroscopic characterization of a Mo-nitrogenase variant expressed in an Azotobacter vinelandii strain containing a deletion of nifV, the gene encoding the homocitrate synthase. Designated NifDKCit , the catalytic component of this Mo-nitrogenase variant contains a citrate-substituted cofactor analogue. Activity analysis of NifDKCit reveals a shift of CO reduction from H2 evolution toward hydrocarbon formation and an opposite shift of N2 reduction from NH3 formation toward H2 evolution. Consistent with a shift in the Mo K-edge energy of NifDKCit relative to that of its wild-type counterpart, EPR analysis demonstrates a broadening of the line-shape and a decrease in the intensity of the cofactor-originated S=3/2 signal, suggesting a change in the spin properties of the cofactor upon citrate substitution. These observations point to a crucial role of homocitrate in substrate reduction by nitrogenase and the possibility to tune product profiles of nitrogenase reactions via organic ligand substitution.


Assuntos
Ácido Cítrico/metabolismo , Metaloproteínas/metabolismo , Molibdênio/metabolismo , Nitrogenase/metabolismo , Azotobacter vinelandii/enzimologia , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Ácido Cítrico/química , Espectroscopia de Ressonância de Spin Eletrônica , Hidrogênio/química , Hidrogênio/metabolismo , Metaloproteínas/química , Metaloproteínas/genética , Molibdênio/química , Nitrogenase/química , Nitrogenase/genética
12.
J Bacteriol ; 202(24)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32989088

RESUMO

Azotobacter vinelandii produces the linear exopolysaccharide alginate, a compound of significant biotechnological importance. The biosynthesis of alginate in A. vinelandii and Pseudomonas aeruginosa has several similarities but is regulated somewhat differently in the two microbes. Here, we show that the second messenger cyclic dimeric GMP (c-di-GMP) regulates the production and the molecular mass of alginate in A. vinelandii The hybrid protein MucG, containing conserved GGDEF and EAL domains and N-terminal HAMP and PAS domains, behaved as a c-di-GMP phosphodiesterase (PDE). This activity was found to negatively affect the amount and molecular mass of the polysaccharide formed. On the other hand, among the diguanylate cyclases (DGCs) present in A. vinelandii, AvGReg, a globin-coupled sensor (GCS) DGC that directly binds to oxygen, was identified as the main c-di-GMP-synthesizing contributor to alginate production. Overproduction of AvGReg in the parental strain phenocopied a ΔmucG strain with regard to alginate production and the molecular mass of the polymer. MucG was previously shown to prevent the synthesis of high-molecular-mass alginates in response to reduced oxygen transfer rates (OTRs). In this work, we show that cultures exposed to reduced OTRs accumulated higher levels of c-di-GMP; this finding strongly suggests that at least one of the molecular mechanisms involved in modulation of alginate production and molecular mass by oxygen depends on a c-di-GMP signaling module that includes the PAS domain-containing PDE MucG and the GCS DGC AvGReg.IMPORTANCE c-di-GMP has been widely recognized for its essential role in the production of exopolysaccharides in bacteria, such as alginate produced by Pseudomonas and Azotobacter spp. This study reveals that the levels of c-di-GMP also affect the physical properties of alginate, favoring the production of high-molecular-mass alginates in response to lower OTRs. This finding opens up new alternatives for the design of tailor-made alginates for biotechnological applications.


Assuntos
Alginatos/metabolismo , Azotobacter vinelandii/metabolismo , GMP Cíclico/análogos & derivados , Polissacarídeos Bacterianos/biossíntese , Alginatos/química , Azotobacter vinelandii/enzimologia , Azotobacter vinelandii/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , GMP Cíclico/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Peso Molecular , Oxigênio/metabolismo , Diester Fosfórico Hidrolases/genética , Diester Fosfórico Hidrolases/metabolismo , Fósforo-Oxigênio Liases/genética , Fósforo-Oxigênio Liases/metabolismo , Polissacarídeos Bacterianos/química
13.
J Bacteriol ; 202(24)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-32989089

RESUMO

The genus Azotobacter, belonging to the Pseudomonadaceae family, is characterized by the formation of cysts, which are metabolically dormant cells produced under adverse conditions and able to resist desiccation. Although this developmental process has served as a model for the study of cell differentiation in Gram-negative bacteria, the molecular basis of its regulation is still poorly understood. Here, we report that the ubiquitous second messenger cyclic dimeric GMP (c-di-GMP) is critical for the formation of cysts in Azotobacter vinelandii Upon encystment induction, the levels of c-di-GMP increased, reaching a peak within the first 6 h. In the absence of the diguanylate cyclase MucR, however, the levels of this second messenger remained low throughout the developmental process. A. vinelandii cysts are surrounded by two alginate layers with variable proportions of guluronic residues, which are introduced into the final alginate chain by extracellular mannuronic C-5 epimerases of the AlgE1 to AlgE7 family. Unlike in Pseudomonas aeruginosa, MucR was not required for alginate polymerization in A. vinelandii Conversely, MucR was necessary for the expression of extracellular alginate C-5 epimerases; therefore, the MucR-deficient strain produced cyst-like structures devoid of the alginate capsule and unable to resist desiccation. Expression of mucR was partially dependent on the response regulator AlgR, which binds to two sites in the mucR promoter, enhancing mucR transcription. Together, these results indicate that the developmental process of A. vinelandii is controlled through a signaling module that involves activation by the response regulator AlgR and c-di-GMP accumulation that depends on MucR.IMPORTANCEA. vinelandii has served as an experimental model for the study of the differentiation processes to form metabolically dormant cells in Gram-negative bacteria. This work identifies c-di-GMP as a critical regulator for the production of alginates with specific contents of guluronic residues that are able to structure the rigid laminated layers of the cyst envelope. Although allosteric activation of the alginate polymerase complex Alg8-Alg44 by c-di-GMP has long been recognized, our results show a previously unidentified role during the polymer modification step, controlling the expression of extracellular alginate epimerases. Our results also highlight the importance of c-di-GMP in the control of the physical properties of alginate, which ultimately determine the desiccation resistance of the differentiated cell.


Assuntos
Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Carboidratos Epimerases/metabolismo , GMP Cíclico/análogos & derivados , Alginatos/metabolismo , Azotobacter vinelandii/genética , Azotobacter vinelandii/crescimento & desenvolvimento , Azotobacter vinelandii/metabolismo , Proteínas de Bactérias/genética , Carboidratos Epimerases/genética , GMP Cíclico/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Fósforo-Oxigênio Liases/genética , Fósforo-Oxigênio Liases/metabolismo , Pseudomonas aeruginosa/enzimologia , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/metabolismo
14.
J Biol Chem ; 294(24): 9367-9376, 2019 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-31043481

RESUMO

The biological route for nitrogen gas entering the biosphere is reduction to ammonia by the nitrogenase enzyme, which is inactivated by oxygen. Three types of nitrogenase exist, the least-studied of which is the iron-only nitrogenase. The Anf3 protein in the bacterium Rhodobacter capsulatus is essential for diazotrophic (i.e. nitrogen-fixing) growth with the iron-only nitrogenase, but its enzymatic activity and function are unknown. Here, we biochemically and structurally characterize Anf3 from the model diazotrophic bacterium Azotobacter vinelandii Determining the Anf3 crystal structure to atomic resolution, we observed that it is a dimeric flavocytochrome with an unusually close interaction between the heme and the FAD cofactors. Measuring the reduction potentials by spectroelectrochemical redox titration, we observed values of -420 ± 10 and -330 ± 10 mV for the two FAD potentials and -340 ± 1 mV for the heme. We further show that Anf3 accepts electrons from spinach ferredoxin and that Anf3 consumes oxygen without generating superoxide or hydrogen peroxide. We predict that Anf3 protects the iron-only nitrogenase from oxygen inactivation by functioning as an oxidase in respiratory protection, with flavodoxin or ferredoxin as the physiological electron donors.


Assuntos
Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Ferro/metabolismo , Nitrogênio/metabolismo , Oxirredutases/metabolismo , Oxigênio/metabolismo , Proteínas de Bactérias/química , Cristalografia por Raios X , Fixação de Nitrogênio , Oxirredução , Oxirredutases/química , Conformação Proteica
15.
J Am Chem Soc ; 142(52): 21679-21690, 2020 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-33326225

RESUMO

Nitrogen fixation by nitrogenase begins with the accumulation of four reducing equivalents at the active-site FeMo-cofactor (FeMo-co), generating a state (denoted E4(4H)) with two [Fe-H-Fe] bridging hydrides. Recently, photolytic reductive elimination (re) of the E4(4H) hydrides showed that enzymatic re of E4(4H) hydride yields an H2-bound complex (E4(H2,2H)), in a process corresponding to a formal 2-electron reduction of the metal-ion core of FeMo-co. The resulting electron-density redistribution from Fe-H bonds to the metal ions themselves enables N2 to bind with concomitant H2 release, a process illuminated here by QM/MM molecular dynamics simulations. What is the nature of this redistribution? Although E4(H2,2H) has not been trapped, cryogenic photolysis of E4(4H) provides a means to address this question. Photolysis of E4(4H) causes hydride-re with release of H2, generating doubly reduced FeMo-co (denoted E4(2H)*), the extreme limit of the electron-density redistribution upon formation of E4(H2,2H). Here we examine the doubly reduced FeMo-co core of the E4(2H)* limiting-state by 1H, 57Fe, and 95Mo ENDOR to illuminate the partial electron-density redistribution upon E4(H2,2H) formation during catalysis, complementing these results with corresponding DFT computations. Inferences from the E4(2H)* ENDOR results as extended by DFT computations include (i) the Mo-site participates negligibly, and overall it is unlikely that Mo changes valency throughout the catalytic cycle; and (ii) two distinctive E4(4H) 57Fe signals are suggested as associated with structurally identified "anchors" of one bridging hydride, two others with identified anchors of the second, with NBO-analysis further identifying one anchor of each hydride as a major recipient of electrons released upon breaking Fe-H bonds.


Assuntos
Hidrogênio/química , Molibdoferredoxina/química , Nitrogenase/química , Animais , Azotobacter vinelandii/enzimologia , Domínio Catalítico , Transporte de Elétrons
16.
J Am Chem Soc ; 142(33): 14324-14330, 2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32787260

RESUMO

Coupling the nitrogenase MoFe protein to light-harvesting semiconductor nanomaterials replaces the natural electron transfer complex of Fe protein and ATP and provides low-potential photoexcited electrons for photocatalytic N2 reduction. A central question is how direct photochemical electron delivery from nanocrystals to MoFe protein is able to support the multielectron ammonia production reaction. In this study, low photon flux conditions were used to identify the initial reaction intermediates of CdS quantum dot (QD):MoFe protein nitrogenase complexes under photochemical activation using EPR. Illumination of CdS QD:MoFe protein complexes led to redox changes in the MoFe protein active site FeMo-co observed as the gradual decline in the E0 resting state intensity that was accompanied by an increase in the intensity of a new "geff = 4.5" EPR signal. The magnetic properties of the geff = 4.5 signal support assignment as a reduced S = 3/2 state, and reaction modeling was used to define it as a two-electron-reduced "E2" intermediate. Use of a MoFe protein variant, ß-188Cys, which poises the P cluster in the oxidized P+ state, demonstrated that the P cluster can function as a site of photoexcited electron delivery from CdS to MoFe protein. Overall, the results establish the initial steps for how photoexcited CdS delivers electrons into the MoFe protein during reduction of N2 to ammonia and the role of electron flux in the photochemical reaction cycle.


Assuntos
Compostos de Cádmio/metabolismo , Molibdoferredoxina/metabolismo , Pontos Quânticos/metabolismo , Sulfetos/metabolismo , Azotobacter vinelandii/enzimologia , Compostos de Cádmio/química , Transporte de Elétrons , Molibdoferredoxina/química , Oxirredução , Processos Fotoquímicos , Pontos Quânticos/química , Sulfetos/química
17.
Chembiochem ; 21(12): 1742-1748, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-31747483

RESUMO

Nitrogenases catalyze the ambient reduction of N2 and CO at its cofactor site. Herein we present a biochemical and spectroscopic characterization of an Azotobacter vinelandii V nitrogenase variant expressing a citrate-substituted cofactor. Designated VnfDGKCit , the catalytic component of this V nitrogenase variant has an αß2 (δ) subunit composition and carries an 8Fe P* cluster and a citrate-substituted V cluster analogue in the αß dimer, as well as a 4Fe cluster in the "orphaned" ß-subunit. Interestingly, when normalized based on the amount of cofactor, VnfDGKCit shows a shift of N2 reduction from H2 evolution toward NH3 formation and an opposite shift of CO reduction from hydrocarbon formation toward H2 evolution. These observations point to a role of the organic ligand in proton delivery during catalysis and imply the use of different reaction sites/mechanisms by nitrogenase for different substrate reductions. Moreover, the increased NH3 /H2 ratio upon citrate substitution suggests the possibility to modify the organic ligand for improved ammonia synthesis in the future.


Assuntos
Azotobacter vinelandii/enzimologia , Proteínas de Bactérias/metabolismo , Ácido Cítrico/metabolismo , Nitrogenase/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Espectroscopia de Ressonância de Spin Eletrônica , Nitrogenase/química , Nitrogenase/genética , Conformação Proteica
18.
Chembiochem ; 21(12): 1749-1759, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32202031

RESUMO

Biological nitrogen fixation is an inherent trait exclusive to a select number of prokaryotes. Although molybdenum nitrogenase is the dominant catalyst for dinitrogen reduction, some diazotrophs also contain one or two additional types of nitrogenase that use alternative metal content as the active-site cofactor. The occurrence of alternative nitrogenases has not been well studied due to the discriminatory expression of the molybdenum nitrogenase and lack of comprehensive genomic data. This study reports on the genomic analysis of 87 unique species containing alternative nitrogenase sequences. The distribution of nitrogen-fixing genes within these species from distinct taxonomic groups shows the presence of the minimum gene set required for nitrogen fixation, including catalytic and biosynthetic enzymes of the Mo-dependent system (NifHDKENB) and the varying occurrence of additional Nif-dedicated components. These include NifS and NifU, found primarily in aerobic species, thus suggesting that these genes are necessary to accommodate the high demand for Fe-S clusters during aerobic nitrogen fixation.


Assuntos
Azotobacter vinelandii/enzimologia , Fixação de Nitrogênio/genética , Nitrogenase/metabolismo , Células Procarióticas/metabolismo
19.
Acc Chem Res ; 52(5): 1168-1176, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30977994

RESUMO

FeS proteins are metalloproteins prevalent in the metabolic pathways of most organisms, playing key roles in a wide range of essential cellular processes. A member of this protein family, the Fe protein of nitrogenase, is a homodimer that contains a redox-active [Fe4S4] cluster at the subunit interface and an ATP-binding site within each subunit. During catalysis, the Fe protein serves as the obligate electron donor for its catalytic partner, transferring electrons concomitant with ATP hydrolysis to the cofactor site of the catalytic component to enable substrate reduction. The effectiveness of Fe protein in electron transfer is reflected by the unique reactivity of nitrogenase toward small-molecule substrates. Most notably, nitrogenase is capable of catalyzing the ambient reduction of N2 and CO into NH4+ and hydrocarbons, respectively, in reactions that parallel the important industrial Haber-Bosch and Fischer-Tropsch processes. Other than participating in nitrogenase catalysis, the Fe protein also functions as an essential factor in nitrogenase assembly, which again highlights its capacity as an effective, ATP-dependent electron donor. Recently, the Fe protein of a soil bacterium, Azotobacter vinelandii, was shown to act as a reductase on its own and catalyze the ambient conversion of CO2 to CO at its [Fe4S4] cluster either under in vitro conditions when a strong reductant is supplied or under in vivo conditions through the action of an unknown electron donor(s) in the cell. Subsequently, the Fe protein of a mesophilic methanogenic organism, Methanosarcina acetivorans, was shown to catalyze the in vitro reduction of CO2 and CO into hydrocarbons under ambient conditions, illustrating an impact of protein scaffold on the redox properties of the [Fe4S4] cluster and the reactivity of the cluster toward C1 substrates. This reactivity was further traced to the [Fe4S4] cluster itself, as a synthetic [Fe4S4] compound was shown to catalyze the reduction of CO2 and CO to hydrocarbons in solutions in the presence of a strong reductant. Together, these observations pointed to an inherent ability of the [Fe4S4] clusters and, possibly, the FeS clusters in general to catalyze C1-substrate reduction. Theoretical calculations have led to the proposal of a plausible reaction pathway that involves the formation of hydrocarbons via aldehyde-like intermediates, providing an important framework for further mechanistic investigations of FeS-based activation and reduction of C1 substrates. In this Account, we summarize the recent work leading to the discovery of C1-substrate reduction by protein-bound and free [Fe4S4] clusters as well as the current mechanistic understanding of this FeS-based reactivity. In addition, we briefly discuss the evolutionary implications of this discovery and potential applications that could be developed to enable FeS-based strategies for the ambient recycling of unwanted C1 waste into useful chemical commodities.


Assuntos
Proteínas Ferro-Enxofre/química , Oxirredutases/química , Azotobacter vinelandii/enzimologia , Dióxido de Carbono/química , Monóxido de Carbono/química , Evolução Molecular , Hidrocarbonetos/síntese química , Methanosarcina/enzimologia , Modelos Químicos , Oxirredução
20.
J Biol Inorg Chem ; 25(3): 521-540, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32266560

RESUMO

We have made a systematic combined quantum mechanical and molecular mechanical (QM/MM) investigation of possible structures of the N2 bound state of nitrogenase. We assume that N2 is immediately protonated to a N2H2 state, thereby avoiding the problem of determining the position of the protons in the cluster. We have systematically studied both end-on and side-on structures, as well as both HNNH and NNH2 states. Our results indicate that the binding of N2H2 is determined more by interactions and steric clashes with the surrounding protein than by the intrinsic preferences of the ligand and the cluster. The best binding mode with both the TPSS and B3LYP density-functional theory methods has trans-HNNH terminally bound to Fe2. It is stabilised by stacking of the substrate with His-195 and Ser-278. However, several other structures come rather close in energy (within 3-35 kJ/mol) at least in some calculations: The corresponding cis-HNNH structure terminally bound to Fe2 is second best with B3LYP. A structure with HNNH2 terminally bound to Fe6 is second most stable with TPSS (where the third proton is transferred to the substrate from the homocitrate ligand). Structures with trans-HNNH, bound to Fe4 or Fe6, or cis-HNNH bound to Fe6 are also rather stable. Finally, with the TPSS functional, a structure with cis-HNNH side-on binding to the Fe3-Fe4-Fe5-Fe7 face of the cluster is also rather low in energy, but all side-on structures are strongly disfavoured by the B3LYP method.


Assuntos
Teoria da Densidade Funcional , Molibdoferredoxina/química , Compostos de Nitrogênio/química , Nitrogenase/química , Azotobacter vinelandii/enzimologia , Sítios de Ligação , Molibdoferredoxina/metabolismo , Compostos de Nitrogênio/metabolismo , Nitrogenase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA