Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 335(3): 580-8, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20823195

RESUMO

In recent years immunotherapy-based approaches for treating Alzheimer's disease have become the subject of intensive research. However, an important mechanistic-related safety concern is exacerbation of the risk of microhemorrhage that may be associated with fast removal of amyloid-ß (Aß) deposits found in blood vessels or brain parenchyma. Rapid in vivo detection of microhemorrhages in living amyloid precursor protein transgenic mice has not been described, and histological analysis can take several months before this risk is assessed. Aged transgenic mice were divided into two groups that would undergo longitudinal passive immunotherapy for 12 or 18 weeks. 6G1, a nonselective anti-Aß monoclonal antibody, and 8F5, a more selective antioligomeric Aß monoclonal antibody, were examined in both longitudinal studies. High-resolution T2*-weighted magnetic resonance microscopy (100 × 100 × 400 µm) was used for microhemorrhage detection in vivo. Cerebral microhemorrhages by magnetic resonance imaging were compared with histological hemosiderin staining in each animal; results showed that T2*-weighted magnetic resonance microscopy can reliably detect microhemorrhages of ≥60 µm in diameter at baseline and after 12 to 18 weeks of treatment in the same animals in vivo. This correlated significantly with histological readings. This new imaging safety biomarker can be readily applied to preclinical antibody screening in a longitudinal manner. 6G1 and 8F5, however, both increased microhemorrhage incidence in aged amyloid precursor protein transgenic mice compared with their baseline and vehicle treatment. A highly selective antibody for soluble Aß is needed to address the question of whether antibodies that do not bind to deposited Aß have microhemorrhage liability.


Assuntos
Doença de Alzheimer/terapia , Precursor de Proteína beta-Amiloide/genética , Hemorragia Cerebral/diagnóstico , Imunização Passiva/efeitos adversos , Imageamento por Ressonância Magnética/métodos , Peptídeos beta-Amiloides/imunologia , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Córtex Cerebral/patologia , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/patologia , Modelos Animais de Doenças , Feminino , Humanos , Estudos Longitudinais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Tempo
2.
Chem Res Toxicol ; 20(12): 1752-9, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18001056

RESUMO

We have recently reported on the development of a La assay to detect reactive molecules by nuclear magnetic resonance (ALARM NMR) to detect reactive false positive hits from high-throughput screening, in which we observed a surprisingly large number of compounds that can oxidize or form covalent adducts with protein thiols groups. In the vast majority of these cases, the covalent interactions are largely nonspecific (e.g., affect many protein targets) and therefore unsuitable for drug development. However, certain thiol-reactive species do appear to inhibit the target of interest in a specific manner. The question then arises as to the potential toxicology risks of developing a drug that can react with protein thiol groups. Here, we report on the evaluation of a large set of ALARM-reactive and -nonreactive compounds against a panel of additional proteins (aldehyde dehydrogenase, superoxide dismutase, and three cytochrome P450 enzymes). It was observed that ALARM-reactive compounds have significantly increased risks of interacting with one or more of these enzymes in vitro. Thus, ALARM NMR seems to be a sensitive tool to rapidly identify compounds with an enhanced risk of producing side effects in humans, including alcohol intolerance, the formation of reactive oxygen species, and drug-drug interactions. In conjunction with other toxicology assays, ALARM NMR should be a valuable tool for prioritizing compounds for lead optimization and animal testing.


Assuntos
Aldeído Desidrogenase/química , Autoantígenos/química , Inibidores das Enzimas do Citocromo P-450 , Preparações Farmacêuticas , Ribonucleoproteínas/química , Compostos de Sulfidrila/química , Superóxido Dismutase/química , Aldeído Desidrogenase/metabolismo , Desenho de Fármacos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Humanos , Espectroscopia de Ressonância Magnética , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/enzimologia , Estrutura Molecular , Preparações Farmacêuticas/análise , Ligação Proteica , Compostos de Sulfidrila/metabolismo , Superóxido Dismutase/metabolismo , Antígeno SS-B
3.
Chem Biol Drug Des ; 70(1): 1-12, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17630989

RESUMO

The molecular chaperone HSP90 has been shown to facilitate cancer cell survival by stabilizing key proteins responsible for a malignant phenotype. We report here the results of parallel fragment-based drug design approaches in the design of novel HSP90 inhibitors. Initial aminopyrimidine leads were elaborated using high-throughput organic synthesis to yield nanomolar inhibitors of the enzyme. Second site leads were also identified which bound to HSP90 in two distinct conformations, an 'open' and 'closed' form. Intriguingly, linked fragment approaches targeting both of these conformations were successful in producing novel, micromolar inhibitors. Overall, this study shows that, with only a few fragment hits, multiple lead series can be generated for HSP90 due to the inherent flexibility of the active site. Thus, ample opportunities exist to use these lead series in the development of clinically useful HSP90 inhibitors for the treatment of cancers.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Fragmentos de Peptídeos , Aminopiridinas/química , Aminopiridinas/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Ligantes , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Conformação Proteica
4.
J Comput Aided Mol Des ; 21(1-3): 121-30, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17294246

RESUMO

Two new proteins of approximately 70 amino acids in length, corresponding to an unnaturally-linked N- and C-helix of the ectodomain of the gp41 protein from the human immunodeficiency virus (HIV) type 1, were designed and characterized. A designed tripeptide links the C-terminus of the C-helix with the N-terminus of the N-helix in a circular permutation so that the C-helix precedes the N-helix in sequence. In addition to the artificial peptide linkage, the C-helix is truncated at its N-terminus to expose a region of the N-helix known as the "Trp-Trp-Ile" binding pocket. Sedimentation, crystallographic, and nuclear magnetic resonance studies confirmed that the protein had the desired trimeric structure with an unoccupied binding site. Spectroscopic and centrifugation studies demonstrated that the engineered protein had ligand binding characteristics similar to previously reported constructs. Unlike previous constructs which expose additional, shallow, non-conserved, and undesired binding pockets, only the single deep and conserved Trp-Trp-Ile pocket is exposed in the proteins of this study. This engineered version of gp41 protein will be potentially useful in research programs aimed at discovery of new drugs for therapy of HIV-infection in humans.


Assuntos
Desenho de Fármacos , Proteína gp41 do Envelope de HIV/química , HIV-1/química , Engenharia de Proteínas , Sequência de Aminoácidos , Sequência de Bases , Sítios de Ligação , Proteína gp41 do Envelope de HIV/genética , HIV-1/genética , Humanos , Dados de Sequência Molecular , Conformação Proteica
5.
J Med Chem ; 49(23): 6726-31, 2006 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-17154503

RESUMO

Adenosine kinase (AK) is an enzyme responsible for converting endogenous adenosine (ADO) to adenosine monophosphate (AMP) in an adenosine triphosphate- (ATP-) dependent manner. The structure of AK consists of two domains, the first a large alpha/beta Rossmann-like nucleotide binding domain that forms the ATP binding site, and a smaller mixed alpha/beta domain, which, in combination with the larger domain, forms the ADO binding site and the site of phosphoryl transfer. AK inhibitors have been under investigation as antinociceptive, antiinflammatory, and anticonvulsant as well as antiinfective agents. In this work, we report the structures of AK in complex with two classes of inhibitors: the first, ADO-like, and the second, a novel alkynylpyrimidine series. The two classes of structures, which contain structurally similar substituents, reveal distinct binding modes in which the AK structure accommodates the inhibitor classes by a 30 degrees rotation of the small domain relative to the large domain. This change in binding mode stabilizes an open and a closed intermediate structural state and provide structural insight into the transition required for catalysis. This results in a significant rearrangement of both the protein active site and the orientation of the alkynylpyrimidine ligand when compared to the observed orientation of nucleosidic inhibitors or substrates.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Adenosina Quinase/química , Inibidores Enzimáticos/química , Morfolinas/química , Pirimidinas/química , Tubercidina/análogos & derivados , Animais , Sítios de Ligação , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Conformação Proteica , Toxoplasma/enzimologia , Tubercidina/química
6.
J Med Chem ; 47(7): 1709-18, 2004 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15027862

RESUMO

Potent inhibitors of 7,8-dihydroneopterin aldolase (DHNA; EC 4.1.2.25) have been discovered using CrystaLEAD X-ray crystallographic high-throughput screening followed by structure-directed optimization. Screening of a 10 000 compound random library provided several low affinity leads and their corresponding X-ray crystal structures bound to the enzyme. The presence of a common structural feature in each of the leads suggested a strategy for the construction of a directed library of approximately 1000 compounds that were screened for inhibitory activity in a traditional enzyme assay. Several lead compounds with IC(50) values of about 1 microM against DHNA were identified, and crystal structures of their enzyme-bound complexes were obtained by cocrystallization. Structure-directed optimization of one of the leads thus identified afforded potent inhibitors with submicromolar IC(50) values.


Assuntos
Aldeído Liases/antagonistas & inibidores , Aldeído Liases/química , Benzoatos/química , Inibidores Enzimáticos/química , Neopterina/química , Pirimidinas/química , Triazóis/química , Benzoatos/síntese química , Sítios de Ligação , Cristalografia por Raios X , Bases de Dados Factuais , Inibidores Enzimáticos/síntese química , Guanina/análogos & derivados , Guanina/síntese química , Guanina/química , Modelos Moleculares , Estrutura Molecular , Purinas/química , Pirimidinas/síntese química , Relação Estrutura-Atividade , Triazóis/síntese química
7.
Anticancer Res ; 24(6): 3907-10, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15736430

RESUMO

Chk1 (checkpoint kinase 1) is a serine-threonine kinase that is critical for G2/M arrest in response to DNA damage. Chk1 phosphorylates Cdc25C at serine-216, a major regulatory site, in response to DNA damage. Furthermore, Chk1 also phosphorylates Cdc25A on serine 123 which accelerates its degradation through the ubiquitin-proteasome pathway and arrests cells in late G2-phase after DNA damage. In the present study, we demonstrated that Chk1 phosphorylates pro-apoptotic protein BAD (Bcl-2/Bcl-XL-Antagonist, causing cell Death) in vitro. In vitro phosphorylation analysis with various mouse BAD peptides has revealed two phosphorylation sites for Chk1 at serine-155 and serine-170. When wild-type and mutant BAD (S155A) constructs were transfected into 293T cells, an association between BAD and Chk1 was observed by co-immunoprecipitation. In addition, there was an increase in the phosphorylation of serine-155 following DNA damage by adriamycin treatment. Our results suggest that Chk1 associates with BAD and phosphorylates the BAD protein at serine-155. Taken together, our results suggest that Chk1 may inactivate BAD by associating with and phosphorylating residues critical for BAD function in response to DNA damage.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas Quinases/metabolismo , Sequência de Aminoácidos , Linhagem Celular , Quinase 1 do Ponto de Checagem , Embrião de Mamíferos , Humanos , Rim/citologia , Dados de Sequência Molecular , Fosfatos de Poli-Isoprenil , Ligação Proteica , Proteína de Morte Celular Associada a bcl
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...