Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.017
Filtrar
1.
FEBS Open Bio ; 14(10): 1731-1745, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39123319

RESUMO

Pectocin M1 (PM1), the bacteriocin from phytopathogenic Pectobacterium carotovorum which causes soft rot disease, has a unique ferredoxin domain that allows it to use FusA of the plant ferredoxin uptake system. To probe the structure-based mechanism of PM1 uptake, we determined the X-ray structure of full-length PM1, containing an N-terminal ferredoxin and C-terminal catalytic domain connected by helical linker, at 2.04 Å resolution. Based on published FusA structure and NMR data for PM1 ferredoxin domain titrated with FusA, we modeled docking of the ferredoxin domain with FusA. Combining the docking models with the X-ray structures of PM1 and FusA enables us to propose the mechanism by which PM1 undergoes dynamic domain rearrangement to translocate across the target cell outer membrane.


Assuntos
Ferredoxinas , Ferredoxinas/metabolismo , Ferredoxinas/química , Cristalografia por Raios X , Bacteriocinas/química , Bacteriocinas/metabolismo , Pectobacterium carotovorum/metabolismo , Pectobacterium carotovorum/química , Conformação Proteica , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Simulação de Acoplamento Molecular
2.
Chembiochem ; 25(20): e202400380, 2024 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-38985090

RESUMO

In the green alga Chlamydomonas reinhardtii, hydrogenase HydA1 converts protons and electrons to H2 at the H-cluster, which includes a [4Fe-4S] cluster linked to a [2Fe] cluster. The yield of H2 is limited by the electron transfer to HydA1, mediated by the iron-sulfur unit of a photosynthetic electron transfer ferredoxin (PetF). In this study, I have investigated by molecular dynamics and the hybrid quantum mechanics/molecular mechanics method two canonical iron-sulfur peptides (PM1 and FBM) that hold potential as PetF replacements. Using a docking approach, I predict that the distance between the two iron-sulfur clusters in FBM/HydA1 is shorter than in PM1/HydA1, ensuring a greater electron transfer rate. This finding is in line with the reported higher H2 production rates for FBM/HydA1. I also show that the redox potential of these peptides, and therefore their electron transfer properties, can be changed by single-residue mutations in the secondary coordination sphere of their cluster. In particular, I have designed a PM1 variant that disrupts the hydrogen-bonding network between water and the cluster, shifting the redox potential negatively compared to PM1. These results will guide experiments aimed at replacing PetF with peptides that can unlock the biotechnological potential of the alga.


Assuntos
Chlamydomonas reinhardtii , Ferredoxinas , Hidrogenase , Proteínas Ferro-Enxofre , Peptídeos , Hidrogenase/química , Hidrogenase/metabolismo , Ferredoxinas/metabolismo , Ferredoxinas/química , Transporte de Elétrons , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/metabolismo , Chlamydomonas reinhardtii/enzimologia , Chlamydomonas reinhardtii/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Simulação de Dinâmica Molecular
3.
Fish Shellfish Immunol ; 152: 109796, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39074519

RESUMO

Ferredoxin (FDX) is a highly conserved iron-sulfur protein that participates in redox reactions and plays an important role as an electron transport protein in biological processes. However, its function in marine fish remains unclear. We identified two ferrodoxin proteins, FDX1 and FDX2, from black scraper (Thamnaconus modestus) to confirm their genetic structures and expression profiles and to investigate their antimicrobial activity properties by fabricating them with antimicrobial peptides based on sequences. The two TmFDXs mRNAs were most abundant in peripheral blood leukocytes of healthy T. modestus. After artificial infection with Vibrio anguillarum, a major pathogen of T. modestus, TmFDX1 mRNA was significantly upregulated in the gills, heart, intestines, kidneys, liver, and spleen, but was consistently downregulated in the brain. The expression levels of TmFDX2 mRNA were significantly upregulated in the heart, intestines, kidneys, liver, and spleen; however, no significant changes in expression were observed in the brain or gills. Based on the 2Fe-2S ferredoxin-type iron-sulfur-binding domain sequence, two peptides (pFDX1 and pFDX2) were synthesized. The bactericidal effect, biofilm formation inhibition, and gDNA-binding activity of these peptides were investigated. These findings highlight the potential as a natural peptide candidate for TmFDXs.


Assuntos
Sequência de Aminoácidos , Peptídeos Antimicrobianos , Ferredoxinas , Doenças dos Peixes , Proteínas de Peixes , Vibrioses , Vibrio , Animais , Proteínas de Peixes/genética , Proteínas de Peixes/química , Proteínas de Peixes/imunologia , Doenças dos Peixes/imunologia , Vibrio/fisiologia , Peptídeos Antimicrobianos/química , Peptídeos Antimicrobianos/farmacologia , Peptídeos Antimicrobianos/genética , Ferredoxinas/genética , Ferredoxinas/química , Vibrioses/veterinária , Vibrioses/imunologia , Imunidade Inata/genética , Alinhamento de Sequência/veterinária , Perfilação da Expressão Gênica/veterinária , Filogenia , Regulação da Expressão Gênica/efeitos dos fármacos , Perciformes/imunologia , Perciformes/genética
4.
Acta Crystallogr D Struct Biol ; 80(Pt 8): 599-604, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38984904

RESUMO

The Azotobacter vinelandii FeSII protein forms an oxygen-resistant complex with the nitrogenase MoFe and Fe proteins. FeSII is an adrenodoxin-type ferredoxin that forms a dimer in solution. Previously, the crystal structure was solved [Schlesier et al. (2016), J. Am. Chem. Soc. 138, 239-247] with five copies in the asymmetric unit. One copy is a normal adrenodoxin domain that forms a dimer with its crystallographic symmetry mate. The other four copies are in an `open' conformation with a loop flipped out exposing the 2Fe-2S cluster. The open and closed conformations were interpreted as oxidized and reduced, respectively, and the large conformational change in the open configuration allowed binding to nitrogenase. Here, the structure of FeSII was independently solved in the same crystal form. The positioning of the atoms in the unit cell is similar to the earlier report. However, the interpretation of the structure is different. The `open' conformation is interpreted as the product of a crystallization-induced domain swap. The 2Fe-2S cluster is not exposed to solvent, but in the crystal its interacting helix is replaced by the same helix residues from a crystal symmetry mate. The domain swap is complicated, as it is unusual in being in the middle of the protein rather than at a terminus, and it creates arrangements of molecules that can be interpreted in multiple ways. It is also cautioned that crystal structures should be interpreted in terms of the contents of the entire crystal rather than of one asymmetric unit.


Assuntos
Azotobacter vinelandii , Proteínas de Bactérias , Modelos Moleculares , Azotobacter vinelandii/química , Cristalografia por Raios X , Proteínas de Bactérias/química , Conformação Proteica , Domínios Proteicos , Ferredoxinas/química , Multimerização Proteica , Proteínas Ferro-Enxofre/química
5.
J Biochem ; 176(3): 237-244, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-38861409

RESUMO

In the chloroplast stroma, dynamic pH changes occur from acidic to alkaline in response to fluctuating light conditions. We investigated the pH dependency of the electron transfer reaction of ferredoxin-NADP+ reductase (FNR) with ferredoxin (Fd) isoproteins, Fd1 and Fd2, which are localized in mesophyll cells and bundle sheath cells, respectively, in the leaves of C4 plant maize. The pH-dependent profile of the electron transfer activity with FNR was quite different between Fd1 and Fd2, which was mainly explained by the opposite pH dependency of the Km value of these Fds for FNR. Replacement of the amino acid residue at position of 65 (D65N) and 78 (H78A) between the two Fds conferred different effect on their pH dependency of the Km value. Double mutations of the two residues between Fd1 and Fd2 (Fd1D65N/H78A and Fd2N65D/A78H) led to the mutual exchange of the pH dependency of the electron transfer activity. This exchange was mainly explained by the changes in the pH-dependent profile of the Km values. Therefore, the differences in Asp/Asn at position 65 and His/Ala at position 78 between Fd1 and Fd2 were shown to be the major determinants for their different pH dependency in the electron transfer reaction with FNR.


Assuntos
Ferredoxina-NADP Redutase , Ferredoxinas , Folhas de Planta , Zea mays , Zea mays/metabolismo , Zea mays/enzimologia , Zea mays/genética , Concentração de Íons de Hidrogênio , Ferredoxina-NADP Redutase/metabolismo , Ferredoxina-NADP Redutase/química , Ferredoxina-NADP Redutase/genética , Folhas de Planta/metabolismo , Folhas de Planta/enzimologia , Transporte de Elétrons , Ferredoxinas/metabolismo , Ferredoxinas/química , Aminoácidos/metabolismo
6.
Chembiochem ; 25(19): e202400098, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-38787654

RESUMO

Cytochrome P450 monooxygenases (CYPs) are valuable biocatalysts for the oxyfunctionalization of non-activated carbon-hydrogen bonds. Most CYPs rely on electron transport proteins as redox partners. In this study, the ferredoxin reductase (FdR) and ferredoxin (FD) for a cytochrome P450 monooxygenase from Acinetobacter sp. OC4 are investigated. Upon heterologous production of both proteins independently in Escherichia coli, spectral analysis showed their reduction capability towards reporter electron acceptors, e. g., cytochrome c. The individual proteins' specific activity towards cytochrome c reduction was 25 U mg-1. Furthermore, the possibility to enhance electron transfer by artificial fusion of the units was elucidated. FdR and FD were linked by helical linkers [EAAAK]n, flexible glycine linkers [GGGGS]n or rigid proline linkers [EPPPP]n of n=1-4 sequence repetitions. The system with a glycine linker (n=4) reached an appreciable specific activity of 19 U mg-1 towards cytochrome c. Moreover, their ability to drive different members of the CYP153A subfamily is demonstrated. By creating artificial self-sufficient P450s with FdR, FD, and a panel of four CYP153A representatives, effective hydroxylation of n-hexane in a whole-cell system was achieved. The results indicate this protein combination to constitute a functional and versatile surrogate electron transport system for this subfamily.


Assuntos
Acinetobacter , Alcanos , Sistema Enzimático do Citocromo P-450 , Acinetobacter/enzimologia , Acinetobacter/metabolismo , Transporte de Elétrons , Sistema Enzimático do Citocromo P-450/metabolismo , Alcanos/metabolismo , Alcanos/química , Ferredoxinas/metabolismo , Ferredoxinas/química , Escherichia coli/metabolismo , Oxirredução
7.
Biochemistry ; 63(12): 1588-1598, 2024 06 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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
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
14.
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
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA