Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 43(4): 114056, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38581678

RESUMO

Little is known of the brain mechanisms that mediate sex-specific autism symptoms. Here, we demonstrate that deletion of the autism spectrum disorder (ASD)-risk gene, Pten, in neocortical pyramidal neurons (NSEPten knockout [KO]) results in robust cortical circuit hyperexcitability selectively in female mice observed as prolonged spontaneous persistent activity states. Circuit hyperexcitability in females is mediated by metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) signaling to mitogen-activated protein kinases (Erk1/2) and de novo protein synthesis. Pten KO layer 5 neurons have a female-specific increase in mGluR5 and mGluR5-dependent protein synthesis. Furthermore, mGluR5-ERα complexes are generally elevated in female cortices, and genetic reduction of ERα rescues enhanced circuit excitability, protein synthesis, and neuron size selectively in NSEPten KO females. Female NSEPten KO mice display deficits in sensory processing and social behaviors as well as mGluR5-dependent seizures. These results reveal mechanisms by which sex and a high-confidence ASD-risk gene interact to affect brain function and behavior.


Assuntos
Transtorno Autístico , Modelos Animais de Doenças , Receptor alfa de Estrogênio , Camundongos Knockout , Neocórtex , PTEN Fosfo-Hidrolase , Receptor de Glutamato Metabotrópico 5 , Animais , Feminino , Masculino , Camundongos , Transtorno Autístico/metabolismo , Transtorno Autístico/fisiopatologia , Transtorno Autístico/genética , Transtorno Autístico/patologia , Receptor alfa de Estrogênio/metabolismo , Camundongos Endogâmicos C57BL , Neocórtex/metabolismo , Neocórtex/patologia , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Células Piramidais/metabolismo , Receptor de Glutamato Metabotrópico 5/metabolismo , Comportamento Social
2.
bioRxiv ; 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-37609208

RESUMO

Autism manifests differently in males and females and the brain mechanisms that mediate these sex-dependent differences are unknown. Here, we demonstrate that deletion of the ASD-risk gene, Pten, in neocortical pyramidal neurons (NSE Pten KO) results in robust hyperexcitability of local neocortical circuits in female, but not male, mice, observed as prolonged, spontaneous persistent activity states (UP states). Circuit hyperexcitability in NSE Pten KO mice is mediated by enhanced and/or altered signaling of metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) to ERK and protein synthesis selectively in Pten deleted female neurons. In support of this idea, Pten deleted Layer 5 cortical neurons have female-specific increases in mGluR5 and mGluR5-driven protein synthesis. In addition, mGluR5-ERα complexes are elevated in female cortex and genetic reduction of ERα in Pten KO cortical neurons rescues circuit excitability, protein synthesis and enlarged neurons selectively in females. Abnormal timing and hyperexcitability of neocortical circuits in female NSE Pten KO mice are associated with deficits in temporal processing of sensory stimuli and social behaviors as well as mGluR5-dependent seizures. Female-specific cortical hyperexcitability and mGluR5-dependent seizures are also observed in a human disease relevant mouse model, germline Pten +/- mice. Our results reveal molecular mechanisms by which sex and a high impact ASD-risk gene interact to affect brain function and behavior.

3.
Cell Rep ; 37(7): 109997, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34788630

RESUMO

The anorexigenic effect of serotonergic compounds has largely been attributed to activation of serotonin 2C receptors (Htr2cs). Using mouse genetic models in which Htr2c can be selectively deleted or restored (in Htr2c-null mice), we investigate the role of Htr2c in forebrain Sim1 neurons. Unexpectedly, we find that Htr2c acts in these neurons to promote food intake and counteract the anorectic effect of serotonergic appetite suppressants. Furthermore, Htr2c marks a subset of Sim1 neurons in the paraventricular nucleus of the hypothalamus (PVH). Chemogenetic activation of these neurons in adult mice suppresses hunger, whereas their silencing promotes feeding. In support of an orexigenic role of PVH Htr2c, whole-cell patch-clamp experiments demonstrate that activation of Htr2c inhibits PVH neurons. Intriguingly, this inhibition is due to Gαi/o-dependent activation of ATP-sensitive K+ conductance, a mechanism of action not identified previously in the mammalian nervous system.


Assuntos
Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Animais , Anorexia , Depressores do Apetite/metabolismo , Depressores do Apetite/farmacologia , Metabolismo Energético/fisiologia , Comportamento Alimentar/fisiologia , Fome/fisiologia , Hipotálamo/metabolismo , Hipotálamo/fisiologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Neurônios/fisiologia , Núcleo Hipotalâmico Paraventricular/citologia , Núcleo Hipotalâmico Paraventricular/fisiologia , Potássio/metabolismo , Receptor 5-HT2C de Serotonina/genética , Serotonina/metabolismo , Serotonina/farmacologia , Serotoninérgicos
4.
ACS Chem Neurosci ; 12(16): 3038-3048, 2021 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-34370453

RESUMO

Novel treatments, screening, and detection methods have prolonged the lives of numerous cancer patients worldwide. Unfortunately, existing and many promising new chemotherapeutics can cause deleterious, off-target side effects in normal tissue and organ systems. The central and peripheral nervous systems are widely recognized as frequent off-target effectors of anticancer drugs which can produce persistent neurological and neuropsychiatric symptoms collectively termed "chemobrain". Following chemotherapy, patients report several forms of cognitive impairment occurring acutely and sometimes persisting years after treatment. There are no effective treatments for cognitive decline induced by chemotherapeutics, and the underlying molecular mechanisms are poorly characterized and understood. In this study, we find that chronic treatment with two common chemotherapeutic agents, cisplatin and gemcitabine, impairs brain region-specific metabolism, hippocampus-dependent memory formation, and stress response behavior. This corresponds to reduced hippocampal synaptic excitability, altered neuronal signal transduction, and neuroinflammation. These findings underline that a better understanding of the basic pathological consequences of chemotherapy-induced cognitive impairment is the first step toward improving cancer treatment survivorship.


Assuntos
Antineoplásicos , Doenças do Sistema Nervoso , Preparações Farmacêuticas , Antineoplásicos/efeitos adversos , Cisplatino , Hipocampo , Humanos
5.
J Exp Med ; 218(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33978701

RESUMO

Atypical antipsychotics such as risperidone cause drug-induced metabolic syndrome. However, the underlying mechanisms remain largely unknown. Here, we report a new mouse model that reliably reproduces risperidone-induced weight gain, adiposity, and glucose intolerance. We found that risperidone treatment acutely altered energy balance in C57BL/6 mice and that hyperphagia accounted for most of the weight gain. Transcriptomic analyses in the hypothalamus of risperidone-fed mice revealed that risperidone treatment reduced the expression of Mc4r. Furthermore, Mc4r in Sim1 neurons was necessary for risperidone-induced hyperphagia and weight gain. Moreover, we found that the same pathway underlies the obesogenic effect of olanzapine-another commonly prescribed antipsychotic drug. Remarkably, whole-cell patch-clamp recording demonstrated that risperidone acutely inhibited the activity of hypothalamic Mc4r neurons via the opening of a postsynaptic potassium conductance. Finally, we showed that treatment with setmelanotide, an MC4R-specific agonist, mitigated hyperphagia and obesity in both risperidone- and olanzapine-fed mice.


Assuntos
Antipsicóticos/farmacologia , Receptor Tipo 4 de Melanocortina/metabolismo , Risperidona/farmacologia , Aumento de Peso/efeitos dos fármacos , Animais , Feminino , Hiperfagia/metabolismo , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Obesidade/metabolismo , Olanzapina/farmacologia , Potássio/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , alfa-MSH/análogos & derivados , alfa-MSH/farmacologia
6.
Sci Rep ; 10(1): 2737, 2020 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-32066765

RESUMO

Astronauts on interplanetary missions - such as to Mars - will be exposed to space radiation, a spectrum of highly-charged, fast-moving particles that includes 56Fe and 28Si. Earth-based preclinical studies show space radiation decreases rodent performance in low- and some high-level cognitive tasks. Given astronaut use of touchscreen platforms during training and space flight and given the ability of rodent touchscreen tasks to assess functional integrity of brain circuits and multiple cognitive domains in a non-aversive way, here we exposed 6-month-old C57BL/6J male mice to whole-body space radiation and subsequently assessed them on a touchscreen battery. Relative to Sham treatment, 56Fe irradiation did not overtly change performance on tasks of visual discrimination, reversal learning, rule-based, or object-spatial paired associates learning, suggesting preserved functional integrity of supporting brain circuits. Surprisingly, 56Fe irradiation improved performance on a dentate gyrus-reliant pattern separation task; irradiated mice learned faster and were more accurate than controls. Improved pattern separation performance did not appear to be touchscreen-, radiation particle-, or neurogenesis-dependent, as 56Fe and 28Si irradiation led to faster context discrimination in a non-touchscreen task and 56Fe decreased new dentate gyrus neurons relative to Sham. These data urge revisitation of the broadly-held view that space radiation is detrimental to cognition.


Assuntos
Cognição/efeitos da radiação , Radiação Cósmica , Giro Denteado/efeitos da radiação , Aprendizagem por Associação de Pares/efeitos da radiação , Reconhecimento Visual de Modelos/efeitos da radiação , Reversão de Aprendizagem/efeitos da radiação , Animais , Astronautas , Ciências Biocomportamentais , Cognição/fisiologia , Giro Denteado/fisiologia , Isótopos de Ferro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Neurônios/fisiologia , Neurônios/efeitos da radiação , Aprendizagem por Associação de Pares/fisiologia , Reconhecimento Visual de Modelos/fisiologia , Reversão de Aprendizagem/fisiologia , Voo Espacial , Irradiação Corporal Total
7.
Mol Psychiatry ; 25(11): 2832-2843, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-30038231

RESUMO

Recent findings from in vivo-imaging and human post-mortem tissue studies in schizophrenic psychosis (SzP), have demonstrated functional and molecular changes in hippocampal subfields that can be associated with hippocampal hyperexcitability. In this study, we used a subfield-specific GluN1 knockout mouse with a disease-like molecular perturbation expressed only in hippocampal dentate gyrus (DG) and assessed its association with hippocampal physiology and psychosis-like behaviors. First, we used whole-cell patch-clamp recordings to measure the physiological changes in hippocampal subfields and cFos immunohistochemistry to examine cellular excitability. DG-GluN1 KO mice show CA3 cellular hyperactivity, detected using two approaches: (1) increased excitatory glutamate transmission at mossy fibers (MF)-CA3 synapses, and (2) an increased number of cFos-activated pyramidal neurons in CA3, an outcome that appears to project downstream to CA1 and basolateral amygdala (BLA). Furthermore, we examined psychosis-like behaviors and pathological memory processing; these show an increase in fear conditioning (FC), a reduction in prepulse inhibition (PPI) in the KO animal, along with a deterioration in memory accuracy with Morris Water Maze (MWM) and reduced social memory (SM). Moreover, with DREADD vectors, we demonstrate a remarkably similar behavioral profile when we induce CA3 hyperactivity. These hippocampal subfield changes could provide the basis for the observed increase in human hippocampal activity in SzP, based on the shared DG-specific GluN1 reduction. With further characterization, these animal model systems may serve as targets to test psychosis mechanisms related to hippocampus and assess potential hippocampus-directed treatments.


Assuntos
Região CA3 Hipocampal/fisiopatologia , Giro Denteado/metabolismo , Proteínas do Tecido Nervoso/deficiência , Transtornos Psicóticos/fisiopatologia , Receptores de N-Metil-D-Aspartato/deficiência , Animais , Região CA3 Hipocampal/citologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Piramidais
8.
Hippocampus ; 29(8): 726-735, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30779299

RESUMO

Dentate gyrus adult neurogenesis is implicated in the formation of hippocampal-dependent contextual associations. However, the role of adult neurogenesis during reward-based context-dependent paradigms-such as conditioned place preference (CPP)-is understudied. Therefore, we used image-guided, hippocampal-targeted X-ray irradiation (IG-IR) and morphine CPP to explore whether dentate gyrus adult neurogenesis plays a role in reward memories created in adult C57BL/6J male mice. In addition, as adult neurogenesis appears to participate to a greater extent in retrieval and extinction of recent (<48 hr posttraining) versus remote (>1 week posttraining) memories, we specifically examined the role of adult neurogenesis in reward-associated contextual memories probed at recent and remote timepoints. Six weeks post-IG-IR or Sham treatment, mice underwent morphine CPP. Using separate groups, retrieval of recent and remote reward memories was found to be similar between IG-IR and Sham treatments. Interestingly, IG-IR mice showed impaired extinction-or increased persistence-of the morphine-associated reward memory when it was probed 24-hr (recent) but not 3-weeks (remote) postconditioning relative to Sham mice. Taken together, these data show that hippocampal-directed irradiation and the associated decrease in dentate gyrus adult neurogenesis affect the persistence of recently-but not remotely-probed reward memory. These data indicate a novel role for adult neurogenesis in reward-based memories and particularly the extinction rate of these memories. Consideration of this work may lead to better understanding of extinction-based behavioral interventions for psychiatric conditions characterized by dysregulated reward processing.


Assuntos
Giro Denteado/fisiologia , Extinção Psicológica/fisiologia , Memória/fisiologia , Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Neurogênese/fisiologia , Recompensa , Animais , Irradiação Craniana/métodos , Giro Denteado/efeitos dos fármacos , Giro Denteado/efeitos da radiação , Extinção Psicológica/efeitos dos fármacos , Extinção Psicológica/efeitos da radiação , Masculino , Memória/efeitos dos fármacos , Memória/efeitos da radiação , Camundongos , Neurogênese/efeitos dos fármacos , Neurogênese/efeitos da radiação , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/efeitos da radiação
9.
Autism Res ; 11(2): 234-244, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29028156

RESUMO

Neuroligin-3 (NLGN3) is a postsynaptic cell adhesion protein that interacts with presynaptic ligands including neurexin-1 (NRXN1) [Ichtchenko et al., Journal of Biological Chemistry, 271, 2676-2682, 1996]. Mice harboring a mutation in the NLGN3 gene (NL3R451C) mimicking a mutation found in two brothers with autism spectrum disorder (ASD) were previously generated and behaviorally phenotyped for autism-related behaviors. In these NL3R451C mice generated and tested on a hybrid C57BL6J/129S2/SvPasCrl background, we observed enhanced spatial memory and reduced social interaction [Tabuchi et al., Science, 318, 71-76, 2007]. Curiously, an independently generated second line of mice harboring the same mutation on a C57BL6J background exhibited minimal aberrant behavior, thereby providing apparently discrepant results. To investigate the origin of the discrepancy, we previously replicated the original findings of Tabuchi et al. by studying the same NL3R451C mutation on a pure 129S2/SvPasCrl genetic background. Here we complete the behavioral characterization of the NL3R451C mutation on a pure C57BL6J genetic background to determine if background genetics play a role in the discrepant behavioral outcomes involving NL3R451C mice. NL3R451C mutant mice on a pure C57BL6J background did not display spatial memory enhancements or social interaction deficits. We only observed a decreased startle response and mildly increased locomotor activity in these mice suggesting that background genetics influences behavioral outcomes involving the NL3R451C mutation. Autism Res 2018, 11: 234-244. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Behavioral symptoms of autism can be highly variable, even in cases that involve identical genetic mutations. Previous studies in mice with a mutation of the Neuroligin-3 gene showed enhanced learning and social deficits. We replicated these findings on the same and different genetic backgrounds. In this study, however, the same mutation in mice on a different genetic background did not reproduce our previous findings. Our results suggest that genetic background influences behavioral symptoms of this autism-associated mutation.


Assuntos
Transtorno do Espectro Autista/genética , Moléculas de Adesão Celular Neuronais/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Comportamento Problema/psicologia , Animais , Transtorno do Espectro Autista/psicologia , Modelos Animais de Doenças , Feminino , Patrimônio Genético , Relações Interpessoais , Aprendizagem/fisiologia , Masculino , Rememoração Mental/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Fenótipo , Reflexo de Sobressalto/genética , Aprendizagem Espacial/fisiologia
10.
Nat Commun ; 8(1): 1668, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29162814

RESUMO

Adult neurogenesis persists in the rodent dentate gyrus and is stimulated by chronic treatment with conventional antidepressants through BDNF/TrkB signaling. Ketamine in low doses produces both rapid and sustained antidepressant effects in patients. Previous studies have shed light on post-transcriptional synaptic NMDAR mediated mechanisms underlying the acute effect, but how ketamine acts at the cellular level to sustain this anti-depressive function for prolonged periods remains unclear. Here we report that ketamine accelerates differentiation of doublecortin-positive adult hippocampal neural progenitors into functionally mature neurons. This process requires TrkB-dependent ERK pathway activation. Genetic ablation of TrkB in neural stem/progenitor cells, or pharmacologic disruption of ERK signaling, or inhibition of adult neurogenesis, each blocks the ketamine-induced behavioral responses. Conversely, enhanced ERK activity via Nf1 gene deletion extends the response and rescues both neurogenic and behavioral deficits in mice lacking TrkB. Thus, TrkB-dependent neuronal differentiation is involved in the sustained antidepressant effects of ketamine.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Ketamina/farmacologia , Células-Tronco Neurais/metabolismo , Receptor trkB/metabolismo , Analgésicos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Feminino , Hipocampo/citologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco Neurais/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/fisiologia , Receptor trkB/genética
11.
J Neurosci ; 37(45): 10917-10931, 2017 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-28978667

RESUMO

Genetic perturbations of the transcription factor Forkhead Box P1 (FOXP1) are causative for severe forms of autism spectrum disorder that are often comorbid with intellectual disability. Recent work has begun to reveal an important role for FoxP1 in brain development, but the brain-region-specific contributions of Foxp1 to autism and intellectual disability phenotypes have yet to be determined fully. Here, we describe Foxp1 conditional knock-out (Foxp1cKO) male and female mice with loss of Foxp1 in the pyramidal neurons of the neocortex and the CA1/CA2 subfields of the hippocampus. Foxp1cKO mice exhibit behavioral phenotypes that are of potential relevance to autism spectrum disorder, including hyperactivity, increased anxiety, communication impairments, and decreased sociability. In addition, Foxp1cKO mice have gross deficits in learning and memory tasks of relevance to intellectual disability. Using a genome-wide approach, we identified differentially expressed genes in the hippocampus of Foxp1cKO mice associated with synaptic function and development. Furthermore, using magnetic resonance imaging, we uncovered a significant reduction in the volumes of both the entire hippocampus as well as individual hippocampal subfields of Foxp1cKO mice. Finally, we observed reduced maintenance of LTP in area CA1 of the hippocampus in these mutant mice. Together, these data suggest that proper expression of Foxp1 in the pyramidal neurons of the forebrain is important for regulating gene expression pathways that contribute to specific behaviors reminiscent of those seen in autism and intellectual disability. In particular, Foxp1 regulation of gene expression appears to be crucial for normal hippocampal development, CA1 plasticity, and spatial learning.SIGNIFICANCE STATEMENT Loss-of-function mutations in the transcription factor Forkhead Box P1 (FOXP1) lead to autism spectrum disorder and intellectual disability. Understanding the potential brain-region-specific contributions of FOXP1 to disease-relevant phenotypes could be a critical first step in the management of patients with these mutations. Here, we report that Foxp1 conditional knock-out (Foxp1cKO) mice with loss of Foxp1 in the neocortex and hippocampus display autism and intellectual-disability-relevant behaviors. We also show that these phenotypes correlate with changes in both the genomic and physiological profiles of the hippocampus in Foxp1cKO mice. Our work demonstrates that brain-region-specific FOXP1 expression may relate to distinct, clinically relevant phenotypes.


Assuntos
Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/fisiologia , Expressão Gênica/genética , Expressão Gênica/fisiologia , Plasticidade Neuronal/genética , Plasticidade Neuronal/fisiologia , Células Piramidais/fisiologia , Proteínas Repressoras/genética , Proteínas Repressoras/fisiologia , Aprendizagem Espacial/fisiologia , Sinapses/fisiologia , Animais , Transtorno do Espectro Autista , Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiologia , Feminino , Deficiências da Aprendizagem/genética , Deficiências da Aprendizagem/psicologia , Masculino , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neocórtex/citologia , Neocórtex/fisiologia , Células Piramidais/metabolismo , Vocalização Animal/fisiologia
12.
Dev Biol ; 431(2): 179-193, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28947178

RESUMO

While several studies indicate the importance of ephrin-B/EphB bidirectional signaling in excitatory neurons, potential roles for these molecules in inhibitory neurons are largely unknown. We identify here an autonomous receptor-like role for ephrin-B reverse signaling in the tangential migration of interneurons into the neocortex using ephrin-B (EfnB1/B2/B3) conditional triple mutant (TMlz) mice and a forebrain inhibitory neuron specific Cre driver. Inhibitory neuron deletion of the three EfnB genes leads to reduced interneuron migration, abnormal cortical excitability, and lethal audiogenic seizures. Truncated and intracellular point mutations confirm the importance of ephrin-B reverse signaling in interneuron migration and cortical excitability. A non-autonomous ligand-like role was also identified for ephrin-B2 that is expressed in neocortical radial glial cells and required for proper tangential migration of GAD65-positive interneurons. Our studies thus define both receptor-like and ligand-like roles for the ephrin-B molecules in controlling the migration of interneurons as they populate the neocortex and help establish excitatory/inhibitory (E/I) homeostasis.


Assuntos
Movimento Celular , Efrinas/metabolismo , Interneurônios/citologia , Interneurônios/metabolismo , Animais , Feminino , Deleção de Genes , Ligantes , Camundongos , Modelos Biológicos , Mutação/genética , Neocórtex/citologia , Neocórtex/metabolismo , Inibição Neural , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Pseudópodes/metabolismo
13.
Elife ; 62017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28695822

RESUMO

Sequencing studies have implicated haploinsufficiency of ARID1B, a SWI/SNF chromatin-remodeling subunit, in short stature (Yu et al., 2015), autism spectrum disorder (O'Roak et al., 2012), intellectual disability (Deciphering Developmental Disorders Study, 2015), and corpus callosum agenesis (Halgren et al., 2012). In addition, ARID1B is the most common cause of Coffin-Siris syndrome, a developmental delay syndrome characterized by some of the above abnormalities (Santen et al., 2012; Tsurusaki et al., 2012; Wieczorek et al., 2013). We generated Arid1b heterozygous mice, which showed social behavior impairment, altered vocalization, anxiety-like behavior, neuroanatomical abnormalities, and growth impairment. In the brain, Arid1b haploinsufficiency resulted in changes in the expression of SWI/SNF-regulated genes implicated in neuropsychiatric disorders. A focus on reversible mechanisms identified Insulin-like growth factor (IGF1) deficiency with inadequate compensation by Growth hormone-releasing hormone (GHRH) and Growth hormone (GH), underappreciated findings in ARID1B patients. Therapeutically, GH supplementation was able to correct growth retardation and muscle weakness. This model functionally validates the involvement of ARID1B in human disorders, and allows mechanistic dissection of neurodevelopmental diseases linked to chromatin-remodeling.


Assuntos
Deficiências do Desenvolvimento/genética , Haploinsuficiência , Transtornos Mentais/genética , Fatores de Transcrição/deficiência , Animais , Comportamento Animal , Encéfalo/patologia , Deficiências do Desenvolvimento/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Hormônio Liberador de Hormônio do Crescimento/metabolismo , Heterozigoto , Fator de Crescimento Insulin-Like I/metabolismo , Transtornos Mentais/fisiopatologia , Camundongos , Proteínas Serina-Treonina Quinases/metabolismo
14.
J Neuroinflammation ; 13(1): 171, 2016 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-27357503

RESUMO

BACKGROUND: A traumatic brain injury (TBI) event is a devastating injury to the brain that may result in heightened inflammation, neurodegeneration, and subsequent cognitive and mood deficits. TBI victims with co-morbidities such as heart disease, diabetes, or obesity may be more vulnerable to the secondary brain injury that follows the initial insult. Compared to lean individuals, obese subjects tend to have worse clinical outcomes and higher mortality rates after trauma. METHODS: To elucidate whether obesity predisposes individuals to worse outcomes after TBI, we subjected adult lean and obese male/female mice to a mild TBI. The injury was administered using a controlled skull impact (CSI) device. Lean or obese 6-month-old C57 BL/6 mice were subjected once to a mild TBI. Additionally, at day 30 after injury, both the lean and obese mice were tested for increased anxiety using the open field test. RESULTS: At day 30 after TBI, compared to the lean mice, we found heightened microglial (MG) activation in the cerebral cortex, corpus callosum, and hypothalamus. Another compelling finding was that, compared to the non-injured obese male control mice, the obese TBI mice had a decrease in the rate of weight gain and serum corticosterone levels at day 30 after injury. Additionally, the injured obese mice displayed higher levels of anxiety as determined by a significant decrease in time spent in the non-peripheral zones in the open field test. In contrast to the obese males, the obese female mice did not exhibit increases in the number of active MG in the brain, changes in weight gain/corticosterone levels, or increased anxiety at day 30 after TBI. CONCLUSIONS: The data presented here suggests that obese mice have worse outcomes compared to lean mice after mild TBI. Also, the obese males have worse outcomes than the injured female mice. This data may explain the sequela of chronic secondary brain injury in obese adults after a single mild TBI. Also, this report may help shape how the overweight/obese populations are monitored over the days and months following a TBI.


Assuntos
Concussão Encefálica/metabolismo , Concussão Encefálica/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Obesidade/metabolismo , Obesidade/patologia , Animais , Concussão Encefálica/complicações , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Obesidade/complicações
15.
J Biol Chem ; 291(22): 11647-56, 2016 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-27008863

RESUMO

The biological underpinnings and the pathological lesions of psychiatric disorders are centuries-old questions that have yet to be understood. Recent studies suggest that schizophrenia and related disorders likely have their origins in perturbed neurodevelopment and can result from a large number of common genetic variants or multiple, individually rare genetic alterations. It is thus conceivable that key neurodevelopmental pathways underline the various genetic changes and the still unknown pathological lesions in schizophrenia. Here, we report that mice defective of the nicastrin subunit of γ-secretase in oligodendrocytes have hypomyelination in the central nervous system. These mice have altered dopamine signaling and display profound abnormal phenotypes reminiscent of schizophrenia. In addition, we identify an association of the nicastrin gene with a human schizophrenia cohort. These observations implicate γ-secretase and its mediated neurodevelopmental pathways in schizophrenia and provide support for the "myelination hypothesis" of the disease. Moreover, by showing that schizophrenia and obsessive-compulsive symptoms could be modeled in animals wherein a single genetic factor is altered, our work provides a biological basis that schizophrenia with obsessive-compulsive disorder is a distinct subtype of schizophrenia.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Comportamento Compulsivo , Glicoproteínas de Membrana/metabolismo , Bainha de Mielina/metabolismo , Oligodendroglia/metabolismo , Esquizofrenia/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Animais , Feminino , Humanos , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Esquizofrenia/genética
16.
Mol Cell Biol ; 36(1): 70-83, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26459759

RESUMO

Synapse development requires normal neuronal activities and the precise expression of synapse-related genes. Dysregulation of synaptic genes results in neurological diseases such as autism spectrum disorders (ASD). Mutations in genes encoding chromatin-remodeling factor Brg1/SmarcA4 and its associated proteins are the genetic causes of several developmental diseases with neurological defects and autistic symptoms. Recent large-scale genomic studies predicted Brg1/SmarcA4 as one of the key nodes of the ASD gene network. We report that Brg1 deletion in early postnatal hippocampal neurons led to reduced dendritic spine density and maturation and impaired synapse activities. In developing mice, neuronal Brg1 deletion caused severe neurological defects. Gene expression analyses indicated that Brg1 regulates a significant number of genes known to be involved in synapse function and implicated in ASD. We found that Brg1 is required for dendritic spine/synapse elimination mediated by the ASD-associated transcription factor myocyte enhancer factor 2 (MEF2) and that Brg1 regulates the activity-induced expression of a specific subset of genes that overlap significantly with the targets of MEF2. Our analyses showed that Brg1 interacts with MEF2 and that MEF2 is required for Brg1 recruitment to target genes in response to neuron activation. Thus, Brg1 plays important roles in both synapse development/maturation and MEF2-mediated synapse remodeling. Our study reveals specific functions of the epigenetic regulator Brg1 in synapse development and provides insights into its role in neurological diseases such as ASD.


Assuntos
DNA Helicases/metabolismo , Fatores de Transcrição MEF2/metabolismo , Neurônios/metabolismo , Proteínas Nucleares/metabolismo , Sinapses/metabolismo , Fatores de Transcrição/metabolismo , Animais , Transtorno Autístico/genética , Transtorno Autístico/metabolismo , Cromatina/metabolismo , DNA Helicases/genética , Espinhas Dendríticas/genética , Hipocampo/metabolismo , Fatores de Transcrição MEF2/genética , Camundongos , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Fatores de Transcrição/genética
17.
Mol Genet Metab ; 116(1-2): 98-105, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25982063

RESUMO

The neuronal ceroid lipofuscinoses (NCLs) are a group of related hereditary lysosomal storage disorders characterized by progressive loss of neurons in the central nervous system resulting in dementia, loss of motor skills, seizures and blindness. A characteristic intralysosomal accumulation of autofluorescent storage material occurs in the brain and other tissues. Three major forms and nearly a dozen minor forms of NCL are recognized. Infantile-onset NCL (CLN1 disease) is caused by severe deficiency in a soluble lysosomal enzyme, palmitoyl-protein thioesterase-1 (PPT1) and no therapy beyond supportive care is available. Homozygous Ppt1 knockout mice reproduce the known features of the disease, developing signs of motor dysfunction at 5 months of age and death around 8 months. Direct delivery of lysosomal enzymes to the cerebrospinal fluid is an approach that has gained traction in small and large animal models of several other neuropathic lysosomal storage diseases, and has advanced to clinical trials. In the current study, Ppt1 knockout mice were treated with purified recombinant human PPT1 enzyme delivered to the lumbar intrathecal space on each of three consecutive days at 6 weeks of age. Untreated PPT1 knockout mice and wild-type mice served as additional controls. Four enzyme concentration levels (0, 2.6, 5.3 and 10.6 mg/ml of specific activity 20 U/mg) were administered in a volume of 80 µl infused over 8 min. Each group consisted of 16-20 mice. The treatment was well tolerated. Disease-specific survival was 233, 267, 272, and 284days for each of the four treatment groups, respectively, and the effect of treatment was highly significant (p<0.0001). The timing of motor deterioration was also delayed. Neuropathology was improved as evidenced by decreased autofluorescent storage material in the spinal cord and a decrease in CD68 staining in the cortex and spinal cord. The improvements in motor function and survival are similar to results reported for preclinical studies involving other lysosomal storage disorders, such as CLN2/TPP1 deficiency, for which intraventricular ERT is being offered in clinical trials. If ERT delivery to the CSF proves to be efficacious in these disorders, PPT1 deficiency may also be amenable to this approach.


Assuntos
Modelos Animais de Doenças , Terapia de Reposição de Enzimas , Proteínas de Membrana/uso terapêutico , Movimento (Física) , Lipofuscinoses Ceroides Neuronais/tratamento farmacológico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/imunologia , Encéfalo/patologia , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Humanos , Injeções Espinhais , Proteínas de Membrana/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Tioléster Hidrolases/genética , Tripeptidil-Peptidase 1
18.
Nat Commun ; 6: 6606, 2015 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-25808087

RESUMO

Acute seizures after a severe brain insult can often lead to epilepsy and cognitive impairment. Aberrant hippocampal neurogenesis follows the insult but the role of adult-generated neurons in the development of chronic seizures or associated cognitive deficits remains to be determined. Here we show that the ablation of adult neurogenesis before pilocarpine-induced acute seizures in mice leads to a reduction in chronic seizure frequency. We also show that ablation of neurogenesis normalizes epilepsy-associated cognitive deficits. Remarkably, the effect of ablating adult neurogenesis before acute seizures is long lasting as it suppresses chronic seizure frequency for nearly 1 year. These findings establish a key role of neurogenesis in chronic seizure development and associated memory impairment and suggest that targeting aberrant hippocampal neurogenesis may reduce recurrent seizures and restore cognitive function following a pro-epileptic brain insult.


Assuntos
Transtornos Cognitivos/fisiopatologia , Epilepsia do Lobo Temporal/fisiopatologia , Hipocampo/crescimento & desenvolvimento , Neurogênese/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/etiologia , Modelos Animais de Doenças , Proteínas do Domínio Duplacortina , Epilepsia/induzido quimicamente , Epilepsia/complicações , Epilepsia/fisiopatologia , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/complicações , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Agonistas Muscarínicos/toxicidade , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais , Neurogênese/fisiologia , Neurônios/metabolismo , Neuropeptídeos/metabolismo , Pilocarpina/toxicidade
19.
Hippocampus ; 25(11): 1374-9, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25786918

RESUMO

CA1 hippocampal N-methyl-d-aspartate-receptors (NMDARs) are necessary for contextually related learning and memory processes. Extinction, a form of learning, has been shown to require intact hippocampal NMDAR signalling. Renewal of fear expression can occur after fear extinction training, when the extinguished fear stimulus is presented in an environmental context different from the training context and thus, renewal is dependent on contextual memory. In this study, we show that a Grin1 knock-out (loss of the essential NR1 subunit for the NMDAR) restricted to the bilateral CA1 subfield of the dorsal hippocampus does not affect acquisition of learned fear, but does attenuate extinction of a cued fear response even when presented in the extinction-training context. We propose that failure to remember the (safe) extinction context is responsible for the abnormal fear response and suggest it is a dysfunctional renewal. The results highlight the difference in outcome of extinguished fear memory resulting from a partial rather than complete loss of function of the hippocampus and suggest a potential mechanism for abnormally increased fear expression in PTSD.


Assuntos
Comportamento Animal/fisiologia , Região CA1 Hipocampal/fisiopatologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Animais , Sinais (Psicologia) , Modelos Animais de Doenças , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética , Transtornos de Estresse Pós-Traumáticos/fisiopatologia
20.
Hippocampus ; 25(4): 409-14, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25424867

RESUMO

Addiction has been proposed to emerge from associations between the drug and the reward-associated contexts. This associative learning has a cellular correlate, as there are more cFos+ neurons in the hippocampal dentate gyrus (DG) after psychostimulant conditioned place preference (CPP) versus saline controls. However, it is unknown whether morphine CPP leads to a similar DG activation, or whether DG activation is due to locomotion, handling, pharmacological effects, or-as data from contextual fear learning suggests-exposure to the drug-associated context. To explore this, we employed an unbiased, counterbalanced, and shortened CPP design that led to place preference and more DG cFos+ cells. Next, mice underwent morphine CPP but were then sequestered into the morphine-paired (conditioned stimulus+ [CS+]) or saline-paired (CS-) context on test day. Morphine-paired mice sequestered to CS+ had ∼30% more DG cFos+ cells than saline-paired mice. Furthermore, Bregma analysis revealed morphine-paired mice had more cFos+ cells in CS+ compared to CS- controls. Notably, there was no significant difference in DG cFos+ cell number after handling alone or after receiving morphine in home cage. Thus, retrieval of morphine-associated context is accompanied by activation of hippocampal DG granule cell neurons.


Assuntos
Giro Denteado/citologia , Rememoração Mental/efeitos dos fármacos , Morfina/administração & dosagem , Entorpecentes/administração & dosagem , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Análise de Variância , Animais , Condicionamento Operante/efeitos dos fármacos , Condicionamento Operante/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Camundongos , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...