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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nucl Med Biol ; 33(4): 555-63, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16720249

RESUMEN

We have characterized the interaction of the serotonin transporter ligand [3H]-N,N-dimethyl-2-(2-amino-4-cyanophenylthio)-benzylamine (DASB) with rhesus monkey brain in vitro using tissue homogenate binding and autoradiographic mapping. [3H]-DASB, a tritiated version of the widely used [11C] positron emission tomography tracer, was found to selectively bind to a single population of sites with high affinity (K(d)=0.20+/-0.04 nM). The serotonin transporter density (B(max)) obtained for rhesus frontal cortex was found to be 66+/-8 fmol/mg protein using [3H]-DASB, similar to the B(max) value obtained using the reference radioligand [3H]-citalopram, a well-characterized and highly selective serotonin reuptake inhibitor (83+/-22 fmol/mg protein). Specific binding sites of both [3H]-DASB and [3H]-citalopram were similarly and nonuniformly distributed throughout the rhesus central nervous system, in a pattern consistent with serotonin transporter localization reported for human brain. Regional serotonin transporter densities, estimated from optical densities of the autoradiographic images, were well correlated between the two radioligands. Finally, DASB and fluoxetine showed dose-dependent full inhibition of [3H]-citalopram binding in a competition autoradiographic study, with K(i) values in close agreement with those obtained from rhesus brain homogenates. This side-by-side comparison of [3H]-DASB and [3H]-citalopram binding sites in rhesus tissue homogenates and in adjacent rhesus brain slices provides additional support for the use of [11C]-DASB to assess the availability and distribution of serotonin transporters in nonhuman primates.


Asunto(s)
Bencilaminas/farmacocinética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Citalopram/farmacocinética , Proteínas de Transporte de Serotonina en la Membrana Plasmática/metabolismo , Animales , Autorradiografía , Sitios de Unión , Técnicas In Vitro , Macaca mulatta , Tasa de Depuración Metabólica , Unión Proteica , Cintigrafía , Radiofármacos/farmacocinética , Distribución Tisular , Tritio/farmacocinética
2.
J Comp Neurol ; 524(1): 90-118, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26105175

RESUMEN

Functional imaging studies have revealed that certain brainstem areas are activated during migraine attacks. The neuropeptide calcitonin gene-related peptide (CGRP) is associated with activation of the trigeminovascular system and transmission of nociceptive information and plays a key role in migraine pathophysiology. Therefore, to elucidate the role of CGRP, it is critical to identify the regions within the brainstem that process CGRP signaling. In situ hybridization and immunofluorescence were performed to detect mRNA expression and define cellular localization of calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1), respectively. To define CGRP receptor binding sites, in vitro autoradiography was performed with [(3)H]MK-3207 (a CGRP receptor antagonist). CLR and RAMP1 mRNA and protein expression were detected in the pineal gland, medial mammillary nucleus, median eminence, infundibular stem, periaqueductal gray, area postrema, pontine raphe nucleus, gracile nucleus, spinal trigeminal nucleus, and spinal cord. RAMP1 mRNA expression was also detected in the posterior hypothalamic area, trochlear nucleus, dorsal raphe nucleus, medial lemniscus, pontine nuclei, vagus nerve, inferior olive, abducens nucleus, and motor trigeminal nucleus; protein coexpression of CLR and RAMP1 was observed in these areas via immunofluorescence. [(3)H]MK-3207 showed high binding densities concordant with mRNA and protein expression. The present study suggests that several regions in the brainstem may be involved in CGRP signaling. Interestingly, we found receptor expression and antagonist binding in some areas that are not protected by the blood-brain barrier, which suggests that drugs inhibiting CGRP signaling may not be able to penetrate the central nervous system to antagonize receptors in these brain regions.


Asunto(s)
Tronco Encefálico/metabolismo , Proteína Similar al Receptor de Calcitonina/metabolismo , Macaca mulatta/metabolismo , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Animales , Autorradiografía , Tronco Encefálico/anatomía & histología , Compuestos Bicíclicos Heterocíclicos con Puentes , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina , Femenino , Técnica del Anticuerpo Fluorescente , Hibridación in Situ , Macaca mulatta/anatomía & histología , Masculino , Unión Proteica , ARN Mensajero/metabolismo , Radiofármacos , Transducción de Señal , Médula Espinal/anatomía & histología , Médula Espinal/metabolismo , Compuestos de Espiro , Tritio
3.
Eur J Pharmacol ; 527(1-3): 44-51, 2005 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-16310181

RESUMEN

This study used behavioural and in vivo electrophysiological paradigms to examine the effects of systemic and spinal administration of a bradykinin B1 receptor antagonist, compound X, on acute nociceptive responses in the rat. In behavioural experiments, compound X significantly increased the latency to withdraw the hindpaw from a radiant heat source after both intravenous and intrathecal administration, without affecting motor performance on the rotarod. In electrophysiological experiments, both intravenous and direct spinal administration of compound X attenuated the responses of single dorsal horn neurones to noxious thermal stimulation of the hindpaw. These data show that the antinociceptive effects of a bradykinin B1 receptor antagonist are mediated, at least in part, at the level of the spinal cord and suggest a role for spinal bradykinin B1 receptors in acute nociception.


Asunto(s)
Amidas/farmacocinética , Antagonistas del Receptor de Bradiquinina B1 , Naftalenos/farmacocinética , Dimensión del Dolor/métodos , Pirrolidinas/farmacocinética , Médula Espinal/efectos de los fármacos , Amidas/administración & dosificación , Animales , Carragenina/administración & dosificación , Carragenina/toxicidad , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Electrofisiología/métodos , Pie , Miembro Posterior , Calor/efectos adversos , Hiperalgesia/etiología , Hiperalgesia/fisiopatología , Hiperalgesia/prevención & control , Hipersensibilidad/etiología , Hipersensibilidad/fisiopatología , Inyecciones Intravenosas , Inyecciones Espinales , Masculino , Morfina/farmacología , Naftalenos/administración & dosificación , Nociceptores/efectos de los fármacos , Nociceptores/fisiología , Desempeño Psicomotor/efectos de los fármacos , Pirrolidinas/administración & dosificación , Ratas , Ratas Sprague-Dawley , Médula Espinal/fisiología
4.
Brain Res ; 1600: 93-109, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25463029

RESUMEN

Calcitonin gene-related peptide (CGRP) receptor antagonists have demonstrated anti-migraine efficacy. One remaining question is where do these blockers act? We hypothesized that the trigeminal ganglion could be one possible site. We examined the binding sites of a CGRP receptor antagonist (MK-3207) and related this to the expression of CGRP and its receptor in rhesus trigeminal ganglion. Pituitary adenylate cyclase-activating polypeptide (PACAP) and glutamate were examined and related to the CGRP system. Furthermore, we examined if the trigeminal ganglion is protected by the blood-brain barrier (BBB). Autoradiography was performed with [(3)H]MK-3207 to demonstrate receptor binding sites in rhesus trigeminal ganglion (TG). Immunofluorescence was used to correlate binding and the presence of CGRP and its receptor components, calcitonin receptor-like receptor (CLR) and receptor activity-modifying protein 1 (RAMP1), and the distribution of PACAP and glutamate in rhesus and rat TG. Evans blue was used to examine large molecule penetration into the rat TG. High receptor binding densities were found in rhesus TG. Immunofluorescence revealed expression of CGRP, CLR and RAMP1 in trigeminal cells. CGRP positive neurons expressed PACAP but not glutamate. Some neurons expressing CLR and RAMP1 co-localized with glutamate. Evans blue revealed that the TG is not protected by BBB. This study demonstrates CGRP receptor binding sites and expression of the CGRP receptor in rhesus and rat TG. The expression pattern of PACAP and glutamate suggests a possible interaction between the glutamatergic and CGRP system. In rat the TG is outside the BBB, suggesting that molecules do not need to be CNS-penetrant to block these receptors.


Asunto(s)
Péptido Relacionado con Gen de Calcitonina/análisis , Ácido Glutámico/análisis , Neuronas/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/análisis , Receptores de Péptido Relacionado con el Gen de Calcitonina/análisis , Ganglio del Trigémino/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Antagonistas del Receptor Peptídico Relacionado con el Gen de la Calcitonina , Proteína Similar al Receptor de Calcitonina/análisis , Femenino , Macaca mulatta , Masculino , Cintigrafía , Ratas , Ratas Sprague-Dawley , Proteína 1 Modificadora de la Actividad de Receptores/análisis , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Compuestos de Espiro/farmacología , Ganglio del Trigémino/diagnóstico por imagen
5.
J Med Chem ; 46(10): 1803-6, 2003 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-12723943

RESUMEN

Antagonism of the bradykinin B(1) receptor was demonstrated to be a potential treatment for chronic pain and inflammation. Novel benzodiazepines were designed that display subnanomolar affinity for the bradykinin B(1) receptor (K(i) = 0.59 nM) and high selectivity against the bradykinin B(2) receptor (K(i) > 10 microM). In vivo efficacy, comparable to morphine, was demonstrated for lead compounds in a rodent hyperalgesia model.


Asunto(s)
Benzodiazepinas/síntesis química , Antagonistas de los Receptores de Bradiquinina , Animales , Benzodiazepinas/química , Benzodiazepinas/farmacología , Células CHO , Cricetinae , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptor de Bradiquinina B1 , Receptor de Bradiquinina B2 , Relación Estructura-Actividad
6.
Int Immunopharmacol ; 2(13-14): 1747-54, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12489788

RESUMEN

The pharmacological properties of the kinin B1 receptor in binding the endogenous kinin peptides are known to differ across species. Molecular cloning has revealed that these pharmacological differences arise from the diversity within the BDKRB gene. In this report, the molecular diversity of the human BDKRB1 gene is expanded by the identification of eight single nucleotide polymorphisms (SNPs) in the coding sequence of the receptor, three of which change the amino acid sequence of the receptor. The molecular cloning and pharmacological characterization of two primate B1 receptors, rhesus and African Green monkey, reveals that they exhibit the same high degree of selectivity for des-Arg10 kallidin (Lys-bradykinin) relative to des-Arg9 bradykinin that is observed with the human kinin B1 receptor. Previous mutagenesis studies of the human B1 receptor have implicated extracellular domain (EC) IV in conferring this selectivity for des-Arg10 kallidin, by interacting with the N-terminal Lys residue of the peptide. The pharmacological analysis of chimeric B1 receptors, in which EC-IV of the human B1 receptor is replaced with the corresponding domain of either rat or dog, supports the proposal that EC-IV is an important determinant in conferring ligand selectivity.


Asunto(s)
Bradiquinina/análogos & derivados , Polimorfismo de Nucleótido Simple , Receptores de Bradiquinina/genética , Receptores de Bradiquinina/metabolismo , Secuencia de Aminoácidos , Animales , Unión Competitiva , Bradiquinina/metabolismo , Bradiquinina/farmacología , Chlorocebus aethiops , Clonación Molecular , Perros , Frecuencia de los Genes , Humanos , Calidina/análogos & derivados , Calidina/metabolismo , Calidina/farmacología , Macaca mulatta , Datos de Secuencia Molecular , Ratas , Receptor de Bradiquinina B1 , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Especificidad de la Especie
7.
PLoS One ; 9(9): e106693, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25198535

RESUMEN

BACKGROUND: Type 2 diabetes results from failure of the ß-cells to compensate for increased insulin demand due to abnormal levels of metabolic factors. The ob/ob(lep-/-) mouse has been extensively studied as an animal model of type 2 diabetes. Previous studies have shown a correlation between ß-cell function and bioluminescent imaging in lean genetically engineered mice. The ability to noninvasively monitor ß-cell function in ob/ob mice could provide new information on ß-cell regulation in type 2 diabetes. METHODS: To create the B6 Albino ob/ob MIP-luc mice (ob/ob-luc), the ob/ob mouse was crossed with the CD1 MIP-luc mouse. All mice were backcrossed over multiple generations to ensure the genetic background of the transgenic mice was over 96% similar to the background of the original ob/ob mouse. Animal weight, blood glucose levels, insulin in plasma, and in vivo bioluminescence (BLI) were monitored weekly or biweekly for up to 70 weeks of age. BL imaging was performed using IVIS Spectrum (Perkin Elmer) and calculated by integrating the bioluminescence signal between 5 and 10 min after i.v. injection of D-luciferin. Insulin immunohistochemistry determined islet beta cell count and insulin secretion assay determined islet insulin function. RESULTS: There were significant increases in BLI and insulin levels as the ob/ob-luc mice aged while glucose levels gradually decreased. Ob/ob-luc were sacrificed at different time points to determine ex vivo BLI, islet function and total ß-cell numbers using a cell counting training algorithm developed for the Vectra image analysis system (Perkin Elmer). The number of ß-cells increased as the mice aged and all three ex vivo measurements correlated with BLI. CONCLUSIONS: The ob/ob-luc mice can serve as a model of metabolic stress, similar to human type 2 diabetes using BLI as a surrogate marker for ß-cell function.


Asunto(s)
Hiperglucemia/fisiopatología , Islotes Pancreáticos/fisiopatología , Obesidad/fisiopatología , Animales , Diabetes Mellitus Tipo 2/fisiopatología , Luminiscencia , Ratones
8.
Mol Imaging Biol ; 15(4): 431-40, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23440602

RESUMEN

PURPOSE: Visualization of the cell cycle in living subjects has long been a big challenge. The present study aimed to noninvasively visualize mitotic arrest of the cell cycle with an optical reporter in living subjects. PROCEDURES: An N-terminal cyclin B1-luciferase fusion construct (cyclin B-Luc) controlled by the cyclin B promoter, as a mitosis reporter, was generated. HeLa or HCT116 cells stably expressing cyclin B-Luc reporter were used to evaluate its cell cycle-dependent regulation and ubiquitination-mediated degradation. We also evaluated its feasibility to monitor the mitotic arrest caused by Taxotere both in vitro and in vivo. RESULTS: We showed that the cyclin B-Luc fusion protein was regulated in a cell cycle-dependent manner and accumulated in the mitotic phase (M phase) in cellular assays. The regulation of cyclin B-Luc reporter was mediated by proteasome ubiquitination. In the present study, in vitro imaging showed that antimitotic reagents like Taxotere upregulated the reporter through cell cycle arrest in the M phase. Noninvasive longitudinal bioluminescence imaging further demonstrated an upregulation of the reporter consistent with mitotic arrest induced in tumor xenograft models. Induction of this reporter was also observed with a kinesin spindle protein inhibitor, which causes cell cycle blockage in the M phase. CONCLUSIONS: Our results demonstrate that the cyclin B-Luc reporter can be used to image whether compounds are capable, in vivo, of causing an M phase arrest and/or altering cyclin B turnover. This reporter can also be potentially used in high-throughput screening efforts aimed at discovering novel molecules that will cause cell cycle arrest at the M phase in cultivated cell lines and animal models.


Asunto(s)
Puntos de Control del Ciclo Celular , Luminiscencia , Mitosis , Imagen Molecular/métodos , Animales , Antineoplásicos/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Ciclina B1/metabolismo , Docetaxel , Células HCT116 , Células HeLa , Histonas/metabolismo , Humanos , Luciferasas/metabolismo , Ratones , Mitosis/efectos de los fármacos , Nocodazol/farmacología , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tejido Subcutáneo/efectos de los fármacos , Taxoides/farmacología , Ubiquitinación/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
ACS Med Chem Lett ; 4(6): 509-13, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-24900701

RESUMEN

We report herein the discovery of a fatty acid amide hydrolase (FAAH) positron emission tomography (PET) tracer. Starting from a pyrazole lead, medicinal chemistry efforts directed toward reducing lipophilicity led to the synthesis of a series of imidazole analogues. Compound 6 was chosen for further profiling due to its appropriate physical chemical properties and excellent FAAH inhibition potency across species. [(11)C]-6 (MK-3168) exhibited good brain uptake and FAAH-specific signal in rhesus monkeys and is a suitable PET tracer for imaging FAAH in the brain.

11.
Assay Drug Dev Technol ; 7(4): 391-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19689207

RESUMEN

beta-Galactosidase (beta-gal) (encoded by the lacZ gene) has been widely used as a transgene reporter enzyme. The ability to image lacZ expression in living transgenic animals would further extend the use of this reporter. It has been reported that 7-hydroxy-9H-(1,3-dichloro-9,9-dimethylacridin-2-one)-beta-d-galactopyranoside (DDAOG), a conjugate of beta-galactose and 7-hydroxy-9H-(1,3-dichloro-9,9-dimethylacridin-2-one), is not only a chromogenic lacZ substrate but that the cleavage product has far-red fluorescence properties detectable by in vivo imaging. In an attempt to noninvasively image lacZ expression in vivo, we applied fluorescence imaging to a G protein-coupled receptor (GPR56), knockout (KO) mouse model, in which the lacZ gene is introduced in the GPR56 locus. Compared to wild-type (WT) mice, GPR56KO/LacZ mice showed three- to fourfold higher fluorescence intensity in the head area 5 min after tail-vein injection of DDAOG. beta-Gal staining in sections of whole brain showed strong lacZ expression in homozygotes, but not in WT mice, consistent with lacZ activity detected by in vivo imaging. The kidneys were also visualized with fluorescence imaging both in vivo and ex vivo, consistent with beta-gal staining findings. Our results demonstrate that fluorescence imaging can be used for in vivo real-time detection of lacZ activity by fluorescence imaging in lacZ transgenic mice. Thus, this technology can potentially be used to noninvasively image changes of certain endogenous molecules and/or molecular pathways in transgenic animals.


Asunto(s)
Expresión Génica/genética , Operón Lac/genética , Animales , Colorantes , Galactosidasas/química , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Noqueados , Ratones Transgénicos , Microscopía Fluorescente , Fenotipo , Receptores Acoplados a Proteínas G/genética , Procesamiento de Señales Asistido por Computador , Transfección , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
12.
Nat Protoc ; 3(5): 891-9, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18451797

RESUMEN

We have applied noninvasive optical imaging technology to the in vivo hollow fiber assay, using tumor cell lines in which optical reporters are expressed in response to activation/inhibition of a specific molecular pathway. In vivo noninvasive imaging of molecular pathways in cells within hollow fibers enables a rapid and accurate evaluation of drug targets and provides useful insights to guide novel drug discovery. In this protocol we show, as an example, that a luciferase reporter, driven by the responsive element of nuclear factor NF-kappaB, was induced in cells within hollow fibers implanted in living mice, and a detailed procedure for in vivo bioluminescence imaging of hollow fibers is described. This approach can, in principle, be applied to image any molecular pathways of interest when appropriate reporter cells are generated. Hollow fiber encapsulation and implantation takes 2 d, and in vivo validation of reporter takes 1-2 weeks.


Asunto(s)
Diagnóstico por Imagen/métodos , Mediciones Luminiscentes/métodos , Polivinilos/química , Animales , Materiales Biocompatibles , Línea Celular , Luciferasas , Ratones , FN-kappa B
13.
Neoplasia ; 9(8): 652-61, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17786184

RESUMEN

The in vivo hollow fiber assay, in which semipermeable hollow fibers filled with tumor cells, are implanted into animals, was originally developed to screen for anticancer compounds before assessment in more complex tumor models. To enhance screening and evaluation of anticancer drugs, we have applied optical imaging technology to this assay. To demonstrate that tumor cells inside hollow fibers can communicate with the host mice, we have used fluorescence imaging in vivo and CD31 immunostaining ex vivo to show that angiogenesis occurs around cell-filled hollow fibers by 2 weeks after subcutaneous implantation. Bioluminescence imaging has been used to follow the number of luciferase-expressing tumor cells within implanted hollow fibers; proliferation of those cells was found to be significantly inhibited by docetaxel or irinotecan. We also used bioluminescence imaging of hollow fibers to monitor the nuclear factor kappaB (NFkappaB) pathway in vivo; NFkappaB activation by lipopolysaccharide and tumor necrosis factor-alpha was evaluated in tumor cell lines genetically engineered to express luciferase controlled by an NFkappaB-responsive element. These results demonstrate that optical imaging of hollow fibers containing reporter tumor cells can be used for the rapid and accurate evaluation of antitumor activities of anticancer drugs and for measurement of molecular pathways.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Mediciones Luminiscentes/métodos , Animales , Antineoplásicos/análisis , Ensayos de Selección de Medicamentos Antitumorales/instrumentación , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias/instrumentación , Trasplante de Neoplasias/métodos , Óptica y Fotónica , Ratas , Células Tumorales Cultivadas
14.
Biochemistry ; 45(48): 14355-61, 2006 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-17128974

RESUMEN

We report the critical residues for the interaction of the kinins with human bradykinin receptor 1 (B1) using site-directed mutagenesis in conjunction with molecular modeling of the binding modes of the kinins in the homology model of the B1 receptor. Mutation of Lys118 in transmembrane (TM) helix 3, Ala270 in TM6, and Leu294 in TM7 causes a significant decrease in the affinity for the peptide agonists des-Arg10kallidin (KD) and des-Arg9BK but not the peptide antagonist des-Arg10Leu9KD. In contrast, mutations in TM2, TM3, TM6, and TM7 cause a significant decrease in the affinity for both the peptide agonists and the antagonist. These data indicate that the B1 bradykinin binding pocket for agonists and antagonists is similar, but the manners in which they interact with the receptor do not completely overlap. Therefore, there is a potential to influence the receptor's ligand selectivity.


Asunto(s)
Cininas/química , Cininas/metabolismo , Modelos Moleculares , Receptor de Bradiquinina B1/química , Receptor de Bradiquinina B1/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Secuencia Conservada , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Mutación/genética , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Secundaria de Proteína , Receptor de Bradiquinina B1/genética , Alineación de Secuencia
15.
Biol Chem ; 387(2): 195-201, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16497152

RESUMEN

Antagonists of the B1 bradykinin receptor (B1R), encoded by the BDKRB1 gene, offer the promise of novel therapeutic agents for inflammatory and neuropathic pain. However, the in vivo characterization of the pharmacodynamics of B1R antagonists is hindered by the low level of B1R expression in healthy tissue and the profound species selectivity exhibited by many compounds for the B1R. To circumvent these issues we generated two genetically engineered rodent models. The first is a transgenic rat over-expressing the human B1R under the control of the neuronal-specific enolase promoter; we previously reported the utility of this model in assessing human B1R receptor occupancy in the central nervous system of the rat. The second model, reported here, utilized gene-targeting by homologous recombination to replace the genomic coding sequence for the endogenous mouse B1R with that of the human B1R. The mRNA expression profile of the humanized Bdkrb1 (hBkdrb1) allele is similar to that of the mouse Bdkrb1 (mBkdrb1) in the wild-type animal. Furthermore, in vitro assays indicate that tissues isolated from the humanized mouse possess pharmacological properties characteristic of the human B1R. Therefore, we have generated a humanized B1R mouse model that is suitable for testing the efficacy of human B1R-selective compounds.


Asunto(s)
Acetamidas/farmacología , Receptor de Bradiquinina B1 , Sulfonas/farmacología , Acetamidas/química , Animales , Antagonistas del Receptor de Bradiquinina B1 , Relación Dosis-Respuesta a Droga , Perfilación de la Expresión Génica , Humanos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Estructura Molecular , ARN Mensajero/genética , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/fisiología , Sulfonas/química , Transcripción Genética
16.
Biochem Biophys Res Commun ; 331(1): 159-66, 2005 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-15845373

RESUMEN

We report the first homology model of human bradykinin receptor B1 generated from the crystal structure of bovine rhodopsin as a template. Using an automated docking procedure, two B1 receptor antagonists of the dihydroquinoxalinone structural class were docked into the receptor model. Site-directed mutagenesis data of the amino acid residues in TM1, TM3, TM6, and TM7 were incorporated to place the compounds in the binding site of the homology model of the human B1 bradykinin receptor. The best pose in agreement with the mutation data was selected for detailed study of the receptor-antagonist interaction. To test the model, the calculated antagonist-receptor binding energy was correlated with the experimentally measured binding affinity (K(i)) for nine dihydroquinoxalinone analogs. The model was used to gain insight into the molecular mechanism for receptor function and to optimize the dihydroquinoxalinone analogs.


Asunto(s)
Modelos Moleculares , Quinoxalinas/química , Receptor de Bradiquinina B1/química , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Receptor de Bradiquinina B1/genética , Receptor de Bradiquinina B1/metabolismo , Rodopsina/química , Alineación de Secuencia , Homología Estructural de Proteína
17.
J Pharmacol Exp Ther ; 313(1): 199-206, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15608073

RESUMEN

We found that 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) is a potent and selective positive allosteric modulator of the metabotropic glutamate receptor subtype 5 (mGluR5). In Chinese hamster ovary cells expressing human mGluR5, CDPPB potentiated threshold responses to glutamate in fluorometric Ca2+ assays more than 7-fold with an EC50 value of approximately 27 nM. At 1 microM, CDPPB shifted mGluR5 agonist concentration response curves to glutamate, quisqualate, and (R,S)-3,5-dihydroxyphenylglycine 3- to 9-fold to the left. At higher concentrations, CDPPB exhibited agonist-like activity on cells expressing mGluR5. No other activity was observed on any other mGluR or cell type at concentrations up to 10 microM. CDPPB had no effect on [3H]quisqualate binding to mGluR5 but did compete for binding of [3H]methoxyPEPy, an analog of the selective mGluR5 negative allosteric modulator MPEP. CDPPB was found to be brain penetrant and reversed amphetamine-induced locomotor activity and amphetamine-induced deficits in prepulse inhibition in rats, two models sensitive to antipsychotic drug treatment. These results demonstrate that positive allosteric modulation of mGluR5 produces behavioral effects, suggesting that such modulation serves as a viable approach to increasing mGluR5 activity in vivo. These effects are consistent with the hypothesis that allosteric potentiation of mGluR5 may provide a novel approach for development of antipsychotic agents.


Asunto(s)
Antipsicóticos/farmacología , Conducta Animal/efectos de los fármacos , Benzamidas/farmacología , Ftalimidas/farmacología , Receptores de Glutamato Metabotrópico/efectos de los fármacos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Aminoácidos/farmacología , Anfetamina/antagonistas & inhibidores , Anfetamina/farmacología , Animales , Antipsicóticos/farmacocinética , Benzamidas/farmacocinética , Células CHO , Línea Celular , Estimulantes del Sistema Nervioso Central/antagonistas & inhibidores , Estimulantes del Sistema Nervioso Central/farmacología , Cricetinae , Perros , Antagonistas de Aminoácidos Excitadores/farmacología , Femenino , Haplorrinos , Humanos , Interpretación de Imagen Asistida por Computador , Técnicas In Vitro , Microsomas Hepáticos/metabolismo , Modelos Estadísticos , Actividad Motora/efectos de los fármacos , Ftalimidas/farmacocinética , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores , Reflejo de Sobresalto/efectos de los fármacos , Xantenos/farmacología
18.
Mol Pharmacol ; 64(3): 731-40, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12920211

RESUMEN

We have identified a family of highly selective allosteric modulators of the group I metabotropic glutamate receptor subtype 5 (mGluR5). This family of closely related analogs exerts a spectrum of effects, ranging from positive to negative allosteric modulation, and includes compounds that do not themselves modulate mGluR5 agonist activity but rather prevent other family members from exerting their modulatory effects. 3,3'-Difluorobenzaldazine (DFB) has no agonist activity, but it acts as a selective positive allosteric modulator of human and rat mGluR5. DFB potentiates threshold responses to glutamate, quisqualate, and 3,5-dihydroxyphenylglycine in fluorometric Ca2+ assays 3- to 6-fold, with EC50 values in the 2 to 5 microM range, and at 10 to 100 microM, it shifts mGluR5 agonist concentration-response curves approximately 2-fold to the left. The analog 3,3'-dimethoxybenzaldazine (DMeOB) acts as a negative modulator of mGluR5 agonist activity, with an IC50 of 3 microM in fluorometric Ca2+ assays, whereas the analog 3,3'-dichlorobenzaldazine (DCB) does not exert any apparent modulatory effect on mGluR5 activity. However, DCB seems to act as an allosteric ligand with neutral cooperativity, preventing the positive allosteric modulation of mGluRs by DFB as well as the negative modulatory effect of DMeOB. None of these analogs affects binding of [3H]quisqualate to the orthosteric (glutamate) site, but they do inhibit [3H]3-methoxy-5-(2-pyridinylethynyl)pyridine binding to the site for 2-methyl-6-(phenylethynyl)-pyridine, a previously identified negative allosteric modulator. With the use of these compounds, we provide evidence that allosteric sites on GPCRs can respond to closely related ligands with a range of pharmacological activities from positive to negative modulation as well as to neutral competition of this modulation.


Asunto(s)
Agonistas de Aminoácidos Excitadores/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Receptores de Glutamato Metabotrópico/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Animales , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Agonistas de Aminoácidos Excitadores/química , Antagonistas de Aminoácidos Excitadores/química , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Receptor del Glutamato Metabotropico 5 , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/antagonistas & inhibidores
19.
J Pharmacol Exp Ther ; 310(2): 488-97, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15051800

RESUMEN

Antagonists of the B1 bradykinin receptor (B1R) offer the promise of novel therapeutic agents for the treatment of inflammatory and neuropathic pain. However, the in vivo characterization of the pharmacodynamics of B1R antagonists is hindered by the low level of B1R expression in healthy tissue and the profound species selectivity exhibited by many compounds for the human B1R. To circumvent these issues, we generated a transgenic rat expressing the human B1R under the control of the neuron-specific enolase promoter. Membranes prepared from whole brain homogenates of heterozygous transgenic rats indicate a B1R expression level of 30 to 40 fmol/mg; there is no detectable B1R expression in control nontransgenic rats. The pharmacological profile of the B1R expressed in the transgenic rat matches that expected of the human, but not the rat receptor. The mapping of the transgene insertion site to rat chromosome 1 permitted the development of a reliable assay for the identification of homozygous transgenic rats. Significantly, homozygous transgenic rats express 2-fold more B1R than heterozygous animals. Autoradiographic analyses of tissue sections from transgenic rats reveal that the B1R is broadly expressed in both the brain and spinal cord. The human B1R expressed in the transgenic rat functions in an in vitro contractile assay and thus has the potential to elicit a functional response in vivo. Using the humanized B1R transgenic rat, an assay was developed that is suitable for the routine evaluation of a test compound's ability to occupy the human B1R in the central nervous system.


Asunto(s)
Animales Modificados Genéticamente/genética , Modelos Animales , Ratas/genética , Receptor de Bradiquinina B1/biosíntesis , Receptor de Bradiquinina B1/genética , Animales , Animales Modificados Genéticamente/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células CHO , Cricetinae , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Íleon/efectos de los fármacos , Íleon/metabolismo , Masculino , Fragmentos de Péptidos/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA