Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuropsychopharmacology ; 43(13): 2586-2596, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30026598

RESUMO

Emerging data continues to point towards a relationship between neuroinflammation and neuropsychiatric disorders. ATP-induced activation of P2X7 results in IL-1ß release causing neuroinflammation and microglial activation. This study describes the in-vitro and in-vivo neuropharmacology of a novel brain-penetrant P2X7 antagonist, JNJ-55308942, currently in clinical development. JNJ-55308942 is a high-affinity, selective, brain-penetrant (brain/plasma of 1) P2X7 functional antagonist. In human blood and in mouse blood and microglia, JNJ-55308942 attenuated IL-1ß release in a potent and concentration-dependent manner. After oral dosing, the compound exhibited both dose and concentration-dependent occupancy of rat brain P2X7 with an ED50 of 0.07 mg/kg. The P2X7 antagonist (3 mg/kg, oral) blocked Bz-ATP-induced brain IL-1ß release in conscious rats, demonstrating functional effects of target engagement in the brain. JNJ-55308942 (30 mg/kg, oral) attenuated LPS-induced microglial activation in mice, assessed at day 2 after a single systemic LPS injection (0.8 mg/kg, i.p.), suggesting a role for P2X7 in microglial activation. In a model of BCG-induced depression, JNJ-55308942 dosed orally (30 mg/kg), reversed the BCG-induced deficits of sucrose preference and social interaction, indicating for the first time a role of P2X7 in the BCG model of depression, probably due to the neuroinflammatory component induced by BCG inoculation. Finally, in a rat model of chronic stress induced sucrose intake deficit, JNJ-55308942 reversed the deficit with concurrent high P2X7 brain occupancy as measured by autoradiography. This body of data demonstrates that JNJ-55308942 is a potent P2X7 antagonist, engages the target in brain, modulates IL-1ß release and microglial activation leading to efficacy in two models of anhedonia in rodents.


Assuntos
Anedonia/efeitos dos fármacos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos/métodos , Mediadores da Inflamação/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Receptores Purinérgicos P2X7/fisiologia , Anedonia/fisiologia , Animais , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Antagonistas do Receptor Purinérgico P2X/química , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Piridinas/química , Piridinas/uso terapêutico , Pirimidinas/química , Pirimidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley , Ratos Wistar
2.
Alzheimers Dement ; 12(5): 527-37, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26555315

RESUMO

INTRODUCTION: Stress and corticotropin-releasing factor (CRF) have been implicated as mechanistically involved in Alzheimer's disease (AD), but agents that impact CRF signaling have not been carefully tested for therapeutic efficacy or long-term safety in animal models. METHODS: To test whether antagonism of the type-1 corticotropin-releasing factor receptor (CRFR1) could be used as a disease-modifying treatment for AD, we used a preclinical prevention paradigm and treated 30-day-old AD transgenic mice with the small-molecule, CRFR1-selective antagonist, R121919, for 5 months, and examined AD pathologic and behavioral end points. RESULTS: R121919 significantly prevented the onset of cognitive impairment in female mice and reduced cellular and synaptic deficits and beta amyloid and C-terminal fragment-ß levels in both genders. We observed no tolerability or toxicity issues in mice treated with R121919. DISCUSSION: CRFR1 antagonism presents a viable disease-modifying therapy for AD, recommending its advancement to early-phase human safety trials.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Cognição/fisiologia , Modelos Animais de Doenças , Receptores de Hormônio Liberador da Corticotropina , Sinapses/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Humanos , Camundongos , Camundongos Transgênicos , Pirimidinas , Receptores de Hormônio Liberador da Corticotropina/deficiência , Receptores de Hormônio Liberador da Corticotropina/genética
3.
J Neuroendocrinol ; 27(11): 827-34, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26510116

RESUMO

Chronic stress is implicated as a risk factor for Alzheimer's disease (AD) and other neurodegenerative disorders. Although the specific mechanisms linking stress exposure and AD vulnerability have yet to be fully determined, our laboratory and others have shown that acute and repeated restraint stress in rodents leads to an increase in hippocampal tau phosphorylation (tau-P) and tau insolubility, a critical component of tau pathology in AD. Although tau phosphorylation induced by acute psychological stress is dependent on intact signaling through the type 1 corticotropin-releasing factor receptor, how sex steroids or other modulators contribute to this effect is unknown. A naturally occurring attenuation of the stress response is observed in female rats at the end of pregnancy and throughout lactation. To test the hypothesis that decreased sensitivity to stress during lactation modulates stress-induced tau-P, cohorts of virgin, lactating and weaned female rats were subjected to 30 min of restraint stress or no stress (control) and were killed 20 min or 24 h after the episode. Exposure to restraint stress induced a significant decrease in tau-P in the hippocampus of lactating rats killed 20 min after stress compared to lactating controls and virgins subjected to stress treatment. Lactating rats killed 24 hr after restraint stress exposure showed significant elevation in tau-P compared to lactating cohorts killed 20 min after stress. Levels of tau-P in these latter cohorts did not differ signficantly from control animals. Furthermore, glycogen synthase kinase (GSK)3-α levels were significantly decreased in stressed lactating animals at both timepoints. This suggests a steep, yet transient stress-induced dephosphorylation of tau, influenced by GSK3, in the hippocampus of lactating rats.


Assuntos
Lactação/metabolismo , Estresse Psicológico/metabolismo , Proteínas tau/metabolismo , Animais , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/metabolismo , Fosforilação/efeitos dos fármacos , Gravidez , Ratos , Restrição Física , Desmame
4.
J Alzheimers Dis ; 45(4): 1175-84, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25697705

RESUMO

Stress exposure and the corticotropin-releasing factor (CRF) system have been implicated as mechanistically involved in both Alzheimer's disease (AD) and associated rodent models. In particular, the major stress receptor, CRF receptor type 1 (CRFR1), modulates cellular activity in many AD-relevant brain areas, and has been demonstrated to impact both tau phosphorylation and amyloid-ß (Aß) pathways. The overarching goal of our laboratory is to develop and characterize agents that impact the CRF signaling system as disease-modifying treatments for AD. In the present study, we developed a novel transgenic mouse to determine whether partial or complete ablation of CRFR1 was feasible in an AD transgenic model and whether this type of treatment could impact Aß pathology. Double transgenic AD mice (PSAPP) were crossed to mice null for CRFR1; resultant CRFR1 heterozygous (PSAPP-R1(+/-)) and homozygous (PSAPP-R1(-/-)) female offspring were used at 12 months of age to examine the impact of CRFR1 disruption on the severity of AD Aß levels and pathology. We found that both PSAPP-R1(+/-) and PSAPP-R1(-/-) had significantly reduced Aß burden in the hippocampus, insular, rhinal, and retrosplenial cortices. Accordingly, we observed dramatic reductions in Aß peptides and AßPP-CTFs, providing support for a direct relationship between CRFR1 and Aß production pathways. In summary, our results suggest that interference of CRFR1 in an AD model is tolerable and is efficacious in impacting Aß neuropathology.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Receptores de Hormônio Liberador da Corticotropina/deficiência , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Presenilina-1/genética , Presenilina-1/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética
5.
J Alzheimers Dis ; 43(3): 967-76, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25125464

RESUMO

Clinical and basic science research suggests that stress and/or changes in central stress signaling intermediates may be involved in Alzheimer's disease (AD) pathogenesis. Although the links between stress and AD remain unsettled, data from our group and others have established that stress exposure in rodents may confer susceptibility to AD pathology by inducing hippocampal tau phosphorylation (tau-P). Work in our laboratory has shown that stress-induced tau-P requires activation of the type-1 corticotropin-releasing factor receptor (CRFR1). CRF overexpressing (CRF-OE) mice are a model of chronic stress that display cognitive impairment at 9-10 month of age. In this study we used 6-7 month old CRF-OE mice to examine whether sustained exposure to CRF and stress steroids would impact hippocampal tau-P and kinase activity in the presence or absence of the CRFR1-specific antagonist, R121919, given daily for 30 days. CRF-OE mice had significantly elevated tau-P compared to wild type (WT) mice at the AT8 (S202/T204), PHF-1 (S396/404), S262, and S422 sites. Treating CRF-OE mice with R121919 blocked phosphorylation at the AT8 (S202/T204) and PHF-1 (S396/404) sites, but not at the S262 and S422 sites and reduced phosphorylation of c-Jun N Terminal Kinase (JNK). Examination of hippocampal extracts from CRF-OE mice at the ultrastructural level revealed negatively stained round/globular aggregates that were positively labeled by PHF-1. These data suggest critical roles for CRF and CRFR1 in tau-P and aggregation and may have implications for the development of AD cognitive decline.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Hipocampo/metabolismo , Agregação Patológica de Proteínas/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Proteínas tau/metabolismo , Animais , Hormônio Liberador da Corticotropina/genética , Hipocampo/patologia , Camundongos , Camundongos Transgênicos , Fosforilação , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/patologia , Receptores de Hormônio Liberador da Corticotropina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...