Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Methods ; 66(3): 466-73, 2014 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23816789

RESUMO

The abilities of lentiviral vectors to carry large transgenes (∼8kb) and to efficiently infect and integrate these genes into the genomes of both dividing and non-dividing cells make them ideal candidates for transport of genetic material into cells and tissues. Given the properties of these vectors, it is somewhat surprising that they have seen only limited use in studies of developing tissues and in particular of the developing nervous system. Over the past several years, we have taken advantage of the large capacity of these vectors to explore the expression characteristics of several dual promoter and 2A peptide bicistronic transgenes in developing chick neural retina, with the goal of identifying transgene designs that reliably express multiple proteins in infected cells. Here we summarize the activities of several of these transgenes in neural retina and provide detailed methodologies for packaging lentivirus and delivering the virus into the developing neural tubes of chicken embryos in ovo, procedures that have been optimized over the course of several years of use in our laboratory. Conditions to hatch injected embryos are also discussed. The chicken-specific techniques will be of highest interest to investigators using avian embryos, development and packaging of lentiviral vectors that reliably express multiple proteins in infected cells should be of interest to all investigators whose experiments demand manipulation and expression of multiple proteins in developing cells and tissues.


Assuntos
Desenvolvimento Embrionário/genética , Engenharia Genética/métodos , Vetores Genéticos , Lentivirus/genética , Transgenes , Animais , Animais Geneticamente Modificados/genética , Embrião de Galinha , Retina/embriologia
2.
J Neurosci ; 33(6): 2626-38, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23392690

RESUMO

The formation of primary cilia is a highly choreographed process that can be disrupted in developing neurons by overexpressing neuromodulatory G-protein-coupled receptors GPCRs or by blocking intraflagellar transport. Here, we examined the effects of overexpressing the ciliary GPCRs, 5HT6 and SSTR3, on cilia structure and the differentiation of neocortical neurons. Neuronal overexpression of 5HT6 and SSTR3 was achieved by electroporating mouse embryo cortex in utero with vectors encoding these receptors. We found that overexpression of ciliary GPCRs in cortical neurons, especially 5HT6, induced the formation of long (>30 µm) and often forked cilia. These changes were associated with increased levels of intraflagellar transport proteins and accelerated ciliogenesis in neonatal neocortex, the induction of which required Kif3a, an anterograde motor critical for cilia protein trafficking and growth. GPCR overexpression also altered the complement of signaling molecules within the cilia. We found that SSTR3 and type III adenylyl cyclase (ACIII), proteins normally enriched in neuronal cilia, were rarely detected in 5HT6-elongated cilia. Intriguingly, the changes in cilia structure were accompanied by changes in neuronal morphology. Specifically, disruption of normal ciliogenesis in developing neocortical neurons, either by overexpressing cilia GPCRs or a dominant-negative form of Kif3a, significantly impaired dendrite outgrowth. Remarkably, coexpression of ACIII with 5HT6 restored ACIII to cilia, normalized cilia structure, and restored dendrite outgrowth, effects that were not observed in neurons coexpressing ACIII and dominant-negative form of Kif3a. Collectively, our data suggest the formation of neuronal dendrites in developing neocortex requires structurally normal cilia enriched with ACIII.


Assuntos
Adenilil Ciclases/fisiologia , Cílios/enzimologia , Dendritos/enzimologia , Neocórtex/enzimologia , Neurônios/enzimologia , Receptores de Serotonina/biossíntese , Animais , Células Cultivadas , Cílios/fisiologia , Feminino , Cinesinas/biossíntese , Masculino , Camundongos , Células NIH 3T3 , Neocórtex/embriologia , Neurogênese/fisiologia , Neurônios/citologia , Gravidez
3.
J Neurosci ; 33(6): 2671-83, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23392694

RESUMO

The expression of the ERα and ERß estrogen receptors in the hippocampus may be important in the etiology of age-related cognitive decline. To examine the role of ERα and ERß in regulating transcription and learning, ovariectomized wild-type (WT) and ERα and ERß knockout (KO) mice were used. Hippocampal gene transcription in young ERαKO mice was similar to WT mice 6 h after a single estradiol treatment. In middle-age ERαKO mice, hormone deprivation was associated with a decrease in the expression of select genes associated with the blood-brain barrier; cyclic estradiol treatment increased transcription of these select genes and improved learning in these mice. In contrast to ERαKO mice, ERßKO mice exhibited a basal hippocampal gene profile similar to WT mice treated with estradiol and, in the absence of estradiol treatment, young and middle-age ERßKO mice exhibited preserved learning on the water maze. The preserved memory performance of middle-age ERßKO mice could be reversed by lentiviral delivery of ERß to the hippocampus. These results suggest that one function of ERß is to regulate ERα-mediated transcription in the hippocampus. This model is supported by our observations that knockout of ERß under conditions of low estradiol allowed ERα-mediated transcription. As estradiol levels increased in the absence of ERα, we observed that other mechanisms, likely including ERß, regulated transcription and maintained hippocampal-dependent memory. Thus, our results indicate that ERα and ERß interact with hormone levels to regulate transcription involved in maintaining hippocampal function during aging.


Assuntos
Envelhecimento/fisiologia , Receptor alfa de Estrogênio/fisiologia , Receptor beta de Estrogênio/fisiologia , Hipocampo/fisiologia , Animais , Feminino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout
4.
J Neurooncol ; 117(1): 15-24, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24510433

RESUMO

Glioblastoma (GBM) is the most common malignant adult brain tumor and carries a poor prognosis due to primary and acquired resistance. While many cellular features of GBM have been documented, it is unclear if cells within these tumors extend a primary cilium, an organelle whose associated signaling pathways may regulate proliferation, migration, and survival of neural precursor and tumor cells. Using immunohistochemical and electron microscopy (EM) techniques, we screened human GBM tumor biopsies and primary cell lines for cilia. Immunocytochemical staining of five primary GBM cell lines revealed that between 8 and 25 % of the cells in each line possessed gamma tubulin-positive basal bodies from which extended acetylated, alpha-tubulin-positive axonemes. EM analyses confirmed the presence of cilia at the cell surface and revealed that their axonemes contained organized networks of microtubules, a structural feature consistent with our detection of IFT88 and Arl13b, two trafficked cilia proteins, along the lengths of the axonemes. Notably, cilia were detected in each of 23 tumor biopsies (22 primary and 1 recurrent) examined. These cilia were distributed across the tumor landscape including regions proximal to the vasculature and within necrotic areas. Moreover, ciliated cells within these tumors co-stained with Ki67, a marker for actively dividing cells, and ZEB1, a transcription factor that is upregulated in GBM and linked to tumor initiation, invasion, and chemoresistance. Collectively, our data show that subpopulations of cells within human GBM tumors are ciliated. In view of mounting evidence supporting roles of primary cilia in tumor initiation and propagation, it is likely that further study of the effects of cilia on GBM tumor cell function will improve our understanding of GBM pathogenesis and may provide new directions for GBM treatment strategies.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/ultraestrutura , Cílios/ultraestrutura , Glioblastoma/metabolismo , Glioblastoma/ultraestrutura , Fatores de Ribosilação do ADP/metabolismo , Idoso de 80 Anos ou mais , Axonema/metabolismo , Axonema/ultraestrutura , Corpos Basais/metabolismo , Corpos Basais/ultraestrutura , Linhagem Celular Tumoral , Cílios/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Masculino , Microscopia Eletrônica , Pessoa de Meia-Idade , Fatores de Transcrição/metabolismo , Tubulina (Proteína)/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Homeobox 1 de Ligação a E-box em Dedo de Zinco
5.
Cereb Cortex ; 21(2): 338-44, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20525772

RESUMO

Most, if not all, cortical neurons possess a single primary cilium; however, little is known about the mechanisms that control neuronal ciliogenesis. The Citron kinase-deficient (Citron-K(fh/fh)) rat, a model in which failed cytokinesis during development produces cortical neurons containing multiple cellular organelles, provides a unique system in which to examine the relationship between centriole inheritance and neuronal ciliogenesis. In this study, we analyzed the cerebral cortex of these animals using immunohistochemistry, serial confocal, and electron microscopy to determine if the multinucleated neurons present in the cortex of these animals also possess multiple centrioles and cilia. We found that neurons containing multiple nuclei possessed multiple centrioles and cilia whose lengths varied across cortical regions. Despite the presence of multiple cilia, we found that perinatal expression of adenylyl cyclase III, a cilia-specific marker, and somatostatin receptor 3, a receptor enriched in cilia, were preserved in developing Citron-K(fh/fh) brain. Together, these results show that multinucleated neurons arising from defective cytokinesis can extend multiple cilia.


Assuntos
Cílios/fisiologia , Citocinese/genética , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Neurônios/citologia , Proteínas Serina-Treonina Quinases/deficiência , Córtex Somatossensorial/citologia , Células-Tronco/metabolismo , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Cílios/ultraestrutura , Hipocampo/citologia , Microscopia Confocal , Microscopia Eletrônica/métodos , Modelos Biológicos , Mutação/genética , Neurônios/metabolismo , Neurônios/ultraestrutura , Fosfopiruvato Hidratase/metabolismo , Transporte Proteico/genética , Ratos , Ratos Wistar , Receptores de Somatostatina/metabolismo , Células-Tronco/ultraestrutura
6.
J Neurochem ; 115(4): 941-55, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20819118

RESUMO

Pleckstrin homology and leucine rich repeat protein phosphatases (PHLPPs) are a novel class of potent protein kinase B (AKT) inhibitors that have been intensely investigated in relation to AKT activity in cancer. Currently, our understanding of the role of PHLPP1α in the central nervous system is limited. In this study, we characterized PHLPP protein expression and target kinases in the adult hippocampus. We directly verify PHLPP1α inhibits AKT in hippocampal neurons and demonstrate a novel role for PHLPP1ß/SCOP, to promote AKT activation. PHLPP1α expression changes dramatically in the hippocampus during development, constituting the most abundant PHLPP protein in adult neurons. Further, while all PHLPP proteins could be observed in the cytosolic fraction, only PHLPP1α could be localized to the nucleus. The results provide unique evidence for a divergence in the function of PHLPP1α and PHLPP1ß/SCOP, and suggest that PHLPP1α plays a major role in regulating AKT signaling in neurons.


Assuntos
Hipocampo/fisiologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Fosfoproteínas Fosfatases/genética , Proteína Quinase C-alfa/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Splicing de RNA , Transdução de Sinais/genética , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Linhagem Celular , Células Cultivadas , Feminino , Hipocampo/embriologia , Hipocampo/enzimologia , Humanos , Isoenzimas/genética , Isoenzimas/fisiologia , Masculino , Neurônios/enzimologia , Neurônios/metabolismo , Proteínas Nucleares/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Splicing de RNA/fisiologia , Ratos , Ratos Endogâmicos BN , Ratos Endogâmicos F344
7.
J Neurosci Res ; 88(12): 2558-68, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20648646

RESUMO

The process of Schwann cell myelination requires precisely coordinated gene expression. At the onset of myelination, there is an increase in the expression of differentiation-promoting transcription factors that regulate key Schwann cell genes. Further control of myelin gene expression occurs at the posttranscriptional level and, in part, is mediated by RNA binding proteins and micro-RNAs (miRNAs). miRNAs are small, endogenously derived RNA molecules that repress gene expression by specifically binding to their mRNA targets. In the experiments described here, we tested whether miRNAs were essential in controlling myelination by reducing the levels of Dicer, an essential endoribonuclease in miRNA biogenesis. We decreased the expression of Dicer by about 60% within Schwann cells using a lentiviral vector expressing an shRNA against Dicer. The reduced levels of Dicer led to a decrease in the steady-state expression of selected miRNAs and of the transcription factors Oct6 and Egr2/Krox20, both of which are critical for Schwann cells differentiation and myelination. In contrast, the levels of c-jun and Sox2 were up-regulated by the reduction in Dicer and were associated with an increase in Schwann cell proliferation. In dorsal root ganglion cocultures, Schwann cells transduced with Dicer shRNA synthesized less myelin, which was accompanied by significant reductions in the levels of myelin basic protein and protein zero. These findings support a critical role for Dicer and miRNAs in Schwann cell differentiation and myelination.


Assuntos
Diferenciação Celular/genética , MicroRNAs/genética , Bainha de Mielina/metabolismo , Ribonuclease III/genética , Ribonuclease III/metabolismo , Células de Schwann/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Técnicas de Cocultura , Regulação para Baixo/fisiologia , Gânglios Espinais/citologia , Gânglios Espinais/metabolismo , Bainha de Mielina/fisiologia , Interferência de RNA/fisiologia , Ratos , Células de Schwann/citologia , Células Receptoras Sensoriais/citologia , Células Receptoras Sensoriais/metabolismo , Células-Tronco/metabolismo , Células-Tronco/fisiologia , Regulação para Cima/fisiologia
8.
Mol Vis ; 16: 916-34, 2010 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-20517486

RESUMO

PURPOSE: Growing evidence suggests that successful treatment of many inherited photoreceptor diseases will require multi-protein therapies that not only correct the genetic defects linked to these diseases but also slow or halt the related degenerative phenotypes. To be effective, it is likely that therapeutic protein expression will need to be targeted to specific cell types. The purpose of this study was to develop dual-promoter lentiviral vectors that target expression of two proteins to retinal cones and rods, rods only, or Müller cells. METHODS: Dual-promoter lentivectors were constructed using the following promoters: Xenopus opsin promoter (XOPS)1.3, murine opsin promoter (MOPS), interphotoreceptor retinoid binding protein promoter (IRBP156), rhodopsin kinase (RK), neural retina leucine zipper (NRLL), vimentin (VIM), cluster differentiation (CD44), and glial fibrillary acidic protein (GFAP). Vectors were packaged and injected into the neural tubes of chicken embryos. The activities of the promoters alone, in duplicate, or when paired with a different promoter were analyzed in transduced, fully-developed retinas, using direct fluorescent and immunofluorescent microscopy. RESULTS: IRBP156, NRLL, and RK were active in cones and rods while XOPS1.3 was active only in rods. Of the glial promoters, only GFAP activity was restricted to Müller cells; both VIM and CD44 were active in Müller and neural cells. Dual-promoter vectors carrying IRBP156 and RK or XOPS1.3 and MOPS, in the order listed, exhibited robust expression of both reporter transgenes in cones and rods or rods only, respectively. Expression of the upstream transgene was much lower than the downstream transgene in dual-promoter vectors constructed using two copies of either RK or IRBP156. Analyses of the expression of a dual-promoter vector carrying CD44 and VIM in the order listed showed that the activity of the VIM promoter was more restricted to glial cells when paired with the CD44 promoter, while the activity of the CD44 promoter was inhibited to the extent that no CD44-driven reporter protein was detected in transduced cells. CONCLUSIONS: We have identified two dual-promoter vectors, one that targets cones and rods and one that targets rods alone. Both vectors reliably express the two proteins encoded by the transgenes they carry. When two well matched promoters are not available, we found that it is possible to target expression of two proteins to single cells using dual-promoter vectors carrying two copies of the same promoter. These vectors should be useful in studies of retina when co-delivery of a reporter protein with an experimental protein is desired or when expression of two exogenous proteins in targeted cells is required.


Assuntos
Marcação de Genes/métodos , Vetores Genéticos , Lentivirus/genética , Células Fotorreceptoras de Vertebrados , Regiões Promotoras Genéticas , Retina/citologia , Animais , Embrião de Galinha , Expressão Gênica , Camundongos/genética , Células Fotorreceptoras de Vertebrados/metabolismo , Retina/embriologia , Retina/metabolismo , Transgenes , Xenopus/genética
9.
Biochem Biophys Res Commun ; 390(4): 1149-53, 2009 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19878658

RESUMO

The gamma subunit of rod-specific cGMP phosphodiesterase 6 (PDE6gamma), an effector of the G-protein GNAT1, is a key regulator of phototransduction. The results of several in vitro biochemical reconstitution experiments conducted to examine the effects of phosphorylation of PDE6gamma on its ability to regulate the PDE6 catalytic core have been inconsistent, showing that phosphorylation of PDE6gamma may increase or decrease the ability of PDE6gamma to deactivate phototransduction. To resolve role of phosphorylation of PDE6gamma in living photoreceptors, we generated transgenic mice in which either one or both Threonine (T) sites in PDE6gamma (T22 and T35), which are candidates for putative regulatory phosphorylation, were substituted with alanine (A). Phosphorylation of these sites was examined as a function of light exposure. We found that phosphorylation of T22 increases with light exposure in intact mouse rods while constitutive phosphorylation of T35 is unaffected by light in intact mouse rods and cones. Phosphorylation of the cone isoform of PDE6gamma, PDE6H, is constitutively phosphorylated at the T20 residue. Light-induced T22 phosphorylation was lost in T35A transgenic rods, and T35 phosphorylation was extinguished in T22A transgenic rods. The interdependency of phosphorylation of T22 and T35 suggests that light-induced, post-translational modification of PDE6gamma is essential for the regulation of G-protein signaling.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Luz , Processamento de Proteína Pós-Traducional , Células Fotorreceptoras Retinianas Cones/efeitos da radiação , Células Fotorreceptoras Retinianas Bastonetes/efeitos da radiação , Animais , Anticorpos Fosfo-Específicos/imunologia , Bovinos , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Transdução de Sinal Luminoso , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Fosforilação , Células Fotorreceptoras Retinianas Cones/enzimologia , Células Fotorreceptoras Retinianas Bastonetes/enzimologia , Transdução de Sinais , Treonina/imunologia , Treonina/metabolismo , Transducina/metabolismo
10.
Mol Ther ; 16(9): 1587-1593, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28189013

RESUMO

Estrogen, which influences both classical genomic and rapid membrane-associated signaling cascades, has been implicated in the regulation of hippocampal function, including spatial learning. Gene mutation studies suggest that estrogen effects are mediated by estrogen receptor-α (ER-α); however, because gonadal steroids influence the organization of the hippocampus during development, it has been difficult to distinguish developmental effects from those specific to adults. In this study we show that lentiviral delivery of the gene encoding ER-α to the hippocampus of adult ER-α-knockout (ER-αKO) mice restores hippocampal responsiveness to estrogen and rescues spatial learning. We propose that constitutive estrogen receptor activity is important for maintaining hippocampus-dependent memory function in adults.

11.
Mol Ther ; 16(9): 1587-93, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18594506

RESUMO

Estrogen, which influences both classical genomic and rapid membrane-associated signaling cascades, has been implicated in the regulation of hippocampal function, including spatial learning. Gene mutation studies suggest that estrogen effects are mediated by estrogen receptor-alpha (ER-alpha); however, because gonadal steroids influence the organization of the hippocampus during development, it has been difficult to distinguish developmental effects from those specific to adults. In this study we show that lentiviral delivery of the gene encoding ER-alpha to the hippocampus of adult ER-alpha-knockout (ER-alphaKO) mice restores hippocampal responsiveness to estrogen and rescues spatial learning. We propose that constitutive estrogen receptor activity is important for maintaining hippocampus-dependent memory function in adults.


Assuntos
Receptor alfa de Estrogênio/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Hipocampo/metabolismo , Aprendizagem em Labirinto , Percepção Espacial , Animais , Benzoatos/farmacologia , Encéfalo/fisiologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Eletrofisiologia , Estradiol/farmacologia , Feminino , Proteínas de Fluorescência Verde/genética , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Elementos de Resposta/fisiologia , Rabdomiossarcoma/metabolismo , Rabdomiossarcoma/terapia , Transmissão Sináptica/fisiologia
12.
Front Cell Neurosci ; 13: 55, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30842728

RESUMO

Primary cilia are microtubule-based organelles that are typically present on cells during the G0 or G1-S/G2 phases of the cell cycle. Recent studies of glioblastoma (GBM) biopsies, a brain tumor that is notorious for its aggressive growth and resistance to treatment, show that many cells in the tumor lack cilia. At this point, it remains unclear whether primary cilia promote or suppress glioma tumorigenesis. In this review, we will discuss the different roles that have been proposed for primary cilia in glioma and how cilia may contribute to the resistance of these tumors to current therapies.

13.
Mol Vis ; 13: 2001-11, 2007 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-17982424

RESUMO

PURPOSE: There is increasing interest in developing viral vectors capable of reliably delivering multiple therapeutic genes to targeted cell populations. Currently, bicistronic vectors carrying two transgenes linked by an internal ribosomal entry site (IRES) are the most commonly employed vectors to accomplish this goal. We and others have found that the protein encoded downstream of the IRES in these vectors is not reliably expressed. The purpose of this study was to determine if replacement of the IRES in our self-inactivating, insulated, lentiviral vectors with a second, independent, cell-specific promoter would produce a vector that reliably expressed two proteins in targeted retinal cells in vivo. METHODS: Five dual promoter lentiviral vectors were constructed using our self-inactivating (SIN), insulated, lentiviral backbone. Each vector carried two independent transgenes encoding a fluorescent protein (GFP or tdTomato) whose expression was driven by three photoreceptor promoters (interphotoreceptor retinoid binding protein-IRPB1783; guanylate cyclase activating protein 1-GCAP292; rhodopsin-mOP500) and one ubiquitously expressed promoter (elongation factor 1alpha-EF1alpha). Constructs were packaged and injected into the optic vesicles of developing chicken embryos. The day before hatching, the retinas were removed and examined as whole mount tissues and as frozen sections using fluorescent microscopy. RESULTS: In our first experiment, we characterized the expression of the three photoreceptor promoters in chicken retina. The activities of GCAP292 and IRBP1783 were restricted to cone cells. GCAP292 was also active in a small sub-group of inner nuclear cells. The activity of mOP500 was restricted to rod cells. In our second experiment, we characterized the activity of three dual promoter vectors: GCAP292-GFP-IRBP1783-tdTomato, IRBP-tdTomato-GCAP292-GFP, and IRBP1783-tdTomato-mOP500-GFP. All three vectors produced easily detectable levels of GFP and tdTomato in transduced retinas, a result that prompted further analyses of the expression characteristics of these vectors. In retinas treated with either of the GCAP292/IRBP1783 dual promoter vectors, GFP and tdTomato were only detected in cone cells. No GFP was detected in the inner retina. In retinas treated with IRBP1783-tdTomato-mOP500-GFP, tdTomato was detected only in cone cells and GFP was detected only in rod cells, a result indicating that these promoters retained their intrinsic expression specificities in this dual promoter vector. In our final experiment, the ubiquitously expressed EF1alpha promoter was paired with either GCAP292 or mOP500 creating EF1alpha-tdTomato-GCAP292-GFP and EF1alpha-tdTomato-mOP-GFP. In retinas treated with EF1alpha-tdTomato-GCAP292-GFP, GFP was only detected in cone cells. In retinas treated with EF1alpha-tdTomato-mOP500-GFP, GFP was detected in rod cells and in several cells within the inner retina. CONCLUSIONS: The results of this study show that it is possible to construct dual promoter lentiviral vectors that reliably express two proteins in a cell-specific manner. Among the dual promoter vectors created for this study, we have identified two vectors that specifically target expression of both transgenes to cone cells and one vector that specifically targets expression of one transgene to cone cells and the other transgene to rod cells. The ability to create one lentiviral vector that is capable of targeting expression of multiple genes to single or multiple cells in vivo should prove very useful in the development and delivery of complex, combination therapies to diseased tissues.


Assuntos
Marcação de Genes/métodos , Vetores Genéticos , Lentivirus/genética , Regiões Promotoras Genéticas , Retina/metabolismo , Transgenes , Animais , Embrião de Galinha , Proteínas do Olho/genética , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas Ativadoras de Guanilato Ciclase/genética , Lentivirus/fisiologia , Proteínas Luminescentes/genética , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Retina/embriologia , Proteínas de Ligação ao Retinol/genética , Rodopsina/genética , Distribuição Tecidual , Inativação de Vírus
14.
PLoS Med ; 3(6): e201, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16700630

RESUMO

BACKGROUND: Leber congenital amaurosis (LCA) is a genetically heterogeneous group of retinal diseases that cause congenital blindness in infants and children. Mutations in the GUCY2D gene that encodes retinal guanylate cyclase-1 (retGC1) were the first to be linked to this disease group (LCA type 1 [LCA1]) and account for 10%-20% of LCA cases. These mutations disrupt synthesis of cGMP in photoreceptor cells, a key second messenger required for function of these cells. The GUCY1*B chicken, which carries a null mutation in the retGC1 gene, is blind at hatching and serves as an animal model for the study of LCA1 pathology and potential treatments in humans. METHODS AND FINDINGS: A lentivirus-based gene transfer vector carrying the GUCY2D gene was developed and injected into early-stage GUCY1*B embryos to determine if photoreceptor function and sight could be restored to these animals. Like human LCA1, the avian disease shows early-onset blindness, but there is a window of opportunity for intervention. In both diseases there is a period of photoreceptor cell dysfunction that precedes retinal degeneration. Of seven treated animals, six exhibited sight as evidenced by robust optokinetic and volitional visual behaviors. Electroretinographic responses, absent in untreated animals, were partially restored in treated animals. Morphological analyses indicated there was slowing of the retinal degeneration. CONCLUSIONS: Blindness associated with loss of function of retGC1 in the GUCY1*B avian model of LCA1 can be reversed using viral vector-mediated gene transfer. Furthermore, this reversal can be achieved by restoring function to a relatively low percentage of retinal photoreceptors. These results represent a first step toward development of gene therapies for one of the more common forms of childhood blindness.


Assuntos
Cegueira/enzimologia , Guanilato Ciclase/genética , Receptores de Superfície Celular/genética , Degeneração Retiniana/enzimologia , Animais , Cegueira/genética , Cegueira/patologia , Linhagem Celular , Embrião de Galinha , Galinhas , Modelos Animais de Doenças , Eletrorretinografia , Movimentos Oculares , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Lentivirus/genética , Mutação , Células Fotorreceptoras de Vertebrados/enzimologia , Proteínas Recombinantes de Fusão/metabolismo , Retina/enzimologia , Retina/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Transfecção , Percepção Visual
15.
Invest Ophthalmol Vis Sci ; 47(9): 3745-53, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16936082

RESUMO

PURPOSE: Cone function and survival are compromised in the guanylate cyclase-1 (GC1) knockout mouse. Disruption of the light-driven translocation of cone arrestin is one of the phenotypes of cone cells in this retina: the cone arrestin in these cells is localized to the outer segments and synaptic terminals, regardless of the state of light adaptation. The purpose of this study was to determine whether the expression of GC1 restores cone arrestin translocation in the cone cells of postnatal GC1 knockout mouse retina. METHODS: Subretinal injections of AAV-GC1 were performed on 3-week-old GC1 KO mice. Electroretinographic and immunohistochemical analyses of treated retinas were carried out 5 weeks after injection. GC1 and cone arrestin antibodies were used to identify photoreceptors transduced by the AAV vector and to localize cone arrestin within cone cells, respectively. RESULTS: Treatment of GC1 knockout retinas with AAV-GC1 restored the light-driven translocation of cone arrestin in transduced cone cells. Staining patterns for cone arrestin in transduced and wild-type cone cells were indistinguishable after dark and light adaptation. In dark-adapted retinas, cone arrestin was distributed throughout the subcellular compartments of the cone cells. In light-adapted retinas, cone arrestin was concentrated in the cone outer segments. Successful restoration of cone arrestin translocation did not translate to a restoration of cone ERG responses, which remained undetectable in the treated retinas. CONCLUSIONS: AAV-mediated expression of GC1 in a subpopulation of cone cells in postnatal GC1 knockout retina restores light-driven translocation of cone arrestin in these cells. These findings, which show that fully developed cone cells that have developed in the absence of GC1 can respond to viral-mediated expression of this enzyme, support further analysis of this animal model of Leber congenital amaurosis type 1 (LCA1), a disease that results from null mutations in the gene encoding this enzyme.


Assuntos
Arrestina/metabolismo , Dependovirus/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Guanilato Ciclase/genética , Luz , Transporte Proteico/efeitos da radiação , Receptores de Superfície Celular/genética , Células Fotorreceptoras Retinianas Cones/metabolismo , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais , Arrestina/química , Eletrorretinografia , Técnica Indireta de Fluorescência para Anticorpo , Técnicas de Transferência de Genes , Vetores Genéticos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microscopia Confocal , Dados de Sequência Molecular , Fragmentos de Peptídeos/imunologia , Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/efeitos da radiação , Transgenes , Visão Ocular
16.
Front Aging Neurosci ; 8: 127, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27303293

RESUMO

The primary cilia of forebrain neurons assemble around birth and become enriched with neuromodulatory receptors. Our understanding of the permanence of these structures and their associated signaling pathways in the aging brain is poor, but they are worthy of investigation because disruptions in neuronal cilia signaling have been implicated in changes in learning and memory, depression-like symptoms, and sleep anomalies. Here, we asked whether neurons in aged forebrain retain primary cilia and whether the staining characteristics of aged cilia for type 3 adenylyl cyclase (ACIII), somatostatin receptor 3 (SSTR3), and pericentrin resemble those of cilia in younger forebrain. To test this, we analyzed immunostained sections of forebrain tissues taken from young and aged male Fischer 344 (F344) and F344 × Brown Norway (F344 × BN) rats. Analyses of ACIII and SSTR3 in young and aged cortices of both strains of rats revealed that the staining patterns in the neocortex and hippocampus were comparable. Virtually every NeuN positive cell examined possessed an ACIII positive cilium. The lengths of ACIII positive cilia in neocortex were similar between young and aged for both strains, whereas in F344 × BN hippocampus, the cilia lengths increased with age in CA1 and CA3, but not in dentate gyrus (DG). Additionally, the percentages of ACIII positive cilia that were also SSTR3 positive did not differ between young and aged tissues in either strain. We also found that pericentrin, a protein that localizes to the basal bodies of neuronal cilia and functions in primary cilia assembly, persisted in aged cortical neurons of both rat strains. Collectively, our data show that neurons in aged rat forebrain possess primary cilia and that these cilia, like those present in younger brain, continue to localize ACIII, SSTR3, and pericentrin. Further studies will be required to determine if the function and signaling pathways regulated by cilia are similar in aged compared to young brain.

17.
Exp Neurol ; 282: 119-27, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27181411

RESUMO

Neonatal or early-life seizures (ELS) are often associated with life-long neurophysiological, cognitive and behavioral deficits, but the underlying mechanisms contributing to these deficits remain poorly understood. Newborn, post-migratory cortical neurons sprout ciliary buds (procilia) that mature into primary cilia. Disruption of the growth or signaling capabilities of these cilia has been linked to atypical neurite outgrowth from neurons and abnormalities in neuronal circuitry. Here, we tested the hypothesis that generalized seizures induced by pentylenetetrazol (PTZ) or kainic acid (KA) during early postnatal development impair neuronal and/or glial ciliogenesis. Mice received PTZ (50 or 100mg/kg), KA (2mg/kg), or saline either once at birth (P0), or once daily from P0 to P4. Using immunohistochemistry and electron microscopy, the cilia of neurons and glia were examined at P7, P14, and P42. A total of 83 regions were analyzed, representing 13 unique neocortical and hippocampal regions. Neuronal cilia were identified by co-expression of NeuN and type 3 adenylyl cyclase (ACIII) or somatostatin receptor 3 (SSTR3), while glial cilia were identified by co-expression of GFAP, Arl13b, and gamma-tubulin. We found that PTZ exposure at either P0 or from P0 to P4 induced convulsive behavior, followed by acute and lasting effects on neuronal cilia lengths that varied depending on the cortical region, PTZ dose, injection frequency, and time post-PTZ. Both increases and decreases in neuronal cilia length were observed. No changes in the length of glial cilia were observed under any of the test conditions. Lastly, we found that a single KA seizure at P0 led to similar abnormalities in neuronal cilia lengths. Our results suggest that seizure(s) occurring during early stages of cortical development induce persistent and widespread changes in neuronal cilia length. Given the impact neuronal cilia have on neuronal differentiation, ELS-induced changes in ciliogenesis may contribute to long-term pathology and abnormal cortical function.


Assuntos
Córtex Cerebral , Convulsivantes/toxicidade , Ácido Caínico/toxicidade , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Pentilenotetrazol/toxicidade , Convulsões/induzido quimicamente , Fatores Etários , Animais , Animais Recém-Nascidos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/crescimento & desenvolvimento , Córtex Cerebral/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Camundongos , Microscopia Imunoeletrônica , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/fisiologia , Neuroglia/ultraestrutura , Neurônios/metabolismo , Neurônios/ultraestrutura , Convulsões/patologia
18.
Oncotarget ; 7(6): 7029-43, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26760767

RESUMO

KIF3A, a component of the kinesin-2 motor, is necessary for the progression of diverse tumor types. This is partly due to its role in regulating ciliogenesis and cell responsiveness to sonic hedgehog (SHH). Notably, primary cilia have been detected in human glioblastoma multiforme (GBM) tumor biopsies and derived cell lines. Here, we asked whether disrupting KIF3A in GBM cells affected ciliogenesis, in vitro growth and responsiveness to SHH, or tumorigenic behavior in vivo. We used a lentiviral vector to create three patient-derived GBM cell lines expressing a dominant negative, motorless form of Kif3a (dnKif3a). In all unmodified lines, we found that most GBM cells were capable of producing ciliated progeny and that dnKif3a expression in these cells ablated ciliogenesis. Interestingly, unmodified and dnKif3a-expressing cell lines displayed differential sensitivities and pathway activation to SHH and variable tumor-associated survival following mouse xenografts. In one cell line, SHH-induced cell proliferation was prevented in vitro by either expressing dnKif3a or inhibiting SMO signaling using cyclopamine, and the survival times of mice implanted with dnKif3a-expressing cells were increased. In a second line, expression of dnKif3a increased the cells' baseline proliferation while, surprisingly, sensitizing them to SHH-induced cell death. The survival times of mice implanted with these dnKif3a-expressing cells were decreased. Finally, expression of dnKif3a in a third cell line had no effect on cell proliferation, SHH sensitivity, or mouse survival times. These findings indicate that KIF3A is essential for GBM cell ciliogenesis, but its role in modulating GBM cell behavior is highly variable.


Assuntos
Carcinogênese/patologia , Cílios/fisiologia , Genes Dominantes/genética , Glioblastoma/patologia , Proteínas Hedgehog/metabolismo , Cinesinas/antagonistas & inibidores , Adulto , Idoso , Animais , Apoptose , Western Blotting , Carcinogênese/genética , Carcinogênese/metabolismo , Proliferação de Células , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas Hedgehog/genética , Humanos , Técnicas Imunoenzimáticas , Cinesinas/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Pessoa de Meia-Idade , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Invest Ophthalmol Vis Sci ; 46(1): 12-6, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15623748

RESUMO

PURPOSE: Light-driven translocation of phototransduction regulatory proteins between the inner and outer segments of photoreceptor cells plays a role in the adaptation of these cells to light. The purpose of this study was to examine the effects of the absence of guanylate cyclase 1 (GC1) on light-driven protein translocation in rod and cone cells. Both cell types express GC1, but differ in sensitivity, saturation, and response times to light. METHODS: Immunohistochemical techniques employing antibodies specific for cone and rod transducin alpha (Talpha) subunits and arrestins were used to examine light-driven translocation of these proteins in the retinas of wild-type and GC1 knockout (KO) mice. RESULTS: Translocation of cone arrestin from cone outer segments to the inner cell regions was disrupted in the absence of GC1, whereas translocation of arrestin and Talpha in rods was not affected. Cone Talpha did not translocate in wild-type and GC1 KO mice, but differed in its subcellular distribution in GC1 KO retina, remaining in the cone outer segment in light and in dark. CONCLUSIONS: These results suggest that multiple, independent pathways regulate the translocation of phototransduction proteins and that GC1, and presumably cGMP, are of key importance in signaling the translocation of cone arrestin.


Assuntos
Arrestina/metabolismo , Deleção de Genes , Guanilato Ciclase/fisiologia , Receptores de Superfície Celular/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Transporte Biológico Ativo/efeitos da radiação , Adaptação à Escuridão , Técnica Indireta de Fluorescência para Anticorpo , Guanilato Ciclase/genética , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Transporte Proteico/efeitos da radiação , Receptores de Superfície Celular/genética , Células Fotorreceptoras Retinianas Cones/efeitos da radiação , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/efeitos da radiação , Transducina/metabolismo , Visão Ocular/fisiologia
20.
Brain Res Mol Brain Res ; 135(1-2): 30-9, 2005 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15857666

RESUMO

Chicken retina contains circadian oscillators that drive rhythmic transcription of several genes expressed in photoreceptor cells. To determine if gap junctions assist in coordinating these transcript rhythms, we examined the effects of two compounds, 18alpha-glycyrrhetnic acid-3-hemisuccinate (ACO) and 18beta-glycyrrhetnic acid (18beta-GA), on photoreceptor iodopsin and arylalkylamine N-acetyltransferase (AANAT) transcript rhythms in embryonic chicken retinal explant cultures that were maintained under different lighting conditions. Both compounds, whose actions include reversibly block gap junction permeability, produced rapid and sustained reductions in iodopsin and AANAT mRNA levels, but did not alter the levels of guanylate cyclase activating protein-1 (GCAP1) mRNA, a noncircadian-regulated, photoreceptor-specific gene. The iodopsin and AANAT mRNA rhythms re-emerged in the cultured retinas within 24 h of removal of the compounds. These results show that the effects of ACO and 18beta-GA on iodopsin and AANAT mRNA levels were not due to generalized suppression of gene transcription. The dramatic reduction in the levels of iodopsin and AANAT mRNA induced by these compounds suggests a mechanism of action that directly affects the synthesis and/or degradation of these transcripts rather than the synchronization or function of the retinal oscillators that drive transcription of these genes.


Assuntos
Carbenoxolona/farmacologia , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Ácido Glicirretínico/farmacologia , Células Fotorreceptoras/efeitos dos fármacos , Retina/citologia , Animais , Arilalquilamina N-Acetiltransferase/genética , Arilalquilamina N-Acetiltransferase/metabolismo , Embrião de Galinha , Regulação da Expressão Gênica/efeitos da radiação , Técnicas de Cultura de Órgãos , Fotoperíodo , Células Fotorreceptoras/efeitos da radiação , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Opsinas de Bastonetes/genética , Opsinas de Bastonetes/metabolismo , Fatores de Tempo , Transcrição Gênica/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa