Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mov Disord ; 38(7): 1336-1340, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37093618

RESUMO

BACKGROUND: Multiple system atrophy (MSA) is a sporadic adult-onset rare neurodegenerative synucleinopathy for which counteracting central nervous system insulin resistance bears the potential of being neuroprotective. G-protein-(heterotrimeric guanine nucleotide-binding protein)-coupled receptor kinase 2 (GRK2) is emerging as a physiologically relevant inhibitor of insulin signaling. OBJECTIVES: We tested whether lowering brain GRK2 abundance may reverse insulin-resistance. METHODS: We lowered brain GRK2 abundance through viral-mediated delivery of a GRK2-specific miRNA and quantified the reversion of a developing or an established insulin-resistant phenotype using the transgenic PLP-SYN mouse model of MSA. RESULTS: Viral vector delivery of a GRK2 miRNA demonstrated a neuroprotective capacity when administered (1) in utero intracerebroventricularly in developing PLP-SYN mice and (2) intrastriatally in adult PLP-SYN mice. Decreased striatal GRK2 levels correlated in both designs with neuroprotection of the substantia nigra dopamine neurons, reduction in high-molecular-weight species of α-synuclein, and reduced insulin resistance. CONCLUSIONS: These data support GRK2 as a potential therapeutic target in MSA. © 2023 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Resistência à Insulina , Insulinas , MicroRNAs , Transtornos dos Movimentos , Atrofia de Múltiplos Sistemas , Camundongos , Animais , Atrofia de Múltiplos Sistemas/terapia , Atrofia de Múltiplos Sistemas/tratamento farmacológico , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Camundongos Transgênicos , Insulinas/uso terapêutico , Modelos Animais de Doenças
2.
Hum Mol Genet ; 27(12): 2138-2153, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29659809

RESUMO

The aristaless-related homeobox (ARX) transcription factor is involved in the development of GABAergic and cholinergic neurons in the forebrain. ARX mutations have been associated with a wide spectrum of neurodevelopmental disorders in humans, among which the most frequent, a 24 bp duplication in the polyalanine tract 2 (c.428_451dup24), gives rise to intellectual disability, fine motor defects with or without epilepsy. To understand the functional consequences of this mutation, we generated a partially humanized mouse model carrying the c.428_451dup24 duplication (Arxdup24/0) that we characterized at the behavior, neurological and molecular level. Arxdup24/0 males presented with hyperactivity, enhanced stereotypies and altered contextual fear memory. In addition, Arxdup24/0 males had fine motor defects with alteration of reaching and grasping abilities. Transcriptome analysis of Arxdup24/0 forebrains at E15.5 showed a down-regulation of genes specific to interneurons and an up-regulation of genes normally not expressed in this cell type, suggesting abnormal interneuron development. Accordingly, interneuron migration was altered in the cortex and striatum between E15.5 and P0 with consequences in adults, illustrated by the defect in the inhibitory/excitatory balance in Arxdup24/0 basolateral amygdala. Altogether, we showed that the c.428_451dup24 mutation disrupts Arx function with a direct consequence on interneuron development, leading to hyperactivity and defects in precise motor movement control and associative memory. Interestingly, we highlighted striking similarities between the mouse phenotype and a cohort of 33 male patients with ARX c.428_451dup24, suggesting that this new mutant mouse line is a good model for understanding the pathophysiology and evaluation of treatment.


Assuntos
Epilepsia/genética , Proteínas de Homeodomínio/genética , Transtornos do Neurodesenvolvimento/genética , Fatores de Transcrição/genética , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Neurônios Colinérgicos/metabolismo , Neurônios Colinérgicos/patologia , Contratura , Modelos Animais de Doenças , Epilepsia/fisiopatologia , Neurônios GABAérgicos/metabolismo , Neurônios GABAérgicos/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Lactente , Deficiência Intelectual , Masculino , Camundongos , Mutação , Transtornos do Neurodesenvolvimento/fisiopatologia , Peptídeos/genética , Prosencéfalo/fisiopatologia , Paraplegia Espástica Hereditária , Transcriptoma/genética , Adulto Jovem
3.
J Neurosci ; 37(46): 11114-11126, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29030432

RESUMO

Classical and systems genetics have identified wide networks of genes associated with cognitive and neurodevelopmental diseases. In parallel to deciphering the role of each of these genes in neuronal or synaptic function, evaluating the response of neuronal and molecular networks to gene loss of function could reveal some pathophysiological mechanisms potentially accessible to nongenetic therapies. Loss of function of the Rho-GAP oligophrenin-1 is associated with cognitive impairments in both human and mouse. Upregulation of both PKA and ROCK has been reported in Ophn1-/y mice, but it remains unclear whether kinase hyperactivity contributes to the behavioral phenotypes. In this study, we thoroughly characterized a prominent perseveration phenotype displayed by Ophn1-deficient mice using a Y-maze spatial working memory (SWM) test. We report that Ophn1 deficiency in the mouse generated severe cognitive impairments, characterized by both a high occurrence of perseverative behaviors and a lack of deliberation during the SWM test. In vivo and in vitro pharmacological experiments suggest that PKA dysregulation in the mPFC underlies cognitive dysfunction in Ophn1-deficient mice, as assessed using a delayed spatial alternation task results. Functionally, mPFC neuronal networks appeared to be affected in a PKA-dependent manner, whereas hippocampal-PFC projections involved in SWM were not affected in Ophn1-/y mice. Thus, we propose that discrete gene mutations in intellectual disability might generate "secondary" pathophysiological mechanisms, which are prone to become pharmacological targets for curative strategies in adult patients.SIGNIFICANCE STATEMENT Here we report that Ophn1 deficiency generates severe impairments in performance at spatial working memory tests, characterized by a high occurrence of perseverative behaviors and a lack of decision making. This cognitive deficit is consecutive to PKA deregulation in the mPFC that prevents Ophn1 KO mice to exploit a correctly acquired rule. Functionally, mPFC neuronal networks appear to be affected in a PKA-dependent manner, whereas behaviorally important hippocampal projections were preserved by the mutation. Thus, we propose that discrete gene mutations in intellectual disability can generate "secondary" pathophysiological mechanisms prone to become pharmacological targets for curative strategies in adults.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteínas do Citoesqueleto/deficiência , Proteínas Ativadoras de GTPase/deficiência , Transtornos da Memória/metabolismo , Memória de Curto Prazo/fisiologia , Proteínas Nucleares/deficiência , Córtex Pré-Frontal/metabolismo , Animais , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Rede Nervosa/metabolismo , Rede Nervosa/fisiopatologia , Técnicas de Cultura de Órgãos , Córtex Pré-Frontal/fisiopatologia , Distribuição Aleatória
4.
Hum Mol Genet ; 24(4): 1106-18, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25305082

RESUMO

Mutations in interleukin-1 receptor accessory protein like 1 (IL1RAPL1) gene have been associated with non-syndromic intellectual disability (ID) and autism spectrum disorder. This protein interacts with synaptic partners like PSD-95 and PTPδ, regulating the formation and function of excitatory synapses. The aim of this work was to characterize the synaptic consequences of three IL1RAPL1 mutations, two novel causing the deletion of exon 6 (Δex6) and one point mutation (C31R), identified in patients with ID. Using immunofluorescence and electrophysiological recordings, we examined the effects of IL1RAPL1 mutant over-expression on synapse formation and function in cultured rodent hippocampal neurons. Δex6 but not C31R mutation leads to IL1RAPL1 protein instability and mislocalization within dendrites. Analysis of different markers of excitatory synapses and sEPSC recording revealed that both mutants fail to induce pre- and post-synaptic differentiation, contrary to WT IL1RAPL1 protein. Cell aggregation and immunoprecipitation assays in HEK293 cells showed a reduction of the interaction between IL1RAPL1 mutants and PTPδ that could explain the observed synaptogenic defect in neurons. However, these mutants do not affect all cellular signaling because their over-expression still activates JNK pathway. We conclude that both mutations described in this study lead to a partial loss of function of the IL1RAPL1 protein through different mechanisms. Our work highlights the important function of the trans-synaptic PTPδ/IL1RAPL1 interaction in synaptogenesis and as such in ID in the patients.


Assuntos
Deficiência Intelectual/genética , Proteína Acessória do Receptor de Interleucina-1/genética , Mutação , Neurogênese/genética , Sinapses/genética , Adulto , Criança , Pré-Escolar , Análise Mutacional de DNA , Éxons , Feminino , Humanos , Deficiência Intelectual/metabolismo , Proteína Acessória do Receptor de Interleucina-1/química , Proteína Acessória do Receptor de Interleucina-1/metabolismo , Íntrons , Masculino , Linhagem , Polimorfismo de Nucleotídeo Único , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Deleção de Sequência , Transdução de Sinais , Sinapses/metabolismo
5.
Hum Mol Genet ; 24(23): 6736-55, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26376863

RESUMO

ATP6AP2, an essential accessory component of the vacuolar H+ ATPase (V-ATPase), has been associated with intellectual disability (ID) and Parkinsonism. ATP6AP2 has been implicated in several signalling pathways; however, little is known regarding its role in the nervous system. To decipher its function in behaviour and cognition, we generated and characterized conditional knockdowns of ATP6AP2 in the nervous system of Drosophila and mouse models. In Drosophila, ATP6AP2 knockdown induced defective phototaxis and vacuolated photoreceptor neurons and pigment cells when depleted in eyes and altered short- and long-term memory when depleted in the mushroom body. In mouse, conditional Atp6ap2 deletion in glutamatergic neurons (Atp6ap2(Camk2aCre/0) mice) caused increased spontaneous locomotor activity and altered fear memory. Both Drosophila ATP6AP2 knockdown and Atp6ap2(Camk2aCre/0) mice presented with presynaptic transmission defects, and with an abnormal number and morphology of synapses. In addition, Atp6ap2(Camk2aCre/0) mice showed autophagy defects that led to axonal and neuronal degeneration in the cortex and hippocampus. Surprisingly, axon myelination was affected in our mutant mice, and axonal transport alterations were observed in Drosophila. In accordance with the identified phenotypes across species, genome-wide transcriptome profiling of Atp6ap2(Camk2aCre/0) mouse hippocampi revealed dysregulation of genes involved in myelination, action potential, membrane-bound vesicles and motor behaviour. In summary, ATP6AP2 disruption in mouse and fly leads to cognitive impairment and neurodegeneration, mimicking aspects of the neuropathology associated with ATP6AP2 mutations in humans. Our results identify ATP6AP2 as an essential gene for the nervous system.


Assuntos
Transtornos Cognitivos/etiologia , Proteínas de Drosophila/genética , Proteínas de Membrana/genética , Degeneração Neural/etiologia , ATPases Translocadoras de Prótons/genética , Receptores de Superfície Celular/genética , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Modelos Animais de Doenças , Drosophila , Feminino , Técnicas de Silenciamento de Genes , Deficiência Intelectual/genética , Masculino , Camundongos , Degeneração Neural/patologia , Neurônios/metabolismo , Neurônios/fisiologia , Neurônios/ultraestrutura , Transtornos Parkinsonianos/genética , Sinapses/metabolismo , Sinapses/fisiologia , Sinapses/ultraestrutura
6.
J Exp Med ; 204(7): 1717-27, 2007 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-17606631

RESUMO

V(D)J recombination and immunoglobulin class switch recombination (CSR) are two somatic rearrangement mechanisms that proceed through the introduction of double-strand breaks (DSBs) in DNA. Although the DNA repair factor XRCC4 is essential for the resolution of DNA DSB during V(D)J recombination, its role in CSR has not been established. To bypass the embryonic lethality of XRCC4 deletion in mice, we developed a conditional XRCC4 knockout (KO) using LoxP-flanked XRCC4 cDNA lentiviral transgenesis. B lymphocyte restricted deletion of XRCC4 in these mice lead to an average two-fold reduction in CSR in vivo and in vitro. Our results connect XRCC4 and the nonhomologous end joining DNA repair pathway to CSR while reflecting the possible use of an alternative pathway in the repair of CSR DSB in the absence of XRCC4. In addition, this new conditional KO approach should be useful in studying other lethal mutations in mice.


Assuntos
Reparo do DNA , Proteínas de Ligação a DNA/genética , Switching de Imunoglobulina , Animais , Linfócitos B/imunologia , Proteínas de Ligação a DNA/deficiência , Genes Letais , Lentivirus/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Mutação
7.
Nat Commun ; 13(1): 3102, 2022 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-35660742

RESUMO

Dopamine transmission is involved in reward processing and motor control, and its impairment plays a central role in numerous neurological disorders. Despite its strong pathophysiological relevance, the molecular and structural organization of the dopaminergic synapse remains to be established. Here, we used targeted labelling and fluorescence activated sorting to purify striatal dopaminergic synaptosomes. We provide the proteome of dopaminergic synapses with 57 proteins specifically enriched. Beyond canonical markers of dopamine neurotransmission such as dopamine biosynthetic enzymes and cognate receptors, we validated 6 proteins not previously described as enriched. Moreover, our data reveal the adhesion of dopaminergic synapses to glutamatergic, GABAergic or cholinergic synapses in structures we named "dopamine hub synapses". At glutamatergic synapses, pre- and postsynaptic markers are significantly increased upon association with dopamine synapses. Dopamine hub synapses may thus support local dopaminergic signalling, complementing volume transmission thought to be the major mechanism by which monoamines modulate network activity.


Assuntos
Dopamina , Sinapses , Animais , Corpo Estriado/fisiologia , Dopamina/metabolismo , Camundongos , Recompensa , Sinapses/metabolismo , Transmissão Sináptica/fisiologia
8.
Elife ; 92020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33252331

RESUMO

Survival depends on the ability of animals to select the appropriate behavior in response to threat and safety sensory cues. However, the synaptic and circuit mechanisms by which the brain learns to encode accurate predictors of threat and safety remain largely unexplored. Here, we show that frontal association cortex (FrA) pyramidal neurons of mice integrate auditory cues and basolateral amygdala (BLA) inputs non-linearly in a NMDAR-dependent manner. We found that the response of FrA pyramidal neurons was more pronounced to Gaussian noise than to pure frequency tones, and that the activation of BLA-to-FrA axons was the strongest in between conditioning pairings. Blocking BLA-to-FrA signaling specifically at the time of presentation of Gaussian noise (but not 8 kHz tone) between conditioning trials impaired the formation of auditory fear memories. Taken together, our data reveal a circuit mechanism that facilitates the formation of fear traces in the FrA, thus providing a new framework for probing discriminative learning and related disorders.


Assuntos
Estimulação Acústica/efeitos adversos , Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Lobo Frontal/fisiologia , Aprendizagem/fisiologia , Animais , Cálcio/metabolismo , Condicionamento Clássico/fisiologia , Masculino , Camundongos , Microscopia Confocal , Plasticidade Neuronal/fisiologia , Optogenética , Técnicas de Patch-Clamp
9.
Elife ; 82019 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-31663854

RESUMO

Glutamate secretion at excitatory synapses is tightly regulated to allow for the precise tuning of synaptic strength. Vesicular Glutamate Transporters (VGLUT) accumulate glutamate into synaptic vesicles (SV) and thereby regulate quantal size. Further, the number of release sites and the release probability of SVs maybe regulated by the organization of active-zone proteins and SV clusters. In the present work, we uncover a mechanism mediating an increased SV clustering through the interaction of VGLUT1 second proline-rich domain, endophilinA1 and intersectin1. This strengthening of SV clusters results in a combined reduction of axonal SV super-pool size and miniature excitatory events frequency. Our findings support a model in which clustered vesicles are held together through multiple weak interactions between Src homology three and proline-rich domains of synaptic proteins. In mammals, VGLUT1 gained a proline-rich sequence that recruits endophilinA1 and turns the transporter into a regulator of SV organization and spontaneous release.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Glutamatos/metabolismo , Vesículas Sinápticas/metabolismo , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo , Animais , Transporte Biológico , Humanos , Camundongos , Camundongos Knockout , Ratos , Proteína Vesicular 1 de Transporte de Glutamato/deficiência
10.
Bioorg Med Chem ; 16(11): 6218-32, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18467104

RESUMO

We describe the discovery of novel potent inhibitors of 2,3-oxidosqualene:lanosterol cyclase inhibitors (OSCi) from a focused pharmacophore-based screen. Optimization of the most tractable hits gave a series of compounds showing inhibition of cholesterol biosynthesis at 2mg/kg in the rat with distinct pharmacokinetic profiles. Two compounds were selected for toxicological study in the rat for 21 days in order to test the hypothesis that low systemic exposure could be used as a strategy to avoid the ocular side effects previously described with OSCi. We demonstrate that for this series of inhibitors, a reduction of systemic exposure is not sufficient to circumvent cataract liabilities.


Assuntos
Catarata/enzimologia , Dislipidemias/enzimologia , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacocinética , Olho/efeitos dos fármacos , Transferases Intramoleculares/antagonistas & inibidores , Animais , Anticolesterolemiantes/efeitos adversos , Anticolesterolemiantes/síntese química , Anticolesterolemiantes/farmacocinética , Catarata/induzido quimicamente , Catarata/tratamento farmacológico , Linhagem Celular Tumoral , Dislipidemias/induzido quimicamente , Inibidores Enzimáticos/efeitos adversos , Olho/metabolismo , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Oxazóis/farmacocinética , Oxazóis/uso terapêutico , Piperazinas/efeitos adversos , Piperazinas/síntese química , Piperazinas/farmacocinética , Piperidinas/farmacocinética , Piperidinas/uso terapêutico , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA