Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 366
Filter
1.
J Biol Chem ; 300(4): 107122, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417793

ABSTRACT

The flavodoxin of Rhodopseudomonas palustris CGA009 (Rp9Fld) supplies highly reducing equivalents to crucial enzymes such as hydrogenase, especially when the organism is iron-restricted. By acquiring those electrons from photodriven electron flow via the bifurcating electron transfer flavoprotein, Rp9Fld provides solar power to vital metabolic processes. To understand Rp9Fld's ability to work with diverse partners, we solved its crystal structure. We observed the canonical flavodoxin (Fld) fold and features common to other long-chain Flds but not all the surface loops thought to recognize partner proteins. Moreover, some of the loops display alternative structures and dynamics. To advance studies of protein-protein associations and conformational consequences, we assigned the 19F NMR signals of all five tyrosines (Tyrs). Our electrochemical measurements show that incorporation of 3-19F-Tyr in place of Tyr has only a modest effect on Rp9Fld's redox properties even though Tyrs flank the flavin on both sides. Meanwhile, the 19F probes demonstrate the expected paramagnetic effect, with signals from nearby Tyrs becoming broadened beyond detection when the flavin semiquinone is formed. However, the temperature dependencies of chemical shifts and linewidths reveal dynamics affecting loops close to the flavin and regions that bind to partners in a variety of systems. These coincide with patterns of amino acid type conservation but not retention of specific residues, arguing against detailed specificity with respect to partners. We propose that the loops surrounding the flavin adopt altered conformations upon binding to partners and may even participate actively in electron transfer.


Subject(s)
Bacterial Proteins , Flavodoxin , Models, Molecular , Oxidation-Reduction , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Crystallography, X-Ray , Flavodoxin/chemistry , Flavodoxin/metabolism , Protein Conformation , Tyrosine/chemistry , Tyrosine/metabolism
2.
J Biol Chem ; 299(7): 104902, 2023 07.
Article in English | MEDLINE | ID: mdl-37302554

ABSTRACT

Fusobacterium nucleatum is an opportunistic oral pathogen that is associated with various cancers. To fulfill its essential need for iron, this anaerobe will express heme uptake machinery encoded at a single genetic locus. The heme uptake operon includes HmuW, a class C radical SAM-dependent methyltransferase that degrades heme anaerobically to release Fe2+ and a linear tetrapyrrole called anaerobilin. The last gene in the operon, hmuF encodes a member of the flavodoxin superfamily of proteins. We discovered that HmuF and a paralog, FldH, bind tightly to both FMN and heme. The structure of Fe3+-heme-bound FldH (1.6 Å resolution) reveals a helical cap domain appended to the ⍺/ß core of the flavodoxin fold. The cap creates a hydrophobic binding cleft that positions the heme planar to the si-face of the FMN isoalloxazine ring. The ferric heme iron is hexacoordinated to His134 and a solvent molecule. In contrast to flavodoxins, FldH and HmuF do not stabilize the FMN semiquinone but instead cycle between the FMN oxidized and hydroquinone states. We show that heme-loaded HmuF and heme-loaded FldH traffic heme to HmuW for degradation of the protoporphyrin ring. Both FldH and HmuF then catalyze multiple reductions of anaerobilin through hydride transfer from the FMN hydroquinone. The latter activity eliminates the aromaticity of anaerobilin and the electrophilic methylene group that was installed through HmuW turnover. Hence, HmuF provides a protected path for anaerobic heme catabolism, offering F. nucleatum a competitive advantage in the colonization of anoxic sites of the human body.


Subject(s)
Flavodoxin , Fusobacterium nucleatum , Heme , Tetrapyrroles , Humans , Flavin Mononucleotide/metabolism , Flavodoxin/chemistry , Flavodoxin/classification , Flavodoxin/genetics , Flavodoxin/metabolism , Fusobacterium nucleatum/chemistry , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Heme/metabolism , Iron/metabolism , Oxidation-Reduction , Tetrapyrroles/metabolism , Biological Transport , Genes, Bacterial , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Protein Domains , Fusobacterium Infections/microbiology
3.
Biochem Biophys Res Commun ; 639: 134-141, 2023 01 08.
Article in English | MEDLINE | ID: mdl-36493556

ABSTRACT

In cyanobacteria and algae (but not plants), flavodoxin (Fld) replaces ferredoxin (Fd) under stress conditions to transfer electrons from photosystem I (PSI) to ferredoxin-NADP+ reductase (FNR) during photosynthesis. Fld constitutes a small electron carrier noncovalently bound to flavin mononucleotide (FMN), and also an ideal model for revealing the protein/flavin-binding mechanism because of its relative simplicity compared to other flavoproteins. Here, we report two crystal structures of apo-Fld from Synechococcus sp. PCC 7942, one dimeric structure of 2.09 Å and one monomeric structure of 1.84 Å resolution. Analytical ultracentrifugation showed that in solution, apo-Fld exists both as monomers and dimers. Our dimer structure contains two ligand-binding pockets separated by a distance of 45 Å, much longer than the previous structures of FMN-bound dimers. These results suggested a potential dimer-monomer transition mechanism of cyanobacterial apo-Fld. We further propose that the dimer represents the "standby" state to stabilize itself, while the monomer constitutes the "ready" state to bind FMN. Furthermore, we generated a new docking model of cyanobacterial Fld-FNR complex based on the recently reported cryo-EM structures, and mapped the special interactions between Fld and FNR in detail.


Subject(s)
Anabaena , Cyanobacteria , Flavodoxin/chemistry , Flavodoxin/metabolism , Ferredoxins/metabolism , Anabaena/metabolism , Flavoproteins , Ferredoxin-NADP Reductase/chemistry , Cyanobacteria/metabolism , Oxidation-Reduction
4.
Brief Bioinform ; 22(5)2021 09 02.
Article in English | MEDLINE | ID: mdl-33569580

ABSTRACT

In this paper, we show that the combination of NMR theoretical and experimental results can help to solve the molecular structure of peptides, here it is used as an example the residue Leucine-67 in Desulfovibrio vulgaris flavodoxin. We apply a computational protocol based on the leucine amino acid dipeptide, which, using calculated and experimental spin-spin coupling constants, allows us to obtain the conformation of the amino acid side chain. Calculated results show that the best agreement is obtained when three conformers around the lateral chain angle $\chi _1$ are considered or when the dynamic effect in the torsional angles is included. The population of each structure is estimated and analyzed according to the correlation between those two approaches. Independently of the approach, the estimated $\chi _1$ angle in solution is close to the staggered value of -60$^\circ $ and deviates significantly from the average x-ray angle of -90$^\circ $.


Subject(s)
Desulfovibrio vulgaris/chemistry , Flavodoxin/chemistry , Leucine/chemistry , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Amino Acid Sequence , Flavodoxin/isolation & purification , Peptides/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Solutions , Solvents/chemistry , Water/chemistry
5.
PLoS Pathog ; 17(2): e1009355, 2021 02.
Article in English | MEDLINE | ID: mdl-33630938

ABSTRACT

A family of eleven glycosylphosphatidylinositol-anchored aspartyl proteases, commonly referred to as CgYapsins, regulate a myriad of cellular processes in the pathogenic yeast Candida glabrata, but their protein targets are largely unknown. Here, using the immunoprecipitation-mass spectrometry approach, we identify the flavodoxin-like protein (Fld-LP), CgPst2, to be an interactor of one of the aspartyl protease CgYps1. We also report the presence of four Fld-LPs in C. glabrata, which are required for survival in kidneys in the murine model of systemic candidiasis. We further demonstrated that of four Fld-LPs, CgPst2 was solely required for menadione detoxification. CgPst2 was found to form homo-oligomers, and contribute to cellular NADH:quinone oxidoreductase activity. CgYps1 cleaved CgPst2 at the C-terminus, and this cleavage was pivotal to oligomerization, activity and function of CgPst2. The arginine-174 residue in CgPst2 was essential for CgYps1-mediated cleavage, with alanine substitution of the arginine-174 residue also leading to elevated activity and oligomerization of CgPst2. Finally, we demonstrate that menadione treatment led to increased CgPst2 and CgYps1 protein levels, diminished CgYps1-CgPst2 interaction, and enhanced CgPst2 cleavage and activity, thereby implicating CgYps1 in activating CgPst2. Altogether, our findings of proteolytic cleavage as a key regulatory determinant of CgPst2, which belongs to the family of highly conserved, electron-carrier flavodoxin-fold-containing proteins, constituting cellular oxidative stress defense system in diverse organisms, unveil a hidden regulatory layer of environmental stress response mechanisms.


Subject(s)
Aspartic Acid Proteases/metabolism , Candida glabrata/metabolism , Candidiasis/microbiology , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal/drug effects , Oxidative Stress , Animals , Benzoquinones/pharmacology , Candida glabrata/drug effects , Candida glabrata/genetics , Candida glabrata/growth & development , Candidiasis/drug therapy , Candidiasis/metabolism , Female , Flavodoxin/chemistry , Indicators and Reagents/pharmacology , Mice , Mice, Inbred BALB C , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidation-Reduction , Protein Conformation , Vitamin K 3/pharmacology , Vitamins/pharmacology
6.
Proc Natl Acad Sci U S A ; 116(51): 25917-25922, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31801875

ABSTRACT

Flavodoxins, electron transfer proteins essential for diverse metabolisms in microbes from the domain Bacteria, are extensively characterized. Remarkably, although genomic annotations of flavodoxins are widespread in microbes from the domain Archaea, none have been isolated and characterized. Herein is described the structural, biochemical, and physiological characterization of an unusual flavodoxin (FldA) from Methanosarcina acetivorans, an acetate-utilizing methane-producing microbe of the domain Archaea In contrast to all flavodoxins, FldA is homodimeric, markedly less acidic, and stabilizes an anionic semiquinone. The crystal structure reveals an flavin mononucleotide (FMN) binding site unique from all other flavodoxins that provides a rationale for stabilization of the anionic semiquinone and a remarkably low reduction potentials for both the oxidized/semiquinone (-301 mV) and semiquinone/hydroquinone couples (-464 mV). FldA is up-regulated in acetate-grown versus methanol-grown cells and shown here to substitute for ferredoxin in mediating the transfer of low potential electrons from the carbonyl of acetate to the membrane-bound electron transport chain that generates ion gradients driving ATP synthesis. FldA offers potential advantages over ferredoxin by (i) sparing iron for abundant iron-sulfur proteins essential for acetotrophic growth and (ii) resilience to oxidative damage.


Subject(s)
Flavodoxin/chemistry , Flavodoxin/metabolism , Methanosarcina/metabolism , Acetates/metabolism , Bacterial Proteins/chemistry , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , Ferredoxins/chemistry , Ferredoxins/metabolism , Flavin Mononucleotide/chemistry , Flavodoxin/genetics , Flavodoxin/isolation & purification , Flavoproteins/chemistry , Global Warming , Hydroquinones , Methane/metabolism , Models, Molecular , Oxidation-Reduction , Protein Conformation
7.
Arch Biochem Biophys ; 703: 108839, 2021 05 30.
Article in English | MEDLINE | ID: mdl-33727041

ABSTRACT

Flavodoxin is a small protein that employs a non-covalently bound flavin to mediate single-electron transfer at low potentials. The long-chain flavodoxins possess a long surface loop that is proposed to interact with partner proteins. We have incorporated 19F-labeled tyrosine in long-chain flavodoxin from Rhodopseudomonas palustris to gain a probe of possible loop dynamics, exploiting the presence of a Tyr in the long loop in addition to Tyr residues near the flavin. We report 19F resonance assignments for all four Tyrs, and demonstration of a pair of resonances in slow exchange, both corresponding to a Tyr adjacent to the flavin. We also provide evidence for dynamics affecting the Tyr in the long loop. Thus, we show that 19F NMR of 19F-Tyr labeled flavodoxin holds promise for monitoring possible changes in conformation upon binding to partner proteins.


Subject(s)
Flavodoxin/chemistry , Flavodoxin/metabolism , Nuclear Magnetic Resonance, Biomolecular , Models, Molecular , Protein Conformation
8.
Phys Chem Chem Phys ; 24(1): 382-391, 2021 Dec 22.
Article in English | MEDLINE | ID: mdl-34889914

ABSTRACT

Short-range protein electron transfer (ET) is ubiquitous in biology and is often observed in photosynthesis, photoreceptors and photoenzymes. These ET processes occur on an ultrafast timescale from femtoseconds to picoseconds at a short donor-acceptor distance within 10 Å, and thus couple with local environmental fluctuations. Here, we use oxidized Anabaena flavodoxin as a model system and have systematically studied the photoinduced redox cycle of the wild type and seven mutant proteins by femtosecond spectroscopy. We observed a series of ultrafast dynamics from the initial charge separation in 100-200 fs, subsequent charge recombination in 1-2 ps and final vibrational cooling process of the products in 3-6 ps. We further characterized the active-site solvation and observed the relaxations in 1-200 ps, indicating a nonergodic ET dynamics. With our new ET model, we uncovered a minor outer (solvent) reorganization energy and a large inner (donor and acceptor) reorganization energy, suggesting a frozen active site in the initial ultrafast ET while the back ET couples with the environment relaxations. The vibronically coupled back ET dynamics was first reported in D. vulgaris flavodoxin and here is observed in Anabaena flavodoxin again, completely due to the faster ET dynamics than the cooling relaxations. We also compared the two flavodoxin structures, revealing a stronger coupling with the donor tyrosine in Anabaena. All ultrafast ET dynamics are from the large donor-acceptor couplings and the minor activation barriers due to the reaction free energies being close to the inner reorganization energies. These observations should be general to many redox reactions in flavoproteins.


Subject(s)
Flavodoxin/metabolism , Molecular Dynamics Simulation , Proteins/metabolism , Anabaena/chemistry , Anabaena/metabolism , Electron Transport , Flavodoxin/chemistry , Proteins/chemistry
9.
Int J Mol Sci ; 22(18)2021 Sep 20.
Article in English | MEDLINE | ID: mdl-34576300

ABSTRACT

Antimicrobial resistant (AMR) bacteria constitute a global health concern. Helicobacter pylori is a Gram-negative bacterium that infects about half of the human population and is a major cause of peptic ulcer disease and gastric cancer. Increasing resistance to triple and quadruple H. pylori eradication therapies poses great challenges and urges the development of novel, ideally narrow spectrum, antimicrobials targeting H. pylori. Here, we describe the antimicrobial spectrum of a family of nitrobenzoxadiazol-based antimicrobials initially discovered as inhibitors of flavodoxin: an essential H. pylori protein. Two groups of inhibitors are described. One group is formed by narrow-spectrum compounds, highly specific for H. pylori, but ineffective against enterohepatic Helicobacter species and other Gram-negative or Gram-positive bacteria. The second group includes extended-spectrum antimicrobials additionally targeting Gram-positive bacteria, the Gram-negative Campylobacter jejuni, and most Helicobacter species, but not affecting other Gram-negative pathogens. To identify the binding site of the inhibitors in the flavodoxin structure, several H. pylori-flavodoxin variants have been engineered and tested using isothermal titration calorimetry. An initial study of the inhibitors capacity to generate resistances and of their synergism with antimicrobials commonly used in H. pylori eradication therapies is described. The narrow-spectrum inhibitors, which are expected to affect the microbiota less dramatically than current antimicrobial drugs, offer an opportunity to develop new and specific H. pylori eradication combinations to deal with AMR in H. pylori. On the other hand, the extended-spectrum inhibitors constitute a new family of promising antimicrobials, with a potential use against AMR Gram-positive bacterial pathogens.


Subject(s)
Anti-Infective Agents/pharmacology , Flavodoxin/antagonists & inhibitors , Helicobacter/drug effects , Anti-Infective Agents/chemical synthesis , Binding Sites , Drug Synergism , Flavodoxin/chemistry , Flavodoxin/metabolism , Molecular Docking Simulation , Protein Binding
10.
Biochemistry ; 58(48): 4790-4793, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31724394

ABSTRACT

Evolutionary processes that led to the emergence of structured protein domains left footprints in the sequences of modern proteins. We searched for such hints employing state-of-the-art sequence analysis and found evidence that the HemD-like fold emerged from the flavodoxin-like fold through segment swap and gene duplication. To verify this hypothesis, we reverted these evolutionary steps experimentally, constructing a HemD-half that resulted in a protein with the canonical flavodoxin-like architecture. These results of fold reconstruction from the sequence of a different fold strongly support our hypothesis of common ancestry. It further illustrates the plasticity of modern proteins to form new folded proteins.


Subject(s)
Bacteria/metabolism , Flavodoxin/chemistry , Flavodoxin/genetics , Bacteria/chemistry , Bacteria/genetics , Flavodoxin/metabolism , Models, Molecular , Protein Conformation , Protein Folding
11.
J Bacteriol ; 200(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29483165

ABSTRACT

Nitrogenase catalyzes the reduction of dinitrogen (N2) using low-potential electrons from ferredoxin (Fd) or flavodoxin (Fld) through an ATP-dependent process. Since its emergence in an anaerobic chemoautotroph, this oxygen (O2)-sensitive enzyme complex has evolved to operate in a variety of genomic and metabolic backgrounds, including those of aerobes, anaerobes, chemotrophs, and phototrophs. However, whether pathways of electron delivery to nitrogenase are influenced by these different metabolic backgrounds is not well understood. Here, we report the distribution of homologs of Fds, Flds, and Fd-/Fld-reducing enzymes in 359 genomes of putative N2 fixers (diazotrophs). Six distinct lineages of nitrogenase were identified, and their distributions largely corresponded to differences in the host cells' ability to integrate O2 or light into energy metabolism. The predicted pathways of electron transfer to nitrogenase in aerobes, facultative anaerobes, and phototrophs varied from those in anaerobes at the levels of Fds/Flds used to reduce nitrogenase, the enzymes that generate reduced Fds/Flds, and the putative substrates of these enzymes. Proteins that putatively reduce Fd with hydrogen or pyruvate were enriched in anaerobes, while those that reduce Fd with NADH/NADPH were enriched in aerobes, facultative anaerobes, and anoxygenic phototrophs. The energy metabolism of aerobic, facultatively anaerobic, and anoxygenic phototrophic diazotrophs often yields reduced NADH/NADPH that is not sufficiently reduced to drive N2 reduction. At least two mechanisms have been acquired by these taxa to overcome this limitation and to generate electrons with potentials capable of reducing Fd. These include the bifurcation of electrons or the coupling of Fd reduction to reverse ion translocation.IMPORTANCE Nitrogen fixation supplies fixed nitrogen to cells from a variety of genomic and metabolic backgrounds, including those of aerobes, facultative anaerobes, chemotrophs, and phototrophs. Here, using informatics approaches applied to genomic data, we show that pathways of electron transfer to nitrogenase in metabolically diverse diazotrophic taxa have diversified primarily in response to host cells' acquired ability to integrate O2 or light into their energy metabolism. The acquisition of two key enzyme complexes enabled aerobic and facultatively anaerobic phototrophic taxa to generate electrons of sufficiently low potential to reduce nitrogenase: the bifurcation of electrons via the Fix complex or the coupling of Fd reduction to reverse ion translocation via the Rhodobacter nitrogen fixation (Rnf) complex.


Subject(s)
Bacteria/genetics , Ferredoxins/chemistry , Flavodoxin/chemistry , Fungi/genetics , Nitrogenase/metabolism , Aerobiosis , Anaerobiosis , Bacteria/enzymology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Electron Transport , Evolution, Molecular , Fungal Proteins/genetics , Fungal Proteins/metabolism , Fungi/enzymology , Genome, Bacterial , Genome, Fungal , Nitrogenase/genetics , Phylogeny , Signal Transduction
12.
Biochemistry ; 57(37): 5427-5436, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30142264

ABSTRACT

Flavodoxins (Flds) are small, bacterial proteins that transfer electrons to various redox enzymes. Flavodoxins are reduced by ferredoxin/flavodoxin NADP+ oxidoreductases (FNRs), but little is known of the FNR-Fld interaction. Here, we compare the interactions of two flavodoxins (Fld1-2), one flavodoxin-like protein (NrdI), and three different thioredoxin reductase (TrxR)-like FNRs (FNR1-3), all from Bacillus cereus. Steady-state kinetics shows that the FNR2-Fld2 electron transfer pair is particularly efficient, and redox potential measurements also indicate that this is the most favorable electron donor/acceptor pair. Furthermore, crystal structures of FNR1 and FNR2 show that the proteins have crystallized in different conformations, a closed and an open conformation, respectively. We suggest that a large-scale conformational rearrangement takes place during the FNR catalytic cycle to allow for the binding and reduction of the Fld and, subsequently, the re-reduction of the FNR by NADPH. Finally, inspection of the residues surrounding the FAD cofactor in the FNR active site shows that a key isoalloxazine ring-stacking residue is different in FNR1 and FNR2, which could explain the large difference in catalytic efficiency between the two FNRs. To date, all of the characterized TrxR-like FNRs have a residue with aromatic character stacking against the FAD isoalloxazine ring, and this has been thought to be a conserved feature of this class of FNRs. FNR1, however, has a valine in this position. Bioinformatic analysis shows that the TrxR-like FNRs can actually be divided into two groups, one group where the FAD-stacking residue has aromatic character and another group where it is valine.


Subject(s)
Bacillus cereus/enzymology , Flavodoxin/metabolism , NADH, NADPH Oxidoreductases/classification , NADH, NADPH Oxidoreductases/metabolism , Binding Sites , Catalytic Domain , Crystallography, X-Ray , Electron Transport , Flavodoxin/chemistry , Models, Molecular , NADH, NADPH Oxidoreductases/chemistry , Oxidation-Reduction , Protein Conformation
13.
J Biol Chem ; 292(34): 14039-14049, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28615449

ABSTRACT

Flavin-based electron transfer bifurcation is emerging as a fundamental and powerful mechanism for conservation and deployment of electrochemical energy in enzymatic systems. In this process, a pair of electrons is acquired at intermediate reduction potential (i.e. intermediate reducing power), and each electron is passed to a different acceptor, one with lower and the other with higher reducing power, leading to "bifurcation." It is believed that a strongly reducing semiquinone species is essential for this process, and it is expected that this species should be kinetically short-lived. We now demonstrate that the presence of a short-lived anionic flavin semiquinone (ASQ) is not sufficient to infer the existence of bifurcating activity, although such a species may be necessary for the process. We have used transient absorption spectroscopy to compare the rates and mechanisms of decay of ASQ generated photochemically in bifurcating NADH-dependent ferredoxin-NADP+ oxidoreductase and the non-bifurcating flavoproteins nitroreductase, NADH oxidase, and flavodoxin. We found that different mechanisms dominate ASQ decay in the different protein environments, producing lifetimes ranging over 2 orders of magnitude. Capacity for electron transfer among redox cofactors versus charge recombination with nearby donors can explain the range of ASQ lifetimes that we observe. Our results support a model wherein efficient electron propagation can explain the short lifetime of the ASQ of bifurcating NADH-dependent ferredoxin-NADP+ oxidoreductase I and can be an indication of capacity for electron bifurcation.


Subject(s)
Bacterial Proteins/metabolism , Flavin-Adenine Dinucleotide/analogs & derivatives , Flavodoxin/metabolism , Models, Molecular , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/metabolism , Nitroreductases/metabolism , Oxidoreductases/metabolism , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Benzoic Acid/chemistry , Benzoic Acid/metabolism , Biocatalysis , Desulfovibrio vulgaris/enzymology , Electron Transport , Enterobacter cloacae/enzymology , Flavin-Adenine Dinucleotide/chemistry , Flavin-Adenine Dinucleotide/metabolism , Flavodoxin/chemistry , Flavodoxin/genetics , Holoenzymes/chemistry , Holoenzymes/genetics , Holoenzymes/metabolism , Multienzyme Complexes/chemistry , Multienzyme Complexes/genetics , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/genetics , Nitroreductases/chemistry , Nitroreductases/genetics , Oxidation-Reduction , Oxidoreductases/chemistry , Oxidoreductases/genetics , Pyrococcus furiosus/enzymology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Silent Mutation , Thermus thermophilus/enzymology , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/metabolism
14.
Phys Chem Chem Phys ; 20(10): 7059-7072, 2018 Mar 07.
Article in English | MEDLINE | ID: mdl-29473921

ABSTRACT

Flavodoxins have a protein topology that can be traced back to the universal ancestor of the three kingdoms of life. Proteins with this type of architecture tend to temporarily misfold during unassisted folding to their native state and form intermediates. Several of these intermediate species are molten globules (MGs), which are characterized by a substantial amount of secondary structure, yet without the tertiary side-chain packing of natively folded proteins. An off-pathway MG is formed at physiological ionic strength in the case of the F44Y variant of Azotobacter vinelandii apoflavodoxin (i.e., flavodoxin without flavin mononucleotide (FMN)). Here, we show that at this condition actually two folding species of this apoprotein co-exist at equilibrium. These species were detected by using a combination of FMN fluorescence quenching upon cofactor binding to the apoprotein and of polarized time-resolved tryptophan fluorescence spectroscopy. Besides the off-pathway MG, we observe the simultaneous presence of an on-pathway folding intermediate, which is native-like. Presence of concurrent intermediates at physiological ionic strength enables future exploration of how aspects of the cellular environment, like for example involvement of chaperones, affect these species.


Subject(s)
Apoproteins/chemistry , Flavodoxin/chemistry , Protein Folding , Azotobacter vinelandii/chemistry , Binding Sites , Kinetics , Models, Molecular , Osmolar Concentration , Protein Binding , Protein Structure, Secondary , Thermodynamics , Tryptophan/chemistry
15.
Biochemistry ; 56(42): 5679-5690, 2017 10 24.
Article in English | MEDLINE | ID: mdl-28956602

ABSTRACT

Bacterial microcompartments (BMCs) are proteinaceous organelles that encapsulate enzymes involved in CO2 fixation (carboxysomes) or carbon catabolism (metabolosomes). Metabolosomes share a common core of enzymes and a distinct signature enzyme for substrate degradation that defines the function of the BMC (e.g., propanediol or ethanolamine utilization BMCs, or glycyl-radical enzyme microcompartments). Loci encoding metabolosomes also typically contain genes for proteins that support organelle function, such as regulation, transport of substrate, and cofactor (e.g., vitamin B12) synthesis and recycling. Flavoproteins are frequently among these ancillary gene products, suggesting that these redox active proteins play an undetermined function in many metabolosomes. Here, we report the first characterization of a BMC-associated flavodoxin (Fld1C), a small flavoprotein, derived from the noncanonical 1,2-propanediol utilization BMC locus (PDU1C) of Lactobacillus reuteri. The 2.0 Å X-ray structure of Fld1C displays the α/ß flavodoxin fold, which noncovalently binds a single flavin mononucleotide molecule. Fld1C is a short-chain flavodoxin with redox potentials of -240 ± 3 mV oxidized/semiquinone and -344 ± 1 mV semiquinone/hydroquinone versus the standard hydrogen electrode at pH 7.5. It can participate in an electron transfer reaction with a photoreductant to form a stable semiquinone species. Collectively, our structural and functional results suggest that PDU1C BMCs encapsulate Fld1C to store and transfer electrons for the reactivation and/or recycling of the B12 cofactor utilized by the signature enzyme.


Subject(s)
Cobamides/chemistry , Flavin Mononucleotide/chemistry , Flavodoxin/chemistry , Limosilactobacillus reuteri/chemistry , Carbon Dioxide/chemistry , Carbon Dioxide/metabolism , Cobamides/metabolism , Flavin Mononucleotide/metabolism , Flavodoxin/metabolism , Limosilactobacillus reuteri/metabolism
16.
Biochim Biophys Acta ; 1860(5): 868-878, 2016 May.
Article in English | MEDLINE | ID: mdl-26476135

ABSTRACT

BACKGROUND: Conformational changes coupled to ligand binding constitute the structural and energetics basis underlying cooperativity, allostery and, in general, protein regulation. These conformational rearrangements are associated with heat capacity changes. ITC is a unique technique for studying binding interactions because of the simultaneous determination of the binding affinity and enthalpy, and for providing the best estimates of binding heat capacity changes. SCOPE OF REVIEW: Still controversial issues in ligand binding are the discrimination between the "conformational selection model" and the "induced fit model", and whether or not conformational changes lead to temperature dependent apparent binding heat capacities. The assessment of conformational changes associated with ligand binding by ITC is discussed. In addition, the "conformational selection" and "induced fit" models are reconciled, and discussed within the context of intrinsically (partially) unstructured proteins. MAJOR CONCLUSIONS: Conformational equilibrium is a major contribution to binding heat capacity changes. A simple model may explain both conformational selection and induced fit scenarios. A temperature-independent binding heat capacity does not necessarily indicate absence of conformational changes upon ligand binding. ITC provides information on the energetics of conformational changes associated with ligand binding (and other possible additional coupled equilibria). GENERAL SIGNIFICANCE: Preferential ligand binding to certain protein states leads to an equilibrium shift that is reflected in the coupling between ligand binding and additional equilibria. This represents the structural/energetic basis of the widespread dependence of ligand binding parameters on temperature, as well as pH, ionic strength and the concentration of other chemical species.


Subject(s)
Bacterial Proteins/chemistry , Models, Chemical , Nucleoplasmins/chemistry , Receptors, LDL/chemistry , Allosteric Regulation , Binding Sites , Flavodoxin/chemistry , HIV Protease/chemistry , Hot Temperature , Humans , Kinetics , Ligands , Protein Binding , Protein Conformation , Thermodynamics , Viral Nonstructural Proteins/chemistry
17.
PLoS Pathog ; 11(9): e1005147, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26325183

ABSTRACT

The fungal pathogen Candida albicans causes lethal systemic infections in humans. To better define how pathogens resist oxidative attack by the immune system, we examined a family of four Flavodoxin-Like Proteins (FLPs) in C. albicans. In agreement with previous studies showing that FLPs in bacteria and plants act as NAD(P)H quinone oxidoreductases, a C. albicans quadruple mutant lacking all four FLPs (pst1Δ, pst2Δ, pst3Δ, ycp4Δ) was more sensitive to benzoquinone. Interestingly, the quadruple mutant was also more sensitive to a variety of oxidants. Quinone reductase activity confers important antioxidant effects because resistance to oxidation was restored in the quadruple mutant by expressing either Escherichia coli wrbA or mammalian NQO1, two distinct types of quinone reductases. FLPs were detected at the plasma membrane in C. albicans, and the quadruple mutant was more sensitive to linolenic acid, a polyunsaturated fatty acid that can auto-oxidize and promote lipid peroxidation. These observations suggested that FLPs reduce ubiquinone (coenzyme Q), enabling it to serve as an antioxidant in the membrane. In support of this, a C. albicans coq3Δ mutant that fails to synthesize ubiquinone was also highly sensitive to oxidative stress. FLPs are critical for survival in the host, as the quadruple mutant was avirulent in a mouse model of systemic candidiasis under conditions where infection with wild type C. albicans was lethal. The quadruple mutant cells initially grew well in kidneys, the major site of C. albicans growth in mice, but then declined after the influx of neutrophils and by day 4 post-infection 33% of the mice cleared the infection. Thus, FLPs and ubiquinone are important new antioxidant mechanisms that are critical for fungal virulence. The potential of FLPs as novel targets for antifungal therapy is further underscored by their absence in mammalian cells.


Subject(s)
Candida albicans/metabolism , Flavoproteins/metabolism , Fungal Proteins/metabolism , Immune Evasion , Macrophages/microbiology , NADH, NADPH Oxidoreductases/metabolism , Oxidative Stress , Animals , Candida albicans/growth & development , Candida albicans/immunology , Candida albicans/pathogenicity , Candidiasis/immunology , Candidiasis/microbiology , Candidiasis/pathology , Cells, Cultured , Female , Flavodoxin/chemistry , Flavodoxin/metabolism , Flavoproteins/genetics , Fungal Proteins/genetics , Gene Deletion , Isoenzymes , Macrophages/immunology , Macrophages/pathology , Mice, Inbred BALB C , Microbial Viability , NADH, NADPH Oxidoreductases/genetics , Oxidation-Reduction , Substrate Specificity , Ubiquinone/chemistry , Ubiquinone/metabolism , Virulence
18.
Phys Chem Chem Phys ; 19(29): 19021-19031, 2017 Jul 26.
Article in English | MEDLINE | ID: mdl-28702545

ABSTRACT

Near the minimum free energy basin of proteins where the native ensemble resides, partly unfolded conformations of slightly higher energy can be significantly populated under native conditions. It has been speculated that they play roles in molecular recognition and catalysis, but they might represent contemporary features of the evolutionary process without functional relevance. Obtaining conclusive evidence on these alternatives is difficult because it requires comparing the performance of a given protein when populating and when not populating one such intermediate, in otherwise identical conditions. Wild type apoflavodoxin populates under native conditions a partly unfolded conformation (10% of molecules) whose unstructured region includes the binding sites for the FMN cofactor and for redox partner proteins. We recently engineered a thermostable variant where the intermediate is no longer detectable. Using the wild type and variant, we assess the relevance of the intermediate comparing folding kinetics, cofactor binding kinetics, cofactor affinity, X-ray structure, intrinsic dynamics, redox potential of the apoflavodoxin-cofactor complex (Fld), its affinity for partner protein FNR, and electron transfer rate within the Fld/FNR physiological complex. Our data strongly suggest the intermediate state, conserved in long-chain apoflavodoxins, is not required for the correct assembly of flavodoxin nor does it contribute to shape its electron transfer properties. This analysis can be applied to evaluate other native basin intermediates.


Subject(s)
Apoproteins/chemistry , Flavodoxin/chemistry , Apoproteins/genetics , Apoproteins/metabolism , Binding Sites , Calorimetry, Differential Scanning , Circular Dichroism , Crystallography, X-Ray , Electron Transport , Flavin Mononucleotide/chemistry , Flavodoxin/genetics , Flavodoxin/metabolism , Kinetics , Molecular Dynamics Simulation , Mutagenesis , Protein Folding , Protein Stability , Protein Structure, Tertiary , Temperature
19.
Biochim Biophys Acta ; 1854(10 Pt A): 1317-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26073784

ABSTRACT

Correct folding of proteins is crucial for cellular homeostasis. More than thirty percent of proteins contain one or more cofactors, but the impact of these cofactors on co-translational folding remains largely unknown. Here, we address the binding of flavin mononucleotide (FMN) to nascent flavodoxin, by generating ribosome-arrested nascent chains that expose either the entire protein or C-terminally truncated segments thereof. The native α/ß parallel fold of flavodoxin is among the most ancestral and widely distributed folds in nature and exploring its co-translational folding is thus highly relevant. In Escherichia coli (strain BL21(DE3) Δtig::kan) FMN turns out to be limiting for saturation of this flavoprotein on time-scales vastly exceeding those of flavodoxin synthesis. Because the ribosome affects protein folding, apoflavodoxin cannot bind FMN during its translation. As a result, binding of cofactor to released protein is the last step in production of this flavoprotein in the cell. We show that once apoflavodoxin is entirely synthesized and exposed outside the ribosome to which it is stalled by an artificial linker containing the SecM sequence, the protein is natively folded and capable of binding FMN.


Subject(s)
Apoproteins/chemistry , Azotobacter vinelandii/chemistry , Bacterial Proteins/chemistry , Flavin Mononucleotide/chemistry , Flavodoxin/chemistry , Ribosomes/chemistry , Apoproteins/genetics , Azotobacter vinelandii/metabolism , Bacterial Proteins/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Flavodoxin/genetics , Gene Expression , Models, Molecular , Protein Binding , Protein Biosynthesis , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Ribosomes/metabolism
20.
J Am Chem Soc ; 138(9): 2889-92, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26871608

ABSTRACT

RimO is a member of the growing radical S-adenosylmethionine (SAM) superfamily of enzymes, which use a reduced [4Fe-4S] cluster to effect reductive cleavage of the 5' C-S bond of SAM to form a 5'-deoxyadenosyl 5'-radical (5'-dA(•)) intermediate. RimO uses this potent oxidant to catalyze the attachment of a methylthio group (-SCH3) to C3 of aspartate 89 of protein S12, one of 21 proteins that compose the 30S subunit of the bacterial ribosome. However, the exact mechanism by which this transformation takes place has remained elusive. Herein, we describe the stereochemical course of the RimO reaction. Using peptide mimics of the S12 protein bearing deuterium at the 3 pro-R or 3 pro-S positions of the target aspartyl residue, we show that RimO from Bacteroides thetaiotaomicron (Bt) catalyzes abstraction of the pro-S hydrogen atom, as evidenced by the transfer of deuterium into 5'-deoxyadenosine (5'-dAH). The observed kinetic isotope effect on H atom versus D atom abstraction is ∼1.9, suggesting that this step is at least partially rate determining. We also demonstrate that Bt RimO can utilize the flavodoxin/flavodoxin oxidoreductase/NADPH reducing system from Escherichia coli as a source of requisite electrons. Use of this in vivo reducing system decreases, but does not eliminate, formation of 5'-dAH in excess of methylthiolated product.


Subject(s)
Sulfurtransferases/chemistry , Sulfurtransferases/metabolism , Catalysis , Escherichia coli/enzymology , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Flavodoxin/chemistry , Flavodoxin/metabolism , Kinetics , Models, Molecular , NADH, NADPH Oxidoreductases/chemistry , NADH, NADPH Oxidoreductases/metabolism , Nuclear Magnetic Resonance, Biomolecular , S-Adenosylmethionine/chemistry , S-Adenosylmethionine/metabolism , Stereoisomerism , Thermotoga maritima/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL