Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Infect Dis ; 77(2): 186-193, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-36996150

RESUMEN

BACKGROUND: The vast majority of coronavirus disease 2019 (COVID-19) disease occurs in outpatients where treatment is limited to antivirals for high-risk subgroups. Acebilustat, a leukotriene B4 inhibitor, has potential to reduce inflammation and symptom duration. METHODS: In a single-center trial spanning Delta and Omicron variants, outpatients were randomized to 100 mg/d of oral acebilustat or placebo for 28 days. Patients reported daily symptoms via electronic query through day 28 with phone follow-up on day 120 and collected nasal swab samples on days 1-10. The primary outcome was sustained symptom resolution to day 28. Secondary 28-day outcomes included time to first symptom resolution, area under the curve (AUC) for longitudinal daily symptom scores, duration of viral shedding through day 10, and symptoms on day 120. RESULTS: Sixty participants were randomized to each study arm. At enrollment, the median duration was 4 days (interquartile range, 3-5 days), and the median number of symptoms was 9 (7-11). Most patients (90%) were vaccinated, with 73% having neutralizing antibodies. A minority of participants (44%; 35% in the acebilustat arm and 53% in placebo) had sustained symptom resolution at day 28 (hazard ratio, 0.6 [95% confidence interval, .34-1.04]; P = .07 favoring placebo). There was no difference in the mean AUC for symptom scores over 28 days (difference in mean AUC, 9.4 [95% confidence interval, -42.1 to 60.9]; P = .72). Acebilustat did not affect viral shedding or symptoms at day 120. CONCLUSIONS: Sustained symptoms through day 28 were common in this low-risk population. Despite this, leukotriene B4 antagonism with acebilustat did not shorten symptom duration in outpatients with COVID-19. Clinical Trials Registration. NCT04662060.


Asunto(s)
COVID-19 , Humanos , SARS-CoV-2 , Leucotrieno B4 , Pacientes Ambulatorios , Método Doble Ciego , Resultado del Tratamiento
2.
Exp Dermatol ; 29(12): 1191-1198, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33047366

RESUMEN

Bullous pemphigoid (BP) is an autoimmune blistering disease characterized by recruitment of leucocytes into skin and release of damaging enzymes, resulting in epidermal detachment and blister formation. To better understand the role of leukotriene B4 (LTB4) and other inflammatory factors in BP pathophysiology, we conducted microscopic and immunohistochemical analyses of preserved skin biopsy sections and conducted flow cytometry and ELISA analyses of matched blood and blister fluid from BP patients. Neutrophils predominated in BP blister fluid, which also contained monocytes/macrophages and T cells, but few to no eosinophils and B cells. In contrast, BP skin histology showed a different pattern, with abundant neutrophils but eosinophils being the predominant immune cell type. LTB4 pathway and neutrophil activation markers were prevalent in BP skin lesions and strongly associated with perivascular neutrophils. Blister fluid neutrophils, monocytes/macrophages and eosinophils all exhibited increased surface expression of leukotriene A4 hydrolase and neutrophil elastase (P = .002 for both). Blister fluid was also enriched in interleukins (IL)-1α, IL-1ß, IL-8, IL-10, IL-18, monocyte colony-stimulating factor (M-CSF) and vascular endothelial growth factor (VEGF). Our findings suggest differential leucocyte recruitment from blood into dermis and from dermis into blister, which correlates with disease activity, and presents potential new treatment opportunities for BP.


Asunto(s)
Exudados y Transudados/citología , Leucotrieno B4/metabolismo , Penfigoide Ampolloso/sangre , Penfigoide Ampolloso/patología , Piel/patología , Anciano , Anciano de 80 o más Años , Eosinófilos , Epóxido Hidrolasas/metabolismo , Exudados y Transudados/metabolismo , Femenino , Citometría de Flujo , Humanos , Interleucinas/metabolismo , Elastasa de Leucocito/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/enzimología , Masculino , Persona de Mediana Edad , Monocitos/enzimología , Infiltración Neutrófila , Neutrófilos/enzimología , Penfigoide Ampolloso/inmunología , Factores Raciales , Factores Sexuales , Piel/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
3.
Invest New Drugs ; 30(1): 90-7, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20820910

RESUMEN

LP-261 is a novel tubulin targeting anticancer agent that binds at the colchicine site on tubulin, inducing G2/M arrest. Screening in the NCI60 cancer cell lines resulted in a mean GI50 of approximately 100 nM. Here, we report the results of testing in multiple mouse xenograft models and angiogenesis assays, along with bioavailability studies. To determine the antiangiogenic activity of LP-261, both in vitro and ex vivo experiments were performed. Human umbilical vein endothelial cells (HUVECs) were incubated with LP-261 at 50 nM to 10 µM. LP-261 was also tested in a rat aortic ring assay, from 20 nM to 10 µM. Multiple mouse xenograft studies were performed to assess in vivo antitumor activity. LP-261 was tested as a single agent in colon adenocarcinoma (SW620) and prostate cancer (LNCaP and PC3) xenografts, evaluating several different dosing schedules. LP-261 was also used in combination with bevacizumab in the SW620 xenograft model. LP-261 also exhibited high oral bioavailability and apparent lack of efflux by intestinal transporters such as ABCB1. LP-261 is a very potent inhibitor of angiogenesis, preventing microvessel outgrowth in the rat aortic ring assay and HUVEC cell proliferation at nanomolar concentrations. Complete inhibition of tumor growth was achieved in the PC3 xenograft model and shown to be schedule dependent. Excellent inhibition of tumor growth in the SW620 model was observed, comparable with paclitaxel. Combining oral, low dose LP-261 with bevacizumab led to significantly improved tumor inhibition. Oral LP-261 is very effective at inhibiting tumor growth in multiple mouse xenograft models and is well tolerated.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Administración Oral , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/sangre , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Bevacizumab , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Absorción Intestinal , Mucosa Intestinal/metabolismo , Ácidos Isonicotínicos/administración & dosificación , Masculino , Ratones , Ratones Desnudos , Neoplasias/irrigación sanguínea , Neoplasias/patología , Neovascularización Fisiológica/efectos de los fármacos , Paclitaxel/administración & dosificación , Permeabilidad , Ratas , Ratas Sprague-Dawley , Sulfonamidas/administración & dosificación , Factores de Tiempo , Moduladores de Tubulina/administración & dosificación , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Bioorg Med Chem Lett ; 21(23): 7155-65, 2011 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-22014550

RESUMEN

Discovery of a new class of DFG-out p38α kinase inhibitors with no hinge interaction is described. A computationally assisted, virtual fragment-based drug design (vFBDD) platform was utilized to identify novel non-aromatic fragments which make productive hydrogen bond interactions with Arg 70 on the αC-helix. Molecules incorporating these fragments were found to be potent inhibitors of p38 kinase. X-ray co-crystal structures confirmed the predicted binding modes. A lead compound was identified as a potent (p38α IC(50)=22 nM) and highly selective (≥ 150-fold against 150 kinase panel) DFG-out p38 kinase inhibitor.


Asunto(s)
Simulación por Computador , Descubrimiento de Drogas , Inhibidores Enzimáticos , Oligopéptidos/química , Tiofenos , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Adenosina Trifosfato/química , Animales , Cristalografía por Rayos X , Dexametasona/farmacología , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Concentración 50 Inhibidora , Ratones , Modelos Moleculares , Estructura Molecular , Ratas , Tiofenos/síntesis química , Tiofenos/química , Tiofenos/farmacología
5.
Contemp Clin Trials ; 108: 106509, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34274494

RESUMEN

More than 3000 clinical trials related to COVID-19 have been registered through clinicaltrials.gov. With so many trials, there is a risk that many will be inconclusive due to being underpowered or due to an inability to recruit patients. At academic medical centers, multiple trials are competing for the same resources; the success of one may come at the expense of another. The COVID-19 Outpatient Pragmatic Protocol Study (COPPS) is a flexible phase 2, multi-site, randomized, blinded trial based at Stanford University designed to overcome these issues by simultaneously evaluating multiple COVID-19 treatments in the outpatient setting in one common platform with shared controls. This approach reduces the overall number of patients required for statistical power, while improving the likelihood that any enrolled patient receives active treatment. The platform study has two main domains designed to evaluate COVID-19 treatments by assessing their ability to reduce viral shedding (Viral Domain), measured with self-collected nasal swabs, or improve clinical outcomes (Clinical Domain), measured through self-reported symptomology data. Data are collected on both domains for all participants enrolled. Participants are followed over a 28-day period. COPPS has the advantage of pragmatism created around its workflow that is also appealing to potential participants because of a lower probability of inactive treatment. At the conclusion of this clinical trial we expect to have identified potentially effective therapeutic strategy/ies for treating COVID-19 in the outpatient setting, which will have a transformative impact on medicine and public health.


Asunto(s)
COVID-19 , Humanos , Pacientes Ambulatorios , Proyectos de Investigación , SARS-CoV-2 , Resultado del Tratamiento
6.
J Cyst Fibros ; 20(6): 1026-1034, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34538755

RESUMEN

BACKGROUND: Cystic fibrosis (CF) is characterized by neutrophilic inflammation in the airways. Leukotriene B4 (LTB4) is a neutrophil chemoattractant and has been implicated in CF pathogenesis. Acebilustat, a novel, synthetic, small-molecule leukotriene A4 hydrolase inhibitor, reduces LTB4 production. We report findings from a randomized placebo-controlled trial of acebilustat in adult subjects with mild-to-moderate lung disease. METHODS: Subjects were randomized (1:1:1) to once-daily acebilustat 50 mg, 100 mg or placebo for 48 weeks, concomitantly with their current therapeutic regimen. Subjects were stratified by use of concomitant CF transmembrane conductance regulator (CFTR) modulators, baseline percent predicted forced expiratory volume in 1 second (ppFEV1) 50-75 and >75, and number of pulmonary exacerbations in the past year (1 or >1). Primary endpoints were the change from baseline in ppFEV1 and safety. Secondary endpoints included the rate of pulmonary exacerbations. RESULTS: Overall, 199 subjects were randomized and dosed (acebilustat 50 mg, n=67; acebilustat 100 mg, n=66; placebo, n=66). Baseline demographics and disease profile were well balanced among treatment groups. Acebilustat had no statistically significant effect on the primary endpoint of change in ppFEV1 at week 48 or the secondary endpoint pulmonary exacerbations. There was a trend towards reduced pulmonary exacerbations in subjects receiving acebilustat in pre-specified populations with ppFEV1>75 (35% rate reduction) and those on concomitant CFTR modulator therapy (20% rate reduction). Acebilustat was well tolerated. CONCLUSIONS: Acebilustat did not improve lung function. A trend towards reduced pulmonary exacerbations in subjects with an earlier stage of lung disease suggests a potential effect in this population.


Asunto(s)
Compuestos de Azabiciclo/uso terapéutico , Benzoatos/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Epóxido Hidrolasas/uso terapéutico , Adolescente , Adulto , Método Doble Ciego , Femenino , Humanos , Masculino , Pruebas de Función Respiratoria , Índice de Severidad de la Enfermedad
7.
Biochemistry ; 49(17): 3611-8, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20337484

RESUMEN

Protein kinases c-Abl, b-Raf, and p38alpha are recognized as important targets for therapeutic intervention. c-Abl and b-Raf are major targets of marketed oncology drugs Imatinib (Gleevec) and Sorafenib (Nexavar), respectively, and BIRB-796 is a p38alpha inhibitor that reached Phase II clinical trials. A shared feature of these drugs is the fact that they bind to the DFG-out forms of their kinase targets. Although the discovery of this class of kinase inhibitors has increased the level of emphasis on the design of DFG-out inhibitors, the structural determinants for their binding and stabilization of the DFG-out conformation remain unclear. To improve our understanding of these determinants, we determined cocrystal structures of Imatinib and Sorafenib with p38alpha. We also conducted a detailed analysis of Imatinib and Sorafenib binding to p38alpha in comparison with BIRB-796, including binding kinetics, binding interactions, the solvent accessible surface area (SASA) of the ligands, and stabilization of key structural elements of the protein upon ligand binding. Our results yield an improved understanding of the structural requirements for stabilizing the DFG-out form and a rationale for understanding the genesis of ligand selectivity among DFG-out inhibitors of protein kinases.


Asunto(s)
Bencenosulfonatos/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Piperazinas/metabolismo , Inhibidores de Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Piridinas/metabolismo , Pirimidinas/metabolismo , Benzamidas , Cristalografía por Rayos X , Humanos , Mesilato de Imatinib , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 14 Activada por Mitógenos/química , Modelos Moleculares , Estructura Molecular , Naftalenos/farmacología , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Unión Proteica , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-abl/química , Pirazoles/farmacología , Sorafenib , Relación Estructura-Actividad
8.
Bioorg Med Chem Lett ; 20(22): 6592-6, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20888224

RESUMEN

The discovery and SAR study of a series of 4,6-diamino-1,3,5-triazin-2-ol compounds as novel HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) are reported. The lead compounds in this series showed excellent activity against wild-type and drug-resistant RT enzymes and viral strains. In addition, compounds from this series demonstrated favorable pharmacokinetic profile in rat. A preliminary modeling study was conducted to understand the binding mode of this series of compounds.


Asunto(s)
Descubrimiento de Drogas , Inhibidores de la Transcriptasa Inversa/química , Inhibidores de la Transcriptasa Inversa/farmacología , Triazinas/síntesis química , Triazinas/farmacología , Animales , Modelos Moleculares , Ratas , Inhibidores de la Transcriptasa Inversa/farmacocinética , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 19(19): 5693-7, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19699090

RESUMEN

A series of substituted biphenyl ethylene ether compounds has been designed to target the gp41N-trimer in order to inhibit formation of the six-helical bundle that represents the end state of gp41-mediated viral fusion. A size exclusion HPLC based helical bundle formation (HBF) assay was developed to evaluate in vitro inhibitory affinity of the inhibitors. The most potent compound 1 had an IC(50) of 31microM. The binding of compound 1 to the proposed hydrophobic pocket of gp41 was further validated by site-directed peptide mutagenesis experiments.


Asunto(s)
Fármacos Anti-VIH/química , Éteres/química , Proteína gp41 de Envoltorio del VIH/química , Naftalenos/química , Tetrazoles/química , Fármacos Anti-VIH/síntesis química , Fármacos Anti-VIH/farmacología , Sitios de Unión , Simulación por Computador , Cristalografía por Rayos X , Diseño de Fármacos , Éteres/síntesis química , Éteres/farmacología , Proteína gp41 de Envoltorio del VIH/antagonistas & inhibidores , Proteína gp41 de Envoltorio del VIH/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Ligandos , Naftalenos/síntesis química , Naftalenos/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Tetrazoles/síntesis química , Tetrazoles/farmacología
10.
Contemp Clin Trials ; 72: 86-94, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30056216

RESUMEN

Inflammation causes irreparable damage in the cystic fibrosis (CF) lung. Despite high standards of care and the advent of new therapies, inflammation continues to cause significant loss of lung function and morbidity. Acebilustat is a once-daily, oral molecule with anti-inflammatory activity through the inhibition of LTA4 hydrolase and modulation of LTB4. It has potential to reduce lung function decline and pulmonary exacerbations in patients with CF and is currently being tested in a Phase II multicenter, randomized, double-blind, placebo-controlled, parallel-group study (EMPIRE-CF). Strict inclusion criteria based on modeling of the Cystic Fibrosis Foundation Patient Registry data were selected to enrich the trial with patients most likely to benefit from chronic anti-inflammatory therapy that reduces lung function decline. 200 patients between 18 and 30 years of age, with an FEV1 percent predicted (pp) ≥50%, and ≥1 exacerbation in the past year have been enrolled. Patients are randomized 1:1:1 to placebo, acebilustat 50 mg or 100 mg for 48 weeks, taken concomitantly with their current standard of care, and stratified based on concomitant CFTR modulator use, baseline FEV1pp (50% to 75% and >75%), and number of exacerbations in the past year (1 or >1). The primary endpoints are absolute change from baseline in FEV1pp and safety outcomes. Secondary endpoints include rate of pulmonary exacerbations and time to first pulmonary exacerbation. Biomarkers of inflammation will also be assessed. EMPIRE-CF is expected to identify the optimal patient population, dose, duration and endpoints for future acebilustat trials, and widen understanding of the drug's efficacy in patients with CF.


Asunto(s)
Antiinflamatorios/uso terapéutico , Compuestos de Azabiciclo/uso terapéutico , Benzoatos/uso terapéutico , Fibrosis Quística/tratamiento farmacológico , Adolescente , Adulto , Proteína C-Reactiva/inmunología , Fibrosis Quística/inmunología , Fibrosis Quística/fisiopatología , Progresión de la Enfermedad , Método Doble Ciego , Epóxido Hidrolasas/antagonistas & inhibidores , Femenino , Volumen Espiratorio Forzado , Humanos , Leucotrieno B4/inmunología , Masculino , Adulto Joven
11.
J Med Chem ; 48(24): 7520-34, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16302794

RESUMEN

We have prepared a series of achiral aminoacetonitriles, bearing tri-ring benzamide moieties and an aminocyclohexanecarboxylate residue at P2. This combination of binding elements resulted in sub-250 pM, reversible, selective, and orally bioavailable cathepsin K inhibitors. Lead compounds displayed single digit nanomolar inhibition in vitro (of rabbit osteoclast-mediated degradation of bovine bone). The best compound in this series, 39n (CRA-013783/L-006235), was orally bioavailable in rats, with a terminal half-life of over 3 h. 39n was dosed orally in ovariectomized rhesus monkeys once per day for 7 days. Collagen breakdown products were reduced by up to 76% dose-dependently. Plasma concentrations of 39n above the bone resorption IC50 after 24 h indicated a correlation between functional cellular and in vivo assays. Inhibition of collagen breakdown by cathepsin K inhibitors suggests this mechanism of action may be useful in osteoporosis and other indications involving bone resorption.


Asunto(s)
Benzamidas/síntesis química , Conservadores de la Densidad Ósea/síntesis química , Catepsinas/antagonistas & inhibidores , Nitrilos/síntesis química , Tiazoles/síntesis química , Administración Oral , Animales , Benzamidas/química , Benzamidas/farmacología , Disponibilidad Biológica , Biomarcadores/orina , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/farmacología , Resorción Ósea/orina , Catepsina K , Catepsinas/química , Bovinos , Colágeno/antagonistas & inhibidores , Colágeno/metabolismo , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Cinética , Macaca mulatta , Modelos Moleculares , Estructura Molecular , Nitrilos/química , Nitrilos/farmacología , Conejos , Ratas , Relación Estructura-Actividad , Tiazoles/química , Tiazoles/farmacología
12.
J Med Chem ; 54(1): 179-200, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21126027

RESUMEN

The synthesis and optimization of a series of orally bioavailable 1-(1H-indol-4-yl)-3,5-disubstituted benzene analogues as antimitotic agents are described. A functionalized dibromobenzene intermediate was used as a key scaffold, which when modified by sequential Suzuki coupling and Buchwald-Hartwig amination provided a flexible entry to 1,3,5-trisubstituted phenyl compounds. A 1H-indol-4-yl moiety at the 1-position was determined to be a critical feature for optimal potency. The compounds have been shown to induce cell cycle arrest at the G2/M phase and demonstrate efficacy in both cell viability and cell proliferation assays. The primary site of action for these agents is revealed by their colchicine competitive inhibition of tubulin polymerization, and a computational model has been developed for the association of these compounds to tubulin. An optimized lead LP-261 significantly inhibits growth of a human non-small-cell lung tumor (NCI-H522) in a mouse xenograft model.


Asunto(s)
Indoles/síntesis química , Ácidos Isonicotínicos/síntesis química , Sulfonamidas/síntesis química , Moduladores de Tubulina/síntesis química , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colchicina/química , Ensayos de Selección de Medicamentos Antitumorales , Fase G2 , Humanos , Indoles/química , Indoles/farmacología , Ácidos Isonicotínicos/química , Ácidos Isonicotínicos/farmacología , Ratones , Ratones Desnudos , Modelos Moleculares , Trasplante de Neoplasias , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Sulfonamidas/química , Sulfonamidas/farmacología , Trasplante Heterólogo , Tubulina (Proteína)/química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
13.
Biochemistry ; 46(19): 5687-96, 2007 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-17441692

RESUMEN

In order to study the role of Phe169 in p38alpha MAP kinase structure and function, wild-type p38alpha and five p38alpha DFG motif mutants were examined in vitro for phosphorylation by MKK6, kinase activity toward ATF2 substrate, thermal stability, and X-ray crystal structure. All six p38alpha variants were efficiently phosphorylated by MKK6. However, only one activated p38alpha mutant (F169Y) possessed measurable kinase activity (1% compared to wild-type). The loss of kinase activity among the DFG mutants may result from an inability to correctly position Asp168 in the activated form of p38alpha. Two mutations significantly increased the thermal stability of p38alpha (F169A DeltaTm = 1.3 degrees C and D168G DeltaTm = 3.8 degrees C), and two mutations significantly decreased the stability of p38alpha (F169R DeltaTm = -3.2 degrees C and F169G DeltaTm = -4.7 degrees C). Interestingly, X-ray crystal structures of two thermally destabilized p38alpha-F169R and p38alpha-F169G mutants revealed a DFG-OUT conformation in the absence of an inhibitor molecule. This DFG-OUT conformation, termed alpha-DFG-OUT, is different from the ones previously identified in p38alpha crystal structures with bound inhibitors and postulated from high-temperature molecular dynamics simulations. Taken together, these results indicate that Phe169 is optimized for p38alpha functional activity and structural dynamics, rather than for structural stability. The alpha-DFG-OUT conformation observed for p38alpha-F169R and p38alpha-F169G may represent a naturally occurring intermediate state of p38alpha that provides access for binding of allosteric inhibitors. A model of the local forces driving the DFG IN-OUT transition in p38alpha is proposed.


Asunto(s)
Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/genética , Fenilalanina/fisiología , Regulación Alostérica , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Cristalización , Cristalografía por Rayos X , Escherichia coli/metabolismo , Calor , MAP Quinasa Quinasa 6/metabolismo , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Mutagénesis Sitio-Dirigida , Conformación Proteica , Desnaturalización Proteica
14.
Bioorg Med Chem Lett ; 15(23): 5274-9, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16169718

RESUMEN

Two new classes of diphenylether inhibitors of p38alpha MAP kinase are described. Both chemical classes are based on a common diphenylether core that is identified by simulated fragment annealing as one of the most favored chemotypes within a prominent hydrophobic pocket of the p38alpha ATP-binding site. In the fully elaborated molecules, the diphenylether moiety acts as an anchor occupying the deep pocket, while polar extensions make specific interactions with either the adenine binding site or the phosphate binding site of ATP. The synthesis, crystallographic analysis, and biological activity of these p38alpha inhibitors are discussed.


Asunto(s)
Compuestos de Bifenilo/química , Compuestos de Bifenilo/farmacología , Éteres/química , Éteres/farmacología , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Estructura Molecular , Unión Proteica , Inhibidores de Proteínas Quinasas/clasificación , Relación Estructura-Actividad
15.
J Biol Chem ; 280(49): 41077-89, 2005 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16199530

RESUMEN

Plasma kallikrein is a serine protease that has many important functions, including modulation of blood pressure, complement activation, and mediation and maintenance of inflammatory responses. Although plasma kallikrein has been purified for 40 years, its structure has not been elucidated. In this report, we described two systems (Pichia pastoris and baculovirus/Sf9 cells) for expression of the protease domain of plasma kallikrein, along with the purification and high resolution crystal structures of the two recombinant forms. In the Pichia pastoris system, the protease domain was expressed as a heterogeneously glycosylated zymogen that was activated by limited trypsin digestion and treated with endoglycosidase H deglycosidase to reduce heterogeneity from the glycosylation. The resulting protein was chromatographically resolved into four components, one of which was crystallized. In the baculovirus/Sf9 system, homogeneous, crystallizable, and nonglycosylated protein was expressed after mutagenizing three asparagines (the glycosylation sites) to glutamates. When assayed against the peptide substrates, pefachrome-PK and oxidized insulin B chain, both forms of the protease domain were found to have catalytic activity similar to that of the full-length protein. Crystallization and x-ray crystal structure determination of both forms have yielded the first three-dimensional views of the catalytic domain of plasma kallikrein. The structures, determined at 1.85 A for the endoglycosidase H-deglycosylated protease domain produced from P. pastoris and at 1.40 A for the mutagenically deglycosylated form produced from Sf9 cells, show that the protease domain adopts a typical chymotrypsin-like serine protease conformation. The structural information provides insights into the biochemical and enzymatic properties of plasma kallikrein and paves the way for structure-based design of protease inhibitors that are selective either for or against plasma kallikrein.


Asunto(s)
Expresión Génica , Calicreína Plasmática/química , Calicreína Plasmática/genética , Secuencia de Aminoácidos , Animales , Asparagina/genética , Baculoviridae/genética , Sitios de Unión/genética , Catálisis , Cristalización , Cristalografía por Rayos X , Electroforesis en Gel de Poliacrilamida , Vectores Genéticos , Ácido Glutámico , Glicósido Hidrolasas/metabolismo , Glicosilación , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Molecular , Mutagénesis , Pichia/genética , Calicreína Plasmática/metabolismo , Conformación Proteica , Proteínas Recombinantes/química , Inhibidores de Serina Proteinasa/farmacología , Spodoptera/metabolismo , Transfección , Tripsina/metabolismo
16.
Anal Biochem ; 325(1): 126-36, 2004 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-14715293

RESUMEN

Protein kinases are emerging as one of the most intensely studied classes of enzymes as their central roles in physiologically and clinically important cellular signaling events become more clearly understood. We report here the development of a real-time, label-free method to study protein kinase inhibitor binding kinetics using surface plasmon resonance-based biomolecular interaction analysis (Biacore). Utilizing p38alpha mitogen-activated protein kinase as a model system, we studied the binding properties of two known small molecule p38alpha inhibitors (SB-203580 and SKF-86002). Direct coupling of p38alpha to the biosensor surface in the presence of a reversible structure-stabilizing ligand (SB-203580) consistently produced greater than 90% active protein on the biosensor surface. The dissociation and kinetic constants derived using this Biacore method are in excellent agreement with values determined by other methods. Additionally, we extend the method to study the thermodynamics of small molecule binding to p38alpha and derive a detailed thermodynamic reaction pathway for SB-203580. The Biacore method reported here provides an efficient way to directly and reproducibly examine dissociation constants, kinetics, and thermodynamics for small molecules binding to p38alpha and possibly other protein kinases. Immobilization in the presence of a stabilizing ligand may further represent a broadly applicable paradigm for creation of highly active biosensor surfaces.


Asunto(s)
Imidazoles/análisis , Proteínas Quinasas Activadas por Mitógenos , Piridinas/análisis , Tiazoles/análisis , Animales , Escherichia coli , Imidazoles/metabolismo , Cinética , Ratones , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/química , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Estructura Molecular , Fosforilación , Unión Proteica , Desnaturalización Proteica , Piridinas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Resonancia por Plasmón de Superficie , Tiazoles/metabolismo
17.
Protein Expr Purif ; 37(1): 154-61, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15294293

RESUMEN

p38alpha mitogen-activated protein (MAP) kinase is widely expressed in many mammalian tissues and is activated as a part of signal transduction cascades that respond to inflammatory stimuli. The activation of p38 is known to trigger various biological effects, including cell death, differentiation, and proliferation. The central role played by p38alpha in cellular signaling events, including those that control a wide range of inflammatory and autoimmune diseases, makes it an attractive drug target. To develop optimized small molecule therapeutics targeting p38alpha, different techniques must be employed for the detailed biochemical, biophysical, and structural characterization of the interactions of p38alpha with lead compounds. These methods typically require large quantities of highly purified p38alpha protein. We describe here an improved expression and purification method for recombinant p38alpha production that reproducibly yields over 70 mg of highly purified protein per liter of shake flask bacterial culture. This yield is significantly higher than that previously reported for p38alpha production in Escherichia coli. We achieved a significant increase in soluble p38alpha protein expression by using the genetically modified E. coli strain BL21 DE3 Rosetta, which is optimized for expression of eukaryotic proteins with codons rarely used in E. coli. The p38alpha protein was purified to near homogeneity using a simple two-step procedure including nickel-chelating Sepharose chromatography followed by anion-exchange chromatography using MonoQ resin. Purified p38alpha was characterized using the standard commercially available small molecule inhibitor SB-203580. The binding association and dissociation rate constants determined by Biacore are in excellent agreement with previously reported values. The purified p38alpha protein was efficiently activated by MKK6 kinase to yield phosphorylated p38alpha. Purified p38alpha protein was also successfully crystallized, producing crystals diffracting to 1.9 angstroms, exceeding the highest resolution for p38alpha reported in the Protein DataBank. The simplicity and efficiency of this approach should prove useful for many laboratories that are interested in production of p38alpha for biochemical and biophysical studies and structure-based drug design.


Asunto(s)
Cromatografía/métodos , Escherichia coli/genética , Escherichia coli/metabolismo , Proteína Quinasa 14 Activada por Mitógenos , Animales , Cristalografía por Rayos X , Ratones , Proteína Quinasa 14 Activada por Mitógenos/química , Proteína Quinasa 14 Activada por Mitógenos/aislamiento & purificación , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Datos de Secuencia Molecular , Transducción de Señal/fisiología
18.
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA