Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 125(22): 3484-90, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25788700

RESUMO

Ticagrelor is a direct-acting reversibly binding P2Y12 antagonist and is widely used as an antiplatelet therapy for the prevention of cardiovascular events in acute coronary syndrome patients. However, antiplatelet therapy can be associated with an increased risk of bleeding. Here, we present data on the identification and the in vitro and in vivo pharmacology of an antigen-binding fragment (Fab) antidote for ticagrelor. The Fab has a 20 pM affinity for ticagrelor, which is 100 times stronger than ticagrelor's affinity for its target, P2Y12. Despite ticagrelor's structural similarities to adenosine, the Fab is highly specific and does not bind to adenosine, adenosine triphosphate, adenosine 5'-diphosphate, or structurally related drugs. The antidote concentration-dependently neutralized the free fraction of ticagrelor and reversed its antiplatelet activity both in vitro in human platelet-rich plasma and in vivo in mice. Lastly, the antidote proved effective in normalizing ticagrelor-dependent bleeding in a mouse model of acute surgery. This specific antidote for ticagrelor may prove valuable as an agent for patients who require emergency procedures.


Assuntos
Adenosina/análogos & derivados , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/farmacologia , Antídotos/química , Antídotos/farmacologia , Adenosina/antagonistas & inibidores , Adenosina/imunologia , Animais , Anticorpos/isolamento & purificação , Anticorpos/metabolismo , Especificidade de Anticorpos , Anticorpos Amplamente Neutralizantes , Células CHO , Cricetinae , Cricetulus , Cristalografia por Raios X , Hemorragia/prevenção & controle , Humanos , Fragmentos Fab das Imunoglobulinas/farmacologia , Camundongos , Modelos Moleculares , Agregação Plaquetária/efeitos dos fármacos , Engenharia de Proteínas , Ticagrelor
2.
J Biol Chem ; 288(51): 36636-47, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24194519

RESUMO

The neutrophil enzyme myeloperoxidase (MPO) promotes oxidative stress in numerous inflammatory pathologies by producing hypohalous acids. Its inadvertent activity is a prime target for pharmacological control. Previously, salicylhydroxamic acid was reported to be a weak reversible inhibitor of MPO. We aimed to identify related hydroxamates that are good inhibitors of the enzyme. We report on three hydroxamates as the first potent reversible inhibitors of MPO. The chlorination activity of purified MPO was inhibited by 50% by a 5 nm concentration of a trifluoromethyl-substituted aromatic hydroxamate, HX1. The hydroxamates were specific for MPO in neutrophils and more potent toward MPO compared with a broad range of redox enzymes and alternative targets. Surface plasmon resonance measurements showed that the strength of binding of hydroxamates to MPO correlated with the degree of enzyme inhibition. The crystal structure of MPO-HX1 revealed that the inhibitor was bound within the active site cavity above the heme and blocked the substrate channel. HX1 was a mixed-type inhibitor of the halogenation activity of MPO with respect to both hydrogen peroxide and halide. Spectral analyses demonstrated that hydroxamates can act variably as substrates for MPO and convert the enzyme to a nitrosyl ferrous intermediate. This property was unrelated to their ability to inhibit MPO. We propose that aromatic hydroxamates bind tightly to the active site of MPO and prevent it from producing hypohalous acids. This mode of reversible inhibition has potential for blocking the activity of MPO and limiting oxidative stress during inflammation.


Assuntos
Inibidores Enzimáticos/farmacologia , Hidrocarbonetos Aromáticos/farmacologia , Ácidos Hidroxâmicos/farmacologia , Peroxidase/química , Sequência de Aminoácidos , Sítios de Ligação , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , Humanos , Hidrocarbonetos Aromáticos/síntese química , Hidrocarbonetos Aromáticos/química , Ácidos Hidroxâmicos/química , Cinética , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Neutrófilos/enzimologia , Peroxidase/antagonistas & inibidores , Peroxidase/metabolismo , Ligação Proteica
3.
Proc Natl Acad Sci U S A ; 110(10): 3806-11, 2013 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-23431194

RESUMO

Prostaglandin E2 (PGE2) is a key mediator in inflammatory response. The main source of inducible PGE2, microsomal PGE2 synthase-1 (mPGES-1), has emerged as an interesting drug target for treatment of pain. To support inhibitor design, we have determined the crystal structure of human mPGES-1 to 1.2 Å resolution. The structure reveals three well-defined active site cavities within the membrane-spanning region in each monomer interface of the trimeric structure. An important determinant of the active site cavity is a small cytosolic domain inserted between transmembrane helices I and II. This extra domain is not observed in other structures of proteins within the MAPEG (Membrane-Associated Proteins involved in Eicosanoid and Glutathione metabolism) superfamily but is likely to be present also in microsomal GST-1 based on sequence similarity. An unexpected feature of the structure is a 16-Å-deep cone-shaped cavity extending from the cytosolic side into the membrane-spanning region. We suggest a potential role for this cavity in substrate access. Based on the structure of the active site, we propose a catalytic mechanism in which serine 127 plays a key role. We have also determined the structure of mPGES-1 in complex with a glutathione-based analog, providing insight into mPGES-1 flexibility and potential for structure-based drug design.


Assuntos
Oxirredutases Intramoleculares/química , Sequência de Aminoácidos , Domínio Catalítico , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Glutationa/análogos & derivados , Glutationa/química , Humanos , Oxirredutases Intramoleculares/antagonistas & inibidores , Oxirredutases Intramoleculares/genética , Microssomos/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Prostaglandina-E Sintases , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Homologia de Sequência de Aminoácidos
4.
ACS Med Chem Lett ; 3(9): 705-9, 2012 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900538

RESUMO

Trk receptor tyrosine kinases have been implicated in cancer and pain. A crystal structure of TrkA with AZ-23 (1a) was obtained, and scaffold hopping resulted in two 5/6-bicyclic series comprising either imidazo[4,5-b]pyridines or purines. Further optimization of these two fusion series led to compounds with subnanomolar potencies against TrkA kinase in cellular assays. Antitumor effects in a TrkA-driven mouse allograft model were demonstrated with compounds 2d and 3a.

5.
J Biol Chem ; 284(16): 10672-83, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19211551

RESUMO

Cyclotides are a family of plant defense proteins that are highly resistant to adverse chemical, thermal, and enzymatic treatment. Here, we present the first crystal structure of a cyclotide, varv F, from the European field pansy, Viola arvensis, determined at a resolution of 1.8 A. The solution state NMR structure was also determined and, combined with measurements of biophysical parameters for several cyclotides, provided an insight into the structural features that account for the remarkable stability of the cyclotide family. The x-ray data confirm the cystine knot topology and the circular backbone, and delineate a conserved network of hydrogen bonds that contribute to the stability of the cyclotide fold. The structural role of a highly conserved Glu residue that has been shown to regulate cyclotide function was also determined, verifying its involvement in a stabilizing hydrogen bond network. We also demonstrate that varv F binds to dodecylphosphocholine micelles, defining the binding orientation and showing that its structure remains unchanged upon binding, further demonstrating that the cyclotide fold is rigid. This study provides a biological insight into the mechanism by which cyclotides maintain their native activity in the unfavorable environment of predator insect guts. It also provides a structural basis for explaining how a cluster of residues important for bioactivity may be involved in self-association interactions in membranes. As well as being important for their bioactivity, the structural rigidity of cyclotides makes them very suitable as a stable template for peptide-based drug design.


Assuntos
Ciclotídeos , Motivos Nó de Cisteína , Desenho de Fármacos , Inseticidas , Conformação Proteica , Dobramento de Proteína , Sequência de Aminoácidos , Animais , Cristalografia por Raios X , Ciclotídeos/química , Ciclotídeos/genética , Ciclotídeos/metabolismo , Humanos , Ligação de Hidrogênio , Inseticidas/química , Inseticidas/metabolismo , Dados de Sequência Molecular , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA