Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
Cell ; 184(7): 1740-1756.e16, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33705688

RESUMO

The core symptoms of many neurological disorders have traditionally been thought to be caused by genetic variants affecting brain development and function. However, the gut microbiome, another important source of variation, can also influence specific behaviors. Thus, it is critical to unravel the contributions of host genetic variation, the microbiome, and their interactions to complex behaviors. Unexpectedly, we discovered that different maladaptive behaviors are interdependently regulated by the microbiome and host genes in the Cntnap2-/- model for neurodevelopmental disorders. The hyperactivity phenotype of Cntnap2-/- mice is caused by host genetics, whereas the social-behavior phenotype is mediated by the gut microbiome. Interestingly, specific microbial intervention selectively rescued the social deficits in Cntnap2-/- mice through upregulation of metabolites in the tetrahydrobiopterin synthesis pathway. Our findings that behavioral abnormalities could have distinct origins (host genetic versus microbial) may change the way we think about neurological disorders and how to treat them.


Assuntos
Microbioma Gastrointestinal , Locomoção , Comportamento Social , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Biopterinas/análogos & derivados , Biopterinas/metabolismo , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores , Transplante de Microbiota Fecal , Fezes/microbiologia , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/fisiologia , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/microbiologia , Transtornos do Neurodesenvolvimento/patologia , Transtornos do Neurodesenvolvimento/terapia , Análise de Componente Principal , Agitação Psicomotora/patologia , Transmissão Sináptica
2.
Cell ; 165(7): 1762-1775, 2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-27315483

RESUMO

Maternal obesity during pregnancy has been associated with increased risk of neurodevelopmental disorders, including autism spectrum disorder (ASD), in offspring. Here, we report that maternal high-fat diet (MHFD) induces a shift in microbial ecology that negatively impacts offspring social behavior. Social deficits and gut microbiota dysbiosis in MHFD offspring are prevented by co-housing with offspring of mothers on a regular diet (MRD) and transferable to germ-free mice. In addition, social interaction induces synaptic potentiation (LTP) in the ventral tegmental area (VTA) of MRD, but not MHFD offspring. Moreover, MHFD offspring had fewer oxytocin immunoreactive neurons in the hypothalamus. Using metagenomics and precision microbiota reconstitution, we identified a single commensal strain that corrects oxytocin levels, LTP, and social deficits in MHFD offspring. Our findings causally link maternal diet, gut microbial imbalance, VTA plasticity, and behavior and suggest that probiotic treatment may relieve specific behavioral abnormalities associated with neurodevelopmental disorders. VIDEO ABSTRACT.


Assuntos
Transtorno do Espectro Autista/microbiologia , Dieta Hiperlipídica , Microbioma Gastrointestinal , Obesidade/complicações , Comportamento Social , Animais , Disbiose/fisiopatologia , Feminino , Vida Livre de Germes , Abrigo para Animais , Limosilactobacillus reuteri , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ocitocina/análise , Ocitocina/metabolismo , Gravidez , Área Tegmentar Ventral
3.
Proc Natl Acad Sci U S A ; 121(31): e2407472121, 2024 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-39047038

RESUMO

The integrated stress response (ISR), a pivotal protein homeostasis network, plays a critical role in the formation of long-term memory (LTM). The precise mechanism by which the ISR controls LTM is not well understood. Here, we report insights into how the ISR modulates the mnemonic process by using targeted deletion of the activating transcription factor 4 (ATF4), a key downstream effector of the ISR, in various neuronal and non-neuronal cell types. We found that the removal of ATF4 from forebrain excitatory neurons (but not from inhibitory neurons, cholinergic neurons, or astrocytes) enhances LTM formation. Furthermore, the deletion of ATF4 in excitatory neurons lowers the threshold for the induction of long-term potentiation, a cellular model for LTM. Transcriptomic and proteomic analyses revealed that ATF4 deletion in excitatory neurons leads to upregulation of components of oxidative phosphorylation pathways, which are critical for ATP production. Thus, we conclude that ATF4 functions as a memory repressor selectively within excitatory neurons.


Assuntos
Fator 4 Ativador da Transcrição , Memória de Longo Prazo , Neurônios , Animais , Camundongos , Fator 4 Ativador da Transcrição/metabolismo , Fator 4 Ativador da Transcrição/genética , Astrócitos/metabolismo , Potenciação de Longa Duração , Memória de Longo Prazo/fisiologia , Camundongos Knockout , Neurônios/metabolismo , Prosencéfalo/metabolismo , Masculino
4.
Cell ; 147(6): 1384-96, 2011 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-22153080

RESUMO

The double-stranded RNA-activated protein kinase (PKR) was originally identified as a sensor of virus infection, but its function in the brain remains unknown. Here, we report that the lack of PKR enhances learning and memory in several behavioral tasks while increasing network excitability. In addition, loss of PKR increases the late phase of long-lasting synaptic potentiation (L-LTP) in hippocampal slices. These effects are caused by an interferon-γ (IFN-γ)-mediated selective reduction in GABAergic synaptic action. Together, our results reveal that PKR finely tunes the network activity that must be maintained while storing a given episode during learning. Because PKR activity is altered in several neurological disorders, this kinase presents a promising new target for the treatment of cognitive dysfunction. As a first step in this direction, we show that a selective PKR inhibitor replicates the Pkr(-/-) phenotype in WT mice, enhancing long-term memory storage and L-LTP.


Assuntos
Hipocampo/fisiologia , Interferon gama/metabolismo , Potenciação de Longa Duração , eIF-2 Quinase/antagonistas & inibidores , eIF-2 Quinase/metabolismo , Animais , Eletrofisiologia , Técnicas In Vitro , Interferon gama/genética , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Sinapses , eIF-2 Quinase/genética
5.
Nature ; 586(7829): 412-416, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33029011

RESUMO

An important tenet of learning and memory is the notion of a molecular switch that promotes the formation of long-term memory1-4. The regulation of proteostasis is a critical and rate-limiting step in the consolidation of new memories5-10. One of the most effective and prevalent ways to enhance memory is by regulating the synthesis of proteins controlled by the translation initiation factor eIF211. Phosphorylation of the α-subunit of eIF2 (p-eIF2α), the central component of the integrated stress response (ISR), impairs long-term memory formation in rodents and birds11-13. By contrast, inhibiting the ISR by mutating the eIF2α phosphorylation site, genetically11 and pharmacologically inhibiting the ISR kinases14-17, or mimicking reduced p-eIF2α with the ISR inhibitor ISRIB11, enhances long-term memory in health and disease18. Here we used molecular genetics to dissect the neuronal circuits by which the ISR gates cognitive processing. We found that learning reduces eIF2α phosphorylation in hippocampal excitatory neurons and a subset of hippocampal inhibitory neurons (those that express somatostatin, but not parvalbumin). Moreover, ablation of p-eIF2α in either excitatory or somatostatin-expressing (but not parvalbumin-expressing) inhibitory neurons increased general mRNA translation, bolstered synaptic plasticity and enhanced long-term memory. Thus, eIF2α-dependent mRNA translation controls memory consolidation via autonomous mechanisms in excitatory and somatostatin-expressing inhibitory neurons.


Assuntos
Fator de Iniciação 2 em Eucariotos/metabolismo , Hipocampo/citologia , Consolidação da Memória , Neurônios/metabolismo , Somatostatina/metabolismo , Animais , Região CA1 Hipocampal/citologia , Região CA1 Hipocampal/fisiologia , Fator de Iniciação 2 em Eucariotos/deficiência , Fator de Iniciação 2 em Eucariotos/genética , Potenciais Pós-Sinápticos Excitadores , Hipocampo/fisiologia , Potenciação de Longa Duração , Masculino , Memória de Longo Prazo , Camundongos , Camundongos Endogâmicos C57BL , Inibição Neural , Plasticidade Neuronal , Parvalbuminas , Fosforilação , Células Piramidais/fisiologia , Transmissão Sináptica
6.
Nat Immunol ; 13(6): 543-550, 2012 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-22544393

RESUMO

Type I interferon is an integral component of the antiviral response, and its production is tightly controlled at the levels of transcription and translation. The eukaryotic translation-initiation factor eIF4E is a rate-limiting factor whose activity is regulated by phosphorylation of Ser209. Here we found that mice and fibroblasts in which eIF4E cannot be phosphorylated were less susceptible to virus infection. More production of type I interferon, resulting from less translation of Nfkbia mRNA (which encodes the inhibitor IκBα), largely explained this phenotype. The lower abundance of IκBα resulted in enhanced activity of the transcription factor NF-κB, which promoted the production of interferon-ß (IFN-ß). Thus, regulated phosphorylation of eIF4E has a key role in antiviral host defense by selectively controlling the translation of an mRNA that encodes a critical suppressor of the innate antiviral response.


Assuntos
Fator de Iniciação 4E em Eucariotos/metabolismo , Interferon Tipo I/biossíntese , NF-kappa B/metabolismo , Estomatite Vesicular/imunologia , Vírus da Estomatite Vesicular Indiana/fisiologia , Animais , Ensaio de Desvio de Mobilidade Eletroforética , Fator de Iniciação 4E em Eucariotos/imunologia , Feminino , Proteínas I-kappa B/biossíntese , Proteínas I-kappa B/genética , Proteínas I-kappa B/imunologia , Imunidade Inata/imunologia , Immunoblotting , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor de NF-kappaB alfa , NF-kappa B/imunologia , Fosforilação , Biossíntese de Proteínas , RNA Mensageiro/química , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Organismos Livres de Patógenos Específicos , Estomatite Vesicular/genética , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia , Vírus da Estomatite Vesicular Indiana/imunologia , Replicação Viral
7.
Cell ; 134(6): 1042-54, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18805096

RESUMO

Strong evidence indicates that regulated mRNA translation in neuronal dendrites underlies synaptic plasticity and brain development. The fragile X mental retardation protein (FMRP) is involved in this process; here, we show that it acts by inhibiting translation initiation. A binding partner of FMRP, CYFIP1/Sra1, directly binds the translation initiation factor eIF4E through a domain that is structurally related to those present in 4E-BP translational inhibitors. Brain cytoplasmic RNA 1 (BC1), another FMRP binding partner, increases the affinity of FMRP for the CYFIP1-eIF4E complex in the brain. Levels of proteins encoded by known FMRP target mRNAs are increased upon reduction of CYFIP1 in neurons. Translational repression is regulated in an activity-dependent manner because BDNF or DHPG stimulation of neurons causes CYFIP1 to dissociate from eIF4E at synapses, thereby resulting in protein synthesis. Thus, the translational repression activity of FMRP in the brain is mediated, at least in part, by CYFIP1.


Assuntos
Encéfalo/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Biossíntese de Proteínas , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Células Cultivadas , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Proteína do X Frágil da Deficiência Intelectual/química , Proteína do X Frágil da Deficiência Intelectual/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Alinhamento de Sequência , Sinapses
8.
BMC Biol ; 20(1): 28, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35086530

RESUMO

BACKGROUND: The functional understanding of genetic interaction networks and cellular mechanisms governing health and disease requires the dissection, and multifaceted study, of discrete cell subtypes in developing and adult animal models. Recombinase-driven expression of transgenic effector alleles represents a significant and powerful approach to delineate cell populations for functional, molecular, and anatomical studies. In addition to single recombinase systems, the expression of two recombinases in distinct, but partially overlapping, populations allows for more defined target expression. Although the application of this method is becoming increasingly popular, its experimental implementation has been broadly restricted to manipulations of a limited set of common alleles that are often commercially produced at great expense, with costs and technical challenges associated with production of intersectional mouse lines hindering customized approaches to many researchers. Here, we present a simplified CRISPR toolkit for rapid, inexpensive, and facile intersectional allele production. RESULTS: Briefly, we produced 7 intersectional mouse lines using a dual recombinase system, one mouse line with a single recombinase system, and three embryonic stem (ES) cell lines that are designed to study the way functional, molecular, and anatomical features relate to each other in building circuits that underlie physiology and behavior. As a proof-of-principle, we applied three of these lines to different neuronal populations for anatomical mapping and functional in vivo investigation of respiratory control. We also generated a mouse line with a single recombinase-responsive allele that controls the expression of the calcium sensor Twitch-2B. This mouse line was applied globally to study the effects of follicle-stimulating hormone (FSH) and luteinizing hormone (LH) on calcium release in the ovarian follicle. CONCLUSIONS: The lines presented here are representative examples of outcomes possible with the successful application of our genetic toolkit for the facile development of diverse, modifiable animal models. This toolkit will allow labs to create single or dual recombinase effector lines easily for any cell population or subpopulation of interest when paired with the appropriate Cre and FLP recombinase mouse lines or viral vectors. We have made our tools and derivative intersectional mouse and ES cell lines openly available for non-commercial use through publicly curated repositories for plasmid DNA, ES cells, and transgenic mouse lines.


Assuntos
Cálcio , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Animais , Feminino , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Recombinases/genética , Recombinases/metabolismo
9.
Annu Rev Neurosci ; 37: 17-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25032491

RESUMO

Activity-dependent changes in the strength of synaptic connections are fundamental to the formation and maintenance of memory. The mechanisms underlying persistent changes in synaptic strength in the hippocampus, specifically long-term potentiation and depression, depend on new protein synthesis. Such changes are thought to be orchestrated by engaging the signaling pathways that regulate mRNA translation in neurons. In this review, we discuss the key regulatory pathways that govern translational control in response to synaptic activity and the mRNA populations that are specifically targeted by these pathways. The critical contribution of regulatory control over new protein synthesis to proper cognitive function is underscored by human disorders associated with either silencing or mutation of genes encoding proteins that directly regulate translation. In light of these clinical implications, we also consider the therapeutic potential of targeting dysregulated translational control to treat cognitive disorders of synaptic dysfunction.


Assuntos
Transtornos Cognitivos/genética , Transtornos Cognitivos/fisiopatologia , Plasticidade Neuronal/genética , Biossíntese de Proteínas/genética , Animais , Hipocampo/fisiologia , Humanos , Modelos Biológicos , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais/genética
10.
Nat Immunol ; 9(10): 1097-9, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18800159

RESUMO

The production of type I interferon -- the first line of defense against virus infection and critical for innate immunity -- in plasmacytoid dendritic cells relies on the mammalian target of rapamycin.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/biossíntese , Proteínas Quinases/imunologia , Transdução de Sinais/imunologia , Animais , Células Dendríticas/metabolismo , Humanos , Serina-Treonina Quinases TOR
11.
Proc Natl Acad Sci U S A ; 114(31): E6420-E6426, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28696288

RESUMO

Traumatic brain injury (TBI) is a leading cause of long-term neurological disability, yet the mechanisms underlying the chronic cognitive deficits associated with TBI remain unknown. Consequently, there are no effective treatments for patients suffering from the long-lasting symptoms of TBI. Here, we show that TBI persistently activates the integrated stress response (ISR), a universal intracellular signaling pathway that responds to a variety of cellular conditions and regulates protein translation via phosphorylation of the translation initiation factor eIF2α. Treatment with ISRIB, a potent drug-like small-molecule inhibitor of the ISR, reversed the hippocampal-dependent cognitive deficits induced by TBI in two different injury mouse models-focal contusion and diffuse concussive injury. Surprisingly, ISRIB corrected TBI-induced memory deficits when administered weeks after the initial injury and maintained cognitive improvement after treatment was terminated. At the physiological level, TBI suppressed long-term potentiation in the hippocampus, which was fully restored with ISRIB treatment. Our results indicate that ISR inhibition at time points late after injury can reverse memory deficits associated with TBI. As such, pharmacological inhibition of the ISR emerges as a promising avenue to combat head trauma-induced chronic cognitive deficits.


Assuntos
Acetamidas/uso terapêutico , Lesões Encefálicas Traumáticas/tratamento farmacológico , Disfunção Cognitiva/tratamento farmacológico , Cicloexilaminas/uso terapêutico , Fator de Iniciação 2 em Eucariotos/metabolismo , Fosforilação/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/patologia , Disfunção Cognitiva/patologia , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/tratamento farmacológico , Memória Episódica , Camundongos , Camundongos Endogâmicos C57BL , Aprendizagem Espacial/efeitos dos fármacos , Aprendizagem Espacial/fisiologia
12.
Mol Cell ; 37(6): 797-808, 2010 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-20347422

RESUMO

The eIF4E-binding proteins (4E-BPs) repress translation initiation by preventing eIF4F complex formation. Of the three mammalian 4E-BPs, only 4E-BP2 is enriched in the mammalian brain and plays an important role in synaptic plasticity and learning and memory formation. Here we describe asparagine deamidation as a brain-specific posttranslational modification of 4E-BP2. Deamidation is the spontaneous conversion of asparagines to aspartates. Two deamidation sites were mapped to an asparagine-rich sequence unique to 4E-BP2. Deamidated 4E-BP2 exhibits increased binding to the mammalian target of rapamycin (mTOR)-binding protein raptor, which effects its reduced association with eIF4E. 4E-BP2 deamidation occurs during postnatal development, concomitant with the attenuation of the activity of the PI3K-Akt-mTOR signaling pathway. Expression of deamidated 4E-BP2 in 4E-BP2(-/-) neurons yielded mEPSCs exhibiting increased charge transfer with slower rise and decay kinetics relative to the wild-type form. 4E-BP2 deamidation may represent a compensatory mechanism for the developmental reduction of PI3K-Akt-mTOR signaling.


Assuntos
Encéfalo/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Processamento de Proteína Pós-Traducional , Transmissão Sináptica , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Células Cultivadas , Fatores de Iniciação em Eucariotos/química , Fatores de Iniciação em Eucariotos/deficiência , Fatores de Iniciação em Eucariotos/genética , Humanos , Cinética , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Especificidade de Órgãos , Fosforilação , Transporte Proteico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
13.
Brain Inj ; 32(1): 113-122, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29156991

RESUMO

PRIMARY OBJECTIVE: Repeated traumatic brain injuries (rmTBI) are frequently associated with debilitating neuropsychiatric conditions such as cognitive impairment, mood disorders, and post-traumatic stress disorder. We tested the hypothesis that repeated mild traumatic brain injury impairs spatial memory and enhances anxiety-like behaviour. RESEARCH DESIGN: We used a between groups design using single (smTBI) or repeated (rmTBI) controlled cranial closed skull impacts to mice, compared to a control group. METHODS AND PROCEDURES: We assessed the effects of smTBI and rmTBI using measures of motor performance (Rotarod Test [RT]), anxiety-like behaviour (Elevated Plus Maze [EPM] and Open Field [OF] tests), and spatial memory (Morris Water Maze [MWM]) within 12 days of the final injury. In separate groups of mice, astrocytosis and microglial activation were assessed 24 hours after the final injury using GFAP and IBA-1 immunohistochemistry. MAIN OUTCOMES AND RESULTS: RmTBI impaired spatial memory in the MWM and increased anxiety-like behaviour in the EPM and OFT. In addition, rmTBI elevated GFAP and IBA-1 immunohistochemistry throughout the mouse brain. RmTBI produced astrocytosis and microglial activation, and elicited impaired spatial memory and anxiety-like behaviour. CONCLUSIONS: rmTBI produces acute cognitive and anxiety-like disturbances associated with inflammatory changes in brain regions involved in spatial memory and anxiety.


Assuntos
Ansiedade/etiologia , Comportamento Animal/fisiologia , Concussão Encefálica/complicações , Encefalite/etiologia , Transtornos da Memória/etiologia , Memória Espacial/fisiologia , Animais , Ansiedade/patologia , Ansiedade/psicologia , Astrócitos/patologia , Encéfalo/patologia , Concussão Encefálica/patologia , Concussão Encefálica/psicologia , Encefalite/patologia , Encefalite/psicologia , Gliose/etiologia , Gliose/patologia , Gliose/psicologia , Masculino , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/patologia , Transtornos da Memória/psicologia , Camundongos , Microglia/patologia , Modelos Animais , Atividade Motora/fisiologia , Recidiva
14.
Hum Mol Genet ; 24(9): 2662-72, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25634563

RESUMO

Rett syndrome (RTT) is a severe neurodevelopmental disorder that is usually caused by mutations in Methyl-CpG-binding Protein 2 (MECP2). Four of the eight common disease causing mutations in MECP2 are nonsense mutations and are responsible for over 35% of all cases of RTT. A strategy to overcome disease-causing nonsense mutations is treatment with nonsense mutation suppressing drugs that allow expression of full-length proteins from mutated genes with premature in-frame stop codons. To determine if this strategy is useful in RTT, we characterized a new mouse model containing a knock-in nonsense mutation (p.R255X) in the Mecp2 locus (Mecp2(R255X)). To determine whether the truncated gene product acts as a dominant negative allele and if RTT-like phenotypes could be rescued by expression of wild-type protein, we genetically introduced an extra copy of MECP2 via an MECP2 transgene. The addition of MECP2 transgene to Mecp2(R255X) mice abolished the phenotypic abnormalities and resulted in near complete rescue. Expression of MECP2 transgene Mecp2(R255X) allele also rescued mTORC1 signaling abnormalities discovered in mice with loss of function and overexpression of Mecp2. Finally, we treated Mecp2(R255X) embryonic fibroblasts with the nonsense mutation suppressing drug gentamicin and we were able to induce expression of full-length MeCP2 from the mutant p.R255X allele. These data provide proof of concept that the p.R255X mutation of MECP2 is amenable to the nonsense suppression therapeutic strategy and provide guidelines for the extent of rescue that can be expected by re-expressing MeCP2 protein.


Assuntos
Alelos , Estudos de Associação Genética , Proteína 2 de Ligação a Metil-CpG/genética , Mutação , Fenótipo , Substituição de Aminoácidos , Animais , Comportamento Animal , Modelos Animais de Doenças , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica , Gentamicinas/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Proteína 2 de Ligação a Metil-CpG/metabolismo , Camundongos , Camundongos Transgênicos , Complexos Multiproteicos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome de Rett/diagnóstico , Síndrome de Rett/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transgenes
15.
J Neurosci ; 35(41): 13836-42, 2015 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-26468183

RESUMO

The mammalian target of rapamycin (mTOR) is a central regulator of a diverse array of cellular processes, including cell growth, proliferation, autophagy, translation, and actin polymerization. Components of the mTOR cascade are present at synapses and influence synaptic plasticity and spine morphogenesis. A prevailing view is that the study of mTOR and its role in autism spectrum disorders (ASDs) will elucidate the molecular mechanisms by which mTOR regulates neuronal function under physiological and pathological conditions. Although many ASDs arise as a result of mutations in genes with multiple molecular functions, they appear to converge on common biological pathways that give rise to autism-relevant behaviors. Dysregulation of mTOR signaling has been identified as a phenotypic feature common to fragile X syndrome, tuberous sclerosis complex 1 and 2, neurofibromatosis 1, phosphatase and tensin homolog, and potentially Rett syndrome. Below are a summary of topics covered in a symposium that presents dysregulation of mTOR as a unifying theme in a subset of ASDs.


Assuntos
Transtorno Autístico/metabolismo , Transtorno Autístico/patologia , Modelos Animais de Doenças , Transdução de Sinais/fisiologia , Sirolimo/metabolismo , Animais , Transtorno Autístico/fisiopatologia , Humanos , Modelos Biológicos
16.
PLoS Genet ; 9(12): e1004039, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24385930

RESUMO

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by maternal deficiency of the imprinted gene UBE3A. Individuals with AS suffer from intellectual disability, speech impairment, and motor dysfunction. Currently there is no cure for the disease. Here, we evaluated the phenotypic effect of activating the silenced paternal allele of Ube3a by depleting its antisense RNA Ube3a-ATS in mice. Premature termination of Ube3a-ATS by poly(A) cassette insertion activates expression of Ube3a from the paternal chromosome, and ameliorates many disease-related symptoms in the AS mouse model, including motor coordination defects, cognitive deficit, and impaired long-term potentiation. Studies on the imprinting mechanism of Ube3a revealed a pattern of biallelic transcription initiation with suppressed elongation of paternal Ube3a, implicating transcriptional collision between sense and antisense polymerases. These studies demonstrate the feasibility and utility of unsilencing the paternal copy of Ube3a via targeting Ube3a-ATS as a treatment for Angelman syndrome.


Assuntos
Síndrome de Angelman/genética , Deficiência Intelectual/patologia , Distúrbios da Fala/genética , Ubiquitina-Proteína Ligases/genética , Síndrome de Angelman/complicações , Síndrome de Angelman/patologia , Animais , Modelos Animais de Doenças , Inativação Gênica , Impressão Genômica , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Camundongos , Terapia de Alvo Molecular , Neurônios/metabolismo , RNA Antissenso/genética , Distúrbios da Fala/complicações , Distúrbios da Fala/patologia , Transcrição Gênica , Ubiquitina-Proteína Ligases/biossíntese , Ubiquitina-Proteína Ligases/metabolismo
17.
Nature ; 452(7185): 323-8, 2008 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-18272964

RESUMO

Transcriptional activation of cytokines, such as type-I interferons (interferon (IFN)-alpha and IFN-beta), constitutes the first line of antiviral defence. Here we show that translational control is critical for induction of type-I IFN production. In mouse embryonic fibroblasts lacking the translational repressors 4E-BP1 and 4E-BP2, the threshold for eliciting type-I IFN production is lowered. Consequently, replication of encephalomyocarditis virus, vesicular stomatitis virus, influenza virus and Sindbis virus is markedly suppressed. Furthermore, mice with both 4E- and 4E-BP2 genes (also known as Eif4ebp1 and Eif4ebp2, respectively) knocked out are resistant to vesicular stomatitis virus infection, and this correlates with an enhanced type-I IFN production in plasmacytoid dendritic cells and the expression of IFN-regulated genes in the lungs. The enhanced type-I IFN response in 4E-BP1-/- 4E-BP2-/- double knockout mouse embryonic fibroblasts is caused by upregulation of interferon regulatory factor 7 (Irf7) messenger RNA translation. These findings highlight the role of 4E-BPs as negative regulators of type-I IFN production, via translational repression of Irf7 mRNA.


Assuntos
Imunidade Inata/imunologia , Fator Regulador 7 de Interferon/biossíntese , Biossíntese de Proteínas , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Células Dendríticas/imunologia , Embrião de Mamíferos/citologia , Fatores de Iniciação em Eucariotos/deficiência , Fatores de Iniciação em Eucariotos/genética , Fatores de Iniciação em Eucariotos/metabolismo , Fibroblastos/virologia , Deleção de Genes , Imunidade Inata/genética , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Camundongos , Camundongos Knockout , Fosfoproteínas/deficiência , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vírus da Estomatite Vesicular Indiana/fisiologia , Fenômenos Fisiológicos Virais , Replicação Viral
18.
Proc Natl Acad Sci U S A ; 108(9): 3791-6, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21307309

RESUMO

Both the formation of long-term memory (LTM) and late-long-term potentiation (L-LTP), which is thought to represent the cellular model of learning and memory, require de novo protein synthesis. The mammalian target of Rapamycin (mTOR) complex I (mTORC1) integrates information from various synaptic inputs and its best characterized function is the regulation of translation. Although initial studies have shown that rapamycin reduces L-LTP and partially blocks LTM, recent genetic and pharmacological evidence indicating that mTORC1 promotes L-LTP and LTM is controversial. Thus, the role of mTORC1 in L-LTP and LTM is unclear. To selectively inhibit mTORC1 activity in the adult brain, we used a "pharmacogenetic" approach that relies on the synergistic action of a drug (rapamycin) and a genetic manipulation (mTOR heterozygotes, mTOR(+/-) mice) on the same target (mTORC1). Although L-LTP and LTM are normal in mTOR(+/-) mice, application of a low concentration of rapamycin-one that is subthreshold for WT mice-prevented L-LTP and LTM only in mTOR(+/-) mice. Furthermore, we found that mTORC1-mediated translational control is required for memory reconsolidation. We provide here direct genetic evidence supporting the role of mTORC1 in L-LTP and behavioral memory.


Assuntos
Potenciação de Longa Duração/efeitos dos fármacos , Memória de Longo Prazo/efeitos dos fármacos , Proteínas/antagonistas & inibidores , Proteínas/genética , Sirolimo/farmacologia , Estimulação Acústica , Animais , Medo/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Farmacogenética , Serina-Treonina Quinases TOR , Transativadores/metabolismo , Fatores de Transcrição
19.
Cell Host Microbe ; 32(1): 106-116.e6, 2024 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-38113884

RESUMO

Autism spectrum disorder (ASD) is characterized by the presence of restricted/repetitive behaviors and social communication deficits. Because effective treatments for ASD remain elusive, novel therapeutic strategies are necessary. Preclinical studies show that L. reuteri selectively reversed social deficits in several models for ASD. Here, in a double-blind, randomized, placebo-controlled trial, we tested the effect of L. reuteri (a product containing a combination of strains ATCC-PTA-6475 and DSM-17938) in children with ASD. The treatment does not alter overall autism severity, restricted/repetitive behaviors, the microbiome composition, or the immune profile. However, L. reuteri combination yields significant improvements in social functioning that generalized across different measures. Interestingly, ATCC-PTA-6475, but not the parental strain of DSM-17938, reverses the social deficits in a preclinical mouse model for ASD. Collectively, our findings show that L. reuteri enhances social behavior in children with ASD, thereby warranting larger trials in which strain-specific effects should also be investigated.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Criança , Camundongos , Animais , Humanos , Transtorno Autístico/terapia , Transtorno do Espectro Autista/terapia , Comportamento Social , Resultado do Tratamento , Método Duplo-Cego
20.
Prog Neurobiol ; 237: 102612, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38642602

RESUMO

Recurrent seizures lead to accumulation of the activity-dependent transcription factor ∆FosB in hippocampal dentate granule cells in both mouse models of epilepsy and mouse models of Alzheimer's disease (AD), which is also associated with increased incidence of seizures. In patients with AD and related mouse models, the degree of ∆FosB accumulation corresponds with increasing severity of cognitive deficits. We previously found that ∆FosB impairs spatial memory in mice by epigenetically regulating expression of target genes such as calbindin that are involved in synaptic plasticity. However, the suppression of calbindin in conditions of neuronal hyperexcitability has been demonstrated to provide neuroprotection to dentate granule cells, indicating that ∆FosB may act over long timescales to coordinate neuroprotective pathways. To test this hypothesis, we used viral-mediated expression of ∆JunD to interfere with ∆FosB signaling over the course of several months in transgenic mice expressing mutant human amyloid precursor protein (APP), which exhibit spontaneous seizures and develop AD-related neuropathology and cognitive deficits. Our results demonstrate that persistent ∆FosB activity acts through discrete modes of hippocampal target gene regulation to modulate neuronal excitability, limit recurrent seizure activity, and provide neuroprotection to hippocampal dentate granule cells in APP mice.


Assuntos
Precursor de Proteína beta-Amiloide , Giro Denteado , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-fos , Convulsões , Animais , Giro Denteado/metabolismo , Camundongos , Convulsões/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/genética , Neuroproteção/fisiologia , Modelos Animais de Doenças , Doença de Alzheimer/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA