Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Phys Chem B ; 128(22): 5336-5343, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38780400

RESUMO

Copper, an essential metal for various cellular processes, requires tight regulation to prevent cytotoxicity. Intracellular pathways crucial for maintaining optimal copper levels involve soluble and membrane transporters, namely, metallochaperones and P-type ATPases, respectively. In this study, we used a simulation workflow based on free-energy perturbation (FEP) theory and parallel bias metadynamics (PBMetaD) to predict the Cu(I) exchange mechanism between the human Cu(I) chaperone, Atox1, and one of its two physiological partners, ATP7A. ATP7A, also known as the Menkes disease protein, is a transmembrane protein and one of the main copper-transporting ATPases. It pumps copper into the trans-Golgi network for the maturation of cuproenzymes and is also essential for the efflux of excess copper across the plasma membrane. In this analysis, we utilized the nuclear magnetic resonance (NMR) structure of the Cu(I)-mediated complex between Atox1 and the first soluble domain of the Menkes protein (Mnk1) as a starting point. Independent free-energy simulations were conducted to investigate the dissociation of both Atox1 and Mnk1. The calculations revealed that the two dissociations require free energy values of 6.3 and 6.2 kcal/mol, respectively, following a stepwise dissociation mechanism.


Assuntos
Proteínas de Transporte de Cobre , ATPases Transportadoras de Cobre , Cobre , Metalochaperonas , Chaperonas Moleculares , Simulação de Dinâmica Molecular , Cobre/química , Cobre/metabolismo , Proteínas de Transporte de Cobre/química , Proteínas de Transporte de Cobre/metabolismo , Humanos , Metalochaperonas/química , Metalochaperonas/metabolismo , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Termodinâmica , Multimerização Proteica
2.
J Inorg Biochem ; 251: 112433, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38043136

RESUMO

The p53 protein plays a major role in cancer prevention, and over 50% of cancer diagnoses can be attributed to p53 malfunction. p53 incorporates a structural Zn site that is required for proper protein folding and function, and in many cases point mutations can result in loss of the Zn2+ ion, destabilization of the tertiary structure, and eventual amyloid aggregation. Herein, we report a series of compounds designed to act as small molecule stabilizers of mutant p53, and feature Zn-binding fragments to chaperone Zn2+ to the metal depleted site and restore wild-type (WT) function. Many Zn metallochaperones (ZMCs) have been shown to generate intracellular reactive oxygen species (ROS), likely by chelating redox-active metals such as Fe2+/3+ and Cu+/2+ and undergoing associated Fenton chemistry. High levels of ROS can result in off-target effects and general toxicity, and thus, careful tuning of ligand Zn2+ affinity, in comparison to the affinity for other endogenous metals, is important for selective mutant p53 targeting. In this work we show that by using carboxylate donors in place of pyridine we can change the relative Zn2+/Cu2+ binding ability in a series of ligands, and we investigate the impact of donor group changes on metallochaperone activity and overall cytotoxicity in two mutant p53 cancer cell lines (NUGC3 and SKGT2).


Assuntos
Metalochaperonas , Proteína Supressora de Tumor p53 , Zinco , Humanos , Linhagem Celular Tumoral , Quelantes , Metalochaperonas/química , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Zinco/metabolismo , Ligação Proteica
3.
mBio ; 14(5): e0096723, 2023 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-37584558

RESUMO

IMPORTANCE: Correct folding of proteins represents a crucial step for their functions. Among the chaperones that control protein folding, the ubiquitous PPIases catalyze the cis/trans-isomerization of peptidyl-prolyl bonds. Only few protein targets of PPIases have been reported in bacteria. To fill this knowledge gap, we performed a large-scale two-hybrid screen to search for targets of the Escherichia coli and Helicobacter pylori SlyD PPIase-metallochaperone. SlyD from both organisms interacts with enzymes (i) containing metal cofactors, (ii) from the central metabolism tricarboxylic acid (TCA) cycle, and (iii) involved in the formation of the essential and ancestral Fe-S cluster cofactor. E. coli and H. pylori ∆slyD mutants present similar phenotypes of diminished susceptibility to antibiotics and to oxidative stress. In H. pylori, measurements of the intracellular ATP content, proton motive force, and activity of TCA cycle proteins suggest that SlyD regulates TCA cycle enzymes by controlling the formation of their indispensable Fe-S clusters.


Assuntos
Proteínas de Escherichia coli , Peptidilprolil Isomerase , Peptidilprolil Isomerase/genética , Escherichia coli , Metalochaperonas/química , Metalochaperonas/metabolismo , Ferro , Dobramento de Proteína , Proteínas de Escherichia coli/metabolismo
4.
J Mol Med (Berl) ; 101(5): 527-542, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37017692

RESUMO

Copper (Cu) was recently demonstrated to play a critical role in cellular physiological and biochemical processes, including energy production and maintenance, antioxidation and enzymatic activity, and signal transduction. Antioxidant 1 (ATOX1), a chaperone of Cu previously named human ATX1 homologue (HAH1), has been found to play an indispensable role in maintaining cellular Cu homeostasis, antioxidative stress, and transcriptional regulation. In the past decade, it has also been found to be involved in a variety of diseases, including numerous neurodegenerative diseases, cancers, and metabolic diseases. Recently, increasing evidence has revealed that ATOX1 is involved in the regulation of cell migration, proliferation, autophagy, DNA damage repair (DDR), and death, as well as in organism development and reproduction. This review summarizes recent advances in the research on the diverse physiological and cytological functions of ATOX1 and the underlying mechanisms of its action in human health and diseases. The potential of ATOX1 as a therapeutic target is also discussed. This review aims to pose unanswered questions related to ATOX1 biology and explore the potential use of ATOX1 as a therapeutic target.


Assuntos
Proteínas de Transporte de Cátions , Cobre , Humanos , Cobre/química , Cobre/metabolismo , Antioxidantes/uso terapêutico , Metalochaperonas/química , Metalochaperonas/genética , Metalochaperonas/metabolismo , Proteínas de Transporte de Cobre , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Chaperonas Moleculares/genética
5.
Protein Sci ; 31(12): e4464, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36208051

RESUMO

Copper is an essential element in nature but in excess, it is toxic to the living cell. The human metallochaperone Atox1 participates in copper homeostasis and is responsible for copper transmission. In a previous multiscale simulation study, we noticed a change in the coordination state of the Cu(I) ion, from 4 bound cysteine residues to 3, in agreement with earlier studies. Here, we perform and analyze classical molecular dynamic simulations of various coordination states: 2, 3, and 4. The main observation is an increase in protein flexibility as a result of a decrease in the coordination state. In addition, we identified several populated conformations that correlate well with double electron-electron resonance distance distributions or an X-ray structure of Cu(I)-bound Atox1. We suggest that the increased flexibility might benefit the process of ion transmission between interacting proteins. Further experiments can scrutinize this hypothesis and shed additional light on the mechanism of action of Atox1.


Assuntos
Proteínas de Transporte de Cátions , Metalochaperonas , Humanos , Metalochaperonas/química , Cobre/química , Proteínas de Transporte de Cobre , Simulação de Dinâmica Molecular , Proteínas de Transporte de Cátions/química , Chaperonas Moleculares/química
6.
Biomolecules ; 12(10)2022 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-36291703

RESUMO

The bioavailability of copper (Cu) in human cells may depend on a complex interplay with zinc (Zn) ions. We investigated the ability of the Zn ion to target the human Cu-chaperone Atox1, a small cytosolic protein capable of anchoring Cu(I), by a conserved surface-exposed Cys-X-X-Cys (CXXC) motif, and deliver it to Cu-transporting ATPases in the trans-Golgi network. The crystal structure of Atox1 loaded with Zn displays the metal ion bridging the CXXC motifs of two Atox1 molecules in a homodimer. The identity and location of the Zn ion were confirmed through the anomalous scattering of the metal by collecting X-ray diffraction data near the Zn K-edge. Furthermore, soaking experiments of the Zn-loaded Atox1 crystals with a strong chelating agent, such as EDTA, caused only limited removal of the metal ion from the tetrahedral coordination cage, suggesting a potential role of Atox1 in Zn metabolism and, more generally, that Cu and Zn transport mechanisms could be interlocked in human cells.


Assuntos
Cobre , Metalochaperonas , Humanos , Proteínas de Transporte de Cobre , Metalochaperonas/química , Metalochaperonas/metabolismo , Cobre/química , ATPases Transportadoras de Cobre , Zinco/metabolismo , Ácido Edético , Chaperonas Moleculares/metabolismo , Quelantes , Íons/metabolismo
7.
J Inorg Biochem ; 225: 111588, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34530332

RESUMO

Nickel insertion into nickel-dependent carbon monoxide dehydrogenase (CODH) represents a key step in the enzyme activation. This is the last step of the biosynthesis of the active site, which contains an atypical heteronuclear NiFe4S4 cluster known as the C-cluster. The enzyme maturation is performed by three accessory proteins, namely CooC, CooT and CooJ. Among them, CooJ from Rhodospirillum rubrum is a histidine-rich protein containing two distinct and spatially separated Ni(II)-binding sites: a N-terminal high affinity site (HAS) and a histidine tail at the C-terminus. In 46 CooJ homologues, the HAS motif was found to be strictly conserved with a H(W/F)XXHXXXH sequence. Here, a proteome database search identified at least 150 CooJ homologues and revealed distinct motifs for HAS, featuring 2, 3 or 4 histidines. The purification and biophysical characterization of three representative members of this protein family showed that they are all homodimers able to bind Ni(II) ions via one or two independent binding sites. Initially thought to be present only in R. rubrum, this study strongly suggests that CooJ could play a significant role in CODH maturation or in nickel homeostasis.


Assuntos
Metalochaperonas , Níquel , Aldeído Oxirredutases/genética , Motivos de Aminoácidos , Archaea/química , Proteínas Arqueais/química , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Bactérias/química , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Metalochaperonas/química , Metalochaperonas/genética , Metalochaperonas/metabolismo , Complexos Multienzimáticos/genética , Família Multigênica , Níquel/metabolismo , Ligação Proteica
8.
Nat Struct Mol Biol ; 27(8): 735-742, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32632277

RESUMO

The cytosolic iron-sulfur (Fe-S) assembly (CIA) pathway is required for the insertion of Fe-S clusters into cytosolic and nuclear client proteins, including many DNA replication and repair factors. The molecular mechanisms of client protein recognition and Fe-S cluster transfer remain unknown. Here, we report crystal structures of the CIA targeting complex (CTC), revealing that its CIAO2B subunit is centrally located and bridges CIAO1 and the client adaptor protein MMS19. Cryo-EM reconstructions of human CTC bound either to the DNA replication factor primase or to the DNA helicase DNA2, combined with biochemical, biophysical and yeast complementation assays, reveal an evolutionarily conserved, bipartite client recognition mode facilitated by CIAO1 and the structural flexibility of the MMS19 subunit. Unexpectedly, the primase Fe-S cluster is located ~70 Å away from the CTC reactive cysteine, implicating conformational dynamics of the CTC or additional maturation factors in the mechanism of Fe-S cluster transfer.


Assuntos
Proteínas Ferro-Enxofre/metabolismo , Metalochaperonas/metabolismo , Metaloproteínas/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Cristalografia por Raios X , Drosophila , Células HEK293 , Humanos , Proteínas Ferro-Enxofre/química , Metalochaperonas/química , Metaloproteínas/química , Camundongos , Modelos Moleculares , Proteínas Nucleares/química , Ligação Proteica , Conformação Proteica , Fatores de Transcrição/química
9.
J Biol Inorg Chem ; 25(3): 501-508, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32222833

RESUMO

The CIAO3 protein operates at a crossroad of the cytosolic iron-sulfur protein assembly (CIA) machinery. Although the functional role of CIAO3 has been recently characterized, a description of its interaction network is still not complete. Literature data suggested that CIAO3 interacts individually with CIA2A and CIAO1 protein, with the latter two interacting each other. However, no experimental data are available yet showing the formation of a possible ternary complex composed by CIAO3, CIAO1, and CIA2A. This work shows, for the first time, via size exclusion chromatography coupled with multiangle light scattering, UV-vis absorption and electron paramagnetic resonance (EPR) spectroscopies, the formation of a stable, [4Fe-4S]-bound, complex, composed by CIAO3 and the hetero-CIA2A-CIAO1 complex. Moreover, site-directed mutagenesis data suggested a structural role for the C-terminal [4Fe-4S] cluster of the CIAO3 protein. These findings can provide solid bases for further investigation of the molecular mechanisms involving these CIA machinery proteins.


Assuntos
Citosol/química , Proteínas Ferro-Enxofre/química , Citosol/metabolismo , Humanos , Proteínas Ferro-Enxofre/genética , Proteínas Ferro-Enxofre/metabolismo , Metalochaperonas/química , Metalochaperonas/metabolismo , Metaloproteínas/química , Metaloproteínas/metabolismo , Estrutura Terciária de Proteína
10.
Chemistry ; 25(67): 15351-15360, 2019 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-31486181

RESUMO

In Rhodospirillum rubrum, the maturation of carbon monoxide dehydrogenase (CODH) requires three nickel chaperones, namely RrCooC, RrCooT and RrCooJ. Recently, the biophysical characterisation of the RrCooT homodimer and the X-ray structure of its apo form revealed the existence of a solvent-exposed NiII -binding site at the dimer interface, involving the strictly conserved Cys2. Here, a multifaceted approach that used NMR and X-ray absorption spectroscopies, complemented with structural bio-modelling methodologies, was used to characterise the binding mode of NiII in RrCooT. This study suggests that NiII adopts a square-planar geometry through a N2 S2 coordinating environment that comprises the two thiolate and amidate groups of both Cys2 residues at the dimer interface. The existence of a diamagnetic mononuclear NiII centre with bis-amidate/bis-thiolate ligands, coordinated by a single-cysteine motif, is unprecedented in biology and raises the question of its role in the activation of CODH at the molecular level.


Assuntos
Cisteína/química , Metalochaperonas/química , Níquel/química , Rhodospirillum rubrum/química , Sequência de Aminoácidos , Sítios de Ligação , Cátions Bivalentes/química , Complexos de Coordenação/química , Ligantes , Modelos Moleculares , Nitrogênio/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Solventes/química , Enxofre/química , Termodinâmica
11.
J Biol Chem ; 294(44): 15876-15888, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31444272

RESUMO

The human pathogen Salmonella enterica serovar Typhimurium (S Typhimurium) contains a complex disulfide bond (Dsb) catalytic machinery. This machinery encompasses multiple Dsb thiol-disulfide oxidoreductases that mediate oxidative protein folding and a less-characterized suppressor of copper sensitivity (scs) gene cluster, associated with increased tolerance to copper. To better understand the function of the Salmonella Scs system, here we characterized two of its key components, the membrane protein ScsB and the periplasmic protein ScsC. Our results revealed that these two proteins form a redox pair in which the electron transfer from the periplasmic domain of ScsB (n-ScsB) to ScsC is thermodynamically driven. We also demonstrate that the Scs reducing pathway remains separate from the Dsb oxidizing pathways and thereby avoids futile redox cycles. Additionally, we provide new insight into the molecular mechanism underlying Scs-mediated copper tolerance in Salmonella We show that both ScsB and ScsC can bind toxic copper(I) with femtomolar affinities and transfer it to the periplasmic copper metallochaperone CueP. Our results indicate that the Salmonella Scs machinery has evolved a dual mode of action, capable of transferring reducing power to the oxidizing periplasm and protecting against copper stress by cooperating with the cue regulon, a major copper resistance mechanism in Salmonella. Overall, these findings expand our understanding of the functional diversity of Dsb-like systems, ranging from those mediating oxidative folding of proteins required for infection to those contributing to defense mechanisms against oxidative stress and copper toxicity, critical traits for niche adaptation and survival.


Assuntos
Proteínas de Bactérias/metabolismo , Cobre/metabolismo , Farmacorresistência Bacteriana , Metalochaperonas/metabolismo , NADH NADPH Oxirredutases/metabolismo , Salmonella/metabolismo , Adaptação Fisiológica , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Cobre/toxicidade , Metalochaperonas/química , Metalochaperonas/genética , NADH NADPH Oxirredutases/química , NADH NADPH Oxirredutases/genética , Oxirredução , Periplasma/metabolismo , Ligação Proteica , Dobramento de Proteína , Regulon , Salmonella/efeitos dos fármacos , Salmonella/enzimologia
12.
Sci Adv ; 5(7): eaaw8478, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31392273

RESUMO

The mechanisms underlying the biogenesis of the structurally unique, binuclear Cu1.5+•Cu1.5+ redox center (CuA) on subunit II (CoxB) of cytochrome oxidases have been a long-standing mystery. Here, we reconstituted the CoxB•CuA center in vitro from apo-CoxB and the holo-forms of the copper transfer chaperones ScoI and PcuC. A previously unknown, highly stable ScoI•Cu2+•CoxB complex was shown to be rapidly formed as the first intermediate in the pathway. Moreover, our structural data revealed that PcuC has two copper-binding sites, one each for Cu1+ and Cu2+, and that only PcuC•Cu1+•Cu2+ can release CoxB•Cu2+ from the ScoI•Cu2+•CoxB complex. The CoxB•CuA center was then formed quantitatively by transfer of Cu1+ from a second equivalent of PcuC•Cu1+•Cu2+ to CoxB•Cu2+. This metalation pathway is consistent with all available in vivo data and identifies the sources of the Cu ions required for CuA center formation and the order of their delivery to CoxB.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cobre/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Metalochaperonas/química , Metalochaperonas/metabolismo , Apoproteínas/metabolismo , Sítios de Ligação , Bradyrhizobium/metabolismo , Cristalografia por Raios X , Modelos Biológicos , Oxirredução , Domínios Proteicos , Relação Estrutura-Atividade
13.
Int J Mol Sci ; 20(15)2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31387303

RESUMO

Copper is an essential cofactor for aerobic respiration, since it is required as a redox cofactor in Cytochrome c Oxidase (COX). This ancient and highly conserved enzymatic complex from the family of heme-copper oxidase possesses two copper sites: CuA and CuB. Biosynthesis of the oxidase is a complex, stepwise process that requires a high number of assembly factors. In this review, we summarize the state-of-the-art in the assembly of COX, with special emphasis in the assembly of copper sites. Assembly of the CuA site is better understood, being at the same time highly variable among organisms. We also discuss the current challenges that prevent the full comprehension of the mechanisms of assembly and the pending issues in the field.


Assuntos
Cobre/metabolismo , Heme/metabolismo , Oxirredutases/metabolismo , Animais , Transporte Biológico , Catálise , Cobre/química , Complexo IV da Cadeia de Transporte de Elétrons/química , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Heme/química , Humanos , Íons/química , Íons/metabolismo , Metalochaperonas/química , Metalochaperonas/metabolismo , Modelos Biológicos , Conformação Molecular , Oxirredução , Oxirredutases/química , Ligação Proteica
14.
Int J Mol Sci ; 20(14)2019 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-31337158

RESUMO

Appropriate maintenance of Cu(I) homeostasis is an essential requirement for proper cell function because its misregulation induces the onset of major human diseases and mortality. For this reason, several research efforts have been devoted to dissecting the inner working mechanism of Cu(I)-binding proteins and transporters. A commonly adopted strategy relies on mutations of cysteine residues, for which Cu(I) has an exquisite complementarity, to serines. Nevertheless, in spite of the similarity between these two amino acids, the structural and functional impact of serine mutations on Cu(I)-binding biomolecules remains unclear. Here, we applied various biochemical and biophysical methods, together with all-atom simulations, to investigate the effect of these mutations on the stability, structure, and aggregation propensity of Cu(I)-binding proteins, as well as their interaction with specific partner proteins. Among Cu(I)-binding biomolecules, we focused on the eukaryotic Atox1-ATP7B system, and the prokaryotic CueR metalloregulator. Our results reveal that proteins containing cysteine-to-serine mutations can still bind Cu(I) ions; however, this alters their stability and aggregation propensity. These results contribute to deciphering the critical biological principles underlying the regulatory mechanism of the in-cell Cu(I) concentration, and provide a basis for interpreting future studies that will take advantage of cysteine-to-serine mutations in Cu(I)-binding systems.


Assuntos
Substituição de Aminoácidos , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Cisteína/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação , Serina/genética , ATPases Transportadoras de Cobre/genética , Humanos , Metalochaperonas/química , Metalochaperonas/genética , Metalochaperonas/metabolismo , Modelos Moleculares , Conformação Proteica , Análise Espectral , Relação Estrutura-Atividade
15.
Biochemistry ; 58(28): 3097-3108, 2019 07 16.
Artigo em Inglês | MEDLINE | ID: mdl-31243953

RESUMO

Mononuclear copper monooxygenases peptidylglycine monooxygenase (PHM) and dopamine ß-monooxygenase (DBM) catalyze the hydroxylation of high energy C-H bonds utilizing a pair of chemically distinct copper sites (CuH and CuM) separated by 11 Å. In earlier work, we constructed single-site PHM variants that were designed to allow the study of the M- and H-centers independently in order to place their reactivity sequentially along the catalytic pathway. More recent crystallographic studies suggest that these single-site variants may not be truly representative of the individual active sites. In this work, we describe an alternative approach that uses a rational design to construct an artificial PHM model in a small metallochaperone scaffold. Using site-directed mutagenesis, we constructed variants that provide a His2Met copper-binding ligand set that mimics the M-center of PHM. The results show that the model accurately reproduces the chemical and spectroscopic properties of the M-center, including details of the methionine coordination, and the properties of Cu(I) and Cu(II) states in the presence of endogenous ligands such as CO and azide. The rate of reduction of the Cu(II) form of the model by the chromophoric reductant N,N'-dimethyl phenylenediamine (DMPD) has been compared with that of the PHM M-center, and the reaction chemistry of the Cu(I) forms with molecular oxygen has also been explored, revealing an unusually low reactivity toward molecular oxygen. This latter finding emphasizes the importance of substrate triggering of oxygen reactivity and implies that the His2Met ligand set, while necessary, is insufficient on its own to activate oxygen in these enzyme systems.


Assuntos
Cobre/metabolismo , Histidina/metabolismo , Metalochaperonas/metabolismo , Metionina/metabolismo , Oxigenases de Função Mista/metabolismo , Modelos Químicos , Animais , Sítios de Ligação/fisiologia , Cobre/química , Histidina/química , Metalochaperonas/química , Metionina/química , Oxigenases de Função Mista/química , Estrutura Secundária de Proteína
16.
Inorg Chem ; 58(20): 13661-13672, 2019 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-31247880

RESUMO

Members of the COG0523 subfamily of candidate GTPase metallochaperones function in bacterial transition-metal homeostasis, but the nature of the cognate metal, mechanism of metal transfer, and identification of target protein(s) for metal delivery remain open questions. Here, we explore the multifunctionality of members of the subfamily linked to delivering ZnII to apoprotein targets under conditions of host-imposed transition-metal depletion. We examine two zinc-uptake repressor (Zur)-regulated COG0523 family members, each from a major human pathogen, Acinetobacter baumannii (AbZigA) and Staphylococcus aureus (SaZigA), in an effort to develop a model for ZnII metallochaperone activity. ZnII chelator competition experiments reveal one high-affinity (KZn1 ≈ 1010-1011 M-1) metal-binding site in each GTPase, while AbZigA and SaZigA are characterized by an additional one and two (lower-affinity) metal-binding sites, respectively. CoII titrations reveal that both metallochaperones have similar electronic absorption characteristics that indicate the presence of two tetrahedral metal coordination sites. High-affinity metal binding at the CXCC motif activates the GTPase activity of both enzymes, with ZnII more effective than CoII. Both GTPases bind the product, GDP, more tightly in the apoprotein than the ZnII-bound state and exhibit what is best described as a "locked" conformation around the GTP substrate. Negative thermodynamic linkage is observed between nucleotide binding and metal binding, leading to a new mechanistic model for COG0523-catalyzed metal delivery.


Assuntos
Metalochaperonas/metabolismo , Zinco/metabolismo , Sítios de Ligação , Metalochaperonas/química , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Zinco/química
17.
Rapid Commun Mass Spectrom ; 33(10): 951-958, 2019 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-30812058

RESUMO

RATIONALE: The binding ratio of metal complexes with cysteinyl thiols in proteins plays an important role in deciphering the mechanisms of action of therapeutic metal complexes, but its analysis is still a significant challenge. In this work, a quantitative mass spectrometry method is developed to determine the binding ratio of metal-based anticancer complexes with cysteines in human copper chaperone protein Atox1. METHODS: A novel strategy based on a thiol-specific stable isotopic labelling reagent was developed to determine the binding ratio of metal-based anticancer complexes, namely cisplatin and organometallic ruthenium complex [(η6 -biphenyl)RuCl(en)]PF6 (en = ethylenediamine), with the cysteinyl residues of Atox1. RESULTS: Both cisplatin and the ruthenium complex were reactive not only to Cys15 and/or Cys18, the copper(I) binding site of Atox1, but also to Cys44. The binding ratios of the ruthenium complex with the cysteinyl residues were much higher than those of cisplatin. However, the addition of copper(I) could markedly increase the binding ratios of cysteinyl residues of Atox1 with cisplatin, but not with the ruthenium complex. CONCLUSIONS: This strategy can not only precisely determine the binding ratios of metal complexes to protein thiols, but also be helpful in distinguishing thiol-binding sites from other binding sites of metal complexes in proteins. We expect wide application of this method to the research of covalent/coordinative interactions between metal complexes and protein thiols.


Assuntos
Antineoplásicos/química , Cisplatino/química , Cobre/química , Espectrometria de Massas/métodos , Metalochaperonas/química , Rutênio/química , Sítios de Ligação , Proteínas de Transporte de Cobre , Cisteína/química , Humanos , Modelos Moleculares , Chaperonas Moleculares , Ligação Proteica , Compostos de Sulfidrila/química
18.
Biometals ; 32(3): 521-532, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30758762

RESUMO

The biosynthesis of the dinuclear metal cluster at the active sites of the [NiFe]-hydrogenase enzymes is a multi-step process executed by a suite of accessory proteins. Nickel insertion during maturation of Escherichia coli [NiFe]-hydrogenase 3 is achieved by the metallochaperones HypA, SlyD and the GTPase HypB, but how these proteins cooperate to ensure nickel delivery is not known. In this study, the complexes formed between the individual purified proteins were examined by using several methods. Size exclusion chromatography (SEC) indicated that SlyD and HypB interact primarily in a 1:1 complex. The affinity of HypB-SlyD was measured by using surface plasmon resonance, which revealed a KD of 24 ± 10 nM in the absence of nucleotide and an interaction several fold tighter in the presence of GDP. A ternary complex between all three proteins was not detected, and instead SlyD blocked the interaction of HypA with HypB in competitive binding experiments. Furthermore, cross-linking experiments suggest a weak interaction between HypA and SlyD, which is not detectable by SEC. Electrochemical analysis confirmed each of the pairwise interactions and that the relative affinities of these complexes are on the order of HypB-SlyD > HypB-HypA > HypA-SlyD. These results indicate a hierarchy of interactions, as opposed to a single multiprotein complex, and provide insight into the nickel delivery process during hydrogenase enzyme maturation.


Assuntos
Escherichia coli/enzimologia , Hidrogenase/metabolismo , Níquel/metabolismo , Cromatografia em Gel , Hidrogenase/química , Metalochaperonas/química , Metalochaperonas/metabolismo , Níquel/química
19.
Biotechnol Bioeng ; 116(3): 481-489, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30418672

RESUMO

The transition metal (iron or cobalt) is a mandatory part that constitutes the catalytic center of nitrile hydratase (NHase). The incorporation of the cobalt ion into cobalt-containing NHase (Co-NHase) was reported to depend on self-subunit swapping and the activator of the Co-NHase acts as a self-subunit swapping chaperone for subunit exchange. Here we discovered that the activator acting as a metallochaperone transferred the cobalt ion into subunit-fused Co-NHase. We successfully isolated two activators, P14K and NhlE, which were the activators of NHases from Pseudomonas putida NRRL-18668 and the activator of low-molecular-mass NHase from Rhodococcus rhodochrous J1, respectively. Cobalt content determination demonstrated that NhlE and P14K were two cobalt-containing proteins. Substitution of the amino acids involved in the C-terminus of the activators affected the activity of the two NHases, indicating that the potential cobalt-binding sites might be located at the flexible C-terminal region. The cobalt-free NHases could be activated by either of the two activators, and both the two activators activated their cognate NHase more efficiently than did the noncognate ones. This study provided insights into the maturation of subunit-fused NHases and confirmed the metallochaperone function of the self-subunit swapping chaperone.


Assuntos
Cobalto , Hidroliases , Metalochaperonas , Subunidades Proteicas , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cobalto/química , Cobalto/metabolismo , Hidroliases/química , Hidroliases/metabolismo , Metalochaperonas/química , Metalochaperonas/metabolismo , Modelos Moleculares , Ligação Proteica , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
20.
J Biol Inorg Chem ; 23(8): 1309-1330, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30264175

RESUMO

Helicobacter pylori HypA (HpHypA) is a metallochaperone necessary for maturation of [Ni,Fe]-hydrogenase and urease, the enzymes required for colonization and survival of H. pylori in the gastric mucosa. HpHypA contains a structural Zn(II) site and a unique Ni(II) binding site at the N-terminus. X-ray absorption spectra suggested that the Zn(II) coordination depends on pH and on the presence of Ni(II). This study was performed to investigate the structural properties of HpHypA as a function of pH and Ni(II) binding, using NMR spectroscopy combined with DFT and molecular dynamics calculations. The solution structure of apo,Zn-HpHypA, containing Zn(II) but devoid of Ni(II), was determined using 2D, 3D and 4D NMR spectroscopy. The structure suggests that a Ni-binding and a Zn-binding domain, joined through a short linker, could undergo mutual reorientation. This flexibility has no physiological effect on acid viability or urease maturation in H. pylori. Atomistic molecular dynamics simulations suggest that Ni(II) binding is important for the conformational stability of the N-terminal helix. NMR chemical shift perturbation analysis indicates that no structural changes occur in the Zn-binding domain upon addition of Ni(II) in the pH 6.3-7.2 range. The structure of the Ni(II) binding site was probed using 1H NMR spectroscopy experiments tailored to reveal hyperfine-shifted signals around the paramagnetic metal ion. On this basis, two possible models were derived using quantum-mechanical DFT calculations. The results provide a comprehensive picture of the Ni(II) mode to HpHypA, important to rationalize, at the molecular level, the functional interactions of this chaperone with its protein partners.


Assuntos
Proteínas de Bactérias/metabolismo , Helicobacter pylori/química , Metalochaperonas/metabolismo , Níquel/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Teoria da Densidade Funcional , Escherichia coli/genética , Glicina/genética , Concentração de Íons de Hidrogênio , Metalochaperonas/química , Metalochaperonas/genética , Modelos Químicos , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Níquel/química , Ressonância Magnética Nuclear Biomolecular/métodos , Ligação Proteica , Conformação Proteica em alfa-Hélice , Domínios Proteicos , Zinco/química , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...