Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neuroinflammation ; 20(1): 29, 2023 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-36755323

RESUMO

Triggering receptor expressed on myeloid cells 2 (TREM2) is a cell-surface immunoreceptor expressed on microglia, osteoclasts, dendritic cells and macrophages. Heterozygous loss-of-function mutations in TREM2, including mutations enhancing shedding form the cell surface, have been associated with myelin/neuronal loss and neuroinflammation in neurodegenerative diseases, such as Alzheimer`s disease and Frontotemporal Dementia. Using the cuprizone model, we investigated the involvement of soluble and cleavage-reduced TREM2 on central myelination processes in cleavage-reduced (TREM2-IPD), soluble-only (TREM2-sol), knockout (TREM2-KO) and wild-type (WT) mice. The TREM2-sol mouse is a new model with selective elimination of plasma membrane TREM2 and a reduced expression of soluble TREM2. In the acute cuprizone model demyelination and remyelination events were reflected by a T2-weighted signal intensity change in magnetic resonance imaging (MRI), most prominently in the external capsule (EC). In contrast to WT and TREM2-IPD, TREM2-sol and TREM2-KO showed an additional increase in MRI signal during the recovery phase. Histological analyses of TREM2-IPD animals revealed no recovery of neuroinflammation as well as of the lysosomal marker LAMP-1 and displayed enhanced cytokine/chemokine levels in the brain. TREM2-sol and, to a much lesser extent, TREM2-KO, however, despite presenting reduced levels of some cytokines/chemokines, showed persistent microgliosis and astrocytosis during recovery, with both homeostatic (TMEM119) as well as activated (LAMP-1) microglia markers increased. This was accompanied, specifically in the EC, by no myelin recovery, with appearance of myelin debris and axonal pathology, while oligodendrocytes recovered. In the chronic model consisting of 12-week cuprizone administration followed by 3-week recovery TREM2-IPD displayed sustained microgliosis and enhanced remyelination in the recovery phase. Taken together, our data suggest that sustained microglia activation led to increased remyelination, whereas microglia without plasma membrane TREM2 and only soluble TREM2 had reduced phagocytic activity despite efficient lysosomal function, as observed in bone marrow-derived macrophages, leading to a dysfunctional phenotype with improper myelin debris removal, lack of remyelination and axonal pathology following cuprizone intoxication.


Assuntos
Doenças Desmielinizantes , Glicoproteínas de Membrana , Receptores Imunológicos , Animais , Camundongos , Cuprizona/toxicidade , Citocinas/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Modelos Genéticos , Bainha de Mielina/metabolismo , Doenças Neuroinflamatórias , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
2.
Int J Mol Sci ; 24(13)2023 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-37445700

RESUMO

Mild cognitive impairment (MCI) is characterized by an abnormal decline in mental and cognitive function compared with normal cognitive aging. It is an underlying condition of Alzheimer's disease (AD), an irreversible neurodegenerative disease. In recent years, neuroinflammation has been investigated as a new leading target that contributes to MCI progression into AD. Understanding the mechanism underlying inflammatory processes involved in the early onset of the disease could help find a safe and effective way to diagnose and treat patients. In this article, we assessed over twenty different blood and cerebrospinal fluid (CSF) inflammatory biomarker concentrations with immunoassay methods in patients with MCI (mild cognitive impairment), non-impaired control (NIC), and serum healthy control (HC). We performed group comparisons and analyzed in-group correlations between the biomarkers. We included 107 participants (mean age: 64.7 ± 7.8, women: 58.9%). CSF osteopontin and YKL-40 were significantly increased in the MCI group, whereas serum C-reactive protein and interleukin-6 were significantly higher (p < 0.001) in the NIC group compared with the MCI and HC groups. Stronger correlations between interleukin-1ß and inflammasome markers were observed in the serum of the MCI group. We confirmed specific inflammatory activation in the central nervous system and interleukin-1ß pathway upregulation in the serum of the MCI cohort.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Doenças Neurodegenerativas , Humanos , Feminino , Pessoa de Meia-Idade , Idoso , Doença de Alzheimer/diagnóstico , Interleucina-1beta , Disfunção Cognitiva/diagnóstico , Inflamação , Biomarcadores/líquido cefalorraquidiano , Progressão da Doença , Peptídeos beta-Amiloides/líquido cefalorraquidiano , Proteínas tau/líquido cefalorraquidiano
3.
Neuroimage ; 230: 117785, 2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33545349

RESUMO

Mavoglurant binds to same allosteric site on metabotropic glutamate receptor 5 (mGluR5) as [11C]-ABP688, a radioligand. This open-label, single-center pilot study estimates extent of occupancy of mGluR5 receptors following single oral doses of mavoglurant, using [11C]-ABP688 positron emission tomography (PET) imaging, in six healthy males aged 20-40 years. This study comprised three periods and six subjects were divided into two cohorts. On Day 1 (Period 1), baseline clinical data and safety samples were obtained along with PET scan. During Period 2 (1-7 days after Period 1), cohort 1 and 2 received mavoglurant 25 mg and 100 mg, respectively. During Period 3 (7 days after Period 2), cohort 1 and 2 received mavoglurant 200 mg and 400 mg, respectively. Mavoglurant showed the highest distribution volumes in the cingulate region with lower uptake in cerebellum and white matter, possibly because myelinated axonal sheets maybe devoid of mGlu5 receptors. Maximum concentrations of mavoglurant were observed around 2-3.25 h post-dose. Mavoglurant passed the blood-brain barrier and induced dose- and exposure-dependent displacement of [11C]-ABP688 from the mGluR5 receptors, 3-4 h post-administration (27%, 59%, 74%, 85% receptor occupancy for mavoglurant 25 mg, 100 mg, 200 mg, 400 mg dose, respectively). There were no severe adverse effects or clinically significant changes in safety parameters. This is the first human receptor occupancy study completed with Mavoglurant. It served to guide the dosing of mavoglurant in the past and currently ongoing clinical studies. Furthermore, it confirms the utility of [11C]-ABP688 as a unique tool to study drug-induced occupancy of mGlu5 receptors in the living human brain.


Assuntos
Encéfalo/metabolismo , Radioisótopos de Carbono/metabolismo , Indóis/metabolismo , Oximas/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Piridinas/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Administração Oral , Adulto , Encéfalo/efeitos dos fármacos , Estudos de Coortes , Relação Dose-Resposta a Droga , Voluntários Saudáveis , Humanos , Indóis/administração & dosagem , Masculino , Projetos Piloto , Ligação Proteica/fisiologia , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores
4.
Hum Mol Genet ; 27(12): 2039-2051, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29590342

RESUMO

Fragile X syndrome (FXS) is a monogenic form of intellectual disability and autism spectrum disorder caused by the absence of the fragile X mental retardation protein (FMRP). In biological models for the disease, this leads to upregulated mRNA translation and as a consequence, deficits in synaptic architecture and plasticity. Preclinical studies revealed that pharmacological interventions restore those deficits, which are thought to mediate the FXS cognitive and behavioral symptoms. Here, we characterized the de novo rate of protein synthesis in patients with FXS and their relationship with clinical severity. We measured the rate of protein synthesis in fibroblasts derived from 32 individuals with FXS and from 17 controls as well as in fibroblasts and primary neurons of 27 Fmr1 KO mice and 20 controls. Here, we show that levels of protein synthesis are increased in fibroblasts of individuals with FXS and Fmr1 KO mice. However, this cellular phenotype displays a broad distribution and a proportion of fragile X individuals and Fmr1 KO mice do not show increased levels of protein synthesis, having measures in the normal range. Because the same Fmr1 KO animal measures in fibroblasts predict those in neurons we suggest the validity of this peripheral biomarker. Our study offers a potential explanation for the comprehensive drug development program undertaken thus far yielding negative results and suggests that a significant proportion, but not all individuals with FXS, may benefit from the reduction of excessive levels of protein synthesis.


Assuntos
Transtorno do Espectro Autista/genética , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Adolescente , Adulto , Idoso , Animais , Transtorno do Espectro Autista/fisiopatologia , Criança , Modelos Animais de Doenças , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteína do X Frágil da Deficiência Intelectual/biossíntese , Síndrome do Cromossomo X Frágil/fisiopatologia , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Humanos , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Neurônios/metabolismo , Neurônios/patologia , Adulto Jovem
5.
Neuroimage ; 191: 392-402, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30807820

RESUMO

Previous work has demonstrated that neuroimaging biomarkers which capture functional connectivity of the brain can be used to define a specific and robust endophenotype in Fmr1-/y mice, a well-established animal model of human Fragile-X Syndrome (FXS). However, it is currently unknown whether this macroscopic measure of brain connectivity is sufficiently sensitive to reliably detect changes caused by pharmacological interventions. Here we inhibited the activity of the metabotropic glutamate receptor-5 (mGluR5) using AFQ056/Mavoglurant, a drug that is assumed to normalize excitatory/inhibitory neural signaling imbalances in FXS. We employed resting-state-fMRI (rs-fMRI) and diffusion-weighted imaging (DWI) to test whether Mavoglurant re-established brain connectivity - at least partly - within some of the affected circuits in Fmr1-/y mice that are related to social behavior deficits. In line with previous findings, we observed that Fmr1-/y mice exhibited impaired social interaction, reduced connectivity in three main functional networks and altered network topology. At the group level, Mavoglurant did neither rescue abnormal social behavioral nor white matter abnormalities; however, for some, but not all of these circuits Mavoglurant had a genotype-specific effect of restoring functional connectivity. These results show that rs-fMRI connectivity is sufficiently sensitive to pick up system-level changes after the pharmacological inhibition of mGluR5 activity. However, our results also show that the effects of Mavoglurant are confined to specific networks suggesting that behavioral benefits might be restricted to narrow functional domains.


Assuntos
Encéfalo/efeitos dos fármacos , Indóis/farmacologia , Vias Neurais/efeitos dos fármacos , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/metabolismo , Neuroimagem/métodos
6.
PLoS Pathog ; 13(11): e1006733, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29176838

RESUMO

Prion infections cause inexorable, progressive neurological dysfunction and neurodegeneration. Expression of the cellular prion protein PrPC is required for toxicity, suggesting the existence of deleterious PrPC-dependent signaling cascades. Because group-I metabotropic glutamate receptors (mGluR1 and mGluR5) can form complexes with the cellular prion protein (PrPC), we investigated the impact of mGluR1 and mGluR5 inhibition on prion toxicity ex vivo and in vivo. We found that pharmacological inhibition of mGluR1 and mGluR5 antagonized dose-dependently the neurotoxicity triggered by prion infection and by prion-mimetic anti-PrPC antibodies in organotypic brain slices. Prion-mimetic antibodies increased mGluR5 clustering around dendritic spines, mimicking the toxicity of Aß oligomers. Oral treatment with the mGluR5 inhibitor, MPEP, delayed the onset of motor deficits and moderately prolonged survival of prion-infected mice. Although group-I mGluR inhibition was not curative, these results suggest that it may alleviate the neurological dysfunctions induced by prion diseases.


Assuntos
Proteínas PrPC/toxicidade , Doenças Priônicas/tratamento farmacológico , Doenças Priônicas/metabolismo , Piridinas/administração & dosagem , Receptor de Glutamato Metabotrópico 5/metabolismo , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Animais , Anticorpos/administração & dosagem , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteínas PrPC/genética , Proteínas PrPC/metabolismo , Doenças Priônicas/genética , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Receptor de Glutamato Metabotrópico 5/genética , Receptores de Glutamato Metabotrópico/genética , Receptores de Glutamato Metabotrópico/metabolismo
7.
J Neuroinflammation ; 13(1): 189, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27549131

RESUMO

BACKGROUND: The fractalkine (CX3CR1) ligand is expressed in astrocytes and reported to be neuroprotective. When cleaved from the membrane, soluble fractalkine (sCX3CL1) activates the receptor CX3CR1. Although somewhat controversial, CX3CR1 is reported to be expressed in neurons and microglia. The membrane-bound form of CX3CL1 additionally acts as an adhesion molecule for microglia and infiltrating white blood cells. Much research has been done on the role of fractalkine in neuronal cells; however, little is known about the regulation of the CX3CL1 ligand in astrocytes. METHODS: The mechanisms involved in the up-regulation and cleavage of CX3CL1 from human astrocytes were investigated using immunocytochemistry, Q-PCR and ELISA. All statistical analysis was performed using GraphPad Prism 5. RESULTS: A combination of ADAM17 (TACE) and ADAM10 protease inhibitors was found to attenuate IL-1ß-, TNF-α- and IFN-γ-induced sCX3CL1 levels in astrocytes. A specific ADAM10 (but not ADAM17) inhibitor also attenuated these effects, suggesting ADAM10 proteases induce release of sCX3CL1 from stimulated human astrocytes. A p38 MAPK inhibitor also attenuated the levels of sCX3CL1 upon treatment with IL-1ß, TNF-α or IFN-γ. In addition, an IKKß inhibitor significantly reduced the levels of sCX3CL1 induced by IL-1ß or TNF-α in a concentration-dependent manner, suggesting a role for the NF-kB pathway. CONCLUSIONS: In conclusion, this study shows that the release of soluble astrocytic fractalkine is regulated by ADAM10 proteases with p38 MAPK also playing a role in the fractalkine shedding event. These findings are important for understanding the role of CX3CL1 in healthy and stimulated astrocytes and may benefit our understanding of this pathway in neuro-inflammatory and neurodegenerative diseases.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Quimiocina CX3CL1/metabolismo , Citocinas/farmacologia , Proteínas de Membrana/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Células Cultivadas , Quimiocina CX3CL1/genética , Meios de Cultura Livres de Soro/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Metaloproteinases da Matriz/metabolismo , RNA Mensageiro/metabolismo , Fatores de Tempo
9.
Mov Disord ; 30(3): 427-31, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25689146

RESUMO

BACKGROUND: This study investigated the hypothesis that AFQ056 (mavoglurant), a selective metabotropic glutamate receptor 5 antagonist, reduces chorea in Huntington's disease (HD). METHODS: This 32-day randomized, double-blind, parallel-group, proof-of-concept study investigated AFQ056 (25-150 mg [incremental doses], twice-daily) versus placebo in patients with HD. Primary efficacy assessments were the chorea-sum score and orientation index (nondominant hand) from the quantitative motor (Q-Motor) grasping task at day 28. Key secondary efficacy assessments included finger-tapping in the Unified Huntington's Disease Rating Scale-Total Motor Score and Q-Motor measures. Safety and tolerability were assessed. RESULTS: Overall, 42 patients were randomized. At day 28, no improvement was observed on the primary efficacy assessments (P > 0.10) with AFQ056 versus placebo. The Q-Motor speeded-tapping interonset interval variability was reduced with AFQ056 versus placebo for the nondominant hand (P = 0.01). The incidence of adverse events was 66.7% with AFQ056 and 57.1% with placebo. CONCLUSIONS: AFQ056 did not reduce choreatic movements in HD, but was well tolerated. The clinical relevance of the Q-Motor findings (speeded-tapping) are unknown and may warrant further investigation.


Assuntos
Coreia/tratamento farmacológico , Coreia/etiologia , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Doença de Huntington/complicações , Indóis/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Avaliação da Deficiência , Método Duplo-Cego , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento
10.
Synapse ; 68(12): 565-573, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25098663

RESUMO

Fenobam is a negative allosteric modulator of the metabotropic glutamate receptor subtype 5 (mGluR5) with inverse agonist activity and is expected to contribute to the treatment of neuropsychiatric disorders involving dysfunction of mGluR5 including Fragile X syndrome. This study examined whether [11 C]ABP688, an antagonist PET radioligand, competes with fenobam for the same binding site in the nonhuman primate brain and would allow examination of occupancy-plasma concentration relationships in the evaluation of the drug for target disorders in the human brain. Four paired PET studies with [11 C]ABP688 were performed in baboons at a baseline condition and after intravenous treatment with fenobam at different dose levels (0.3-1.33 mg/kg). Total distribution volume (VT ) and binding potential (BPND ) using the cerebellum as a reference region were obtained by the plasma reference graphical method. Then it was examined whether occupancy follows a dose-dependent, saturating pattern that was predicted by a modified first-order Hill equation in individual regions. Baseline regional VT and BPND values agreed with previously published data. Occupancy showed dose-dependent and saturating patterns in individual regions, reaching >90% occupancy at 1.33 mg/kg dose of fenobam in the majority of regions. To our knowledge, this is the first use of PET to characterize the mGluR5 therapeutic drug fenobam. This study demonstrates a proof of principle for determining the in vivo occupancy of fenobam in primates. The results indicate that [11 C]ABP688 and PET may be useful for examination of occupancy of mGluR5 by fenobam, which should prove to be useful for designing future studies and treatment of human disease states. Synapse 68:565-573, 2014. © 2014 Wiley Periodicals, Inc.

11.
Bioorg Med Chem ; 22(21): 5790-803, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25316499

RESUMO

Here we describe the identification, structure-activity relationship and the initial pharmacological characterization of AFQ056/mavoglurant, a structurally novel, non-competitive mGlu5 receptor antagonist. AFQ056/mavoglurant was identified by chemical derivatization of a lead compound discovered in a HTS campaign. In vitro, AFQ056/mavoglurant had an IC50 of 30 nM in a functional assay with human mGluR5 and was selective over the other mGluR subtypes, iGluRs and a panel of 238 CNS relevant receptors, transporter or enzymes. In vivo, AFQ056/mavoglurant showed an improved pharmacokinetic profile in rat and efficacy in the stress-induced hyperthermia test in mice as compared to the prototypic mGluR5 antagonist MPEP. The efficacy of AFQ056/mavoglurant in humans has been assessed in L-dopa induced dyskinesia in Parkinson's disease and Fragile X syndrome in proof of principle clinical studies.


Assuntos
Indóis/química , Receptor de Glutamato Metabotrópico 5/antagonistas & inibidores , Animais , Encéfalo/metabolismo , Discinesia Induzida por Medicamentos/tratamento farmacológico , Meia-Vida , Ensaios de Triagem em Larga Escala , Humanos , Hipertermia Induzida , Indóis/farmacocinética , Indóis/farmacologia , Indóis/uso terapêutico , Levodopa/toxicidade , Masculino , Camundongos , Ligação Proteica/efeitos dos fármacos , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Receptor de Glutamato Metabotrópico 5/metabolismo , Relação Estrutura-Atividade
12.
BMC Med Genet ; 13: 13, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22397687

RESUMO

BACKGROUND: Fragile X syndrome (FXS), the leading cause of inherited mental retardation, is due to expansion and methylation of a CGG sequence in the FMR1 gene, which result in its silencing and consequent absence of FMRP protein. This absence causes loss of repression of metabotropic glutamate receptor 5 (mGluR5)-mediated pathways resulting in the behavioral and cognitive impairments associated with FXS. In a randomized, double-blind trial it was recently demonstrated a beneficial effect of AFQ056, a selective inhibitor of metabotrobic glutamate receptor type 5 (mGluR5), on fully methylated FXS patients respect to partially methylated FXS ones. METHODS: To determine whether AFQ056 may have secondary effects on the methylation and transcription of FMR1, here we treated three FXS lymphoblastoid cell lines and one normal control male line. A quantitative RT-PCR was performed to assess transcriptional reactivation of the FMR1 gene. To assess the methylation status of the FMR1 gene promoter it was carried out a bisulphite sequencing analysis. RESULTS: Both FMR1-mRNA levels and DNA methylation were unmodified with respect to untreated controls. CONCLUSIONS: These results demonstrate that the AFQ056 effect on fully methylated FXS patients is not due to a secondary effect on DNA methylation and consequent transcriptional activation of FMR1.


Assuntos
Metilação de DNA/efeitos dos fármacos , Proteína do X Frágil da Deficiência Intelectual/genética , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Linhagem Celular , Síndrome do Cromossomo X Frágil , Humanos , Masculino , Mutação , Receptor de Glutamato Metabotrópico 5
13.
Addict Biol ; 17(2): 235-47, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21392179

RESUMO

Growing evidence supports a role for the central nervous system (CNS) neurotransmitter L-glutamate and its metabotropic receptors (mGluRs) in drug addiction in general and alcohol-use disorders in particular. Alcohol dependence, for instance, has a genetic component, and the recent discovery that variations in the gene coding for mGluR7 modulate alcohol consumption further validates involvement of the L-glutamate system. Consequently, increasing interest emerges in developing L-glutamatergic therapies for the treatment of alcohol abuse and dependence. To this end, we performed a detailed behavioral pharmacology study to investigate the regulation of alcohol consumption and preference following administration of the mGluR7-selective drugs N,N'-dibenzyhydryl-ethane-1,2-diamine dihydrochloride (AMN082) and 6-(4-Methoxyphenyl)-5-methyl-3-(4-pyridinyl)-isoxazolo[4,5-c]pyridin-4(5H)-one hydrochloride (MMPIP). Upon administration of the allosteric agonist AMN082 (10 mg/kg, i.p.) in rats, there was a significant decrease in ethanol consumption and preference, without affecting ethanol blood metabolism. In contrast, mGluR7 blockade with MMPIP (10 mg/kg, i.p.) showed an increase in alcohol intake and reversed AMN082's effect on ethanol consumption and preference. Both mGluR7-directed pharmacological tools had no effect on total fluid intake, taste preference, or on spontaneous locomotor activity. In conclusion, these findings support a specific regulatory role for mGluR7 on alcohol drinking and preference and provide evidence for the use of AMN082-type drugs as potential new treatments for alcohol-use disorders in man.


Assuntos
Consumo de Bebidas Alcoólicas/tratamento farmacológico , Compostos Benzidrílicos/farmacologia , Piridonas/farmacologia , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Consumo de Bebidas Alcoólicas/psicologia , Animais , Etanol/metabolismo , Preferências Alimentares/efeitos dos fármacos , Masculino , Atividade Motora/efeitos dos fármacos , Quinina/farmacologia , Ratos , Ratos Wistar , Sacarina/farmacologia , Autoadministração , Edulcorantes/farmacologia , Paladar/efeitos dos fármacos
14.
Cell Rep ; 39(9): 110883, 2022 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-35649351

RESUMO

TREM2 is a transmembrane protein expressed exclusively in microglia in the brain that regulates inflammatory responses to pathological conditions. Proteolytic cleavage of membrane TREM2 affects microglial function and is associated with Alzheimer's disease, but the consequence of reduced TREM2 proteolytic cleavage has not been determined. Here, we generate a transgenic mouse model of reduced Trem2 shedding (Trem2-Ile-Pro-Asp [IPD]) through amino-acid substitution of an ADAM-protease recognition site. We show that Trem2-IPD mice display increased Trem2 cell-surface-receptor load, survival, and function in myeloid cells. Using single-cell transcriptomic profiling of mouse cortex, we show that sustained Trem2 stabilization induces a shift of fate in microglial maturation and accelerates microglial responses to Aß pathology in a mouse model of Alzheimer's disease. Our data indicate that reduction of Trem2 proteolytic cleavage aggravates neuroinflammation during the course of Alzheimer's disease pathology, suggesting that TREM2 shedding is a critical regulator of microglial activity in pathological states.


Assuntos
Doença de Alzheimer , Glicoproteínas de Membrana , Microglia , Receptores Imunológicos , Doença de Alzheimer/metabolismo , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo
15.
Neurobiol Dis ; 42(3): 311-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21316452

RESUMO

Fragile X syndrome, the most common form of inherited intellectual disability, is caused by a lack of FMRP, which is the product of the Fmr1 gene. FMRP is an RNA-binding protein and a component of RNA-granules found in the dendrites of neurons. At the synapse, FMRP is involved in regulation of translation of specific target mRNAs upon stimulation of mGluR5 receptors. In this study, we test the effects of a new mGluR5 antagonist (AFQ056) on the prepulse inhibition of startle response in mice. We show that Fmr1 KO mice have a deficit in inhibition of the startle response after a prepulse and that AFQ056 can rescue this phenotype. We also studied the effect of AFQ056 on cultured Fmr1 KO hippocampal neurons; untreated neurons showed elongated spines and treatment resulted in shortened spines. These results suggest that AFQ056 might be a potent mGluR5 antagonist to rescue various aspects of the fragile X phenotype.


Assuntos
Hipocampo/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Reflexo de Sobressalto/efeitos dos fármacos , Filtro Sensorial/efeitos dos fármacos , Animais , Células Cultivadas , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Hipocampo/metabolismo , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Receptor de Glutamato Metabotrópico 5
16.
Mov Disord ; 26(7): 1243-50, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21484867

RESUMO

Study objectives were to assess the efficacy, safety, and tolerability of AFQ056 in Parkinson's disease patients with levodopa-induced dyskinesia. Two randomized, double-blind, placebo-controlled, parallel-group, in-patient studies for Parkinson's disease patients with moderate to severe levodopa-induced dyskinesia (study 1) and severe levodopa-induced dyskinesia (study 2) on stable dopaminergic therapy were performed. Patients received 25-150 mg AFQ056 or placebo twice daily for 16 days (both studies). Study 2 included a 4-day down-titration. Primary outcomes were the Lang-Fahn Activities of Daily Living Dyskinesia Scale (study 1), the modified Abnormal Involuntary Movement Scale (study 2), and the Unified Parkinson's Disease Rating Scale-part III (both studies). Secondary outcomes included the Unified Parkinson's Disease Rating Scale-part IV items 32-33. The primary analysis was change from baseline to day 16 on all outcomes. Treatment differences were assessed. Fifteen patients were randomized to AFQ056 and 16 to placebo in study 1; 14 patients were randomized to each group in study 2. AFQ056-treated patients showed significant improvements in dyskinesias on day 16 versus placebo (eg, Lang-Fahn Activities of Daily Living Dyskinesia Scale, P = .021 [study 1]; modified Abnormal Involuntary Movement Scale, P = .032 [study 2]). No significant changes were seen from baseline on day 16 on the Unified Parkinson's Disease Rating Scale-part III in either study. Adverse events were reported in both studies, including dizziness. Serious adverse events (most commonly worsening of dyskinesias, apparently associated with stopping treatment) were reported by 4 AFQ056-treated patients in study 1, and 3 patients (2 AFQ056-treated patient and 1 in the placebo group) in study 2. AFQ056 showed a clinically relevant and significant antidyskinetic effect without changing the antiparkinsonian effects of dopaminergic therapy. © 2011 Movement Disorder Society.


Assuntos
Discinesia Induzida por Medicamentos/tratamento farmacológico , Antagonistas de Aminoácidos Excitatórios/administração & dosagem , Levodopa/efeitos adversos , Doença de Parkinson/tratamento farmacológico , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Antiparkinsonianos/efeitos adversos , Interações Medicamentosas , Antagonistas de Aminoácidos Excitatórios/efeitos adversos , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Receptor de Glutamato Metabotrópico 5 , Resultado do Tratamento
17.
J Neurochem ; 113(3): 715-24, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20132464

RESUMO

Behavioral investigations of selective and potent metabotropic glutamate receptor type 5 (mGluR5) antagonists in animal models suggest involvement of mGluR5 in compensatory mechanisms of the basal ganglia circuitry in Parkinson's disease and levodopa (L-Dopa) induced motor complications. This study investigated mGluR5 changes in MPTP lesioned monkeys. The effect of a chronic 1 month treatment with L-Dopa on mGluR5-specific binding and mRNA levels was investigated in MPTP monkeys killed 4 or 24 h after their last L-Dopa administration. [(3)H]ABP688 specific binding in the putamen was elevated in L-Dopa-treated MPTP monkeys killed 24 h but not 4 h after their last L-Dopa dose compared with vehicle-treated MPTP monkeys. Caudate nucleus [(3)H]ABP688-specific binding was elevated in both groups of L-Dopa treated compared with vehicle-treated MPTP monkeys. In contrast, caudate nucleus and putamen mGluR5 mRNA levels were elevated only in L-Dopa-treated MPTP monkeys killed 4 h after their last L-Dopa administration. MPTP monkeys killed 4 h after their last L-Dopa treatment showed higher caudate nucleus and putamen L-Dopa concentrations compared with those killed after 24 h. Hence, mGluR5 in the putamen are sensitive to presence of L-Dopa leading to a rapid decrease of [(3)H]ABP688-specific binding possibly involving a direct mGluR5/dopamine receptors interaction.


Assuntos
Antiparkinsonianos/farmacologia , Química Encefálica/efeitos dos fármacos , Levodopa/farmacologia , Intoxicação por MPTP/metabolismo , Doença de Parkinson Secundária/metabolismo , Receptores de Glutamato Metabotrópico/biossíntese , Animais , Autorradiografia , Aminas Biogênicas/metabolismo , Catecolaminas/metabolismo , Núcleo Caudado/efeitos dos fármacos , Núcleo Caudado/metabolismo , Interpretação Estatística de Dados , Proteínas da Membrana Plasmática de Transporte de Dopamina/metabolismo , Feminino , Hibridização In Situ , Macaca fascicularis , Ovariectomia , Oximas/farmacologia , Putamen/efeitos dos fármacos , Putamen/metabolismo , Piridinas/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Reserpina/farmacologia
18.
Curr Opin Drug Discov Devel ; 11(5): 655-65, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18729017

RESUMO

Disturbances of glutamate-mediated neurotransmission have been implicated in a broad range of nervous system disorders. Numerous attempts to correct nervous system dysfunction by pharmacological intervention at glutamate receptors have been made, and some of the approaches have achieved a high level of preclinical validation. However, in a number of cases involving agents acting as blockers of the ionotropic glutamate receptors, clinical success could not be achieved, mostly because of the lack of a therapeutic window. The identification of the metabotropic glutamate receptor (mGluR) family and their modulatory role in the control of neurotransmission provided a new means to alter glutamatergic transmission. Furthermore, selective agents acting as allosteric antagonists at the mGluR5 subtype have demonstrated therapeutic potential. The identification and characterization of mGluR5 antagonists and recent progress in clinical development are summarized.


Assuntos
Desenho de Fármacos , Antagonistas de Aminoácidos Excitatórios/uso terapêutico , Doenças do Sistema Nervoso/tratamento farmacológico , Receptores de Glutamato Metabotrópico/antagonistas & inibidores , Animais , Antagonistas de Aminoácidos Excitatórios/efeitos adversos , Antagonistas de Aminoácidos Excitatórios/química , Ácido Glutâmico/metabolismo , Humanos , Ligantes , Estrutura Molecular , Doenças do Sistema Nervoso/diagnóstico por imagem , Doenças do Sistema Nervoso/metabolismo , Tomografia por Emissão de Pósitrons , Conformação Proteica , Receptor de Glutamato Metabotrópico 5 , Receptores de Glutamato Metabotrópico/química , Receptores de Glutamato Metabotrópico/metabolismo , Relação Estrutura-Atividade
19.
Nat Rev Drug Discov ; 17(4): 280-299, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29217836

RESUMO

Neurodevelopmental disorders such as fragile X syndrome (FXS) result in lifelong cognitive and behavioural deficits and represent a major public health burden. FXS is the most frequent monogenic form of intellectual disability and autism, and the underlying pathophysiology linked to its causal gene, FMR1, has been the focus of intense research. Key alterations in synaptic function thought to underlie this neurodevelopmental disorder have been characterized and rescued in animal models of FXS using genetic and pharmacological approaches. These robust preclinical findings have led to the implementation of the most comprehensive drug development programme undertaken thus far for a genetically defined neurodevelopmental disorder, including phase IIb trials of metabotropic glutamate receptor 5 (mGluR5) antagonists and a phase III trial of a GABAB receptor agonist. However, none of the trials has been able to unambiguously demonstrate efficacy, and they have also highlighted the extent of the knowledge gaps in drug development for FXS and other neurodevelopmental disorders. In this Review, we examine potential issues in the previous studies and future directions for preclinical and clinical trials. FXS is at the forefront of efforts to develop drugs for neurodevelopmental disorders, and lessons learned in the process will also be important for such disorders.


Assuntos
Síndrome do Cromossomo X Frágil/tratamento farmacológico , Transtornos do Neurodesenvolvimento/tratamento farmacológico , Neurotransmissores/farmacologia , Neurotransmissores/uso terapêutico , Animais , Ensaios Clínicos como Assunto , Desenvolvimento de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
20.
J Nucl Med ; 48(7): 1207-15, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17574984

RESUMO

UNLABELLED: (11)C-ABP688 is a new PET ligand to assess the subtype 5 metabotropic glutamate receptor (mGlu(5)). The purpose of this study was to evaluate different methods for the analysis of human (11)C-ABP688 data acquired from 6 healthy, young volunteers. METHODS: The methods were a 1-tissue-compartment model (K(1), k(2)''), a 2-tissue-compartment model (K1-k4), and the noncompartmental method developed by Logan. Parameters related to receptor density were the total distribution volume (DV), DV'' (= K(1)/k(2)'', 1 tissue compartment); specific DV, DV(C2) (= K(1)/k(2)' x k(3)'/k(4), 2 tissue compartments); and DV(tot) for the noncompartmental method. RESULTS: The 1-tissue-compartment model was too simple to adequately fit the data. DV(C2) calculated with the 2-tissue-compartment model ranged from 5.45 +/- 1.47 (anterior cingulate) to 1.91 +/- 0.32 (cerebellum). The corresponding values for DV(tot), calculated with the 2-tissue-compartment model and the Logan method (in parentheses), were 6.57 +/- 1.45 (6.35 +/- 1.32) and 2.93 +/- 0.53 (2.48 +/- 0.40). There was no clear evidence of a region devoid of mGlu(5) receptors. The first-pass extraction fraction exceeded 95%. The minimal scan duration to obtain stable results was estimated to be 45 min. CONCLUSION: (11)C-ABP688 displays favorable kinetics for assessing mGlu(5) receptors. For tracer kinetic modeling, 2-tissue-compartment models are clearly superior to models with only 1 tissue compartment. In comparison to the compartmental models, the Logan method is equally useful if only DV(tot) values are required and fast pixelwise parametric maps are desired. The lack of regions devoid of receptors limits the use of reference region methods that do not require arterial blood sampling. Another advantage of the tracer is the fast kinetics that allow for relatively short acquisitions.


Assuntos
Encéfalo/metabolismo , Oximas/farmacocinética , Piridinas/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Receptores de Glutamato Metabotrópico/metabolismo , Adulto , Encéfalo/diagnóstico por imagem , Radioisótopos de Carbono , Estudos de Avaliação como Assunto , Humanos , Imageamento por Ressonância Magnética , Masculino , Modelos Biológicos , Tomografia por Emissão de Pósitrons/métodos , Receptor de Glutamato Metabotrópico 5
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA