Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Synchrotron Radiat ; 29(Pt 6): 1495-1503, 2022 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-36345757

RESUMO

The microscopy research at the Bionanoprobe (currently at beamline 9-ID and later 2-ID after APS-U) of Argonne National Laboratory focuses on applying synchrotron X-ray fluorescence (XRF) techniques to obtain trace elemental mappings of cryogenic biological samples to gain insights about their role in critical biological activities. The elemental mappings and the morphological aspects of the biological samples, in this instance, the bacterium Escherichia coli (E. Coli), also serve as label-free biological fingerprints to identify E. coli cells that have been treated differently. The key limitations of achieving good identification performance are the extraction of cells from raw XRF measurements via binary conversion, definition of features, noise floor and proportion of cells treated differently in the measurement. Automating cell extraction from raw XRF measurements across different types of chemical treatment and the implementation of machine-learning models to distinguish cells from the background and their differing treatments are described. Principal components are calculated from domain knowledge specific features and clustered to distinguish healthy and poisoned cells from the background without manual annotation. The cells are ranked via fuzzy clustering to recommend regions of interest for automated experimentation. The effects of dwell time and the amount of data required on the usability of the software are also discussed.


Assuntos
Escherichia coli , Síncrotrons , Raios X , Microscopia de Fluorescência , Aprendizado de Máquina
2.
Mol Hum Reprod ; 20(11): 1077-89, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25143461

RESUMO

Rapid cellular zinc influx regulates early mammalian development during the oocyte-to-egg transition through modulation of the meiotic cell cycle. Despite the physiological necessity of this zinc influx, the molecular mechanisms that govern such accumulation are unknown. Here we show that the fully grown mammalian oocyte does not employ a transcriptionally based mechanism of zinc regulation involving metal response element-binding transcription factor-1 (MTF-1), as demonstrated by a lack of MTF-1 responsiveness to environmental zinc manipulation. Instead, the mammalian oocyte controls zinc uptake through two maternally derived and cortically distributed zinc transporters, ZIP6 and ZIP10. Targeted disruption of these transporters using several approaches during meiotic maturation perturbs the intracellular zinc quota and results in a cell cycle arrest at a telophase I-like state. This arrest phenocopies established models of zinc insufficiency during the oocyte-to-egg transition, indicating the essential function of these maternally expressed transporters. Labile zinc localizes to punctate cytoplasmic structures in the human oocyte, and ZIP6 and ZIP10 are enriched in the cortex. Altogether, we demonstrate a mechanism of metal regulation required for female gamete development that may be evolutionarily conserved.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Zinco/metabolismo , Adolescente , Adulto , Animais , Transporte Biológico/genética , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Ciclo Celular/genética , Feminino , Regulação da Expressão Gênica , Homeostase , Humanos , Camundongos Endogâmicos , Oócitos/metabolismo , Elementos de Resposta , Telófase
3.
Ultramicroscopy ; 128: 24-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23500508

RESUMO

A dedicated analytical scanning transmission electron microscope (STEM) with dual energy dispersive spectroscopy (EDS) detectors has been designed for complementary high performance imaging as well as high sensitivity elemental analysis and mapping of biological structures. The performance of this new design, based on a Hitachi HD-2300A model, was evaluated using a variety of biological specimens. With three imaging detectors, both the surface and internal structure of cells can be examined simultaneously. The whole-cell elemental mapping, especially of heavier metal species that have low cross-section for electron energy loss spectroscopy (EELS), can be faithfully obtained. Optimization of STEM imaging conditions is applied to thick sections as well as thin sections of biological cells under low-dose conditions at room and cryogenic temperatures. Such multimodal capabilities applied to soft/biological structures usher a new era for analytical studies in biological systems.


Assuntos
Eritrócitos/ultraestrutura , Ilhotas Pancreáticas/ultraestrutura , Microscopia Eletrônica de Transmissão e Varredura/instrumentação , Microscopia Eletrônica de Transmissão e Varredura/métodos , Espectrometria por Raios X/instrumentação , Espectroscopia de Perda de Energia de Elétrons/instrumentação , Vírus do Mosaico do Tabaco/ultraestrutura , Animais , Microscopia Crioeletrônica/métodos , Humanos , Masculino , Metais Pesados/análise , Espectrometria por Raios X/métodos , Espectroscopia de Perda de Energia de Elétrons/métodos , Espermatozoides/citologia , Espermatozoides/ultraestrutura
4.
Nat Struct Biol ; 8(9): 751-5, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11524675

RESUMO

The copper chaperone for superoxide dismutase (CCS) activates the eukaryotic antioxidant enzyme copper, zinc superoxide dismutase (SOD1). The 2.9 A resolution structure of yeast SOD1 complexed with yeast CCS (yCCS) reveals that SOD1 interacts with its metallochaperone to form a complex comprising one monomer of each protein. The heterodimer interface is remarkably similar to the SOD1 and yCCS homodimer interfaces. Striking conformational rearrangements are observed in both the chaperone and target enzyme upon complex formation, and the functionally essential C-terminal domain of yCCS is well positioned to play a key role in the metal ion transfer mechanism. This domain is linked to SOD1 by an intermolecular disulfide bond that may facilitate or regulate copper delivery.


Assuntos
Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae , Superóxido Dismutase/química , Superóxido Dismutase/metabolismo , Leveduras/enzimologia , Sítios de Ligação , Cobre/metabolismo , Cristalografia por Raios X , Dimerização , Dissulfetos/metabolismo , Ativação Enzimática , Modelos Moleculares , Conformação Proteica
6.
J Biol Chem ; 276(44): 41365-76, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11500502

RESUMO

The interaction of the copper chaperone Atx1 and the first cytosolic domain of Ccc2 ATPase, Ccc2a, was investigated by NMR in solution. In particular, a solution of Cu(I)-15NAtx1 was titrated with apo-Ccc2a, and, vice versa, a solution of Cu(I)-15NCcc2a was titrated with apo-Atx1. By following the 15N and 1H chemical shifts, a new species is detected in both experiments. This species is the same in both titrations and is in fast exchange with the parent species on the NMR time scale. Nuclear relaxation data are consistent with the formation of an adduct. Judging from the nuclear Overhauser effect spectroscopy patterns, the structure of Cu(I)-15NCcc2a in the presence of apo-Atx1 is not significantly altered, whereas Cu(I)-15NAtx1 in the presence of apo-Ccc2a experiences some changes with respect to both the apoproteins and the Cu(I)-loaded proteins. The structure of the Cu(I)-15NAtx1 moiety in the adduct was obtained from 1137 nuclear Overhauser effects to a final root mean square deviation to the mean structure of 0.76 +/- 0.13 A for the backbone and 1.11 +/- 0.11 A for the heavy atoms. 15N and 1H chemical shifts suggest the regions of interaction that, together with independent information, allow a structural model of the adduct to be proposed. The apo form of Atx1 displays significant mobility in loops 1 and 5, the N-terminal part of helix alpha1, and the C-terminal part of helix alpha2 on the ms-micros time scale. These regions correspond to the metal binding site. Such mobility is largely reduced in the free Cu(I)-Atx1 and in the adduct with apo-Ccc2a. The analogous mobility of Ccc2a in both Cu(I) and apo forms is reduced with respect to Atx1. Such an adduct is relevant as a structural and kinetic model for copper transfer from Atx1 to Ccc2a in physiological conditions.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte , Proteínas de Transporte de Cátions , Cobre/metabolismo , Citosol/enzimologia , Proteínas Fúngicas/metabolismo , Proteínas de Saccharomyces cerevisiae , Adenosina Trifosfatases/química , Sequência de Aminoácidos , Sítios de Ligação , Transporte Biológico , Proteínas de Transporte de Cobre , Proteínas Fúngicas/química , Modelos Moleculares , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica
7.
Biochemistry ; 40(35): 10417-23, 2001 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-11523983

RESUMO

The Escherichia coli Zur protein is a Fur homologue that regulates expression of Zn(II) uptake systems. The zinc-loaded form of Zur is proposed to bind DNA and repress transcription of the znuABC genes. Recent in vitro data indicate that the transcriptional activity of Zur is half-maximal when free Zn(II) concentrations are in the sub-femtomolar range, making it the most sensitive Zn(II) metalloregulatory protein reported to date. Previous results indicate that Zur binds at least one zinc; however, little else is known about Zn(II) binding. We have purified E. coli Zur to homogeneity and found that it has two Zn(II) binding sites per monomer with different coordination environments. Using Zn(II) binding assays, ICP-AES analysis, and Zn EXAFS analysis, we show that one zinc is tightly bound in an S(3)(N/O) coordination environment. Both Co(II) and Zn(II) were substituted into the second metal binding site and probed by EXAFS and UV-visible absorption spectroscopy. These studies indicate that Co(II) is bound in an S(N/O)(3) coordination environment with tetrahedral geometry. The Zn(II) EXAFS of Zn(2)Zur, which is consistent with the results for both sites, indicates an average coordination environment of S(2)(N/O)(2), presumably due to one S(N/O)(3) site and one S(3)(N/O) site. These studies reveal the coordination environments that confer such exceptional zinc sensitivity and may provide the foundation for understanding the molecular basis of metal ion selectivity. A comparison of the metal binding sites in Zur with its Fe(II)-sensing homologue Fur provides clues as to why these two proteins with similar structures respond to two very different metal ions.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli , Escherichia coli/química , Zinco/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Sítios de Ligação , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/isolamento & purificação , Metaloproteínas/química , Dados de Sequência Molecular , Proteínas Repressoras/química , Espectrofotometria
8.
J Biol Chem ; 276(42): 38410-6, 2001 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-11473116

RESUMO

The copper chaperone for superoxide dismutase (CCS) activates the antioxidant enzyme Cu,Zn-SOD (SOD1) by directly inserting the copper cofactor into the apo form of SOD1. Neither the mechanism of protein-protein recognition nor of metal transfer is clear. The metal transfer step has been proposed to occur within a transient copper donor/acceptor complex that is either a heterodimer or heterotetramer (i.e. a dimer of dimers). To determine the nature of this intermediate, we generated a mutant form of SOD1 by replacing a copper binding residue His-48 with phenylalanine. This protein cannot accept copper from CCS but does form a stable complex with apo- and Cu-CCS, as observed by immunoprecipitation and native gel electrophoresis. Fluorescence anisotropy measurements corroborate the formation of this species and further indicate that copper enhances the stability of the dimer by an order of magnitude. The copper form of the heterodimer was isolated by gel filtration chromatography and contains one copper and one zinc atom per heterodimer. These results support a mechanism for copper transfer in which CCS and SOD1 dock via their highly conserved dimer interfaces in a manner that precisely orients the Cys-rich copper donor sites of CCS and the His-rich acceptor sites of SOD1 to form a copper-bridged intermediate.


Assuntos
Cobre/química , Cobre/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae , Superóxido Dismutase/metabolismo , Anisotropia , Sítios de Ligação , Western Blotting , Cromatografia em Gel , Dimerização , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Histidina/química , Mutação , Fenilalanina/metabolismo , Testes de Precipitina , Ligação Proteica , Superóxido Dismutase/genética , Fatores de Tempo
9.
Science ; 292(5526): 2488-92, 2001 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-11397910

RESUMO

Intracellular zinc is thought to be available in a cytosolic pool of free or loosely bound Zn(II) ions in the micromolar to picomolar range. To test this, we determined the mechanism of zinc sensors that control metal uptake or export in Escherichia coli and calibrated their response against the thermodynamically defined free zinc concentration. Whereas the cellular zinc quota is millimolar, free Zn(II) concentrations that trigger transcription of zinc uptake or efflux machinery are femtomolar, or six orders of magnitude less than one atom per cell. This is not consistent with a cytosolic pool of free Zn(II) and suggests an extraordinary intracellular zinc-binding capacity. Thus, cells exert tight control over cytosolic metal concentrations, even for relatively low-toxicity metals such as zinc.


Assuntos
Proteínas de Bactérias , Proteínas de Ligação a DNA/metabolismo , Proteínas de Escherichia coli , Escherichia coli/metabolismo , Fatores de Transcrição/metabolismo , Zinco/metabolismo , Sequência de Bases , Meios de Cultura , Citosol/metabolismo , Pegada de DNA , DNA Bacteriano/metabolismo , Proteínas de Ligação a DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Desoxirribonuclease I/metabolismo , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Etilenodiaminas/metabolismo , Genes Bacterianos , Homeostase , Concentração de Íons de Hidrogênio , Transporte de Íons , Dados de Sequência Molecular , Concentração Osmolar , Regiões Promotoras Genéticas , Termodinâmica , Fatores de Transcrição/genética , Transcrição Gênica
10.
Annu Rev Biochem ; 70: 677-701, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11395420

RESUMO

Genetic, biochemical, and spectroscopic studies have established a new function for an intracellular protein, i.e., guiding and inserting a copper cofactor into the active site of a target enzyme. Studies of these new proteins have revealed a fundamental aspect of copper physiology, namely the vast overcapacity of the cytoplasm for copper sequestration. This finding framed the mechanistic, energetic, and structural aspects of intracellular copper trafficking proteins. One hallmark of the copper chaperones is the similarity of the protein fold between the chaperone and its target enzyme. The surface residues presented by each partner, however, are quite different, and some initial findings concerning the complementarity of these interfaces have led to mechanistic insights. The copper chaperones appear to lower the activation barrier for metal transfer into specific protein-binding sites. The manner in which they facilitate metal insertion appears to involve a docking of the metal donor and acceptor sites in close proximity to one another. Although the intimate mechanism is still open, it appears that a low activation barrier for metal transfer is achieved by a network of coordinate-covalent, electrostatic, and hydrogen bonding interactions in the vicinity of the metal-binding site itself.


Assuntos
Proteínas de Transporte , Cobre/metabolismo , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Chaperonas Moleculares , Proteínas de Saccharomyces cerevisiae , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Transporte Biológico , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Sequência Conservada , Proteínas de Transporte de Cobre , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Metalochaperonas , Metaloproteínas/química , Metaloproteínas/metabolismo , Dados de Sequência Molecular , Superóxido Dismutase/metabolismo
11.
J Biol Chem ; 276(33): 30670-7, 2001 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-11399769

RESUMO

Copper is essential but can be toxic even at low concentrations. Coping with this duality requires multiple pathways to control intracellular copper availability. Three copper-inducible promoters, controlling expression of six copper tolerance genes, were recently identified in Escherichia coli. The cue system employs an inner membrane copper transporter, whereas the cus system includes a tripartite transporter spanning the entire cell envelope. Although cus is not essential for aerobic copper tolerance, we show here that a copper-sensitive phenotype can be observed when cus is inactivated in a cueR background. Furthermore, a clear copper-sensitive phenotype for the cus system is revealed in the absence of O(2). These results indicate that the cue pathway, which includes a copper exporter, CopA, and a periplasmic oxidase, CueO, is the primary aerobic system for copper tolerance. During anaerobic growth, however, copper toxicity increases, and the independent cus copper exporter is also necessary for full copper tolerance. We conclude that the cytosolic (CueR) and periplasmic (CusRS) sensor systems differentially regulate copper export systems in response to changes in copper and oxygen availability. These results underscore the increased toxicity of copper under anaerobic conditions and the complex adaptation of copper export in E. coli.


Assuntos
Proteínas de Bactérias/fisiologia , Cobre/toxicidade , Proteínas de Ligação a DNA/fisiologia , Escherichia coli/efeitos dos fármacos , Genes Bacterianos , Aerobiose , Anaerobiose , Cobre/metabolismo , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento
12.
Biochemistry ; 40(6): 1528-39, 2001 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-11327811

RESUMO

The (1)H NMR solution structure of the Cu(I)-bound form of Atx1, a 73-amino acid metallochaperone protein from the yeast Saccharomyces cerevisiae, has been determined. Ninety percent of the (1)H and 95% of the (15)N resonances were assigned, and 1184 meaningful NOEs and 42 (3)J(HNH)(alpha) and 60 (1)J(HN) residual dipolar couplings provided a family of structures with rmsd values to the mean structure of 0.37 +/- 0.07 A for the backbone and 0.83 +/- 0.08 A for all heavy atoms. The structure is constituted by four antiparallel beta strands and two alpha helices in a betaalphabetabetaalphabeta fold. Following EXAFS data [Pufahl, R., Singer, C. P., Peariso, K. L., Lin, S.-J., Schmidt, P. J., Fahrni, C. J., Cizewski Culotta, V., Penner-Hahn, J. E., and O'Halloran, T. V. (1997) Science 278, 853-856], a copper ion can be placed between two sulfur atoms of Cys15 and Cys18. The structure of the reduced apo form has also been determined with similar resolution using 1252 meaningful NOEs (rmsd values for the family to the mean structure are 0.67 +/- 0.12 A for the backbone and 1.00 +/- 0.12 A for all heavy atoms). Comparison of the Cu(I) and apo conformations of the protein reveals that the Cu(I) binding cysteines move from a buried site in the bound metal form to a solvent-exposed conformation on the surface of the protein after copper release. Furthermore, copper release leads to a less helical character in the metal binding site. Comparison with the Hg(II)-Atx1 solid-state structure [Rosenzweig, A. C., Huffman, D. L., Hou, M. Y., Wernimont, A. K., Pufahl, R. A., and O'Halloran, T. V. (1999) Structure 7, 605-617] provides insights into the copper transfer mechanism, and a pivotal role for Lys65 in the metal capture and release process is proposed.


Assuntos
Apoproteínas/química , Proteínas de Transporte , Cobre/química , Proteínas Fúngicas/química , Metaloproteínas/química , Chaperonas Moleculares/química , Proteínas de Saccharomyces cerevisiae , Sequência de Aminoácidos , Apoproteínas/metabolismo , Sequência de Bases , Cristalografia por Raios X , Proteínas Fúngicas/metabolismo , Computação Matemática , Mercúrio/química , Metaloproteínas/metabolismo , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Oxirredução , Conformação Proteica , Estrutura Secundária de Proteína , Saccharomyces cerevisiae , Soluções , Termodinâmica
13.
J Biol Chem ; 276(7): 5166-76, 2001 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-11018045

RESUMO

The mechanism for copper loading of the antioxidant enzyme copper, zinc superoxide dismutase (SOD1) by its partner metallochaperone protein is not well understood. Here we show the human copper chaperone for Cu,Zn-SOD1 (hCCS) activates either human or yeast enzymes in vitro by direct protein to protein transfer of the copper cofactor. Interestingly, when denatured with organic solvents, the apo-form of human SOD1 cannot be reactivated by added copper ion alone, suggesting an additional function of hCCS such as facilitation of an active folded state of the enzyme. While hCCS can bind several copper ions, metal binding studies in the presence of excess copper scavengers that mimic the intracellular chelation capacity indicate a limiting stoichiometry of one copper and one zinc per hCCS monomer. This protein is active and unlike the yeast protein, is a homodimer regardless of copper occupancy. Matrix-assisted laser desorption ionization-mass spectrometry and metal binding studies suggest that Cu(I) is bound by residues from the first and third domains and no bound copper is detected for the second domain of hCCS in either the full-length or truncated forms of the protein. Copper-induced conformational changes in the essential C-terminal peptide of hCCS are consistent with a "pivot, insert, and release" mechanism that is similar to one proposed for the well characterized metal handling enzyme, mercuric ion reductase.


Assuntos
Chaperonas Moleculares/química , Superóxido Dismutase/metabolismo , Sequência de Aminoácidos , Cobre/química , Ativação Enzimática , Humanos , Modelos Químicos , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Superóxido Dismutase/química , Superóxido Dismutase-1 , Leveduras/enzimologia , Zinco/química
14.
J Biol Chem ; 276(11): 8415-26, 2001 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-11083871

RESUMO

Ccc2 is an intracellular copper transporter in Saccharomyces cerevisiae and is a physiological target of the copper chaperone Atx1. Here we describe the solution structure of the first N-terminal MTCXXC metal-binding domain, Ccc2a, both in the presence and absence of Cu(I). For Cu(I)-Ccc2a, 1944 meaningful nuclear Overhauser effects were used to obtain a family of 35 structures with root mean square deviation to the average structure of 0.36 +/- 0.06 A for the backbone and 0.79 +/- 0.05 A for the heavy atoms. For apo-Ccc2a, 1970 meaningful nuclear Overhauser effects have been used with 35 (3)J(HNHalpha) to obtain a family of 35 structures with root mean square deviation to the average structure of 0.38 +/- 0.06 A for the backbone and 0.82 +/- 0.07 A for the heavy atoms. The protein exhibits a betaalphabetabetaalphabeta, ferrodoxin-like fold similar to that of its target Atx1 and that of a human counterpart, the fourth metal-binding domain of the Menkes protein. The overall fold remains unchanged upon copper loading, but the copper-binding site itself becomes less disordered. The helical context of the copper-binding site, and the copper-induced conformational changes in Ccc2a differ from those in Atx1. Ccc2a presents a conserved acidic surface which complements the basic surface of Atx1 and a hydrophobic surface. These results open new mechanistic aspects of copper transporter domains with physiological copper donor and acceptor proteins.


Assuntos
Proteínas de Transporte de Cátions , Cobre/química , Proteínas Fúngicas/química , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/metabolismo , Sequência de Aminoácidos , Cobre/metabolismo , Proteínas de Transporte de Cobre , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Soluções , Eletricidade Estática
15.
Biochemistry ; 39(48): 14720-7, 2000 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-11101286

RESUMO

Copper, zinc superoxide dismutase (SOD1) is activated in vivo by the copper chaperone for superoxide dismutase (CCS). The molecular mechanisms by which CCS recognizes and docks with SOD1 for metal ion insertion are not well understood. Two models for the oligomerization state during copper transfer have been proposed: a heterodimer comprising one monomer of CCS and one monomer of SOD1 and a dimer of dimers involving interactions between the two homodimers. We have investigated protein-protein complex formation between copper-loaded and apo yeast CCS (yCCS) and yeast SOD1 for both wild-type SOD1 (wtSOD1) and a mutant SOD1 in which copper ligand His 48 has been replaced with phenylalanine (H48F-SOD1). According to gel filtration chromatography, dynamic light scattering, analytical ultracentrifugation, and chemical cross-linking experiments, yCCS and this mutant SOD1 form a complex with the correct molecular mass for a heterodimer. No higher order oligomers were detected. Heterodimer formation is facilitated by the presence of zinc but does not depend on copper loading of yCCS. The complex formed with H48F-SOD1 is more stable than that formed with wtSOD1, suggesting that the latter is a more transient species. Notably, heterodimer formation between copper-loaded yCCS and wtSOD1 is accompanied by SOD1 activation only in the presence of zinc. These findings, taken together with structural, biochemical, and genetic studies, strongly suggest that in vivo copper loading of yeast SOD1 occurs via a heterodimeric intermediate.


Assuntos
Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae , Superóxido Dismutase/metabolismo , Transporte Biológico , Dimerização , Modelos Químicos , Ligação Proteica , Superóxido Dismutase-1 , Ultracentrifugação
16.
J Bacteriol ; 182(20): 5864-71, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11004187

RESUMO

Using a genetic screen we have identified two chromosomal genes, cusRS (ylcA ybcZ), from Escherichia coli K-12 that encode a two-component, signal transduction system that is responsive to copper ions. This regulatory system is required for copper-induced expression of pcoE, a plasmid-borne gene from the E. coli copper resistance operon pco. The closest homologs of CusR and CusS are plasmid-borne two-component systems that are also involved in metal responsive gene regulation: PcoR and PcoS from the pco operon of E. coli; CopR and CopS from the cop operon, which provides copper resistance to Pseudomonas syringae; and SilR and SilS from the sil locus, which provides silver ion resistance to Salmonella enterica serovar Typhimurium. The genes cusRS are also required for the copper-dependent expression of at least one chromosomal gene, designated cusC (ylcB), which is allelic to the recently identified virulence gene ibeB in E. coli K1. The cus locus may comprise a copper ion efflux system, because the expression of cusC is induced by high concentrations of copper ions. Furthermore, the translation products of cusC and additional downstream genes are homologous to known metal ion antiporters.


Assuntos
Cromossomos Bacterianos/genética , Sulfato de Cobre/farmacologia , Escherichia coli/genética , Genes Bacterianos , Óperon , Bacteriófago lambda/genética , Sequência de Bases , Mapeamento Cromossômico , Cromossomos Bacterianos/efeitos dos fármacos , Escherichia coli/efeitos dos fármacos , Biblioteca Genômica , Dados de Sequência Molecular , Plasmídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/metabolismo , Alinhamento de Sequência , Homologia de Sequência do Ácido Nucleico , Transdução de Sinais , beta-Galactosidase/genética
17.
Nat Struct Biol ; 7(9): 766-71, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10966647

RESUMO

The Hah1 metallochaperone protein is implicated in copper delivery to the Menkes and Wilson disease proteins. Hah1 and the N-termini of its target proteins belong to a family of metal binding domains characterized by a conserved MT/HCXXC sequence motif. The crystal structure of Hah1 has been determined in the presence of Cu(I), Hg(II), and Cd(II). The 1.8 A resolution structure of CuHah1 reveals a copper ion coordinated by Cys residues from two adjacent Hah1 molecules. The CuHah1 crystal structure is the first of a copper chaperone bound to copper and provides structural support for direct metal ion exchange between conserved MT/HCXXC motifs in two domains. The structures of HgHah1 and CdHah1, determined to 1.75 A resolution, also reveal metal ion coordination by two MT/HCXXC motifs. An extended hydrogen bonding network, unique to the complex of two Hah1 molecules, stabilizes the metal binding sites and suggests specific roles for several conserved residues. Taken together, the structures provide models for intermediates in metal ion transfer and suggest a detailed molecular mechanism for protein recognition and metal ion exchange between MT/HCXXC containing domains.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte/química , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions , Cobre/metabolismo , Metaloproteínas/química , Metaloproteínas/metabolismo , Motivos de Aminoácidos , Sítios de Ligação , Cádmio/metabolismo , Sequência Conservada , Proteínas de Transporte de Cobre , ATPases Transportadoras de Cobre , Cristalografia por Raios X , Cisteína/metabolismo , Humanos , Ligação de Hidrogênio , Mercúrio/metabolismo , Metalochaperonas , Modelos Moleculares , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
18.
J Biol Chem ; 275(40): 31024-9, 2000 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-10915804

RESUMO

Because copper ions are both essential cofactors and cytotoxic agents, the net accumulation of this element in a cell must be carefully balanced. Depending upon the cellular copper status, copper ions must either be imported or ejected. CopA, the principal copper efflux ATPase in Escherichia coli, is induced by elevated copper in the medium, but the copper-sensing regulatory factor is unknown. Inspection of the copA promoter reveals signature elements of promoters controlled by metalloregulatory proteins in the MerR family. These same elements are also present upstream of yacK, which encodes a putative multi-copper oxidase. Homologues of YacK are found in copper resistance determinants that facilitate copper efflux. Here we show by targeted gene deletion and promoter fusion assays that both copA and yacK are regulated in a copper-responsive manner by the MerR homologue, ybbI. We have designated ybbI as cueR for the Cu efflux regulator. This represents the first example of a copper-responsive regulon on the E. coli chromosome and further extends the roles of MerR family members in prokaryotic stress response.


Assuntos
Proteínas de Bactérias/genética , Cobre/metabolismo , Proteínas de Ligação a DNA/genética , Escherichia coli/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Proteínas de Bactérias/metabolismo , Sequência de Bases , Cromossomos Bacterianos , Clonagem Molecular , Sulfato de Cobre/farmacologia , Primers do DNA/metabolismo , Escherichia coli/genética , Íons , Dados de Sequência Molecular , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , Homologia de Sequência de Aminoácidos
20.
Curr Opin Chem Biol ; 4(2): 140-7, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10742187

RESUMO

Major advances have been made in the past year towards an understanding of the structure and chemistry of copper chaperone proteins. Three-dimensional structures of Atx1, CopZ, yCCS, and hCCSdII were determined, and reveal a remarkable structural similarity between chaperones and target proteins. In addition, biochemical studies of CCS suggested that chaperones are required in vivo because intracellular copper concentrations are extremely low and also indicated that copper transfer occurs via a direct protein-protein interaction.


Assuntos
Cobre , Metaloproteínas/química , Chaperonas Moleculares/química , Sequência de Aminoácidos , Animais , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Superóxido Dismutase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA