Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 26(11): 6427-6450, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33879865

RESUMO

Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) has been shown to activate the eIF2α kinase PERK to directly regulate translation initiation. Tight control of PERK-eIF2α signaling has been shown to be necessary for normal long-lasting synaptic plasticity and cognitive function, including memory. In contrast, chronic activation of PERK-eIF2α signaling has been shown to contribute to pathophysiology, including memory impairments, associated with multiple neurological diseases, making this pathway an attractive therapeutic target. Herein, using multiple genetic approaches we show that selective deletion of the PERK in mouse midbrain dopaminergic (DA) neurons results in multiple cognitive and motor phenotypes. Conditional expression of phospho-mutant eIF2α in DA neurons recapitulated the phenotypes caused by deletion of PERK, consistent with a causal role of decreased eIF2α phosphorylation for these phenotypes. In addition, deletion of PERK in DA neurons resulted in altered de novo translation, as well as changes in axonal DA release and uptake in the striatum that mirror the pattern of motor changes observed. Taken together, our findings show that proper regulation of PERK-eIF2α signaling in DA neurons is required for normal cognitive and motor function in a non-pathological state, and also provide new insight concerning the onset of neuropsychiatric disorders that accompany UPR failure.


Assuntos
Neurônios Dopaminérgicos , Fator de Iniciação 2 em Eucariotos , Animais , Cognição , Neurônios Dopaminérgicos/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático , Fator de Iniciação 2 em Eucariotos/genética , Camundongos , Fosforilação , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
2.
Proc Natl Acad Sci U S A ; 116(7): 2707-2712, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30692248

RESUMO

Inositol polyphosphate multikinase (IPMK), the key enzyme for the biosynthesis of higher inositol polyphosphates and phosphatidylinositol 3,4,5-trisphosphate, also acts as a versatile signaling player in regulating tissue growth and metabolism. To elucidate neurobehavioral functions of IPMK, we generated mice in which IPMK was deleted from the excitatory neurons of the postnatal forebrain. These mice showed no deficits in either novel object recognition or spatial memory. IPMK conditional knockout mice formed cued fear memory normally but displayed enhanced fear extinction. Signaling analyses revealed dysregulated expression of neural genes accompanied by selective activation of the mechanistic target of rapamycin (mTOR) regulatory enzyme p85 S6 kinase 1 (S6K1) in the amygdala following fear extinction. The IPMK mutants also manifested facilitated hippocampal long-term potentiation. These findings establish a signaling action of IPMK that mediates fear extinction.


Assuntos
Extinção Psicológica , Medo/psicologia , Memória , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Ativação Enzimática , Deleção de Genes , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Prosencéfalo/fisiologia , Transdução de Sinais , Regulação para Cima
3.
Mov Disord ; 36(5): 1137-1146, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33460487

RESUMO

BACKGROUND: Autophagy is intensively studied in cancer, metabolic and neurodegenerative diseases, but little is known about its role in pathological conditions linked to altered neurotransmission. We examined the involvement of autophagy in levodopa (l-dopa)-induced dyskinesia, a frequent motor complication developed in response to standard dopamine replacement therapy in parkinsonian patients. METHODS: We used mouse and non-human primate models of Parkinson's disease to examine changes in autophagy associated with chronic l-dopa administration and to establish a causative link between impaired autophagy and dyskinesia. RESULTS: We found that l-dopa-induced dyskinesia is associated with accumulation of the autophagy-specific substrate p62, a marker of autophagy deficiency. Increased p62 was observed in a subset of projection neurons located in the striatum and depended on l-dopa-mediated activation of dopamine D1 receptors, and mammalian target of rapamycin. Inhibition of mammalian target of rapamycin complex 1 with rapamycin counteracted the impairment of autophagy produced by l-dopa, and reduced dyskinesia. The anti-dyskinetic effect of rapamycin was lost when autophagy was constitutively suppressed in D1 receptor-expressing striatal neurons, through inactivation of the autophagy-related gene protein 7. CONCLUSIONS: These findings indicate that augmented responsiveness at D1 receptors leads to dysregulated autophagy, and results in the emergence of l-dopa-induced dyskinesia. They further suggest the enhancement of autophagy as a therapeutic strategy against dyskinesia. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Discinesia Induzida por Medicamentos , Transtornos Parkinsonianos , Animais , Antiparkinsonianos/toxicidade , Autofagia , Corpo Estriado , Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/tratamento farmacológico , Discinesia Induzida por Medicamentos/etiologia , Humanos , Levodopa/toxicidade , Camundongos , Oxidopamina
4.
Nature ; 493(7432): 411-5, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23263185

RESUMO

Autism spectrum disorders (ASDs) are an early onset, heterogeneous group of heritable neuropsychiatric disorders with symptoms that include deficits in social interaction skills, impaired communication abilities, and ritualistic-like repetitive behaviours. One of the hypotheses for a common molecular mechanism underlying ASDs is altered translational control resulting in exaggerated protein synthesis. Genetic variants in chromosome 4q, which contains the EIF4E locus, have been described in patients with autism. Importantly, a rare single nucleotide polymorphism has been identified in autism that is associated with increased promoter activity in the EIF4E gene. Here we show that genetically increasing the levels of eukaryotic translation initiation factor 4E (eIF4E) in mice results in exaggerated cap-dependent translation and aberrant behaviours reminiscent of autism, including repetitive and perseverative behaviours and social interaction deficits. Moreover, these autistic-like behaviours are accompanied by synaptic pathophysiology in the medial prefrontal cortex, striatum and hippocampus. The autistic-like behaviours displayed by the eIF4E-transgenic mice are corrected by intracerebroventricular infusions of the cap-dependent translation inhibitor 4EGI-1. Our findings demonstrate a causal relationship between exaggerated cap-dependent translation, synaptic dysfunction and aberrant behaviours associated with autism.


Assuntos
Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Biossíntese de Proteínas , Sinapses/metabolismo , Sinapses/patologia , Animais , Transtorno Autístico/tratamento farmacológico , Transtorno Autístico/patologia , Comportamento Animal/efeitos dos fármacos , Dendritos/metabolismo , Dendritos/patologia , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação Eucariótico 4G/metabolismo , Feminino , Hipocampo/metabolismo , Hidrazonas , Infusões Intraventriculares , Masculino , Camundongos , Camundongos Transgênicos , Neostriado/metabolismo , Plasticidade Neuronal , Nitrocompostos/administração & dosagem , Nitrocompostos/farmacologia , Nitrocompostos/uso terapêutico , Córtex Pré-Frontal/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/genética , Capuzes de RNA/metabolismo , Tiazóis/administração & dosagem , Tiazóis/farmacologia , Tiazóis/uso terapêutico
5.
Cereb Cortex ; 27(2): 1670-1685, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-26826102

RESUMO

Loss-of-function (LOF) mutations in CC2D1A cause a spectrum of neurodevelopmental disorders, including intellectual disability, autism spectrum disorder, and seizures, identifying a critical role for this gene in cognitive and social development. CC2D1A regulates intracellular signaling processes that are critical for neuronal function, but previous attempts to model the human LOF phenotypes have been prevented by perinatal lethality in Cc2d1a-deficient mice. To overcome this challenge, we generated a floxed Cc2d1a allele for conditional removal of Cc2d1a in the brain using Cre recombinase. While removal of Cc2d1a in neuronal progenitors using Cre expressed from the Nestin promoter still causes death at birth, conditional postnatal removal of Cc2d1a in the forebrain via calcium/calmodulin-dependent protein kinase II-alpha (CamKIIa) promoter-driven Cre generates animals that are viable and fertile with grossly normal anatomy. Analysis of neuronal morphology identified abnormal cortical dendrite organization and a reduction in dendritic spine density. These animals display deficits in neuronal plasticity and in spatial learning and memory that are accompanied by reduced sociability, hyperactivity, anxiety, and excessive grooming. Cc2d1a conditional knockout mice therefore recapitulate features of both cognitive and social impairment caused by human CC2D1A mutation, and represent a model that could provide much needed insights into the developmental mechanisms underlying nonsyndromic neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista/genética , Deficiência Intelectual/genética , Neurônios/citologia , Prosencéfalo/patologia , Proteínas Repressoras/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Dendritos/metabolismo , Dendritos/patologia , Modelos Animais de Doenças , Humanos , Camundongos Transgênicos , Plasticidade Neuronal/genética , Proteínas Repressoras/deficiência , Transdução de Sinais/fisiologia
6.
J Neurosci ; 36(45): 11402-11410, 2016 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-27911742

RESUMO

Autism spectrum disorder (ASD) is a constellation of neurodevelopmental presentations with high heritability and both phenotypic and genetic heterogeneity. To date, mutations in hundreds of genes have been associated to varying degrees with increased ASD risk. A better understanding of the functions of these genes and whether they fit together in functional groups or impact similar neuronal circuits is needed to develop rational treatment strategies. We will review current areas of emphasis in ASD research, starting from human genetics and exploring how mouse models of human mutations have helped identify specific molecular pathways (protein synthesis and degradation, chromatin remodeling, intracellular signaling), which are linked to alterations in circuit function and cognitive/social behavior. We will conclude by discussing how we can leverage the findings on molecular and cellular alterations found in ASD to develop therapies for neurodevelopmental disorders.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/terapia , Encéfalo/metabolismo , Terapia Genética/métodos , Proteínas do Tecido Nervoso/genética , Transtorno do Espectro Autista/diagnóstico , Medicina Baseada em Evidências , Marcadores Genéticos/genética , Predisposição Genética para Doença/genética , Testes Genéticos/métodos , Humanos , Terapia de Alvo Molecular/métodos , Proteínas do Tecido Nervoso/metabolismo , Resultado do Tratamento
7.
J Neurosci ; 35(49): 16213-20, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26658871

RESUMO

Angelman syndrome (AS) is a neurodevelopmental disorder associated with developmental delay, lack of speech, motor dysfunction, and epilepsy. In the majority of the patients, AS is caused by the deletion of small portions of maternal chromosome 15 harboring the UBE3A gene. This results in a lack of expression of the UBE3A gene because the paternal allele is genetically imprinted. The UBE3A gene encodes an enzyme termed ubiquitin ligase E3A (E6-AP) that targets proteins for degradation by the 26S proteasome. Because neurodegenerative disease and other neurodevelopmental disorders have been linked to oxidative stress, we asked whether mitochondrial reactive oxygen species (ROS) played a role in impaired synaptic plasticity and memory deficits exhibited by AS model mice. We discovered that AS mice have increased levels of superoxide in area CA1 of the hippocampus that is reduced by MitoQ 10-methanesuflonate (MitoQ), a mitochondria-specific antioxidant. In addition, we found that MitoQ rescued impairments in hippocampal synaptic plasticity and deficits in contextual fear memory exhibited by AS model mice. Our findings suggest that mitochondria-derived oxidative stress contributes to hippocampal pathophysiology in AS model mice and that targeting mitochondrial ROS pharmacologically could benefit individuals with AS. SIGNIFICANCE STATEMENT: Oxidative stress has been hypothesized to contribute to the pathophysiology of neurodevelopmental disorders, including autism spectrum disorders and Angelman syndrome (AS). Herein, we report that AS model mice exhibit elevated levels of mitochondria-derived reactive oxygen species in pyramidal neurons in hippocampal area CA1. Moreover, we demonstrate that the administration of MitoQ (MitoQ 10-methanesuflonate), a mitochondria-specific antioxidant, to AS model mice normalizes synaptic plasticity and restores memory. Finally, our findings suggest that antioxidants that target the mitochondria could be used therapeutically to ameliorate synaptic and cognitive deficits in individuals with AS.


Assuntos
Síndrome de Angelman/complicações , Hipocampo , Mitocôndrias/metabolismo , Transtornos dos Movimentos/etiologia , Transtornos dos Movimentos/patologia , Superóxidos/metabolismo , Sinapses/fisiologia , Análise de Variância , Animais , Condicionamento Psicológico , Modelos Animais de Doenças , Estimulação Elétrica , Medo , Hipocampo/metabolismo , Hipocampo/patologia , Hipocampo/ultraestrutura , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/fisiologia , Compostos Organofosforados/metabolismo , Ubiquinona/análogos & derivados , Ubiquinona/metabolismo
8.
J Neurosci ; 34(27): 9034-9, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24990923

RESUMO

Memory retrieval, often termed reconsolidation, can render previously consolidated memories susceptible to manipulation that can lead to alterations in memory strength. Although it is known that reconsolidation requires mammalian target of rapamycin complex 1 (mTORC1)-dependent translation, the specific contributions of its downstream effectors in reconsolidation are unclear. Using auditory fear conditioning in mice, we investigated the role of eukaryotic translation initiation factor 4E (eIF4E)-eIF4G interactions and p70 S6 kinase polypeptide 1 (S6K1) in reconsolidation. We found that neither 4EGI-1 (2-[(4-(3,4-dichlorophenyl)-thiazol-2-ylhydrazono)-3-(2-nitrophenyl)]propionic acid), an inhibitor of eFI4E-eIF4G interactions, nor PF-4708671 [2-((4-(5-ethylpyrimidin-4-yl)piperazin-1-yl)methyl)-5-(trifluoromethyl)-1H-benzo[d]imidazole], an inhibitor of S6K1, alone blocked the reconsolidation of auditory fear memory. In contrast, using these drugs in concert to simultaneously block eIF4E-eIF4G interactions and S6K1 immediately after memory reactivation significantly attenuated fear memory reconsolidation. Moreover, the combination of 4EGI-1 and PF-4708671 further destabilized fear memory 10 d after memory reactivation, which was consistent with experiments using rapamycin, an mTORC1 inhibitor. Furthermore, inhibition of S6K1 immediately after retrieval resulted in memory destabilization 10 d after reactivation, whereas inhibition of eIF4E-eIF4G interactions did not. These results indicate that the reconsolidation of fear memory requires concomitant association of eIF4E to eIF4G as well as S6K1 activity and that the persistence of memory at longer intervals after memory reactivation also requires mTORC1-dependent processes that involve S6K1. These findings suggest a potential mechanism for how mTORC1-dependent translation is fine tuned to alter memory persistence.


Assuntos
Aprendizagem da Esquiva/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Rememoração Mental/fisiologia , Complexos Multiproteicos/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Estimulação Acústica , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Sinais (Psicologia) , Eletrochoque , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/fisiologia , Fator de Iniciação Eucariótico 4G/antagonistas & inibidores , Fator de Iniciação Eucariótico 4G/fisiologia , Hidrazonas , Imidazóis/farmacologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Memória de Longo Prazo/efeitos dos fármacos , Memória de Longo Prazo/fisiologia , Rememoração Mental/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrocompostos/farmacologia , Piperazinas/farmacologia , Proteínas Quinases S6 Ribossômicas 90-kDa/deficiência , Proteínas Quinases S6 Ribossômicas 90-kDa/fisiologia , Sirolimo/farmacologia , Tiazóis/farmacologia
9.
Neurobiol Dis ; 83: 67-74, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26306459

RESUMO

Autism spectrum disorder (ASD) is a group of heritable disorders with complex and unclear etiology. Classic ASD symptoms include social interaction and communication deficits as well as restricted, repetitive behaviors. In addition, ASD is often comorbid with intellectual disability. Fragile X syndrome (FXS) is the leading genetic cause of ASD, and is the most commonly inherited form of intellectual disability. Several mouse models of ASD and FXS exist, however the intellectual disability observed in ASD patients is not well modeled in mice. Using the Fmr1 knockout mouse and the eIF4E transgenic mouse, two previously characterized mouse models of fragile X syndrome and ASD, respectively, we generated the eIF4E/Fmr1 double mutant mouse. Our study shows that the eIF4E/Fmr1 double mutant mice display classic ASD behaviors, as well as cognitive dysfunction. Importantly, the learning impairments displayed by the double mutant mice spanned multiple cognitive tasks. Moreover, the eIF4E/Fmr1 double mutant mice display increased levels of basal protein synthesis. The results of our study suggest that the eIF4E/Fmr1 double mutant mouse may be a reliable model to study cognitive dysfunction in the context of ASD.


Assuntos
Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/psicologia , Transtornos Cognitivos/genética , Modelos Animais de Doenças , Fator de Iniciação 4E em Eucariotos/fisiologia , Proteína do X Frágil da Deficiência Intelectual/fisiologia , Memória/fisiologia , Animais , Ansiedade/genética , Comportamento Animal/fisiologia , Condicionamento Clássico/fisiologia , Fator de Iniciação 4E em Eucariotos/genética , Medo/fisiologia , Proteína do X Frágil da Deficiência Intelectual/genética , Hipocampo/metabolismo , Relações Interpessoais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação
10.
bioRxiv ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38559006

RESUMO

The substantia nigra pars reticulata (SNr), a crucial basal ganglia output nucleus, contains a dense expression of dopamine D1 receptors (D1Rs), along with dendrites belonging to dopaminergic neurons of substantia nigra pars compacta. These D1Rs are primarily located on the terminals of striatonigral medium spiny neurons, suggesting their involvement in the regulation of neurotransmitter release from the direct pathway in response to somatodendritic dopamine release. To explore the hypothesis that D1Rs modulate GABA release from striatonigral synapses, we conducted optical recordings of striatonigral activity and postsynaptic patch-clamp recordings from SNr neurons in the presence of dopamine and D1R agonists. We found that dopamine inhibits optogenetically triggered striatonigral GABA release by modulating vesicle fusion and Ca 2+ influx in striatonigral boutons. Notably, the effect of DA was independent of D1R activity but required activation of 5-HT1B receptors. Our results suggest a serotonergic mechanism involved in the therapeutic actions of dopaminergic medications for Parkinson's disease and psychostimulant-related disorders.

11.
bioRxiv ; 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38352367

RESUMO

Autism Spectrum Disorders (ASD) consist of diverse neurodevelopmental conditions where core behavioral symptoms are critical for diagnosis. Altered dopamine neurotransmission in the striatum has been suggested to contribute to the behavioral features of ASD. Here, we examine dopamine neurotransmission in a mouse model of ASD characterized by elevated expression of the eukaryotic initiation factor 4E (eIF4E), a key regulator of cap-dependent translation, using a comprehensive approach that encompasses genetics, behavior, synaptic physiology, and imaging. The results indicate that increased eIF4E expression leads to behavioral inflexibility and impaired striatal dopamine release. The loss of normal dopamine neurotransmission is due to a defective nicotinic receptor signaling that regulates calcium dynamics in dopaminergic axons. These findings reveal an intricate interplay between eIF4E, DA neurotransmission, and behavioral flexibility, provide a mechanistic understanding of ASD symptoms and offer a foundation for targeted therapeutic interventions.

12.
J Neurosci ; 32(17): 5900-10, 2012 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-22539851

RESUMO

Although L-3,4-dihydroxyphenylalanine (L-DOPA) remains the reference treatment of Parkinson's disease, its long-term beneficial effects are hindered by L-DOPA-induced dyskinesia (LID). In the dopamine (DA)-denervated striatum, L-DOPA activates DA D1 receptor(D1R) signaling, including cAMP-dependent protein kinase A (PKA) and extracellular signal-regulated kinase (ERK), two responses associated with LID. However, the cause of PKA and ERK activation, their respective contribution to LID, and their relationship are not known. In striatal neurons, D1R activates adenylyl-cyclase through Gα(olf), a protein upregulated after lesion of DA neurons in rats and inpatients. We report here that increased Gα(olf) levels in hemiparkinsonian mice are correlated with LID after chronic L-DOPA treatment. To determine the role of this upregulation, we performed unilateral lesion in mice lacking one allele of the Gnal gene coding for Gα(olf) (Gnal⁺/⁻). Despite an increase in the lesioned striatum,Gα(olf) levels remained below those of unlesioned wild-type mice. In Gnal⁺/⁻ mice, the lesion-induced L-DOPA stimulation of cAMP/PKA-mediated phosphorylation of GluA1 Ser845 and DARPP-32 (32 kDa DA- and cAMP-regulated phosphoprotein) Thr34 was dramatically reduced, whereas ERK activation was preserved. LID occurrence was similar in Gnal⁺/⁺ and Gnal⁺/⁻ mice after a 10-d L-DOPA (20 mg/kg) treatment. Thus, in lesioned animals, Gα(olf) upregulation is critical for the activation by L-DOPA of D1R-stimulated cAMP/PKA but not ERK signaling. Although the cAMP/PKA pathway appears to be required for LID development, our results indicate that its activation is unlikely to be the main source of LID. In contrast, the persistence of L-DOPA-induced ERK activation in Gnal⁺/⁻ mice supports its causal role in LID development.


Assuntos
Discinesia Induzida por Medicamentos/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Mutação/fisiologia , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Antiparkinsonianos/efeitos adversos , Benserazida/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Corpo Estriado/efeitos dos fármacos , Interações Medicamentosas , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/genética , Inibidores Enzimáticos/farmacologia , Lateralidade Funcional/efeitos dos fármacos , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Histonas/metabolismo , Levodopa/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Movimento/efeitos dos fármacos , Mutação/genética , Oxidopamina/farmacologia , Desempenho Psicomotor/efeitos dos fármacos , Receptores de AMPA/metabolismo , Transdução de Sinais/genética , Simpatolíticos/farmacologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/metabolismo
13.
J Biol Chem ; 287(33): 27806-12, 2012 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-22753408

RESUMO

Dyskinesia, a motor complication caused by prolonged administration of the antiparkinsonian drug l-3,4-dihydroxyphenylalanine (l-DOPA), is accompanied by activation of cAMP signaling and hyperphosphorylation of the dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32). Here, we show that the abnormal phosphorylation of DARPP-32 occurs specifically in medium spiny neurons (MSNs) expressing dopamine D1 receptors (D1R). Using mice in which DARPP-32 is selectively deleted in D1R-expressing MSNs, we demonstrate that this protein is required for l-DOPA-induced activation of the extracellular signal-regulated protein kinases 1 and 2 and the mammalian target of rapamycin complex 1 (mTORC1) pathways, which are implicated in dyskinesia. We also show that mutation of the phosphorylation site for cAMP-dependent protein kinase on DARPP-32 attenuates l-DOPA-induced dyskinesia and reduces the concomitant activations of ERK and mTORC1 signaling. These studies demonstrate that, in D1R-expressing MSNs, l-DOPA-induced activation of ERK and mTORC1 requires DARPP-32 and indicates the importance of the cAMP/DARPP-32 signaling cascade in dyskinesia.


Assuntos
AMP Cíclico/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Dopamina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Proteínas/metabolismo , Animais , Antiparkinsonianos/farmacologia , AMP Cíclico/genética , Dopamina/genética , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/genética , Humanos , Levodopa/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos , Proteínas do Tecido Nervoso/genética , Transtornos Parkinsonianos/tratamento farmacológico , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Proteínas/genética , Serina-Treonina Quinases TOR
14.
J Neurophysiol ; 109(1): 68-76, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23054596

RESUMO

Persistent forms of synaptic plasticity are widely thought to require the synthesis of new proteins. This feature of long-lasting forms of plasticity largely has been demonstrated using inhibitors of general protein synthesis, such as either anisomycin or emetine. However, these drugs, which inhibit elongation, cannot address detailed questions about the regulation of translation initiation, where the majority of translational control occurs. Moreover, general protein synthesis inhibitors cannot distinguish between cap-dependent and cap-independent modes of translation initiation. In the present study, we took advantage of two novel compounds, 4EGI-1 and hippuristanol, each of which targets a different component of the eukaryotic initiation factor (eIF)4F initiation complex, and investigated their effects on long-term potentiation (LTP) at CA3-CA1 synapses in the hippocampus. We found that 4EGI-1 and hippuristanol both attenuated long-lasting late-phase LTP induced by two different stimulation paradigms. We also found that 4EGI-1 and hippuristanol each were capable of blocking the expression of newly synthesized proteins immediately after the induction of late-phase LTP. These new pharmacological tools allow for a more precise dissection of the role played by translational control pathways in synaptic plasticity and demonstrate the importance of multiple aspects of eIF4F in processes underlying hippocampal LTP, laying the foundation for future studies investigating the role of eIF4F in hippocampus-dependent memory processes.


Assuntos
Fator de Iniciação 4F em Eucariotos/metabolismo , Hipocampo/fisiologia , Potenciação de Longa Duração/fisiologia , Biossíntese de Proteínas/fisiologia , Animais , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hidrazonas , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitrocompostos/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Esteróis/farmacologia , Sinapses/efeitos dos fármacos , Sinapses/fisiologia , Tiazóis/farmacologia
15.
Proc Natl Acad Sci U S A ; 107(33): 14845-50, 2010 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-20682746

RESUMO

The direct and indirect pathways of the basal ganglia have been proposed to oppositely regulate locomotion and differentially contribute to pathological behaviors. Analysis of the distinct contributions of each pathway to behavior has been a challenge, however, due to the difficulty of selectively investigating the neurons comprising the two pathways using conventional techniques. Here we present two mouse models in which the function of striatonigral or striatopallidal neurons is selectively disrupted due to cell type-specific deletion of the striatal signaling protein dopamine- and cAMP-regulated phosphoprotein Mr 32kDa (DARPP-32). Using these mice, we found that the loss of DARPP-32 in striatonigral neurons decreased basal and cocaine-induced locomotion and abolished dyskinetic behaviors in response to the Parkinson's disease drug L-DOPA. Conversely, the loss of DARPP-32 in striatopallidal neurons produced a robust increase in locomotor activity and a strongly reduced cataleptic response to the antipsychotic drug haloperidol. These findings provide insight into the selective contributions of the direct and indirect pathways to striatal motor behaviors.


Assuntos
Corpo Estriado/metabolismo , Fosfoproteína 32 Regulada por cAMP e Dopamina/fisiologia , Atividade Motora/fisiologia , Neurônios/metabolismo , Animais , Catalepsia/induzido quimicamente , Catalepsia/fisiopatologia , Cocaína/farmacologia , Corpo Estriado/citologia , Dopaminérgicos/toxicidade , Inibidores da Captação de Dopamina/farmacologia , Fosfoproteína 32 Regulada por cAMP e Dopamina/genética , Fosfoproteína 32 Regulada por cAMP e Dopamina/metabolismo , Discinesia Induzida por Medicamentos/etiologia , Discinesia Induzida por Medicamentos/fisiopatologia , Feminino , Imunofluorescência , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Haloperidol/toxicidade , Imuno-Histoquímica , Levodopa/toxicidade , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/classificação , Neurônios/citologia , Potenciais Sinápticos/fisiologia
16.
Cell Rep ; 42(8): 112901, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37505982

RESUMO

Individuals with fragile X syndrome (FXS) are frequently diagnosed with autism spectrum disorder (ASD), including increased risk for restricted and repetitive behaviors (RRBs). Consistent with observations in humans, FXS model mice display distinct RRBs and hyperactivity that are consistent with dysfunctional cortico-striatal circuits, an area relatively unexplored in FXS. Using a multidisciplinary approach, we dissect the contribution of two populations of striatal medium spiny neurons (SPNs) in the expression of RRBs in FXS model mice. Here, we report that dysregulated protein synthesis at cortico-striatal synapses is a molecular culprit of the synaptic and ASD-associated motor phenotypes displayed by FXS model mice. Cell-type-specific translational profiling of the FXS mouse striatum reveals differentially translated mRNAs, providing critical information concerning potential therapeutic targets. Our findings uncover a cell-type-specific impact of the loss of fragile X messenger ribonucleoprotein (FMRP) on translation and the sequence of neuronal events in the striatum that drive RRBs in FXS.


Assuntos
Transtorno do Espectro Autista , Síndrome do Cromossomo X Frágil , Animais , Humanos , Camundongos , Síndrome do Cromossomo X Frágil/metabolismo , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Neurônios/metabolismo , Sinapses/metabolismo , Camundongos Knockout , Modelos Animais de Doenças
17.
Biol Open ; 12(10)2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37815090

RESUMO

Genetic variants affecting Heterogeneous Nuclear Ribonucleoprotein U (HNRNPU) have been identified in several neurodevelopmental disorders (NDDs). HNRNPU is widely expressed in the human brain and shows the highest postnatal expression in the cerebellum. Recent studies have investigated the role of HNRNPU in cerebral cortical development, but the effects of HNRNPU deficiency on cerebellar development remain unknown. Here, we describe the molecular and cellular outcomes of HNRNPU locus deficiency during in vitro neural differentiation of patient-derived and isogenic neuroepithelial stem cells with a hindbrain profile. We demonstrate that HNRNPU deficiency leads to chromatin remodeling of A/B compartments, and transcriptional rewiring, partly by impacting exon inclusion during mRNA processing. Genomic regions affected by the chromatin restructuring and host genes of exon usage differences show a strong enrichment for genes implicated in epilepsies, intellectual disability, and autism. Lastly, we show that at the cellular level HNRNPU downregulation leads to an increased fraction of neural progenitors in the maturing neuronal population. We conclude that the HNRNPU locus is involved in delayed commitment of neural progenitors to differentiate in cell types with hindbrain profile.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo U , Transtornos do Neurodesenvolvimento , Humanos , Cromatina , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/genética , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo , Transtornos do Neurodesenvolvimento/genética , Neurogênese/genética , Rombencéfalo/metabolismo
18.
Neurobiol Dis ; 45(1): 156-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21827857

RESUMO

Tuberous sclerosis complex (TSC) is a genetic disorder characterized by the development of hamartomas in multiple organs. Neurological manifestation includes cortical dysplasia, epilepsy, and cognitive deficits such as mental impairment and autism. We measured the impact of TSC2-GAP mutations on cognitive processes and behavior in, ΔRG transgenic mice that express a dominant/negative TSC2 that binds to TSC1, but has mutations affecting its GAP domain and its rabaptin-5 binding motif, resulting in inactivation of the TSC1/2 complex. We performed a behavioral characterization of the ΔRG transgenic mice and found that they display mild, but significant impairments in social behavior and rotarod motor learning. These findings suggest that the ΔRG transgenic mice recapitulate some behavioral abnormalities observed in human TSC patients.


Assuntos
Comportamento Animal/fisiologia , Aprendizagem/fisiologia , Atividade Motora/fisiologia , Comportamento Social , Esclerose Tuberosa/fisiopatologia , Animais , Condicionamento Psicológico/fisiologia , Modelos Animais de Doenças , Medo/fisiologia , Asseio Animal/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Teste de Desempenho do Rota-Rod , Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
19.
Neurobiol Dis ; 45(3): 1101-10, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22198573

RESUMO

Tuberous sclerosis complex (TSC) and fragile X syndrome (FXS) are caused by mutations in negative regulators of translation. FXS model mice exhibit enhanced metabotropic glutamate receptor-dependent long-term depression (mGluR-LTD). Therefore, we hypothesized that a mouse model of TSC, ΔRG transgenic mice, also would exhibit enhanced mGluR-LTD. We measured the impact of TSC2-GAP mutations on the mTORC1 and ERK signaling pathways and protein synthesis-dependent hippocampal synaptic plasticity in ΔRG transgenic mice. These mice express a dominant/negative TSC2 that binds to TSC1, but has a deletion and substitution mutation in its GAP-domain, resulting in inactivation of the complex. Consistent with previous studies of several other lines of TSC model mice, we observed elevated S6 phosphorylation in the brains of ΔRG mice, suggesting upregulated translation. Surprisingly, mGluR-LTD was not enhanced, but rather was impaired in the ΔRG transgenic mice, indicating that TSC and FXS have divergent synaptic plasticity phenotypes. Similar to patients with TSC, the ΔRG transgenic mice exhibit elevated ERK signaling. Moreover, the mGluR-LTD impairment displayed by the ΔRG transgenic mice was rescued with the MEK-ERK inhibitor U0126. Our results suggest that the mGluR-LTD impairment observed in ΔRG mice involves aberrant TSC1/2-ERK signaling.


Assuntos
Depressão Sináptica de Longo Prazo/genética , Sistema de Sinalização das MAP Quinases/genética , Receptores de Glutamato Metabotrópico/metabolismo , Esclerose Tuberosa/complicações , Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/deficiência , Análise de Variância , Animais , Animais Recém-Nascidos , Biofísica , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Fármacos Atuantes sobre Aminoácidos Excitatórios/farmacologia , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Hipocampo/patologia , Técnicas In Vitro , Metoxi-Hidroxifenilglicol/análogos & derivados , Metoxi-Hidroxifenilglicol/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Técnicas de Patch-Clamp , Receptores de Glutamato Metabotrópico/genética , Proteínas Quinases S6 Ribossômicas/genética , Proteínas Quinases S6 Ribossômicas/metabolismo , Esclerose Tuberosa/metabolismo , Esclerose Tuberosa/patologia , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
20.
Neurobiol Dis ; 38(3): 434-45, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20227500

RESUMO

DYT1 dystonia is an inherited disease linked to mutation in the TOR1A gene encoding for the protein torsinA. Although the mechanism by which this genetic alteration leads to dystonia is unclear, multiple lines of clinical evidence suggest a link between dystonia and a reduced dopamine D2 receptor (D2R) availability. Based on this evidence, herein we carried out a comprehensive analysis of electrophysiological, behavioral and signaling correlates of D2R transmission in transgenic mice with the DYT1 dystonia mutation. Electrophysiological recordings from nigral dopaminergic neurons showed a normal responsiveness to D2-autoreceptor function. Conversely, postsynaptic D2R function in hMT mice was impaired, as suggested by the inability of a D2R agonist to re-establish normal corticostriatal synaptic plasticity and supported by the reduced sensitivity to haloperidol-induced catalepsy. Although an in situ hybridization analysis showed normal D1R and D2R mRNA expression levels in the striata of hMT mice, we found a significant decrease of D2R protein, coupled to a reduced ability of D2Rs to activate their cognate Go/i proteins. Of relevance, we found that pharmacological blockade of adenosine A2A receptors (A2ARs) fully restored the impairment of synaptic plasticity observed in hMT mice. Together, our findings demonstrate an important link between torsinA mutation and D2R dysfunction and suggest that A2AR antagonism is able to counteract the deficit in D2R-mediated transmission observed in mutant mice, opening new perspectives for the treatment of this movement disorder.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Distonia/tratamento farmacológico , Distonia/fisiopatologia , Chaperonas Moleculares/metabolismo , Receptor A2A de Adenosina/metabolismo , Receptores de Dopamina D2/metabolismo , Animais , Fármacos do Sistema Nervoso Central/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Dopamina/metabolismo , Distonia/genética , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Transgênicos , Chaperonas Moleculares/genética , Vias Neurais/efeitos dos fármacos , Vias Neurais/fisiopatologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , RNA Mensageiro/metabolismo , Receptores de Dopamina D1/metabolismo , Substância Negra/efeitos dos fármacos , Substância Negra/fisiopatologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA