Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 15(1): 307, 2018 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-30400912

RESUMEN

BACKGROUND: Causal associations between microglia activation and ß-amyloid (Aß) accumulation during the progression of Alzheimer's disease (AD) remain a matter of controversy. Therefore, we used longitudinal dual tracer in vivo small animal positron emission tomography (µPET) imaging to resolve the progression of the association between Aß deposition and microglial responses during aging of an Aß mouse model. METHODS: APP-SL70 mice (N = 17; baseline age 3.2-8.5 months) and age-matched C57Bl/6 controls (wildtype (wt)) were investigated longitudinally for 6 months using Aß (18F-florbetaben) and 18 kDa translocator protein (TSPO) µPET (18F-GE180). Changes in cortical binding were transformed to Z-scores relative to wt mice, and microglial activation relative to amyloidosis was defined as the Z-score difference (TSPO-Aß). Using 3D immunohistochemistry for activated microglia (Iba-1) and histology for fibrillary Aß (methoxy-X04), we measure microglial brain fraction relative to plaque size and the distance from plaque margins. RESULTS: Aß-PET binding increased exponentially as a function of age in APP-SL70 mice, whereas TSPO binding had an inverse U-shape growth function. Longitudinal Z-score differences declined with aging, suggesting that microglial response declined relative to increasing amyloidosis in aging APP-SL70 mice. Microglial brain volume fraction was inversely related to adjacent plaque size, while the proximity to Aß plaques increased with age. CONCLUSIONS: Microglial activity decreases relative to ongoing amyloidosis with aging in APP-SL70 mice. The plaque-associated microglial brain fraction saturated and correlated negatively with increasing plaque size with aging.


Asunto(s)
Envejecimiento , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Microglía/metabolismo , Tomografía de Emisión de Positrones , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Amiloidosis/diagnóstico por imagen , Animales , Proteínas de Unión al Calcio/metabolismo , Carbazoles/farmacocinética , Modelos Animales de Enfermedad , Fluorodesoxiglucosa F18/farmacocinética , Estudios Longitudinales , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Microglía/efectos de los fármacos , Radioquímica , Receptores de GABA/metabolismo
2.
J Neurosci ; 34(35): 11621-30, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25164658

RESUMEN

Therapeutic approaches for prevention or reduction of amyloidosis are currently a main objective in basic and clinical research on Alzheimer's disease. Among the agents explored in clinical trials are anti-Aß peptide antibodies and secretase inhibitors. Most anti-Aß antibodies are considered to act via inhibition of amyloidosis and enhanced clearance of existing amyloid, although secretase inhibitors reduce the de novo production of Aß. Limited information is currently available on the efficacy and potential advantages of combinatorial antiamyloid treatment. We performed a chronic study in APPLondon transgenic mice that received treatment with anti-Aß antibody gantenerumab and BACE inhibitor RO5508887, either as mono- or combination treatment. Treatment aimed to evaluate efficacy on amyloid progression, similar to preexisting amyloidosis as present in Alzheimer's disease patients. Mono-treatments with either compound caused a dose-dependent reduction of total brain Aß and amyloid burden. Combination treatment with both compounds significantly enhanced the antiamyloid effect. The observed combination effect was most pronounced for lowering of amyloid plaque load and plaque number, which suggests effective inhibition of de novo plaque formation. Moreover, significantly enhanced clearance of pre-existing amyloid plaques was observed when gantenerumab was coadministered with RO5508887. BACE inhibition led to a significant time- and dose-dependent decrease in CSF Aß, which was not observed for gantenerumab treatment. Our results demonstrate that combining these two antiamyloid agents enhances overall efficacy and suggests that combination treatments may be of clinical relevance.


Asunto(s)
Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Anticuerpos Monoclonales/administración & dosificación , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Encéfalo/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Péptidos beta-Amiloides/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados , Encéfalo/patología , Modelos Animales de Enfermedad , Quimioterapia Combinada , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Placa Amiloide/patología
3.
Brain ; 137(Pt 10): 2834-46, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25085375

RESUMEN

The severity of tau pathology in Alzheimer's disease brain correlates closely with disease progression. Tau immunotherapy has therefore been proposed as a new therapeutic approach to Alzheimer's disease and encouraging results have been obtained by active or passive immunization of tau transgenic mice. This work investigates the mechanism by which immunotherapy can impact tau pathology. We demonstrate the development of Alzheimer's disease-like tau pathology in a triple transgenic mouse model of Alzheimer's disease and show that tau/pS422 is present in membrane microdomains on the neuronal cell surface. Chronic, peripheral administration of anti-tau/pS422 antibody reduces the accumulation of tau pathology. The unequivocal presence of anti-tau/pS422 antibody inside neurons and in lysosomes is demonstrated. We propose that anti-tau/pS422 antibody binds to membrane-associated tau/pS422 and that the antigen-antibody complexes are cleared intracellularly, thereby offering one explanation for how tau immunotherapy can ameliorate neuronal tau pathology.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Enfermedad de Alzheimer/patología , Anticuerpos/metabolismo , Proteínas tau/inmunología , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Western Blotting , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Lisosomas/metabolismo , Lisosomas/patología , Microdominios de Membrana/patología , Ratones , Ratones Transgénicos , Fosforilación , Sarcosina/análogos & derivados , Sarcosina/química
4.
Proc Natl Acad Sci U S A ; 108(20): 8485-90, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21525407

RESUMEN

The trace amine-associated receptor 1 (TAAR1), activated by endogenous metabolites of amino acids like the trace amines p-tyramine and ß-phenylethylamine, has proven to be an important modulator of the dopaminergic system and is considered a promising target for the treatment of neuropsychiatric disorders. To decipher the brain functions of TAAR1, a selective TAAR1 agonist, RO5166017, was engineered. RO5166017 showed high affinity and potent functional activity at mouse, rat, cynomolgus monkey, and human TAAR1 stably expressed in HEK293 cells as well as high selectivity vs. other targets. In mouse brain slices, RO5166017 inhibited the firing frequency of dopaminergic and serotonergic neurons in regions where Taar1 is expressed (i.e., the ventral tegmental area and dorsal raphe nucleus, respectively). In contrast, RO5166017 did not change the firing frequency of noradrenergic neurons in the locus coeruleus, an area devoid of Taar1 expression. Furthermore, modulation of TAAR1 activity altered the desensitization rate and agonist potency at 5-HT(1A) receptors in the dorsal raphe, suggesting that TAAR1 modulates not only dopaminergic but also serotonergic neurotransmission. In WT but not Taar1(-/-) mice, RO5166017 prevented stress-induced hyperthermia and blocked dopamine-dependent hyperlocomotion in cocaine-treated and dopamine transporter knockout mice as well as hyperactivity induced by an NMDA antagonist. These results tie TAAR1 to the control of monoamine-driven behaviors and suggest anxiolytic- and antipsychotic-like properties for agonists such as RO5166017, opening treatment opportunities for psychiatric disorders.


Asunto(s)
Monoaminas Biogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transmisión Sináptica/fisiología , Animales , Benzodioxoles/farmacología , Dopamina/metabolismo , Glutamina/metabolismo , Células HEK293 , Humanos , Trastornos Mentales , Ratones , Fenilpropionatos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/deficiencia
5.
Neurobiol Dis ; 47(1): 1-12, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22426397

RESUMEN

Alzheimer's disease (AD) is a neurodegenerative disorder characterized by brain accumulation of amyloid-ß peptide and neurofibrillary tangles, which are believed to initiate a pathological cascade that results in progressive impairment of cognitive functions and eventual neuronal death. To obtain a mouse model displaying the typical AD histopathology of amyloidosis and tauopathy, we generated a triple-transgenic mouse line (TauPS2APP) by overexpressing human mutations of the amyloid precursor protein, presenilin2 and tau genes. Stereological analysis of TauPS2APP mice revealed significant neurodegeneration of GABAergic septo-hippocampal projection neurons as well as their target cells, the GABAergic hippocampal interneurons. In contrast, the cholinergic medial septum neurons remained unaffected. Moreover, the degeneration of hippocampal GABAergic interneurons was dependent on the hippocampal subfield and interneuronal subtype investigated, whereby the dentate gyrus and the NPY-positive interneurons, respectively, were most strongly affected. Neurodegeneration was also accompanied by a change in the mRNA expression of markers for inhibitory interneurons. In line with the loss of inhibitory neurons, we observed functional changes in TauPS2APP mice relative to WT mice, with strongly enhanced long-term potentiation in the medial-perforant pathway input to the dentate gyrus, and stereotypic hyperactivity. Our data indicate that inhibitory neurons are the targets of neurodegeneration in a mouse model of amyloidosis and tauopathy, thus pointing to a possible role of the inhibitory network in the pathophysiological and functional cascade of Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuronas Colinérgicas/metabolismo , Neuronas GABAérgicas/metabolismo , Hipocampo/metabolismo , Interneuronas/metabolismo , Potenciación a Largo Plazo , Núcleos Septales/metabolismo , Enfermedad de Alzheimer/patología , Amiloidosis/patología , Animales , Neuronas Colinérgicas/patología , Modelos Animales de Enfermedad , Neuronas GABAérgicas/patología , Hipocampo/patología , Interneuronas/patología , Ratones , Ratones Transgénicos , Neuropéptido Y/metabolismo , Presenilina-2/genética , Núcleos Septales/patología , Tauopatías/patología , Proteínas tau/genética
6.
J Pharmacokinet Pharmacodyn ; 39(3): 227-37, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22481485

RESUMEN

Inhibition of the enzyme(s) that produce the Amyloid beta (Aß) peptide, namely BACE and γ-secretase, is considered an attractive target for Alzheimer's disease therapy. However, the optimal pharmacokinetic-pharmacodynamic modelling method to describe the changes in Aß levels after drug treatment is unclear. In this study, turnover models were employed to describe Aß levels following treatment with the γ-secretase inhibitor RO5036450, in the wild type rat. Initially, Aß level changes in the brain, cerebral spinal fluid (CSF) and plasma were modeled as separate biological compartments, which allowed the estimation of a compound IC50 and Aß turnover. While the data were well described, the model did not take into consideration that the CSF pool of Aß most likely originates from the brain via the CSF drainage pathway. Therefore, a separate model was carried out, with the assumption that CSF Aß levels originated from the brain. The optimal model that described the data involved two brain Aß 40 sub-compartments, one with a rapid turnover, from which CSF Aß 40 is derived, and a second quasi-static pool of ~20%. Importantly, the estimated in vivo brain IC50 was in a good range of the in vitro IC50 (ratio, 1.4). In conclusion, the PK/PD models presented here are well suited for describing the temporal changes in Aß levels that occur after treatment with an Aß lowering drug, and identifying physiological parameters.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Modelos Biológicos , Inhibidores de Proteasas/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/enzimología , Encéfalo/metabolismo , Masculino , Ratas , Ratas Wistar , Resultado del Tratamiento
7.
Proc Natl Acad Sci U S A ; 106(47): 20081-6, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19892733

RESUMEN

Trace amine-associated receptor 1 (TAAR1) is a G protein-coupled receptor (GPCR) that is nonselectively activated by endogenous metabolites of amino acids. TAAR1 is considered a promising drug target for the treatment of psychiatric and neurodegenerative disorders. However, no selective ligand to identify TAAR1-specific signaling mechanisms is available yet. Here we report a selective TAAR1 antagonist, EPPTB, and characterize its physiological effects at dopamine (DA) neurons of the ventral tegmental area (VTA). We show that EPPTB prevents the reduction of the firing frequency of DA neurons induced by p-tyramine (p-tyr), a nonselective TAAR1 agonist. When applied alone, EPPTB increases the firing frequency of DA neurons, suggesting that TAAR1 either exhibits constitutive activity or is tonically activated by ambient levels of endogenous agonist(s). We further show that EPPTB blocks the TAAR1-mediated activation of an inwardly rectifying K(+) current. When applied alone, EPPTB induces an apparent inward current, suggesting the closure of tonically activated K(+) channels. Importantly, these EPPTB effects were absent in Taar1 knockout mice, ruling out off-target effects. We additionally found that both the acute application of EPPTB and the constitutive genetic lack of TAAR1 increase the potency of DA at D2 receptors in DA neurons. In summary, our data support that TAAR1 tonically activates inwardly rectifying K(+) channels, which reduces the basal firing frequency of DA neurons in the VTA. We hypothesize that the EPPTB-induced increase in the potency of DA at D2 receptors is part of a homeostatic feedback mechanism compensating for the lack of inhibitory TAAR1 tone.


Asunto(s)
Benzamidas/química , Benzamidas/metabolismo , Dopamina/metabolismo , Sistema Límbico , Neuronas/metabolismo , Pirrolidinas/química , Pirrolidinas/metabolismo , Receptores Acoplados a Proteínas G , Área Tegmental Ventral , Potenciales de Acción/fisiología , Animales , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Humanos , Sistema Límbico/citología , Sistema Límbico/metabolismo , Ratones , Ratones Noqueados , Estructura Molecular , Neuronas/citología , Oocitos/citología , Oocitos/fisiología , Técnicas de Placa-Clamp , Ratas , Receptores de Dopamina D2/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/metabolismo , Área Tegmental Ventral/citología , Área Tegmental Ventral/metabolismo , Xenopus laevis
8.
Proc Natl Acad Sci U S A ; 106(47): 20057-62, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19897719

RESUMEN

Alzheimer's disease (AD) is characterized by amyloid-beta (Abeta)-containing plaques, neurofibrillary tangles, and neuron and synapse loss. Tangle formation has been reproduced in P301L tau transgenic pR5 mice, whereas APP(sw)PS2(N141I) double-transgenic APP152 mice develop Abeta plaques. Cross-breeding generates triple transgenic ((triple)AD) mice that combine both pathologies in one model. To determine functional consequences of the combined Abeta and tau pathologies, we performed a proteomic analysis followed by functional validation. Specifically, we obtained vesicular preparations from (triple)AD mice, the parental strains, and nontransgenic mice, followed by the quantitative mass-tag labeling proteomic technique iTRAQ and mass spectrometry. Within 1,275 quantified proteins, we found a massive deregulation of 24 proteins, of which one-third were mitochondrial proteins mainly related to complexes I and IV of the oxidative phosphorylation system (OXPHOS). Notably, deregulation of complex I was tau dependent, whereas deregulation of complex IV was Abeta dependent, both at the protein and activity levels. Synergistic effects of Abeta and tau were evident in 8-month-old (triple)AD mice as only they showed a reduction of the mitochondrial membrane potential at this early age. At the age of 12 months, the strongest defects on OXPHOS, synthesis of ATP, and reactive oxygen species were exhibited in the (triple)AD mice, again emphasizing synergistic, age-associated effects of Abeta and tau in perishing mitochondria. Our study establishes a molecular link between Abeta and tau protein in AD pathology in vivo, illustrating the potential of quantitative proteomics.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides/metabolismo , Ratones Transgénicos , Fosforilación Oxidativa , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/genética , Animales , Humanos , Espectrometría de Masas/métodos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas tau/genética
9.
Bioorg Med Chem Lett ; 21(21): 6554-8, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21924610

RESUMEN

We herein report the discovery of a new γ-secretase modulator class with an aminothiazole core starting from a HTS hit (3). Synthesis and SAR of this series are discussed. These novel compounds demonstrate moderate to good in vitro potency in inhibiting amyloid beta (Aß) peptide production. Overall γ-secretase is not inhibited but the formation of the aggregating, toxic Aß42 peptide is shifted to smaller non-aggregating Aß peptides. Compound 15 reduced brain Aß42 in vivo in APPSwe transgenic mice at 30mg/kg p.o.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/efectos de los fármacos , Tiazoles/farmacología , Animales , Humanos , Ratones , Ratones Transgénicos , Relación Estructura-Actividad , Tiazoles/química
10.
Neurobiol Dis ; 37(2): 294-306, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19781645

RESUMEN

Amyloid beta peptides and microtubule-associated protein Tau are misfolded and form aggregates in brains of Alzheimer's disease patients. To examine their specific roles in the pathogenesis of Alzheimer's disease and their relevance in neurodegenerative processes, we have created TauPS2APP triple transgenic mice that express human mutated Amyloid Precursor Protein, presenilin 2 and Tau. We present a cross-sectional analysis of these mice at 4, 8, 12 and 16 months of age. By comparing with single transgenic Tau mice, we demonstrate that accumulation of Abeta in TauPS2APP triple transgenic mice impacts on Tau pathology by increasing the phosphorylation of Tau at serine 422, as determined by a novel immunodetection method that is able to reliably measure phospho-Tau species in transgenic mouse brains. The TauPS2APP triple transgenic mouse model will be very useful for studying the effect of new therapeutic paradigms on amyloid deposition and downstream neurofibrillary tangle development.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Presenilina-2/metabolismo , Proteínas tau/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Animales , Biomarcadores/análisis , Biomarcadores/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Estudios Transversales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Ovillos Neurofibrilares/metabolismo , Fosforilación , Placa Amiloide/metabolismo , Presenilina-2/genética , Regulación hacia Arriba/fisiología , Proteínas tau/genética
12.
Neurodegener Dis ; 6(1-2): 29-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19066434

RESUMEN

BACKGROUND: Alzheimer's disease is the most common cause of dementia occurring in the elderly. The identification of the genetic factors in the familial forms of the disease enabled the generation of transgenic animals which reproduce an essential part of its pathology. Various lines of transgenic mice expressing human wild-type or mutated APP have been reported. The phenotype expressed in these mice varies according to the transgene itself, the promoter used for its expression, and the level of expression achieved in the brain, but is also determined by the genetic background of the mice. METHODS: We analyzed the variability in the amyloid load by ELISA and the levels of huAPP transcripts by quantitative PCR in our PS2APP double-transgenic mice when expressed in a mixed C57Bl/6, DBA/2 background. RESULTS: In 12-month-old PS2APP double-transgenic mice, the amount of cerebral amyloid deposits is directly correlated with the levels of the huAPP transgenic transcript. Furthermore, a reduction in human APP transcripts by 50% leads to a complete absence of cerebral deposits at the age of 12 months. CONCLUSION: Our results demonstrate that a 2-fold reduction in APP expression can result in a profound decrease in brain pathology.


Asunto(s)
Péptidos beta-Amiloides/genética , Precursor de Proteína beta-Amiloide/genética , Presenilina-2/genética , ARN Mensajero/genética , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Sustitución de Aminoácidos , Animales , Encéfalo/patología , Cromosomas Humanos Par 1 , Cromosomas Humanos Par 21 , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/patología , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa
13.
J Nucl Med ; 60(4): 548-554, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30262517

RESUMEN

Neuroinflammation may have beneficial or detrimental net effects on the cognitive outcome of Alzheimer disease (AD) patients. PET imaging with 18-kDa translocator protein (TSPO) enables longitudinal monitoring of microglial activation in vivo. Methods: We compiled serial PET measures of TSPO and amyloid with terminal cognitive assessment (water maze) in an AD transgenic mouse model (PS2APP) from 8 to 13 mo of age, followed by immunohistochemical analyses of microglia, amyloid, and synaptic density. Results: Better cognitive outcome and higher synaptic density in PS2APP mice was predicted by higher TSPO expression at 8 mo. The progression of TSPO activation to 13 mo also showed a moderate association with spared cognition, but amyloidosis did not correlate with the cognitive outcome, regardless of the time point. Conclusion: This first PET investigation with longitudinal TSPO and amyloid PET together with terminal cognitive testing in an AD mouse model indicates that continuing microglial response seems to impart preserved cognitive performance.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/patología , Cognición , Microglía/patología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Proteínas Amiloidogénicas/metabolismo , Animales , Femenino , Estudios Longitudinales , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tomografía de Emisión de Positrones , Pronóstico , Receptores de GABA/metabolismo
14.
J Pharmacol Exp Ther ; 324(3): 948-56, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18083911

RESUMEN

The recent identification of the trace amine-associated receptor (TAAR)1 provides an opportunity to dissociate the effects of trace amines on the dopamine transporter from receptor-mediated effects. To separate both effects on a physiological level, a Taar1 knockout mouse line was generated. Taar1 knockout mice display increased sensitivity to amphetamine as revealed by enhanced amphetamine-triggered increases in locomotor activity and augmented striatal release of dopamine compared with wild-type animals. Under baseline conditions, locomotion and extracellular striatal dopamine levels were similar between Taar1 knockout and wild-type mice. Electrophysiological recordings revealed an elevated spontaneous firing rate of dopaminergic neurons in the ventral tegmental area of Taar1 knock-out mice. The endogenous TAAR1 agonist p-tyramine specifically decreased the spike frequency of these neurons in wild-type but not in Taar1 knockout mice, consistent with the prominent expression of Taar1 in the ventral tegmental area. Taken together, the data reveal TAAR1 as regulator of dopaminergic neurotransmission.


Asunto(s)
Antagonistas de Dopamina/metabolismo , Dopamina/metabolismo , Actividad Motora/fisiología , Receptores Acoplados a Proteínas G/biosíntesis , Área Tegmental Ventral/fisiología , Anfetamina/farmacología , Animales , Dopamina/genética , Dopamina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Área Tegmental Ventral/citología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
15.
Bioorg Med Chem Lett ; 18(1): 304-8, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17983746

RESUMEN

A hydroxamic acid screening hit 1 was elaborated to 5,5-dimethyl-2-oxoazepane derivatives exhibiting low nanomolar inhibition of gamma-secretase, a key proteolytic enzyme involved in Alzheimer's disease. Early ADME data showed a high metabolic clearance for the geminal dimethyl analogs which could be overcome by replacement with the bioisosteric geminal difluoro group. Synthesis and structure-activity relationship are discussed and in vivo active compounds are presented.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Azepinas/química , Azepinas/farmacología , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Animales , Azepinas/síntesis química , Humanos , Ácidos Hidroxámicos/química , Ratones , Ratones Transgénicos , Modelos Moleculares , Inhibidores de Proteasas/síntesis química , Relación Estructura-Actividad
16.
Mol Neurobiol ; 55(6): 4745-4762, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28717969

RESUMEN

Trisomy 21 or Down syndrome (DS) is the most common cause of intellectual disability of a genetic origin. The Ts65Dn (TS) mouse, which is the most commonly used and best-characterized mouse model of DS, displays many of the cognitive, neuromorphological, and biochemical anomalies that are found in the human condition. One of the mechanisms that have been proposed to be responsible for the cognitive deficits in this mouse model is impaired GABA-mediated inhibition. Because of the well-known modulatory role of GABAA α5 subunit-containing receptors in cognitive processes, these receptors are considered to be potential targets for improving the intellectual disability in DS. The chronic administration of GABAA α5-negative allosteric modulators has been shown to be procognitive without anxiogenic or proconvulsant side effects. In the present study, we use a genetic approach to evaluate the contribution of GABAA α5 subunit-containing receptors to the cognitive, electrophysiological, and neuromorphological deficits in TS mice. We show that reducing the expression of GABAA α5 receptors by deleting one or two copies of the Gabra5 gene in TS mice partially ameliorated the cognitive impairments, improved long-term potentiation, enhanced neural differentiation and maturation, and normalized the density of the GABAergic synapse markers. Reducing the gene dosage of Gabra5 in TS mice did not induce motor alterations and anxiety or affect the viability of the mice. Our results provide further evidence of the role of GABAA α5 receptor-mediated inhibition in cognitive impairment in the TS mouse model of DS.


Asunto(s)
Cognición , Síndrome de Down/patología , Síndrome de Down/fisiopatología , Fenómenos Electrofisiológicos , Hipocampo/patología , Hipocampo/fisiopatología , Receptores de GABA-A/metabolismo , Animales , Recuento de Células , Diferenciación Celular , Proliferación Celular , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Dosificación de Gen , Masculino , Ratones Noqueados , Neuronas/patología , Receptores de GABA-A/genética , Factores de Tiempo
17.
J Clin Invest ; 110(10): 1429-39, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12438441

RESUMEN

The pathological modifications of alpha-synuclein (alphaS) in Parkinson disease and related diseases are poorly understood. We have detected misfolded alphaS in situ based on the proteinase K resistance (PK resistance) of alphaS fibrils, and using specific antibodies against S129-phosphorylated alphaS as well as oxidized alphaS. Unexpectedly massive neuritic pathology was found in affected human brain regions, in addition to classical alphaS pathology. PK resistance and abnormal phosphorylation of alphaS developed with increasing age in (Thy1)-h[A30P] alphaS transgenic mice, concomitant with formation of argyrophilic, thioflavin S-positive, and electron-dense inclusions that were occasionally ubiquitinated. alphaS pathology in the transgenic mice was predominantly in the brainstem and spinal cord. Astrogliosis was found in these heavily affected tissues. Homozygous mice showed the same pathology approximately one year earlier. The transgenic mice showed a progressive deterioration of locomotor function. Thus, misfolding and hyperphosphorylation of alphaS may cause dysfunction of affected brain regions.


Asunto(s)
Endopeptidasa K/metabolismo , Proteínas del Tejido Nervioso/química , Enfermedades Neurodegenerativas/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/patología , Resistencia a Medicamentos , Femenino , Dosificación de Gen , Humanos , Técnicas In Vitro , Enfermedad por Cuerpos de Lewy/metabolismo , Enfermedad por Cuerpos de Lewy/patología , Locomoción , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Fosforilación , Pliegue de Proteína , Sinucleínas , alfa-Sinucleína
18.
J Cereb Blood Flow Metab ; 37(12): 3683-3694, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28273726

RESUMEN

The blood-brain barrier (BBB) regulates differing needs of the various brain regions by controlling transport of blood-borne components from the neurovascular circulation into the brain parenchyma. The mechanisms underlying region-specific transport across the BBB are not completely understood. Previous work showed that pericytes are key regulators of BBB function. Here we investigated whether pericytes influence BBB permeability in a region-specific manner by analysing the regional permeability of the BBB in the pdgf-b ret/ret mouse model of pericyte depletion. We show that BBB permeability is heterogeneous in pdgf-b ret/ret mice, being significantly higher in the cortex, striatum and hippocampus compared to the interbrain and midbrain. However, we show that this regional heterogeneity in BBB permeability is not explained by local differences in pericyte coverage. Region-specific differences in permeability were not associated with disruption of tight junctions but may result from changes in transcytosis across brain endothelial cells. Our data show that certain brain regions are able to maintain low BBB permeability despite substantial pericyte loss and suggest that additional, locally-acting mechanisms may contribute to control of transport.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/irrigación sanguínea , Permeabilidad Capilar , Pericitos/metabolismo , Animales , Barrera Hematoencefálica/citología , Encéfalo/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Pericitos/citología , Uniones Estrechas/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-28197095

RESUMEN

Heterozygous missense mutations in the triggering receptor expressed on myeloid cells 2 (TREM2) have been reported to significantly increase the risk of developing Alzheimer's disease (AD). Since TREM2 is specifically expressed by microglia in the brain, we hypothesized that soluble TREM2 (sTREM2) levels may increase together with in vivo biomarkers of microglial activity and amyloidosis in an AD mouse model as assessed by small animal positron-emission-tomography (µPET). In this cross-sectional study, we examined a strong amyloid mouse model (PS2APP) of four age groups by µPET with [18F]-GE180 (glial activation) and [18F]-florbetaben (amyloidosis), followed by measurement of sTREM2 levels and amyloid levels in the brain. Pathology affected brain regions were compared between tracers (dice similarity coefficients) and pseudo-longitudinally. µPET results of both tracers were correlated with terminal TREM2 levels. The brain sTREM2 levels strongly increased with age of PS2APP mice (5 vs. 16 months: +211%, p < 0.001), and correlated highly with µPET signals of microglial activity (R = 0.89, p < 0.001) and amyloidosis (R = 0.92, p < 0.001). Dual µPET enabled regional mapping of glial activation and amyloidosis in the mouse brain, which progressed concertedly leading to a high overlap in aged PS2APP mice (dice similarity 67%). Together, these results substantiate the use of in vivo µPET measurements in conjunction with post mortem sTREM2 in future anti-inflammatory treatment trials. Taking human data into account sTREM2 may increase during active amyloid deposition.

20.
Sci Rep ; 6: 25658, 2016 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-27149947

RESUMEN

The Blood-Brain Barrier (BBB) restricts access of large molecules to the brain. The low endocytic activity of brain endothelial cells (BECs) is believed to limit delivery of immunoglobulins (IgG) to the brain parenchyma. Here, we report that endogenous mouse IgG are localized within intracellular vesicles at steady state in BECs in vivo. Using high-resolution quantitative microscopy, we found a fraction of endocytosed IgG in lysosomes. We observed that loss of pericytes (key components of the BBB) in pdgf-b(ret/ret) mice affects the intracellular distribution of endogenous mouse IgG in BECs. In these mice, endogenous IgG was not detected within lysosomes but instead accumulate at the basement membrane and brain parenchyma. Such IgG accumulation could be due to reduced lysosomal clearance and increased sorting to the abluminal membrane of BECs. Our results suggest that, in addition to low uptake from circulation, IgG lysosomal degradation may be a downstream mechanism by which BECs further restrict IgG access to the brain.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Inmunoglobulinas/metabolismo , Animales , Barrera Hematoencefálica/ultraestructura , Inmunoglobulina G , Espacio Intracelular/metabolismo , Lisosomas/metabolismo , Lisosomas/ultraestructura , Ratones , Pericitos/metabolismo , Pericitos/ultraestructura , Fosforilación , Proteínas tau/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA