Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurosci ; 44(5)2024 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-38123360

RESUMO

Mutations in the activity-dependent transcription factor MEF2C have been associated with several neuropsychiatric disorders. Among these, autism spectrum disorder (ASD)-related behavioral deficits are manifested. Multiple animal models that harbor mutations in Mef2c have provided compelling evidence that Mef2c is indeed an ASD gene. However, studies in mice with germline or global brain knock-out of Mef2c are limited in their ability to identify the precise neural substrates and cell types that are required for the expression of Mef2c-mediated ASD behaviors. Given the role of hippocampal neurogenesis in cognitive and social behaviors, in this study we aimed to investigate the role of Mef2c in the structure and function of newly generated dentate granule cells (DGCs) in the postnatal hippocampus and to determine whether disrupted Mef2c function is responsible for manifesting ASD behaviors. Overexpression of Mef2c (Mef2cOE ) arrested the transition of neurogenesis at progenitor stages, as indicated by sustained expression of Sox2+ in Mef2cOE DGCs. Conditional knock-out of Mef2c (Mef2ccko ) allowed neuronal commitment of Mef2ccko cells; however, Mef2ccko impaired not only dendritic arborization and spine formation but also synaptic transmission onto Mef2ccko DGCs. Moreover, the abnormal structure and function of Mef2ccko DGCs led to deficits in social interaction and social novelty recognition, which are key characteristics of ASD behaviors. Thus, our study revealed a dose-dependent requirement of Mef2c in the control of distinct steps of neurogenesis, as well as a critical cell-autonomous function of Mef2c in newborn DGCs in the expression of proper social behavior in both sexes.


Assuntos
Transtorno do Espectro Autista , Transtorno Autístico , Masculino , Feminino , Camundongos , Animais , Transtorno do Espectro Autista/genética , Hipocampo , Neurônios/fisiologia , Neurogênese/fisiologia , Fatores de Transcrição MEF2/genética
2.
J Exp Med ; 219(9)2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35984475

RESUMO

Hippocampal neurogenesis is impaired in Alzheimer's disease (AD) patients and familial Alzheimer's disease (FAD) mouse models. However, it is unknown whether new neurons play a causative role in memory deficits. Here, we show that immature neurons were actively recruited into the engram following a hippocampus-dependent task. However, their recruitment is severely deficient in FAD. Recruited immature neurons exhibited compromised spine density and altered transcript profile. Targeted augmentation of neurogenesis in FAD mice restored the number of new neurons in the engram, the dendritic spine density, and the transcription signature of both immature and mature neurons, ultimately leading to the rescue of memory. Chemogenetic inactivation of immature neurons following enhanced neurogenesis in AD, reversed mouse performance, and diminished memory. Notably, AD-linked App, ApoE, and Adam10 were of the top differentially expressed genes in the engram. Collectively, these observations suggest that defective neurogenesis contributes to memory failure in AD.


Assuntos
Doença de Alzheimer , Transtornos da Memória , Neurogênese , Animais , Camundongos , Doença de Alzheimer/complicações , Modelos Animais de Doenças , Hipocampo , Transtornos da Memória/genética , Camundongos Transgênicos , Neurogênese/genética , Neurônios
3.
Brain Plast ; 6(1): 27-39, 2020 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-33680844

RESUMO

Chronic alcohol consumption results in alcohol use disorder (AUD). Interestingly, however, sudden alcohol withdrawal (AW) after chronic alcohol exposure also leads to a devastating series of symptoms, referred to as alcohol withdrawal syndromes. One key feature of AW syndromes is to produce phenotypes that are opposite to AUD. For example, while the brain is characterized by a hypoactive state in the presence of alcohol, AW induces a hyperactive state, which is manifested as seizure expression. In this review, we discuss the idea that hippocampal neurogenesis and neural circuits play a key role in neuroadaptation and establishment of allostatic states in response to alcohol exposure and AW. The intrinsic properties of dentate granule cells (DGCs), and their contribution to the formation of a potent feedback inhibitory loop, endow the dentate gyrus with a "gate" function, which can limit the entry of excessive excitatory signals from the cortex into the hippocampus. We discuss the possibility that alcohol exposure and withdrawal disrupts structural development and circuitry integration of hippocampal newborn neurons, and that this altered neurogenesis impairs the gate function of the hippocampus. Failure of this gate function is expected to alter the ratio of excitatory to inhibitory (E/I) signals in the hippocampus and to induce seizure expression during AW. Recent functional studies have shown that specific activation and inhibition of hippocampal newborn DGCs are both necessary and sufficient for the expression of AW-associated seizures, further supporting the concept that neurogenesis-induced neuroadaptation is a critical target to understand and treat AUD and AW-associated seizures.

4.
J Neurosci ; 40(2): 447-458, 2020 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-31719166

RESUMO

Cognitive impairments are key features in multiple sclerosis (MS), a progressive disorder characterized by neuroinflammation-induced demyelination in the central nervous system. To understand the neural substrates that link demyelination to cognitive deficits in MS, we investigated hippocampal neurogenesis and synaptic connectivity of adult-born neurons, which play an essential role in cognitive function. The administration and withdrawal of the combination of cuprizone and rapamycin (Cup/Rap) in C57BL/6J male mice efficiently demyelinated and remyelinated the hippocampus, respectively. In the demyelinated hippocampus, neurogenesis was nearly absent in the dentate gyrus, which was due to inhibited proliferation of neural stem cells (NSCs). Specifically, radial glia-like type 1 NSCs were shifted from a proliferative state to a mitotically-quiescent state in the demyelinated hippocampus. In addition, dendritic spine densities of adult-born neurons were significantly decreased, indicating a reduction in synaptic connections between hippocampal newborn neurons and excitatory input neurons. Concomitant with hippocampal remyelination induced by withdrawal of Cup/Rap, proliferation of type 1 NSCs and dendritic spine densities of adult-born neurons reverted to normal in the hippocampus. Our study shows that proliferation of hippocampal NSCs and synaptic connectivity of adult-born neurons are inversely correlated with the level of demyelination, providing critical insight into hippocampal neurogenesis as a potential therapeutic target to treat cognitive deficits associated with MS.SIGNIFICANCE STATEMENT To identify the neural substrates that mediate cognitive dysfunctions associated with a majority of MS patients, we investigated hippocampal neurogenesis and structural development of adult-born neurons using a Cup/Rap model, which recapitulates the hippocampal demyelination that occurs in MS patients. A shift of NSCs from a proliferatively-active state to mitotically-quiescent state dramatically decreased neurogenesis in the demyelinated hippocampus. Formation of dendritic spines on newborn neurons was also impaired following demyelination. Interestingly, the altered neurogenesis and synaptic connectivity of newborn neurons were reversed to normal levels during remyelination. Thus, our study revealed reversible genesis and synaptic connectivity of adult-born neurons between the demyelinated and remyelinated hippocampus, suggesting hippocampal neurogenesis as a potential target to normalize cognitive impairments in MS patients.


Assuntos
Disfunção Cognitiva/patologia , Hipocampo/patologia , Esclerose Múltipla , Vias Neurais/patologia , Neurogênese/fisiologia , Animais , Proliferação de Células/fisiologia , Disfunção Cognitiva/etiologia , Cuprizona/toxicidade , Modelos Animais de Doenças , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Inibidores da Monoaminoxidase/toxicidade , Esclerose Múltipla/induzido quimicamente , Esclerose Múltipla/complicações , Esclerose Múltipla/patologia , Células-Tronco Neurais/patologia , Neurônios/patologia
5.
JCI Insight ; 4(19)2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31578307

RESUMO

Alcohol withdrawal (AW) after chronic alcohol exposure produces a series of symptoms, with AW-associated seizures being among the most serious and dangerous. However, the mechanism underlying AW seizures has yet to be established. In our mouse model, a sudden AW produced 2 waves of seizures: the first wave includes a surge of multiple seizures that occurs within hours to days of AW, and the second wave consists of sustained expression of epileptiform spikes and wave discharges (SWDs) during a protracted period of abstinence. We revealed that the structural and functional adaptations in newborn dentate granule cells (DGCs) in the hippocampus underlie the second wave of seizures but not the first wave. While the general morphology of newborn DGCs remained unchanged, AW increased the dendritic spine density of newborn DGCs, suggesting that AW induced synaptic connectivity of newborn DGCs with excitatory afferent neurons and enhanced excitability of newborn DGCs. Indeed, specific activation and suppression of newborn DGCs by the chemogenetic DREADD method increased and decreased the expression of epileptiform SWDs, respectively, during abstinence. Thus, our study unveiled that the pathological plasticity of hippocampal newborn DGCs underlies AW seizures during a protracted period of abstinence, providing critical insight into hippocampal neural circuits as a foundation to understand and treat AW seizures.


Assuntos
Convulsões por Abstinência de Álcool/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Abstinência de Álcool , Convulsões por Abstinência de Álcool/patologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Eletroencefalografia , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Coluna Vertebral/patologia , Tamoxifeno/farmacologia
6.
Mol Psychiatry ; 24(11): 1627-1640, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30967683

RESUMO

Pten mutations are associated with autism spectrum disorder. Pten loss of function in neurons increases excitatory synaptic connectivity, contributing to an imbalance between excitation and inhibition. We aimed to determine whether Pten loss results in aberrant connectivity in neural circuits. We compared postnatally generated wild-type and Pten knockout granule neurons integrating into the dentate gyrus using a variety of methods to examine their connectivity. We found that postsynaptic Pten loss provides an advantage to dendritic spines in competition over a limited pool of presynaptic boutons. Retrograde monosynaptic tracing with rabies virus reveals that this results in synaptic contact with more presynaptic partners. Using independently excitable opsins to interrogate multiple inputs onto a single neuron, we found that excess connectivity is established indiscriminately from among glutamatergic afferents. Therefore, Pten loss results in inappropriate connectivity whereby neurons are coupled to a greater number of synaptic partners.


Assuntos
Potenciais Pós-Sinápticos Excitadores/fisiologia , PTEN Fosfo-Hidrolase/metabolismo , Terminações Pré-Sinápticas/fisiologia , Animais , Espinhas Dendríticas/fisiologia , Feminino , Hipocampo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/fisiologia , Opsinas/metabolismo , PTEN Fosfo-Hidrolase/genética , Sinapses/fisiologia
7.
Transl Psychiatry ; 9(1): 24, 2019 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-30655503

RESUMO

SETD5, a gene linked to intellectual disability (ID) and autism spectrum disorder (ASD), is a member of the SET-domain family and encodes a putative histone methyltransferase (HMT). To date, the mechanism by which SETD5 haploinsufficiency causes ASD/ID remains an unanswered question. Setd5 is the highly conserved mouse homolog, and although the Setd5 null mouse is embryonic lethal, the heterozygote is viable. Morphological tracing and multielectrode array was used on cultured cortical neurons. MRI was conducted of adult mouse brains and immunohistochemistry of juvenile mouse brains. RNA-Seq was used to investigate gene expression in the developing cortex. Behavioral assays were conducted on adult mice. Setd5+/- cortical neurons displayed significantly reduced synaptic density and neuritic outgrowth in vitro, with corresponding decreases in network activity and synchrony by electrophysiology. A specific subpopulation of fetal Setd5+/- cortical neurons showed altered gene expression of neurodevelopment-related genes. Setd5+/- animals manifested several autism-like behaviors, including hyperactivity, cognitive deficit, and altered social interactions. Anatomical differences were observed in Setd5+/- adult brains, accompanied by a deficit of deep-layer cortical neurons in the developing brain. Our data converge on a picture of abnormal neurodevelopment driven by Setd5 haploinsufficiency, consistent with a highly penetrant risk factor.


Assuntos
Transtorno do Espectro Autista/genética , Comportamento Animal , Haploinsuficiência/genética , Metiltransferases/genética , Neurônios/metabolismo , Animais , Transtorno do Espectro Autista/psicologia , Encéfalo/patologia , Feminino , Predisposição Genética para Doença , Heterozigoto , Imageamento por Ressonância Magnética , Masculino , Camundongos , Camundongos Knockout , Mutação
8.
J Clin Invest ; 129(1): 310-323, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30507615

RESUMO

We investigated how pathological changes in newborn hippocampal dentate granule cells (DGCs) lead to epilepsy. Using a rabies virus-mediated retrograde tracing system and a designer receptors exclusively activated by designer drugs (DREADD) chemogenetic method, we demonstrated that newborn hippocampal DGCs are required for the formation of epileptic neural circuits and the induction of spontaneous recurrent seizures (SRS). A rabies virus-mediated mapping study revealed that aberrant circuit integration of hippocampal newborn DGCs formed excessive de novo excitatory connections as well as recurrent excitatory loops, allowing the hippocampus to produce, amplify, and propagate excessive recurrent excitatory signals. In epileptic mice, DREADD-mediated-specific suppression of hippocampal newborn DGCs dramatically reduced epileptic spikes and SRS in an inducible and reversible manner. Conversely, specific activation of hippocampal newborn DGCs increased both epileptic spikes and SRS. Our study reveals an essential role for hippocampal newborn DGCs in the formation and function of epileptic neural circuits, providing critical insights into DGCs as a potential therapeutic target for treating epilepsy.


Assuntos
Giro Denteado/fisiopatologia , Epilepsia/fisiopatologia , Rede Nervosa/fisiopatologia , Animais , Animais Recém-Nascidos , Giro Denteado/metabolismo , Giro Denteado/patologia , Drogas Desenhadas/farmacologia , Epilepsia/tratamento farmacológico , Epilepsia/metabolismo , Epilepsia/patologia , Masculino , Camundongos , Camundongos Transgênicos , Rede Nervosa/metabolismo , Rede Nervosa/patologia
9.
Front Mol Neurosci ; 11: 415, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30498432

RESUMO

Using a lentivirus-mediated labeling method, we investigated whether the adult hippocampus retains long-lasting, self-renewing neural stem cells (NSCs). We first showed that a single injection of a lentiviral vector expressing a green fluorescent protein (LV PGK-GFP) into the subgranular zone (SGZ) of the adult hippocampus enabled an efficient, robust, and long-term marking of self-renewing NSCs and their progeny. Interestingly, a subset of labeled cells showed the ability to proliferate multiple times and give rise to Sox2+ cells, clearly suggesting the ability of NSCs to self-renew for an extensive period of time (up to 6 months). In addition, using GFP+ cells isolated from the SGZ of mice that received a LV PGK-GFP injection 3 months earlier, we demonstrated that some GFP+ cells displayed the essential properties of NSCs, such as self-renewal and multipotency. Furthermore, we investigated the plasticity of NSCs in a perforant path transection, which has been shown to induce astrocyte formation in the molecular layer of the hippocampus. Our lentivirus (LV)-mediated labeling study revealed that hippocampal NSCs are not responsible for the burst of astrocyte formation, suggesting that signals released from the injured perforant path did not influence NSC fate determination. Therefore, our studies showed that a gene delivery system using LVs is a unique method to be used for understanding the complex nature of NSCs and may have translational impact in gene therapy by efficiently targeting NSCs.

10.
Sci Rep ; 7(1): 17702, 2017 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-29255203

RESUMO

We previously showed increased growth associated protein 43 (GAP-43) expression in brain samples resected from patients with cortical dysplasia (CD), which was correlated with duration of epilepsy. Here, we used a rat model of CD to examine the regulation of GAP-43 in the brain and serum over the course of epileptogenesis. Baseline GAP-43 expression was higher in CD animals compared to control non-CD rats. An acute seizure increased GAP-43 expression in both CD and control rats. However, GAP-43 expression decreased by day 15 post-seizure in control rats, which did not develop spontaneous seizures. In contrast, GAP-43 remained up-regulated in CD rats, and over 50% developed chronic epilepsy with increased GAP-43 levels in their serum. GAP-43 protein was primarily located in excitatory neurons, suggesting its functional significance in epileptogenesis. Inhibition of GAP-43 expression by shRNA significantly reduced seizure duration and severity in CD rats after acute seizures with subsequent reduction in interictal spiking. Serum GAP-43 levels were significantly higher in CD rats that developed spontaneous seizures. Together, these results suggest GAP-43 as a key factor promoting epileptogenesis, a possible therapeutic target for treatment of progressive epilepsy and a potential biomarker for epilepsy progression in CD.


Assuntos
Epilepsia/fisiopatologia , Epilepsia/terapia , Proteína GAP-43/metabolismo , Animais , Biomarcadores/sangue , Modelos Animais de Doenças , Eletroencefalografia/métodos , Epilepsia/prevenção & controle , Proteína GAP-43/fisiologia , Regulação da Expressão Gênica/genética , Masculino , Malformações do Desenvolvimento Cortical/genética , Malformações do Desenvolvimento Cortical/metabolismo , Ratos , Ratos Sprague-Dawley , Convulsões/fisiopatologia
11.
Stem Cell Reports ; 9(2): 543-556, 2017 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-28757168

RESUMO

The molecular mechanism of memory formation remains a mystery. Here, we show that TERT, the catalytic subunit of telomerase, gene knockout (Tert-/-) causes extremely poor ability in spatial memory formation. Knockdown of TERT in the dentate gyrus of adult hippocampus impairs spatial memory processes, while overexpression facilitates it. We find that TERT plays a critical role in neural development including dendritic development and neuritogenesis of hippocampal newborn neurons. A monosynaptic pseudotyped rabies virus retrograde tracing method shows that TERT is required for neural circuit integration of hippocampal newborn neurons. Interestingly, TERT regulated neural development and spatial memory formation in a reverse transcription activity-independent manner. Using X-ray irradiation, we find that hippocampal newborn neurons mediate the modulation of spatial memory processes by TERT. These observations reveal an important function of TERT through a non-canonical pathway and encourage the development of a TERT-based strategy to treat neurological disease-associated memory impairment.


Assuntos
Regulação da Expressão Gênica , Hipocampo/fisiologia , Neurogênese/genética , Memória Espacial , Telomerase/genética , Animais , Linhagem Celular , Dendritos/metabolismo , Imunofluorescência , Genes Reporter , Humanos , Masculino , Camundongos , Camundongos Knockout , Células Piramidais/metabolismo , Proteínas Recombinantes de Fusão , Telomerase/metabolismo
12.
Stem Cell Reports ; 9(1): 217-230, 2017 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-28669600

RESUMO

BACE1 is validated as Alzheimer's ß-secretase and a therapeutic target for Alzheimer's disease. In examining BACE1-null mice, we discovered that BACE1 deficiency develops abnormal clusters of immature neurons, forming doublecortin-positive neuroblasts, in the developing dentate gyrus, mainly in the subpial zone (SPZ). Such clusters were rarely observed in wild-type SPZ and not reported in other mouse models. To understand their origins and fates, we examined how neuroblasts in BACE1-null SPZ mature and migrate during early postnatal development. We show that such neuroblasts are destined to form Prox1-positive granule cells in the dentate granule cell layer, and mainly mature to form excitatory neurons, but not inhibitory neurons. Mechanistically, higher levels of reelin potentially contribute to abnormal neurogenesis and timely migration in BACE1-null SPZ. Altogether, we demonstrate that BACE1 is a critical regulator in forming the dentate granule cell layer through timely maturation and migration of SPZ neuroblasts.


Assuntos
Secretases da Proteína Precursora do Amiloide/genética , Ácido Aspártico Endopeptidases/genética , Giro Denteado/patologia , Deleção de Genes , Neurônios/patologia , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Ácido Aspártico Endopeptidases/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular , Giro Denteado/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Neurogênese , Neurônios/metabolismo , Proteína Reelina , Serina Endopeptidases/metabolismo
13.
Sci Rep ; 6: 35572, 2016 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-27759049

RESUMO

Hippocampus-dependent cognitive and emotional function appears to be regionally dissociated along the dorsoventral (DV) axis of the hippocampus. Recent observations that adult hippocampal neurogenesis plays a critical role in both cognition and emotion raised an interesting question whether adult neurogenesis within specific subregions of the hippocampus contributes to these distinct functions. We examined the regional-specific and cell type-specific effects of fluoxetine, which requires adult hippocampal neurogenesis to function as an antidepressant, on the proliferation of hippocampal neural stem cells (NSCs). Fluoxetine specifically increased proliferation of NSCs located in the ventral region of the hippocampus while the mitotic index of NSCs in the dorsal portion of the hippocampus remained unaltered. Moreover, within the ventral hippocampus, type II NSC and neuroblast populations specifically responded to fluoxetine, showing increased proliferation; however, proliferation of type I NSCs was unchanged in response to fluoxetine. Activation or inhibition of serotonin receptor 1A (5-HTR1A) recapitulated or abolished the effect of fluoxetine on proliferation of type II NSCs and neuroblast populations in the ventral hippocampus. Our study showed that the effect of fluoxetine on proliferation is dependent upon the type and the position of the NSCs along the DV axis of the hippocampus.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Fluoxetina/uso terapêutico , Hipocampo/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Animais , Proliferação de Células , Células Cultivadas , Cognição , Modelos Animais de Doenças , Feminino , Hipocampo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/patologia , Neurogênese , Receptor 5-HT1A de Serotonina/metabolismo
14.
Cold Spring Harb Protoc ; 2015(10): 925-31, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26430255

RESUMO

Retroviral vectors are powerful tools for genetic manipulation. This protocol discusses the production, purification, and use of replication-deficient retroviral vectors based on Moloney murine leukemia virus and lentivirus. It also describes the injection of a retroviral vector into the dentate gyrus of young adult mice to fluorescently label live murine brain tissue.


Assuntos
Vetores Genéticos/isolamento & purificação , Lentivirus/crescimento & desenvolvimento , Lentivirus/isolamento & purificação , Vírus da Leucemia Murina de Moloney/crescimento & desenvolvimento , Vírus da Leucemia Murina de Moloney/isolamento & purificação , Animais , Lentivirus/genética , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Replicação Viral
15.
Cold Spring Harb Protoc ; 2015(10): 932-3, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26430256

RESUMO

This protocol describes the harvesting of brain tissue from mice that have had the retroviral vector CAG-GFP injected into the dentate gyrus. Brain tissue from these mice is dissected, the tissue is fixed, and the sections are prepared. The fixed sections are imaged using fluorescent confocal microscopy, and newborn granule cells containing GFP are visualized and are characterized.


Assuntos
Encéfalo/citologia , Microscopia Confocal/métodos , Neurônios/citologia , Animais , Animais Recém-Nascidos , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Camundongos , Microtomia , Coloração e Rotulagem/métodos , Fixação de Tecidos
16.
Cold Spring Harb Protoc ; 2015(10): 934-6, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26430257

RESUMO

In this protocol, acute brain slices are prepared from mice in which newborn granule cells have been labeled using retroviral vector technology. Using a live-cell imaging stage and confocal microscopy coupled to imaging software, dendritic spines are analyzed.


Assuntos
Encéfalo/citologia , Espinhas Dendríticas/fisiologia , Plasticidade Neuronal , Neurônios/citologia , Coluna Vertebral/citologia , Animais , Humanos , Processamento de Imagem Assistida por Computador , Recém-Nascido , Camundongos , Microscopia Confocal/métodos , Coloração e Rotulagem/métodos
17.
Cold Spring Harb Protoc ; 2015(10): 883-8, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26430260

RESUMO

Retroviral vectors are a powerful technology for achieving long-term genetic manipulation. This introduction provides some background on replication-deficient retroviral vectors based on Moloney murine leukemia virus and lentivirus. Details, examples, and associated protocols are provided for using these vectors to fluorescently label, genetically alter, and image both live and fixed murine brain tissue.


Assuntos
Vetores Genéticos/isolamento & purificação , Lentivirus/crescimento & desenvolvimento , Lentivirus/isolamento & purificação , Vírus da Leucemia Murina de Moloney/crescimento & desenvolvimento , Vírus da Leucemia Murina de Moloney/isolamento & purificação , Animais , Encéfalo/citologia , Lentivirus/genética , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Coloração e Rotulagem/métodos , Transformação Genética , Replicação Viral
18.
Alcohol Clin Exp Res ; 39(10): 1967-77, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26365148

RESUMO

BACKGROUND: Neurological deficits of alcohol use disorder (AUD) have been attributed to dysfunctions of specific brain structures. Studies of alcoholic patients and chronic alcohol exposure animal models consistently identify reduced hippocampal mass and cogntive dysfunctions as a key alcohol-induced brain adaptation. However, the precise substrate of chronic alcohol exposure that leads to structural and functional impairments of the hippocampus is largely unknown. METHODS: Using a calorie-matched alcohol feeding method, we tested whether chronic alcohol exposure targets neural stem cells and neurogenesis in the adult hippocampus. The effect of alcohol on proliferation of neural stem cells as well as cell fate determination and survival of newborn cells was evaluated via bromodeoxyuridine pulse and chase methods. A retrovirus-mediated single-cell labeling method was used to determine the effect of alcohol on the morphological development and circuitry incorporation of individual hippocampal newborn neurons. Finally, novel object recognition (NOR) and Y-maze tests were performed to examine whether disrupted neurogenesis is associated with hippocampus-dependent functional deficits in alcohol-fed mice. RESULTS: Chronic alcohol exposure reduced proliferation of neural stem cells and survival rate of newborn neurons; however, the fate determination of newborn cells remained unaltered. Moreover, the dendritic spine density of newborn neurons significantly decreased in alcohol-fed mice. Impaired spine formation indicates that alcohol interfered the synaptic connectivity of newborn neurons with excitatory neurons originating from various areas of the brain. In the NOR test, alcohol-fed mice displayed deficits in the ability to discriminate the novel object. CONCLUSIONS: Our study revealed that chronic alcohol exposure disrupted multiple steps of neurogenesis, including the production and development of newborn neurons. In addition, chronic alcohol exposure altered connectivity of newborn neurons with other input neurons. Decreased neurogenesis and aberrant integration of newborn neurons into hippocampal networks are closely associated with deficits in hippocampus-dependent cognitive functions of alcohol-fed mice.


Assuntos
Encéfalo/efeitos dos fármacos , Transtornos Cognitivos/induzido quimicamente , Etanol/administração & dosagem , Etanol/farmacologia , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Consumo de Bebidas Alcoólicas/patologia , Consumo de Bebidas Alcoólicas/fisiopatologia , Consumo de Bebidas Alcoólicas/psicologia , Animais , Encéfalo/patologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Células-Tronco Neurais/efeitos dos fármacos , Reconhecimento Psicológico/efeitos dos fármacos , Teste de Desempenho do Rota-Rod
19.
PLoS One ; 9(5): e97689, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24831808

RESUMO

Hypothalamus-pituitary-adrenal (HPA) hyperactivity is observed in many patients suffering from depression and the mechanism underling the dysfunction of HPA axis is not well understood. Chronic stress has a causal relationship with the hyperactivity of HPA axis. Stress induces the over-synthesis of glucocorticoids, which will arrive at all the body containing the brain. It is still complicated whether glucocorticoids account for chronic stress-induced HPA axis hyperactivity and in which part of the brain the glucocorticoids account for chronic stress-induced HPA axis hyperactivity. Here, we demonstrated that glucocorticoids were indispensable and sufficient for chronic stress-induced hyperactivity of HPA axis. Although acute glucocorticoids elevation in the hippocampus and hypothalamus exerted a negative regulation of HPA axis, we found that chronic glucocorticoids elevation in the hippocampus but not in the hypothalamus accounted for chronic stress-induced hyperactivity of HPA axis. Chronic glucocorticoids exposure in the hypothalamus still exerted a negative regulation of HPA axis activity. More importantly, we found mineralocorticoid receptor (MR) - neuronal nitric oxide synthesis enzyme (nNOS) - nitric oxide (NO) pathway mediated the different roles of glucocorticoids in the hippocampus and hypothalamus in regulating HPA axis activity. This study suggests that the glucocorticoids in the hippocampus play an important role in the development of HPA axis hyperactivity and the glucocorticoids in the hypothalamus can't induce hyperactivity of HPA axis, revealing new insights into understanding the mechanism of depression.


Assuntos
Glucocorticoides/fisiologia , Hipocampo/metabolismo , Sistema Hipotálamo-Hipofisário , Hipotálamo/metabolismo , Sistema Hipófise-Suprarrenal , Estresse Psicológico/metabolismo , Animais , Doença Crônica , Depressão/metabolismo , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Óxido Nítrico Sintase Tipo I/metabolismo
20.
Proc Natl Acad Sci U S A ; 111(13): E1240-8, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24639535

RESUMO

Breast cancer susceptibility gene 1 (BRCA1) is a breast and ovarian cancer tumor suppressor whose loss leads to DNA damage and defective centrosome functions. Despite its tumor suppression functions, BRCA1 is most highly expressed in the embryonic neuroepithelium when the neural progenitors are highly proliferative. To determine its functional significance, we deleted BRCA1 in the developing brain using a neural progenitor-specific driver. The phenotype is characterized by severe agenesis of multiple laminated cerebral structures affecting most notably the neocortex, hippocampus, cerebellum, and olfactory bulbs. Major phenotypes are caused by excess apoptosis, as these could be significantly suppressed by the concomitant deletion of p53. Certain phenotypes attributable to centrosomal and cell polarity functions could not be rescued by p53 deletion. A double KO with the DNA damage sensor kinase ATM was able to rescue BRCA1 loss to a greater extent than p53. Our results suggest distinct apoptotic and centrosomal functions of BRCA1 in neural progenitors, with important implications to understand the sensitivity of the embryonic brain to DNA damage, as well as the developmental regulation of brain size.


Assuntos
Proteína BRCA1/metabolismo , Encéfalo/embriologia , Encéfalo/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Encéfalo/citologia , Polaridade Celular , Proliferação de Células , Sobrevivência Celular , Cognição/fisiologia , Deleção de Genes , Aprendizagem , Camundongos , Camundongos Knockout , Atividade Motora/fisiologia , Nestina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fenótipo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...