Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 991, 2022 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-35181664

RESUMO

The homo-dimeric bacterial membrane protein EmrE effluxes polyaromatic cationic substrates in a proton-coupled manner to cause multidrug resistance. We recently determined the structure of substrate-bound EmrE in phospholipid bilayers by measuring hundreds of protein-ligand HN-F distances for a fluorinated substrate, 4-fluoro-tetraphenylphosphonium (F4-TPP+), using solid-state NMR. This structure was solved at low pH where one of the two proton-binding Glu14 residues is protonated. Here, to understand how substrate transport depends on pH, we determine the structure of the EmrE-TPP complex at high pH, where both Glu14 residues are deprotonated. The high-pH complex exhibits an elongated and hydrated binding pocket in which the substrate is similarly exposed to the two sides of the membrane. In contrast, the low-pH complex asymmetrically exposes the substrate to one side of the membrane. These pH-dependent EmrE conformations provide detailed insights into the alternating-access model, and suggest that the high-pH conformation may facilitate proton binding in the presence of the substrate, thus accelerating the conformational change of EmrE to export the substrate.


Assuntos
Antiporters/metabolismo , Proteínas de Escherichia coli/metabolismo , Prótons , Antiporters/ultraestrutura , Farmacorresistência Bacteriana Múltipla , Proteínas de Escherichia coli/ultraestrutura , Concentração de Íons de Hidrogênio , Simulação de Acoplamento Molecular , Ressonância Magnética Nuclear Biomolecular , Oniocompostos/metabolismo , Compostos Organofosforados/metabolismo
2.
Sci Rep ; 11(1): 12328, 2021 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-34112901

RESUMO

Boron has essential roles in plant growth and development. BOR proteins are key in the active uptake and distribution of boron, and regulation of intracellular boron concentrations. However, their mechanism of action remains poorly studied. BOR proteins are homologues of the human SLC4 family of transporters, which includes well studied mammalian transporters such as the human Anion Exchanger 1 (hAE1). Here we generated Arabidopsis thaliana BOR1 (AtBOR1) variants based (i) on known disease causing mutations of hAE1 (S466R, A500R) and (ii) a loss of function mutation (D311A) identified in the yeast BOR protein, ScBOR1p. The AtBOR1 variants express in yeast and localise to the plasma membrane, although both S466R and A500R exhibit lower expression than the WT AtBOR1 and D311A. The D311A, S466R and A500R mutations result in a loss of borate efflux activity in a yeast bor1p knockout strain. A. thaliana plants containing these three individual mutations exhibit substantially decreased growth phenotypes in soil under conditions of low boron. These data confirm an important role for D311 in the function of the protein and show that mutations equivalent to disease-causing mutations in hAE1 have major effects in AtBOR1. We also obtained a low resolution cryo-EM structure of a BOR protein from Oryza sativa, OsBOR3, lacking the 30 C-terminal amino acid residues. This structure confirms the gate and core domain organisation previously observed for related proteins, and is strongly suggestive of an inward facing conformation.


Assuntos
Proteína 1 de Troca de Ânion do Eritrócito/genética , Antiporters/genética , Proteínas de Arabidopsis/genética , Proteínas de Membrana Transportadoras/genética , Desenvolvimento Vegetal/genética , Proteínas de Saccharomyces cerevisiae/genética , Antiporters/ultraestrutura , Arabidopsis/genética , Arabidopsis/crescimento & desenvolvimento , Arabidopsis/metabolismo , Proteínas de Arabidopsis/ultraestrutura , Boratos/metabolismo , Boro/metabolismo , Regulação da Expressão Gênica de Plantas , Humanos , Transporte de Íons/genética , Mutação , Oryza/genética , Oryza/crescimento & desenvolvimento , Saccharomyces cerevisiae/genética
3.
J Biol Chem ; 296: 100262, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33837745

RESUMO

In both prokaryotes and eukaryotes, multidrug and toxic-compound extrusion (MATE) transporters catalyze the efflux of a broad range of cytotoxic compounds, including human-made antibiotics and anticancer drugs. MATEs are secondary-active antiporters, i.e., their drug-efflux activity is coupled to, and powered by, the uptake of ions down a preexisting transmembrane electrochemical gradient. Key aspects of this mechanism, however, remain to be delineated, such as its ion specificity and stoichiometry. We previously revealed the existence of a Na+-binding site in a MATE transporter from Pyroccocus furiosus (PfMATE) and hypothesized that this site might be broadly conserved among prokaryotic MATEs. Here, we evaluate this hypothesis by analyzing VcmN and ClbM, which along with PfMATE are the only three prokaryotic MATEs whose molecular structures have been determined at atomic resolution, i.e. better than 3 Å. Reinterpretation of existing crystallographic data and molecular dynamics simulations indeed reveal an occupied Na+-binding site in the N-terminal lobe of both structures, analogous to that identified in PfMATE. We likewise find this site to be strongly selective against K+, suggesting it is mechanistically significant. Consistent with these computational results, DEER spectroscopy measurements for multiple doubly-spin-labeled VcmN constructs demonstrate Na+-dependent changes in protein conformation. The existence of this binding site in three MATE orthologs implicates Na+ in the ion-coupled drug-efflux mechanisms of this class of transporters. These results also imply that observations of H+-dependent activity likely stem either from a site elsewhere in the structure, or from H+ displacing Na+ under certain laboratory conditions, as has been noted for other Na+-driven transport systems.


Assuntos
Antiporters/química , Proteínas de Transporte de Cátions Orgânicos/química , Conformação Proteica/efeitos dos fármacos , Sódio/química , Antibacterianos/efeitos adversos , Antibacterianos/farmacologia , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Antiporters/ultraestrutura , Sítios de Ligação/efeitos dos fármacos , Cristalografia por Raios X , Humanos , Íons/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Proteínas de Transporte de Cátions Orgânicos/ultraestrutura , Células Procarióticas/química , Células Procarióticas/ultraestrutura , Domínios Proteicos/efeitos dos fármacos
4.
Elife ; 92020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32735215

RESUMO

Multiple resistance and pH adaptation (Mrp) antiporters are multi-subunit Na+ (or K+)/H+ exchangers representing an ancestor of many essential redox-driven proton pumps, such as respiratory complex I. The mechanism of coupling between ion or electron transfer and proton translocation in this large protein family is unknown. Here, we present the structure of the Mrp complex from Anoxybacillus flavithermus solved by cryo-EM at 3.0 Å resolution. It is a dimer of seven-subunit protomers with 50 trans-membrane helices each. Surface charge distribution within each monomer is remarkably asymmetric, revealing probable proton and sodium translocation pathways. On the basis of the structure we propose a mechanism where the coupling between sodium and proton translocation is facilitated by a series of electrostatic interactions between a cation and key charged residues. This mechanism is likely to be applicable to the entire family of redox proton pumps, where electron transfer to substrates replaces cation movements.


Assuntos
Anoxybacillus/metabolismo , Antiporters/metabolismo , Proteínas de Bactérias/metabolismo , Antiporters/ultraestrutura , Proteínas de Bactérias/ultraestrutura , Transporte Biológico Ativo , Cátions/metabolismo , Microscopia Crioeletrônica , Escherichia coli , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/ultraestrutura , Conformação Proteica , Prótons , Sódio/metabolismo
5.
Elife ; 82019 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-31339488

RESUMO

The epithelial anion transporter SLC26A9 contributes to airway surface hydration and gastric acid production. Colocalizing with CFTR, SLC26A9 has been proposed as a target for the treatment of cystic fibrosis. To provide molecular details of its transport mechanism, we present cryo-EM structures and a functional characterization of murine Slc26a9. These structures define the general architecture of eukaryotic SLC26 family members and reveal an unusual mode of oligomerization which relies predominantly on the cytosolic STAS domain. Our data illustrates conformational transitions of Slc26a9, supporting a rapid alternate-access mechanism which mediates uncoupled chloride transport with negligible bicarbonate or sulfate permeability. The characterization of structure-guided mutants illuminates the properties of the ion transport path, including a selective anion binding site located in the center of a mobile module within the transmembrane domain. This study thus provides a structural foundation for the understanding of the entire SLC26 family and potentially facilitates their therapeutic exploitation.


Assuntos
Antiporters/metabolismo , Antiporters/ultraestrutura , Cloretos/metabolismo , Microscopia Crioeletrônica , Transportadores de Sulfato/metabolismo , Transportadores de Sulfato/ultraestrutura , Animais , Antiporters/química , Sítios de Ligação , Células HEK293 , Humanos , Transporte de Íons , Camundongos , Modelos Moleculares , Domínios Proteicos , Proteolipídeos/metabolismo , Eletricidade Estática , Especificidade por Substrato , Transportadores de Sulfato/química
6.
Biochim Biophys Acta ; 1848(10 Pt A): 2065-74, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26014489

RESUMO

EmrE protein transports positively charged aromatic drugs (xenobiotics) in exchange for two protons and thus provides bacteria resistance to variety of drugs. In order to understand how this protein may recognize ligands, the monomer and asymmetric apo-form of the EmrE dimer embedded in a heterogeneous phospholipid (POPE+POPG) membrane were studied by molecular dynamics simulations. Dimer is regarded as a functional form of the transporter, but to understand molecular aspects of its mode of action, a monomer was also included in our work. We analyzed hydrogen bonds which include inter- and intra-molecular interactions. Analyzing the long-lasting H-bond interactions, we found that water access to the internal transmembrane segments is regulated by residues with aromatic or basic side chains and fluctuating transmembrane helices. Our finding supports that GLU14 in EmrE apo-form is ready to interact or bind with substrate molecule. The analysis of distance center of masses and water entrance area indicate the feasibility of the dimer to undergo induced fit in order to accommodate a ligand. The results indicate that a binding pattern can be formed in the EmrE in such a way that GLU14 binds to the positively charged fragment of a substrate molecule, and other aromatic residues (i.e., TRP63 and TYR40) located in vicinity may accommodate other non-polar parts of substrate molecule. The results of our simulation also allow us to support experimentally testable hypotheses concerning functional inward-outward conformational changes of the protein.


Assuntos
Antiporters/química , Antiporters/ultraestrutura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/ultraestrutura , Bicamadas Lipídicas/química , Modelos Químicos , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Sítios de Ligação , Simulação por Computador , Dados de Sequência Molecular , Ligação Proteica , Conformação Proteica
7.
J Biomol NMR ; 61(3-4): 209-26, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631353

RESUMO

CLC transporters catalyze the exchange of Cl(-) for H(+) across cellular membranes. To do so, they must couple Cl(-) and H(+) binding and unbinding to protein conformational change. However, the sole conformational changes distinguished crystallographically are small movements of a glutamate side chain that locally gates the ion-transport pathways. Therefore, our understanding of whether and how global protein dynamics contribute to the exchange mechanism has been severely limited. To overcome the limitations of crystallography, we used solution-state (13)C-methyl NMR with labels on methionine, lysine, and engineered cysteine residues to investigate substrate (H(+)) dependent conformational change outside the restraints of crystallization. We show that methyl labels in several regions report H(+)-dependent spectral changes. We identify one of these regions as Helix R, a helix that extends from the center of the protein, where it forms the part of the inner gate to the Cl(-)-permeation pathway, to the extracellular solution. The H(+)-dependent spectral change does not occur when a label is positioned just beyond Helix R, on the unstructured C-terminus of the protein. Together, the results suggest that H(+) binding is mechanistically coupled to closing of the intracellular access-pathway for Cl(-).


Assuntos
Antiporters/ultraestrutura , Espectroscopia de Ressonância Magnética Nuclear de Carbono-13/métodos , Antiportadores de Cloreto-Bicarbonato/ultraestrutura , Proteínas de Escherichia coli/ultraestrutura , Ressonância Magnética Nuclear Biomolecular/métodos , Radioisótopos de Carbono , Cristalografia por Raios X , Cisteína/química , Escherichia coli/metabolismo , Lisina/química , Metionina/química , Metilação , Modelos Moleculares , Conformação Proteica , Estrutura Terciária de Proteína
8.
J Biol Chem ; 290(2): 805-14, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25406320

RESUMO

Secondary active transporters undergo large conformational changes to facilitate the efflux of substrates across the lipid bilayer. Among the smallest known transport proteins are members of the small multidrug resistance (SMR) family that are composed of four transmembrane (TM) domains and assemble into dimers. An unanswered question in the SMR field is how the dimerization domain (TM4) is coupled with the substrate-binding chamber (TM1-3). To provide insight for this essential aspect of ion-coupled transport, we carried out a structure-function study on the SMR protein EmrE using solid-state NMR spectroscopy in lipid bilayers and resistance assays in Escherichia coli. The chemical shifts for EmrE were consistent with ß-strand secondary structure for the loop connecting TM3 and TM4. Based on these structural results, EmrE mutants were created to ascertain whether a specific loop length and composition were necessary for function. A linker encompassing six extra Gly residues relative to wild-type EmrE failed to give resistance; however, the number of residues in the loop was not the only criterion for a functional efflux pump. Replacement of the central hydrophobic residue with Gly (L83G) also conferred no ethidium resistance phenotype, which supported the conclusion that the structure and length of the loop were both essential for ion-coupled transport. Taken together with a bioinformatics analysis, a structured linker is likely conserved across the SMR family to play an active role in mediating the conformational switch between inward-open and outward-open states necessary for drug efflux. These findings underscore the important role loops can play in mediating efflux.


Assuntos
Antiporters/química , Resistência a Múltiplos Medicamentos/genética , Proteínas de Escherichia coli/química , Bicamadas Lipídicas/química , Relação Estrutura-Atividade , Antiporters/genética , Antiporters/ultraestrutura , Sítios de Ligação , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/ultraestrutura , Bicamadas Lipídicas/metabolismo , Espectroscopia de Ressonância Magnética , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Estrutura Terciária de Proteína/genética , Especificidade por Substrato
9.
Chem Phys Lipids ; 167-168: 33-42, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23384478

RESUMO

Understanding lipid-protein interactions to enhance our knowledge of membrane architecture is a critical step in the development of novel therapeutic measures to respond to the drastic rise of drug resistant microorganisms. Escherichia coli contains a small archetypal inner membrane multidrug resistance protein, EmrE, that must multimerize to be functional but this multimerization is difficult to demonstrate in vivo. We studied three major E. coli lipids (phosphatidylethanolamine, phosphatidylglycerol and cardiolipin) that varied in head group structure, acyl chain length and saturation. These were investigated both in the presence and absence of EmrE to determine which lipid(s) EmrE influenced most strongly. Langmuir monolayers and Brewster angle microscopy demonstrated that varying each head group, acyl chain length and saturation contributed to differences in membrane packing and affected lipid-protein associations. Long unsaturated anionic lipids were influenced most strongly by EmrE. Shorter acyl chains initiated string-like formations of EmrE clusters, whereas longer chains contributed to enhance protein clustering. Longer partially unsaturated acyl chains in phosphatidylglycerol showed a significant surface pressure decrease in the presence of the protein, indicating that the monolayer was destabilized. Interestingly, longer unsaturated chains of cardiolipin formed the most stable monolayer in the presence of EmrE. These studies indicate cardiolipin acyl chains that hydrophobically match protein helical lengths stabilize EmrE structural forms.


Assuntos
Antiporters/ultraestrutura , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/ultraestrutura , Escherichia coli/citologia , Lipídeos de Membrana/química , Antiporters/análise , Cardiolipinas/química , Farmacorresistência Bacteriana , Infecções por Escherichia coli/tratamento farmacológico , Proteínas de Escherichia coli/análise , Humanos , Fosfatidiletanolaminas/química , Fosfatidilgliceróis/química
10.
Acta Crystallogr D Biol Crystallogr ; 65(Pt 2): 186-92, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19171974

RESUMO

The archetypical member of the small multidrug-resistance family is EmrE, a multidrug transporter that extrudes toxic polyaromatic cations from the cell coupled to the inward movement of protons down a concentration gradient. The architecture of EmrE was first defined from the analysis of two-dimensional crystals by cryoelectron microscopy (cryo-EM), which showed that EmrE was an unusual asymmetric dimer formed from a bundle of eight alpha-helices. The most favoured interpretation of the structure was that the monomers were oriented in opposite orientations in the membrane in an antiparallel orientation. A model was subsequently built based upon the cryo-EM data and evolutionary constraints and this model was consistent with mutagenic data indicating which amino-acid residues were important for substrate binding and transport. Two X-ray structures that differed significantly from the cryo-EM structure were subsequently retracted owing to a data-analysis error. However, the revised X-ray structure with substrate bound is extremely similar to the model built from the cryo-EM structure (r.m.s.d. of 1.4 A), suggesting that the proposed antiparallel orientation of the monomers is indeed correct; this represents a new structural paradigm in membrane-protein structures. The vast majority of mutagenic and biochemical data corroborate this structure, although cross-linking studies and recent EPR data apparently support a model of EmrE that contains parallel dimers.


Assuntos
Antiporters/química , Proteínas de Escherichia coli/química , Antiporters/ultraestrutura , Microscopia Crioeletrônica , Cristalografia por Raios X , Proteínas de Escherichia coli/ultraestrutura , Estrutura Secundária de Proteína
11.
Biochim Biophys Acta ; 1768(12): 3036-43, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17976529

RESUMO

We study the uniformly 13C,15N isotopically enriched Escherichia coli multidrug resistance transporter EmrE using MAS solid-state NMR. Solid-state NMR can provide complementary structural information as the method allows studying membrane proteins in their native environment as no detergent is required for reconstitution. We compare the spectra obtained from wildtype EmrE to those obtained from the mutant EmrE-E14C. To resolve the critical amino acid E14, glutamic/aspartic acid selective experiments are carried out. These experiments allow to assign the chemical shift of the carboxylic carbon of E14. In addition, spectra are analyzed which are obtained in the presence and absence of the ligand TPP+.


Assuntos
Antiporters/química , Proteínas de Escherichia coli/química , Espectroscopia de Ressonância Magnética/métodos , Antiporters/ultraestrutura , Isótopos de Carbono , Proteínas de Escherichia coli/ultraestrutura , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Eletrônica , Mutação , Isótopos de Nitrogênio , Oniocompostos/metabolismo , Compostos Organofosforados/metabolismo , Ligação Proteica
12.
J Biol Chem ; 280(9): 7487-92, 2005 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-15623511

RESUMO

EmrE is a small multidrug transporter in Escherichia coli that extrudes various positively charged drugs across the plasma membrane in exchange with protons, thereby rendering cells resistant to these compounds. Biochemical experiments indicate that the basic functional unit of EmrE is a dimer where the common binding site for protons and substrate is formed by the interaction of an essential charged residue (Glu-14) from both EmrE monomers. Carbodiimide modification of EmrE has been studied using functional assays, and the evidence suggests that Glu-14 is the target of the reaction. Here we exploited electrospray ionization mass spectrometry to directly monitor the reaction with each monomer rather than following inactivation of the functional unit. A cyanogen bromide peptide containing Glu-14 allows the extent of modification by the carboxyl-specific modification reagent diisopropylcarbodiimide (DiPC) to be monitored and reveals that peptide 2NPYIYLGGAILAEVIGTTLM(21) is approximately 80% modified in a time-dependent fashion, indicating that each Glu-14 residue in the oligomer is accessible to DiPC. Furthermore, preincubation with tetraphenylphosphonium reduces the reaction of Glu-14 with DiPC by up to 80%. Taken together with other biochemical data, the findings support a "time sharing" mechanism in which both Glu-14 residues in a dimer are involved in tetraphenylphosphonium and H(+) binding.


Assuntos
Antiporters/química , Espectrometria de Massas/métodos , Proteínas de Membrana/química , Antiporters/ultraestrutura , Sítios de Ligação , Transporte Biológico , Carbodi-Imidas/farmacologia , Brometo de Cianogênio/química , Dimerização , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Ácido Glutâmico/química , Concentração de Íons de Hidrogênio , Proteínas de Membrana/ultraestrutura , Modelos Biológicos , Modelos Químicos , Oniocompostos/química , Compostos Organofosforados/química , Peptídeos/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Prótons , Espectrometria de Massas por Ionização por Electrospray , Fatores de Tempo
13.
EMBO J ; 20(1-2): 77-81, 2001 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11226157

RESUMO

EmrE belongs to a family of eubacterial multidrug transporters that confer resistance to a wide variety of toxins by coupling the influx of protons to toxin extrusion. EmrE was purified and crystallized in two dimensions by reconstitution with dimyristoylphosphatidylcholine into lipid bilayers. Images of frozen hydrated crystals were collected by cryo-electron microscopy and a projection structure of EmrE was calculated to 7 A resolution. The projection map shows an asymmetric EmrE dimer with overall dimensions approximately 31 x 40 A, comprising an arc of highly tilted helices separating two helices nearly perpendicular to the membrane from another two helices, one tilted and the other nearly perpendicular. There is no obvious 2-fold symmetry axis perpendicular to the membrane within the dimer, suggesting that the monomers may have different structures in the functional unit.


Assuntos
Antiporters/química , Antiporters/ultraestrutura , Escherichia coli/fisiologia , Proteínas de Membrana/química , Proteínas de Membrana/ultraestrutura , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Microscopia Crioeletrônica , Cristalografia por Raios X/métodos , Dimerização , Dimiristoilfosfatidilcolina , Resistência a Múltiplos Medicamentos , Proteínas de Escherichia coli , Bicamadas Lipídicas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/ultraestrutura
14.
Mol Microbiol ; 38(3): 482-92, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11069672

RESUMO

TetA, a tetracycline cation/proton antiporter, was expressed in Escherichia coli with a C-terminal tag of six histidines, solubilized in dodecyl maltoside and purified in a single step using Ni2+ affinity chromatography. Two-dimensional crystals were obtained after reconstitution of purified protein with lipids. Electron microscopy of negatively stained crystals revealed a trigonal symmetry, from which we infer that this secondary transporter has a trimeric structure. An overall molecular envelope can be described by a triangle of side approximately 100 A enclosing a central stain-filled depression. These dimensions are consistent with those obtained from projection views of single, isolated TetA particles that also display a trimeric architecture, confirming that the threefold symmetry is not simply a consequence of crystal-packing interactions. These data represent the first direct view of the quarternary arrangement of any antibiotic efflux pump. They are fully consistent with biochemical data on TetA, which indicate that it functions as a multimer and that the monomer consists of two domains, one of which plays the major part in oligomerization interactions.


Assuntos
Antiporters/química , Antiporters/ultraestrutura , Proteínas de Bactérias/química , Proteínas de Bactérias/ultraestrutura , Escherichia coli/química , Antiporters/metabolismo , Proteínas de Bactérias/metabolismo , Cristalização , Escherichia coli/crescimento & desenvolvimento , Histidina/química , Processamento de Imagem Assistida por Computador , Microscopia Eletrônica , Estrutura Quaternária de Proteína
15.
J Biol Chem ; 272(1): 580-5, 1997 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-8995300

RESUMO

The transposon (Tn) 10-encoded metal-tetracycline/H+ antiporter (Tn10-TetA) is predicted to have a membrane topology involving 12 transmembrane domains on the basis of the hydropathy profile of its sequence and the results of limited proteolysis; however, the experimental results of limited proteolysis are not enough to confirm the topology because proteases cannot gain access from the periplasmic side (Eckert, B., and Beck, C. F. (1989) J. Biol. Chem. 264, 11663-11670). One or two cysteine residues were introduced into each predicted hydrophilic loop or the N-terminal segment of Tn10-TetA by site-directed mutagenesis, and then the topology of the protein was determined by examining whether labeling of the introduced Cys residue by membrane-permeant [14C]N-ethylmaleimide ([14C]NEM) was prevented by preincubation of intact cells with the membrane-impermeant maleimide, 4-acetamido-4'-maleimidylstilbene-2,2'-disulfonic acid (AMS). The binding of [14C]NEM to the S36C (loop 1-2), L97C (loop 3-4), S156C (loop 5-6), R238C (loop 7-8), S296C (loop 9-10), Y357C, and D365C (loop 10-11) mutants was completely blocked by pretreatment with AMS, indicating that these residues are located on the periplasmic surface. In contrast, [14C]NEM binding to the S4C (N-terminal segment), S65C (loop 2-3), D120C (loop 4-5), S199C and S201C (loop 6-7), T270C (loop 8-9), and S328C (loop 10-11) mutants was not affected by pretreatment with AMS, indicating that these residues are on the cytoplasmic surface. These results for the first time thoroughly confirm the 12-transmembrane topology of the metal-tetracycline/H+ antiporter.


Assuntos
Antiporters/ultraestrutura , Proteínas de Bactérias/ultraestrutura , Elementos de DNA Transponíveis , Proteínas de Membrana/ultraestrutura , Resistência a Tetraciclina , Sequência de Aminoácidos , Antiporters/química , Proteínas de Bactérias/química , Cisteína/química , Etilmaleimida/metabolismo , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Plasmídeos , Reagentes de Sulfidrila
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...