Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Front Cell Neurosci ; 13: 55, 2019.
Article in English | MEDLINE | ID: mdl-30842728

ABSTRACT

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.

2.
Front Aging Neurosci ; 8: 127, 2016.
Article in English | MEDLINE | ID: mdl-27303293

ABSTRACT

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.

3.
Exp Neurol ; 282: 119-27, 2016 08.
Article in English | MEDLINE | ID: mdl-27181411

ABSTRACT

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.


Subject(s)
Cerebral Cortex , Convulsants/toxicity , Kainic Acid/toxicity , Neuroglia/drug effects , Neurons/drug effects , Pentylenetetrazole/toxicity , Seizures/chemically induced , Age Factors , Animals , Animals, Newborn , Cerebral Cortex/drug effects , Cerebral Cortex/growth & development , Cerebral Cortex/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Mice , Microscopy, Immunoelectron , Nerve Tissue Proteins/metabolism , Neuroglia/physiology , Neuroglia/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Seizures/pathology
4.
Oncotarget ; 7(6): 7029-43, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26760767

ABSTRACT

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.


Subject(s)
Carcinogenesis/pathology , Cilia/physiology , Genes, Dominant/genetics , Glioblastoma/pathology , Hedgehog Proteins/metabolism , Kinesins/antagonists & inhibitors , Adult , Aged , Animals , Apoptosis , Blotting, Western , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Proliferation , Glioblastoma/genetics , Glioblastoma/metabolism , Hedgehog Proteins/genetics , Humans , Immunoenzyme Techniques , Kinesins/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
J Neurooncol ; 117(1): 15-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24510433

ABSTRACT

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.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/ultrastructure , Cilia/ultrastructure , Glioblastoma/metabolism , Glioblastoma/ultrastructure , ADP-Ribosylation Factors/metabolism , Aged, 80 and over , Axoneme/metabolism , Axoneme/ultrastructure , Basal Bodies/metabolism , Basal Bodies/ultrastructure , Cell Line, Tumor , Cilia/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Microscopy, Electron , Middle Aged , Transcription Factors/metabolism , Tubulin/metabolism , Tumor Suppressor Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1
6.
Methods ; 66(3): 466-73, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-23816789

ABSTRACT

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.


Subject(s)
Embryonic Development/genetics , Genetic Engineering/methods , Genetic Vectors , Lentivirus/genetics , Transgenes , Animals , Animals, Genetically Modified/genetics , Chick Embryo , Retina/embryology
7.
J Neurosci ; 33(6): 2626-38, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392690

ABSTRACT

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.


Subject(s)
Adenylyl Cyclases/physiology , Cilia/enzymology , Dendrites/enzymology , Neocortex/enzymology , Neurons/enzymology , Receptors, Serotonin/biosynthesis , Animals , Cells, Cultured , Cilia/physiology , Female , Kinesins/biosynthesis , Male , Mice , NIH 3T3 Cells , Neocortex/embryology , Neurogenesis/physiology , Neurons/cytology , Pregnancy
8.
J Neurosci ; 33(6): 2671-83, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23392694

ABSTRACT

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.


Subject(s)
Aging/physiology , Estrogen Receptor alpha/physiology , Estrogen Receptor beta/physiology , Hippocampus/physiology , Animals , Female , Maze Learning/physiology , Mice , Mice, Knockout
9.
PLoS One ; 8(12): e83378, 2013.
Article in English | MEDLINE | ID: mdl-24386190

ABSTRACT

Many aspects of retinal photoreceptor function and physiology are regulated by the circadian clocks in these cells. It is well established that light is the primary stimulus that entrains these clocks; yet, the biochemical cascade(s) mediating light's effects on these clocks remains unknown. This deficiency represents a significant gap in our fundamental understanding of photoreceptor signaling cascades and their functions. In this study, we utilized re-aggregated spheroid cultures prepared from embryonic chick retina to determine if activation of phospholipase C in photoreceptors in the absence of light can phase shift the melatonin secretion rhythms of these cells in a manner similar to that induced by light. We show that spheroid cultures rhythmically secrete melatonin and that these melatonin rhythms can be dynamically phase shifted by exposing the cultures to an appropriately timed light pulse. Importantly, we show that activation of phospholipase C using m-3M3FBS in the absence of light induces a phase delay in photoreceptor melatonin rhythms that mirrors that induced by light. The implication of this finding is that the light signaling cascade that entrains photoreceptor melatonin rhythms involves activation of phospholipase C.


Subject(s)
Circadian Rhythm/physiology , Light , Melatonin/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Type C Phospholipases/metabolism , Animals , Chick Embryo , Enzyme Activation , Tissue Culture Techniques
10.
PLoS One ; 7(12): e51385, 2012.
Article in English | MEDLINE | ID: mdl-23240018

ABSTRACT

We previously demonstrated that aged ovariectomized rats that had received prior estradiol treatment in middle-age exhibited increased levels of estrogen receptor alpha (ERα) in the hippocampus as well as enhanced hippocampal dependent memory as compared to aged rats that had not received mid-life estradiol treatment. These effects persisted long after the estradiol treatment had been terminated. The goal of the current experiment was to determine if increased expression of ERα in the hippocampus, in the absence of exogenously administered estrogens, can impact the hippocampus and cognitive function in aging ovariectomized rats. Middle-aged rats were trained for 24 days on an eight-arm radial maze spatial memory task. All rats were then ovariectomized. Forty days later, rats received either lentiviral delivery to the hippocampus of the gene encoding ERα (lenti-ERα) or a control virus. Rats were tested on delay trials in the radial-maze in which delays of varying lengths were imposed between the fourth and fifth arm choices. Following behavior testing, hippocampi were immunostained using western blotting for ERα, the ERα-regulated protein choline acetyltransferase, and phosphorylation of the ERα-regulated kinases, ERK/MAPK and Akt. Results revealed that aging ovariectomized rats that received delivery of lenti-ERα to the hippocampus exhibited enhanced spatial memory as indicated by increased arm-choice accuracy across delays as compared to ovariectomized rats that received control virus. Western blot data revealed that lenti-ERα delivery significantly increased levels of ERα and phosphorylated ERK/MAPK and had no impact on levels of ChAT or phosphorylation of Akt. Results indicate that increasing hippocampal levels of ERα in aging females in the absence of ovarian or exogenously administered estrogens leads to increases in phosphorylation of ERK/MAPK as well as in enhanced memory.


Subject(s)
Aging , Estrogen Receptor alpha , Memory , Aging/genetics , Aging/metabolism , Animals , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Genetic Vectors , Hippocampus/metabolism , Maze Learning , Memory/drug effects , Memory/physiology , Mitogen-Activated Protein Kinases/metabolism , Ovariectomy , Rats
11.
PLoS One ; 7(4): e34097, 2012.
Article in English | MEDLINE | ID: mdl-22496779

ABSTRACT

Transplantation of neural stems cells (NSCs) could be a useful means to deliver biologic therapeutics for late-stage Alzheimer's disease (AD). In this study, we conducted a small preclinical investigation of whether NSCs could be modified to express metalloproteinase 9 (MMP9), a secreted protease reported to degrade aggregated Aß peptides that are the major constituents of the senile plaques. Our findings illuminated three issues with using NSCs as delivery vehicles for this particular application. First, transplanted NSCs generally failed to migrate to amyloid plaques, instead tending to colonize white matter tracts. Second, the final destination of these cells was highly influenced by how they were delivered. We found that our injection methods led to cells largely distributing to white matter tracts, which are anisotropic conduits for fluids that facilitate rapid distribution within the CNS. Third, with regard to MMP9 as a therapeutic to remove senile plaques, we observed high concentrations of endogenous metalloproteinases around amyloid plaques in the mouse models used for these preclinical tests with no evidence that the NSC-delivered enzymes elevated these activities or had any impact. Interestingly, MMP9-expressing NSCs formed substantially larger grafts. Overall, we observed long-term survival of NSCs in the brains of mice with high amyloid burden. Therefore, we conclude that such cells may have potential in therapeutic applications in AD but improved targeting of these cells to disease-specific lesions may be required to enhance efficacy.


Subject(s)
Amyloidosis/prevention & control , Brain/pathology , Disease Models, Animal , Matrix Metalloproteinase 9/metabolism , Nerve Fibers, Myelinated/pathology , Neural Stem Cells/transplantation , Plaque, Amyloid/prevention & control , Amyloid/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Amyloidosis/enzymology , Amyloidosis/pathology , Animals , Brain/metabolism , Cells, Cultured , Immunoenzyme Techniques , Lentivirus/genetics , Mice , Nerve Fibers, Myelinated/metabolism , Neural Stem Cells/cytology , Plaque, Amyloid/enzymology , Plaque, Amyloid/pathology
12.
PLoS One ; 6(5): e20553, 2011.
Article in English | MEDLINE | ID: mdl-21647387

ABSTRACT

The disease processes underlying inherited retinal disease are complex and are not completely understood. Many of the corrective gene therapies designed to treat diseases linked to mutations in genes specifically expressed in photoreceptor cells restore function to these cells but fail to stop progression of the disease. There is growing consensus that effective treatments for these diseases will require delivery of multiple therapeutic proteins that will be selected to treat specific aspects of the disease process. The purpose of this study was to design a lentiviral transgene that reliably expresses all of the proteins it encodes and does so in a consistent manner among infected cells. We show, using both in vitro and in vivo analyses, that bicistronic lentiviral transgenes encoding two fluorescent proteins fused to a viral 2A-like cleavage peptide meet these expression criteria. To determine if this transgene design is suitable for therapeutic applications, we replaced one of the fluorescent protein genes with the gene encoding guanylate cyclase-1 (GC1) and delivered lentivirus carrying this transgene to the retinas of the GUCY1*B avian model of Leber congenital amaurosis-1 (LCA1). GUCY1*B chickens carry a null mutation in the GC1 gene that disrupts photoreceptor function and causes blindness at hatching, a phenotype that closely matches that observed in humans with LCA1. We found that treatment of these animals with the 2A lentivector encoding GC1 restored vision to these animals as evidenced by the presence of optokinetic reflexes. We conclude that 2A-like peptides, with proper optimization, can be successfully incorporated into therapeutic vectors designed to deliver multiple proteins to neural retinal. These results highlight the potential of this vector design to serve as a platform for the development of combination therapies designed to enhance or prolong the benefits of corrective gene therapies.


Subject(s)
Genes, Viral/genetics , Genetic Vectors/genetics , Leber Congenital Amaurosis/physiopathology , Lentivirus/genetics , Luminescent Proteins/genetics , Recovery of Function/genetics , Vision, Ocular/physiology , Animals , Chick Embryo , Disease Models, Animal , Gene Expression , Guanylate Cyclase/genetics , HEK293 Cells , Humans , Leber Congenital Amaurosis/genetics , Leber Congenital Amaurosis/therapy , Promoter Regions, Genetic/genetics , Receptors, Cell Surface/genetics , Retina/metabolism , Retina/physiopathology , Transfection , Transgenes/genetics
13.
Cereb Cortex ; 21(2): 338-44, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20525772

ABSTRACT

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.


Subject(s)
Cilia/physiology , Cytokinesis/genetics , Intracellular Signaling Peptides and Proteins/deficiency , Neurons/cytology , Protein Serine-Threonine Kinases/deficiency , Somatosensory Cortex/cytology , Stem Cells/metabolism , Animals , Animals, Genetically Modified , Animals, Newborn , Cilia/ultrastructure , Hippocampus/cytology , Microscopy, Confocal , Microscopy, Electron/methods , Models, Biological , Mutation/genetics , Neurons/metabolism , Neurons/ultrastructure , Phosphopyruvate Hydratase/metabolism , Protein Transport/genetics , Rats , Rats, Wistar , Receptors, Somatostatin/metabolism , Stem Cells/ultrastructure
14.
J Neurochem ; 115(4): 941-55, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20819118

ABSTRACT

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.


Subject(s)
Hippocampus/physiology , Neurons/physiology , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/genetics , Protein Kinase C-alpha/physiology , Proto-Oncogene Proteins c-akt/physiology , RNA Splicing , Signal Transduction/genetics , Aging/genetics , Aging/physiology , Animals , Cell Line , Cells, Cultured , Female , Hippocampus/embryology , Hippocampus/enzymology , Humans , Isoenzymes/genetics , Isoenzymes/physiology , Male , Neurons/enzymology , Neurons/metabolism , Nuclear Proteins/physiology , Phosphoprotein Phosphatases/physiology , Protein Kinase C-alpha/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , RNA Splicing/physiology , Rats , Rats, Inbred BN , Rats, Inbred F344
15.
J Neurosci Res ; 88(12): 2558-68, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20648646

ABSTRACT

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.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/genetics , Myelin Sheath/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Schwann Cells/metabolism , Animals , Animals, Newborn , Cells, Cultured , Coculture Techniques , Down-Regulation/physiology , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Myelin Sheath/physiology , RNA Interference/physiology , Rats , Schwann Cells/cytology , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Stem Cells/metabolism , Stem Cells/physiology , Up-Regulation/physiology
16.
Mol Vis ; 16: 916-34, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20517486

ABSTRACT

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.


Subject(s)
Gene Targeting/methods , Genetic Vectors , Lentivirus/genetics , Photoreceptor Cells, Vertebrate , Promoter Regions, Genetic , Retina/cytology , Animals , Chick Embryo , Gene Expression , Mice/genetics , Photoreceptor Cells, Vertebrate/metabolism , Retina/embryology , Retina/metabolism , Transgenes , Xenopus/genetics
17.
Biochem Biophys Res Commun ; 390(4): 1149-53, 2009 Dec 25.
Article in English | MEDLINE | ID: mdl-19878658

ABSTRACT

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.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 6/metabolism , Light , Protein Processing, Post-Translational , Retinal Cone Photoreceptor Cells/radiation effects , Retinal Rod Photoreceptor Cells/radiation effects , Animals , Antibodies, Phospho-Specific/immunology , Cattle , GTP-Binding Protein alpha Subunits/metabolism , Light Signal Transduction , Mice , Mice, Inbred Strains , Mice, Transgenic , Phosphorylation , Retinal Cone Photoreceptor Cells/enzymology , Retinal Rod Photoreceptor Cells/enzymology , Signal Transduction , Threonine/immunology , Threonine/metabolism , Transducin/metabolism
18.
Mol Ther ; 16(9): 1587-93, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18594506

ABSTRACT

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.


Subject(s)
Estrogen Receptor alpha/genetics , Gene Transfer Techniques , Genetic Vectors , Hippocampus/metabolism , Maze Learning , Space Perception , Animals , Benzoates/pharmacology , Brain/physiology , COS Cells , Cells, Cultured , Chlorocebus aethiops , Electrophysiology , Estradiol/pharmacology , Female , Green Fluorescent Proteins/genetics , Immunoenzyme Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Response Elements/physiology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/therapy , Synaptic Transmission/physiology
19.
Mol Ther ; 16(9): 1587-1593, 2008 Sep.
Article in English | MEDLINE | ID: mdl-28189013

ABSTRACT

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.

20.
Mol Vis ; 13: 2001-11, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17982424

ABSTRACT

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.


Subject(s)
Gene Targeting/methods , Genetic Vectors , Lentivirus/genetics , Promoter Regions, Genetic , Retina/metabolism , Transgenes , Animals , Chick Embryo , Eye Proteins/genetics , Gene Expression , Green Fluorescent Proteins/genetics , Guanylate Cyclase-Activating Proteins/genetics , Lentivirus/physiology , Luminescent Proteins/genetics , Mice , Peptide Elongation Factor 1/genetics , Retina/embryology , Retinol-Binding Proteins/genetics , Rhodopsin/genetics , Tissue Distribution , Virus Inactivation
SELECTION OF CITATIONS
SEARCH DETAIL
...