Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 1327, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36899003

RESUMO

During biosynthesis by multi-modular trans-AT polyketide synthases, polyketide structural space can be expanded by conversion of initially-formed electrophilic ß-ketones into ß-alkyl groups. These multi-step transformations are catalysed by 3-hydroxy-3-methylgluratryl synthase cassettes of enzymes. While mechanistic aspects of these reactions have been delineated, little information is available concerning how the cassettes select the specific polyketide intermediate(s) to target. Here we use integrative structural biology to identify the basis for substrate choice in module 5 of the virginiamycin M trans-AT polyketide synthase. Additionally, we show in vitro that module 7, at minimum, is a potential additional site for ß-methylation. Indeed, analysis by HPLC-MS coupled with isotopic labelling and pathway inactivation identifies a metabolite bearing a second ß-methyl at the expected position. Collectively, our results demonstrate that several control mechanisms acting in concert underpin ß-branching programming. Furthermore, variations in this control - whether natural or by design - open up avenues for diversifying polyketide structures towards high-value derivatives.


Assuntos
Streptomyces , Metilação , Virginiamicina/biossíntese , Virginiamicina/química , Streptomyces/metabolismo , Ligação Proteica , Modelos Moleculares , Estrutura Terciária de Proteína , Especificidade por Substrato
2.
J Struct Biol ; 212(1): 107581, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32717326

RESUMO

Modular polyketide synthases (PKSs) are molecular-scale assembly lines comprising multiple gigantic polypeptide subunits. Faithful ordering of the subunits is mediated by extreme C- and N-terminal regions called docking domains (DDs). Decrypting how specificity is achieved by these elements is important both for understanding PKS function and modifying it to generate useful polyketide analogues for biological evaluation. Here we report the biophysical and structural characterisation of all six PKS/PKS interfaces in the unusual, chimaeric cis-AT/trans-AT PKS pathway responsible for biosynthesis of the antibiotic enacyloxin IIa in Burkholderia ambifaria. Taken together with previous work, our data reveal that specificity is achieved in the enacyloxin PKS by deploying at least three functionally orthogonal classes of DDs. We also demonstrate for the first time that cis-AT PKS subunits incorporate DDs with intrinsically disordered character, reinforcing the utility of such regions for achieving both medium affinity and high specificity at PKS intersubunit junctions. Overall, this work substantially increases the number of orthogonal DDs available for creating novel, highly-specific interfaces within cis- and trans-AT PKSs and their hybrids. It also reveals unexpected sequence/structure relationships in PKS DDs, identifying major current limitations to both accurately predicting and categorising these useful protein-protein interaction motifs.


Assuntos
Policetídeo Sintases/metabolismo , Policetídeos/metabolismo , Subunidades Proteicas/metabolismo , Burkholderia/metabolismo , Peptídeos/metabolismo , Polienos/metabolismo , Mapas de Interação de Proteínas/fisiologia
3.
Nat Commun ; 11(1): 683, 2020 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-31996686

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

4.
Nat Commun ; 10(1): 553, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30696828

RESUMO

In the original version of this Article, the final concentration of riboflavin in the supplemented LB medium for recombinant LkcE expression was incorrectly stated as 1 g L-1 (this was the concentration of the stock solution) and should have read 10-50 mg L-1. This error has been corrected in both the PDF and HTML versions of the Article.

5.
Nat Commun ; 9(1): 3998, 2018 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-30266997

RESUMO

Acquisition of new catalytic activity is a relatively rare evolutionary event. A striking example appears in the pathway to the antibiotic lankacidin, as a monoamine oxidase (MAO) family member, LkcE, catalyzes both an unusual amide oxidation, and a subsequent intramolecular Mannich reaction to form the polyketide macrocycle. We report evidence here for the molecular basis for this dual activity. The reaction sequence involves several essential active site residues and a conformational change likely comprising an interdomain hinge movement. These features, which have not previously been described in the MAO family, both depend on a unique dimerization mode relative to all structurally characterized members. Taken together, these data add weight to the idea that designing new multifunctional enzymes may require changes in both architecture and catalytic machinery. Encouragingly, however, our data also show LkcE to bind alternative substrates, supporting its potential utility as a general cyclization catalyst in synthetic biology.


Assuntos
Proteínas de Bactérias/metabolismo , Macrolídeos/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/metabolismo , Streptomyces/metabolismo , Amidas/química , Amidas/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Vias Biossintéticas/genética , Macrolídeos/síntese química , Macrolídeos/química , Modelos Químicos , Modelos Moleculares , Estrutura Molecular , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/química , Oxirredutases atuantes sobre Doadores de Grupo CH-NH/genética , Conformação Proteica , Multimerização Proteica , Homologia de Sequência de Aminoácidos , Streptomyces/enzimologia , Streptomyces/genética , Especificidade por Substrato
6.
J Am Chem Soc ; 138(12): 4155-67, 2016 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-26982529

RESUMO

Modular polyketide synthases (PKSs) direct the biosynthesis of clinically valuable secondary metabolites in bacteria. The fidelity of chain growth depends on specific recognition between successive subunits in each assembly line: interactions mediated by C- and N-terminal "docking domains" (DDs). We have identified a new family of DDs in trans-acyl transferase PKSs, exemplified by a matched pair from the virginiamycin (Vir) system. In the absence of C-terminal partner (VirA (C)DD) or a downstream catalytic domain, the N-terminal DD (VirFG (N)DD) exhibits multiple characteristics of an intrinsically disordered protein. Fusion of the two docking domains results in a stable fold for VirFG (N)DD and an overall protein-protein complex of unique topology whose structure we support by site-directed mutagenesis. Furthermore, using small-angle X-ray scattering (SAXS), the positions of the flanking acyl carrier protein and ketosynthase domains have been identified, allowing modeling of the complete intersubunit interface.


Assuntos
Aciltransferases/metabolismo , Policetídeo Sintases/metabolismo , Virginiamicina/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Virginiamicina/metabolismo
7.
J Biol Chem ; 290(6): 3836-49, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25525268

RESUMO

The majority of known bacteriophages have long tails that serve for bacterial target recognition and viral DNA delivery into the host. These structures form a tube from the viral capsid to the bacterial cell. The tube is formed primarily by a helical array of tail tube protein (TTP) subunits. In phages with a contractile tail, the TTP tube is surrounded by a sheath structure. Here, we report the first evidence that a phage TTP, gp17.1 of siphophage SPP1, self-assembles into long tubes in the absence of other viral proteins. gp17.1 does not exhibit a stable globular structure when monomeric in solution, even if it was confidently predicted to adopt the ß-sandwich fold of phage λ TTP. However, Fourier transform infrared and nuclear magnetic resonance spectroscopy analyses showed that its ß-sheet content increases significantly during tube assembly, suggesting that gp17.1 acquires a stable ß-sandwich fold only after self-assembly. EM analyses revealed that the tube is formed by hexameric rings stacked helicoidally with the same organization and helical parameters found for the tail of SPP1 virions. These parameters were used to build a pseudo-atomic model of the TTP tube. The large loop spanning residues 40-56 is located on the inner surface of the tube, at the interface between adjacent monomers and hexamers. In line with our structural predictions, deletion of this loop hinders gp17.1 tube assembly in vitro and interferes with SPP1 tail assembly during phage particle morphogenesis in bacteria.


Assuntos
Dobramento de Proteína , Proteínas Virais/química , Sequência de Aminoácidos , Bacteriófagos/química , Dados de Sequência Molecular , Estrutura Terciária de Proteína
8.
PLoS One ; 8(7): e68915, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894374

RESUMO

Given the fundamental roles of histone deacetylases (HDACs) in the regulation of DNA repair, replication, transcription and chromatin structure, it is fitting that therapies targeting HDAC activities are now being explored as anti-cancer agents. In fact, two histone deacetylase inhibitors (HDIs), SAHA and Depsipeptide, are FDA approved for single-agent treatment of refractory cutaneous T cell lymphoma (CTCL). An important target of these HDIs, histone deacetylase 3 (HDAC3), regulates processes such as DNA repair, metabolism, and tumorigenesis through the regulation of chromatin structure and gene expression. Here we show that HDAC3 inhibition using a first in class selective inhibitor, RGFP966, resulted in decreased cell growth in CTCL cell lines due to increased apoptosis that was associated with DNA damage and impaired S phase progression. Through isolation of proteins on nascent DNA (iPOND), we found that HDAC3 was associated with chromatin and is present at and around DNA replication forks. DNA fiber labeling analysis showed that inhibition of HDAC3 resulted in a significant reduction in DNA replication fork velocity within the first hour of drug treatment. These results suggest that selective inhibition of HDAC3 could be useful in treatment of CTCL by disrupting DNA replication of the rapidly cycling tumor cells, ultimately leading to cell death.


Assuntos
Antineoplásicos/farmacologia , Replicação do DNA/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Linfoma Cutâneo de Células T/genética , Linfoma Cutâneo de Células T/patologia , Estresse Fisiológico/genética , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cromatina/efeitos dos fármacos , Cromatina/genética , Dano ao DNA , Sinergismo Farmacológico , Humanos , Fase S/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos
9.
Methods Mol Biol ; 963: 173-86, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23296611

RESUMO

Calmodulin (CaM) is a ubiquitous EF-hand calcium sensor protein that transduces calcium signals in a wide range of signaling pathways. Structural analysis of complexes with peptides has provided valuable insights into the remarkable variety in the way in which CaM interacts with and activates its targets. Among these various targets, CaM has been shown to be an essential component of a calcium-sensing regulatory apparatus for a number of voltage-gated ion channels. NMR spectroscopy has proven to be a powerful tool for the structural characterization of CaM-peptide complexes, in particular for the study of IQ motifs, which bind CaM at the basal level of calcium in cells and thereby serve to localize CaM to its sites of action. We describe here methods for the robust expression and purification of CaM isotopically enriched for NMR analysis, as well as for the complex of CaM with a peptide derived from the IQ motif sequence of the human cardiac sodium channel Na(V)1.5. We also describe methods for NMR analysis of titrations of CaM with IQ motif peptides to determine the stoichiometry of the complex and to identify the residues at the binding interface.


Assuntos
Ressonância Magnética Nuclear Biomolecular/métodos , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Motivos de Aminoácidos , Apoproteínas/química , Apoproteínas/metabolismo , Cálcio/metabolismo , Calmodulina/química , Humanos , Espaço Intracelular/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/química , Ligação Proteica , Transdução de Sinais
11.
J Mol Biol ; 406(1): 106-19, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21167176

RESUMO

The function of the human voltage-gated sodium channel Na(V)1.5 is regulated in part by intracellular calcium signals. The ubiquitous calcium sensor protein calmodulin (CaM) is an important part of the complex calcium-sensing apparatus in Na(V)1.5. CaM interacts with an IQ (isoleucine-glutamine) motif in the large intracellular C-terminal domain of the channel. Using co-expression and co-purification, we have been able to isolate a CaM-IQ motif complex and to determine its high-resolution structure in absence of calcium using multi-dimensional solution NMR. Under these conditions, the Na(V)1.5 IQ motif interacts with the C-terminal domain (C-lobe) of CaM, with the N-terminal domain remaining free in solution. The structure reveals that the C-lobe adopts a semi-open conformation with the IQ motif bound in a narrow hydrophobic groove. Sequence similarities between voltage-gated sodium channels and voltage-gated calcium channels suggest that the structure of the CaM-Na(V)1.5 IQ motif complex can serve as a general model for the interaction between CaM and ion channel IQ motifs under low-calcium conditions. The structure also provides insight into the biochemical basis for disease-associated mutations that map to the IQ motif in Na(V)1.5.


Assuntos
Calmodulina/química , Proteínas Musculares/química , Domínios e Motivos de Interação entre Proteínas , Canais de Sódio/química , Sequência de Aminoácidos , Cálcio/química , Canais de Cálcio/química , Humanos , Dados de Sequência Molecular , Proteínas Musculares/genética , Mutação , Canal de Sódio Disparado por Voltagem NAV1.5 , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Estrutura Secundária de Proteína , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Canais de Sódio/genética
12.
J Biol Chem ; 285(30): 22942-9, 2010 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-20489205

RESUMO

The eukaryotic DNA replication protein Mcm10 associates with chromatin in early S-phase and is required for assembly and function of the replication fork protein machinery. Xenopus laevis (X) Mcm10 binds DNA via a highly conserved internal domain (ID) and a C-terminal domain (CTD) that is unique to higher eukaryotes. Although the structural basis of the interactions of the ID with DNA and polymerase alpha is known, little information is available for the CTD. We have identified the minimal DNA binding region of the XMcm10-CTD and determined its three-dimensional structure by solution NMR. The CTD contains a globular domain composed of two zinc binding motifs. NMR chemical shift perturbation and mutational analysis show that ssDNA binds only to the N-terminal (CCCH-type) zinc motif, whose structure is unique to Mcm10. The second (CCCC-type) zinc motif is not involved in DNA binding. However, it is structurally similar to the CCCC zinc ribbon in the N-terminal oligomerization domain of eukaryotic and archaeal MCM helicases. NMR analysis of a construct spanning both the ID and CTD reveals that the two DNA binding domains are structurally independent in solution, supporting a modular architecture for vertebrate Mcm10. Our results provide insight in the action of Mcm10 in the replisome and support a model in which it serves as a central scaffold through coupling of interactions with partner proteins and the DNA.


Assuntos
Sequência Conservada , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , DNA/metabolismo , Ressonância Magnética Nuclear Biomolecular , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Humanos , Camundongos , Proteínas de Manutenção de Minicromossomo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Xenopus laevis , Zinco
13.
J Biol Chem ; 284(33): 21934-21940, 2009 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-19553670

RESUMO

The action of Clostridium difficile toxins A and B depends on inactivation of host small G-proteins by glucosylation. Cellular inositol hexakisphosphate (InsP6) induces an autocatalytic cleavage of the toxins, releasing an N-terminal glucosyltransferase domain into the host cell cytosol. We have defined the cysteine protease domain (CPD) responsible for autoprocessing within toxin A (TcdA) and report the 1.6 A x-ray crystal structure of the domain bound to InsP6. InsP6 is bound in a highly basic pocket that is separated from an unusual active site by a beta-flap structure. Functional studies confirm an intramolecular mechanism of cleavage and highlight specific residues required for InsP6-induced TcdA processing. Analysis of the structural and functional data in the context of sequences from similar and diverse origins highlights a C-terminal extension and a pi-cation interaction within the beta-flap that appear to be unique among the large clostridial cytotoxins.


Assuntos
Toxinas Bacterianas/química , Clostridioides difficile/metabolismo , Enterotoxinas/química , Ácido Fítico/farmacologia , Domínio Catalítico , Cátions , Cristalografia por Raios X/métodos , Espectroscopia de Ressonância Magnética , Modelos Biológicos , Modelos Moleculares , Conformação Molecular , Mutação Puntual , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Espectrofotometria/métodos , Fatores de Tempo
14.
J Biol Chem ; 284(13): 8846-54, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19171938

RESUMO

Sodium channels are fundamental signaling molecules in excitable cells, and are molecular targets for local anesthetic agents and intracellular free Ca(2+) ([Ca(2+)](i)). Two regions of Na(V)1.5 have been identified previously as [Ca(2+)](i)-sensitive modulators of channel inactivation. These include a C-terminal IQ motif that binds calmodulin (CaM) in different modes depending on Ca(2+) levels, and an immediately adjacent C-terminal EF-hand domain that directly binds Ca(2+). Here we show that a mutation of the IQ domain (A1924T; Brugada Syndrome) that reduces CaM binding stabilizes Na(V)1.5 inactivation, similarly and more extensively than even reducing [Ca(2+)](i). Because the DIII-DIV linker is an essential structure in Na(V)1.5 inactivation, we evaluated this domain for a potential CaM binding interaction. We identified a novel CaM binding site within the linker, validated its interaction with CaM by NMR spectroscopy, and revealed its micromolar affinity by isothermal titration calorimetry. Mutation of three consecutive hydrophobic residues (Phe(1520)-Ile(1521)-Phe(1522)) to alanines in this CaM-binding domain recapitulated the electrophysiology phenotype observed with mutation of the C-terminal IQ domain: Na(V)1.5 inactivation was stabilized; moreover, mutations of either CaM-binding domain abolish the well described stabilization of inactivation by lidocaine. The direct physical interaction of CaM with the C-terminal IQ domain and the DIII-DIV linker, combined with the similarity in phenotypes when CaM-binding sites in either domain are mutated, suggests these cytoplasmic structures could be functionally coupled through the action of CaM. These findings have bearing upon Na(+) channel function in genetically altered channels and under pathophysiologic conditions where [Ca(2+)](i) impacts cardiac conduction.


Assuntos
Cálcio/metabolismo , Calmodulina/metabolismo , Proteínas Musculares/metabolismo , Canais de Sódio/metabolismo , Motivos de Aminoácidos/genética , Substituição de Aminoácidos , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Cálcio/química , Calmodulina/química , Calmodulina/genética , Linhagem Celular , Citoplasma/química , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Proteínas Musculares/química , Proteínas Musculares/genética , Mutação de Sentido Incorreto , Canal de Sódio Disparado por Voltagem NAV1.5 , Ressonância Magnética Nuclear Biomolecular , Estabilidade Proteica , Estrutura Quaternária de Proteína/genética , Estrutura Terciária de Proteína/genética , Canais de Sódio/química , Canais de Sódio/genética
15.
J Biol Chem ; 284(10): 6436-45, 2009 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-19074138

RESUMO

The voltage-gated sodium channel NaV1.5 is responsible for the initial upstroke of the action potential in cardiac tissue. Levels of intracellular calcium modulate inactivation gating of NaV1.5, in part through a C-terminal EF-hand calcium binding domain. The significance of this structure is underscored by the fact that mutations within this domain are associated with specific cardiac arrhythmia syndromes. In an effort to elucidate the molecular basis for calcium regulation of channel function, we have determined the solution structure of the C-terminal EF-hand domain using multidimensional heteronuclear NMR. The structure confirms the existence of the four-helix bundle common to EF-hand domain proteins. However, the location of this domain is shifted with respect to that predicted on the basis of a consensus 12-residue EF-hand calcium binding loop in the sequence. This finding is consistent with the weak calcium affinity reported for the isolated EF-hand domain; high affinity binding is observed only in a construct with an additional 60 residues C-terminal to the EF-hand domain, including the IQ motif that is central to the calcium regulatory apparatus. The binding of an IQ motif peptide to the EF-hand domain was characterized by isothermal titration calorimetry and nuclear magnetic resonance spectroscopy. The peptide binds between helices I and IV in the EF-hand domain, similar to the binding of target peptides to other EF-hand calcium-binding proteins. These results suggest a molecular basis for the coupling of the intrinsic (EF-hand domain) and extrinsic (calmodulin) components of the calcium-sensing apparatus of NaV1.5.


Assuntos
Proteínas de Ligação ao Cálcio/química , Cálcio/química , Proteínas Musculares/química , Miocárdio/química , Canais de Sódio/química , Motivos de Aminoácidos/fisiologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Calmodulina/química , Calmodulina/genética , Calmodulina/metabolismo , Humanos , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Miocárdio/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Peptídeos/genética , Peptídeos/metabolismo , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Canais de Sódio/genética , Canais de Sódio/metabolismo , Síndrome
16.
Protein Sci ; 15(3): 628-34, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16452619

RESUMO

Psalmopeotoxin I (PcFK1) is a 33-amino-acid residue peptide isolated from the venom of the tarantula Psalmopoeus cambridgei. It has been recently shown to possess strong antiplasmodial activity against the intra-erythrocyte stage of Plasmodium falciparum in vitro. Although the molecular target for PcFK1 is not yet determined, this peptide does not lyse erythrocytes, is not cytotoxic to nucleated mammalian cells, and does not inhibit neuromuscular function. We investigated the structural properties of PcFK1 to help understand the unique mechanism of action of this peptide and to enhance its utility as a lead compound for rational development of new antimalarial drugs. In this paper, we have determined the three-dimensional solution structure by (1)H two-dimensional NMR means of recombinant PcFK1, which is shown to belong to the ICK structural superfamily with structural determinants common to several neurotoxins acting as ion channels effectors.


Assuntos
Antimaláricos/química , Modelos Moleculares , Venenos de Aranha/química , Animais , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química , Plasmodium falciparum/efeitos dos fármacos , Soluções
17.
Calcium Bind Proteins ; 1(4): 203-212, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-28757812

RESUMO

Calcium plays an important role in regulating hundreds of biological processes due to its primary role as one of the most ubiquitous second messengers. As a result, the levels of calcium are tightly regulated as are the peak and trough calcium concentrations during a calcium signal. Calcium levels are controlled via a variety of feedback mechanisms and exchangers/transporters. Here the role of calcium in the feedback regulation of ion channel function is reviewed, with an emphasis on the molecular mechanisms governing calcium-dependent function. In particular, the role of calcium in the regulation of voltage-gated sodium, calcium, and potassium channels are reviewed as well as its effects on the ryanodine receptor.

18.
Protein Sci ; 14(8): 2003-10, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15987885

RESUMO

Acid-sensing ion channels (ASIC) are proton-gated sodium channels that have been implicated in pain transduction associated with acidosis in inflamed or ischemic tissues. APETx2, a peptide toxin effector of ASIC3, has been purified from an extract of the sea anemone Anthopleura elegantissima. APETx2 is a 42-amino-acid peptide cross-linked by three disulfide bridges. Its three-dimensional structure, as determined by conventional two-dimensional 1H-NMR, consists of a compact disulfide-bonded core composed of a four-stranded beta-sheet. It belongs to the disulfide-rich all-beta structural family encompassing peptide toxins commonly found in animal venoms. The structural characteristics of APETx2 are compared with that of PcTx1, another effector of ASIC channels but specific to the ASIC1a subtype and to APETx1, a toxin structurally related to APETx2, which targets the HERG potassium channel. Structural comparisons, coupled with the analysis of the electrostatic characteristics of these various ion channel effectors, led us to suggest a putative channel interaction surface for APETx2, encompassing its N terminus together with the type I-beta turn connecting beta-strands III and IV. This basic surface (R31 and R17) is also rich in aromatic residues (Y16, F15, Y32, and F33). An additional region made of the type II'-beta turn connecting beta-strands I and II could also play a role in the specificity observed for these different ion effectors.


Assuntos
Venenos de Cnidários/química , Proteínas de Membrana/química , Modelos Moleculares , Proteínas do Tecido Nervoso/química , Bloqueadores dos Canais de Sódio/química , Canais de Sódio/química , Canais Iônicos Sensíveis a Ácido , Sequência de Aminoácidos , Dados de Sequência Molecular , Ressonância Magnética Nuclear Biomolecular , Estrutura Secundária de Proteína , Soluções
19.
Proteins ; 60(3): 401-11, 2005 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15971207

RESUMO

Scorpion toxins interact with their target ion channels through multiple molecular contacts. Because a "gain of function" approach has never been described to evaluate the importance of the molecular contacts in defining toxin affinity, we experimentally examined whether increasing the molecular contacts between a toxin and an ion channel directly impacts toxin affinity. For this purpose, we focused on two scorpion peptides, the well-characterized maurotoxin with its variant Pi1-like disulfide bridging (MTX(Pi1)), used as a molecular template, and butantoxin (BuTX), used as an N-terminal domain provider. BuTX is found to be 60-fold less potent than MTX(Pi1) in blocking Kv1.2 (IC(50) values of 165 nM for BuTX versus 2.8 nM for MTX(Pi1)). Removal of its N-terminal domain (nine residues) further decreases BuTX affinity for Kv1.2 by 5.6-fold, which is in agreement with docking simulation data showing the importance of this domain in BuTX-Kv1.2 interaction. Transfer of the BuTX N-terminal domain to MTX(Pi1) results in a chimera with five disulfide bridges (BuTX-MTX(Pi1)) that exhibits 22-fold greater affinity for Kv1.2 than MTX(Pi1) itself, in spite of the lower affinity of BuTX as compared to MTX(Pi1). Docking experiments performed with the 3-D structure of BuTX-MTX(Pi1) in solution, as solved by (1)H-NMR, reveal that the N-terminal domain of BuTX participates in the increased affinity for Kv1.2 through additional molecular contacts. Altogether, the data indicate that acting on molecular contacts between a toxin and a channel is an efficient strategy to modulate toxin affinity.


Assuntos
Biologia Computacional/métodos , Canal de Potássio Kv1.2/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana/química , Proteômica/métodos , Venenos de Escorpião/química , Sequência de Aminoácidos , Animais , Dicroísmo Circular , Cisteína/química , Dissulfetos/química , Eletrofisiologia , Concentração Inibidora 50 , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Escorpiões , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ácido Trifluoracético/química
20.
Proteins ; 59(2): 380-6, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15726634

RESUMO

APETx1 is a 42-amino acid toxin purified from the venom of the sea anemone Anthopleura elegantissima. This cysteine-rich peptide possesses three disulfide bridges (C4-C37, C6-C30, and C20-C38). Its pharmacological target is the Ether-a-gogo potassium channel. We herein determine the solution structure of APETx1 by use of conventional two-dimensional 1H-NMR techniques followed by torsion angle dynamics and refinement protocols. The calculated structure of APETx1 belongs to the disulfide-rich all-beta structural family, in which a three-stranded anti-parallel beta-sheet is the only secondary structure. APETx1 is the first Ether-a-gogo effector discovered to fold in this way. We therefore compare the structure of APETx1 to those of the two other known effectors of the Ether-a-gogo potassium channel, CnErg1 and BeKm-1, and analyze the topological disposition of key functional residues proposed by analysis of the electrostatic anisotropy. The interacting surface is made of a patch of aromatic residues (Y5, Y32, and F33) together with two basic residues (K8 and K18) at the periphery of the surface. We pinpoint the absence of the central lysine present in the functional surface of the two other Ether-a-gogo effectors.


Assuntos
Venenos de Cnidários/química , Anêmonas-do-Mar , Animais , Venenos de Cnidários/metabolismo , Espectroscopia de Ressonância Magnética , Conformação Proteica , Dobramento de Proteína , Estrutura Secundária de Proteína , Soluções
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...