Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Hippocampus ; 29(8): 710-725, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30734387

RESUMO

Advanced cognitive tasks are encoded in distributed neocortical circuits that span multiple forebrain areas. Nonetheless, synaptic plasticity and neural network theories hypothesize that essential information for performing these tasks is encoded in specific ensembles within these circuits. Relatively simpler subcortical areas contain specific ensembles that encode learning, suggesting that neocortical circuits contain such ensembles. Previously, using localized gene transfer of a constitutively active protein kinase C (PKC), we established that a genetically-modified circuit in rat postrhinal cortex, part of the hippocampal formation, can encode some essential information for performing specific visual shape discriminations. However, these studies did not identify any specific neurons that encode learning; the entire circuit might be required. Here, we show that both learning and recall require fast neurotransmitter release from an identified ensemble within this circuit, the transduced neurons; we blocked fast release from these neurons by coexpressing a Synaptotagmin I siRNA with the constitutively active PKC. During learning or recall, specific signaling pathways required for learning are activated in this ensemble; during learning, calcium/calmodulin-dependent protein kinase II, MAP kinase, and CREB are activated; and, during recall, dendritic protein synthesis and CREB are activated. Using activity-dependent gene imaging, we showed that during learning, activity in this ensemble is required to recruit and activate the circuit. Further, after learning, during image presentation, blocking activity in this ensemble reduces accuracy, even though most of the rest of the circuit is activated. Thus, an identified ensemble within a neocortical circuit encodes essential information for performing an advanced cognitive task.


Assuntos
Percepção de Forma/fisiologia , Hipocampo/fisiologia , Aprendizagem/fisiologia , Rede Nervosa/fisiologia , Aprendizagem Espacial/fisiologia , Animais , Rememoração Mental/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , RNA Interferente Pequeno , Ratos , Transdução de Sinais/fisiologia , Sinaptotagmina I/genética , Sinaptotagmina I/metabolismo
2.
Proc Natl Acad Sci U S A ; 107(32): 14478-83, 2010 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-20660720

RESUMO

Learning theories hypothesize specific circuits encode essential information for performance. For simple tasks in invertebrates and mammals, the essential circuits are known, but for cognitive functions, the essential circuits remain unidentified. Here, we show that some essential information for performing a choice task is encoded in a specific circuit in a neocortical area. Rat postrhinal (POR) cortex is required for visual shape discriminations, protein kinase C (PKC) pathways mediate changes in neuronal physiology that support learning, and specific PKC genes are required for multiple learning tasks. We used direct gene transfer of a constitutively active PKC to prime a specific POR cortex circuit for learning visual shape discriminations. In the experiment, rats learned a discrimination, received gene transfer, learned new discriminations, received a small lesion that ablated approximately 21% of POR cortex surrounding the gene transfer site, and were tested for performance for discriminations learned either before or after gene transfer. Lesions of the genetically targeted circuit selectively interfered with performance for discriminations learned after gene transfer. Activity-dependent gene imaging confirmed increased activity in the genetically targeted circuit during learning and showed the essential information was sparse-coded in approximately 500 neurons in the lesioned area. Wild-type rats contained circuits with similar increases in activity during learning, but these circuits were located at unpredictable, different positions in POR cortex. These results establish that some essential information for performing specific visual discriminations can be encoded in a small, identified, neocortical circuit and provide a foundation for characterizing the circuit and essential information.


Assuntos
Córtex Cerebral/fisiologia , Aprendizagem por Discriminação/fisiologia , Proteína Quinase C/metabolismo , Percepção Visual/fisiologia , Animais , Discriminação Psicológica , Vias Neurais , Proteína Quinase C/genética , Ratos , Transfecção
3.
Hippocampus ; 22(12): 2276-89, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22736516

RESUMO

Current theories postulate that the essential information for specific cognitive tasks is widely dispersed in multiple forebrain areas. Nonetheless, synaptic plasticity and neural network theories hypothesize that activation of specific signaling pathways, in specific neurons, modifies synaptic strengths, thereby encoding essential information for performance in localized circuits. Consistent with these latter theories, we have shown that gene transfer of a constitutively active protein kinase C into several hundred glutamatergic and GABAergic neurons in rat postrhinal cortex enhances choice accuracy in visual shape discriminations, and the genetically-modified circuit encodes some of the essential information for performance. However, little is known about the role of specific signaling pathways required for learning, in specific neurons within a critical circuit. Here we show that three learning-associated signaling pathways are coactivated in the transduced neurons during both learning and performance. After gene transfer, but before learning a new discrimination, the calcium/calmodulin-dependent protein kinase (CaMKII), MAP kinase, and CREB pathways were inactive. During learning, these three pathways were coactivated in the transduced neurons. During later performance of the discrimination, CaMKII activity declined, but MAP kinase and CREB activity persisted. Because the transduced neurons are part of a circuit that encodes essential information for performance, activation of these learning-associated signaling pathways, in these identified neurons, is likely important for both learning and performance.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Neocórtex/metabolismo , Neurônios/metabolismo , Reconhecimento Visual de Modelos/fisiologia , Animais , Ativação Enzimática/fisiologia , Aprendizagem/fisiologia , Vias Neurais/metabolismo , Plasticidade Neuronal/fisiologia , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Ratos , Transdução Genética
4.
J Chem Neuroanat ; 118: 102024, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34492329

RESUMO

Neocortical areas comprised of multiple neuronal circuits which are encoded with innumerable advanced cognitive tasks. Studies focused on neuronal network and synaptic plasticity has hypothesized that every specific neuron and the circuit process the explicit essential information for the specific tasks. However, the structure of these circuits and the involved critical neurons remain to be elucidated. Considering our previous studies, showing the specificity of rat postrhinal cortex comprising specific neuronal circuit for encoding both the learning and recall of shape discrimination through a fast neurotransmitter release from the transduced neurons, here we have demonstrated that postsynaptic neurons in two distinct areas, perirhinal cortex and the ventral temporal association areas are required for the specific visual shape discriminations learning. The constitutively active PKC was delivered into neuronal cells in postrhinal cortex, and the animals were allowed to learn the new shape discriminations, and then the silencing siRNA was delivered into postsynaptic neurons in either perirhinal cortex or ventral temporal association areas, using a novel technology for gene transfer into connected neurons. We observed that expression of the siRNA caused the deficits in visual performance, via blocking the activity in the neurons, as displayed by activity-dependent gene imaging, and also subsequently obstructed the activation of specific signaling pathways required for further learning, and dendritic protein synthesis and CREB. Thus, ratifying the conclusion that the two parallel circuits are both required for the visual shape discrimination learning.


Assuntos
Percepção de Forma/fisiologia , Aprendizagem/fisiologia , Neocórtex/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Percepção Visual/fisiologia , Animais , Linhagem Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dendritos/metabolismo , Humanos , Neocórtex/citologia , Rede Nervosa/citologia , Córtex Perirrinal/crescimento & desenvolvimento , Córtex Perirrinal/metabolismo , Proteína Quinase C/metabolismo , RNA Interferente Pequeno , Ratos , Transdução de Sinais/fisiologia , Lobo Temporal/crescimento & desenvolvimento , Lobo Temporal/metabolismo
5.
Hippocampus ; 19(5): 413-23, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-18942114

RESUMO

Age-related decline in human cognition is well known, and there are correlative changes in the function of neocortical and hippocampal neurons. Similarly, age-related decline in learning has been observed in rodents, including deficits in a hippocampal-dependent learning paradigm, the Morris water maze. Furthermore, there are correlative deficits in specific signaling pathways, including protein kinase C (PKC) pathways, in cerebellar, hippocampal, or neocortical neurons. PKC pathways are strong candidates for mediating the molecular changes that underlie spatial learning, as they play critical roles in neurotransmitter release and synaptic plasticity, including long-term potentiation (LTP) and long-term depression (LTD), and deletion of specific PKC genes results in deficits in learning. Conversely, genetic activation of PKC pathways in small groups of hippocampal or cortical neurons enhances learning in specific paradigms. In this study, the authors delivered a constitutively active PKC into small groups of hippocampal dentate granule neurons in aged rats (using a herpes simplex virus-1 vector). Aged 2-year-old rats that received the constitutively active PKC displayed improved performance in the Morris water maze relative to controls in three different measures. These results indicate that PKC pathways play an important role in mediating spatial learning in aged rats. Additionally, these results represent a system for studying the neural mechanisms underlying aging-related learning deficits, and potentially developing gene therapies for cognitive and age-related deficits.


Assuntos
Envelhecimento , Hipocampo/fisiologia , Aprendizagem em Labirinto/fisiologia , Neurônios/fisiologia , Proteína Quinase C/metabolismo , Percepção Espacial/fisiologia , Análise de Variância , Animais , Contagem de Células , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Herpesvirus Humano 1 , Hipocampo/citologia , Imuno-Histoquímica , Deficiências da Aprendizagem/metabolismo , Masculino , Proteína Quinase C/genética , Ratos , Ratos Transgênicos , Comportamento Espacial/fisiologia
6.
BMC Mol Biol ; 10: 58, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19531264

RESUMO

BACKGROUND: Herpes Simplex Virus (HSV-1) gene expression is thought to shut off recombinant gene expression from HSV-1 vectors; however, in a helper virus-free HSV-1 vector system, a number of promoters support only short-term expression. These results raise the paradox that recombinant gene expression remains short-term even in the absence of almost all (approximately 99%) of the HSV-1 genome, HSV-1 genes, and HSV-1 gene expression. To resolve this paradox, we hypothesized that specific proteins in the HSV-1 virus particle shut off recombinant gene expression. In two earlier studies, we examined the effects on recombinant gene expression of packaging vectors using specific mutated HSV-1 proteins. We found that vectors packaged using mutated UL13 (a protein kinase), or VP16, or UL46 and/or UL47 (components of the VP16 transcriptional complex) supported improved long-term expression, and vectors packaged using mutated UL46 and/or UL47 also supported improved gene transfer (numbers of cells at 4 days). These results suggested the hypothesis that specific proteins in the HSV-1 particle act by multiple pathways to reduce recombinant gene expression. To test this hypothesis, we examined combinations of mutated proteins that included both UL13 and specific components of the VP16 transcriptional complex. RESULTS: A HSV-1 vector containing a neuronal-specific promoter was packaged using specific combinations of mutated proteins, and the resulting vector stocks were tested in the rat striatum. For supporting long-term expression, the preferred combination of mutated HSV-1 proteins was mutated UL13, UL46, and UL47. Vectors packaged using this combination of mutated proteins supported a higher efficiency of gene transfer and high levels expression for 3 months, the longest time examined. CONCLUSION: Vector particles containing this combination of mutated HSV-1 proteins improve recombinant gene expression. Implications of these results for strategies to further improve long-term expression are discussed. Moreover, long-term expression will benefit specific gene therapy applications.


Assuntos
Expressão Gênica , Vetores Genéticos , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Humano 1/genética , Proteínas Quinases/metabolismo , Transcrição Gênica , Animais , Corpo Estriado/metabolismo , Vírus Auxiliares , Proteína Vmw65 do Vírus do Herpes Simples/genética , Masculino , Mutação , Ligação Proteica , Proteínas Quinases/genética , Ratos , Ratos Sprague-Dawley
7.
J Mol Neurosci ; 68(4): 549-564, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30972540

RESUMO

mGluR5-containing synapses have essential roles in synaptic plasticity, circuit physiology, and learning, and dysfunction at these synapses is implicated in specific neurological disorders. As mGluR5-containing synapses are embedded in large and complex distributed circuits containing many neuron and synapse types, it is challenging to elucidate the roles of these synapses and to develop treatments for the associated disorders. Thus, it would be advantageous to deliver different genes into pre- and postsynaptic neurons connected by a mGluR5-containing synapse. Here, we develop this capability: The first gene transfer, into the presynaptic neurons, uses standard techniques to deliver a vector that expresses a synthetic peptide neurotransmitter. This peptide neurotransmitter has three domains: a dense core vesicle sorting domain, a mGluR5-binding domain composed of a single-chain variable fragment anti-mGluR5, and the His tag. Upon release, this peptide neurotransmitter binds to mGluR5, predominately located on the postsynaptic neurons. Selective gene transfer into these neurons uses antibody-mediated, targeted gene transfer and anti-His tag antibodies, as the synthetic peptide neurotransmitter contains the His tag. For the model system, we studied the connection between neurons in two neocortical areas: postrhinal and perirhinal cortices. Targeted gene transfer was over 80% specific for mGluR5-containing synapses, but untargeted gene transfer was only ~ 15% specific for these synapses. This technology may enable studies on the roles of mGluR5-containing neurons and synapses in circuit physiology and learning and support gene therapy treatments for specific disorders that involve dysfunction at these synapses.


Assuntos
Técnicas de Transferência de Genes , Neocórtex/citologia , Neurônios/metabolismo , Neurotransmissores/genética , Sinapses/metabolismo , Animais , Linhagem Celular , Cricetinae , Vetores Genéticos/genética , Masculino , Neurotransmissores/metabolismo , Ratos , Ratos Long-Evans , Receptor de Glutamato Metabotrópico 5/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
8.
Brain Res ; 1712: 16-24, 2019 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-30710509

RESUMO

Brain-Derived Neurotrophic Factor (BDNF) signaling through TrkB receptors has important roles in synapse formation, synaptic plasticity, learning, and specific diseases. However, it is challenging to relate BDNF-TrkB synapses to circuit physiology or learning, as BDNF-TrkB synapses are embedded in complex circuits that contain numerous neuron and synapse types. Thus, analyzing the physiology of neurons connected by BDNF-TrkB synapses would be advanced by a technology to deliver different genes into presynaptic and postsynaptic neurons, connected by a BDNF-TrkB synapse. Here, we report selective gene transfer across BDNF-TrkB synapses: The model system was the large projection from rat postrhinal to perirhinal cortex. The first gene transfer, into presynaptic neurons in postrhinal cortex, used a virus vector and standard gene transfer procedures. This vector expresses a synthetic peptide neurotransmitter composed of three domains, a dense core vesicle sorting domain, BDNF, and the His tag. Upon release, this peptide neurotransmitter binds to TrkB receptors on postsynaptic neurons. The second gene transfer, into connected postsynaptic neurons in perirhinal cortex, uses antibody-mediated, targeted gene transfer and an anti-His tag antibody, as the synthetic peptide neurotransmitter contains the His tag. Confocal microscope images showed that using untargeted gene transfer, only 10-15% of the transduced presynaptic axons were proximal to a transduced postsynaptic dendrite. But using targeted gene transfer, ∼70% of the transduced presynaptic axons were proximal to a transduced postsynaptic dendrite. This technology may support studies on the roles of neurons connected by BDNF-TrkB synapses in circuit physiology and learning.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Receptor trkB/metabolismo , Sinapses/metabolismo , Animais , Axônios/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Dendritos/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Interneurônios/metabolismo , Masculino , Neocórtex/metabolismo , Plasticidade Neuronal/fisiologia , Neurônios/metabolismo , Neurotransmissores/metabolismo , Terminações Pré-Sinápticas/metabolismo , Ratos , Ratos Long-Evans , Receptor trkB/genética , Sinapses/genética
9.
J Neurosci Methods ; 327: 108390, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31404560

RESUMO

BACKGROUND: Within a circuit, specific neurons and synapses are hypothesized to have essential roles in circuit physiology and learning, and dysfunction in these neurons and synapses causes specific disorders. These critical neurons and synapses are embedded in complex circuits containing many neuron and synapse types. NEW METHOD: We established technology that can deliver different genes into pre- and post-synaptic neurons connected by a specific synapse type. The first, presynaptic gene transfer employs standard gene transfer technology to express a synthetic peptide neurotransmitter which has three domains, a dense core vesicle sorting domain for processing the protein as a peptide neurotransmitter, a receptor-binding domain, here a small peptide that binds to NMDA NR1 subunits, and the His tag. Upon release, this peptide neurotransmitter binds to its cognate receptor on postsynaptic neurons. Gene transfer selectively into these postsynaptic neurons employs antibody-mediated, targeted gene transfer and anti-His tag antibodies, which recognize the His tag domain in the synthetic peptide neurotransmitter. RESULTS: For the model system, we studied the connection from projection neurons in postrhinal cortex to specific neurons in perirhinal cortex. In our initial report, gene transfer to connected neurons was 20+1% specific. Here, we optimized the technology; we improved the transfection for packaging by using a modern using a modern lipid, Lipofectamine 3000, and used a modern confocal microscope to collect data. We now report 80+2% specific gene transfer to connected neurons. COMPARISON WITH EXISTING METHODS: There is no existing method with this capability. CONCLUSIONS: This technology may enable studies on the roles of specific neurons and synapses in circuit physiology and learning, and support gene therapy treatments for specific disorders.


Assuntos
Neurônios , Receptores de N-Metil-D-Aspartato , Sinapses , Transfecção/métodos , Animais , Vetores Genéticos , Lipídeos , Neocórtex , Ratos
10.
PLoS One ; 14(5): e0217094, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31125364

RESUMO

Local neocortical circuits play critical roles in information processing, including synaptic plasticity, circuit physiology, and learning, and GABAergic inhibitory interneurons have key roles in these circuits. Moreover, specific neurological disorders, including schizophrenia and autism, are associated with deficits in GABAergic transmission in these circuits. GABAergic synapses represent a small fraction of neocortical synapses, and are embedded in complex local circuits that contain many neuron and synapse types. Thus, it is challenging to study the physiological roles of GABAergic inhibitory interneurons and their synapses, and to develop treatments for the specific disorders caused by dysfunction at these GABAergic synapses. To these ends, we report a novel technology that can deliver different genes into pre- and post-synaptic neocortical interneurons connected by a GABAergic synapse: First, standard gene transfer into the presynaptic neurons delivers a synthetic peptide neurotransmitter, containing three domains, a dense core vesicle sorting domain, a GABAA receptor-binding domain, a single-chain variable fragment anti-GABAA ß2 or ß3, and the His tag. Second, upon release, this synthetic peptide neurotransmitter binds to GABAA receptors on the postsynaptic neurons. Third, as the synthetic peptide neurotransmitter contains the His tag, antibody-mediated, targeted gene transfer using anti-His tag antibodies is selective for these neurons. We established this technology by expressing the synthetic peptide neurotransmitter in GABAergic neurons in the middle layers of postrhinal cortex, and the delivering the postsynaptic vector into connected GABAergic neurons in the upper neocortical layers. Targeted gene transfer was 61% specific for the connected neurons, but untargeted gene transfer was only 21% specific for these neurons. This technology may support studies on the roles of GABAergic inhibitory interneurons in circuit physiology and learning, and support gene therapy treatments for specific disorders associated with deficits at GABAergic synapses.


Assuntos
Neurônios GABAérgicos/metabolismo , Interneurônios/metabolismo , Neocórtex/metabolismo , Neurotransmissores/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-B/genética , Sinapses/metabolismo , Animais , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Neurotransmissores/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Receptores de GABA-A/imunologia , Receptores de GABA-A/metabolismo , Receptores de GABA-B/imunologia , Receptores de GABA-B/metabolismo , Anticorpos de Cadeia Única/imunologia
11.
BMC Neurosci ; 9: 37, 2008 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-18402684

RESUMO

BACKGROUND: Direct gene transfer into neurons has potential for developing gene therapy treatments for specific neurological conditions, and for elucidating neuronal physiology. Due to the complex cellular composition of specific brain areas, neuronal type-specific recombinant gene expression is required for many potential applications of neuronal gene transfer. One approach is to target gene transfer to a specific type of neuron. We developed modified Herpes Simplex Virus (HSV-1) particles that contain chimeric glycoprotein C (gC) - glial cell line-derived neurotrophic factor (GDNF) or brain-derived neurotrophic factor (BDNF) proteins. HSV-1 vector particles containing either gC - GDNF or gC - BDNF target gene transfer to nigrostriatal neurons, which contain specific receptors for GDNF or BDNF. A second approach to achieve neuronal type-specific expression is to use a cell type-specific promoter, and we have used the tyrosine hydroxylase (TH) promoter to restrict expression to catecholaminergic neurons or a modified neurofilament heavy gene promoter to restrict expression to neurons, and both of these promoters support long-term expression from HSV-1 vectors. To both improve nigrostriatal-neuron specific expression, and to establish that targeted gene transfer can be followed by long-term expression, we performed targeted gene transfer with vectors that support long-term, neuronal-specific expression. RESULTS: Helper virus-free HSV-1 vector packaging was performed using either gC - GDNF or gC - BDNF and vectors that contain either the TH promoter or the modified neurofilament heavy gene promoter. Vector stocks were injected into the midbrain proximal to the substantia nigra, and the rats were sacrificed at either 4 days or 1 month after gene transfer. Immunofluorescent costaining was performed to detect both recombinant gene products and nigrostriatal neurons. The combination of targeted gene transfer with neuronal-specific promoters improved nigrostriatal neuron-specific expression (83 to 93%) compared to either approach alone, and supported long-term (1 month) expression at levels similar to those observed using untargeted gene transfer. CONCLUSION: Targeted gene transfer can be used in combination with neuronal-specific promoters to achieve a high level of nigrostriatal neuron-specific expression. Targeted gene transfer can be followed by long-term expression. Nigrostriatal neuron-specific expression may be useful for specific gene therapy approaches to Parkinson's disease or for genetic analyses of nigrostriatal neuron physiology.


Assuntos
Corpo Estriado/metabolismo , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Substância Negra/metabolismo , Animais , Transporte Axonal/genética , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Linhagem Celular , Cricetinae , Marcação de Genes/métodos , Técnicas de Transferência de Genes , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Masculino , Vias Neurais/metabolismo , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Substância Negra/citologia , Fatores de Tempo , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo
12.
Brain Res ; 1130(1): 1-16, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17169349

RESUMO

Direct gene transfer into neurons, using a virus vector, has been used to study neuronal physiology and learning, and has potential for supporting gene therapy treatments for specific neurological diseases. Many of these applications require high-level, long-term recombinant gene expression, in forebrain neurons. We previously showed that addition of upstream sequences from the rat tyrosine hydroxylase (TH) promoter to a neurofilament heavy gene (NF-H) promoter supports long-term expression in forebrain neurons, from helper virus-free Herpes Simplex Virus (HSV-1) vectors. This element in the TH promoter satisfied the definition of an enhancer; it displayed activity at a distance from the basal promoter, and in both orientations. This enhancer supported physiological studies that required long-term expression; a modified neurofilament promoter, containing an insulator upstream of the TH-NFH promoter, supported expression in approximately 11,400 striatal neurons at 6 months after gene transfer, and expression for 7, 8, or 14 months, the longest times tested. In contrast, the NF-H promoter alone does not support long-term expression, indicating that the critical sequences are in the 6.3 kb fragment of the TH promoter. In this study, we performed a deletion analysis to identify the critical sequences in the TH promoter that support long-term expression. We localized these critical sequences to an approximately 320 bp fragment, and two subfragments of approximately 100 bp each. Vectors that contained each of these small fragments supported levels of long-term, neuronal-specific expression that were similar to the levels supported by a vector that contained the initial 6.3 kb fragment of the TH promoter. These small fragments of the TH promoter may benefit construction of vectors for physiological studies, and may support studies on the mechanism by which this enhancer supports long-term expression.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica/genética , Técnicas de Transferência de Genes , Proteínas de Neurofilamentos/metabolismo , Regiões Promotoras Genéticas/genética , Tirosina 3-Mono-Oxigenase/genética , Animais , Linhagem Celular , DNA Recombinante/genética , DNA Recombinante/isolamento & purificação , Vetores Genéticos/genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Masculino , Neostriado/citologia , Neostriado/metabolismo , Proteínas de Neurofilamentos/genética , Neurônios/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
13.
Brain Res ; 1144: 19-32, 2007 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-17331479

RESUMO

Many potential uses of direct gene transfer into neurons require restricting expression to one of the two major types of forebrain neurons, glutamatergic or GABAergic neurons. Thus, it is desirable to develop virus vectors that contain either a glutamatergic or GABAergic neuron-specific promoter. The brain/kidney phosphate-activated glutaminase (PAG), the product of the GLS1 gene, produces the majority of the glutamate for release as neurotransmitter, and is a marker for glutamatergic neurons. A PAG promoter was partially characterized using a cultured kidney cell line. The three vesicular glutamate transporters (VGLUTs) are expressed in distinct populations of neurons, and VGLUT1 is the predominant VGLUT in the neocortex, hippocampus, and cerebellar cortex. Glutamic acid decarboxylase (GAD) produces GABA; the two molecular forms of the enzyme, GAD65 and GAD67, are expressed in distinct, but largely overlapping, groups of neurons, and GAD67 is the predominant form in the neocortex. In transgenic mice, an approximately 9 kb fragment of the GAD67 promoter supports expression in most classes of GABAergic neurons. Here, we constructed plasmid (amplicon) Herpes Simplex Virus (HSV-1) vectors that placed the Lac Z gene under the regulation of putative PAG, VGLUT1, or GAD67 promoters. Helper virus-free vector stocks were delivered into postrhinal cortex, and the rats were sacrificed 4 days or 2 months later. The PAG or VGLUT1 promoters supported approximately 90% glutamatergic neuron-specific expression. The GAD67 promoter supported approximately 90% GABAergic neuron-specific expression. Long-term expression was observed using each promoter. Principles for obtaining long-term expression from HSV-1 vectors, based on these and other results, are discussed. Long-term glutamatergic or GABAergic neuron-specific expression may benefit specific experiments on learning or specific gene therapy approaches. Of note, promoter analyses might identify regulatory elements that determine a glutamatergic or GABAergic neuron.


Assuntos
Regulação da Expressão Gênica/fisiologia , Ácido Glutâmico/metabolismo , Neocórtex/citologia , Neurônios/metabolismo , Regiões Promotoras Genéticas/fisiologia , Ácido gama-Aminobutírico/metabolismo , Animais , Contagem de Células/métodos , Linhagem Celular Transformada , Cricetinae , Vetores Genéticos/fisiologia , Glutamato Descarboxilase/genética , Glutaminase/genética , Herpesvirus Humano 1/fisiologia , Masculino , Neocórtex/virologia , Neurônios/virologia , Ratos , Ratos Sprague-Dawley , Proteína Vesicular 1 de Transporte de Glutamato/genética
14.
Behav Brain Res ; 331: 261-275, 2017 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-28511982

RESUMO

Synaptic plasticity and neural network theories hypothesize that the essential information for advanced cognitive tasks is encoded in specific circuits and neurons within distributed neocortical networks. However, these circuits are incompletely characterized, and we do not know if a specific discrimination is encoded in characteristic circuits among multiple animals. Here, we determined the spatial distribution of active neurons for a circuit that encodes some of the essential information for a cognitive task. We genetically activated protein kinase C pathways in several hundred spatially-grouped glutamatergic and GABAergic neurons in rat postrhinal cortex, a multimodal associative area that is part of a distributed circuit that encodes visual object discriminations. We previously established that this intervention enhances accuracy for specific discriminations. Moreover, the genetically-modified, local circuit in POR cortex encodes some of the essential information, and this local circuit is preferentially activated during performance, as shown by activity-dependent gene imaging. Here, we mapped the positions of the active neurons, which revealed that two image sets are encoded in characteristic and different circuits. While characteristic circuits are known to process sensory information, in sensory areas, this is the first demonstration that characteristic circuits encode specific discriminations, in a multimodal associative area. Further, the circuits encoding the two image sets are intermingled, and likely overlapping, enabling efficient encoding. Consistent with reconsolidation theories, intermingled and overlapping encoding could facilitate formation of associations between related discriminations, including visually similar discriminations or discriminations learned at the same time or place.


Assuntos
Cognição/fisiologia , Aprendizagem/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Percepção Visual/fisiologia , Animais , Masculino , Neurônios/fisiologia , Estimulação Luminosa/métodos , Ratos Long-Evans , Sensação/fisiologia
15.
J Neurosci ; 25(37): 8468-81, 2005 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-16162929

RESUMO

Although learning and memory theories hypothesize that memories are encoded by specific circuits, it has proven difficult to localize learning within a cortical area. Neural network theories predict that activation of a small fraction of the neurons in a circuit can activate that circuit. Consequently, altering the physiology of a small group of neurons might potentiate a specific circuit and enhance learning, thereby localizing learning to that circuit. In this study, we activated protein kinase C (PKC) pathways in small groups of neurons in rat postrhinal (POR) cortex. We microinjected helper virus-free herpes simplex virus vectors that expressed a constitutively active PKC into POR cortex. This PKC was expressed predominantly in glutamatergic and GABAergic neurons in POR cortex. This intervention increased phosphorylation of five PKC substrates that play critical roles in neurotransmitter release (GAP-43 and dynamin) or glutamatergic neurotransmission (specific subunits of AMPA or NMDA receptors and myristoylated alanine-rich C kinase substrate). Additionally, activation of PKC pathways in cultured cortical neurons supported activation-dependent increases in release of glutamate and GABA. This intervention enhanced the learning rate and accuracy of visual object discriminations. In individual rats, the numbers of transfected neurons positively correlated with this learning. During learning, neuronal activity was increased in neurons proximal to the transfected neurons. These results demonstrate that potentiating small groups of glutamatergic and GABAergic neurons in POR cortex enhances visual object learning. More generally, these results suggest that learning can be mediated by specific cortical circuits.


Assuntos
Córtex Cerebral/fisiologia , Aprendizagem/fisiologia , Neurônios/fisiologia , Estimulação Luminosa , Proteína Quinase C/deficiência , Proteína Quinase C/metabolismo , Animais , Córtex Cerebral/enzimologia , Ativação Enzimática , Modelos Animais , Neurônios/enzimologia , Proteína Quinase C/genética , Ratos
16.
Brain Res ; 1083(1): 1-13, 2006 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-16545782

RESUMO

Direct gene transfer into neurons in the brain via a virus vector system has potential for both examining neuronal physiology and for developing gene therapy treatments for neurological diseases. Many of these applications require precise control of the levels of recombinant gene expression. The preferred method for controlling the levels of expression is by use of an inducible promoter system, and the tetracycline (tet)-inducible promoter system is the preferred system. Helper virus-free Herpes Simplex Virus (HSV-1) vectors have a number of the advantages, including their large size and efficient gene transfer. Also, we have reported long-term (14 months) expression from HSV-1 vectors that contain a modified neurofilament heavy gene promoter. A number of studies have reported short-term, inducible expression from helper virus-containing HSV-1 vector systems. However, long-term, inducible expression has not been reported using HSV-1 vectors. The goal of this study was to obtain long-term, inducible expression from helper virus-free HSV-1 vectors. We examined two different vector designs for adapting the tet promoter system to HSV-1 vectors. One design was an autoregulatory design; one transcription unit used a tet-regulated promoter to express the tet-regulated transcription factor tet-off, and another transcription unit used a tet-regulated promoter to express the Lac Z gene. In the other vector design, one transcription unit used the modified neurofilament heavy gene promoter to express tet-off, and another transcription unit used a tet-regulated promoter to express the Lac Z gene. The results showed that both vector designs supported inducible expression in cultured fibroblast or neuronal cell lines and for a short time (4 days) in the rat striatum. Of note, only the vector design that used the modified neurofilament promoter to express tet-off supported long-term (2 months) inducible expression in striatal neurons.


Assuntos
Regulação Viral da Expressão Gênica/genética , Técnicas de Transferência de Genes/tendências , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Animais , Corpo Estriado/metabolismo , Cricetinae , Fibroblastos/metabolismo , Vírus Auxiliares/genética , Óperon Lac/genética , Masculino , Proteínas de Neurofilamentos/genética , Células PC12 , Ratos , Ratos Sprague-Dawley , Elementos Reguladores de Transcrição/genética , Proteínas Repressoras/genética , Fatores de Tempo
17.
Brain Res Mol Brain Res ; 139(1): 88-102, 2005 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-15993510

RESUMO

Direct gene transfer into neurons has potential for both studying neuronal physiology and for developing gene therapy treatments for specific neurological conditions. Due to the heterogeneous cellular composition of the brain, cell-type-specific recombinant gene expression is required for many potential applications of neuronal gene transfer. The two prevalent approaches for achieving cell-type-specific expression are to use a cell-type-specific promoter to control recombinant gene expression or to modify a virus vector particle to target gene transfer to a specific cell type. Targeted gene transfer to multiple peripheral cell types has been described, but targeted gene transfer to a specific type of neuron in the brain has yet to be reported. Targeted gene transfer approaches with Herpes Simplex Virus (HSV-1) vectors have focused on modifying glycoprotein C (gC) to remove the heparin binding domain and add a binding activity for a specific protein on the cell surface. This study was designed to develop HSV-1 vectors that target gene transfer to cells that contain receptors for either glial-cell-line-derived neurotrophic factor (GDNF) or brain-derived neurotrophic factor (BDNF), such as nigrostriatal neurons. We isolated chimeric gC-GDNF or chimeric gC-BDNF constructs, and the resulting proteins were incorporated into HSV-1 virus particles. We performed helper virus-free HSV-1 vector packaging in the presence of each chimeric protein. The resulting vector stocks supported 2.2- to 5.0-fold targeted gene transfer to nigrostriatal neurons in the rat brain, compared to vector particles that contained wild-type (wt) gC. Gene transfer to nigrostriatal neurons by vector particles that contained chimeric gC-BDNF was reduced by preincubation with an anti-BDNF antibody. Targeted gene transfer to neurons that contain specific neurotrophic factor receptors may benefit specific physiological and gene therapy studies.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/metabolismo , Vírus Auxiliares/metabolismo , Neurônios/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Linhagem Celular , Corpo Estriado/citologia , Cosmídeos/genética , Cosmídeos/metabolismo , Cricetinae , Vetores Genéticos/genética , Vírus Auxiliares/genética , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Masculino , Neurônios/citologia , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Substância Negra/citologia , Proteínas do Envelope Viral/genética
18.
J Neurosci Methods ; 145(1-2): 1-9, 2005 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-15922021

RESUMO

Herpes simplex virus (HSV-1) gene expression is hypothesized to shut off recombinant gene expression from HSV-1 vectors, but in a helper virus-free HSV-1 vector system, a number of promoters support only short-term expression. Thus paradoxically, recombinant gene expression remains short-term in the absence of almost all (approximately 99%) of the HSV-1 genome. To resolve this paradox, we hypothesize that specific HSV-1 proteins that affect the virion can shut off recombinant gene expression. In an earlier study, we examined the effects on recombinant gene expression of five different proteins that affect the HSV-1 virion. We found that vectors packaged in the presence of mutated vhs or U S 11 exhibited minimal changes in gene expression, vectors packaged in the presence of a mutated U S 3 supported improved gene transfer (numbers of cells at 4 days), and vectors packaged in the presence of mutated U L 13 or VP16 supported improved long-term expression. The capability of the VP16 transcriptional complex to reduce gene expression deserves additional study because VP16 is a powerful enhancer that interacts with a number of cellular and viral proteins. In particular, U L 46 and U L 47 are known to modulate the effects of VP16 on immediate early promoters. In this study, we examined expression from a HSV-1 vector that contains a neuronal-specific promoter and was packaged in the presence of deletions in U L 46, or U L 47, or both U L 46 and U L 47. In the rat striatum, each of these vector stocks supported both improved gene transfer (numbers of cells at 4 days) and improved long-term expression (2 months). Vectors packaged in the presence of a deletion in both U L 46 and U L 47 supported larger improvements in gene expression compared to vectors packaged in the presence of deletions in either gene alone. The implications of these results for strategies to improve long-term expression are discussed.


Assuntos
Antígenos Virais/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Vírus Auxiliares/fisiologia , Herpesvirus Humano 1/fisiologia , Proteínas Virais de Fusão/genética , Proteínas Virais/genética , Animais , Corpo Estriado/virologia , Cosmídeos/genética , Cricetinae , Deleção de Genes , Expressão Gênica , Proteína Vmw65 do Vírus do Herpes Simples/genética , Microinjeções , Regiões Promotoras Genéticas , Ratos , Fatores de Tempo
19.
Brain Res ; 1052(2): 119-29, 2005 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-16018990

RESUMO

Both glial cell line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) can protect nigrostriatal dopaminergic neurons from neurotoxins in rodent and monkey models of Parkinson's disease (PD). These two neurotrophic factors are usually tested individually. This study was designed to compare GDNF, BDNF, or both, for their capabilities to correct behavioral deficits and protect nigrostriatal dopaminergic neurons in a rat model of PD. Gene transfer used a helper virus-free Herpes Simplex Virus (HSV-1) vector system and a modified neurofilament heavy gene promoter that supports long-term expression in forebrain neurons. Rats received unilateral intrastriatal injections of HSV-1 vectors that express either GDNF or BDNF, or both vectors, followed by intrastriatal injections of 6-hydroxydopamine (6-OHDA). Recombinant GDNF or BDNF was detected in striatal neurons in rats sacrificed at 7 months after gene transfer. Of note, GDNF was significantly more effective than BDNF for both correcting behavioral deficits and protecting nigrostriatal dopaminergic neurons. Expression of both neurotrophic factors was no more effective than expression of only GDNF. These results suggest that GDNF is more effective than BDNF for correcting the rat model of PD, and that there are no detectable benefits from expressing both of these neurotrophic factors.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/farmacologia , Corpo Estriado/patologia , Fatores de Crescimento Neural/farmacologia , Neurônios/efeitos dos fármacos , Substância Negra/patologia , Adrenérgicos/toxicidade , Análise de Variância , Animais , Apomorfina/farmacologia , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Fator Neurotrófico Derivado do Encéfalo/uso terapêutico , Contagem de Células , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática/métodos , Lateralidade Funcional , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Imuno-Histoquímica/métodos , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/uso terapêutico , Oxidopamina/toxicidade , Doença de Parkinson/complicações , Doença de Parkinson/etiologia , Doença de Parkinson/terapia , Ratos , Rotação , Simplexvirus/fisiologia
20.
J Neurosci Methods ; 240: 77-88, 2015 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-25448383

RESUMO

BACKGROUND: A central problem in neuroscience is elucidating synaptic connections, the connectome. Because mammalian forebrains contain many neurons, labeling specific neurons with unique tags is desirable. A novel technology, Brainbow, creates hundreds of hues by combinatorial expression of multiple fluorescent proteins (FPs). NEW METHOD: We labeled small numbers of neurons, and their axons, with unique hues, by expressing Brainbow from a helper virus-free Herpes Simplex Virus (HSV-1) vector. RESULTS: The vector expresses a Brainbow cassette containing four FPs from a glutamatergic-specific promoter. Packaging HSV-Brainbow produced arrays of seven to eight Brainbow cassettes, and using Cre, each FP gene was in a position to be expressed, in different cassettes. Delivery into rat postrhinal (POR) cortex or hippocampus labeled small numbers of neurons with different, often unique, hues. An area innervated by POR cortex, perirhinal (PER) cortex, contained axons with different hues. Specific axons in PER cortex were matched to specific cell bodies in POR cortex, using hue. COMPARISON WITH EXISTING METHODS: HSV-Brainbow is the only technology for labeling small numbers of neurons with unique hues. In Brainbow mice, many neurons contain the same hue. Brainbow-adeno-associated virus vectors require transduction of the same neuron with multiple vector particles, confounding neuroanatomical studies. Replication-competent Brainbow-pseudorabies virus vectors label multiple neurons with the same hue. CONCLUSIONS: Attractive properties of HSV-Brainbow include each vector particle contains multiple cassettes, representing numerous hues, recombination products are stabile, and experimental control of the number of labeled neurons. Labeling neurons with unique hues will benefit mapping forebrain circuits.


Assuntos
Vetores Genéticos , Herpesvirus Humano 1/genética , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Neurônios/metabolismo , Animais , Axônios/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Cricetinae , Fibroblastos/citologia , Fibroblastos/metabolismo , Técnicas de Transferência de Genes , Masculino , Neurônios/citologia , Ratos Sprague-Dawley , Recombinação Genética , Proteína Vesicular 1 de Transporte de Glutamato/genética , Proteína Vesicular 1 de Transporte de Glutamato/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA