Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochem J ; 474(5): 851-864, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28049758

RESUMO

Cathepsin K (CatK) is the predominant mammalian bone-degrading protease and thus an ideal target for antiosteoporotic drug development. Rodent models of osteoporosis are preferred due to their close reflection of the human disease and their ease of handling, genetic manipulation and economic affordability. However, large differences in the potency of CatK inhibitors for the mouse/rat vs. the human protease orthologs have made it impossible to use rodent models. This is even more of a problem considering that the most advanced CatK inhibitors, including odanacatib (ODN) and balicatib, failed in human clinical trials due to side effects and rodent models are not available to investigate the mechanism of these failures. Here, we elucidated the structural elements of the potency differences between mouse and human CatK (hCatK) using ODN. We determined and compared the structures of inhibitor-free mouse CatK (mCatK), hCatK and ODN bound to hCatK. Two structural differences were identified and investigated by mutational analysis. Humanizing subsite 2 in mCatK led to a 5-fold improvement of ODN binding, whereas the replacement of Tyr61 in mCatK with Asp resulted in an hCatK with comparable ODN potency. Combining both sites further improved the inhibition of the mCatK variant. Similar results were obtained for balicatib. These findings will allow the generation of transgenic CatK mice that will facilitate the evaluation of CatK inhibitor adverse effects and to explore routes to avoid them.


Assuntos
Benzamidas/química , Compostos de Bifenilo/química , Conservadores da Densidade Óssea/química , Catepsina K/antagonistas & inibidores , Piperazinas/química , Inibidores de Proteases/química , Sequência de Aminoácidos , Animais , Benzamidas/metabolismo , Sítios de Ligação , Compostos de Bifenilo/metabolismo , Conservadores da Densidade Óssea/metabolismo , Catepsina K/química , Catepsina K/genética , Catepsina K/metabolismo , Clonagem Molecular , Cristalografia por Raios X , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Humanos , Cinética , Ligantes , Camundongos , Mutagênese Sítio-Dirigida , Piperazinas/metabolismo , Inibidores de Proteases/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia Estrutural de Proteína
2.
Nat Chem Biol ; 11(9): 691-6, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26214255

RESUMO

The complex plant flavonol glycoside montbretin A is a potent (Ki = 8 nM) and specific inhibitor of human pancreatic α-amylase with potential as a therapeutic for diabetes and obesity. Controlled degradation studies on montbretin A, coupled with inhibition analyses, identified an essential high-affinity core structure comprising the myricetin and caffeic acid moieties linked via a disaccharide. X-ray structural analyses of the montbretin A-human α-amylase complex confirmed the importance of this core structure and revealed a novel mode of glycosidase inhibition wherein internal π-stacking interactions between the myricetin and caffeic acid organize their ring hydroxyls for optimal hydrogen bonding to the α-amylase catalytic residues D197 and E233. This novel inhibitory motif can be reproduced in a greatly simplified analog, offering potential for new strategies for glycosidase inhibition and therapeutic development.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/química , Flavonóis/química , Glicosídeos/química , alfa-Amilases/química , Sítios de Ligação , Ácidos Cafeicos/química , Sequência de Carboidratos , Inibidores Enzimáticos/síntese química , Flavonas/química , Flavonoides/química , Expressão Gênica , Humanos , Ligação de Hidrogênio , Hidrólise , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Pichia/genética , Pichia/metabolismo , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Trissacarídeos/química , alfa-Amilases/antagonistas & inibidores , alfa-Amilases/genética
3.
Proc Natl Acad Sci U S A ; 111(49): 17474-9, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422423

RESUMO

Cathepsin K is the major collagenolytic protease in bone that facilitates physiological as well as pathological bone degradation. Despite its key role in bone remodeling and for being a highly sought-after drug target for the treatment of osteoporosis, the mechanism of collagen fiber degradation by cathepsin K remained elusive. Here, we report the structure of a collagenolytically active cathepsin K protein dimer. Cathepsin K is organized into elongated C-shaped protease dimers that reveal a putative collagen-binding interface aided by glycosaminoglycans. Molecular modeling of collagen binding to the dimer indicates the participation of nonactive site amino acid residues, Q21 and Q92, in collagen unfolding. Mutations at these sites as well as perturbation of the dimer protein-protein interface completely inhibit cathepsin-K-mediated fiber degradation without affecting the hydrolysis of gelatin or synthetic peptide. Using scanning electron microscopy, we demonstrate the specific binding of cathepsin K at the edge of the fibrillar gap region of collagen fibers, which suggest initial cleavage events at the N- and C-terminal ends of tropocollagen molecules. Edman degradation analysis of collagen fiber degradation products revealed those initial cleavage sites. We propose that one cathepsin K molecule binds to collagen-bound glycosaminoglycans at the gap region and recruits a second protease molecule that provides an unfolding and cleavage mechanism for triple helical collagen. Removal of collagen-associated glycosaminoglycans prevents cathepsin K binding and subsequently fiber hydrolysis. Cathepsin K dimer and glycosaminoglycan binding sites represent novel targeting sites for the development of nonactive site-directed second-generation inhibitors of this important drug target.


Assuntos
Catepsina K/química , Colágeno/química , Aminoácidos/química , Sítios de Ligação , Remodelação Óssea , Osso e Ossos/metabolismo , Cristalografia por Raios X , Glicosaminoglicanos/química , Humanos , Hidrólise , Microscopia Eletrônica , Modelos Moleculares , Mutagênese , Osteoporose , Peptídeo Hidrolases/química , Pichia , Desnaturação Proteica , Dobramento de Proteína , Multimerização Proteica , Estrutura Terciária de Proteína , Especificidade por Substrato
4.
J Nat Prod ; 79(8): 1962-70, 2016 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-27498895

RESUMO

Natural products are an important source of novel drug scaffolds. The highly variable and unpredictable timelines associated with isolating novel compounds and elucidating their structures have led to the demise of exploring natural product extract libraries in drug discovery programs. Here we introduce affinity crystallography as a new methodology that significantly shortens the time of the hit to active structure cycle in bioactive natural product discovery research. This affinity crystallography approach is illustrated by using semipure fractions of an actinomycetes culture extract to isolate and identify a cathepsin K inhibitor and to compare the outcome with the traditional assay-guided purification/structural analysis approach. The traditional approach resulted in the identification of the known inhibitor antipain (1) and its new but lower potency dehydration product 2, while the affinity crystallography approach led to the identification of a new high-affinity inhibitor named lichostatinal (3). The structure and potency of lichostatinal (3) was verified by total synthesis and kinetic characterization. To the best of our knowledge, this is the first example of isolating and characterizing a potent enzyme inhibitor from a partially purified crude natural product extract using a protein crystallographic approach.


Assuntos
Produtos Biológicos/farmacologia , Catepsina K/antagonistas & inibidores , Líquens/química , Peptídeos/isolamento & purificação , Peptídeos/farmacologia , Antipaína/química , Antipaína/farmacologia , Produtos Biológicos/síntese química , Produtos Biológicos/química , Colúmbia Britânica , Cristalografia por Raios X , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Peptídeos/química
5.
Biochim Biophys Acta ; 1834(12): 2546-53, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23954305

RESUMO

The citrate synthase (CS) of Escherichia coli is an allosteric hexameric enzyme specifically inhibited by NADH. The crystal structure of wild type (WT) E. coli CS, determined by us previously, has no substrates bound, and part of the active site is in a highly mobile region that is shifted from the position needed for catalysis. The CS of Acetobacter aceti has a similar structure, but has been successfully crystallized with bound substrates: both oxaloacetic acid (OAA) and an analog of acetyl coenzyme A (AcCoA). We engineered a variant of E. coli CS wherein five amino acids in the mobile region have been replaced by those in the A. aceti sequence. The purified enzyme shows unusual kinetics with a low affinity for both substrates. Although the crystal structure without ligands is very similar to that of the WT enzyme (except in the mutated region), complexes are formed with both substrates and the allosteric inhibitor NADH. The complex with OAA in the active site identifies a novel OAA-binding residue, Arg306, which has no functional counterpart in other known CS-OAA complexes. This structure may represent an intermediate in a multi-step substrate binding process where Arg306 changes roles from OAA binding to AcCoA binding. The second complex has the substrate analog, S-carboxymethyl-coenzyme A, in the allosteric NADH-binding site and the AcCoA site is not formed. Additional CS variants unable to bind adenylates at the allosteric site show that this second complex is not a factor in positive allosteric activation of AcCoA binding.


Assuntos
Acetobacter/enzimologia , Acetilcoenzima A/química , Citrato (si)-Sintase/química , Proteínas de Escherichia coli/química , Escherichia coli/enzimologia , NADP/química , Acetobacter/genética , Acetilcoenzima A/genética , Acetilcoenzima A/metabolismo , Regulação Alostérica , Animais , Domínio Catalítico , Citrato (si)-Sintase/genética , Citrato (si)-Sintase/metabolismo , Escherichia coli/genética , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , NADP/genética , NADP/metabolismo , Ligação Proteica , Suínos
6.
J Clin Invest ; 130(2): 699-714, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-31845908

RESUMO

HSP27 is highly expressed in, and supports oncogene addiction of, many cancers. HSP27 phosphorylation is a limiting step for activation of this protein and a target for inhibition, but its highly disordered structure challenges rational structure-guided drug discovery. We performed multistep biochemical, structural, and computational experiments to define a spherical 24-monomer complex composed of 12 HSP27 dimers with a phosphorylation pocket flanked by serine residues between their N-terminal domains. Ivermectin directly binds this pocket to inhibit MAPKAP2-mediated HSP27 phosphorylation and depolymerization, thereby blocking HSP27-regulated survival signaling and client-oncoprotein interactions. Ivermectin potentiated activity of anti-androgen receptor and anti-EGFR drugs in prostate and EGFR/HER2-driven tumor models, respectively, identifying a repurposing approach for cotargeting stress-adaptive responses to overcome resistance to inhibitors of oncogenic pathway signaling.


Assuntos
Proteínas de Choque Térmico , Ivermectina , Chaperonas Moleculares , Neoplasias Experimentais , Receptor ErbB-2 , Células A549 , Animais , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Ivermectina/química , Ivermectina/farmacologia , Camundongos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/química , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Domínios Proteicos , Multimerização Proteica , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo
7.
Artigo em Inglês | MEDLINE | ID: mdl-19478443

RESUMO

Autotransporters (ATs) are proteins that deliver effectors (the passenger domain) to the surface of Gram-negative bacteria by the type V secretion pathway. The passenger domain of BrkA, a Bordetella pertussis autotransporter mediating serum resistance and adherence, was cloned in a pET expression system and overexpressed in Escherichia coli. The gene product was correctly refolded, purified to homogeneity and crystallized. The crystals diffracted to 2.8 A resolution. The space group was assumed to be P4(1)2(1)2, with unit-cell parameters a = b = 108.19, c = 115.35 A.


Assuntos
Proteínas da Membrana Bacteriana Externa/química , Bordetella pertussis/química , Proteínas de Membrana Transportadoras/química , Sequência de Aminoácidos , Proteínas da Membrana Bacteriana Externa/genética , Proteínas da Membrana Bacteriana Externa/isolamento & purificação , Proteínas da Membrana Bacteriana Externa/fisiologia , Bordetella pertussis/genética , Dicroísmo Circular , Clonagem Molecular , Cristalização , Coleta de Dados , Escherichia coli/genética , Corpos de Inclusão/química , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/fisiologia , Dados de Sequência Molecular , Dobramento de Proteína , Sinais Direcionadores de Proteínas , Estrutura Terciária de Proteína/genética , Proteínas/genética , Proteínas/metabolismo , Via Secretória , Homologia de Sequência de Aminoácidos , Estatística como Assunto , Temperatura , Difração de Raios X
8.
FEBS Lett ; 590(8): 1143-51, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27000970

RESUMO

As part of a search for selective, mechanism-based covalent inhibitors of human pancreatic α-amylase we describe the chemoenzymatic synthesis of the disaccharide analog α-glucosyl epi-cyclophellitol, demonstrate its stoichiometric reaction with human pancreatic α-amylase and evaluate the time dependence of its inhibition. X-ray crystallographic analysis of the covalent derivative so formed confirms its reaction at the active site with formation of a covalent bond to the catalytic nucleophile D197. The structure illuminates the interactions with the active site and confirms OH4' on the nonreducing end sugar as a good site for attachment of fluorescent tags in generating probes for localization and quantitation of amylase in vivo.


Assuntos
Cicloexanóis/farmacologia , Dissacarídeos/farmacologia , Compostos de Epóxi/farmacologia , alfa-Amilases Pancreáticas/química , alfa-Amilases Pancreáticas/metabolismo , Domínio Catalítico , Simulação por Computador , Humanos , Ligação de Hidrogênio , Inositol/análogos & derivados , Inositol/química , Inositol/metabolismo , Cinética , Espectrometria de Massas , Modelos Moleculares , Água , Difração de Raios X
9.
ACS Cent Sci ; 2(3): 154-161, 2016 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-27066537

RESUMO

Selective inhibitors of human pancreatic α-amylase (HPA) are an effective means of controlling blood sugar levels in the management of diabetes. A high-throughput screen of marine natural product extracts led to the identification of a potent (Ki = 10 pM) peptidic HPA inhibitor, helianthamide, from the Caribbean sea anemone Stichodactyla helianthus. Active helianthamide was produced in Escherichia coli via secretion as a barnase fusion protein. X-ray crystallographic analysis of the complex of helianthamide with porcine pancreatic α-amylase revealed that helianthamide adopts a ß-defensin fold and binds into and across the amylase active site, utilizing a contiguous YIYH inhibitory motif. Helianthamide represents the first of a novel class of glycosidase inhibitors and provides an unusual example of functional malleability of the ß-defensin fold, which is rarely seen outside of its traditional role in antimicrobial peptides.

10.
Biochemistry ; 42(19): 5555-65, 2003 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-12741811

RESUMO

Study of the hexameric and allosterically regulated citrate synthases (type II CS) provides a rare opportunity to gain not only an understanding of a novel allosteric mechanism but also insight into how such properties can evolve from an unregulated structural platform (the dimeric type I CS). To address both of these issues, we have determined the structure of the complex of NADH (a negative allosteric effector) with the F383A variant of type II Escherichia coli CS. This variant was chosen because its kinetics indicate it is primarily in the T or inactive allosteric conformation, the state that strongly binds to NADH. Our structural analyses show that the six NADH binding sites in the hexameric CS complex are located at the interfaces between dimer units such that most of each site is formed by one subunit, but a number of key residues are drawn from the adjacent dimer. This arrangement of interactions serves to explain why NADH allosteric regulation is a feature only of hexameric type II CS. Surprisingly, in both the wild-type enzyme and the NADH complex, the two subunits of each dimer within the hexameric conformation are similar but not identical in structure, and therefore, while the general characteristics of NADH binding interactions are similar in each subunit, the details of these are somewhat different between subunits. Detailed examination of the observed NADH binding sites indicates that both direct charged interactions and the overall cationic nature of the sites are likely responsible for the ability of these sites to discriminate between NADH and NAD(+). A particularly novel characteristic of the complex is the horseshoe conformation assumed by NADH, which is strikingly different from the extended conformation found in its complexes with most proteins. Sequence homology studies suggest that this approach to binding NADH may arise out of the evolutionary need to add an allosteric regulatory function to the base CS structure. Comparisons of the amino acid sequences of known type II CS enzymes, from different Gram-negative bacteria taxonomic groups, show that the NADH-binding residues identified in our structure are strongly conserved, while hexameric CS molecules that are insensitive to NADH have undergone key changes in the sequence of this part of the protein.


Assuntos
Citrato (si)-Sintase/química , Citrato (si)-Sintase/metabolismo , NAD/metabolismo , Regulação Alostérica , Sítio Alostérico/genética , Sequência de Aminoácidos , Citrato (si)-Sintase/classificação , Citrato (si)-Sintase/genética , Cristalografia por Raios X , Dimerização , Escherichia coli/enzimologia , Escherichia coli/genética , Evolução Molecular , Variação Genética , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Estrutura Quaternária de Proteína , Homologia de Sequência de Aminoácidos
11.
J Biol Chem ; 278(37): 35435-43, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12824188

RESUMO

The citrate synthase of Escherichia coli is an example of a Type II citrate synthase, a hexamer that is subject to allosteric inhibition by NADH. In previous crystallographic work, we defined the NADH binding sites, identifying nine amino acids whose side chains were proposed to make hydrogen bonds with the NADH molecule. Here, we describe the functional properties of nine sequence variants, in which these have been replaced by nonbonding residues. All of the variants show some changes in NADH binding and inhibition and small but significant changes in kinetic parameters for catalysis. In three cases, Y145A, R163L, and K167A, NADH inhibition has become extremely weak. We have used nanospray/time-of-flight mass spectrometry, under non-denaturing conditions, to show that two of these, R163L and K167A, do not form hexamers in response to NADH binding, unlike the wild type enzyme. One variant, R109L, shows tighter NADH binding. We have crystallized this variant and determined its structure, with and without bound NADH. Unexpectedly, the greatest structural changes in the R109L variant are in two regions outside the NADH binding site, both of which, in wild type citrate synthase, have unusually high mobilities as measured by crystallographic thermal factors. In the R109L variant, both regions (residues 260 -311 and 316-342) are much less mobile and have rearranged significantly. We argue that these two regions are elements in the path of communication between the NADH binding sites and the active sites and are centrally involved in the regulatory conformational change in E. coli citrate synthase.


Assuntos
Citrato (si)-Sintase/química , Citrato (si)-Sintase/metabolismo , Escherichia coli/enzimologia , NAD/metabolismo , Substituição de Aminoácidos , Sítios de Ligação , Cristalografia por Raios X , Variação Genética , Ligação de Hidrogênio , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Eletricidade Estática , Termodinâmica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA