Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Nature ; 586(7829): 407-411, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33029009

RESUMO

To survive in a dynamic environment, animals need to identify and appropriately respond to stimuli that signal danger1. Survival also depends on suppressing the threat-response during a stimulus that predicts the absence of threat (safety)2-5. An understanding of the biological substrates of emotional memories during a task in which animals learn to flexibly execute defensive responses to a threat-predictive cue and a safety cue is critical for developing treatments for memory disorders such as post-traumatic stress disorder5. The centrolateral amygdala is an important node in the neuronal circuit that mediates defensive responses6-9, and a key brain area for processing and storing threat memories. Here we applied intersectional chemogenetic strategies to inhibitory neurons in the centrolateral amygdala of mice to block cell-type-specific translation programs that are sensitive to depletion of eukaryotic initiation factor 4E (eIF4E) and phosphorylation of eukaryotic initiation factor 2α (p-eIF2α). We show that de novo translation in somatostatin-expressing inhibitory neurons in the centrolateral amygdala is necessary for the long-term storage of conditioned-threat responses, whereas de novo translation in protein kinase Cδ-expressing inhibitory neurons in the centrolateral amygdala is necessary for the inhibition of a conditioned response to a safety cue. Our results provide insight into the role of de novo protein synthesis in distinct inhibitory neuron populations in the centrolateral amygdala during the consolidation of long-term memories.


Assuntos
Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/fisiologia , Emoções , Memória/fisiologia , Inibição Neural , Neurônios/fisiologia , Animais , Condicionamento Psicológico , Sinais (Psicologia) , Fator de Iniciação 2 em Eucariotos/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Medo/fisiologia , Feminino , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Masculino , Camundongos , Biossíntese de Proteínas , Capuzes de RNA/genética , Capuzes de RNA/metabolismo , Transdução de Sinais , Somatostatina/metabolismo
2.
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
3.
Cell Rep ; 43(2): 113670, 2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38219147

RESUMO

Neuronal protein synthesis is required for long-lasting plasticity and long-term memory consolidation. Dephosphorylation of eukaryotic initiation factor 2α is one of the key translational control events that is required to increase de novo protein synthesis that underlies long-lasting plasticity and memory consolidation. Here, we interrogate the molecular pathways of translational control that are triggered by neuronal stimulation with brain-derived neurotrophic factor (BDNF), which results in eukaryotic initiation factor 2α (eIF2α) dephosphorylation and increases in de novo protein synthesis. Primary rodent neurons exposed to BDNF display elevated translation of GADD34, which facilitates eIF2α dephosphorylation and subsequent de novo protein synthesis. Furthermore, GADD34 requires G-actin generated by cofilin to dephosphorylate eIF2α and enhance protein synthesis. Finally, GADD34 is required for BDNF-induced translation of synaptic plasticity-related proteins. Overall, we provide evidence that neurons repurpose GADD34, an effector of the integrated stress response, as an orchestrator of rapid increases in eIF2-dependent translation in response to plasticity-inducing stimuli.


Assuntos
Fatores de Despolimerização de Actina , Fator Neurotrófico Derivado do Encéfalo , Actinas , Fator de Iniciação 2 em Eucariotos , Neurônios
4.
Mol Autism ; 13(1): 29, 2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35768828

RESUMO

BACKGROUND: Fragile X syndrome (FXS), the most common genetic cause of autism spectrum disorder and intellectual disability, is caused by the lack of fragile X mental retardation protein (FMRP) expression. FMRP is an mRNA binding protein with functions in mRNA transport, localization, and translational control. In Fmr1 knockout mice, dysregulated translation has been linked to pathophysiology, including abnormal synaptic function and dendritic morphology, and autistic-like behavioral phenotypes. The role of FMRP in morphology and function of excitatory neurons has been well studied in mice lacking Fmr1, but the impact of Fmr1 deletion on inhibitory neurons remains less characterized. Moreover, the contribution of FMRP in different cell types to FXS pathophysiology is not well defined. We sought to characterize whether FMRP loss in parvalbumin or somatostatin-expressing neurons results in FXS-like deficits in mice. METHODS: We used Cre-lox recombinase technology to generate two lines of conditional knockout mice lacking FMRP in either parvalbumin or somatostatin-expressing cells and carried out a battery of behavioral tests to assess motor function, anxiety, repetitive, stereotypic, social behaviors, and learning and memory. In addition, we used fluorescent non-canonical amino acid tagging along with immunostaining to determine whether de novo protein synthesis is dysregulated in parvalbumin or somatostatin-expressing neurons. RESULTS: De novo protein synthesis was elevated in hippocampal parvalbumin and somatostatin-expressing inhibitory neurons in Fmr1 knockout mice. Cell type-specific deletion of Fmr1 in parvalbumin-expressing neurons resulted in anxiety-like behavior, impaired social behavior, and dysregulated de novo protein synthesis. In contrast, deletion of Fmr1 in somatostatin-expressing neurons did not result in behavioral abnormalities and did not significantly impact de novo protein synthesis. This is the first report of how loss of FMRP in two specific subtypes of inhibitory neurons is associated with distinct FXS-like abnormalities. LIMITATIONS: The mouse models we generated are limited by whole body knockout of FMRP in parvalbumin or somatostatin-expressing cells and further studies are needed to establish a causal relationship between cellular deficits and FXS-like behaviors. CONCLUSIONS: Our findings indicate a cell type-specific role for FMRP in parvalbumin-expressing neurons in regulating distinct behavioral features associated with FXS.


Assuntos
Transtorno do Espectro Autista , Proteína do X Frágil da Deficiência Intelectual , Síndrome do Cromossomo X Frágil , Neurônios , Animais , Transtorno do Espectro Autista/metabolismo , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/genética , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/genética , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/patologia , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Neurônios/patologia , Parvalbuminas/metabolismo , RNA Mensageiro/metabolismo , Somatostatina/metabolismo
5.
J Pineal Res ; 51(1): 75-86, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21355879

RESUMO

Mitochondrial dysfunction is a hallmark of Alzheimer's disease (AD) and is observed in mutant amyloid precursor protein (APP) transgenic mouse models of familial AD. Melatonin is a potent antioxidant, can prevent toxic aggregation of Alzheimer's beta-amyloid (Aß) peptide and, when taken long term, can protect against cognitive deficits in APP transgenic mice. To study the effects of melatonin on brain mitochondrial function in an AD model, APP/PS1 transgenic mice were treated for 1 month with melatonin. Analysis of isolated brain mitochondria from mice indicated that melatonin treatment decreased mitochondrial Aß levels by two- to fourfold in different brain regions. This was accompanied by a near complete restoration of mitochondrial respiratory rates, membrane potential, and ATP levels in isolated mitochondria from the hippocampus, cortex, or striatum. When isolated mitochondria from untreated young mice were given melatonin, a slight increase in respiratory rate was observed. No such effect was observed in mitochondria from aged mice. In APP-expressing neuroblastoma cells in culture, mitochondrial function was restored by melatonin or by the structurally related compounds indole-3-propionic acid or N(1)-acetyl-N(2)-formyl-5-methoxykynuramine. This restoration was partially blocked by melatonin receptor antagonists indicating melatonin receptor signaling is required for the full effect. Therefore, treatments that stimulate melatonin receptor signaling may be beneficial for restoring mitochondrial function in AD, and preservation of mitochondrial function may an important mechanism by which long term melatonin treatment delays cognitive dysfunction in AD mice.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Melatonina/farmacologia , Mitocôndrias/efeitos dos fármacos , Receptores de Melatonina/metabolismo , Trifosfato de Adenosina/metabolismo , Doença de Alzheimer/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Análise de Variância , Animais , Encéfalo/citologia , Encéfalo/metabolismo , Fracionamento Celular , Linhagem Celular Tumoral , Indóis/farmacologia , Cinuramina/análogos & derivados , Cinuramina/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/metabolismo , Neuroblastoma , Propionatos/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Estatísticas não Paramétricas
6.
Commun Biol ; 4(1): 823, 2021 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-34193971

RESUMO

Alzheimer's disease (AD) is an age-related neurodegenerative disorder associated with memory loss, but the AD-associated neuropathological changes begin years before memory impairments. Investigation of the early molecular abnormalities in AD might offer innovative opportunities to target memory impairment prior to onset. Decreased protein synthesis plays a fundamental role in AD, yet the consequences of this dysregulation for cellular function remain unknown. We hypothesize that alterations in the de novo proteome drive early metabolic alterations in the hippocampus that persist throughout AD progression. Using a combinatorial amino acid tagging approach to selectively label and enrich newly synthesized proteins, we found that the de novo proteome is disturbed in young APP/PS1 mice prior to symptom onset, affecting the synthesis of multiple components of the synaptic, lysosomal, and mitochondrial pathways. Furthermore, the synthesis of large clusters of ribosomal subunits were affected throughout development. Our data suggest that large-scale changes in protein synthesis could underlie cellular dysfunction in AD.


Assuntos
Envelhecimento , Doença de Alzheimer/metabolismo , Modelos Animais de Doenças , Hipocampo/metabolismo , Proteoma/metabolismo , Proteômica/métodos , Fatores Etários , Doença de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Cromatografia Líquida/métodos , Feminino , Hipocampo/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Presenilina-1/genética , Presenilina-1/metabolismo , Proteoma/classificação , Espectrometria de Massas em Tandem/métodos
7.
Nat Commun ; 10(1): 1710, 2019 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-30979884

RESUMO

Whether fragile X mental retardation protein (FMRP) target mRNAs and neuronal activity contributing to elevated basal neuronal protein synthesis in fragile X syndrome (FXS) is unclear. Our proteomic experiments reveal that the de novo translational profile in FXS model mice is altered at steady state and in response to metabotropic glutamate receptor (mGluR) stimulation, but the proteins expressed differ under these conditions. Several altered proteins, including Hexokinase 1 and Ras, also are expressed in the blood of FXS model mice and pharmacological treatments previously reported to ameliorate phenotypes modify their abundance in blood. In addition, plasma levels of Hexokinase 1 and Ras differ between FXS patients and healthy volunteers. Our data suggest that brain-based de novo proteomics in FXS model mice can be used to find altered expression of proteins in blood that could serve as disease-state biomarkers in individuals with FXS.


Assuntos
Síndrome do Cromossomo X Frágil/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Adolescente , Adulto , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Feminino , Síndrome do Cromossomo X Frágil/genética , Hexoquinase/sangue , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Adulto Jovem , Proteínas ras/metabolismo
8.
BJU Int ; 101(8): 1048-51, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18070171

RESUMO

OBJECTIVE: To evaluate whether fluorescent tracers can consistently label the neurovascular bundles (NVBs) and major pelvic ganglion (MPG) after an intracavernosal penile injection, as the reported incidence of erectile dysfunction (ED) in men after radical prostatectomy (RP) is 55-65% and thus preservation of erectile function, sparing one or both of the NVBs remains one of the most vital factors. MATERIALS AND METHODS: Male Sprague-Dawley rats (3 months old) received penile injections (20 microL; seven rats/group) of either deionized water (DW), Fluoro-Gold (FG), Fast-Blue (FB), Fluoro-Ruby (FR) or green fluorescent pseudorabies virus (GF-PRv). The rats were killed at 2, 3 and 14 days after injection and the NVBs and MPG were harvested and placed directly under fluorescence light. Image analysis was done by computer, coupled to a microscope equipped with a digital camera. Each NVB and MPG were analysed for its staining pattern and consistency. RESULTS: When compared with the FB, FR and GF-PRv rats, the FG-injected rats had better staining of the NVB at 2, 3 and 14 days after injection. Under x200, FG highlighted the axons of the cavernous nerve (CN) and cell bodies (MPG). This indicates that FG injection into the penis induced the strongest CN labelling (positive staining) at 2 and 3 days after injection as compared with FB-, FR- and GF-PRv-injected rats. CONCLUSION: FG injection into the penis has consistent retrograde staining of the NVBs and MPG after 3 days. Therefore, we predict that FG could potentially be used to improve the identification of the NVB in other models. However, further studies need to be carried out before these tracers can be used in humans.


Assuntos
Microscopia de Fluorescência/normas , Ereção Peniana/fisiologia , Pênis/inervação , Vias Aferentes/fisiologia , Animais , Vias Eferentes/fisiologia , Disfunção Erétil/prevenção & controle , Injeções , Masculino , Pênis/fisiopatologia , Prostatectomia/efeitos adversos , Ratos , Ratos Sprague-Dawley
9.
Neuropsychopharmacology ; 41(8): 1991-2000, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26708105

RESUMO

Aberrant neuronal translation is implicated in the etiology of numerous brain disorders. Although mTORC1-p70 ribosomal S6 kinase 1 (S6K1) signaling is critical for translational control, pharmacological manipulation in vivo has targeted exclusively mTORC1 due to the paucity of specific inhibitors to S6K1. However, small molecule inhibitors of S6K1 could potentially ameliorate pathological phenotypes of diseases, which are based on aberrant translation and protein expression. One such condition is fragile X syndrome (FXS), which is considered to be caused by exaggerated neuronal translation and is the most frequent heritable cause of autism spectrum disorder (ASD). To date, potential therapeutic interventions in FXS have focused largely on targets upstream of translational control to normalize FXS-related phenotypes. Here we test the ability of two S6K1 inhibitors, PF-4708671 and FS-115, to normalize translational homeostasis and other phenotypes exhibited by FXS model mice. We found that although the pharmacokinetic profiles of the two S6K1 inhibitors differed, they overlapped in reversing multiple disease-associated phenotypes in FXS model mice including exaggerated protein synthesis, inappropriate social behavior, behavioral inflexibility, altered dendritic spine morphology, and macroorchidism. In contrast, the two inhibitors differed in their ability to rescue stereotypic marble-burying behavior and weight gain. These findings provide an initial pharmacological characterization of the impact of S6K1 inhibitors in vivo for FXS, and have therapeutic implications for other neuropsychiatric conditions involving aberrant mTORC1-S6K1 signaling.


Assuntos
Comportamento Animal/efeitos dos fármacos , Encéfalo/metabolismo , Síndrome do Cromossomo X Frágil/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/administração & dosagem , Piperazinas/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Animais , Transtorno do Espectro Autista/prevenção & controle , Transtorno do Espectro Autista/psicologia , Encéfalo/patologia , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/patologia , Modelos Animais de Doenças , Comportamento Exploratório/efeitos dos fármacos , Feminino , Síndrome do Cromossomo X Frágil/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fenótipo , Fosforilação , Comportamento Social
10.
Neuropharmacology ; 63(8): 1368-79, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22959965

RESUMO

Caffeine and melatonin have been shown to protect the Swedish mutant amyloid precursor protein (APP(sw)) transgenic mouse model of Alzheimer's disease from cognitive dysfunction. But their mechanisms of action remain incompletely understood. These Alzheimer's mice have extensive mitochondrial dysfunction, which likely contributes to their cognitive decline. To further explore the mechanism through which caffeine and melatonin protect cognitive function in these mice, we monitored the function of isolated mitochondria from APP(sw) mice treated with caffeine, melatonin, or both in their drinking water for one month. Melatonin treatment yielded a near complete restoration of mitochondrial function in assays of respiratory rate, membrane potential, reactive oxygen species production, and ATP levels. Caffeine treatment by itself yielded a small increase in mitochondrial function. However, caffeine largely blocked the large enhancement of mitochondrial function provided by melatonin. Studies with N2a neuroblastoma cells stably expressing APP(sw) showed that specific inhibition of cAMP-dependent phosphodiesterase (PDE) 4 or cGMP-dependent PDE5 also blocked melatonin protection of mitochondrial function, but A(2a) and A1 adenosine receptor antagonists were without effect. Melatonin or caffeine at the concentrations used to modulate mitochondrial function in the cells had no effect on cAMP-dependent PDE activity or cellular cAMP or cGMP levels. Therefore, caffeine and increased cyclic nucleotide levels likely block melatonin signaling to mitochondria by independent mechanisms that do not involve adenosine receptor antagonism. The results of this study indicate that melatonin restores mitochondrial function much more potently than caffeine in APP(sw) transgenic mouse and cell models of Alzheimer's disease.


Assuntos
Doença de Alzheimer/patologia , Antioxidantes/farmacologia , Cafeína/farmacologia , Estimulantes do Sistema Nervoso Central/farmacologia , Melatonina/antagonistas & inibidores , Melatonina/farmacologia , Mitocôndrias/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animais , Células Cultivadas , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Ensaio de Imunoadsorção Enzimática , Técnicas In Vitro , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Doenças Mitocondriais/tratamento farmacológico , Doenças Mitocondriais/metabolismo , Neurônios/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transfecção
11.
Brain Res ; 1256: 173-9, 2009 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-19135431

RESUMO

Rodents exposed to environmental enrichment show many differences, including improved cognitive performance, when compared to those living in standard (impoverished) housing. The purpose of the present study was to determine if a selective increase in neurogenesis occurred in cognitively-protected Tg mice raised in an enriched environment compared to those reared in physical activity housing. At weaning, double Tg APP+PS1 mice were placed into one of three environments: complete environmental enrichment (CE), enhanced physical activity (PA), or individual, impoverished housing (IMP). At 9-10 months of age, Tg mice were injected with BrdU (100 mg/kg BID) followed by euthanasia either 24 h or 2 weeks after the last injection. Unbiased estimates of BrdU positive cells in the hippocampal subgranular zone revealed a significant increase in cellular proliferation in Tg mice raised in CE or PA compared to Tg mice reared in IMP housing. However, counts of BrdU birth-dated cells 2 weeks after labeling showed no difference among the three groups, indicating decreased survival of cells in those groups (CE and PA) with higher cellular proliferation rates in the neurogenic niche. Counts of calretinin-expressing cells, a marker of immature neurons, also indicated no difference among the three groups of mice. In view of our prior study showing that enhanced cognitive activity (but not enhanced physical activity) protects Tg mice against cognitive impairment, the present results indicate that increased generation and survival of new neurons in the hippocampal dentate gyrus is not involved with the cognitively-protective effects of complete CE in Alzheimer's transgenic mice.


Assuntos
Cognição/fisiologia , Meio Ambiente , Abrigo para Animais , Atividade Motora/fisiologia , Neurogênese , Precursor de Proteína beta-Amiloide/genética , Animais , Calbindina 2 , Sobrevivência Celular/fisiologia , Feminino , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Neurônios , Presenilina-1/genética , Proteína G de Ligação ao Cálcio S100/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA