Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 4731, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35961955

ABSTRACT

The human pathogen Mycobacterium tuberculosis requires a P1B-ATPase metal exporter, CtpC (Rv3270), for resistance to zinc poisoning. Here, we show that zinc resistance also depends on a chaperone-like protein, PacL1 (Rv3269). PacL1 contains a transmembrane domain, a cytoplasmic region with glutamine/alanine repeats and a C-terminal metal-binding motif (MBM). PacL1 binds Zn2+, but the MBM is required only at high zinc concentrations. PacL1 co-localizes with CtpC in dynamic foci in the mycobacterial plasma membrane, and the two proteins form high molecular weight complexes. Foci formation does not require flotillin nor the PacL1 MBM. However, deletion of the PacL1 Glu/Ala repeats leads to loss of CtpC and sensitivity to zinc. Genes pacL1 and ctpC appear to be in the same operon, and homologous gene pairs are found in the genomes of other bacteria. Furthermore, PacL1 colocalizes and functions redundantly with other PacL orthologs in M. tuberculosis. Overall, our results indicate that PacL proteins may act as scaffolds that assemble P-ATPase-containing metal efflux platforms mediating bacterial resistance to metal poisoning.


Subject(s)
Adenosine Triphosphatases , Mycobacterium tuberculosis , Adenosine Triphosphatases/metabolism , Biological Transport , Humans , Metals/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , Zinc/metabolism
2.
J Hazard Mater ; 422: 126894, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34416697

ABSTRACT

Uranium (U) is a naturally-occurring radionuclide that is toxic for all living organisms. To date, the mechanisms of U uptake are far from being understood. Here we provide a direct characterization of the transport machineries capable of transporting U, using the yeast Saccharomyces cerevisiae as a unicellular eukaryote model. First, we evidenced a metabolism-dependent U transport in yeast. Then, competition experiments with essential metals allowed us to identify calcium, iron and copper entry pathways as potential routes for U uptake. The analysis of various metal transport mutants revealed that mutant affected in calcium (mid1Δ and cch1Δ) and Fe(III) (ftr1Δ) transport, exhibited highly reduced U uptake rates and accumulation, demonstrating the implication of the calcium channel Mid1/Cch1 and the iron permease Ftr1 in U uptake. Finally, expression of the Mid1 gene into the mid1Δ mutant restored U uptake levels of the wild type strain, underscoring the central role of the Mid1/Cch1 calcium channel in U absorption process in yeast. Our results also open up the opportunity for rapid screening of U-transporter candidates by functional expression in yeast, before their validation in more complex higher eukaryote model systems.


Subject(s)
Membrane Glycoproteins/genetics , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Calcium/metabolism , Calcium Channels , Ferric Compounds/metabolism , Iron/metabolism , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
3.
Commun Biol ; 3(1): 46, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992852

ABSTRACT

The hexameric MoxR AAA+ ATPase RavA and the decameric lysine decarboxylase LdcI form a 3.3 MDa cage, proposed to assist assembly of specific respiratory complexes in E. coli. Here, we show that inside the LdcI-RavA cage, RavA hexamers adopt an asymmetric spiral conformation in which the nucleotide-free seam is constrained to two opposite orientations. Cryo-EM reconstructions of free RavA reveal two co-existing structural states: an asymmetric spiral, and a flat C2-symmetric closed ring characterised by two nucleotide-free seams. The closed ring RavA state bears close structural similarity to the pseudo two-fold symmetric crystal structure of the AAA+ unfoldase ClpX, suggesting a common ATPase mechanism. Based on these structures, and in light of the current knowledge regarding AAA+ ATPases, we propose different scenarios for the ATP hydrolysis cycle of free RavA and the LdcI-RavA cage-like complex, and extend the comparison to other AAA+ ATPases of clade 7.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Carboxy-Lyases/chemistry , Carboxy-Lyases/metabolism , Electron Transport Complex I/chemistry , Electron Transport Complex I/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Adenosine Diphosphate/metabolism , Catalytic Domain , Cryoelectron Microscopy , Crystallization , Crystallography, X-Ray , Escherichia coli/metabolism , Hydrogen-Ion Concentration , Hydrolysis , Protein Binding , Protein Conformation, alpha-Helical
4.
Metallomics ; 11(7): 1219-1229, 2019 07 17.
Article in English | MEDLINE | ID: mdl-31143907

ABSTRACT

Biogenic thiols, such as cysteine, have been used to control the speciation of Hg(ii) in bacterial exposure experiments. However, the extracellular biodegradation of excess cysteine leads to the formation of Hg(ii)-sulfide species, convoluting the interpretation of Hg(ii) uptake results. Herein, we test the hypothesis that Hg(ii)-sulfide species formation is a critical step during bacterial Hg(ii) uptake in the presence of excess cysteine. An Escherichia coli (E. coli) wild-type and mutant strain lacking the decR gene that regulates cysteine degradation to sulfide were exposed to 50 and 500 nM Hg with 0 to 2 mM cysteine. The decR mutant released ∼4 times less sulfide from cysteine degradation compared to the wild-type for all tested cysteine concentrations during a 3 hour exposure period. We show with thermodynamic calculations that the predicted concentration of Hg(ii)-cysteine species remaining in the exposure medium (as opposed to forming HgS(s)) is a good proxy for the measured concentration of dissolved Hg(ii) (i.e., not cell-bound). Likewise, the measured cell-bound Hg(ii) correlates with thermodynamic calculations for HgS(s) formation in the presence of cysteine. High resolution X-ray absorption near edge structure (HR-XANES) spectra confirm the existence of cell-associated HgS(s) at 500 nM total Hg and suggest the formation of Hg-S clusters at 50 nM total Hg. Our results indicate that a speciation change to Hg(ii)-sulfide controls Hg(ii) cell-association in the presence of excess cysteine.


Subject(s)
Cysteine/metabolism , Escherichia coli/metabolism , Mercury/metabolism , Sulfides/metabolism , Sulfur/metabolism , Biological Transport , Escherichia coli Infections/microbiology , Humans , Thermodynamics
5.
Biochem J ; 475(1): 289-303, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29229760

ABSTRACT

Schistosoma mansoni is a parasite that causes bilharzia, a neglected tropical disease affecting hundreds of millions of people each year worldwide. In 2012, S. mansoni had been identified as the only invertebrate possessing two SERCA-type Ca2+-ATPases, SMA1 and SMA2. However, our analysis of recent genomic data shows that the presence of two SERCA pumps is rather frequent in parasitic flatworms. To understand the reasons of this redundancy in S. mansoni, we compared SMA1 and SMA2 at different levels. In terms of sequence and organization, the genes SMA1 and SMA2 are similar, suggesting that they might be the result of a duplication event. At the protein level, SMA1 and SMA2 only slightly differ in length and in the sequence of the nucleotide-binding domain. To get functional information on SMA1, we produced it in an active form in Saccharomyces cerevisiae, as previously done for SMA2. Using phosphorylation assays from ATP, we demonstrated that like SMA2, SMA1 bound calcium in a cooperative mode with an apparent affinity in the micromolar range. We also showed that SMA1 and SMA2 had close sensitivities to cyclopiazonic acid but different sensitivities to thapsigargin, two specific inhibitors of SERCA pumps. On the basis of transcriptomic data available in GeneDB, we hypothesize that SMA1 is a housekeeping Ca2+-ATPase, whereas SMA2 might be required in particular striated-like muscles like those present the tail of the cercariae, the infecting form of the parasite.


Subject(s)
Calcium-Transporting ATPases/chemistry , Calcium/chemistry , Helminth Proteins/chemistry , Schistosoma mansoni/enzymology , Amino Acid Motifs , Animals , Calcium/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Calcium-Transporting ATPases/genetics , Calcium-Transporting ATPases/metabolism , Catalytic Domain , Cloning, Molecular , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/genetics , Helminth Proteins/metabolism , Indoles/chemistry , Indoles/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schistosoma mansoni/genetics , Thapsigargin/chemistry , Thapsigargin/metabolism
6.
PLoS One ; 11(11): e0165666, 2016.
Article in English | MEDLINE | ID: mdl-27802305

ABSTRACT

Copper is a crucial ion in cells, but needs to be closely controlled due to its toxic potential and ability to catalyse the formation of radicals. In chloroplasts, an important step for the proper functioning of the photosynthetic electron transfer chain is the delivery of copper to plastocyanin in the thylakoid lumen. The main route for copper transport to the thylakoid lumen is driven by two PIB-type ATPases, Heavy Metal ATPase 6 (HMA6) and HMA8, located in the inner membrane of the chloroplast envelope and in the thylakoid membrane, respectively. Here, the crystal structures of the nucleotide binding domain of HMA6 and HMA8 from Arabidopsis thaliana are reported at 1.5Å and 1.75Å resolution, respectively, providing the first structural information on plants Cu+-ATPases. The structures reveal a compact domain, with two short helices on both sides of a twisted beta-sheet. A double mutant, aiding in the crystallization, provides a new crystal contact, but also avoids an internal clash highlighting the benefits of construct modifications. Finally, the histidine in the HP motif of the isolated domains, unable to bind ATP, shows a side chain conformation distinct from nucleotide bound structures.


Subject(s)
Adenosine Triphosphatases/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Nucleotides/metabolism , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Binding Sites , Copper/metabolism , Crystallography, X-Ray , Models, Molecular , Protein Binding , Protein Domains , Sequence Alignment
7.
J Biol Chem ; 291(38): 20136-48, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27493208

ABSTRACT

Copper is an essential transition metal for living organisms. In the plant model Arabidopsis thaliana, half of the copper content is localized in the chloroplast, and as a cofactor of plastocyanin, copper is essential for photosynthesis. Within the chloroplast, copper delivery to plastocyanin involves two transporters of the PIB-1-ATPases subfamily: HMA6 at the chloroplast envelope and HMA8 in the thylakoid membranes. Both proteins are high affinity copper transporters but share distinct enzymatic properties. In the present work, the comparison of 140 sequences of PIB-1-ATPases revealed a conserved region unusually rich in histidine and cysteine residues in the TMA-L1 region of eukaryotic chloroplast copper ATPases. To evaluate the role of these residues, we mutated them in HMA6 and HMA8. Mutants of interest were selected from phenotypic tests in yeast and produced in Lactococcus lactis for further biochemical characterizations using phosphorylation assays from ATP and Pi Combining functional and structural data, we highlight the importance of the cysteine and the first histidine of the CX3HX2H motif in the process of copper release from HMA6 and HMA8 and propose a copper pathway through the membrane domain of these transporters. Finally, our work suggests a more general role of the histidine residue in the transport of copper by PIB-1-ATPases.


Subject(s)
Adenosine Triphosphatases/chemistry , Arabidopsis Proteins/chemistry , Arabidopsis/enzymology , Copper/chemistry , Thylakoid Membrane Proteins/chemistry , Thylakoids/enzymology , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Copper/metabolism , Histidine/chemistry , Histidine/genetics , Histidine/metabolism , Lactococcus lactis/genetics , Lactococcus lactis/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Thylakoid Membrane Proteins/genetics , Thylakoid Membrane Proteins/metabolism , Thylakoids/genetics
8.
Methods Mol Biol ; 1432: 79-101, 2016.
Article in English | MEDLINE | ID: mdl-27485331

ABSTRACT

Due to their unique properties, expression and study of membrane proteins in heterologous systems remains difficult. Among the bacterial systems available, the Gram-positive lactic bacterium, Lactococcus lactis, traditionally used in food fermentations, is nowadays widely used for large-scale production and functional characterization of bacterial and eukaryotic membrane proteins. The aim of this chapter is to describe the different possibilities for the functional characterization of peripheral or intrinsic membrane proteins expressed in Lactococcus lactis.


Subject(s)
Lactococcus lactis/growth & development , Membrane Proteins/metabolism , Protein Engineering/methods , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Gene Expression , Genetic Vectors , Lactococcus lactis/genetics , Membrane Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
9.
Biosci Rep ; 35(3)2015 Apr 20.
Article in English | MEDLINE | ID: mdl-26182363

ABSTRACT

Copper (Cu) plays a key role in the photosynthetic process as cofactor of the plastocyanin (PC), an essential component of the chloroplast photosynthetic electron transfer chain. Encoded by the nuclear genome, PC is translocated in its apo-form into the chloroplast and the lumen of thylakoids where it is processed to its mature form and acquires Cu. In Arabidopsis, Cu delivery into the thylakoids involves two transporters of the PIB-1 ATPases family, heavy metal associated protein 6 (HMA6) located at the chloroplast envelope and HMA8 at the thylakoid membrane. To gain further insight into the way Cu is delivered to PC, we analysed the enzymatic properties of HMA8 and compared them with HMA6 ones using in vitro phosphorylation assays and phenotypic tests in yeast. These experiments reveal that HMA6 and HMA8 display different enzymatic properties: HMA8 has a higher apparent affinity for Cu(+) but a slower dephosphorylation kinetics than HMA6. Modelling experiments suggest that these differences could be explained by the electrostatic properties of the Cu(+) releasing cavities of the two transporters and/or by the different nature of their cognate Cu(+) acceptors (metallochaperone/PC).


Subject(s)
Adenosine Triphosphatases/metabolism , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/metabolism , Copper/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Arabidopsis Proteins/genetics , Chloroplast Proton-Translocating ATPases/metabolism , Copper/pharmacology , Lactococcus/genetics , Molecular Docking Simulation , Phosphorylation , Plastocyanin/chemistry , Plastocyanin/metabolism , Protein Conformation , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Thylakoids/metabolism
10.
ACS Chem Biol ; 9(12): 2779-86, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25238402

ABSTRACT

The FUR protein (ferric uptake regulator) is an iron-dependent global transcriptional regulator. Specific to bacteria, FUR is an attractive antibacterial target since virulence is correlated to iron bioavailability. Recently, four anti-FUR peptide aptamers, composed of 13 amino acid variable loops inserted into a thioredoxinA scaffold, were identified, which were able to interact with Escherichia coli FUR (EcFUR), inhibit its binding to DNA and to decrease the virulence of pathogenic E. coli in a fly infection model. The first characterization of anti-FUR linear peptides (pF1 6 to 13 amino acids) derived from the variable part of the F1 anti-FUR peptide aptamer is described herein. Theoretical and experimental approaches, in original combination, were used to study interactions of these peptides with FUR in order to understand their mechanism of inhibition. After modeling EcFUR by homology, docking with Autodock was combined with molecular dynamics simulations in implicit solvent to take into account the flexibility of the partners. All calculations were cross-checked either with other programs or with experimental data. As a result, reliable structures of EcFUR and its complex with pF1 are given and an inhibition pocket formed by the groove between the two FUR subunits is proposed. The location of the pocket was validated through experimental mutation of key EcFUR residues at the site of proposed peptide interaction. Cyclisation of pF1, mimicking the peptide constraint in F1, improved inhibition. The details of the interactions between peptide and protein were analyzed and a mechanism of inhibition of these anti-FUR molecules is proposed.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Aptamers, Peptide/chemistry , Bacterial Proteins/chemistry , Escherichia coli/chemistry , Iron/chemistry , Repressor Proteins/chemistry , Thioredoxins/chemistry , Amino Acid Sequence , Antimicrobial Cationic Peptides/chemical synthesis , Aptamers, Peptide/chemical synthesis , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Iron/metabolism , Kinetics , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Structure-Activity Relationship , Thermodynamics , Thioredoxins/genetics , Thioredoxins/metabolism
12.
Protein Expr Purif ; 83(2): 211-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22542587

ABSTRACT

ATAD3 is a mitochondrial integral inner membrane ATPase with unknown function. ATAD3 is absent in yeast and protozoan and present in all pluricellular eucaryotes where its expression is essential for development. To date, bacterial-based expression of full-length ATAD3 has been unsuccessful because of very high levels of endogenous degradation. Based on Saccharomyces cerevisiae as a heterogeneous expression system, we engineered a high copy strain expressing human ATAD3A-Myc-HIS at a relative high level (2.5mg/l of yeast culture) without significantly affecting yeast growth. Most of the expressed human ATAD3A-Myc-HIS co-purified with the yeast mitochondrial fraction thus suggesting that targeting to this organelle is preserved in yeast. Like the endogenous protein in human cells, ATAD3A-Myc-HIS expressed in yeast is found resistant to extraction with salt and certain detergents, suggesting membrane insertion. Sarkosyl, C13-DAO, C12-DAO and ONMG efficiently solubilized ATAD3A-Myc-HIS from yeast extracts, but these soluble species did not bind to agarose-nickel matrix. By contrast, urea-denaturated ATAD3A-Myc-HIS bound to agarose-nickel beads and could be renatured and eluted to obtain highly pure ATAD3A-Myc-HIS. As the native protein in vivo, this recombinant, renatured species specifically bound in vitro to S100B and S100A1 in Far-Western assays.


Subject(s)
Adenosine Triphosphatases/isolation & purification , Histidine/metabolism , Membrane Proteins/isolation & purification , Mitochondrial Proteins/isolation & purification , Nerve Growth Factors/metabolism , Recombinant Fusion Proteins/isolation & purification , S100 Proteins/metabolism , Saccharomyces cerevisiae/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Blotting, Far-Western , Histidine/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Protein Denaturation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , S100 Calcium Binding Protein beta Subunit , Urea/metabolism
13.
J Mol Biol ; 415(3): 584-99, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22119486

ABSTRACT

The accumulation of amyloid fibers due to protein misfolding is associated with numerous human diseases. For example, the formation of amyloid deposits in neurodegenerative pathologies is correlated with abnormal apoptosis. We report here the in vitro formation of various types of aggregates by Bcl-xL, a protein of the Bcl-2 family involved in the regulation of apoptosis. Bcl-xL forms aggregates in three states, micelles, native-like fibrils, and amyloid fibers, and their biophysical characterization has been performed in detail. Bcl-xL remains in its native state within micelles and native-like fibrils, and our results suggest that native-like fibrils are formed by the association of micelles. Formation of amyloid structures, that is, nonnative intermolecular ß-sheets, is favored by the proximity of proteins within fibrils at the expense of the Bcl-xL native structure. Finally, we provide evidence of a direct relationship between the amyloid character of the fibers and the tertiary-structure stability of the native Bcl-xL. The potential causality between the accumulation of Bcl-xL into amyloid deposits and abnormal apoptosis during neurodegenerative diseases is discussed.


Subject(s)
Amyloid/metabolism , bcl-X Protein/metabolism , Amyloid/chemistry , Amyloid/ultrastructure , Humans , Microscopy, Electron , Models, Molecular , Protein Conformation , Protein Denaturation , Protein Multimerization , Protein Stability , bcl-X Protein/chemistry
14.
J Biol Chem ; 286(42): 36188-97, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-21878617

ABSTRACT

Copper is an essential plant micronutrient playing key roles in cellular processes, among them photosynthesis. In Arabidopsis thaliana, copper delivery to chloroplasts, mainly studied by genetic approaches, is thought to involve two P(IB)-type ATPases: AtHMA1 and AtHMA6/PAA1. The lack of biochemical characterization of AtHMA1 and PAA1, and more generally of plant P(IB)-type ATPases, is due to the difficulty of getting high amounts of these membrane proteins in an active form, either from their native environment or after expression in heterologous systems. In this study, we report the first biochemical characterization of PAA1, a plant copper-transporting ATPase. PAA1 produced in Lactococcus lactis is active, forming an aspartyl phosphate intermediate in the presence of ATP and the adequate metal ion. PAA1 can also be phosphorylated using inorganic phosphate in the absence of transition metal. Both phosphorylation types allowed us to demonstrate that PAA1 is activated by monovalent copper ions (and to a lower extent by silver ions) with an apparent affinity in the micromolar range. In agreement with these biochemical data, we also demonstrate that when expressed in yeast, PAA1 induces increased sensitivities to copper and silver. These data provide the first enzymatic characterization of a P(IB-1)-type plant ATPase and clearly identify PAA1 as a high affinity Cu(I) transporter of the chloroplast envelope.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/enzymology , Chloroplast Proton-Translocating ATPases/metabolism , Chloroplasts/enzymology , Copper/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/chemistry , Arabidopsis Proteins/genetics , Cations, Monovalent/metabolism , Chloroplast Proton-Translocating ATPases/chemistry , Chloroplast Proton-Translocating ATPases/genetics , Chloroplasts/genetics , Ion Transport/physiology , Lactococcus lactis/enzymology , Lactococcus lactis/genetics
15.
Mol Microbiol ; 76(4): 1034-48, 2010 May.
Article in English | MEDLINE | ID: mdl-20444096

ABSTRACT

Cadmium (Cd(2+)) is a very toxic metal that causes DNA damage, oxidative stress and apoptosis. Despite many studies, the cellular and molecular mechanisms underlying its high toxicity are not clearly understood. We show here that very low doses of Cd(2+) cause ER stress in Saccharomyces cerevisiae as evidenced by the induction of the unfolded protein response (UPR) and the splicing of HAC1 mRNA. Furthermore, mutant strains (Delta ire1 and Delta hac1) unable to induce the UPR are hypersensitive to Cd(2+), but not to arsenite and mercury. The full functionality of the pathways involved in ER stress response is required for Cd(2+) tolerance. The data also suggest that Cd(2+)-induced ER stress and Cd(2+) toxicity are a direct consequence of Cd(2+) accumulation in the ER. Cd(2+) does not inhibit disulfide bond formation but perturbs calcium metabolism. In particular, Cd(2+) activates the calcium channel Cch1/Mid1, which also contributes to Cd(2+) entry into the cell. The results reinforce the interest of using yeast as a cellular model to study toxicity mechanisms in eukaryotic cells.


Subject(s)
Cadmium/toxicity , Endoplasmic Reticulum/drug effects , Saccharomyces cerevisiae/drug effects , Stress, Physiological , Cadmium/metabolism , Calcium Channels/metabolism , Drug Resistance, Fungal , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/agonists , Membrane Glycoproteins/metabolism , Protein Folding , Saccharomyces cerevisiae/ultrastructure , Saccharomyces cerevisiae Proteins/agonists , Saccharomyces cerevisiae Proteins/metabolism
16.
J Biol Chem ; 281(40): 29533-41, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-16835223

ABSTRACT

CadA, the Cd(2+)-ATPase from Listeria monocytogenes, belongs to the Zn(2+)/Cd(2+)/Pb(2+)-ATPase bacterial subfamily of P(1B)-ATPases that ensure detoxification of the bacteria. Whereas it is the major determinant of Listeria resistance to Cd(2+), CadA expressed in Saccharomyces cerevisiae severely decreases yeast tolerance to Cd(2+) (Wu, C. C., Bal, N., Pérard, J., Lowe, J., Boscheron, C., Mintz, E., and Catty, P. (2004) Biochem. Biophys. Res. Commun. 324, 1034-1040). This phenotype, which reflects in vivo Cd(2+)-transport activity, was used to select from 33 point mutations, shared out among the eight transmembrane (TM) segments of CadA, those that affect the activity of the protein. Six mutations affecting CadA were found: M149A in TM3; E164A in TM4; C354A, P355A, and C356A in TM6; and D692A in TM8. Functional studies of the six mutants produced in Sf9 cells revealed that Cys(354) and Cys(356) in TM6 as well as Asp(692) in TM8 and Met(149) in TM3 could participate at the Cd(2+)-binding site(s). In the canonical Cys-Pro-Cys motif of P(1B)-ATPases, the two cysteines act at distinct steps in the transport mechanism, Cys(354) being directly involved in Cd(2+) binding, while Cys(356) seems to be required for Cd(2+) occlusion. This confirms an earlier observation that the two equivalent Cys of Ccc2, the yeast Cu(+)-ATPase, also act at different steps. In TM4, Glu(164), which is conserved among P(1B)-ATPases, may be required for Cd(2+) release. Finally, analysis of the role of Cd(2+) in the phosphorylation from ATP and from P(i) of the mutants suggests that two Cd(2+) ions are involved in the reaction cycle of CadA.


Subject(s)
Adenosine Triphosphatases/metabolism , Cadmium/metabolism , Listeria monocytogenes/enzymology , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Amino Acid Substitution/genetics , Binding Sites , Biological Transport, Active/genetics , Listeria monocytogenes/genetics , Molecular Sequence Data , Phosphorylation , Protein Binding/genetics
17.
J Mol Biol ; 356(3): 638-50, 2006 Feb 24.
Article in English | MEDLINE | ID: mdl-16388822

ABSTRACT

In bacteria, P1-type ATPases are responsible for resistance to di- and monovalent toxic heavy metals by taking them out of the cell. These ATPases have a cytoplasmic N terminus comprising metal binding domains defined by a betaalphabetabetaalphabeta fold and a CXXC metal binding motif. To check how the structural properties of the metal binding site in the N terminus can influence the metal specificity of the ATPase, the first structure of a Cd(II)-ATPase N terminus was determined by NMR and its coordination sphere was investigated by X-ray absorption spectroscopy. A novel metal binding environment was found, comprising the two conserved Cys residues of the metal binding motif and a Glu in loop 5. A bioinformatic search identifies an ensemble of highly homologous sequences presumably with the same function. Another group of highly homologous sequences is found which can be referred to as zinc-detoxifying P1-type ATPases with the metal binding pattern DCXXC in the N terminus. Because no carboxylate groups participate in Cu(I) or Ag(I) binding sites, we suggest that the acidic residue plays a key role in the coordination properties of divalent cations, hence conferring a function to the N terminus in the metal specificity of the ATPase.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Cadmium/metabolism , Listeria monocytogenes/enzymology , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Apoenzymes/chemistry , Apoenzymes/metabolism , Biological Transport, Active , Cadmium/chemistry , Cation Transport Proteins/genetics , Cations, Divalent/chemistry , Cations, Divalent/metabolism , Copper-Transporting ATPases , Crystallography, X-Ray , Dimerization , Escherichia coli/enzymology , Escherichia coli/genetics , Listeria monocytogenes/genetics , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Solutions , Thermodynamics
18.
Biochem Biophys Res Commun ; 324(3): 1034-40, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15485658

ABSTRACT

CadA, the P1-type ATPase involved in Listeria monocytogenes resistance to Cd(2+), was expressed in Saccharomyces cerevisiae and did just the opposite to what was expected, as it strikingly decreased the Cd(2+) tolerance of these cells. Yeast cells expressing the non-functional mutant Asp(398)Ala could grow on selective medium containing up to 100 microM Cd(2+), whereas those expressing the functional protein could not grow in the presence of 1 microM Cd(2+). The CadA-GFP fusion protein was localized in the endoplasmic reticulum membrane, suggesting that yeast hyper-sensitivity was due to Cd(2+) accumulation in the reticulum lumen. CadA is also known to transport Zn(2+), but Zn(2+) did not protect the cells against Cd(2+) poisoning. In the presence of 10 microM Cd(2+), transformed yeasts survived by rapid loss of their expression vector.


Subject(s)
Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/physiology , Listeria monocytogenes/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/chemistry , Alanine/chemistry , Aspartic Acid/chemistry , Cadmium/chemistry , Cadmium/metabolism , Culture Media/metabolism , Culture Media/pharmacology , Endoplasmic Reticulum/metabolism , Genetic Vectors , Green Fluorescent Proteins/metabolism , Listeria monocytogenes/enzymology , Metals/chemistry , Phenotype , Phosphorylation , Saccharomyces cerevisiae/metabolism , Zinc/chemistry
19.
J Biol Chem ; 279(25): 25986-94, 2004 Jun 18.
Article in English | MEDLINE | ID: mdl-15078884

ABSTRACT

Ccc2p is homologous to the human Menkes and Wilson copper ATPases and is herein studied as a model for human copper transport. Most studies to date have sought to understand how mutations in the human Menkes or Wilson genes impair copper homeostasis and induce disease. Here we analyze whether eight conserved amino acids of the transmembrane domain are important for copper transport. Wild-type Ccc2p and variants were expressed in a ccc2-Delta yeast strain to check whether they were able to restore copper transport by complementation. Wild-type Ccc2p and variants were also expressed in Sf9 cells using baculovirus to study their enzymatic properties on membrane preparations. The latter system allowed us to measure a copper-activated ATPase activity of about 20 nmol/mg/min for the wild-type Ccc2p at 37 degrees C. None of the variants was as efficient as the wild type in restoring copper homeostasis. The mutation of each cysteine of the (583)CPC(585) motif into a serine resulted in nonfunctional proteins that could not restore copper homeostasis in yeast and had no ATPase activity. Phosphorylation by ATP was still possible with the C583S variant, although it was not possible with the C585S variant, suggesting that the cysteines of the CPC motif have a different role in copper transport. Cys(583) would be necessary for copper dissociation and/or enzyme dephosphorylation and Cys(585) would be necessary for ATP phosphorylation, suggesting a role in copper binding.


Subject(s)
Adenosine Triphosphatases/chemistry , Cation Transport Proteins/chemistry , Cation Transport Proteins/physiology , Cysteine/chemistry , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/physiology , Adenosine Triphosphatases/physiology , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Biological Transport , Cell Line , Cell Membrane/metabolism , Chelating Agents/pharmacology , Copper/chemistry , Copper Transport Proteins , Copper-Transporting ATPases , Cytoplasm/metabolism , Dose-Response Relationship, Drug , Genetic Complementation Test , Insecta , Ligands , Models, Biological , Molecular Sequence Data , Mutation , Phenanthrolines/pharmacology , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Quinolines/pharmacology , Saccharomyces cerevisiae/metabolism , Temperature , Time Factors
20.
Biochem J ; 369(Pt 3): 681-5, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12383056

ABSTRACT

CadA, the Cd(2+)-ATPase of Listeria monocytogenes, contains four cysteine residues: two in the CTNC (Cys-Thr-Asn-Cys) sequence in the cytoplasmic metal-binding domain (MBD), and two in the CPC (Cys-Pro-Cys) sequence in the membrane domain. Taking advantage of DeltaMBD, a truncated version of CadA that lacks the MBD but which still acts as a functional Cd(2+)-ATPase [Bal, Mintz, Guillain and Catty (2001) FEBS Lett. 506, 249-252], we analysed the role of the membrane cysteine residues (studied using DeltaMBD) separately from that of the cysteine residues of the MBD, which were studied using full-length CadA. The role of the cysteines was assessed by reacting DeltaMBD and CadA with N -ethylmaleimide (NEM), an SH-specific reagent, in the presence or absence of Cd(2+). We show here that (i) in both DeltaMBD and CadA, the cysteine residues in the CPC motif are essential for phosphorylation; (ii) in both proteins, Cd(2+) protects against alkylation by NEM; and (iii) in the absence of Cd(2+), the MBD of CadA also protects against alkylation by NEM. Our results suggest that the CPC motif is present in the membrane Cd(2+) transport site(s) and that the MBD protects these site(s).


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Cadmium/metabolism , Listeria monocytogenes/enzymology , Adenosine Triphosphatases/genetics , Amino Acid Motifs , Biological Transport, Active , Cell Membrane , Cysteine/chemistry , Ethylmaleimide/chemistry , Phosphorylation , Protein Structure, Tertiary , Sulfhydryl Reagents/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...