Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
J Am Heart Assoc ; 11(10): e025181, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35535614

RESUMO

Background Lung injury, a severe adverse outcome of lipopolysaccharide-induced acute respiratory distress syndrome, is attributed to excessive neutrophil recruitment and effector response. Poldip2 (polymerase δ-interacting protein 2) plays a critical role in regulating endothelial permeability and leukocyte recruitment in acute inflammation. Thus, we hypothesized that myeloid Poldip2 is involved in neutrophil recruitment to inflamed lungs. Methods and Results After characterizing myeloid-specific Poldip2 knockout mice, we showed that at 18 hours post-lipopolysaccharide injection, bronchoalveolar lavage from myeloid Poldip2-deficient mice contained fewer inflammatory cells (8 [4-16] versus 29 [12-57]×104/mL in wild-type mice) and a smaller percentage of neutrophils (30% [28%-34%] versus 38% [33%-41%] in wild-type mice), while the main chemoattractants for neutrophils remained unaffected. In vitro, Poldip2-deficient neutrophils responded as well as wild-type neutrophils to inflammatory stimuli with respect to neutrophil extracellular trap formation, reactive oxygen species production, and induction of cytokines. However, neutrophil adherence to a tumor necrosis factor-α stimulated endothelial monolayer was inhibited by Poldip2 depletion (225 [115-272] wild-type [myePoldip2+/+] versus 133 [62-178] myeloid-specific Poldip2 knockout [myePoldip2-/-] neutrophils) as was transmigration (1.7 [1.3-2.1] versus 1.1 [1.0-1.4] relative to baseline transmigration). To determine the underlying mechanism, we examined the surface expression of ß2-integrin, its binding to soluble intercellular adhesion molecule 1, and Pyk2 phosphorylation. Surface expression of ß2-integrins was not affected by Poldip2 deletion, whereas ß2-integrins and Pyk2 were less activated in Poldip2-deficient neutrophils. Conclusions These results suggest that myeloid Poldip2 is involved in ß2-integrin activation during the inflammatory response, which in turn mediates neutrophil-to-endothelium adhesion in lipopolysaccharide-induced acute respiratory distress syndrome.


Assuntos
Proteínas Mitocondriais , Neutrófilos , Proteínas Nucleares , Pneumonia , Síndrome do Desconforto Respiratório , Animais , Adesão Celular , Modelos Animais de Doenças , Quinase 2 de Adesão Focal/metabolismo , Integrinas/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Neutrófilos/metabolismo , Neutrófilos/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Pneumonia/genética , Pneumonia/metabolismo , Pneumonia/patologia , Síndrome do Desconforto Respiratório/genética , Síndrome do Desconforto Respiratório/metabolismo , Síndrome do Desconforto Respiratório/patologia
2.
Mol Psychiatry ; 26(3): 772-783, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-30976085

RESUMO

The 3q29 deletion confers increased risk for neuropsychiatric phenotypes including intellectual disability, autism spectrum disorder, generalized anxiety disorder, and a >40-fold increased risk for schizophrenia. To investigate consequences of the 3q29 deletion in an experimental system, we used CRISPR/Cas9 technology to introduce a heterozygous deletion into the syntenic interval on C57BL/6 mouse chromosome 16. mRNA abundance for 20 of the 21 genes in the interval was reduced by ~50%, while protein levels were reduced for only a subset of these, suggesting a compensatory mechanism. Mice harboring the deletion manifested behavioral impairments in multiple domains including social interaction, cognitive function, acoustic startle, and amphetamine sensitivity, with some sex-dependent manifestations. In addition, 3q29 deletion mice showed reduced body weight throughout development consistent with the phenotype of 3q29 deletion syndrome patients. Of the genes within the interval, DLG1 has been hypothesized as a contributor to the neuropsychiatric phenotypes. However, we show that Dlg1+/- mice did not exhibit the behavioral deficits seen in mice harboring the full 3q29 deletion. These data demonstrate the following: the 3q29 deletion mice are a valuable experimental system that can be used to interrogate the biology of 3q29 deletion syndrome; behavioral manifestations of the 3q29 deletion may have sex-dependent effects; and mouse-specific behavior phenotypes associated with the 3q29 deletion are not solely due to haploinsufficiency of Dlg1.


Assuntos
Transtorno do Espectro Autista , Deficiência Intelectual , Esquizofrenia , Animais , Criança , Deleção Cromossômica , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Deficiências do Desenvolvimento/genética , Modelos Animais de Doenças , Humanos , Deficiência Intelectual/genética , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Esquizofrenia/genética
3.
Transl Psychiatry ; 8(1): 164, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30135420

RESUMO

Behavioral and molecular characterization of cell-type-specific populations governing fear learning and behavior is a promising avenue for the rational identification of potential therapeutics for fear-related disorders. Examining cell-type-specific changes in neuronal translation following fear learning allows for targeted pharmacological intervention during fear extinction learning, mirroring possible treatment strategies in humans. Here we identify the central amygdala (CeA) Drd2-expressing population as a novel fear-supporting neuronal population that is molecularly distinct from other, previously identified, fear-supporting CeA populations. Sequencing of actively translating transcripts of Drd2 neurons using translating ribosome affinity purification (TRAP) technology identifies mRNAs that are differentially regulated following fear learning. Differentially expressed transcripts with potentially targetable gene products include Npy5r, Rxrg, Adora2a, Sst5r, Fgf3, Erbb4, Fkbp14, Dlk1, and Ssh3. Direct pharmacological manipulation of NPY5R, RXR, and ADORA2A confirms the importance of this cell population and these cell-type-specific receptors in fear behavior. Furthermore, these findings validate the use of functionally identified specific cell populations to predict novel pharmacological targets for the modulation of emotional learning.


Assuntos
Medo/fisiologia , Aprendizagem/fisiologia , Neurônios/fisiologia , Receptores de Dopamina D2/genética , Animais , Comportamento Animal , Núcleo Central da Amígdala/citologia , Condicionamento Psicológico/fisiologia , Extinção Psicológica/fisiologia , Masculino , Camundongos Endogâmicos C57BL , Análise de Sequência de RNA
4.
J Neurosci Res ; 95(5): 1144-1160, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-27859486

RESUMO

Recent studies show that the complex genetic architecture of schizophrenia (SZ) is driven in part by polygenic components, or the cumulative effect of variants of small effect in many genes, as well as rare single-locus variants with large effect sizes. Here we discuss genetic aberrations known as copy number variants (CNVs), which fall in the latter category and are associated with a high risk for SZ and other neuropsychiatric disorders. We briefly review recurrent CNVs associated with SZ, and then highlight one CNV in particular, a recurrent 1.6-Mb deletion on chromosome 3q29, which is estimated to confer a 40-fold increased risk for SZ. Additionally, we describe the use of genetic mouse models, behavioral tools, and patient-derived induced pluripotent stem cells as a means to study CNVs in the hope of gaining mechanistic insight into their respective disorders. Taken together, the genomic data connecting CNVs with a multitude of human neuropsychiatric disease, our current technical ability to model such chromosomal anomalies in mouse, and the existence of precise behavioral measures of endophenotypes argue that the time is ripe for systematic dissection of the genetic mechanisms underlying such disease. © 2016 Wiley Periodicals, Inc.


Assuntos
Variações do Número de Cópias de DNA/genética , Predisposição Genética para Doença , Deficiência Intelectual/genética , Esquizofrenia/genética , Animais , Deleção Cromossômica , Cromossomos Humanos Par 3/genética , Deficiências do Desenvolvimento/genética , Humanos
5.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-666614

RESUMO

Behavioral and molecular characterization of cell- type specific populations governing fear learning and behavior is a promising avenue for the rational identification of potential therapeutics for fear-related disorders. Identification of cell-type specific changes in neuronal translation following fear learning allows for targeted pharmacological intervention during fear extinction learning, mirroring possible treatment strategies in humans. Here we identify the central amygdala (CeA) Drd2-expressing population as a fear-supporting population that is molecularly distinct from other, previously identified fear-supporting CeA populations. Sequencing of actively translating transcripts of Drd2 neurons identifies mRNAs that are differentially regulated following fear learning including Npy5r, Rxrg, Sst5r, Fgf3, ErbB4, Fkbp14, Dlk1,Ssh3 and Adora2a. Direct pharmacological manipulation of NPY5R, RXR, and ADORA2A confirms their importance in fear behavior and validates the present approach of identifying pharmacological targets for the modulation of emotional learning.

6.
Horm Behav ; 76: 136-42, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25888455

RESUMO

This article is part of a Special Issue "SBN 2014". Beginning with Vale and Colleagues in 1981, corticotropin releasing factor (CRF) also called corticotropin releasing hormone (CRH) has repeatedly been identified as an important contributor to fear and anxiety behavior. These findings have proven useful to further our understanding of disorders that have significant fear-dysregulation, such as post-traumatic stress, as well as other stress- and anxiety-related disorders. Unfortunately, the data are not all in agreement. In particular the role of CRF in fear learning is controversial, with studies pointing to contradictory effects from CRF manipulation even within the same brain structure. Further, very few studies address the potentially promising role of CRF manipulation in fear extinction behavior. Here, we briefly review the role of CRF in anxiety, fear learning and extinction, focusing on recent cell-type and neurotransmitter-specific studies in the amygdala and bed nucleus of the stria terminalis (BNST) that may help to synthesize the available data on the role of CRF in fear and anxiety-related behaviors.


Assuntos
Ansiedade/metabolismo , Núcleo Central da Amígdala/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Medo/fisiologia , Neurônios/metabolismo , Receptores de GABA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Núcleos Septais/metabolismo , Animais , Humanos
7.
PLoS One ; 9(10): e111009, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25340785

RESUMO

Corticotropin releasing factor (CRF) dysregulation is implicated in mood and anxiety disorders such as posttraumatic stress disorder (PTSD). CRF is expressed in areas engaged in fear and anxiety processing including the central amygdala (CeA). Complicating our ability to study the contribution of CRF-containing neurons to fear and anxiety behavior is the wide variety of cell types in which CRF is expressed. To manipulate specific subpopulations of CRF containing neurons, our lab has developed a mouse with a Cre recombinase gene driven by a CRF promoter (CRFp3.0Cre) (Martin et al., 2010). In these studies, mice that have the gene that encodes NR1 (Grin1) flanked by loxP sites (floxed) were crossed with our previously developed CRFp3.0Cre mouse to selectively disrupt Grin1 within CRF containing neurons (Cre+/fGrin1+). We find that disruption of Grin1 in CRF neurons did not affect baseline levels of anxiety, locomotion, pain sensitivity or exploration of a novel object. However, baseline expression of Grin1 was decreased in Cre+/fGrin1+ mice as measured by RTPCR. Cre+/fGrin1+ mice showed enhanced auditory fear acquisition and retention without showing any significant effect on fear extinction. We measured Gria1, the gene that encodes AMPAR1 and the CREB activator Creb1 in the amygdala of Cre+/fGrin1+ mice after fear conditioning. Both Gria1 and Creb1 were enhanced in the amygdala after training. To determine if the Grin1-expressing CRF neurons within the CeA are responsible for the enhancement of fear memory in adults, we infused a lentivirus with Cre driven by a CRF promoter (LV pCRF-Cre/fGrin1+) into the CeA of floxed Grin1 mice. Cre driven deletion of Grin1 specifically within CRF expressing cells in the CeA also resulted in enhanced fear memory acquisition and retention. Altogether, these findings suggest that selective disruption of Grin1 within CeA CRF neurons strongly enhances fear memory.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Medo , Memória/fisiologia , Proteínas do Tecido Nervoso/genética , Receptores de N-Metil-D-Aspartato/genética , Tonsila do Cerebelo/metabolismo , Animais , Ansiedade , Comportamento Animal , Cruzamentos Genéticos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Deleção de Genes , Hibridização in Situ Fluorescente , Integrases/metabolismo , Lentivirus/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/fisiologia , Neurônios/metabolismo , Regiões Promotoras Genéticas , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/fisiologia
8.
Hippocampus ; 23(9): 842-7, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23640842

RESUMO

Prior work suggests that hippocampus-dependent memory undergoes a systems consolidation process such that recent memories are stored in the hippocampus, while older memories are independent of the hippocampus and instead dependent on cortical areas. One problem with interpreting these studies is that memory for the contextual stimuli weakens as time passes between the training event and testing and older memories are often less detailed, making it difficult to determine if memory storage in the hippocampus is related to the age or to the accuracy of the memory. Activity of the mammalian target of rapamycin (mTOR) signaling pathway is known to be important for controlling protein translation necessary for both memory consolidation after initial learning and for the reconsolidation of memory after retrieval. We tested whether p70s6 kinase (p70s6K), a key component of the mTOR signaling pathway, is activated following retrieval of context fear memory in the dorsal hippocampus (DH) and anterior cingulate cortex (ACC) at 1, 10, or 36 days after context fear conditioning. We also tested whether strengthening memory for the contextual stimuli changed p70s6K phosphorylation in these structures 36 days after training. We show that under standard training conditions retrieval of a recently formed memory is initially precise and involves the DH. Over time it loses detail, becomes independent of the DH and depends on the ACC. In a subsequent experiment, we preserved the accuracy of older memories through pre-exposure to the training context. We show that remote memory still involved the DH in animals given pre-exposure. These data support the notion that detailed memories depend on the DH regardless of their age.


Assuntos
Condicionamento Psicológico/fisiologia , Medo , Hipocampo/metabolismo , Rememoração Mental/fisiologia , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo , Análise de Variância , Animais , Reação de Congelamento Cataléptica/fisiologia , Masculino , Ratos , Ratos Long-Evans , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Fatores de Tempo
9.
Proc Natl Acad Sci U S A ; 109(40): 16330-5, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22992651

RESUMO

Corticotropin-releasing factor (CRF) is critical for the endocrine, autonomic, and behavioral responses to stressors, and it has been shown to modulate fear and anxiety. The CRF receptor is widely expressed across a variety of cell types, impeding progress toward understanding the contribution of specific CRF-containing neurons to fear dysregulation. We used a unique CRF-Cre driver transgenic mouse line to remove floxed GABA(A)α1 subunits specifically from CRF neurons [CRF-GABA(A)α1 KO]. This process resulted in mice with decreased GABA(A)α1 expression only in CRF neurons and increased CRF mRNA within the amygdala, bed nucleus of the stria terminalis (BNST) and paraventricular nucleus of the hypothalamus. These mice show normal locomotor and pain responses and no difference in depressive-like behavior or Pavlovian fear conditioning. However, CRF-GABA(A)α1 KO increased anxiety-like behavior and impaired extinction of conditioned fear, coincident with an increase in plasma corticosterone concentration. These behavioral impairments were rescued with systemic or BNST infusion of the CRF antagonist R121919. Infusion of Zolpidem, a GABA(A)α1-preferring benzodiazepine-site agonist, into the BNST of the CRF-GABA(A)α1 KO was ineffective at decreasing anxiety. Electrophysiological findings suggest a disruption in inhibitory current may play a role in these changes. These data indicate that disturbance of CRF containing GABA(A)α1 neurons causes increased anxiety and impaired fear extinction, both of which are symptoms diagnostic for anxiety disorders, such as posttraumatic stress disorder.


Assuntos
Ansiedade/fisiopatologia , Hormônio Liberador da Corticotropina/metabolismo , Extinção Psicológica/fisiologia , Medo/fisiologia , Neurônios/metabolismo , Receptores de GABA-A/deficiência , Análise de Variância , Animais , Ansiedade/metabolismo , Condicionamento Psicológico/fisiologia , Corticosterona/sangue , Primers do DNA/genética , Hibridização In Situ , Hibridização in Situ Fluorescente , Aprendizagem em Labirinto , Camundongos , Camundongos Transgênicos , Neurônios/fisiologia , Técnicas de Patch-Clamp , Piridinas , Pirimidinas , Receptores de GABA-A/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Zolpidem
10.
Learn Mem ; 19(7): 300-6, 2012 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-22723052

RESUMO

Numerous studies have indicated that maintaining a fear memory after retrieval requires de novo protein synthesis. However, no study to date has examined how the temporal dynamics of repeated retrieval events affect this protein synthesis requirement. The present study varied the timing of a second retrieval of an established auditory fear memory and followed this second retrieval with infusions of the protein synthesis inhibitor anisomycin (ANI) into the basolateral amygdala. Results indicated that the memory-impairing effects of ANI were not observed when the second retrieval occurred soon after the first (within 1 h), and that the inhibitor gradually regained effectiveness as the retrieval episodes were spaced further apart. Additionally, if the second of the closely timed retrievals was omitted prior to ANI infusions, long-term memory deficits were observed, suggesting that the altered effectiveness of ANI was due specifically to the second retrieval event. Further experiments revealed that the second retrieval was not associated with a change in Zif268 protein expression but did produce a rapid and persistent dephosphorylation of GluR1 receptors at Ser845, an AMPAR trafficking site known to regulate the stability of GluR2 lacking AMPARs, which have been shown to be important in memory updating. This suggests that the precise timing of multiple CS presentations during the reconsolidation window may affect the destabilization state of the memory trace.


Assuntos
Condicionamento Clássico/fisiologia , Medo/fisiologia , Rememoração Mental/fisiologia , Biossíntese de Proteínas/fisiologia , Receptores de AMPA/metabolismo , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Anisomicina/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Rememoração Mental/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Ratos , Ratos Long-Evans , Serina/metabolismo , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Fatores de Tempo
11.
Artigo em Inglês | MEDLINE | ID: mdl-20740074

RESUMO

Recent work has led to a better understanding of the neural mechanisms underlying the extinction of Pavlovian fear conditioning. Long-term synaptic changes in the medial prefrontal cortex (mPFC) are critical for extinction learning, but very little is currently known about how the mPFC and other brain areas interact during extinction. The current study examined the effect of drugs that impair the extinction of fear conditioning on the activation of the extracellular-related kinase/mitogen-activated protein kinase (ERK/MAPK) in brain regions that likely participate in the consolidation of extinction learning. Inhibitors of opioid and N-methyl-d-aspartic acid (NMDA) receptors were applied to the ventrolateral periaqueductal gray matter (vlPAG) and amygdala shortly before extinction training. Results from these experiments show that blocking opioid receptors in the vlPAG prevented the formation of extinction memory, whereas NMDA receptor blockade had no effect. Conversely, blocking NMDA receptors in the amygdala disrupted the formation of fear extinction memory, but opioid receptor blockade in the same brain area did not. Subsequent experiments tested the effect of these drug treatments on the activation of the ERK/MAPK signaling pathway in various brain regions following extinction training. Only opioid receptor blockade in the vlPAG disrupted ERK phosphorylation in the mPFC and amygdala. These data support the idea that opiodergic signaling derived from the vlPAG affects plasticity across the brain circuit responsible for the formation of extinction memory.

12.
Neural Plast ; 2010: 139891, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20592749

RESUMO

Memory consolidation requires transcription and translation of new protein. Arc, an effector immediate early gene, and zif268, a regulatory transcription factor, have been implicated in synaptic plasticity underlying learning and memory. This study explored the temporal expression profiles of these proteins in the rat hippocampus following fear conditioning. We observed a time-dependent increase of Arc protein in the dorsal hippocampus 30-to-90-minute post training, returning to basal levels at 4 h. Zif268 protein levels, however, gradually increased at 30-minute post training before peaking in expression at 60 minute. The timing of hippocampal Arc and zif268 expression coincides with the critical period for protein synthesis-dependent memory consolidation following fear conditioning. However, the expression of Arc protein appears to be driven by context exploration, whereas, zif268 expression may be more specifically related to associative learning. These findings suggest that altered Arc and zif268 expression are related to neural plasticity during the formation of fear memory.


Assuntos
Condicionamento Psicológico/fisiologia , Proteínas do Citoesqueleto/biossíntese , Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Medo/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , Proteínas do Tecido Nervoso/biossíntese , Animais , Western Blotting , Proteínas do Citoesqueleto/genética , Proteína 1 de Resposta de Crescimento Precoce/genética , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/fisiologia , Masculino , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal/genética , Ratos , Ratos Long-Evans , Fatores de Tempo , Regulação para Cima/genética
13.
Neurobiol Learn Mem ; 89(3): 324-37, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17977027

RESUMO

Recent work from a number of laboratories has provided new and important insights about how gene expression is altered by experience and how these molecular changes may provide a substrate for the long-term storage of new memories. Here, we review a series of recent studies using aversive Pavlovian conditioning in rats as a well characterized model system in which experience-dependent alterations in gene expression can be manipulated and quantified within a specific neural circuit. We highlight some of the issues involved in using broad-spectrum inhibitors of mRNA and protein synthesis to study cellular changes underlying the formation and long-term stability of memory and discuss the idea that these changes occur over widespread, behaviorally-defined, networks of cells. We also discuss the idea that the maintenance of memory and its susceptibly to disruption after retrieval may relate to local protein synthesis in dendrites. Finally, a series of recent experiments from our laboratory studying the role of a specific signaling pathway (mTOR) which regulates translational processes and memory formation in the amygdala and hippocampus during fear conditioning are reviewed.


Assuntos
Condicionamento Psicológico/fisiologia , Medo , Complexos Multiproteicos/metabolismo , Plasticidade Neuronal/fisiologia , Biossíntese de Proteínas/fisiologia , Sinapses/fisiologia , Tonsila do Cerebelo/fisiologia , Expressão Gênica/genética , Hipocampo/fisiologia , Humanos , Rede Nervosa/fisiologia , Proteínas Quinases/genética , RNA Mensageiro/genética , Serina-Treonina Quinases TOR , Ativação Transcricional/genética
14.
J Neurosci ; 26(50): 12977-83, 2006 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-17167087

RESUMO

The mammalian target of rapamycin kinase (mTOR) regulates protein synthesis in neurons at the translational level through phosphorylation of several intracellular targets. Recent work in invertebrates indicates that mTOR-dependent translational control may be critical for the induction and maintenance of activity-dependent synaptic plasticity underlying memory formation. Here, we report that training rats in a simple fear conditioning procedure evokes a time-dependent increase in the phosphorylation of p70s6 kinase, a major direct downstream target of mTOR. When the activation of mTOR was prevented by posttraining injection of rapamycin into the amygdala, formation of the memory and the increase in p70s6 kinase phosphorylation was attenuated. Furthermore, when rapamycin was applied to the amygdala after the recall of a previously stored fear memory, subsequent retention was disrupted, indicating that local translational control at active synapses is required for the stability as well as the formation of long-term memory in this system.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Memória/fisiologia , Biossíntese de Proteínas , Proteínas Quinases/fisiologia , Sirolimo/administração & dosagem , Estimulação Acústica/métodos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Condicionamento Psicológico/efeitos dos fármacos , Medo/efeitos dos fármacos , Injeções Intraventriculares , Masculino , Memória/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Ratos , Ratos Long-Evans , Proteínas Quinases S6 Ribossômicas 70-kDa/biossíntese , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Serina-Treonina Quinases TOR , Fatores de Tempo
15.
Eur J Neurosci ; 23(7): 1853-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16623842

RESUMO

Synaptic modification supporting memory formation is thought to depend on gene expression and protein synthesis. Disrupting either process around the time of learning prevents the formation of long-term memory. Recent evidence suggests that memory also becomes susceptible to disruption upon retrieval. Whether or not the molecular events involved in the formation of new memory are the same as what is needed for memory to persist after retrieval has yet to be determined. In the present set of experiments, rats were given inhibitors of protein or messenger ribonucleic acid (mRNA) synthesis into the amygdala just after training or retrieval of fear memory. Results showed that blocking mRNA or protein synthesis immediately after learning prevented the formation of long-term memory, while stability of memory after retrieval required protein, but not mRNA, synthesis. These data suggest that the protein needed for memory reconsolidation after retrieval may be transcribed from pre-existing stores of mRNA.


Assuntos
Tonsila do Cerebelo/metabolismo , Medo , Memória , Biossíntese de Proteínas , RNA Mensageiro/biossíntese , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Anisomicina/farmacologia , Líquido Cefalorraquidiano , Dactinomicina/farmacologia , Diclororribofuranosilbenzimidazol/farmacologia , Masculino , Biossíntese de Proteínas/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/antagonistas & inibidores , Ratos , Ratos Long-Evans , Fatores de Tempo
16.
Learn Mem ; 12(6): 573-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16322359

RESUMO

Benzodiazepines have been useful tools for investigating mechanisms underlying learning and memory. The present set of experiments investigates the role of hippocampal GABA(A)/benzodiazepine receptors in memory consolidation using Pavlovian fear conditioning. Rats were prepared with cannulae aimed at the dorsal hippocampus and trained with a series of white noise-shock pairings. In the first experiment, animals received intrahippocampal infusion of midazolam or vehicle immediately or 3 h after training. Then, 24 h later, freezing to the training context and the white noise were measured independently. Results show infusion of midazolam immediately, but not 3 h, after training selectively attenuates contextual fear conditioning. In the second experiment, animals received intrahippocampal infusions of an antisense oligodeoxynucleotide (ODN) targeting the alpha5 subunit of the GABA(A) receptor or a missense control for several days prior to training and testing. Immediately after training, animals received an infusion of either midazolam or vehicle. Western blots conducted after testing showed a significant decrease in alpha5-containing GABA(A) receptor protein. This reduction did not alter the effectiveness of midazolam immediately after training at impairing context fear memory. Therefore, alpha5-containing GABA(A) receptors may not contribute to the effects of midazolam on context fear conditioning when given immediately post-training.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Medo , Moduladores GABAérgicos/administração & dosagem , Hipocampo/efeitos dos fármacos , Midazolam/administração & dosagem , Animais , Aprendizagem por Associação/efeitos dos fármacos , Aprendizagem por Associação/fisiologia , Condicionamento Clássico/fisiologia , Esquema de Medicação , Reação de Congelamento Cataléptica/efeitos dos fármacos , Reação de Congelamento Cataléptica/fisiologia , Hipocampo/metabolismo , Masculino , Microinjeções , Ratos , Ratos Long-Evans , Receptores de GABA-A/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...