Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.011
Filtrar
1.
Biochemistry ; 63(12): 1588-1598, 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38817151

RESUMO

Thioredoxin reductases (TrxR) activate thioredoxins (Trx) that regulate the activity of diverse target proteins essential to prokaryotic and eukaryotic life. However, very little is understood of TrxR/Trx systems and redox control in methanogenic microbes from the domain Archaea (methanogens), for which genomes are abundant with annotations for ferredoxin:thioredoxin reductases [Fdx/thioredoxin reductase (FTR)] from group 4 of the widespread FTR-like family. Only two from the FTR-like family are characterized: the plant-type FTR from group 1 and FDR from group 6. Herein, the group 4 archetype (AFTR) from Methanosarcina acetivorans was characterized to advance understanding of the family and TrxR/Trx systems in methanogens. The modeled structure of AFTR, together with EPR and Mössbauer spectroscopies, supports a catalytic mechanism similar to plant-type FTR and FDR, albeit with important exceptions. EPR spectroscopy of reduced AFTR identified a transient [4Fe-4S]1+ cluster exhibiting a mixture of S = 7/2 and typical S = 1/2 signals, although rare for proteins containing [4Fe-4S] clusters, it is most likely the on-pathway intermediate in the disulfide reduction. Furthermore, an active site histidine equivalent to residues essential for the activity of plant-type FTR and FDR was found dispensable for AFTR. Finally, a unique thioredoxin system was reconstituted from AFTR, ferredoxin, and Trx2 from M. acetivorans, for which specialized target proteins were identified that are essential for growth and other diverse metabolisms.


Assuntos
Proteínas Ferro-Enxofre , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/genética , Methanosarcina/enzimologia , Methanosarcina/genética , Ferredoxinas/metabolismo , Ferredoxinas/química , Ferredoxinas/genética , Oxirredução , Modelos Moleculares , Tiorredoxinas/metabolismo , Tiorredoxinas/química , Tiorredoxinas/genética , Oxirredutases/metabolismo , Oxirredutases/química , Oxirredutases/genética , Tiorredoxina Dissulfeto Redutase/metabolismo , Tiorredoxina Dissulfeto Redutase/química , Tiorredoxina Dissulfeto Redutase/genética , Proteínas Arqueais/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/genética , Espectroscopia de Ressonância de Spin Eletrônica
2.
FEBS J ; 291(11): 2449-2460, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38468562

RESUMO

In the hydrogenotrophic methanogenic pathway, formylmethanofuran dehydrogenase (Fmd) catalyzes the formation of formylmethanofuran through reducing CO2. Heterodisulfide reductase (Hdr) provides two low potential electrons for the Fmd reaction using a flavin-based electron-bifurcating mechanism. [NiFe]-hydrogenase (Mvh) or formate dehydrogenase (Fdh) complexes with Hdr and provides electrons to Hdr from H2 and formate, or the reduced form of F420, respectively. Recently, an Fdh-Hdr complex was purified as a 3-MDa megacomplex that contained Fmd, and its three-dimensional structure was elucidated by cryo-electron microscopy. In contrast, the Mvh-Hdr complex has been characterized only as a complex without Fmd. Here, we report the isolation and characterization of a 1-MDa Mvh-Hdr-Fmd megacomplex from Methanothermobacter marburgensis. After anion-exchange and hydrophobic chromatography was performed, the proteins with Hdr activity eluted in the 1- and 0.5-MDa fractions during size exclusion chromatography. Considering the apparent molecular mass and the protein profile in the fractions, the 1-MDa megacomplex was determined to be a dimeric Mvh-Hdr-Fmd complex. The megacomplex fraction contained a polyferredoxin subunit MvhB, which contains 12 [4Fe-4S]-clusters. MvhB polyferredoxin has never been identified in the previously purified Mvh-Hdr and Fmd preparations, suggesting that MvhB polyferredoxin is stabilized by the binding between Mvh-Hdr and Fmd in the Mvh-Hdr-Fmd complex. The purified Mvh-Hdr-Fmd megacomplex catalyzed electron-bifurcating reduction of [13C]-CO2 to form [13C]-formylmethanofuran in the absence of extrinsic ferredoxin. These results demonstrated that the subunits in the Mvh-Hdr-Fmd megacomplex are electronically connected for the reduction of CO2, which likely involves MvhB polyferredoxin as an electron relay.


Assuntos
Dióxido de Carbono , Hidrogênio , Methanobacteriaceae , Methanobacteriaceae/metabolismo , Methanobacteriaceae/enzimologia , Hidrogênio/metabolismo , Hidrogênio/química , Dióxido de Carbono/metabolismo , Dióxido de Carbono/química , Oxirredutases/metabolismo , Oxirredutases/química , Ferredoxinas/metabolismo , Ferredoxinas/química , Oxirredução , Proteínas Arqueais/metabolismo , Proteínas Arqueais/química , Proteínas Arqueais/genética , Elétrons , Hidrogenase/metabolismo , Hidrogenase/química
3.
Proteins ; 92(6): 776-794, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38258321

RESUMO

Three-dimensional (3D) structure information, now available at the proteome scale, may facilitate the detection of remote evolutionary relationships in protein superfamilies. Here, we illustrate this with the identification of a novel family of protein domains related to the ferredoxin-like superfold, by combining (i) transitive sequence similarity searches, (ii) clustering approaches, and (iii) the use of AlphaFold2 3D structure models. Domains of this family were initially identified in relation with the intracellular biomineralization of calcium carbonates by Cyanobacteria. They are part of the large heavy-metal-associated (HMA) superfamily, departing from the latter by specific sequence and structural features. In particular, most of them share conserved basic amino acids  (hence their name CoBaHMA for Conserved Basic residues HMA), forming a positively charged surface, which is likely to interact with anionic partners. CoBaHMA domains are found in diverse modular organizations in bacteria, existing in the form of monodomain proteins or as part of larger proteins, some of which are membrane proteins involved in transport or lipid metabolism. This suggests that the CoBaHMA domains may exert a regulatory function, involving interactions with anionic lipids. This hypothesis might have a particular resonance in the context of the compartmentalization observed for cyanobacterial intracellular calcium carbonates.


Assuntos
Sequência de Aminoácidos , Proteínas de Bactérias , Metais Pesados , Modelos Moleculares , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Metais Pesados/química , Metais Pesados/metabolismo , Domínios Proteicos , Cianobactérias/metabolismo , Cianobactérias/química , Cianobactérias/genética , Ferredoxinas/química , Ferredoxinas/metabolismo , Dobramento de Proteína
4.
J Am Chem Soc ; 145(43): 23488-23502, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37867463

RESUMO

We identified a multisubstrate-bound state, hereby referred as a 3site state, in cytochrome P450cam via integrating molecular dynamics simulation with nuclear magnetic resonance (NMR) pseudocontact shift measurements. The 3site state is a result of simultaneous binding of three camphor molecules in three locations around P450cam: (a) in a well-established "catalytic" site near heme, (b) in a kink-separated "waiting" site along channel-1, and (c) in a previously reported "allosteric" site at E, F, G, and H helical junctions. These three spatially distinct binding modes in the 3site state mutually communicate with each other via homotropic allostery and act cooperatively to render P450cam functional. The 3site state shows a significantly superior fit with NMR pseudo contact shift (PCS) data with a Q-score of 0.045 than previously known bound states and consists of D251 free of salt-bridges with K178 and R186, rendering the enzyme functionally primed. To date, none of the reported cocomplex of P450cam with its redox partner putidaredoxin (pdx) has been able to match solution NMR data and controversial pdx-induced opening of P450cam's channel-1 remains a matter of recurrent discourse. In this regard, inclusion of pdx to the 3site state is able to perfectly fit the NMR PCS measurement with a Q-score of 0.08 and disfavors the pdx-induced opening of channel-1, reconciling previously unexplained remarkably fast hydroxylation kinetics with a koff of 10.2 s-1. Together, our findings hint that previous experimental observations may have inadvertently captured the 3site state as an in vitro solution state, instead of the catalytic state alone, and provided a distinct departure from the conventional understanding of cytochrome P450.


Assuntos
Cânfora 5-Mono-Oxigenase , Pseudomonas putida , Cânfora 5-Mono-Oxigenase/química , Ligação Proteica , Ferredoxinas/química , Oxirredução , Sistema Enzimático do Citocromo P-450/metabolismo , Simulação de Dinâmica Molecular
5.
Sci Rep ; 13(1): 10832, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37402767

RESUMO

[4Fe-4S] clusters are essential cofactors in many proteins involved in biological redox-active processes. Density functional theory (DFT) methods are widely used to study these clusters. Previous investigations have indicated that there exist two local minima for these clusters in proteins. We perform a detailed study of these minima in five proteins and two oxidation states, using combined quantum mechanical and molecular mechanical (QM/MM) methods. We show that one local minimum (L state) has longer Fe-Fe distances than the other (S state), and that the L state is more stable for all cases studied. We also show that some DFT methods may only obtain the L state, while others may obtain both states. Our work provides new insights into the structural diversity and stability of [4Fe-4S] clusters in proteins, and highlights the importance of reliable DFT methods and geometry optimization. We recommend r2SCAN for optimizing [4Fe-4S] clusters in proteins, which gives the most accurate structures for the five proteins studied.


Assuntos
Bactérias , Proteínas Ferro-Enxofre , Proteínas Ferro-Enxofre/química , Oxirredução , Bactérias/química , Ferredoxinas/química , Teoria da Densidade Funcional , Cristalografia
6.
Bioelectrochemistry ; 153: 108459, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37263168

RESUMO

In this study, photobioelectrodes based on a ferredoxin-modified photosystem I (PSI-Fd) from Thermosynechococcus vestitus have been prepared and characterized regarding the direct electron transfer between PSI-Fd and the electrode. The modified PSI with the covalently linked ferredoxin (Fd) on its stromal side has been immobilized on indium-tin-oxide (ITO) electrodes with a 3-dimensional inverse-opal structure. Compared to native PSI, a lower photocurrent and a lower onset potential of the cathodic photocurrent have been observed. This can be mainly attributed to a different adsorption behavior of the PSI-Fd-construct onto the 3D ITO. However, the overall behavior is rather similar to PSI. First experiments have been performed for applying this PSI-Fd photobioelectrode for enzyme-driven NADPH generation. By coupling the electrode system with ferredoxin-NADP+-reductase (FNR), first hints for the usage of photoelectrons for biosynthesis have been collected by verifying NADPH generation.


Assuntos
Ferredoxinas , Complexo de Proteína do Fotossistema I , Complexo de Proteína do Fotossistema I/química , Ferredoxinas/química , Ferredoxinas/metabolismo , NADP/metabolismo , Transporte de Elétrons , Ferredoxina-NADP Redutase/química , Ferredoxina-NADP Redutase/metabolismo , Eletrodos
7.
PeerJ ; 11: e15187, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37131988

RESUMO

Background: The spread of artemisinin (ART)-resistant Plasmodium falciparum threatens the control of malaria. Mutations in the propeller domains of P. falciparum Kelch13 (k13) are strongly associated with ART resistance. Ferredoxin (Fd), a component of the ferredoxin/NADP+ reductase (Fd/FNR) redox system, is essential for isoprenoid precursor synthesis in the plasmodial apicoplast, which is important for K13-dependent hemoglobin trafficking and ART activation. Therefore, Fd is an antimalarial drug target and fd mutations may modulate ART sensitivity. We hypothesized that loss of Fd/FNR function enhances the effect of k13 mutation on ART resistance. Methods: In this study, methoxyamino chalcone (C3), an antimalarial compound that has been reported to inhibit the interaction of recombinant Fd and FNR proteins, was used as a chemical inhibitor of the Fd/FNR redox system. We investigated the inhibitory effects of dihydroartemisinin (DHA), C3, and iron chelators including deferiprone (DFP), 1-(N-acetyl-6-aminohexyl)-3-hydroxy-2-methylpyridin-4-one (CM1) and deferiprone-resveratrol hybrid (DFP-RVT) against wild-type (WT), k13 mutant, fd mutant, and k13 fd double mutant P. falciparum parasites. Furthermore, we investigated the pharmacological interaction of C3 with DHA, in which the iron chelators were used as reference ART antagonists. Results: C3 showed antimalarial potency similar to that of the iron chelators. As expected, combining DHA with C3 or iron chelators exhibited a moderately antagonistic effect. No differences were observed among the mutant parasites with respect to their sensitivity to C3, iron chelators, or the interactions of these compounds with DHA. Discussion: The data suggest that inhibitors of the Fd/FNR redox system should be avoided as ART partner drugs in ART combination therapy for treating malaria.


Assuntos
Antimaláricos , Chalcona , Malária Falciparum , Humanos , Antimaláricos/farmacologia , Plasmodium falciparum/genética , Ferredoxinas/química , Chalcona/farmacologia , Deferiprona/farmacologia , Malária Falciparum/tratamento farmacológico , Ferredoxina-NADP Redutase , Quelantes de Ferro/farmacologia
8.
FEBS J ; 290(16): 4107-4125, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37074156

RESUMO

A major electron carrier involved in energy and carbon metabolism in the acetogenic model organism Thermoanaerobacter kivui is ferredoxin, an iron-sulfur-containing, electron-transferring protein. Here, we show that the genome of T. kivui encodes four putative ferredoxin-like proteins (TKV_c09620, TKV_c16450, TKV_c10420 and TKV_c19530). All four genes were cloned, a His-tag encoding sequence was added and the proteins were produced from a plasmid in T. kivui. The purified proteins had an absorption peak at 430 nm typical for ferredoxins. The determined iron-sulfur content is consistent with the presence of two predicted [4Fe4S] clusters in TKV_c09620 and TKV_c19530 or one predicted [4Fe4S] cluster in TKV_c16450 and TKV_c10420 respectively. The reduction potential (Em ) for TKV_c09620, TKV_c16450, TKV_c10420 and TKV_c19530 was determined to be -386 ± 4 mV, -386 ± 2 mV, -559 ± 10 mV and -557 ± 3 mV, respectively. TKV_c09620 and TKV_c16450 served as electron carriers for different oxidoreductases from T. kivui. Deletion of the ferredoxin genes led to only a slight reduction of growth on pyruvate or autotrophically on H2 + CO2 . Transcriptional analysis revealed that TKV_c09620 was upregulated in a ΔTKV_c16450 mutant and vice versa TKV_c16450 in a ΔTKV_c09620 mutant, indicating that TKV_c09620 and TKV_c16450 can replace each other. In sum, our data are consistent with the hypothesis that TKV_c09620 and TKV_c16450 are ferredoxins involved in autotrophic and heterotrophic metabolism of T. kivui.


Assuntos
Ferredoxinas , Thermoanaerobacter , Thermoanaerobacter/química , Thermoanaerobacter/genética , Thermoanaerobacter/metabolismo , Ferredoxinas/química , Ferredoxinas/genética , Ferredoxinas/metabolismo , Genoma Bacteriano/genética , Deleção de Genes , Regulação Bacteriana da Expressão Gênica , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Espectroscopia Fotoeletrônica
9.
Mol Cell Proteomics ; 22(4): 100521, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36858286

RESUMO

Lysine methylation is a conserved and dynamic regulatory posttranslational modification performed by lysine methyltransferases (KMTs). KMTs catalyze the transfer of mono-, di-, or tri-methyl groups to substrate proteins and play a critical regulatory role in all domains of life. To date, only one KMT has been identified in cyanobacteria. Here, we tested all of the predicted KMTs in the cyanobacterium Synechocystis sp. PCC 6803 (Synechocystis), and we biochemically characterized sll1526 that we termed cKMT1 (cyanobacterial lysine methyltransferase 1) and determined that it can catalyze lysine methylation both in vivo and in vitro. Loss of cKMT1 alters photosynthetic electron transfer in Synechocystis. We analyzed cKMT1-regulated methylation sites in Synechocystis using a timsTOF Pro instrument. We identified 305 class I lysine methylation sites within 232 proteins, and of these, 80 methylation sites in 58 proteins were hypomethylated in ΔcKMT1 cells. We further demonstrated that cKMT1 could methylate ferredoxin-NADP(+) oxidoreductase (FNR) and its potential sites of action on FNR were identified. Amino acid residues H118 and Y219 were identified as key residues in the putative active site of cKMT1 as indicated by structure simulation, site-directed mutagenesis, and KMT activity measurement. Using mutations that mimic the unmethylated forms of FNR, we demonstrated that the inability to methylate K139 residues results in a decrease in the redox activity of FNR and affects energy transfer in Synechocystis. Together, our study identified a new KMT in Synechocystis and elucidated a methylation-mediated molecular mechanism catalyzed by cKMT1 for the regulation of energy transfer in cyanobacteria.


Assuntos
Cianobactérias , Ferredoxinas , Synechocystis , Transferência de Energia , Ferredoxina-NADP Redutase/química , Ferredoxina-NADP Redutase/genética , Ferredoxina-NADP Redutase/metabolismo , Ferredoxinas/química , Ferredoxinas/metabolismo , Lisina , Metiltransferases/metabolismo , NADP/metabolismo , Synechocystis/metabolismo , Cianobactérias/metabolismo
10.
J Am Chem Soc ; 145(10): 5696-5709, 2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36811855

RESUMO

Electron bifurcation is a fundamental energy coupling mechanism widespread in microorganisms that thrive under anoxic conditions. These organisms employ hydrogen to reduce CO2, but the molecular mechanisms have remained enigmatic. The key enzyme responsible for powering these thermodynamically challenging reactions is the electron-bifurcating [FeFe]-hydrogenase HydABC that reduces low-potential ferredoxins (Fd) by oxidizing hydrogen gas (H2). By combining single-particle cryo-electron microscopy (cryoEM) under catalytic turnover conditions with site-directed mutagenesis experiments, functional studies, infrared spectroscopy, and molecular simulations, we show that HydABC from the acetogenic bacteria Acetobacterium woodii and Thermoanaerobacter kivui employ a single flavin mononucleotide (FMN) cofactor to establish electron transfer pathways to the NAD(P)+ and Fd reduction sites by a mechanism that is fundamentally different from classical flavin-based electron bifurcation enzymes. By modulation of the NAD(P)+ binding affinity via reduction of a nearby iron-sulfur cluster, HydABC switches between the exergonic NAD(P)+ reduction and endergonic Fd reduction modes. Our combined findings suggest that the conformational dynamics establish a redox-driven kinetic gate that prevents the backflow of the electrons from the Fd reduction branch toward the FMN site, providing a basis for understanding general mechanistic principles of electron-bifurcating hydrogenases.


Assuntos
Elétrons , Hidrogenase , Hidrogenase/química , NAD/metabolismo , Microscopia Crioeletrônica , Ferredoxinas/química , Oxirredução , Hidrogênio/química , Transporte de Elétrons
11.
J Am Chem Soc ; 145(4): 2075-2080, 2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36688844

RESUMO

Synthetic analogues of the three common types of Fe-S clusters found in biology─diamond-core [Fe2S2] clusters, open-cuboidal [Fe3S4] clusters, and cuboidal [Fe4S4] clusters─have been reported in each biologically relevant redox state with one exception: the open-cuboidal [Fe3S4]+ cluster. Here, we describe the synthesis and characterization of an open-cuboidal [Fe3S4] cluster in both biologically relevant redox states: [Fe3S4]+ and [Fe3S4]0. Like their biological counterparts, the oxidized cluster has a spin-canted, S = 1/2 ground state, and the reduced cluster has an S = 2 ground state. Structural analysis reveals that the [Fe3S4] core undergoes substantial contraction upon oxidation, in contrast to the minimal structural changes observed for the only [Fe3S4] protein for which high-resolution structures are available in both redox states (Azotobacter vinelandii ferredoxin I; Av FdI). This difference between the synthetic models and Av FdI is discussed in the context of electron transfer by [Fe3S4] proteins.


Assuntos
Proteínas Ferro-Enxofre , Ferro , Ferro/química , Oxirredução , Ferredoxinas/química , Sulfetos , Proteínas Ferro-Enxofre/química , Espectroscopia de Ressonância de Spin Eletrônica
12.
Elife ; 112022 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-36018003

RESUMO

Electron bifurcation is a fundamental energy conservation mechanism in nature in which two electrons from an intermediate-potential electron donor are split so that one is sent along a high-potential pathway to a high-potential acceptor and the other is sent along a low-potential pathway to a low-potential acceptor. This process allows endergonic reactions to be driven by exergonic ones and is an alternative, less recognized, mechanism of energy coupling to the well-known chemiosmotic principle. The electron-bifurcating [FeFe] hydrogenase from Thermotoga maritima (HydABC) requires both NADH and ferredoxin to reduce protons generating hydrogen. The mechanism of electron bifurcation in HydABC remains enigmatic in spite of intense research efforts over the last few years. Structural information may provide the basis for a better understanding of spectroscopic and functional information. Here, we present a 2.3 Å electron cryo-microscopy structure of HydABC. The structure shows a heterododecamer composed of two independent 'halves' each made of two strongly interacting HydABC heterotrimers connected via a [4Fe-4S] cluster. A central electron transfer pathway connects the active sites for NADH oxidation and for proton reduction. We identified two conformations of a flexible iron-sulfur cluster domain: a 'closed bridge' and an 'open bridge' conformation, where a Zn2+ site may act as a 'hinge' allowing domain movement. Based on these structural revelations, we propose a possible mechanism of electron bifurcation in HydABC where the flavin mononucleotide serves a dual role as both the electron bifurcation center and as the NAD+ reduction/NADH oxidation site.


Assuntos
Hidrogenase , Proteínas Ferro-Enxofre , Proteínas de Bactérias/metabolismo , Elétrons , Ferredoxinas/química , Ferredoxinas/metabolismo , Mononucleotídeo de Flavina/metabolismo , Hidrogênio/metabolismo , Hidrogenase/metabolismo , Ferro/metabolismo , Proteínas Ferro-Enxofre/metabolismo , NAD/metabolismo , Oxirredução , Prótons , Enxofre/metabolismo
13.
J Biol Inorg Chem ; 27(7): 631-640, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36038787

RESUMO

Ferredoxins are essential electron transferring proteins in organisms. Twelve plant-type ferredoxins in the green alga Chlamydomonas reinhardtii determine the fate of electrons, generated in multiple metabolic processes. The two hydrogenases HydA1 and HydA2 of. C. reinhardtii compete for electrons from the photosynthetic ferredoxin PetF, which is the first stromal mediator of the high-energy electrons derived from the absorption of light energy at the photosystems. While being involved in many chloroplast-located metabolic pathways, PetF shows the highest affinity for ferredoxin-NADP+ oxidoreductase (FNR), not for the hydrogenases. Aiming to identify other potential electron donors for the hydrogenases, we screened as yet uncharacterized ferredoxins Fdx7, 8, 10 and 11 for their capability to reduce the hydrogenases. Comparing the performance of the Fdx in presence and absence of competitor FNR, we show that Fdx7 has a higher affinity for HydA1 than for FNR. Additionally, we show that synthetic FeS-cluster-binding maquettes, which can be reduced by NADPH alone, can also be used to reduce the hydrogenases. Our findings pave the way for the creation of tailored electron donors to redirect electrons to enzymes of interest.


Assuntos
Ferredoxinas , Hidrogenase , Transporte de Elétrons , Elétrons , Ferredoxinas/química , Hidrogênio/metabolismo , Hidrogenase/química , NADP/metabolismo
14.
World J Microbiol Biotechnol ; 38(10): 178, 2022 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-35941298

RESUMO

Ferredoxin (Fd) is a small metalloprotein holding one or two Fe-S clusters in its inner shell. Like many other metalloproteins, Fd is redox active and involved in electron transfer during cellular metabolism. The electrons from reduced Fd are mostly used to regenerate NADPH under physiological conditions. Increasing number of attempts have been reported, however, where Fd delivers electrons to enable biosynthesis of valuable compounds. Various compounds ranging from H2 to vitamin D3 have been synthesized successfully using electrons mediated by Fd molecules. In this review, we provide an overview of the engineering studies utilizing Fd for biosynthesis of targeted molecules. The emphasis is on the role and activity of Fd as well as the methods used to improve the rate of electron transfer. Both microbial and electrochemical biosynthesis technologies are described and compared with respect to productivity and the compound being produced. In addition to the ferredoxins from the microbial organisms, artificially designed de novo types are described, highlighting the potential of the emerging computational methods used in metabolic and protein engineering. We believe that the recent advances in utilization of Fd for biosynthesis can result in breakthrough innovation across the biotechnology industry.


Assuntos
Ferredoxinas , Fotossíntese , Transporte de Elétrons , Elétrons , Ferredoxinas/química , Ferredoxinas/metabolismo , Oxirredução
15.
J Phys Chem B ; 126(16): 3059-3066, 2022 04 28.
Artigo em Inglês | MEDLINE | ID: mdl-35435680

RESUMO

Most low-potential Fe4S4 clusters exist in the conserved binding sequence CxxCxxC (CnCn+3Cn+6). Fe(II) and Fe(III) at the first (Cn) and third (Cn+6) cysteine ligand sites form a mixed-valence Fe2.5+···Fe2.5+ pair in the reduced Fe(II)3Fe(III) cluster. Here, we investigate the mechanism of how the conserved protein environment induces mixed-valence pair formation in the Fe4S4 clusters, FX, FA, and FB in photosystem I, using a quantum mechanical/molecular mechanical approach. Exchange coupling between Fe sites is predominantly determined by the shape of the Fe4S4 cluster, which is stabilized by the preorganized protein electrostatic environment. The backbone NH and CO groups in the conserved CxxCxxC and adjacent helix regions orient along the FeCn···FeC(n+6) axis, generating an electric field and stabilizing the FeCn(II)FeC(n+6)(III) state in FA and FB. The overlap of the d orbitals via -S- (superexchange) is observed for the single FeCn(II)···FeC(n+6)(III) pair, leading to the formation of the mixed-valence Fe2.5+···Fe2.5+ pair. In contrast, several superexchange Fe(II)···Fe(III) pairs are observed in FX due to the highly symmetric pair of the CDGPGRGGTC sequences. This is likely the origin of FX serving as an electron acceptor in the two electron transfer branches.


Assuntos
Ferredoxinas , Compostos Férricos , Espectroscopia de Ressonância de Spin Eletrônica , Ferredoxinas/química , Compostos Férricos/metabolismo , Compostos Ferrosos/metabolismo , Complexo de Proteína do Fotossistema I/metabolismo
16.
Acc Chem Res ; 55(3): 373-380, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34965086

RESUMO

This Account summarizes recent findings centered on the role that redox partner binding, allostery, and conformational dynamics plays in cytochrome P450 proton coupled electron transfer. P450s are one of Nature's largest enzyme families and it is not uncommon to find a P450 wherever substrate oxidation is required in the formation of essential molecules critical to the life of the organism or in xenobiotic detoxification. P450s can operate on a remarkably large range of substrates from the very small to the very large, yet the overall P450 three-dimensional structure is conserved. Given this conservation of structure, it is generally assumed that the basic catalytic mechanism is conserved. In nearly all P450s, the O2 O-O bond must be cleaved heterolytically enabling one oxygen atom, the distal oxygen, to depart as water and leave behind a heme iron-linked O atom as the powerful oxidant that is used to activate the nearby substrate. For this process to proceed efficiently, externally supplied electrons and protons are required. Two protons must be added to the departing O atom while an electron is transferred from a redox partner that typically contains either a Fe2S2 or FMN redox center. The paradigm P450 used to unravel the details of these mechanisms has been the bacterial CYP101A1 or P450cam. P450cam is specific for its own Fe2S2 redox partner, putidaredoxin or Pdx, and it has long been postulated that Pdx plays an effector/allosteric role by possibly switching P450cam to an active conformation. Crystal structures, spectroscopic data, and direct binding experiments of the P450cam-Pdx complex provide some answers. Pdx shifts the conformation of P450cam to a more open state, a transition that is postulated to trigger the proton relay network required for O2 activation. An essential part of this proton relay network is a highly conserved Asp (sometimes Glu) that is known to be critical for activity in a number of P450s. How this Asp and proton delivery networks are connected to redox partner binding is quite simple. In the closed state, this Asp is tied down by salt bridges, but these salt bridges are ruptured when Pdx binds, leaving the Asp free to serve its role in proton transfer. An alternative hypothesis suggests that a specific proton relay network is not really necessary. In this scenario, the Asp plays a structural role in the open/close transition and merely opening the active site access channel is sufficient to enable solvent protons in for O2 protonation. Experiments designed to test these various hypotheses have revealed some surprises in both P450cam and other bacterial P450s. Molecular dynamics and crystallography show that P450cam can undergo rather significant conformational gymnastics that result in a large restructuring of the active site requiring multiple cis/trans proline isomerizations. It also has been found that X-ray driven substrate hydroxylation is a useful tool for better understanding the role that the essential Asp and surrounding residues play in catalysis. Here we summarize these recent results which provide a much more dynamic picture of P450 catalysis.


Assuntos
Cânfora 5-Mono-Oxigenase , Ferredoxinas , Sítios de Ligação , Cânfora 5-Mono-Oxigenase/química , Domínio Catalítico , Sistema Enzimático do Citocromo P-450/metabolismo , Ferredoxinas/química , Humanos , Simulação de Dinâmica Molecular , Oxirredução , Conformação Proteica
17.
J Biomol Struct Dyn ; 40(5): 1995-2009, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33073701

RESUMO

In the light reaction of oxygenic photosynthesis, plastocyanin (PC) and ferredoxins (Fd) are small/diffusible redox-active proteins playing key roles in electron transfer/transport phenomena. In the Z-scheme mechanistic purview, they are considered as specific affinity binding-based electron-relay agents, linking the functions of Cytochrome b6f (Cyt. b6f), Photosystem I (PS I) and Fd:NADPH oxidoreductase (FNR). The murburn explanation for photolytic photophosphorylation deems PC/Fd as generic 'redox capacitors', temporally accepting and releasing one-electron equivalents in reaction milieu. Herein, we explore the two theories with respect to structural, distributional and functional aspects of PC/Fd. Amino acid residues located on the surface loci of key patches of PC/Fd vary in electrostatic/contour (topography) signatures. Crystal structures of four different complexes each of Cyt.f-PC and Fd-FNR show little conservation in the contact-surfaces, thereby discrediting 'affinity binding-based electron transfers (ET)' as an evolutionary logic. Further, thermodynamic and kinetic data of wildtype and mutant proteins interactions do not align with Z-scheme. Furthermore, micromolar physiological concentrations of PC and the non-conducive architecture of chloroplasts render the classical model untenable. In the murburn model, as PC is optional, the observation that plants lacking PC survive and grow is justified. Further, the low physiological concentration/distribution of PC in chloroplast lumen/stroma is supported by murburn equilibriums, as higher concentrations would limit electron transfers. Thus, structural evidence, interactive dynamics with redox partners and physiological distribution/role of PC/Fd support the murburn perspective that these proteins serve as generic redox-capacitors in chloroplasts.Communicated by Ramaswamy H. Sarma.


Assuntos
Ferredoxinas , Plastocianina , Transporte de Elétrons , Elétrons , Ferredoxinas/química , Ferredoxinas/metabolismo , Oxirredução , Fotossíntese , Plastocianina/química , Plastocianina/metabolismo
18.
Acta Crystallogr F Struct Biol Commun ; 77(Pt 11): 407-411, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34726179

RESUMO

Azotobacter vinelandii is a model diazotroph and is the source of most nitrogenase material for structural and biochemical work. Azotobacter can grow in above-atmospheric levels of oxygen, despite the sensitivity of nitrogenase activity to oxygen. Azotobacter has many iron-sulfur proteins in its genome, which were identified as far back as the 1960s and probably play roles in the complex redox chemistry that Azotobacter must maintain when fixing nitrogen. Here, the 2.1 Šresolution crystal structure of the [2Fe-2S] protein I (Shethna protein I) from A. vinelandii is presented, revealing a homodimer with the [2Fe-2S] cluster coordinated by the surrounding conserved cysteine residues. It is similar to the structure of the thioredoxin-like [2Fe-2S] protein from Aquifex aeolicus, including the positions of the [2Fe-2S] clusters and conserved cysteine residues. The structure of Shethna protein I will provide information for understanding its function in relation to nitrogen fixation and its evolutionary relationships to other ferredoxins.


Assuntos
Azotobacter vinelandii , Proteínas Ferro-Enxofre , Azotobacter vinelandii/química , Azotobacter vinelandii/genética , Azotobacter vinelandii/metabolismo , Cristalografia por Raios X , Ferredoxinas/química , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Nitrogenase/química , Nitrogenase/metabolismo
19.
Curr Issues Mol Biol ; 43(3): 1374-1390, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34698119

RESUMO

Ferredoxins, iron-sulfur (Fe-S) cluster proteins, play a key role in oxidoreduction reactions. To date, evolutionary analysis of these proteins across the domains of life have been confined to observing the abundance of Fe-S cluster types (2Fe-2S, 3Fe-4S, 4Fe-4S, 7Fe-8S (3Fe-4s and 4Fe-4S) and 2[4Fe-4S]) and the diversity of ferredoxins within these cluster types was not studied. To address this research gap, here we propose a subtype classification and nomenclature for ferredoxins based on the characteristic spacing between the cysteine amino acids of the Fe-S binding motif as a subtype signature to assess the diversity of ferredoxins across the living organisms. To test this hypothesis, comparative analysis of ferredoxins between bacterial groups, Alphaproteobacteria and Firmicutes and ferredoxins collected from species of different domains of life that are reported in the literature has been carried out. Ferredoxins were found to be highly diverse within their types. Large numbers of alphaproteobacterial species ferredoxin subtypes were found in Firmicutes species and the same ferredoxin subtypes across the species of Bacteria, Archaea, and Eukarya, suggesting shared common ancestral origin of ferredoxins between Archaea and Bacteria and lateral gene transfer of ferredoxins from prokaryotes (Archaea/Bacteria) to eukaryotes. This study opened new vistas for further analysis of diversity of ferredoxins in living organisms.


Assuntos
Ferredoxinas/química , Ferredoxinas/genética , Variação Genética , Archaea/classificação , Archaea/genética , Bactérias/classificação , Bactérias/genética , Biologia Computacional/métodos , Bases de Dados Genéticas , Eucariotos/classificação , Eucariotos/genética , Evolução Molecular , Ferredoxinas/classificação , Filogenia , Especificidade da Espécie
20.
Inorg Chem ; 60(18): 13869-13875, 2021 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34488341

RESUMO

The nickel-iron carbon monoxide dehydrogenase (CODH) enzyme catalyzes the reversible and selective interconversion of carbon dioxide (CO2) to carbon monoxide (CO) with high rates and negligible overpotential. Despite decades of research, many questions remain about this complex metalloenzyme system. A simplified model enzyme could provide substantial insight into biological carbon cycling. Here, we demonstrate reversible electron transfer and binding of both CO and cyanide, a substrate and an inhibitor of CODH, respectively, in a Pyrococcus furiosus (Pf) ferredoxin (Fd) protein that has been reconstituted with a nickel-iron sulfide cluster ([NiFe3S4] Fd). The [NiFe3S4] cluster mimics the core of the native CODH active site and thus serves as a protein-based structural model of the CODH subsite. Notably, despite binding cyanide, no CO binding is observed for the physiological [Fe4S4] clusters in Pf Fd, providing chemical rationale underlying the evolution of a site-differentiated cluster for substrate conversion in native CODH. The demonstration of a substrate-binding metalloprotein model of CODH sets the stage for high-resolution spectroscopic and mechanistic studies correlating the subsite structure and function, ultimately guiding the design of anthropogenic catalysts that harness the advantages of CODH for effective CO2 reduction.


Assuntos
Aldeído Oxirredutases/metabolismo , Ferredoxinas/metabolismo , Complexos Multienzimáticos/metabolismo , Pyrococcus furiosus/química , Aldeído Oxirredutases/química , Dióxido de Carbono/química , Dióxido de Carbono/metabolismo , Monóxido de Carbono/química , Monóxido de Carbono/metabolismo , Transporte de Elétrons , Ferredoxinas/química , Modelos Moleculares , Complexos Multienzimáticos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...