Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 374(2): 252-263, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32493725

RESUMEN

Deposition of hyperphosphorylated and aggregated tau protein in the central nervous system is characteristic of Alzheimer disease and other tauopathies. Tau is subject to O-linked N-acetylglucosamine (O-GlcNAc) modification, and O-GlcNAcylation of tau has been shown to influence tau phosphorylation and aggregation. Inhibition of O-GlcNAcase (OGA), the enzyme that removes O-GlcNAc moieties, is a novel strategy to attenuate the formation of pathologic tau. Here we described the in vitro and in vivo pharmacological properties of a novel and selective OGA inhibitor, MK-8719. In vitro, this compound is a potent inhibitor of the human OGA enzyme with comparable activity against the corresponding enzymes from mouse, rat, and dog. In vivo, oral administration of MK-8719 elevates brain and peripheral blood mononuclear cell O-GlcNAc levels in a dose-dependent manner. In addition, positron emission tomography imaging studies demonstrate robust target engagement of MK-8719 in the brains of rats and rTg4510 mice. In the rTg4510 mouse model of human tauopathy, MK-8719 significantly increases brain O-GlcNAc levels and reduces pathologic tau. The reduction in tau pathology in rTg4510 mice is accompanied by attenuation of brain atrophy, including reduction of forebrain volume loss as revealed by volumetric magnetic resonance imaging analysis. These findings suggest that OGA inhibition may reduce tau pathology in tauopathies. However, since hundreds of O-GlcNAcylated proteins may be influenced by OGA inhibition, it will be critical to understand the physiologic and toxicological consequences of chronic O-GlcNAc elevation in vivo. SIGNIFICANCE STATEMENT: MK-8719 is a novel, selective, and potent O-linked N-acetylglucosamine (O-GlcNAc)-ase (OGA) inhibitor that inhibits OGA enzyme activity across multiple species with comparable in vitro potency. In vivo, MK-8719 elevates brain O-GlcNAc levels, reduces pathological tau, and ameliorates brain atrophy in the rTg4510 mouse model of tauopathy. These findings indicate that OGA inhibition may be a promising therapeutic strategy for the treatment of Alzheimer disease and other tauopathies.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Tauopatías/tratamiento farmacológico , Tauopatías/metabolismo , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , Proteínas tau/metabolismo , Animales , Atrofia/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Locomoción/efectos de los fármacos , Masculino , Ratones , Células PC12 , Ratas , Tauopatías/patología , Tauopatías/fisiopatología
2.
J Med Chem ; 62(22): 10062-10097, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31487175

RESUMEN

Inhibition of O-GlcNAcase (OGA) has emerged as a promising therapeutic approach to treat tau pathology in neurodegenerative diseases such as Alzheimer's disease and progressive supranuclear palsy. Beginning with carbohydrate-based lead molecules, we pursued an optimization strategy of reducing polar surface area to align the desired drug-like properties of potency, selectivity, high central nervous system (CNS) exposure, metabolic stability, favorable pharmacokinetics, and robust in vivo pharmacodynamic response. Herein, we describe the medicinal chemistry and pharmacological studies that led to the identification of (3aR,5S,6S,7R,7aR)-5-(difluoromethyl)-2-(ethylamino)-3a,6,7,7a-tetrahydro-5H-pyrano[3,2-d]thiazole-6,7-diol 42 (MK-8719), a highly potent and selective OGA inhibitor with excellent CNS penetration that has been advanced to first-in-human phase I clinical trials.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , Administración Oral , Animales , Disponibilidad Biológica , Encéfalo/efectos de los fármacos , Perros , Descubrimiento de Drogas , Inhibidores Enzimáticos/sangre , Inhibidores Enzimáticos/farmacocinética , Humanos , Macaca mulatta , Masculino , Células PC12 , Ratas , Ratas Wistar , Relación Estructura-Actividad , Tauopatías/tratamiento farmacológico , beta-N-Acetilhexosaminidasas/química , beta-N-Acetilhexosaminidasas/metabolismo
3.
PLoS One ; 13(4): e0195486, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29624602

RESUMEN

Although tau pathology, behavioral deficits, and neuronal loss are observed in patients with tauopathies, the relationship between these endpoints has not been clearly established. Here we found that rTg4510 mice, which overexpress human mutant tau in the forebrain, develop progressive age-dependent increases in locomotor activity (LMA), which correlates with neurofibrillary tangle (NFT) pathology, hyperphosphorylated tau levels, and brain atrophy. To further clarify the relationship between these endpoints, we treated the rTg4510 mice with either doxycycline to reduce mutant tau expression or an O-GlcNAcase inhibitor Thiamet G, which has been shown to ameliorate tau pathology in animal models. We found that both doxycycline and Thiamet G treatments starting at 2 months of age prevented the progression of hyperactivity, slowed brain atrophy, and reduced brain hyperphosphorylated tau. In contrast, initiating doxycycline treatment at 4 months reduced neither brain hyperphosphorylated tau nor hyperactivity, further confirming the relationship between these measures. Collectively, our results demonstrate a unique behavioral phenotype in the rTg4510 mouse model of tauopathy that strongly correlates with disease progression, and that early interventions which reduce tau pathology ameliorate the progression of the locomotor dysfunction. These findings suggest that better understanding the relationship between locomotor deficits and tau pathology in the rTg4510 model may improve our understanding of the mechanisms underlying behavioral disturbances in patients with tauopathies.


Asunto(s)
Tauopatías/tratamiento farmacológico , Proteínas tau/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Doxiciclina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Actividad Motora/genética , Actividad Motora/fisiología , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ovillos Neurofibrilares/patología , Fosforilación , Piranos/uso terapéutico , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tauopatías/patología , Tauopatías/fisiopatología , Tiazoles/uso terapéutico , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , Proteínas tau/genética
4.
Mol Neurodegener ; 10: 14, 2015 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-25881209

RESUMEN

BACKGROUND: Microtubule associated protein tau is the major component of the neurofibrillary tangles (NFTs) found in the brains of patients with Alzheimer's disease and several other neurodegenerative diseases. Tau mutations are associated with frontotemperal dementia with parkinsonism on chromosome 17 (FTDP-17). rTg4510 mice overexpress human tau carrying the P301L FTDP-17 mutation and develop robust NFT-like pathology at 4-5 months of age. The current study is aimed at characterizing the rTg4510 mice to better understand the genesis of tau pathology and to better enable the use of this model in drug discovery efforts targeting tau pathology. RESULTS: Using a panel of immunoassays, we analyzed the age-dependent formation of pathological tau in rTg4510 mice and our data revealed a steady age-dependent accumulation of pathological tau in the insoluble fraction of brain homogenates. The pathological tau was associated with multiple post-translational modifications including aggregation, phosphorylation at a wide variety of sites, acetylation, ubiquitination and nitration. The change of most tau species reached statistical significance at the age of 16 weeks. There was a strong correlation between the different post-translationally modified tau species in this heterogeneous pool of pathological tau. Total tau in the cerebrospinal fluid (CSF) displayed a multiphasic temporal profile distinct from the steady accumulation of pathological tau in the brain. Female rTg4510 mice displayed significantly more aggressive accumulation of pathological tau in the brain and elevation of total tau in CSF than their male littermates. CONCLUSION: The immunoassays described here were used to generate the most comprehensive description of the changes in various tau species across the lifespan of the rTg4510 mouse model. The data indicate that development of tauopathy in rTg4510 mice involves the accumulation of a pool of pathological tau that carries multiple post-translational modifications, a process that can be detected well before the histological detection of NFTs. Therapeutic treatment targeting tau should therefore aim to reduce all tau species associated with the pathological tau pool rather than reduce specific post-translational modifications. There is still much to learn about CSF tau in physiological and pathological processes in order to use it as a translational biomarker in drug discovery.


Asunto(s)
Encéfalo/metabolismo , Procesamiento Proteico-Postraduccional/genética , Tauopatías/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo , Animales , Biomarcadores/análisis , Encéfalo/patología , Modelos Animales de Enfermedad , Humanos , Ratones Transgénicos , Procesamiento Proteico-Postraduccional/fisiología , Tauopatías/genética
5.
J Neurochem ; 118(6): 1016-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21736568

RESUMEN

The EphA4 receptor and its ephrin ligands are involved in astrocytic gliosis following CNS injury. Therefore, a strategy aimed at the blockade of EphA4 signaling could have broad therapeutic interest in brain disorders. We have identified novel small molecule inhibitors of EphA4 kinase in specific enzymatic and cell-based assays. In addition, we have demonstrated in two in vitro models of scratch injury that EphA4 receptor kinase is activated through phosphorylation and is involved in the repopulation of the wound after the scratch. A potent EphA4 kinase inhibitor significantly inhibited wound closure and reduced the accumulation of the reactive astrocytes inside the scratch. We have also shown that after the transient focal cerebral ischemia in rats, a large glial scar is formed by the accumulation of astrocytes and chondroitin sulfate proteoglycan surrounding the infarcted tissue at 7 days and 14 days of reperfusion. EphA4 protein expression is highly up-regulated in the same areas at these time points, supporting its potential role in the glial scar formation and maintenance. Taken together, these results suggest that EphA4 kinase inhibitors might interfere with the astrogliosis reaction and thereby lead to improved neurological outcome after ischemic injury.


Asunto(s)
Gliosis/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor EphA4/antagonistas & inhibidores , Heridas y Lesiones/patología , Animales , Astrocitos/patología , Western Blotting , Células CHO , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Cricetinae , Cricetulus , Gliosis/patología , Humanos , Inmunohistoquímica , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Ratas , Ratas Sprague-Dawley , Bibliotecas de Moléculas Pequeñas , Cicatrización de Heridas/efectos de los fármacos
6.
Curr Top Med Chem ; 7(10): 972-8, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17508929

RESUMEN

The transcription of antioxidant response element (ARE)-containing cytoprotective genes has been proposed as a means to combat oxidative stress-related disorders, such as cancer and Parkinson's disease. Transactivation of the ARE requires the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). Cellular levels of Nrf2 protein are regulated by the Kelch-like ECH-associated protein 1 (Keap1), a substrate adaptor protein for the ubiquitin ligase machinery and subsequent proteasomal degradation. Recently, detailed studies have elucidated the structure and interactions of the Keap1-containing ubiquitin ligase complex. Here, we propose that small molecule modulation of Keap1 protein:protein interactions may permit Nrf2's nuclear accumulation and the transcription of ARE-dependent genes to enhance cellular resistance to oxidative insult.


Asunto(s)
Antioxidantes , Péptidos y Proteínas de Señalización Intracelular , Estrés Oxidativo/efectos de los fármacos , Elementos de Respuesta/genética , Complejos de Ubiquitina-Proteína Ligasa , Animales , Antioxidantes/química , Antioxidantes/metabolismo , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Ensayos Clínicos como Asunto , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Proteína 1 Asociada A ECH Tipo Kelch , Modelos Moleculares , Estrés Oxidativo/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Complejos de Ubiquitina-Proteína Ligasa/antagonistas & inhibidores , Complejos de Ubiquitina-Proteína Ligasa/química , Complejos de Ubiquitina-Proteína Ligasa/genética
7.
J Med Chem ; 50(2): 272-82, 2007 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-17228869

RESUMEN

A series of biphenylaminocyclopropane carboxamide based bradykinin B1 receptor antagonists has been developed that possesses good pharmacokinetic properties and is CNS penetrant. Discovery that the replacement of the trifluoropropionamide in the lead structure with polyhaloacetamides, particularly a trifluoroacetamide, significantly reduced P-glycoprotein mediated efflux for the series proved essential. One of these novel bradykinin B1 antagonists (13b) also exhibited suitable pharmacokinetic properties and efficient ex vivo receptor occupancy for further development as a novel approach for the treatment of pain and inflammation.


Asunto(s)
Acetamidas/síntesis química , Amidas/síntesis química , Compuestos de Aminobifenilo/síntesis química , Benzoatos/síntesis química , Antagonistas del Receptor de Bradiquinina B1 , Encéfalo/metabolismo , Ciclopropanos/síntesis química , Médula Espinal/metabolismo , Acetamidas/farmacocinética , Acetamidas/farmacología , Administración Oral , Amidas/farmacocinética , Amidas/farmacología , Compuestos de Aminobifenilo/farmacocinética , Compuestos de Aminobifenilo/farmacología , Analgésicos/síntesis química , Analgésicos/química , Analgésicos/farmacología , Animales , Animales Modificados Genéticamente , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Benzoatos/farmacocinética , Benzoatos/farmacología , Disponibilidad Biológica , Barrera Hematoencefálica/metabolismo , Células CHO , Chlorocebus aethiops , Cricetinae , Cricetulus , Ciclopropanos/farmacocinética , Ciclopropanos/farmacología , Femenino , Humanos , Macaca mulatta , Masculino , Ratones , Conejos , Ensayo de Unión Radioligante , Ratas , Especificidad de la Especie , Relación Estructura-Actividad
8.
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
9.
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
10.
Curr Top Med Chem ; 5(9): 885-95, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16178733

RESUMEN

Over the last several years a great deal of interest has been focused on the metabotropic glutamate receptors as potential targets for the treatment of a variety of disorders of the central nervous system. Recently, selective agonist or allosteric potentiators of mGluR4, one of the group III mGluRs, have been proposed as potential novel therapeutics for the palliative treatment of Parkinson's disease and some forms of epilepsy. mGluR4 stands out amongst the group III mGluRs due to its relatively restricted localization and apparent role in several key neuronal circuits. Work from a number of laboratories has led to the development of more selective tools for the study of mGluR4, as well as refined models of the structure and function of this receptor. In addition, a growing body of literature suggests that mGluR4 plays a key neuroprotective role in broad spectrum of neurodegenerative disorders. It is hoped that this increased understanding of the role of mGluR4 combined with more detailed structural information will spur the development of better pharmacological tools, and ultimately to novel clinical therapy.


Asunto(s)
Enfermedades del Sistema Nervioso Central/tratamiento farmacológico , Receptores de Glutamato Metabotrópico/agonistas , Animales , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Agonistas de Aminoácidos Excitadores/administración & dosificación , Agonistas de Aminoácidos Excitadores/farmacología , Humanos , Ligandos , Receptores de Glutamato Metabotrópico/metabolismo
11.
J Neurochem ; 95(2): 406-17, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16092930

RESUMEN

The ability of cells to control the balance between the generation and quenching of reactive oxygen species is important in combating potentially damaging effects of oxidative stress. One mechanism that cells use to maintain redox homeostasis is the antioxidant response pathway. Antioxidant response elements (AREs) are cis-acting elements located in regulatory regions of antioxidant and phase II detoxification genes. Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) is a member of the Cap 'n' Collar family of transcription factors that binds to the ARE and regulates the transcription of specific ARE-containing genes such as NAD(P)H:quinone oxidoreductase 1, glutamylcysteine synthetase and heme oxygenase. Activation of Nrf2 results in release from its negative repressor, Kelch-like ECH-associated protein 1 (Keap1), and allows Nrf2 to translocate into the nucleus to induce gene expression. In this study, we demonstrate that increasing Nrf2 activity by various methods, including chemical induction, Nrf2 overexpression or Keap1 siRNA knockdown, protects cells against specific types of oxidative damage. Cells were protected against 6-hydroxydopamine- and 3-morpholinosydnonimine-mediated toxicity but not against 1-methyl-1-4-phenylpyridinium toxicity. As oxidative stress is a hallmark of several neurodegenerative disorders, including Parkinson's disease, pharmacological agents that selectively target the Keap1-Nrf2 pathway may provide a novel neuroprotective strategy for the treatment of these diseases.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Estrés Oxidativo/fisiología , Transactivadores/fisiología , 1-Metil-4-fenilpiridinio/antagonistas & inhibidores , 1-Metil-4-fenilpiridinio/toxicidad , Western Blotting , Supervivencia Celular/efectos de los fármacos , Proteínas de Unión al ADN/biosíntesis , Genes Reporteros/genética , Humanos , Hidroquinonas/farmacología , Péptidos y Proteínas de Señalización Intracelular , Proteína 1 Asociada A ECH Tipo Kelch , Luciferasas/metabolismo , Molsidomina/análogos & derivados , Molsidomina/toxicidad , Factor 2 Relacionado con NF-E2 , Oxidopamina/antagonistas & inhibidores , Oxidopamina/toxicidad , Plásmidos/genética , Proteínas/antagonistas & inhibidores , Proteínas/fisiología , ARN Interferente Pequeño/farmacología , Especies Reactivas de Oxígeno/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Transactivadores/biosíntesis
12.
J Bone Miner Res ; 19(12): 2033-40, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15537447

RESUMEN

UNLABELLED: Humans and mice lacking Lrp5 have low BMD. To evaluate whether Lrp5 and Lrp6 interact genetically to control bone or skeletal development, we created mice carrying mutations in both Lrp5 and the related gene Lrp6. We found that compound mutants had dose-dependent deficits in BMD and limb formation, suggesting functional redundancy between these two genes in bone and limb development. INTRODUCTION: Lrp5 and Lrp6 are closely related members of the low density lipoprotein receptor family and are co-receptors for Wnt ligands. While Lrp5 mutations are associated with low BMD in humans and mice, the role of Lrp6 in bone formation has not been analyzed. MATERIALS AND METHODS: To address whether Lrp5 and Lrp6 play complimentary roles in bone and skeletal development, we created mice with mutations in both genes. We inspected limbs of mice from the different genotypic classes of compound mutants to identify abnormalities. DXA and muCT were used to evaluate the effect of mutations in Lrp5 and Lrp6 on BMD and microarchitecture. RESULTS: Mice heterozygous for mutations in Lrp6 and either heterozygous or homozygous for a mutation in Lrp5 (Lrp6(+/-);Lrp5(+/-) or Lrp6(+/-);Lrp5(-/-)) display limb defects with incomplete penetrance and variable expression. DXA analysis showed that BMD decreased as mice progressively were more deficient in Lrp5 and Lrp6. Lrp6(+/-);Lrp5(-/-) mice were more severely affected than Lrp6(+/+);Lrp5(-/-) mice, whereas Lrp6(+/-);Lrp5(+/-) mice had statistically higher BMD than Lrp6(+/+);Lrp5(-/-) mice and lower BMD compared with wildtype mice and mice heterozygous for either mutation alone. CONCLUSIONS: Lrp6 and Lrp5 genetically interact in limb development in mice. Furthermore, heterozygosity for an inactivating mutation in Lrp6 further reduces BMD in both male and female mice lacking Lrp5.


Asunto(s)
Densidad Ósea , Deformidades Congénitas de las Extremidades/genética , Mutación , Receptores de LDL/genética , Animales , Relación Dosis-Respuesta a Droga , Extremidades/embriología , Femenino , Genotipo , Heterocigoto , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas Relacionadas con Receptor de LDL , Ligandos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Microcirculación , Fenotipo , Unión Proteica , Factores Sexuales , Factores de Tiempo , Tomografía Computarizada por Rayos X , Proteínas Wnt
14.
Eur J Pharmacol ; 499(1-2): 77-84, 2004 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-15363953

RESUMEN

Compound A (N-[2-[4-(4,5-dihydro-1H-imidazol-2-yl)phenyl]ethyl]-2-[(2R)-1-(2-napthylsulfonyl)-3-oxo-1,2,3,4-tetrahydroquinoxalin-2-yl]acetamide) is a member of a new class of aryl sulfonamide dihydroquinoxalinone bradykinin B1 receptor antagonists that should be useful pharmacological tools. Here we report on some of the pharmacological properties of compound A as well as the characterization of [35S]compound A as the first nonpeptide bradykinin B1 receptor radioligand. Compound A inhibited tritiated peptide ligand binding to the cloned human, rabbit, dog, and rat bradykinin B1 receptors expressed in CHO cells with Ki values of 0.016, 0.050, 0.56, and 29 nM, respectively. It was inactive at 10 microM in binding assays with the cloned human bradykinin B2 receptor. In functional antagonist assays with the cloned bradykinin B1 receptors, compound A inhibited agonist-induced signaling with activities consistent with the competition binding results, but had no antagonist activity at the bradykinin B2 receptor. Compound A was also found to be a potent antagonist in a rabbit aorta tissue bath preparation and to effectively block des-Arg9 bradykinin depressor responses in lipopolysaccharide-treated rabbit following intravenous administration. The binding of [35S]compound A was evaluated with the cloned bradykinin B1 receptors. In assays with human, rabbit, and dog receptors, [35S]compound A labeled a single site with Kd values of 0.012, 0.064, and 0.37 nM, respectively, and with binding site densities equivalent to those obtained using the conventional tritiated peptide ligands. Binding assays with the cloned rat bradykinin B1 receptor were not successful, presumably due to the low affinity of the ligand for this species receptor. There was no specific binding of the ligand detected in CHO cells expressing the human bradykinin B2 receptor. In assays with the cloned human bradykinin B1 receptor, the pharmacologies of the binding of [35S]compound A and [3H][Leu9]des-Arg10-kallidin were the same. The high signal-to-noise ratio obtained with [35S]compound A will allow this ligand to be a very useful tool for future investigations of the bradykinin B1 receptor.


Asunto(s)
Antagonistas del Receptor de Bradiquinina B1 , Calidina/análogos & derivados , Receptor de Bradiquinina B1/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/fisiología , Unión Competitiva/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Perros , Relación Dosis-Respuesta a Droga , Humanos , Imidazoles/metabolismo , Imidazoles/farmacología , Técnicas In Vitro , Calidina/metabolismo , Lipopolisacáridos/farmacología , Masculino , Quinoxalinas/metabolismo , Quinoxalinas/farmacología , Conejos , Ensayo de Unión Radioligante , Ratas , Receptor de Bradiquinina B1/genética , Transfección , Tritio , Vasoconstricción/efectos de los fármacos
15.
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
16.
Proc Natl Acad Sci U S A ; 100(23): 13668-73, 2003 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-14593202

RESUMEN

Parkinson's disease (PD) is a debilitating movement disorder that afflicts >1 million people in North America. Current treatments focused on dopamine-replacement strategies ultimately fail in most patients because of loss of efficacy and severe adverse effects that worsen as the disease progresses. The recent success of surgical approaches suggests that a pharmacological intervention that bypasses the dopamine system and restores balance in the basal ganglia motor circuit may provide an effective treatment strategy. We previously identified the metabotropic glutamate receptor 4 (mGluR4) as a potential drug target and predicted that selective activation of mGluR4 could provide palliative benefit in PD. We now report that N-phenyl-7-(hydroxylimino)cyclopropa[b]chromen-1a-carboxamide (PHCCC) is a selective allosteric potentiator of mGluR4. This compound selectively potentiated agonist-induced mGluR4 activity in cultured cells expressing this receptor and did not itself act as an agonist. Furthermore, PHCCC potentiated the effect of l-(+)-2-amino-4-phosphonobutyric acid in inhibiting transmission at the striatopallidal synapse. Modulation of the striatopallidal synapse has been proposed as a potential therapeutic target for PD, in that it may restore balance in the basal ganglia motor circuit. Consistent with this, PHCCC produced a marked reversal of reserpine-induced akinesia in rats. The closely related analogue 7-(hydroxylimino)cyclopropachromen-1a-carboxamide ethyl ester, which does not potentiate mGluR4, had no effect in this model. These results are evidence for in vivo behavioral effects of an allosteric potentiator of mGluRs and suggest that potentiation of mGluR4 may be a useful therapeutic approach to the treatment of PD.


Asunto(s)
Enfermedad de Parkinson/terapia , Receptores de Glutamato Metabotrópico/química , Sitio Alostérico , Animales , Benzopiranos/farmacología , Encéfalo/metabolismo , Línea Celular , Cromonas/farmacología , Dopamina/metabolismo , Electrofisiología , Ésteres/farmacología , Humanos , Masculino , Modelos Biológicos , Modelos Químicos , Ratas , Ratas Sprague-Dawley , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis , Factores de Tiempo
17.
Genome Res ; 13(5): 845-55, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12727905

RESUMEN

Patterns of linkage disequilibrium (LD) in the human genome are beginning to be characterized, with a paucity of haplotype diversity in "LD blocks," interspersed by apparent "hot spots" of recombination. Previously, we cloned and physically characterized the low-density lipoprotein-receptor-related protein 5 (LRP5) gene. Here, we have extensively analysed both LRP5 and its flanking three genes, spanning 269 kb, for single nucleotide polymorphisms (SNPs), and we present a comprehensive SNP map comprising 95 polymorphisms. Analysis revealed high levels of recombination across LRP5, including a hot-spot region from intron 1 to intron 7 of LRP5, where there are 109 recombinants/Mb (4882 meioses), in contrast to flanking regions of 14.6 recombinants/Mb. This region of high recombination could be delineated into three to four hot spots, one within a 601-bp interval. For LRP5, three haplotype blocks were identified, flanked by the hot spots. Each LD block comprised over 80% common haplotypes, concurring with a previous study of 14 genes that showed that common haplotypes account for at least 80% of all haplotypes. The identification of hot spots in between these LD blocks provides additional evidence that LD blocks are separated by areas of higher recombination.


Asunto(s)
Haplotipos/genética , Desequilibrio de Ligamiento/genética , Receptores de LDL/genética , Recombinación Genética/genética , Región de Flanqueo 3'/genética , Región de Flanqueo 5'/genética , Alelos , Mapeo Cromosómico/métodos , Cromosomas Humanos Par 11/genética , Diabetes Mellitus Tipo 1/genética , Frecuencia de los Genes/genética , Marcadores Genéticos/genética , Genética de Población , Genotipo , Humanos , Intrones/genética , Proteínas Relacionadas con Receptor de LDL , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Repeticiones de Microsatélite/genética , Núcleo Familiar , Polimorfismo de Nucleótido Simple/genética
18.
Nature ; 423(6939): 506-11, 2003 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-12724780

RESUMEN

Genes and mechanisms involved in common complex diseases, such as the autoimmune disorders that affect approximately 5% of the population, remain obscure. Here we identify polymorphisms of the cytotoxic T lymphocyte antigen 4 gene (CTLA4)--which encodes a vital negative regulatory molecule of the immune system--as candidates for primary determinants of risk of the common autoimmune disorders Graves' disease, autoimmune hypothyroidism and type 1 diabetes. In humans, disease susceptibility was mapped to a non-coding 6.1 kb 3' region of CTLA4, the common allelic variation of which was correlated with lower messenger RNA levels of the soluble alternative splice form of CTLA4. In the mouse model of type 1 diabetes, susceptibility was also associated with variation in CTLA-4 gene splicing with reduced production of a splice form encoding a molecule lacking the CD80/CD86 ligand-binding domain. Genetic mapping of variants conferring a small disease risk can identify pathways in complex disorders, as exemplified by our discovery of inherited, quantitative alterations of CTLA4 contributing to autoimmune tissue destruction.


Asunto(s)
Antígenos de Diferenciación/genética , Enfermedades Autoinmunes/genética , Predisposición Genética a la Enfermedad/genética , Inmunoconjugados , Abatacept , Empalme Alternativo/genética , Animales , Antígenos CD , Secuencia de Bases , Antígeno CTLA-4 , Diabetes Mellitus Tipo 1/genética , Modelos Animales de Enfermedad , Genotipo , Enfermedad de Graves/genética , Humanos , Hipotiroidismo/genética , Ratones , Polimorfismo de Nucleótido Simple/genética , Isoformas de Proteínas/genética , Linfocitos T/inmunología
19.
Hum Genet ; 113(2): 99-105, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12700977

RESUMEN

Linkage of chromosome 11q13 to type 1 diabetes (T1D) was first reported from genome scans (Davies et al. 1994; Hashimoto et al. 1994) resulting in P <2.2 x 10(-5) (Luo et al. 1996) and designated IDDM4 ( insulin dependent diabetes mellitus 4). Association mapping under the linkage peak using 12 polymorphic microsatellite markers suggested some evidence of association with a two-marker haplotype, D11S1917*03-H0570POLYA*02, which was under-transmitted to affected siblings and over-transmitted to unaffected siblings ( P=1.5 x 10(-6)) (Nakagawa et al. 1998). Others have reported evidence for T1D association of the microsatellite marker D11S987, which is approximately 100 kb proximal to D11S1917 (Eckenrode et al. 2000). We have sequenced a 400-kb interval surrounding these loci and identified four genes, including the low-density lipoprotein receptor related protein (LRP5) gene, which has been considered as a functional candidate gene for T1D (Hey et al. 1998; Twells et al. 2001). Consequently, we have developed a comprehensive SNP map of the LRP5 gene region, and identified 95 SNPs encompassing 269 kb of genomic DNA, characterised the LD in the region and haplotypes (Twells et al. 2003). Here, we present our refined linkage curve of the IDDM4 region, comprising 32 microsatellite markers and 12 SNPs, providing a peak MLS=2.58, P=5 x 10(-4), at LRP5 g.17646G>T. The disease association data, largely focused in the LRP5 region with 1,106 T1D families, provided no further evidence for disease association at LRP5 or at D11S987. A second dataset, comprising 1,569 families from Finland, failed to replicate our previous findings at LRP5. The continued search for the variants of the putative IDDM4 locus will greatly benefit from the future development of a haplotype map of the genome.


Asunto(s)
Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Diabetes Mellitus Tipo 1/genética , Ligamiento Genético , Receptores de LDL/genética , Genotipo , Humanos , Proteínas Relacionadas con Receptor de LDL , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Repeticiones de Microsatélite
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...