Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Chem Inf Model ; 64(7): 2612-2623, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38157481

RESUMO

Structure-based drug discovery is a process for both hit finding and optimization that relies on a validated three-dimensional model of a target biomolecule, used to rationalize the structure-function relationship for this particular target. An ultralarge virtual screening approach has emerged recently for rapid discovery of high-affinity hit compounds, but it requires substantial computational resources. This study shows that active learning with simple linear regression models can accelerate virtual screening, retrieving up to 90% of the top-1% of the docking hit list after docking just 10% of the ligands. The results demonstrate that it is unnecessary to use complex models, such as deep learning approaches, to predict the imprecise results of ligand docking with a low sampling depth. Furthermore, we explore active learning meta-parameters and find that constant batch size models with a simple ensembling method provide the best ligand retrieval rate. Finally, our approach is validated on the ultralarge size virtual screening data set, retrieving 70% of the top-0.05% of ligands after screening only 2% of the library. Altogether, this work provides a computationally accessible approach for accelerated virtual screening that can serve as a blueprint for the future design of low-compute agents for exploration of the chemical space via large-scale accelerated docking. With recent breakthroughs in protein structure prediction, this method can significantly increase accessibility for the academic community and aid in the rapid discovery of high-affinity hit compounds for various targets.


Assuntos
Descoberta de Drogas , Ligação Proteica , Simulação de Acoplamento Molecular , Ligantes
2.
Int J Mol Sci ; 24(3)2023 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-36768442

RESUMO

Ribosome biogenesis is a complex and highly accurate conservative process of ribosomal subunit maturation followed by association. Subunit maturation comprises sequential stages of ribosomal RNA and proteins' folding, modification and binding, with the involvement of numerous RNAses, helicases, GTPases, chaperones, RNA, protein-modifying enzymes, and assembly factors. One such assembly factor involved in bacterial 30S subunit maturation is ribosomal binding factor A (RbfA). In this study, we present the crystal (determined at 2.2 Å resolution) and NMR structures of RbfA as well as the 2.9 Å resolution cryo-EM reconstruction of the 30S-RbfA complex from Staphylococcus aureus (S. aureus). Additionally, we show that the manner of RbfA action on the small ribosomal subunit during its maturation is shared between bacteria and mitochondria. The obtained results clarify the function of RbfA in the 30S maturation process and its role in ribosome functioning in general. Furthermore, given that S. aureus is a serious human pathogen, this study provides an additional prospect to develop antimicrobials targeting bacterial pathogens.


Assuntos
Proteínas de Escherichia coli , Staphylococcus aureus Resistente à Meticilina , Humanos , Proteínas Ribossômicas/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus Resistente à Meticilina/genética , Proteínas de Escherichia coli/metabolismo , Bactérias/metabolismo , Mitocôndrias/metabolismo , RNA Ribossômico 16S/metabolismo
3.
J Am Chem Soc ; 143(3): 1513-1520, 2021 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-33449695

RESUMO

Photopharmacology addresses the challenge of drug selectivity and side effects through creation of photoresponsive molecules activated with light with high spatiotemporal precision. This is achieved through incorporation of molecular photoswitches and photocages into the pharmacophore. However, the structural basis for the light-induced modulation of inhibitory potency in general is still missing, which poses a major design challenge for this emerging field of research. Here we solved crystal structures of the glutamate transporter homologue GltTk in complex with photoresponsive transport inhibitors-azobenzene derivative of TBOA (both in trans and cis configuration) and with the photocaged compound ONB-hydroxyaspartate. The essential role of glutamate transporters in the functioning of the central nervous system renders them potential therapeutic targets in the treatment of neurodegenerative diseases. The obtained structures provide a clear structural insight into the origins of photocontrol in photopharmacology and lay the foundation for application of photocontrolled ligands to study the transporter dynamics by using time-resolved X-ray crystallography.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Compostos Azo/metabolismo , Sistema X-AG de Transporte de Aminoácidos/química , Ácido Aspártico/efeitos da radiação , Compostos Azo/química , Compostos Azo/efeitos da radiação , Cristalografia por Raios X , Ligação Proteica , Estereoisomerismo , Thermococcus/química , Raios Ultravioleta
4.
Bioinformatics ; 36(10): 3064-3071, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32022861

RESUMO

MOTIVATION: Halides are negatively charged ions of halogens, forming fluorides (F-), chlorides (Cl-), bromides (Br-) and iodides (I-). These anions are quite reactive and interact both specifically and non-specifically with proteins. Despite their ubiquitous presence and important roles in protein function, little is known about the preferences of halides binding to proteins. To address this problem, we performed the analysis of halide-protein interactions, based on the entries in the Protein Data Bank. RESULTS: We have compiled a pipeline for the quick analysis of halide-binding sites in proteins using the available software. Our analysis revealed that all of halides are strongly attracted by the guanidinium moiety of arginine side chains, however, there are also certain preferences among halides for other partners. Furthermore, there is a certain preference for coordination numbers in the binding sites, with a correlation between coordination numbers and amino acid composition. This pipeline can be used as a tool for the analysis of specific halide-protein interactions and assist phasing experiments relying on halides as anomalous scatters. AVAILABILITY AND IMPLEMENTATION: All data described in this article can be reproduced via complied pipeline published at https://github.com/rostkick/Halide_sites/blob/master/README.md. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Brometos , Censos , Sítios de Ligação , Iodetos , Proteínas
5.
J Chem Inf Model ; 61(5): 2407-2417, 2021 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-33886304

RESUMO

The CorA family of proteins plays a housekeeping role in the homeostasis of divalent metal ions in many bacteria and archaea as well as in mitochondria of eukaryotes, rendering it an important target to study the mechanisms of divalent transport and regulation across different life domains. Despite numerous studies, the mechanistic details of the channel gating and the transport of the metal ions are still not entirely understood. Here, we use all-atom and coarse-grained molecular dynamics simulations combined with in vitro experiments to investigate the influence of divalent cations on the function of CorA. Simulations reveal pronounced asymmetric movements of monomers that enable the rotation of the α7 helix and the cytoplasmic subdomain with the subsequent formation of new interactions and the opening of the channel. These computational results are functionally validated using site-directed mutagenesis of the intracellular cytoplasmic domain residues and biochemical assays. The obtained results infer a complex network of interactions altering the structure of CorA to allow gating. Furthermore, we attempt to reconcile the existing gating hypotheses for CorA to conclude the mechanism of transport of divalent cations via these proteins.


Assuntos
Proteínas de Transporte de Cátions , Simulação de Dinâmica Molecular , Proteínas de Bactérias/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Magnésio/metabolismo , Mutagênese Sítio-Dirigida
6.
Molecules ; 25(8)2020 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-32331317

RESUMO

NadR is a bifunctional enzyme that converts nicotinamide riboside (NR) into nicotinamide mononucleotide (NMN), which is then converted into nicotinamide adenine dinucleotide (NAD). Although a crystal structure of the enzyme from the Gram-negative bacterium Haemophilus influenzae is known, structural understanding of its catalytic mechanism remains unclear. Here, we purified the NadR enzyme from Lactococcus lactis and established an assay to determine the combined activity of this bifunctional enzyme. The conversion of NR into NAD showed hyperbolic dependence on the NR concentration, but sigmoidal dependence on the ATP concentration. The apparent cooperativity for ATP may be explained because both reactions catalyzed by the bifunctional enzyme (phosphorylation of NR and adenylation of NMN) require ATP. The conversion of NMN into NAD followed simple Michaelis-Menten kinetics for NMN, but again with the sigmoidal dependence on the ATP concentration. In this case, the apparent cooperativity is unexpected since only a single ATP is used in the NMN adenylyltransferase catalyzed reaction. To determine the possible structural determinants of such cooperativity, we solved the crystal structure of NadR from L. lactis (NadRLl). Co-crystallization with NAD, NR, NMN, ATP, and AMP-PNP revealed a 'sink' for adenine nucleotides in a location between two domains. This sink could be a regulatory site, or it may facilitate the channeling of substrates between the two domains.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Lactococcus lactis/enzimologia , Proteínas Repressoras/química , Proteínas Repressoras/metabolismo , Sequência de Aminoácidos , Ativação Enzimática , Cinética , Modelos Moleculares , Conformação Molecular , NAD/metabolismo , Mononucleotídeo de Nicotinamida/química , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Relação Estrutura-Atividade , Especificidade por Substrato
7.
Trends Biochem Sci ; 40(4): 183-8, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25757400

RESUMO

The evolutionary relation between sugar and vitamin transporters from the SWEET and Pnu families is unclear. They have similar 3D structures, but differ in the topology of their secondary structure elements, and lack significant sequence similarity. Here we analyze the structures and sequences of different members of the SWEET and Pnu transporter families and propose an evolutionary pathway by which they may have diverged from a common ancestor. A 3D domain swapping event can explain the topological differences between the families, as well as the puzzling observation that a highly conserved and essential sequence motif of the SWEET family (the PQ loop) is absent from the Pnu transporters.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Bactérias/genética , Evolução Biológica , Transporte Biológico , Proteínas de Membrana Transportadoras/genética , Família Multigênica/genética , Família Multigênica/fisiologia , Estrutura Secundária de Proteína
9.
Chembiochem ; 16(5): 819-26, 2015 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-25676607

RESUMO

Energy-coupling factor (ECF) transporters are membrane-protein complexes that mediate vitamin uptake in prokaryotes. They bind the substrate through the action of a specific integral membrane subunit (S-component) and power transport by hydrolysis of ATP in the three-subunit ECF module. Here, we have studied the binding of thiamine derivatives to ThiT, a thiamine-specific S-component. We designed and synthesized derivatives of thiamine that bind to ThiT with high affinity; this allowed us to evaluate the contribution of the functional groups to the binding affinity. We determined six crystal structures of ThiT in complex with our derivatives. The structure of the substrate-binding site in ThiT remains almost unchanged despite substantial differences in affinity. This work indicates that the structural organization of the binding site is robust and suggests that substrate release, which is required for transport, requires additional changes in conformation in ThiT that might be imposed by the ECF module.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Proteínas de Bactérias/metabolismo , Desenho de Fármacos , Bibliotecas de Moléculas Pequenas/metabolismo , Tiamina/metabolismo , Transportadores de Cassetes de Ligação de ATP/química , Proteínas de Bactérias/química , Transporte Biológico , Lactococcus lactis/química , Modelos Moleculares , Estrutura Molecular , Bibliotecas de Moléculas Pequenas/química , Tiamina/síntese química , Tiamina/química
10.
EMBO Rep ; 14(1): 49-56, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23196367

RESUMO

The 5-HT(3) receptor is a pentameric serotonin-gated ion channel, which mediates rapid excitatory neurotransmission and is the target of a therapeutically important class of anti-emetic drugs, such as granisetron. We report crystal structures of a binding protein engineered to recognize the agonist serotonin and the antagonist granisetron with affinities comparable to the 5-HT(3) receptor. In the serotonin-bound structure, we observe hydrophilic interactions with loop E-binding site residues, which might enable transitions to channel opening. In the granisetron-bound structure, we observe a critical cation-π interaction between the indazole moiety of the ligand and a cationic centre in loop D, which is uniquely present in the 5-HT(3) receptor. We use a series of chemically tuned granisetron analogues to demonstrate the energetic contribution of this electrostatic interaction to high-affinity ligand binding in the human 5-HT(3) receptor. Our study offers the first structural perspective on recognition of serotonin and antagonism by anti-emetics in the 5-HT(3) receptor.


Assuntos
Antieméticos/química , Granisetron/análogos & derivados , Subunidades Proteicas/química , Receptores 5-HT3 de Serotonina/química , Agonistas do Receptor de Serotonina/química , Serotonina/análogos & derivados , Sequência de Aminoácidos , Antieméticos/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Granisetron/metabolismo , Humanos , Interações Hidrofóbicas e Hidrofílicas , Cinética , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Engenharia de Proteínas , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas/metabolismo , Receptores 5-HT3 de Serotonina/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Serotonina/metabolismo , Agonistas do Receptor de Serotonina/metabolismo , Eletricidade Estática , Termodinâmica
11.
Proc Natl Acad Sci U S A ; 109(45): 18459-64, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23091000

RESUMO

Despite the importance of Mg(2+) for numerous cellular activities, the mechanisms underlying its import and homeostasis are poorly understood. The CorA family is ubiquitous and is primarily responsible for Mg(2+) transport. However, the key questions-such as, the ion selectivity, the transport pathway, and the gating mechanism-have remained unanswered for this protein family. We present a 3.2 Å resolution structure of the archaeal CorA from Methanocaldococcus jannaschii, which is a unique complete structure of a CorA protein and reveals the organization of the selectivity filter, which is composed of the signature motif of this family. The structure reveals that polar residues facing the channel coordinate a partially hydrated Mg(2+) during the transport. Based on these findings, we propose a unique gating mechanism involving a helical turn upon the binding of Mg(2+) to the regulatory intracellular binding sites, and thus converting a polar ion passage into a narrow hydrophobic pore. Because the amino acids involved in the uptake, transport, and gating are all conserved within the entire CorA family, we believe this mechanism is general for the whole family including the eukaryotic homologs.


Assuntos
Proteínas Arqueais/química , Proteínas Arqueais/metabolismo , Ativação do Canal Iônico , Magnésio/metabolismo , Methanococcales/metabolismo , Sítios de Ligação , Transporte Biológico , Transporte de Íons , Íons , Modelos Moleculares
12.
Biochem J ; 451(3): 365-74, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23425532

RESUMO

The CorA family of divalent cation transporters utilizes Mg2+ and Co2+ as primary substrates. The molecular mechanism of its function, including ion selectivity and gating, has not been fully characterized. Recently we reported a new structure of a CorA homologue from Methanocaldococcus jannaschii, which provided novel structural details that offered the conception of a unique gating mechanism involving conversion of an open hydrophilic gate into a closed hydrophobic one. In the present study we report functional evidence for this novel gating mechanism in the Thermotoga maritima CorA together with an improved crystal structure of this CorA to 2.7 Å (1 Å=0.1 nm) resolution. The latter reveals the organization of the selectivity filter to be similar to that of M. jannaschii CorA and also the previously unknown organization of the second signature motif of the CorA family. The proposed gating is achieved by a helical rotation upon the binding of a metal ion substrate to the regulatory binding sites. Additionally, our data suggest that the preference of this CorA for Co2+ over Mg2+ is controlled by the presence of threonine side chains in the channel. Finally, the roles of the intracellular metal-binding sites have been assigned to increased thermostability and regulation of the gating. These mechanisms most likely apply to the entire CorA family as they are regulated by the highly conserved amino acids.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/metabolismo , Cobalto/química , Magnésio/química , Thermotoga maritima/metabolismo , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Sítios de Ligação , Transporte Biológico , Proteínas de Transporte de Cátions/genética , Cátions Bivalentes , Cobalto/metabolismo , Cristalografia por Raios X , Escherichia coli/genética , Interações Hidrofóbicas e Hidrofílicas , Ativação do Canal Iônico , Cinética , Magnésio/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Thermotoga maritima/genética
13.
bioRxiv ; 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38328101

RESUMO

The electroosmotic-driven transport of unravelled proteins across nanopores is an important biological process that is now under investigation for the rapid analysis and sequencing of proteins. For this approach to work, however, it is crucial that the polymer is threaded in single file. Here we found that, contrary to the electrophoretic transport of charged polymers such as DNA, during polypeptide translocation blob-like structures typically form inside nanopores. Comparisons between different nanopore sizes, shapes and surface chemistries showed that under electroosmotic-dominated regimes single-file transport of polypeptides can be achieved using nanopores that simultaneously have an entry and an internal diameter that is smaller than the persistence length of the polymer, have a uniform non-sticky ( i . e . non-aromatic) nanopore inner surface, and using moderate translocation velocities.

14.
Cryst Growth Des ; 24(1): 325-330, 2024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38188264

RESUMO

Membrane proteins are indispensable for every living organism, yet their structural organization remains underexplored. Despite the recent advancements in single-particle cryogenic electron microscopy and cryogenic electron tomography, which have significantly increased the structural coverage of membrane proteins across various kingdoms, certain scientific methods, such as time-resolved crystallography, still mostly rely on crystallization techniques, such as lipidic cubic phase (LCP) or in meso crystallization. In this study, we present an open-access blueprint for a humidity control chamber designed for LCP/in meso crystallization experiments using a Gryphon crystallization robot. Using this chamber, we have obtained crystals of a transmembrane aspartate transporter GltTk from Thermococcus kodakarensis in a lipidic environment using in meso crystallization. The data collected from these crystals allowed us to perform an analysis of lipids bound to the transporter. With this publication of our open-access design of a humidity chamber, we aim to improve the accessibility of in meso protein crystallization for the scientific community.

15.
Structure ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38733996

RESUMO

BtuM is a bacterial cobalamin transporter that binds the transported substrate in the base-off state, with a cysteine residue providing the α-axial coordination of the central cobalt ion via a sulfur-cobalt bond. Binding leads to decyanation of cobalamin variants with a cyano group as the ß-axial ligand. Here, we report the crystal structures of untagged BtuM bound to two variants of cobalamin, hydroxycobalamin and cyanocobalamin, and unveil the native residue responsible for the ß-axial coordination, His28. This coordination had previously been obscured by non-native histidines of His-tagged BtuM. A model in which BtuM initially binds cobinamide reversibly with low affinity (KD = 4.0 µM), followed by the formation of a covalent bond (rate constant of 0.163 s-1), fits the kinetics data of substrate binding and decyanation of the cobalamin precursor cobinamide by BtuM. The covalent binding mode suggests a mechanism not used by any other transport protein.

16.
FEBS J ; 290(17): 4238-4255, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37213112

RESUMO

Formate dehydrogenases (Fdhs) mediate the oxidation of formate to carbon dioxide and concomitant reduction of nicotinamide adenine dinucleotide (NAD+ ). The low cost of the substrate formate and importance of the product NADH as a cellular source of reducing power make this reaction attractive for biotechnological applications. However, the majority of Fdhs are sensitive to inactivation by thiol-modifying reagents. In this study, we report a chemically resistant Fdh (FdhSNO ) from the soil bacterium Starkeya novella strictly specific for NAD+ . We present its recombinant overproduction, purification and biochemical characterization. The mechanistic basis of chemical resistance was found to be a valine in position 255 (rather than a cysteine as in other Fdhs) preventing the inactivation by thiol-modifying compounds. To further improve the usefulness of FdhSNO as for generating reducing power, we rationally engineered the protein to reduce the coenzyme nicotinamide adenine dinucleotide phosphate (NADP+ ) with better catalytic efficiency than NAD+ . The single mutation D221Q enabled the reduction of NADP+ with a catalytic efficiency kCAT /KM of 0.4 s-1 ·mm-1 at 200 mm formate, while a quadruple mutant (A198G/D221Q/H379K/S380V) resulted in a fivefold increase in catalytic efficiency for NADP+ compared with the single mutant. We determined the cofactor-bound structure of the quadruple mutant to gain mechanistic evidence behind the improved specificity for NADP+ . Our efforts to unravel the key residues for the chemical resistance and cofactor specificity of FdhSNO may lead to wider use of this enzymatic group in a more sustainable (bio)manufacture of value-added chemicals, as for instance the biosynthesis of chiral compounds.


Assuntos
Formiato Desidrogenases , NAD , NAD/metabolismo , Formiato Desidrogenases/genética , NADP/metabolismo , Formiatos/metabolismo , Compostos de Sulfidrila
17.
Nat Commun ; 14(1): 1799, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37002226

RESUMO

Episodic ataxias (EAs) are rare neurological conditions affecting the nervous system and typically leading to motor impairment. EA6 is linked to the mutation of a highly conserved proline into an arginine in the glutamate transporter EAAT1. In vitro studies showed that this mutation leads to a reduction in the substrates transport and an increase in the anion conductance. It was hypothesised that the structural basis of these opposed functional effects might be the straightening of transmembrane helix 5, which is kinked in the wild-type protein. In this study, we present the functional and structural implications of the mutation P208R in the archaeal homologue of glutamate transporters GltTk. We show that also in GltTk the P208R mutation leads to reduced aspartate transport activity and increased anion conductance, however a cryo-EM structure reveals that the kink is preserved. The arginine side chain of the mutant points towards the lipidic environment, where it may engage in interactions with the phospholipids, thereby potentially interfering with the transport cycle and contributing to stabilisation of an anion conducting state.


Assuntos
Sistema X-AG de Transporte de Aminoácidos , Proteínas Arqueais , Ataxia , Humanos , Sistema X-AG de Transporte de Aminoácidos/metabolismo , Arginina/genética , Ataxia/genética , Transportador 1 de Aminoácido Excitatório/genética , Mutação , Archaea/genética , Archaea/fisiologia , Proteínas Arqueais/genética , Proteínas Arqueais/fisiologia
18.
J Biol Chem ; 286(18): 15964-72, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21367867

RESUMO

Herbicides that target photosystem II (PSII) compete with the native electron acceptor plastoquinone for binding at the Q(B) site in the D1 subunit and thus block the electron transfer from Q(A) to Q(B). Here, we present the first crystal structure of PSII with a bound herbicide at a resolution of 3.2 Å. The crystallized PSII core complexes were isolated from the thermophilic cyanobacterium Thermosynechococcus elongatus. The used herbicide terbutryn is found to bind via at least two hydrogen bonds to the Q(B) site similar to photosynthetic reaction centers in anoxygenic purple bacteria. Herbicide binding to PSII is also discussed regarding the influence on the redox potential of Q(A), which is known to affect photoinhibition. We further identified a second and novel chloride position close to the water-oxidizing complex and in the vicinity of the chloride ion reported earlier (Guskov, A., Kern, J., Gabdulkhakov, A., Broser, M., Zouni, A., and Saenger, W. (2009) Nat. Struct. Mol. Biol. 16, 334-342). This discovery is discussed in the context of proton transfer to the lumen.


Assuntos
Cianobactérias/enzimologia , Herbicidas/química , Complexo de Proteína do Fotossistema II/química , Triazinas/química , Cristalografia por Raios X , Herbicidas/farmacologia , Complexo de Proteína do Fotossistema II/antagonistas & inibidores , Complexo de Proteína do Fotossistema II/metabolismo , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade , Triazinas/farmacologia
19.
J Biol Chem ; 286(6): 4420-8, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21115477

RESUMO

Covalent modification of α7 W55C nicotinic acetylcholine receptors (nAChR) with the cysteine-modifying reagent [2-(trimethylammonium)ethyl] methanethiosulfonate (MTSET(+)) produces receptors that are unresponsive to acetylcholine, whereas methyl methanethiolsulfonate (MMTS) produces enhanced acetylcholine-gated currents. Here, we investigate structural changes that underlie the opposite effects of MTSET(+) and MMTS using acetylcholine-binding protein (AChBP), a homolog of the extracellular domain of the nAChR. Crystal structures of Y53C AChBP show that MTSET(+)-modification stabilizes loop C in an extended conformation that resembles the antagonist-bound state, which parallels our observation that MTSET(+) produces unresponsive W55C nAChRs. The MMTS-modified mutant in complex with acetylcholine is characterized by a contracted C-loop, similar to other agonist-bound complexes. Surprisingly, we find two acetylcholine molecules bound in the ligand-binding site, which might explain the potentiating effect of MMTS modification in W55C nAChRs. Unexpectedly, we observed in the MMTS-Y53C structure that ten phosphate ions arranged in two rings at adjacent sites are bound in the vestibule of AChBP. We mutated homologous residues in the vestibule of α1 GlyR and observed a reduction in the single channel conductance, suggesting a role of this site in ion permeation. Taken together, our results demonstrate that targeted modification of a conserved aromatic residue in loop D is sufficient for a conformational switch of AChBP and that a defined region in the vestibule of the extracellular domain contributes to ion conduction in anion-selective Cys-loop receptors.


Assuntos
Acetilcolina/química , Aplysia/química , Proteínas de Transporte/química , Cisteína/química , Mutação de Sentido Incorreto , Acetilcolina/genética , Acetilcolina/metabolismo , Substituição de Aminoácidos , Animais , Aplysia/genética , Aplysia/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Cristalografia por Raios X , Cisteína/genética , Cisteína/metabolismo , Estabilidade Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
20.
Curr Top Membr ; 69: 393-414, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23046658

RESUMO

The metal ions Mg(2+) and Co(2+) are essential for life, although to different degree. They have similar chemical and physical properties, but their slight differences result in Mg(2+) to be the most abundant metal ion in living cells and the trace element Co(2+) being toxic at relatively low concentrations. Specialized transporters have evolved in living cells to supply and balance the Mg(2+) and Co(2+) need of the cells. The current knowledge of the molecular mechanisms of Mg(2+) and Co(2+) -specific transporters is very limited at this point. Recently, there has been remarkable advances to understand the CorA family, a family of transporters that are able to transport both ions. These new data have increased our insights in how Mg(2+) and Co(2+) are translocated across membranes. Presently, CorA is probably the best system to study the mechanisms of Mg(2+) and Co(2+) transport. This chapter discusses the mechanisms through which CorA selects, transports, and regulates the translocation of its substrate. In addition, we highlight the physical and chemical properties of the substrates, which are important parameters required for better understanding of the transporter action.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobalto/metabolismo , Magnésio/metabolismo , Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Transporte de Cátions/química , Cobalto/química , Transporte de Íons , Magnésio/química , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA