Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
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
2.
Brain Res ; 1436: 157-67, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22208646

RESUMO

Long-term expression from helper virus-free Herpes Simplex Virus (HSV-1) vectors is required for many specific neural gene therapies and studies on neuronal physiology. We previously developed a promoter that supports long-term, neuron-specific expression by fusing the chicken ß-globin insulator (INS), followed by an upstream enhancer from the rat tyrosine hydroxylase (TH) promoter, to a neurofilament heavy gene (NFH) promoter. Here, we examined the capability of specific transcription factors to further improve long-term expression from this promoter. Following a HSV-1 virus infection, the virus genome is localized to promyelocytic leukemia protein (PML) nuclear bodies (NB). At these sites, specific cellular transcription factors interact with HSV-1 encoded transcription factors, and together regulate HSV-1 gene expression. Importantly, lysine-specific demethylase-1 (LSD1), CLOCK, and Co-Rest each activate HSV-1 gene expression. However, gene expression from HSV-1 vectors differs in a number of important aspects from the virus, including no HSV-1 genes are expressed. Nonetheless, these observations raise the possibility that specific transcription factors may improve long-term expression from specific promoters in HSV-1 vectors. Here, we show that overexpression of either LSD1 or CLOCK improves long-term expression from the INS-TH-NFH promoter, but overexpression of Co-Rest supports levels of long-term expression similar to those supported by a control vector. Further, overexpression of LSD1 is compatible with neuron-specific expression. Thus, overexpressing specific transcription factors can improve long-term expression from specific cellular promoters in HSV-1 vectors, and the chromatin structure of the vector has an important role in enabling expression.


Assuntos
Proteínas CLOCK/genética , Expressão Gênica , Vetores Genéticos , Histona Desmetilases/genética , Neurônios/metabolismo , Animais , Galinhas , Terapia Genética , Herpesvirus Humano 1/genética , Regiões Promotoras Genéticas , Ratos , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transativadores/genética , Transativadores/metabolismo
3.
Brain Res ; 1415: 127-35, 2011 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-21885042

RESUMO

Because of the numerous types of neurons in the brain, and particularly the forebrain, neuron type-specific expression will benefit many potential applications of direct gene transfer. The two most promising approaches for achieving neuron type-specific expression are targeted gene transfer to a specific type of neuron and using a neuron type-specific promoter. We previously developed antibody-mediated targeted gene transfer with Herpes Simplex Virus (HSV-1) vectors by modifying glycoprotein C (gC) to replace the heparin binding domain, which mediates the initial binding of HSV-1 particles to many cell types, with the Staphylococcus A protein ZZ domain, which binds immunoglobulin (Ig) G. We showed that a chimeric gC-ZZ protein is incorporated into vector particles and binds IgG. As a proof-of-principle for antibody-mediated targeted gene transfer, we isolated complexes of these vector particles and an anti-NMDA NR1 subunit antibody, and demonstrated targeted gene transfer to neocortical cells that contain NR1 subunits. However, because most forebrain neurons contain NR1, we obtained only a modest increase in the specificity of gene transfer, and this targeting specificity is of limited utility for physiological experiments. Here, we report efficient antibody-mediated targeted gene transfer to NMDA NR2B- or NR2A-containing cells in rat postrhinal cortex, and a neuron-specific promoter further restricted recombinant expression to neurons. Of note, because NR2A-containing neurons are relatively rare, these results show that antibody-mediated targeted gene transfer with HSV-1 vectors containing neuron type-specific promoters can restrict recombinant expression to specific types of forebrain neurons of physiological significance.


Assuntos
Expressão Gênica/efeitos dos fármacos , Imunoglobulina G/farmacologia , Neocórtex/citologia , Neurônios/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Animais , Linhagem Celular Transformada , Cricetinae , Expressão Gênica/genética , Marcação de Genes/métodos , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Masculino , Regiões Promotoras Genéticas/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ratos , Ratos Long-Evans , Receptores de N-Metil-D-Aspartato/genética , Simplexvirus/genética , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
4.
Brain Res ; 1351: 1-12, 2010 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-20599821

RESUMO

Because of the heterogeneous cellular composition of the brain, and especially the forebrain, cell type-specific expression will benefit many potential applications of direct gene transfer. The two prevalent approaches for achieving cell type-specific expression are using a cell type-specific promoter or targeting gene transfer to a specific cell type. Targeted gene transfer with Herpes Simplex Virus (HSV-1) vectors modifies glycoprotein C (gC) to replace the heparin binding domain, which binds to many cell types, with a binding activity for a specific cell surface protein. We previously reported targeted gene transfer to nigrostriatal neurons using chimeric gC-glial cell line-derived neurotrophic factor or gC-brain-derived neurotrophic factor protein. Unfortunately, this approach is limited to cells that express the cognate receptor for either neurotrophic factor. Thus, a general strategy for targeting gene transfer to many different types of neurons is desirable. Antibody-mediated targeted gene transfer has been developed for targeting specific virus vectors to specific peripheral cell types; a specific vector particle protein is modified to contain the Staphylococcus A protein ZZ domain, which binds immunoglobulin (Ig) G. Here, we report antibody-mediated targeted gene transfer of HSV-1 vectors to a specific type of forebrain neuron. We constructed a chimeric gC-ZZ protein, and showed this protein is incorporated into vector particles and binds Ig G. Complexes of these vector particles and an antibody to the NMDA receptor NR1 subunit supported targeted gene transfer to NR1-containing neocortical neurons in the rat brain, with long-term (2 months) expression.


Assuntos
Anticorpos Antivirais/genética , Técnicas de Transferência de Genes , Herpesvirus Humano 1/genética , Receptores de N-Metil-D-Aspartato/genética , Proteína Estafilocócica A/genética , Proteínas do Envelope Viral/genética , Animais , Anticorpos Antivirais/biossíntese , Linhagem Celular , Marcação de Genes/métodos , Vetores Genéticos , Vírus Auxiliares/genética , Vírus Auxiliares/metabolismo , Herpesvirus Humano 1/metabolismo , Imunoglobulina G/biossíntese , Imunoglobulina G/genética , Masculino , Proteínas Mutantes Quiméricas/biossíntese , Proteínas Mutantes Quiméricas/genética , Neocórtex/fisiologia , Neurônios/fisiologia , Ratos , Ratos Long-Evans , Receptores de N-Metil-D-Aspartato/biossíntese , Simplexvirus/genética , Simplexvirus/metabolismo , Proteína Estafilocócica A/biossíntese , Proteínas do Envelope Viral/biossíntese
5.
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
6.
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
7.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA