Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 273
Filter
1.
Protein Expr Purif ; 219: 106487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38657915

ABSTRACT

The bacterial Efe system functions as an importer of free Fe2+ into cells independently of iron-chelating compounds such as siderophores and consisted of iron-binding protein EfeO, peroxidase EfeB, and transmembrane permease EfeU. While we and other researchers reported crystal structures of EfeO and EfeB, that of EfeU remains undetermined. In this study, we constructed expression system of EfeU derived from Escherichia coli, selected E. coli Rosetta-gami 2 (DE3) as an expression host, and succeeded in purification of the proteins which were indicated to form an oligomer by blue native PAGE. We obtained preliminary data of the X-ray crystallography, suggesting that expression and purification methods we established in this study enable structural analysis of the bacterial Efe system.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Iron , Escherichia coli/genetics , Escherichia coli/metabolism , Crystallography, X-Ray , Escherichia coli Proteins/genetics , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli Proteins/isolation & purification , Iron/metabolism , Iron/chemistry , Gene Expression , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Recombinant Proteins/biosynthesis , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/isolation & purification , Iron-Binding Proteins/metabolism
2.
J Mol Biol ; 436(10): 168555, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38552947

ABSTRACT

The funneled energy landscape theory suggests that the folding pathway of homologous proteins should converge at the late stages of folding. In this respect, proteins displaying a broad energy landscape for folding are particularly instructive, allowing inferring both the early, intermediate and late stages of folding. In this paper we explore the folding mechanisms of human frataxin, an essential mitochondrial protein linked to the neurodegenerative disorder Friedreich's ataxia. Building upon previous studies on the yeast homologue, the folding pathway of human frataxin is thoroughly examined, revealing a mechanism implying the presence of a broad energy barrier, reminiscent of the yeast counterpart. Through an extensive site-directed mutagenesis, we employed a Φ -value analysis to map native-like contacts in the folding transition state. The presence of a broad energy barrier facilitated the exploration of such contacts in both early and late folding events. We compared results from yeast and human frataxin providing insights into the impact of native topology on the folding mechanism and elucidating the properties of the underlying free energy landscape. The findings are discussed in the context of the funneled energy landscape theory of protein folding.


Subject(s)
Frataxin , Iron-Binding Proteins , Protein Folding , Saccharomyces cerevisiae , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Iron-Binding Proteins/genetics , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae/genetics , Thermodynamics , Models, Molecular , Protein Conformation , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/genetics , Mutagenesis, Site-Directed , Kinetics
3.
ACS Chem Biol ; 18(7): 1534-1547, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37410592

ABSTRACT

The mitochondrial cysteine desulfurase NFS1 is an essential PLP-dependent enzyme involved in iron-sulfur cluster assembly. The enzyme catalyzes the desulfurization of the l-Cys substrate, producing a persulfide and l-Ala as products. In this study, we set the measurement of the product l-Ala by NMR in vitro by means of 1H NMR spectra acquisition. This methodology provided us with the possibility of monitoring the reaction in both fixed-time and real-time experiments, with high sensitivity and accuracy. By studying I452A, W454A, Q456A, and H457A NFS1 variants, we found that the C-terminal stretch (CTS) of the enzyme is critical for function. Specifically, mutation of the extremely conserved position W454 resulted in highly decreased activity. Additionally, we worked on two singular variants: "GGG" and C158A. In the former, the catalytic Cys-loop was altered by including two Gly residues to increase the flexibility of this loop. This variant had significantly impaired activity, indicating that the Cys-loop motions are fine-tuned in the wild-type enzyme. In turn, for C158A, we found an unanticipated increase in l-Cys desulfurase activity. Furthermore, we carried out molecular dynamics simulations of the supercomplex dedicated to iron-sulfur cluster biosynthesis, which includes NFS1, ACP, ISD11, ISCU2, and FXN subunits. We identified CTS as a key element that established interactions with ISCU2 and FXN concurrently; we found specific interactions that are established when FXN is present, reinforcing the idea that FXN not only forms part of the iron-sulfur cluster assembly site but also modulates the internal motions of ISCU2.


Subject(s)
Iron-Sulfur Proteins , Humans , Iron-Sulfur Proteins/chemistry , Carbon-Sulfur Lyases/metabolism , Sulfur/chemistry , Iron/chemistry , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics
4.
Acta Crystallogr D Struct Biol ; 79(Pt 1): 22-30, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36601804

ABSTRACT

Friedreich's ataxia (FRDA) is a hereditary cardiodegenerative and neurodegenerative disease that affects 1 in 50 000 Americans. FRDA arises from either a cellular inability to produce sufficient quantities or the production of a nonfunctional form of the protein frataxin, a key molecule associated with mitochondrial iron-sulfur cluster biosynthesis. Within the mitochondrial iron-sulfur cluster (ISC) assembly pathway, frataxin serves as an allosteric regulator for cysteine desulfurase, the enzyme that provides sulfur for [2Fe-2S] cluster assembly. Frataxin is a known iron-binding protein and is also linked to the delivery of ferrous ions to the scaffold protein, the ISC molecule responsible for the direct assembly of [2Fe-2S] clusters. The goal of this report is to provide structural details of the Drosophila melanogaster frataxin ortholog (Dfh), using both X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, in order to provide the foundational insight needed to understand the structure-function correlation of the protein. Additionally, NMR iron(II) titrations were used to provide metal contacts on the protein to better understand how it binds iron and aids its delivery to the ISC scaffold protein. Here, the structural and functional similarities of Dfh to its orthologs are also outlined. Structural data show that bacterial, yeast, human and Drosophila frataxins are structurally similar, apart from a structured C-terminus in Dfh that is likely to aid in protein stability. The iron-binding location on helix 1 and strand 1 of Dfh is also conserved across orthologs.


Subject(s)
Drosophila melanogaster , Neurodegenerative Diseases , Animals , Humans , Drosophila melanogaster/metabolism , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Iron/metabolism , Sulfur/metabolism , Frataxin
5.
Biotechnol Bioeng ; 120(2): 409-425, 2023 02.
Article in English | MEDLINE | ID: mdl-36225115

ABSTRACT

Frataxin is a kinetic activator of the mitochondrial supercomplex for iron-sulfur cluster assembly. Low frataxin expression or a decrease in its functionality results in Friedreich's Ataxia (FRDA). With the aim of creating new molecular tools to study this metabolic pathway, and ultimately, to explore new therapeutic strategies, we have investigated the possibility of obtaining small proteins exhibiting a high affinity for frataxin. In this study, we applied the ribosome display approach, using human frataxin as the target. We focused on Affi_224, one of the proteins that we were able to select after five rounds of selection. We have studied the interaction between both proteins and discussed some applications of this specific molecular tutor, concerning the modulation of the supercomplex activity. Affi_224 and frataxin showed a KD value in the nanomolar range, as judged by surface plasmon resonance analysis. Most likely, it binds to the frataxin acidic ridge, as suggested by the analysis of chemical shift perturbations (nuclear magnetic resonance) and computational simulations. Affi_224 was able to increase Cys NFS1 desulfurase activation exerted by the FRDA frataxin variant G130V. Importantly, Affi_224 interacts with frataxin in a human cellular model. Our results suggest quaternary addition may be a new tool to modulate frataxin function in vivo. Nevertheless, more functional experiments under physiological conditions should be carried out to evaluate Affi_224 effectiveness in FRDA cell models.


Subject(s)
Carbon-Sulfur Lyases , Iron-Binding Proteins , Humans , Iron-Binding Proteins/genetics , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/metabolism , Frataxin
6.
Int J Mol Sci ; 23(21)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36361939

ABSTRACT

Friedreich's ataxia is a neurodegenerative disease caused by mutations in the frataxin gene. Frataxin homologues, including bacterial CyaY proteins, can be found in most species and play a fundamental role in mitochondrial iron homeostasis, either promoting iron assembly into metaloproteins or contributing to iron detoxification. While several lines of evidence suggest that eukaryotic frataxins are more effective than bacterial ones in iron detoxification, the residues involved in this gain of function are unknown. In this work, we analyze conservation of amino acid sequence and protein structure among frataxins and CyaY proteins to identify four highly conserved residue clusters and group them into potential functional clusters. Clusters 1, 2, and 4 are present in eukaryotic frataxins and bacterial CyaY proteins. Cluster 3, containing two serines, a tyrosine, and a glutamate, is only present in eukaryotic frataxins and on CyaY proteins from the Rickettsia genus. Residues from cluster 3 are blocking a small cavity of about 40 Å present in E. coli's CyaY. The function of this cluster is unknown, but we hypothesize that its tyrosine may contribute to prevent formation of reactive oxygen species during iron detoxification. This cluster provides an example of gain of function during evolution in a protein involved in iron homeostasis, as our results suggests that Cluster 3 was present in the endosymbiont ancestor of mitochondria and was conserved in eukaryotic frataxins.


Subject(s)
Iron-Binding Proteins , Neurodegenerative Diseases , Rickettsia , Humans , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Eukaryota/metabolism , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Iron/metabolism , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Rickettsia/metabolism , Tyrosine/metabolism , Mitochondria/metabolism , Mitochondria/microbiology , Frataxin
7.
J Biol Chem ; 298(4): 101808, 2022 04.
Article in English | MEDLINE | ID: mdl-35271852

ABSTRACT

Iron is an essential element for nearly all organisms, and under anoxic and/or reducing conditions, Fe2+ is the dominant form of iron available to bacteria. The ferrous iron transport (Feo) system is the primary prokaryotic Fe2+ import machinery, and two constituent proteins (FeoA and FeoB) are conserved across most bacterial species. However, how FeoA and FeoB function relative to one another remains enigmatic. In this work, we explored the distribution of feoAB operons encoding a fusion of FeoA tethered to the N-terminal, G-protein domain of FeoB via a connecting linker region. We hypothesized that this fusion poises FeoA to interact with FeoB to affect function. To test this hypothesis, we characterized the soluble NFeoAB fusion protein from Bacteroides fragilis, a commensal organism implicated in drug-resistant infections. Using X-ray crystallography, we determined the 1.50-Å resolution structure of BfFeoA, which adopts an SH3-like fold implicated in protein-protein interactions. Using a combination of structural modeling, small-angle X-ray scattering, and hydrogen-deuterium exchange mass spectrometry, we show that FeoA and NFeoB interact in a nucleotide-dependent manner, and we mapped the protein-protein interaction interface. Finally, using guanosine triphosphate (GTP) hydrolysis assays, we demonstrate that BfNFeoAB exhibits one of the slowest known rates of Feo-mediated GTP hydrolysis that is not potassium-stimulated. Importantly, truncation of FeoA from this fusion demonstrates that FeoA-NFeoB interactions function to stabilize the GTP-bound form of FeoB. Taken together, our work reveals a role for FeoA function in the fused FeoAB system and suggests a function for FeoA among prokaryotes.


Subject(s)
Bacterial Proteins , Bacteroides fragilis , Cation Transport Proteins , Iron-Binding Proteins , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacteroides fragilis/genetics , Bacteroides fragilis/metabolism , Cation Transport Proteins/metabolism , Crystallography, X-Ray , Guanosine Triphosphate/chemistry , Guanosine Triphosphate/metabolism , Hydrolysis , Iron/metabolism , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Protein Stability
8.
Arch Biochem Biophys ; 715: 109086, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34801473

ABSTRACT

In humans, the loss of frataxin results in Friedreich's Ataxia, a neurodegenerative disease, in which a deficit in the iron-sulfur cluster assembly is observed. In this work, we analyzed three frataxin variants in which one tryptophan was replaced by a glycine: W155G, W168G and W173G. As expected, given its localization in the assembly site, W155G was not able to activate the desulfurase activity of the supercomplex for iron-sulfur cluster assembly. In turn, W168G, which was significantly more unstable than W155G, was fully active. W173G, which was highly unstable as W168G, showed a significantly decreased activity, only slightly higher than W155G. As W168G and W173G were highly sensitive to proteolysis, we investigated the protein motions by molecular dynamic simulations. We observed that W173G may display altered motions at the Trp155 site. Furthermore, we revealed a H-bond network in which Trp155 takes part, involving residues Gln148, Asn151, Gln153 and Arg165. We suggest that this motion modulation that specifically alters the population of different Trp155 rotamers can be directly transferred to the assembly site, altering the dynamics of the ISCU His137 key residue. This hypothesis was also contrasted by means of molecular dynamic simulations of frataxin in the context of the complete supercomplex. We propose that the supercomplex requires very definite motions of Trp155 to consolidate the assembly site.


Subject(s)
Iron-Binding Proteins/chemistry , Tryptophan/chemistry , Humans , Iron-Binding Proteins/genetics , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Mutation , Protein Conformation , Protein Stability , Frataxin
9.
Int J Mol Sci ; 22(23)2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34884815

ABSTRACT

BACKGROUND: New strategies are needed to combat multidrug-resistant bacteria. The restriction of iron uptake by bacteria is a promising way to inhibit their growth. We aimed to suppress the growth of Vibrio bacterial species by inhibiting their ferric ion-binding protein (FbpA) using food components. METHODS: Twenty spices were selected for the screening of FbpA inhibitors. The candidate was applied to antibacterial tests, and the mechanism was further studied. RESULTS: An active compound, rosmarinic acid (RA), was screened out. RA binds competitively and more tightly than Fe3+ to VmFbpA, the FbpA from V. metschnikovii, with apparent KD values of 8 µM vs. 17 µM. Moreover, RA can inhibit the growth of V. metschnikovii to one-third of the control at 1000 µM. Interestingly, sodium citrate (SC) enhances the growth inhibition effect of RA, although SC only does not inhibit the growth. The combination of RA/SC completely inhibits the growth of not only V. metschnikovii at 100/100 µM but also the vibriosis-causative pathogens V. vulnificus and V. parahaemolyticus, at 100/100 and 1000/100 µM, respectively. However, RA/SC does not affect the growth of Escherichia coli. CONCLUSIONS: RA/SC is a potential bacteriostatic agent against Vibrio species while causing little damage to indigenous gastrointestinal bacteria.


Subject(s)
Cinnamates/pharmacology , Depsides/pharmacology , Iron/metabolism , Sodium Citrate/pharmacology , Vibrio parahaemolyticus/drug effects , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cinnamates/chemistry , Cinnamates/metabolism , Depsides/chemistry , Depsides/metabolism , Drug Synergism , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Molecular Docking Simulation , Plant Extracts/chemistry , Protein Binding , Vibrio parahaemolyticus/metabolism , Rosmarinic Acid
10.
Int J Mol Sci ; 22(10)2021 May 12.
Article in English | MEDLINE | ID: mdl-34065957

ABSTRACT

The presented research concerns the triple activity of trans-cinnamic (tCA), ferulic (FA) and syringic acids (SA). They act as thyroid peroxidase (TPO) activators, lipoxygenase (LOX) inhibitors and show antiradical activity. All compounds showed a dose-dependent TPO activatory effect, thus the AC50 value (the concentration resulting in 50% activation) was determined. The tested compounds can be ranked as follows: tCA > FA > SA with AC50 = 0.10, 0.39, 0.69 mM, respectively. Strong synergism was found between FA and SA. The activatory effects of all tested compounds may result from interaction with the TPO allosteric site. It was proposed that conformational change resulting from activator binding to TPO allosteric pocket results from the flexibility of a nearby loop formed by residues Val352-Tyr363. All compounds act as uncompetitive LOX inhibitors. The most effective were tCA and SA, whereas the weakest was FA (IC50 = 0.009 mM and IC50 0.027 mM, respectively). In all cases, an interaction between the inhibitors carboxylic groups and side-chain atoms of Arg102 and Arg139 in an allosteric pocket of LOX was suggested. FA/tCA and FA/SA acted synergistically, whereas tCA/SA demonstrated antagonism. The highest antiradical activity was found in the case of SA (IC50 = 0.22 mM). FA/tCA and tCA/SA acted synergistically, whereas antagonism was found for the SA/FA mixture.


Subject(s)
Autoantigens/metabolism , Enzyme Activators/pharmacology , Iodide Peroxidase/metabolism , Iron-Binding Proteins/metabolism , Lipoxygenase Inhibitors/pharmacology , Phytochemicals/pharmacology , Protein-Lysine 6-Oxidase/metabolism , Autoantigens/chemistry , Cinnamates/chemistry , Cinnamates/pharmacology , Coumaric Acids/chemistry , Coumaric Acids/pharmacology , Dose-Response Relationship, Drug , Enzyme Activators/chemistry , Gallic Acid/analogs & derivatives , Gallic Acid/chemistry , Gallic Acid/pharmacology , Humans , Inhibitory Concentration 50 , Iodide Peroxidase/chemistry , Iron-Binding Proteins/chemistry , Lipoxygenase Inhibitors/chemistry , Models, Molecular , Phytochemicals/chemistry , Protein-Lysine 6-Oxidase/chemistry , Structure-Activity Relationship
11.
IUBMB Life ; 73(3): 543-553, 2021 03.
Article in English | MEDLINE | ID: mdl-33675183

ABSTRACT

Friedreich Ataxia is a neuro-cardiodegenerative disease caused by the deficiency of frataxin, a mitochondrial protein. Many evidences indicate that frataxin deficiency causes an unbalance of iron homeostasis. Nevertheless, in the last decade many results also highlighted the importance of calcium unbalance in the deleterious downstream effects caused by frataxin deficiency. In this review, the role of these two metals has been gathered to give a whole view of how iron and calcium dyshomeostasys impacts on cellular functions and, as a result, which strategies can be followed to find an effective therapy for the disease.


Subject(s)
Calcium/metabolism , Friedreich Ataxia/metabolism , Iron-Binding Proteins/metabolism , Iron/metabolism , Mitochondria/metabolism , Friedreich Ataxia/drug therapy , Friedreich Ataxia/physiopathology , Homeostasis , Humans , Iron Chelating Agents/pharmacology , Iron-Binding Proteins/chemistry , Frataxin
12.
Int J Mol Sci ; 21(24)2020 Dec 17.
Article in English | MEDLINE | ID: mdl-33348670

ABSTRACT

Frataxin is a highly conserved protein whose deficiency results in the neurodegenerative disease Friederich's ataxia. Frataxin's actual physiological function has been debated for a long time without reaching a general agreement; however, it is commonly accepted that the protein is involved in the biosynthetic iron-sulphur cluster (ISC) machinery, and several authors have pointed out that it also participates in iron homeostasis. In this work, we use site-directed spin labeling coupled to electron paramagnetic resonance (SDSL EPR) to add new information on the effects of ferric and ferrous iron binding on the properties of human frataxin in vitro. Using SDSL EPR and relating the results to fluorescence experiments commonly performed to study iron binding to FXN, we produced evidence that ferric iron causes reversible aggregation without preferred interfaces in a concentration-dependent fashion, starting at relatively low concentrations (micromolar range), whereas ferrous iron binds without inducing aggregation. Moreover, our experiments show that the ferrous binding does not lead to changes of protein conformation. The data reported in this study reveal that the currently reported binding stoichiometries should be taken with caution. The use of a spin label resistant to reduction, as well as the comparison of the binding effect of Fe2+ in wild type and in the pathological D122Y variant of frataxin, allowed us to characterize the Fe2+ binding properties of different protein sites and highlight the effect of the D122Y substitution on the surrounding residues. We suggest that both Fe2+ and Fe3+ might play a relevant role in the context of the proposed FXN physiological functions.


Subject(s)
Circular Dichroism/methods , Ferric Compounds/chemistry , Ferrous Compounds/chemistry , Iron-Binding Proteins/chemistry , Iron/chemistry , Spin Labels , Electron Spin Resonance Spectroscopy/methods , Humans , Hydrogen-Ion Concentration , Protein Aggregation, Pathological , Protein Binding , Protein Conformation , Spectrometry, Fluorescence/methods , Frataxin
13.
PLoS One ; 15(11): e0242206, 2020.
Article in English | MEDLINE | ID: mdl-33196673

ABSTRACT

Artemin is an abundant thermostable protein in Artemia embryos and it is considered as a highly efficient molecular chaperone against extreme environmental stress conditions. The conformational dynamics of artemin have been suggested to play a critical role in its biological functions. In this study, we have investigated the conformational and functional changes of artemin under heat and oxidative stresses to identify the relationship between its structure and function. The tertiary and quaternary structures of artemin were evaluated by fluorescence measurements, protein cross-linking analysis, and dynamic light scattering. Based on the structural analysis, artemin showed irreversible substantial conformational lability in responses to heat and oxidant, which was mainly mediated through the hydrophobic interactions and dimerization of the chaperone. In addition, the chaperone-like activity of heated and oxidized artemin was examined using lysozyme refolding assay and the results showed that although both factors, i.e. heat and oxidant, at specific levels improved artemin potency, simultaneous incubation with both stressors significantly triggered the chaperone activation. Moreover, the heat-induced dimerization of artemin was found to be the most critical factor for its activation. It was suggested that oxidation presumably acts through stabilizing the dimer structures of artemin through formation of disulfide bridges between the subunits and strengthens its chaperoning efficacy. Accordingly, it is proposed that artemin probably exists in a monomer-oligomer equilibrium in Artemia cysts and environmental stresses and intracellular portion of protein substrates may shift the equilibrium towards the active dimer forms of the chaperone.


Subject(s)
Arthropod Proteins/chemistry , Iron-Binding Proteins/chemistry , Oxidants/chemistry , RNA-Binding Proteins/chemistry , Animals , Artemia/metabolism , Arthropod Proteins/genetics , Arthropod Proteins/metabolism , Dimerization , Electrophoresis, Polyacrylamide Gel , Hydrogen Peroxide/chemistry , Hydrophobic and Hydrophilic Interactions , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Protein Structure, Tertiary , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Spectrometry, Fluorescence , Temperature
14.
Int J Mol Sci ; 21(18)2020 Sep 17.
Article in English | MEDLINE | ID: mdl-32957566

ABSTRACT

Several biological activities depend on iron-sulfur clusters ([Fe-S]). Even though they are well-known in several organisms their function and metabolic pathway were poorly understood in the majority of the organisms. We propose to use the amoeba Dictyostelium discoideum, as a biological model to study the biosynthesis of [Fe-S] at the molecular, cellular and organism levels. First, we have explored the D. discoideum genome looking for genes corresponding to the subunits that constitute the molecular machinery for Fe-S cluster assembly and, based on the structure of the mammalian supercomplex and amino acid conservation profiles, we inferred the full functionality of the amoeba machinery. After that, we expressed the recombinant mature form of D. discoideum frataxin protein (DdFXN), the kinetic activator of this pathway. We characterized the protein and its conformational stability. DdFXN is monomeric and compact. The analysis of the secondary structure content, calculated using the far-UV CD spectra, was compatible with the data expected for the FXN fold, and near-UV CD spectra were compatible with the data corresponding to a folded protein. In addition, Tryptophan fluorescence indicated that the emission occurs from an apolar environment. However, the conformation of DdFXN is significantly less stable than that of the human FXN, (4.0 vs. 9.0 kcal mol-1, respectively). Based on a sequence analysis and structural models of DdFXN, we investigated key residues involved in the interaction of DdFXN with the supercomplex and the effect of point mutations on the energetics of the DdFXN tertiary structure. More than 10 residues involved in Friedreich's Ataxia are conserved between the human and DdFXN forms, and a good correlation between mutational effect on the energetics of both proteins were found, suggesting the existence of similar sequence/function/stability relationships. Finally, we integrated this information in an evolutionary context which highlights particular variation patterns between amoeba and humans that may reflect a functional importance of specific protein positions. Moreover, the complete pathway obtained forms a piece of evidence in favor of the hypothesis of a shared and highly conserved [Fe-S] assembly machinery between Human and D. discoideum.


Subject(s)
Dictyostelium/metabolism , Friedreich Ataxia/genetics , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Iron-Sulfur Proteins/metabolism , Amino Acid Sequence/genetics , Chromatography, High Pressure Liquid , Circular Dichroism , Computational Biology , Crystallography , Dictyostelium/genetics , Humans , Iron-Binding Proteins/genetics , Iron-Sulfur Proteins/biosynthesis , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/genetics , Kinetics , Molecular Dynamics Simulation , Phylogeny , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins , Sequence Alignment , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Frataxin
15.
Arch Biochem Biophys ; 691: 108491, 2020 09 30.
Article in English | MEDLINE | ID: mdl-32707090

ABSTRACT

The relationships between conformational dynamics, stability and protein function are not obvious. Frataxin (FXN) is an essential protein that forms part of a supercomplex dedicated to the iron-sulfur (Fe-S) cluster assembly within the mitochondrial matrix. In humans, the loss of FXN expression or a decrease in its functionality results in Friedreich's Ataxia, a cardio-neurodegenerative disease. Recently, the way in which FXN interacts with the rest of the subunits of the supercomplex was uncovered. This opens a window to explore relationships between structural dynamics and function. In this study, we prepared a set of FXN variants spanning a broad range of conformational stabilities. Variants S160I, S160M and A204R were more stable than the wild-type and showed similar biological activity. Additionally, we prepared SILCAR, a variant that combines S160I, L203C and A204R mutations. SILCAR was 2.4 kcal mol-1 more stable and equally active. Some of the variants were significantly more resistant to proteolysis than the wild-type FXN. SILCAR showed the highest resistance, suggesting a more rigid structure. It was corroborated by means of molecular dynamics simulations. Relaxation dispersion NMR experiments comparing SILCAR and wild-type variants suggested similar internal motions in the microsecond to millisecond timescale. Instead, variant S157I showed higher denaturation resistance but a significant lower function, similarly to that observed for the FRDA variant N146K. We concluded that the contribution of particular side chains to the conformational stability of FXN might be highly subordinated to their impact on both the protein function and the stability of the functional supercomplex.


Subject(s)
Iron-Binding Proteins/chemistry , Carbon-Sulfur Lyases/chemistry , Computational Biology , Humans , Iron-Binding Proteins/genetics , Molecular Dynamics Simulation , Point Mutation , Protein Conformation , Protein Engineering , Protein Stability , Proteolysis , Frataxin
16.
Immunol Lett ; 220: 71-78, 2020 04.
Article in English | MEDLINE | ID: mdl-32027873

ABSTRACT

BACKGROUND: Human proteins such as interleukin-24 (IL24), thyroperoxidase (TPO) and thyroglobulin (Tg) are targets of IgE or IgG autoantibodies. Why these proteins are recognized by autoantibodies in some patients with chronic spontaneous urticaria (CSU) or hypothyroidism is unknown. OBJECTIVE: Through in silico analysis, identify antigen patches of TPO, Tg and IL24 and compare the sequences of these human proteins with some prevalent allergens. METHODS: The amino acids sequences of IL24, thyroperoxidase and thyroglobulin were compared between them and with 22 environmental allergens. Phylogenetic studies and multiple pairing were carried out to explore the degree of protein identity and cover. The proteins without 3D structure reported in the database, were modeled by homology with "Swiss Modeller" and compared through PYMOL. Residues conserved and accessible to the solvent (rASA> 0.25) were located in the 3D model to identify possible areas of cross-reactivity and antigen binding. RESULTS: We build a 3D model of the TPO and thyroglobulin protein base on proteins closely related. Five epitopes for TPO, six for IL24 and six for thyroglobulin were predicted. The amino acid sequences of allergens from different sources (Dermatophagoides pteronyssinus, Blomia tropicalis, Betula verrucosa, Cynodon dactylon, Aspergillus fumigatus, Canis domesticus, Felis domesticus) were compared with human TPO, Tg and IL24. The cover and alignments between allergens and human proteins were low. CONCLUSION: We identify possible linear and conformational epitopes of TPO, Tg and IL24 that could be the target of IgE or IgG binding in patients with urticaria or hypothyroidism; These epitopes do not appear to be present among common environmental allergens, suggesting that autoreactivity to these human proteins are not by cross-reactivity.


Subject(s)
Allergens/immunology , Autoantigens/immunology , Chronic Urticaria/immunology , Epitopes/immunology , Hypothyroidism/immunology , Interleukins/immunology , Iodide Peroxidase/immunology , Iron-Binding Proteins/immunology , Thyroglobulin/immunology , Animals , Aspergillus fumigatus/immunology , Autoantibodies/immunology , Autoantigens/chemistry , Autoantigens/classification , Cats , Cross Reactions , Dogs , Epitope Mapping , Epitopes/chemistry , Epitopes/classification , Humans , Interleukins/chemistry , Interleukins/classification , Iodide Peroxidase/chemistry , Iodide Peroxidase/classification , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/classification , Models, Chemical , Phylogeny , Thyroglobulin/chemistry , Thyroglobulin/classification
17.
Curr Opin Chem Biol ; 55: 34-44, 2020 04.
Article in English | MEDLINE | ID: mdl-31918395

ABSTRACT

The recently solved crystal structures of the human cysteine desulfurase NFS1, in complex with the LYR protein ISD11, the acyl carrier protein ACP, and the main scaffold ISCU, have shed light on the molecular interactions that govern initial cluster assembly on ISCU. Here, we aim to highlight recent insights into iron-sulfur (Fe-S) cluster (ISC) biogenesis in mammalian cells that have arisen from the crystal structures of the core ISC assembly complex. We will also discuss how ISCs are delivered to recipient proteins and the challenges that remain in dissecting the pathways that deliver clusters to numerous Fe-S recipient proteins in both the mitochondrial matrix and cytosolic compartments of mammalian cells.


Subject(s)
Acyl Carrier Protein/chemistry , Adenosine Triphosphatases/metabolism , Iron-Binding Proteins/chemistry , Iron/chemistry , Sulfur/chemistry , Acyl Carrier Protein/metabolism , Amino Acid Sequence , Carbon-Sulfur Lyases/chemistry , Computational Biology , Crystallization , Cytosol/metabolism , Cytosol/ultrastructure , Humans , Iron-Binding Proteins/metabolism , Iron-Regulatory Proteins/chemistry , Mitochondria/metabolism , Mitochondria/ultrastructure , Models, Molecular , Protein Binding , Protein Conformation , Frataxin
18.
Annu Rev Biochem ; 89: 471-499, 2020 06 20.
Article in English | MEDLINE | ID: mdl-31935115

ABSTRACT

Mitochondria are essential in most eukaryotes and are involved in numerous biological functions including ATP production, cofactor biosyntheses, apoptosis, lipid synthesis, and steroid metabolism. Work over the past two decades has uncovered the biogenesis of cellular iron-sulfur (Fe/S) proteins as the essential and minimal function of mitochondria. This process is catalyzed by the bacteria-derived iron-sulfur cluster assembly (ISC) machinery and has been dissected into three major steps: de novo synthesis of a [2Fe-2S] cluster on a scaffold protein; Hsp70 chaperone-mediated trafficking of the cluster and insertion into [2Fe-2S] target apoproteins; and catalytic conversion of the [2Fe-2S] into a [4Fe-4S] cluster and subsequent insertion into recipient apoproteins. ISC components of the first two steps are also required for biogenesis of numerous essential cytosolic and nuclear Fe/S proteins, explaining the essentiality of mitochondria. This review summarizes the molecular mechanisms underlying the ISC protein-mediated maturation of mitochondrial Fe/S proteins and the importance for human disease.


Subject(s)
Friedreich Ataxia/genetics , Iron-Sulfur Proteins/genetics , Mitochondria/genetics , Mitochondrial Diseases/genetics , Mitochondrial Proteins/genetics , Molecular Chaperones/genetics , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Carbon-Sulfur Lyases/chemistry , Carbon-Sulfur Lyases/genetics , Carbon-Sulfur Lyases/metabolism , Ferredoxins/chemistry , Ferredoxins/genetics , Ferredoxins/metabolism , Friedreich Ataxia/metabolism , Friedreich Ataxia/pathology , Gene Expression Regulation , Glutaredoxins/chemistry , Glutaredoxins/genetics , Glutaredoxins/metabolism , Humans , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Iron-Sulfur Proteins/metabolism , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Biosynthesis , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Frataxin
19.
Biochemistry ; 58(49): 4935-4949, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31713418

ABSTRACT

The acquisition of iron is essential to establishing virulence among most pathogens. Under acidic and/or anaerobic conditions, most bacteria utilize the widely distributed ferrous iron (Fe2+) uptake (Feo) system to import metabolically-required iron. The Feo system is inadequately understood at the atomic, molecular, and mechanistic levels, but we do know it is composed of a main membrane component (FeoB) essential for iron translocation, as well as two small, cytosolic proteins (FeoA and FeoC) hypothesized to function as accessories to this process. FeoC has many hypothetical functions, including that of an iron-responsive transcriptional regulator. Here, we demonstrate for the first time that Escherichia coli FeoC (EcFeoC) binds an [Fe-S] cluster. Using electronic absorption, X-ray absorption, and electron paramagnetic resonance spectroscopies, we extensively characterize the nature of this cluster. Under strictly anaerobic conditions after chemical reconstitution, we demonstrate that EcFeoC binds a redox-active [4Fe-4S]2+/+ cluster that is rapidly oxygen-sensitive and decays to a [2Fe-2S]2+ cluster (t1/2 ≈ 20 s), similar to the [Fe-S] cluster in the fumarate and nitrate reductase (FNR) transcriptional regulator. We further show that this behavior is nearly identical to the homologous K. pneumoniae FeoC, suggesting a redox-active, oxygen-sensitive [4Fe-4S]2+ cofactor is a general phenomenon of cluster-binding FeoCs. Finally, in contrast to FNR, we show that the [4Fe-4S]2+ cluster binding to FeoC is associated with modest conformational changes of the polypeptide, but not protein dimerization. We thus posit a working hypothesis in which the cluster-binding FeoCs may function as oxygen-sensitive iron sensors that fine-tune pathogenic ferrous iron acquisition.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Iron-Binding Proteins/chemistry , Iron-Binding Proteins/metabolism , Iron-Sulfur Proteins/chemistry , Oxygen/metabolism , Repressor Proteins/chemistry , Repressor Proteins/metabolism , Catalytic Domain , Electron Spin Resonance Spectroscopy , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Iron/chemistry , Iron/metabolism , Iron-Binding Proteins/genetics , Iron-Sulfur Proteins/genetics , Iron-Sulfur Proteins/metabolism , Kinetics , Oxidation-Reduction , Oxygen/chemistry , Repressor Proteins/genetics , Sulfur/chemistry , Sulfur/metabolism
20.
J Bacteriol ; 202(1)2019 12 06.
Article in English | MEDLINE | ID: mdl-31611288

ABSTRACT

The obligate intracellular pathogen Chlamydia trachomatis is a globally significant cause of sexually transmitted bacterial infections and the leading etiological agent of preventable blindness. The first-row transition metal iron (Fe) plays critical roles in chlamydial cell biology, and acquisition of this nutrient is essential for the survival and virulence of the pathogen. Nevertheless, how C. trachomatis acquires Fe from host cells is not well understood, since it lacks genes encoding known siderophore biosynthetic pathways, receptors for host Fe storage proteins, and the Fe acquisition machinery common to many bacteria. Recent studies have suggested that C. trachomatis directly acquires host Fe via the ATP-binding cassette permease YtgABCD. Here, we characterized YtgA, the periplasmic solute binding protein component of the transport pathway, which has been implicated in scavenging Fe(III) ions. The structure of Fe(III)-bound YtgA was determined at 2.0-Å resolution with the bound ion coordinated via a novel geometry (3 Ns, 2 Os [3N2O]). This unusual coordination suggested a highly plastic metal binding site in YtgA capable of interacting with other cations. Biochemical analyses showed that the metal binding site of YtgA was not restricted to interaction with only Fe(III) ions but could bind all transition metal ions examined. However, only Mn(II), Fe(II), and Ni(II) ions bound reversibly to YtgA, with Fe being the most abundant cellular transition metal in C. trachomatis Collectively, these findings show that YtgA is the metal-recruiting component of the YtgABCD permease and is most likely involved in the acquisition of Fe(II) and Mn(II) from host cells.IMPORTANCEChlamydia trachomatis is the most common bacterial sexually transmitted infection in developed countries, with an estimated global prevalence of 4.2% in the 15- to 49-year age group. Although infection is asymptomatic in more than 80% of infected women, about 10% of cases result in serious disease. Infection by C. trachomatis is dependent on the ability to acquire essential nutrients, such as the transition metal iron, from host cells. In this study, we show that iron is the most abundant transition metal in C. trachomatis and report the structural and biochemical properties of the iron-recruiting protein YtgA. Knowledge of the high-resolution structure of YtgA will provide a platform for future structure-based antimicrobial design approaches.


Subject(s)
Antigens, Bacterial/chemistry , Iron-Binding Proteins/chemistry , Iron/metabolism , Antigens, Bacterial/metabolism , Binding Sites , Iron-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...