Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Diabetes Obes Metab ; 23(8): 1795-1805, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33822469

RESUMEN

AIM: To gain further insights into the efficacy of SAR425899, a dual glucagon-like peptide-1/glucagon receptor agonist, by providing direct comparison with the glucagon-like peptide-1 receptor agonist, liraglutide, in terms of key outcomes of glucose metabolism. RESEARCH DESIGN AND METHODS: Seventy overweight to obese subjects with type 2 diabetes (T2D) were randomized to receive once-daily subcutaneous administrations of SAR425899 (0.12, 0.16 or 0.20 mg), liraglutide (1.80 mg) or placebo for 26 weeks. Mixed meal tolerance tests were conducted at baseline (BSL) and at the end of treatment (EOT). Metabolic indices of insulin action and secretion were assessed via Homeostasis Model Assessment (HOMA2) and oral minimal model (OMM) methods. RESULTS: From BSL to EOT (median [25th, 75th] percentile), HOMA2 quantified a significant improvement in basal insulin action in liraglutide (35% [21%, 74%]), while secretion enhanced both in SAR425899 (125% [63%, 228%]) and liraglutide (73% [43%, 147%]). OMM quantified, both in SAR425899 and liraglutide, a significant improvement in insulin sensitivity (203% [58%, 440%] and 36% [21%, 197%]), basal beta-cell responsiveness (67% [34%, 112%] and 40% [16%, 59%]), and above-basal beta-cell responsiveness (139% [64%, 261%] and 69% [-15%, 120%]). A significant delay in glucose absorption was highlighted in SAR425899 (37% [52%,18%]). CONCLUSIONS: SAR425899 and liraglutide improved postprandial glucose control in overweight to obese subjects with T2D. A significantly higher enhancement in beta-cell function was shown by SAR425899 than liraglutide.


Asunto(s)
Diabetes Mellitus Tipo 2 , Liraglutida , Glucemia , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón , Glucosa , Humanos , Hipoglucemiantes/uso terapéutico , Insulina , Liraglutida/uso terapéutico , Receptores de Glucagón
2.
Diabetes Technol Ther ; 22(8): 553-561, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32125178

RESUMEN

Background: Second-generation long-acting insulin glargine 300 U/mL (Gla-300) and degludec 100 U/mL (Deg-100) provide novel basal insulin therapies for the treatment of type 1 diabetes (T1D). Both offer a flatter pharmacokinetic (PK) profile than the previous generation of long-acting insulins, thus improving glycemic control while reducing hypoglycemic events. This work describes an in silico head-to-head comparison of the two basal insulins on 24-h glucose profiles and was used to guide the design of a clinical trial. Materials and Methods: The Universities of Virginia (UVA)/Padova T1D simulator describes the intra-/interday variability of glucose-insulin dynamics and thus provides a robust bench-test for assessing glucose control for basal insulin therapies. A PK model describing subcutaneous absorption of Deg-100, in addition to the one already available for Gla-300, has been developed based on T1D clinical data and incorporated into the simulator. One hundred in silico T1D subjects received a basal insulin dose (Gla-300 or Deg-100) for 12 weeks (8 weeks uptitration, 4 weeks stable dosing) by morning or evening administration in a basal/bolus regimen. The virtual patients were uptitrated to their individual doses with two different titration rules. Results: The last 2-week simulated continuous glucose monitoring data were used to calculate various outcome metrics for both basal insulin treatments, with primary outcome being the percent time in glucose target (70-140 mg/dL). The simulations show no statistically significant difference for Gla-300 versus Deg-100 in the main endpoints. Conclusions: This work suggests comparable glucose control using either Gla-300 or Deg-100 and was used to guide the design of a clinical trial intended to compare second-generation long-acting insulin analogues.


Asunto(s)
Diabetes Mellitus Tipo 1 , Hipoglucemiantes/uso terapéutico , Insulina Glargina/uso terapéutico , Insulina de Acción Prolongada/uso terapéutico , Glucemia , Automonitorización de la Glucosa Sanguínea , Simulación por Computador , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Humanos , Hipoglucemiantes/farmacocinética , Insulina Glargina/farmacocinética , Insulina de Acción Prolongada/farmacocinética
3.
IEEE Trans Biomed Eng ; 67(2): 624-631, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31150327

RESUMEN

OBJECTIVE: Subcutaneous (sc) administration of long-acting insulin analogs is often employed in multiple daily injection (MDI) therapy of type 1 diabetes (T1D) to cover patient's basal insulin needs. Among these, insulin glargine 100 U/mL (Gla-100) and 300 U/mL (Gla-300) are formulations indicated for once daily sc administration in MDI therapy of T1D. A few semi-mechanistic models of sc absorption of insulin glargine have been proposed in the literature, but were not quantitatively assessed on a large dataset. The aim of this paper is to propose a model of sc absorption of insulin glargine able to describe the data and provide precise model parameters estimates with a clear physiological interpretation. METHODS: Three candidate models were identified on a total of 47 and 77 insulin profiles of T1D subjects receiving a single or repeated sc administration of Gla-100 or Gla-300, respectively. Model comparison and selection were performed on the basis of their ability to describe the data and numerical identifiability. RESULTS: The most parsimonious model is linear two-compartment and accounts for the insulin distribution between the two compartments after sc administration through parameter k. Between the two formulations, we report a lower fraction of insulin in the first versus second compartment (k = 86% versus 94% in Gla-100 versus Gla-300, p < 0.05), a lower dissolution rate from the first to the second compartment ([Formula: see text] versus 0.0008 min-1 in Gla-100 versus Gla-300, p << 0.001), and a similar rate of insulin absorption from the second compartment to plasma ([Formula: see text] versus 0.0016 min-1 in Gla-100 versus Gla-300, p = NS), in accordance with the mechanisms of insulin glargine protraction. CONCLUSIONS: The proposed model is able to both accurately describe plasma insulin data after sc administration and precisely estimate physiologically plausible parameters. SIGNIFICANCE: The model can be incorporated in simulation platforms potentially usable for optimizing basal insulin treatment strategies.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Insulina Glargina/farmacocinética , Modelos Biológicos , Absorción Subcutánea/fisiología , Adulto , Simulación por Computador , Femenino , Humanos , Insulina/sangre , Insulina/metabolismo , Insulina Glargina/administración & dosificación , Insulina Glargina/uso terapéutico , Masculino , Persona de Mediana Edad , Ensayos Clínicos Controlados Aleatorios como Asunto
4.
Diabetes Obes Metab ; 22(4): 640-647, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31808298

RESUMEN

AIM: To evaluate the change in insulin sensitivity, ß-cell function and glucose absorption after 28 days of treatment with high and low doses of SAR425899, a novel dual glucagon-like peptide-1 receptor/glucagon receptor agonist, versus placebo. MATERIALS AND METHODS: Thirty-six overweight to obese subjects with type 2 diabetes were randomized to receive daily subcutaneous administrations of low-dose SAR425899 (0.03, 0.06 and 0.09 mg) and high-dose SAR425899 (0.06, 0.12 and 0.18 mg) or placebo for 28 days; dose escalation occurred after days 7 and 14. Mixed meal tolerance tests were conducted before treatment (day -1) and on days 1 and 28. Oral glucose and C-peptide minimal models were used to quantify metabolic indices of insulin sensitivity, ß-cell responsiveness and glucose absorption. RESULTS: With low-dose SAR425899, high-dose SAR425899 and placebo, ß-cell function from day -1 to day 28 increased by 163%, 95% and 23%, respectively. The change in area under the curve for the rate of meal glucose appearance between 0 and 120 minutes was -32%, -20% and 8%, respectively. CONCLUSIONS: After 28 days of treatment, SAR425899 improved postprandial glucose control by significantly enhancing ß-cell function and slowing glucose absorption rate.


Asunto(s)
Diabetes Mellitus Tipo 2 , Glucemia , Péptido C , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Receptor del Péptido 1 Similar al Glucagón , Humanos , Hipoglucemiantes/uso terapéutico , Insulina , Receptores de Glucagón
5.
IEEE Trans Biomed Eng ; 66(10): 2889-2896, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30735983

RESUMEN

OBJECTIVE: Glargine 100 U/mL (Gla-100) and 300 U/mL (Gla-300) are long-acting insulin analogs providing basal insulin supply in multiple daily injection (MDI) therapy of type 1 diabetes (T1D). Both insulins require extensive testing to arrive at the optimal dosing regimen, e.g., timing and amount. Here we aim at a simulation tool for evaluating benefits/risks of different dosing schemes and up-titration rules for both Gla-100 and Gla-300 before clinical testing. METHODS: A new pharmacokinetic (PK) model of both Gla-100 and Gla-300 was incorporated into the FDA-accepted University of Virginia/Padova T1D simulator: Specifically, a joint parameter distribution, built from PK parameter estimates, was used to generate individual PK parametrizations for each in silico subject. A virtual trial comparing Gla-100 vs. Gla-300 was performed and assessed against a clinical study to validate the glargine simulator. RESULTS: Like in vivo, in silico both insulins performed similarly with respect to glucose control: percent time of glucose between [80-140] mg/dL with Gla-100 vs. Gla-300 (primary endpoint) were 41.5 ± 1.1% vs. 39.0 ± 1.2% (P = 0.11) in silico, 31.0 ± 1.6% vs. 31.8 ± 1.5% (P = 0.73) in vivo. CONCLUSIONS: The glargine simulator reproduced the main findings of the clinical trial, proving its validity for testing MDI therapies. SIGNIFICANCE: In silico testing of MDI therapies can help designing clinical trials. Due to the more standardized settings in silico (e.g., standardized meals and strict adherence to titration rule), any potential treatment effect is reaching statistical significance in simulation vs. clinical trial.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacocinética , Insulina Glargina/administración & dosificación , Insulina Glargina/farmacocinética , Glucemia/análisis , Simulación por Computador , Esquema de Medicación , Humanos , Inyecciones , Insulina de Acción Prolongada
6.
Annu Int Conf IEEE Eng Med Biol Soc ; 2018: 4905-4908, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30441443

RESUMEN

The University of Virginia /Padova Type 1 Diabetes (TID) simulator has been widely used for testing artificial pancreas controllers, and, recently, novel insulin formulations and glucose sensors. However, a module describing the pharmacokinetics of the new long-acting insulin analogues is not available. The aim of this contribution is to reproduce multiple daily insulin injection (MDI) therapy, with insulin glargine 100 U/mL (Gla-100) as basal insulin, using the TID simulator. This was achieved by developing a model of Gla-100 and by incorporating it into the simulator. The methodology described here can be extended to other insulins, allowing an extensive in silico testing of different long-acting insulin analogues under various settings before starting human trials.


Asunto(s)
Diabetes Mellitus Tipo 1 , Páncreas Artificial , Glucemia , Humanos , Hipoglucemiantes , Insulina , Insulina Glargina , Insulina de Acción Prolongada
7.
Nat Commun ; 7: 13250, 2016 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-27841257

RESUMEN

Insulin-secreting beta cells play an essential role in maintaining physiological blood glucose levels, and their dysfunction leads to the development of diabetes. To elucidate the signalling events regulating insulin secretion, we applied a recently developed phosphoproteomics workflow. We quantified the time-resolved phosphoproteome of murine pancreatic cells following their exposure to glucose and in combination with small molecule compounds that promote insulin secretion. The quantitative phosphoproteome of 30,000 sites clustered into three main groups in concordance with the modulation of the three key kinases: PKA, PKC and CK2A. A high-resolution time course revealed key novel regulatory sites, revealing the importance of methyltransferase DNMT3A phosphorylation in the glucose response. Remarkably a significant proportion of these novel regulatory sites is significantly downregulated in diabetic islets. Control of insulin secretion is embedded in an unexpectedly broad and complex range of cellular functions, which are perturbed by drugs in multiple ways.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular Tumoral , Análisis por Conglomerados , Diabetes Mellitus Tipo 1/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fosfoproteínas/clasificación , Fosforilación/efectos de los fármacos , Proteómica/métodos
8.
Diabetes Technol Ther ; 18(9): 574-85, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27333446

RESUMEN

BACKGROUND: Technosphere(®) insulin (TI), an inhaled human insulin with a fast onset of action, provides a novel option for the control of prandial glucose. We used the University of Virginia (UVA)/Padova simulator to explore in-silico the potential benefit of different dosing regimens on postprandial glucose (PPG) control to support the design of further clinical trials. Tested dosing regimens included at-meal or postmeal dosing, or dosing before and after a meal (split dosing). METHODS: Various dosing regimens of TI were compared among one another and to insulin lispro in 100 virtual type-1 patients. Individual doses were identified for each regimen following different titration rules. The resulting postprandial glucose profiles were analyzed to quantify efficacy and the risk for hypoglycemic events. RESULTS: This approach allowed us to assess the benefit/risk for each TI dosing regimen and to compare results with simulations of insulin lispro. We identified a new titration rule for TI that could significantly improve the efficacy of treatment with TI. CONCLUSION: In-silico clinical trials comparing the treatment effect of different dosing regimens with TI and of insulin lispro suggest that postmeal dosing or split dosing of TI, in combination with an appropriate titration rule, can achieve a superior postprandial glucose control while providing a lower risk for hypoglycemic events than conventional treatment with subcutaneously administered rapid-acting insulin products.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/administración & dosificación , Insulina/administración & dosificación , Administración por Inhalación , Simulación por Computador , Esquema de Medicación , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Periodo Posprandial
9.
Bioinformatics ; 32(3): 424-31, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26628587

RESUMEN

MOTIVATION: Phosphoproteomics measurements are widely applied in cellular biology to detect changes in signalling dynamics. However, due to the inherent complexity of phosphorylation patterns and the lack of knowledge on how phosphorylations are related to functions, it is often not possible to directly deduce protein activities from those measurements. Here, we present a heuristic machine learning algorithm that infers the activities of kinases from Phosphoproteomics data using kinase-target information from the PhosphoSitePlus database. By comparing the estimated kinase activity profiles to the measured phosphosite profiles, it is furthermore possible to derive the kinases that are most likely to phosphorylate the respective phosphosite. RESULTS: We apply our approach to published datasets of the human cell cycle generated from HeLaS3 cells, and insulin signalling dynamics in mouse hepatocytes. In the first case, we estimate the activities of 118 at six cell cycle stages and derive 94 new kinase-phosphosite links that can be validated through either database or motif information. In the second case, the activities of 143 kinases at eight time points are estimated and 49 new kinase-target links are derived. AVAILABILITY AND IMPLEMENTATION: The algorithm is implemented in Matlab and be downloaded from github. It makes use of the Optimization and Statistics toolboxes. https://github.com/marcel-mischnik/IKAP.git. CONTACT: marcel.mischnik@gmail.com SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Algoritmos , Hepatocitos/metabolismo , Heurística , Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Proteómica/métodos , Programas Informáticos , Animales , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Bases de Datos Factuales , Células HeLa , Hepatocitos/citología , Humanos , Insulina/metabolismo , Ratones , Fosforilación
10.
Artículo en Inglés | MEDLINE | ID: mdl-26736985

RESUMEN

The University of Virginia/Padova Type 1 Diabetes (T1DM) Simulator has been extensively used in artificial pancreas research mostly for testing and design of control algorithms. However, it also offers the possibility of testing new insulin analogs and alternative routes of delivery given that subcutaneous insulin administration present significant delays & variability. Inhaled insulin appears an important candidate to improve post-prandial glucose control given its rapid appearance in plasma. In this contribution, we present the results of incorporating a pharmacokinetic model of inhaled Technosphere(®) Insulin (TI) into the T1DM simulator. In particular, we successfully reproduced in silico the post-prandial glucose control observed in T1DM subjects treated with TI given at meal time, and the post-prandial glucose dynamics in response to different timing of TI dose.


Asunto(s)
Simulación por Computador , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Insulina/administración & dosificación , Insulina/uso terapéutico , United States Food and Drug Administration , Universidades , Administración por Inhalación , Algoritmos , Glucemia/análisis , Diabetes Mellitus Tipo 1/sangre , Humanos , Insulina/farmacocinética , Comidas , Modelos Biológicos , Estados Unidos
11.
Am J Physiol Endocrinol Metab ; 306(6): E627-34, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24425760

RESUMEN

The experimental protocol of the perfused rat pancreas is commonly used to evaluate ß-cell function. In this context, mathematical models become useful tools through the determination of indexes that allow the assessment of ß-cell function in different experimental groups and the quantification of the effects of antidiabetic drugs, secretagogues, or treatments. However, a minimal model applicable to the isolated perfused rat pancreas has so far been unavailable. In this work, we adapt the C-peptide minimal model applied previously to the intravenous glucose tolerance test to obtain a specific model for the experimental settings of the perfused pancreas. Using the model, it is possible to estimate indexes describing ß-cell responsivity for first (ΦD) and second phase (ΦS, T) of insulin secretion. The model was initially applied to untreated pancreata and afterward used for the assessment of pharmacologically relevant agents (the gut hormone GLP-1, the potent GLP-1 receptor agonist lixisenatide, and a GPR40/FFAR1 agonist, SAR1) to quantify and differentiate their effect on insulin secretion. Model fit was satisfactory, and parameters were estimated with good precision for both untreated and treated pancreata. Model application showed that lixisenatide reaches improvement of ß-cell function similarly to GLP-1 (11.7- vs. 13.1-fold increase in ΦD and 2.3- vs. 2.8-fold increase in ΦS) and demonstrated that SAR1 leads to an additional improvement of ß-cell function in the presence of postprandial GLP-1 levels.


Asunto(s)
Péptido 1 Similar al Glucagón/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Modelos Biológicos , Receptores de Glucagón/metabolismo , Transducción de Señal , Algoritmos , Animales , Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Hipoglucemiantes/agonistas , Hipoglucemiantes/metabolismo , Hipoglucemiantes/farmacología , Técnicas In Vitro , Secreción de Insulina , Células Secretoras de Insulina/efectos de los fármacos , Cinética , Masculino , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas de Unión al GTP Monoméricas/farmacología , Páncreas/efectos de los fármacos , Páncreas/metabolismo , Péptidos/farmacología , Perfusión , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glucagón/agonistas , Reproducibilidad de los Resultados , Transducción de Señal/efectos de los fármacos
12.
Bioorg Med Chem Lett ; 23(5): 1168-76, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23337596

RESUMEN

Systems biology aims to provide a holistic and in many cases dynamic picture of biological function and malfunction, in case of disease. Technology developments in the generation of genome-wide datasets and massive improvements in computer power now allow to obtain new insights into complex biological networks and to copy nature by computing these interactions and their kinetics and by generating in silico models of cells, tissues and organs. The expectations are high that systems biology will pave the way to the identification of novel disease genes, to the selection of successful drug candidates--that do not fail in clinical studies due to toxicity or lack of human efficacy--and finally to a more successful discovery of novel therapeutics. However, further research is necessary to fully unleash the potential of systems biology. Within this review we aim to highlight the most important and promising top-down and bottom-up systems biology applications in drug discovery.


Asunto(s)
Enfermedad , Descubrimiento de Drogas , Biología de Sistemas , Animales , Humanos
13.
Bioinformatics ; 29(1): 132-4, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23129297

RESUMEN

SUMMARY: Drug versus Disease (DvD) provides a pipeline, available through R or Cytoscape, for the comparison of drug and disease gene expression profiles from public microarray repositories. Negatively correlated profiles can be used to generate hypotheses of drug-repurposing, whereas positively correlated profiles may be used to infer side effects of drugs. DvD allows users to compare drug and disease signatures with dynamic access to databases Array Express, Gene Expression Omnibus and data from the Connectivity Map. AVAILABILITY AND IMPLEMENTATION: R package (submitted to Bioconductor) under GPL 3 and Cytoscape plug-in freely available for download at www.ebi.ac.uk/saezrodriguez/DVD/.


Asunto(s)
Reposicionamiento de Medicamentos , Programas Informáticos , Transcriptoma/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
14.
J Chem Inf Model ; 52(8): 2022-30, 2012 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-22794356

RESUMEN

The three-dimensional (3D) superimposition of molecules of one biological target reflecting their relative bioactive orientation is key for several ligand-based drug design studies (e.g., QSAR studies, pharmacophore modeling). However, with the lack of sufficient ligand-protein complex structures, an experimental alignment is difficult or often impossible to obtain. Several computational 3D alignment tools have been developed by academic or commercial groups to address this challenge. Here, we present a new approach, MARS (Multiple Alignments by ROCS-based Similarity), that is based on the pairwise alignment of all molecules within the data set using the tool ROCS (Rapid Overlay of Chemical Structures). Each pairwise alignment is scored, and the results are captured in a score matrix. The ideal superimposition of the compounds in the set is then identified by the analysis of the score matrix building stepwise a superimposition of all molecules. The algorithm exploits similarities among all molecules in the data set to compute an optimal 3D alignment. This alignment tool presented here can be used for several applications, including pharmacophore model generation, 3D QSAR modeling, 3D clustering, identification of structural outliers, and addition of compounds to an already existing alignment. Case studies are shown, validating the 3D alignments for six different data sets.


Asunto(s)
Algoritmos , Diseño de Fármacos , Modelos Moleculares , Proteínas/metabolismo , Cristalografía por Rayos X , Ligandos , Conformación Molecular , Unión Proteica , Proteínas/antagonistas & inhibidores , Proteínas/química
16.
J Med Chem ; 52(9): 2923-32, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19374402

RESUMEN

G-protein-coupled receptors (GPCRs) comprise a large protein family of significant past and current interest of pharmaceutical research. X-ray crystallography and molecular modeling combined with site-directed mutagenesis studies suggest that most family A GPCRs share a small-molecule binding site located in the outer part of the seven-transmembrane (7TM) bundle. Here we describe an automated method to derive sequence-derived three-dimensional (3D) pharmacophore models capturing the key elements for addressing this binding site by a small-molecule ligand. We have generated structure-based pharmacophore models from 10 homology models and 3 X-ray structures of receptor-ligand complexes. These 13 pharmacophores have been dissected into 35 different single-feature pharmacophore elements, each associated with a sequence motif or chemoprint, describing its molecular interaction partner(s) in the receptor. Subsequently, the protein sequences of 270 GPCRs have been searched for the presence of chemoprints and the appropriate single-feature pharmacophores have been assembled into three- to seven-feature 3D-pharmacophore models for each human family A GPCR. These models can be applied for virtual screening and for the design of subfamily directed libraries. A case study demonstrates the successful application of this approach for the identification of potent agonists for the complement component 3a receptor 1 (C3AR1) by virtual screening.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Modelos Moleculares , Receptores Acoplados a Proteínas G/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Descubrimiento de Drogas , Humanos , Ligandos , Datos de Secuencia Molecular , Conformación Proteica , Receptores de Complemento/agonistas , Receptores de Complemento/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Reproducibilidad de los Resultados
17.
Biochemistry ; 47(16): 4683-91, 2008 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-18373353

RESUMEN

Glycogen phosphorylase (GP) is a validated target for the treatment of type 2 diabetes. Here we describe highly potent GP inhibitors, AVE5688, AVE2865, and AVE9423. The first two compounds are optimized members of the acyl urea series. The latter represents a novel quinolone class of GP inhibitors, which is introduced in this study. In the enzyme assay, both inhibitor types compete with the physiological activator AMP and act synergistically with glucose. Isothermal titration calorimetry (ITC) shows that the compounds strongly bind to nonphosphorylated, inactive GP (GPb). Binding to phosphorylated, active GP (GPa) is substantially weaker, and the thermodynamic profile reflects a coupled transition to the inactive (tense) conformation. Crystal structures confirm that the three inhibitors bind to the AMP site of tense state GP. These data provide the first direct evidence that acyl urea and quinolone compounds are allosteric inhibitors that selectively bind to and stabilize the inactive conformation of the enzyme. Furthermore, ITC reveals markedly different thermodynamic contributions to inhibitor potency that can be related to the binding modes observed in the cocrystal structures. For AVE5688, which occupies only the lower part of the bifurcated AMP site, binding to GPb (Kd = 170 nM) is exclusively enthalpic (Delta H = -9.0 kcal/mol, TDelta S = 0.3 kcal/mol). The inhibitors AVE2865 (Kd = 9 nM, Delta H = -6.8 kcal/mol, TDelta S = 4.2 kcal/mol) and AVE9423 (Kd = 24 nM, Delta H = -5.9 kcal/mol, TDelta S = 4.6 kcal/mol) fully exploit the volume of the binding pocket. Their pronounced binding entropy can be attributed to the extensive displacement of solvent molecules as well as to ionic interactions with the phosphate recognition site.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Glucógeno Fosforilasa/antagonistas & inhibidores , Glucógeno Fosforilasa/metabolismo , Termodinámica , Regulación Alostérica/efectos de los fármacos , Animales , Tampones (Química) , Calorimetría , Glucosa/metabolismo , Glucógeno Fosforilasa/química , Cinética , Modelos Moleculares , Estructura Molecular , Unión Proteica , Conejos , Temperatura , Volumetría
18.
J Med Chem ; 48(20): 6178-93, 2005 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-16190745

RESUMEN

Using a focused screening approach, acyl ureas have been discovered as a new class of inhibitors of human liver glycogen phosphorylase (hlGPa). The X-ray structure of screening hit 1 (IC50 = 2 microM) in a complex with rabbit muscle glycogen phosphorylase b reveals that 1 binds at the AMP site, the main allosteric effector site of the dimeric enzyme. A first cycle of chemical optimization supported by X-ray structural data yielded derivative 21, which inhibited hlGPa with an IC50 of 23 +/- 1 nM, but showed only moderate cellular activity in isolated rat hepatocytes (IC50 = 6.2 microM). Further optimization was guided by (i) a 3D pharmacophore model that was derived from a training set of 24 compounds and revealed the key chemical features for the biological activity and (ii) the 1.9 angstroms crystal structure of 21 in complex with hlGPa. A second set of compounds was synthesized and led to 42 with improved cellular activity (hlGPa IC50 = 53 +/- 1 nM; hepatocyte IC50 = 380 nM). Administration of 42 to anaesthetized Wistar rats caused a significant reduction of the glucagon-induced hyperglycemic peak. These findings are consistent with the inhibition of hepatic glycogenolysis and support the use of acyl ureas for the treatment of type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Glucógeno Fosforilasa de Forma Hepática/antagonistas & inhibidores , Urea/análogos & derivados , Urea/síntesis química , Adenosina Monofosfato/química , Sitio Alostérico , Animales , Sitios de Unión , Cristalografía por Rayos X , Glucógeno Fosforilasa de Forma Hepática/química , Glucógeno Fosforilasa de Forma Muscular/química , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Humanos , Técnicas In Vitro , Modelos Moleculares , Relación Estructura-Actividad Cuantitativa , Conejos , Ratas , Urea/química
19.
J Med Chem ; 48(17): 5448-65, 2005 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-16107144

RESUMEN

In this paper, we compare protein- and ligand-based virtual screening techniques for identifying the ligands of four biogenic amine-binding G-protein coupled receptors (GPCRs). For the screening of the virtual compound libraries, we used (1) molecular docking into GPCR homology models, (2) ligand-based pharmacophore and Feature Tree models, (3) three-dimensional (3D)-similarity searches, and (4) statistical methods [partial least squares (PLS) and partial least squares discriminant analysis (PLS-DA) models] based on two-dimensional (2D) molecular descriptors. The comparison of the different methods in retrieving known antagonists from virtual libraries shows that in our study the ligand-based pharmacophore-, Feature Tree-, and 2D quantitative structure-activity relationship (QSAR)-based screening techniques provide enrichment factors that are higher than those provided by molecular docking into the GPCR homology models. Nevertheless, the hit rates achieved when docking with GOLD and ranking the ligands with GoldScore (up to 60% among the top-ranked 1% of the screened databases) are still satisfying. These results suggest that docking into GPCR homology models can be a useful approach for lead finding by virtual screening when either little or no information about the active ligands is available.


Asunto(s)
Aminas Biogénicas/química , Ligandos , Relación Estructura-Actividad Cuantitativa , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/química , Antagonistas de Receptores Adrenérgicos alfa 1 , Sitios de Unión , Bases de Datos Factuales , Antagonistas de los Receptores de Dopamina D2 , Análisis de los Mínimos Cuadrados , Modelos Moleculares , Receptor Muscarínico M1/antagonistas & inhibidores , Receptor Muscarínico M1/química , Receptor de Serotonina 5-HT2A/química , Receptores Adrenérgicos alfa 1/química , Receptores de Dopamina D2/química , Antagonistas del Receptor de Serotonina 5-HT2
20.
Protein Sci ; 14(7): 1760-71, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15987904

RESUMEN

Acyl ureas were discovered as a novel class of inhibitors for glycogen phosphorylase, a molecular target to control hyperglycemia in type 2 diabetics. This series is exemplified by 6-{2,6-Dichloro- 4-[3-(2-chloro-benzoyl)-ureido]-phenoxy}-hexanoic acid, which inhibits human liver glycogen phosphorylase a with an IC(50) of 2.0 microM. Here we analyze four crystal structures of acyl urea derivatives in complex with rabbit muscle glycogen phosphorylase b to elucidate the mechanism of inhibition of these inhibitors. The structures were determined and refined to 2.26 Angstroms resolution and demonstrate that the inhibitors bind at the allosteric activator site, where the physiological activator AMP binds. Acyl ureas induce conformational changes in the vicinity of the allosteric site. Our findings suggest that acyl ureas inhibit glycogen phosphorylase by direct inhibition of AMP binding and by indirect inhibition of substrate binding through stabilization of the T' state.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Glucógeno Fosforilasa de Forma Muscular/antagonistas & inhibidores , Músculos/enzimología , Conformación Proteica/efectos de los fármacos , Urea/metabolismo , Adenosina Monofosfato/metabolismo , Sitio Alostérico , Animales , Sitios de Unión , Cristalografía por Rayos X , Inhibidores Enzimáticos/farmacología , Estabilidad de Enzimas , Glucógeno Fosforilasa de Forma Hepática/antagonistas & inhibidores , Glucógeno Fosforilasa de Forma Hepática/química , Glucógeno Fosforilasa de Forma Hepática/metabolismo , Glucógeno Fosforilasa de Forma Muscular/química , Glucógeno Fosforilasa de Forma Muscular/metabolismo , Humanos , Hipoglucemiantes , Cinética , Modelos Moleculares , Estructura Molecular , Unión Proteica , Conejos , Urea/análogos & derivados , Urea/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA