Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
BMC Biol ; 22(1): 2, 2024 01 02.
Article in English | MEDLINE | ID: mdl-38167120

ABSTRACT

BACKGROUND: Dysregulation of nucleocytoplasmic shuttling of histone deacetylase 4 (HDAC4) is associated with several neurodevelopmental and neurodegenerative disorders. Consequently, understanding the roles of nuclear and cytoplasmic HDAC4 along with the mechanisms that regulate nuclear entry and exit is an area of concerted effort. Efficient nuclear entry is dependent on binding of the transcription factor MEF2, as mutations in the MEF2 binding region result in cytoplasmic accumulation of HDAC4. It is well established that nuclear exit and cytoplasmic retention are dependent on 14-3-3-binding, and mutations that affect binding are widely used to induce nuclear accumulation of HDAC4. While regulation of HDAC4 shuttling is clearly important, there is a gap in understanding of how the nuclear and cytoplasmic distribution of HDAC4 impacts its function. Furthermore, it is unclear whether other features of the protein including the catalytic site, the MEF2-binding region and/or the ankyrin repeat binding motif influence the distribution and/or activity of HDAC4 in neurons. Since HDAC4 functions are conserved in Drosophila, and increased nuclear accumulation of HDAC4 also results in impaired neurodevelopment, we used Drosophila as a genetic model for investigation of HDAC4 function. RESULTS: Here we have generated a series of mutants for functional dissection of HDAC4 via in-depth examination of the resulting subcellular distribution and nuclear aggregation, and correlate these with developmental phenotypes resulting from their expression in well-established models of neuronal morphogenesis of the Drosophila mushroom body and eye. We found that in the mushroom body, forced sequestration of HDAC4 in the nucleus or the cytoplasm resulted in defects in axon morphogenesis. The actions of HDAC4 that resulted in impaired development were dependent on the MEF2 binding region, modulated by the ankyrin repeat binding motif, and largely independent of an intact catalytic site. In contrast, disruption to eye development was largely independent of MEF2 binding but mutation of the catalytic site significantly reduced the phenotype, indicating that HDAC4 acts in a neuronal-subtype-specific manner. CONCLUSIONS: We found that the impairments to mushroom body and eye development resulting from nuclear accumulation of HDAC4 were exacerbated by mutation of the ankyrin repeat binding motif, whereas there was a differing requirement for the MEF2 binding site and an intact catalytic site. It will be of importance to determine the binding partners of HDAC4 in nuclear aggregates and in the cytoplasm of these tissues to further understand its mechanisms of action.


Subject(s)
Ankyrin Repeat , Drosophila , Histone Deacetylases , Animals , Catalytic Domain , Cell Nucleus/metabolism , Drosophila/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , MEF2 Transcription Factors/genetics , MEF2 Transcription Factors/metabolism , Morphogenesis , Neurons/metabolism
2.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-37396791

ABSTRACT

Spatial and temporal control of gene expression in Drosophila is essential in elucidating gene function. Spatial control is facilitated by the UAS/GAL4 system, and this can be coupled with additional adaptations for precise temporal control and fine tuning of gene expression levels. Here we directly compare the level of pan-neuronal transgene expression governed by nSyb-GAL4 and elav-GAL4, as well as mushroom body-specific expression alongside OK107-GAL4. We also compare the temporal modulation of gene expression in neurons with the auxin-inducible gene expression system (AGES) and temporal and regional gene expression targeting (TARGET) systems.

3.
Mol Brain ; 16(1): 42, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37194019

ABSTRACT

Dysregulation of HDAC4 expression and/or nucleocytoplasmic shuttling results in impaired neuronal morphogenesis and long-term memory in Drosophila melanogaster. A recent genetic screen for genes that interact in the same molecular pathway as HDAC4 identified the cytoskeletal adapter Ankyrin2 (Ank2). Here we sought to investigate the role of Ank2 in neuronal morphogenesis, learning and memory. We found that Ank2 is expressed widely throughout the Drosophila brain where it localizes predominantly to axon tracts. Pan-neuronal knockdown of Ank2 in the mushroom body, a region critical for memory formation, resulted in defects in axon morphogenesis. Similarly, reduction of Ank2 in lobular plate tangential neurons of the optic lobe disrupted dendritic branching and arborization. Conditional knockdown of Ank2 in the mushroom body of adult Drosophila significantly impaired long-term memory (LTM) of courtship suppression, and its expression was essential in the γ neurons of the mushroom body for normal LTM. In summary, we provide the first characterization of the expression pattern of Ank2 in the adult Drosophila brain and demonstrate that Ank2 is critical for morphogenesis of the mushroom body and for the molecular processes required in the adult brain for the formation of long-term memories.


Subject(s)
Ankyrins , Drosophila Proteins , Drosophila melanogaster , Animals , Ankyrins/metabolism , Courtship , Drosophila melanogaster/metabolism , Drosophila Proteins/metabolism , Memory, Long-Term/physiology , Morphogenesis , Mushroom Bodies/metabolism , Neurons/metabolism
4.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-36798589

ABSTRACT

Herzog (Hzg, CG5830) shares similarity to members of the haloacid dehalogenase subfamily of small CTD phosphatases. In Drosophila it is a maternal gene essential for establishment of embryonic segment polarity, and oligomerization is required for activation of phosphatase activity. While Hzg is expressed in the brain, its role has not been investigated. To that end, we further characterized Hzg expression in the brain and found that it is highly expressed in neurons of the mushroom body where it localises to axons, and is also expressed in cortical glia. We investigated its role in mushroom body development as well as courtship learning and memory, but found that knockdown of Hzg had no impact on these processes. In contrast, knockdown in post-mitotic neurons in the eye resulted in disruption to ommatidial patterning and pigmentation, indicating it plays an important role in eye development.

5.
Mol Brain ; 15(1): 15, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35139868

ABSTRACT

Coenzyme Q8A encodes the homologue of yeast coq8, an ATPase that is required for the biosynthesis of Coenzyme Q10, an essential component of the electron transport chain. Mutations in COQ8A in humans result in CoQ10 deficiency, the clinical features of which include early-onset cerebellar ataxia, seizures and intellectual disability. The rapid advancement of massively parallel sequencing has resulted in the identification of more than 40 new mutations in COQ8A and functional studies are required to confirm causality and to further research into determining the specific mechanisms through which the mutations result in loss of function. To that end, a Drosophila model of Coq8 deficiency was developed and characterized to determine its appropriateness as a model system to further explore the role of Coq8 in the brain, and for functional characterisation of Coq8 mutations. Pan-neuronal RNAi knockdown of Coq8 was largely lethal, with female escapers displaying severe locomotor deficits. Knockdown of Coq8 in the eye resulted in degeneration of photoreceptors, progressive necrosis and increased generation of reactive oxygen species. Reintroduction of wild-type Coq8 restored normal function, however expression of human wild-type COQ8A exacerbated the eye phenotype, suggesting it was acting as a dominant-negative. This model is therefore informative for investigating the function of Drosophila Coq8, however human COQ8A mutations cannot be assessed as hCOQ8A does not rescue Coq8 deficiency.


Subject(s)
Mitochondrial Diseases , Saccharomyces cerevisiae Proteins , Animals , Ataxia/genetics , Drosophila/metabolism , Female , Mitochondrial Diseases/genetics , Muscle Weakness , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
6.
Front Mol Neurosci ; 14: 616642, 2021.
Article in English | MEDLINE | ID: mdl-33859551

ABSTRACT

Dysregulation of the histone deacetylase HDAC4 is associated with both neurodevelopmental and neurodegenerative disorders, and a feature common to many of these disorders is impaired cognitive function. HDAC4 shuttles between the nucleus and cytoplasm in both vertebrates and invertebrates and alterations in the amounts of nuclear and/or cytoplasmic HDAC4 have been implicated in these diseases. In Drosophila, HDAC4 also plays a critical role in the regulation of memory, however, the mechanisms through which it acts are unknown. Nuclear and cytoplasmically-restricted HDAC4 mutants were expressed in the Drosophila brain to investigate a mechanistic link between HDAC4 subcellular distribution, transcriptional changes and neuronal dysfunction. Deficits in mushroom body morphogenesis, eye development and long-term memory correlated with increased abundance of nuclear HDAC4 but were associated with minimal transcriptional changes. Although HDAC4 sequesters MEF2 into punctate foci within neuronal nuclei, no alteration in MEF2 activity was observed on overexpression of HDAC4, and knockdown of MEF2 had no impact on long-term memory, indicating that HDAC4 is likely not acting through MEF2. In support of this, mutation of the MEF2 binding site within HDAC4 also had no impact on nuclear HDAC4-induced impairments in long-term memory or eye development. In contrast, the defects in mushroom body morphogenesis were ameliorated by mutation of the MEF2 binding site, as well as by co-expression of MEF2 RNAi, thus nuclear HDAC4 acts through MEF2 to disrupt mushroom body development. These data provide insight into the mechanisms through which dysregulation of HDAC4 subcellular distribution impairs neurological function and provides new avenues for further investigation.

7.
Mol Brain ; 10(1): 41, 2017 08 29.
Article in English | MEDLINE | ID: mdl-28851405

ABSTRACT

Moesin is a cytoskeletal adaptor protein that plays an important role in modification of the actin cytoskeleton. Rearrangement of the actin cytoskeleton drives both neuronal morphogenesis and the structural changes in neurons that are required for long-term memory formation. Moesin has been identified as a candidate memory gene in Drosophila, however, whether it is required for memory formation has not been evaluated. Here, we investigate the role of Moesin in neuronal morphogenesis and in short- and long-term memory formation in the courtship suppression assay, a model of associative memory. We found that both knockdown and overexpression of Moesin led to defects in axon growth and guidance as well as dendritic arborization. Moreover, reduction of Moesin expression or expression of a constitutively active phosphomimetic in the adult Drosophila brain had no effect on short term memory, but prevented long-term memory formation, an effect that was independent of its role in development. These results indicate a critical role for Moesin in both neuronal morphogenesis and long-term memory formation.


Subject(s)
Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Memory, Long-Term , Morphogenesis , Neurons/metabolism , Animals , Brain/cytology , Brain/metabolism , Dendrites/metabolism , Drosophila melanogaster/cytology , Gene Knockdown Techniques , Mushroom Bodies/cytology , Mushroom Bodies/metabolism , Neurons/cytology , Protein Transport , Subcellular Fractions/metabolism
8.
Mol Microbiol ; 103(4): 657-677, 2017 02.
Article in English | MEDLINE | ID: mdl-27882646

ABSTRACT

Cell-cell fusion in fungi is required for colony formation, nutrient transfer and signal transduction. Disruption of genes required for hyphal fusion in Epichloë festucae, a mutualistic symbiont of Lolium grasses, severely disrupts the host interaction phenotype. They examined whether symB and symC, the E. festucae homologs of Podospora anserina self-signaling genes IDC2 and IDC3, are required for E. festucae hyphal fusion and host symbiosis. Deletion mutants of these genes were defective in hyphal cell fusion, formed intra-hyphal hyphae, and had enhanced conidiation. SymB-GFP and SymC-mRFP1 localize to plasma membrane, septa and points of hyphal cell fusion. Plants infected with ΔsymB and ΔsymC strains were severely stunted. Hyphae of the mutants colonized vascular bundles, were more abundant than wild type in the intercellular spaces and formed intra-hyphal hyphae. Although these phenotypes are identical to those previously observed for cell wall integrity MAP kinase mutants no difference was observed in the basal level of MpkA phosphorylation or its cellular localization in the mutant backgrounds. Both genes contain binding sites for the transcription factor ProA. Collectively these results show that SymB and SymC are key components of a conserved signaling network for E. festucae to maintain a mutualistic symbiotic interaction within L. perenne.


Subject(s)
Epichloe/genetics , Fungal Proteins/genetics , Hyphae/genetics , Lolium/growth & development , Membrane Proteins/genetics , Spores, Fungal/growth & development , Symbiosis/genetics , Cell Fusion , Epichloe/physiology , Fungal Proteins/metabolism , Gene Expression Regulation, Fungal , Hyphae/physiology , Lolium/microbiology , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Sequence Deletion/genetics , Spores, Fungal/genetics , Transcription Factors/metabolism
9.
Mol Plant Pathol ; 17(9): 1480-1492, 2016 12.
Article in English | MEDLINE | ID: mdl-27277141

ABSTRACT

In both Sordaria macrospora and Neurospora crassa, components of the conserved STRIPAK (striatin-interacting phosphatase and kinase) complex regulate cell-cell fusion, hyphal network development and fruiting body formation. Interestingly, a number of Epichloë festucae genes that are required for hyphal cell-cell fusion, such as noxA, noxR, proA, mpkA and mkkA, are also required for the establishment of a mutualistic symbiotic interaction with Lolium perenne. To determine whether MobC, a homologue of the STRIPAK complex component MOB3 in S. macrospora and N. crassa, is required for E. festucae hyphal fusion and symbiosis, a mobC deletion strain was generated. The ΔmobC mutant showed reduced rates of hyphal cell-cell fusion, formed intrahyphal hyphae and exhibited enhanced conidiation. Plants infected with ΔmobC were severely stunted. Hyphae of ΔmobC showed a proliferative pattern of growth within the leaves of Lolium perenne with increased colonization of the intercellular spaces and vascular bundles. Although hyphae were still able to form expressoria, structures allowing the colonization of the leaf surface, the frequency of formation was significantly reduced. Collectively, these results show that the STRIPAK component MobC is required for the establishment of a mutualistic symbiotic association between E. festucae and L. perenne, and plays an accessory role in the regulation of hyphal cell-cell fusion and expressorium development in E. festucae.


Subject(s)
Epichloe/metabolism , Fungal Proteins/metabolism , Lolium/microbiology , Multiprotein Complexes/metabolism , Sequence Homology, Amino Acid , Symbiosis/physiology , Cell Fusion , Epichloe/cytology , Host-Pathogen Interactions , Hyphae/cytology , Lolium/ultrastructure , Mutation/genetics , Phenotype , Plant Stems/ultrastructure
10.
Genetics ; 203(3): 1249-64, 2016 07.
Article in English | MEDLINE | ID: mdl-27182943

ABSTRACT

HDAC4 is a potent memory repressor with overexpression of wild type or a nuclear-restricted mutant resulting in memory deficits. Interestingly, reduction of HDAC4 also impairs memory via an as yet unknown mechanism. Although histone deacetylase family members are important mediators of epigenetic mechanisms in neurons, HDAC4 is predominantly cytoplasmic in the brain and there is increasing evidence for interactions with nonhistone proteins, suggesting HDAC4 has roles beyond transcriptional regulation. To that end, we performed a genetic interaction screen in Drosophila and identified 26 genes that interacted with HDAC4, including Ubc9, the sole SUMO E2-conjugating enzyme. RNA interference-induced reduction of Ubc9 in the adult brain impaired long-term memory in the courtship suppression assay, a Drosophila model of associative memory. We also demonstrate that HDAC4 and Ubc9 interact genetically during memory formation, opening new avenues for investigating the mechanisms through which HDAC4 regulates memory formation and other neurological processes.


Subject(s)
Brain/metabolism , Drosophila Proteins/genetics , Histone Deacetylases/genetics , Memory, Long-Term , Ubiquitin-Conjugating Enzymes/genetics , Animals , Brain/growth & development , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental/genetics , Histone Deacetylases/metabolism , RNA Interference , Sumoylation/genetics , Ubiquitin-Conjugating Enzymes/metabolism
11.
BMC Biotechnol ; 16: 30, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-27006073

ABSTRACT

BACKGROUND: Diabetes and its concurrent complications impact a significant proportion of the population of the US and create a large financial burden on the American health care system. FDA-approved maggot debridement therapy (MDT), the application of sterile laboratory-reared Lucilia sericata (green bottle fly) larvae to wounds, is a cost-effective and successful treatment for diabetic foot ulcers and other medical conditions. Human platelet derived growth factor-BB (PDGF-BB) is a secreted dimeric peptide growth factor that binds the PDGF receptor. PDGF-BB stimulates cell proliferation and survival, promotes wound healing, and has been investigated as a possible topical treatment for non-healing wounds. Genetic engineering has allowed for expression and secretion of human growth factors and other proteins in transgenic insects. Here, we present a novel concept in MDT technology that combines the established benefits of MDT with the power of genetic engineering to promote healing. The focus of this study is to create and characterize strains of transgenic L. sericata that express and secrete PDGF-BB at detectable levels in adult hemolymph, whole larval lysate, and maggot excretions/ secretions (ES), with potential for clinical utility in wound healing. RESULTS: We have engineered and confirmed transgene insertion in several strains of L. sericata that express human PDGF-BB. Using a heat-inducible promoter to control the pdgf-b gene, pdgf-b mRNA was detected via semi-quantitative PCR upon heat shock. PDGF-BB protein was also detectable in larval lysates and adult hemolymph but not larval ES. An alternative, tetracycline-repressible pdgf-b system mediated expression of pdgf-b mRNA when maggots were raised on diet that lacked tetracycline. Further, PDGF-BB protein was readily detected in whole larval lysate as well as larval ES. CONCLUSIONS: Here we show robust, inducible expression and production of human PDGF-BB protein from two conditional expression systems in transgenic L. sericata larvae. The tetracycline-repressible system appears to be the most promising as PDGF-BB protein was detectable in larval ES following induction. Our system could potentially be used to deliver a variety of growth factors and anti-microbial peptides to the wound environment with the aim of enhancing wound healing, thereby improving patient outcome in a cost-effective manner.


Subject(s)
Animals, Genetically Modified/genetics , Debridement/methods , Diptera/genetics , Larva , Platelet-Derived Growth Factor/metabolism , Recombinant Proteins/metabolism , Animals , Animals, Genetically Modified/metabolism , Diabetic Foot , Diptera/metabolism , Female , Gene Expression/drug effects , Humans , Male , Platelet-Derived Growth Factor/genetics , Recombinant Proteins/genetics , Tetracycline/pharmacology , Wound Healing
12.
Neurobiol Learn Mem ; 123: 149-58, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26074448

ABSTRACT

Histone deacetylase (HDAC) family members are important mediators of epigenetic mechanisms in the regulation of memory formation. Recent studies have revealed that Class IIa HDAC family member HDAC4 contributes essential functions to synaptic plasticity and memory as well as other neurological processes. HDAC4 is localized to both the nucleus and cytoplasm and undergoes subcellular shuttling in response to synaptic activity. In the nucleus, HDAC4 is a repressor of transcriptional programmes, although vertebrate HDAC4 is itself catalytically inactive and repression is facilitated through direct inhibition of transcription factors such as MEF2. In the brain, however, HDAC4 is predominantly cytoplasmic, and a pool of HDAC4 is concentrated in dendritic spines. This review explores and synthesizes the evidence that specific subcellular pools of HDAC4 mediate differential effects on neurological function. Nuclear-restriction of HDAC4 results in down-regulation of plasticity-related genes and memory impairment. However loss of HDAC4 also results in memory deficits through unknown mechanisms. The localization to dendritic shafts and spines, combined with the evidence that cytoplasmic HDAC4 is neuroprotective in some models of neurological dysfunction point to an essential cytoplasmic role. Investigation of extra-nuclear roles of HDAC4, including identification of cytoplasmic binding partners is paramount in order to understand and therefore manipulate its function for potential therapeutic benefit.


Subject(s)
Brain/metabolism , Cytoplasm/metabolism , Histone Deacetylases/physiology , Memory/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Repressor Proteins/physiology , Animals , Histone Deacetylases/metabolism , Humans , Neuronal Plasticity/genetics , Repressor Proteins/metabolism
13.
PLoS One ; 8(12): e83903, 2013.
Article in English | MEDLINE | ID: mdl-24349558

ABSTRACT

A growing body of research indicates that pharmacological inhibition of histone deacetylases (HDACs) correlates with enhancement of long-term memory and current research is concentrated on determining the roles that individual HDACs play in cognitive function. Here, we investigate the role of HDAC4 in long-term memory formation in Drosophila. We show that overexpression of HDAC4 in the adult mushroom body, an important structure for memory formation, resulted in a specific impairment in long-term courtship memory, but had no affect on short-term memory. Overexpression of an HDAC4 catalytic mutant also abolished LTM, suggesting a mode of action independent of catalytic activity. We found that overexpression of HDAC4 resulted in a redistribution of the transcription factor MEF2 from a relatively uniform distribution through the nucleus into punctate nuclear bodies, where it colocalized with HDAC4. As MEF2 has also been implicated in regulation of long-term memory, these data suggest that the repressive effects of HDAC4 on long-term memory may be through interaction with MEF2. In the same genetic background, we also found that RNAi-mediated knockdown of HDAC4 impairs long-term memory, therefore we demonstrate that HDAC4 is not only a repressor of long-term memory, but also modulates normal memory formation.


Subject(s)
Drosophila Proteins/biosynthesis , Gene Expression Regulation, Enzymologic/physiology , Histone Deacetylases/biosynthesis , Memory, Long-Term/physiology , Mushroom Bodies/enzymology , Animals , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Gene Knockdown Techniques , Histone Deacetylases/genetics , Mushroom Bodies/cytology , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism
14.
PLoS One ; 6(12): e29171, 2011.
Article in English | MEDLINE | ID: mdl-22195015

ABSTRACT

There is increasing evidence that regulation of local chromatin structure is a critical mechanism underlying the consolidation of long-term memory (LTM), however considerably less is understood about the specific mechanisms by which these epigenetic effects are mediated. Furthermore, the importance of histone acetylation in Drosophila memory has not been reported. The histone deacetylase (HDAC) Rpd3 is abundant in the adult fly brain, suggesting a post-mitotic function. Here, we investigated the role of Rpd3 in long-term courtship memory in Drosophila. We found that while modulation of Rpd3 levels predominantly in the adult mushroom body had no observed impact on immediate recall or one-hour memory, 24-hour LTM was severely impaired. Surprisingly, both overexpression as well as RNAi-mediated knockdown of Rpd3 resulted in impairment of long-term courtship memory, suggesting that the dose of Rpd3 is critical for normal LTM.


Subject(s)
Courtship , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Histone Deacetylase 1/genetics , Memory, Long-Term/physiology , Animals , Brain/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Gene Knockdown Techniques , Histone Deacetylase 1/metabolism , Histone Deacetylases/metabolism , Phenotype , RNA Interference , Survival Analysis , Tubulin/metabolism
15.
J Gene Med ; 12(4): 385-98, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20352617

ABSTRACT

BACKGROUND: The steps necessary to translate promising new biological therapies to the clinic are poorly documented. For gene therapy, there are unique aspects that need to be addressed in biodistribution studies. Notably, the spread of the vector beyond the intended target cells or tissue may result in persistent unwanted biological activity or unpredictable biological events; thus, it is critical to evaluate the risks associated with viral vector-mediated gene transfer prior to embarking on human clinical trials. METHODS: In the present study, we conducted a comprehensive assessment of vector biodistribution throughout the brain, blood and major organs of rats that had been injected via the subthalamic nucleus with recombinant adeno-associated virus (AAV) expressing glutamic acid decarboxylase (GAD). In addition, behavioral and histological analyses were also performed. RESULTS: AAV genomes were not detected in blood or cerebrospinal fluid, and did not disseminate to organs outside of the brain in the majority of animals. In the brain, an average of 97.3% of AAV2-GAD genomes were restricted to the area of the ipsilateral subthalamic nucleus (STN). There were no discernable effects of AAV2-GAD on general health, and a behavioral assessment of the animals did not reveal any alteration in general behavior, exploration, locomotion or motor symmetry. CONCLUSIONS: The present study met Food and Drug Administration requirements, in addition to efficacy and toxicity studies in rodents and nonhuman primates, to support and supplement a Phase II clinical trial invloving the gene transfer of AAV2-GAD to the human STN for the potential therapy of Parkinson's disease.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/pharmacokinetics , Parkinson Disease/therapy , Subthalamus/metabolism , Animals , Cryoultramicrotomy , DNA Primers/genetics , DNA, Viral/blood , DNA, Viral/cerebrospinal fluid , Dependovirus/metabolism , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Glutamate Decarboxylase/metabolism , Immunohistochemistry , Motor Activity/drug effects , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Subthalamus/virology
16.
Epilepsia ; 50(1): 24-32, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18717707

ABSTRACT

Results from animal models suggest gene therapy is a promising new approach for the treatment of epilepsy. Several candidate genes such as neuropeptide Y and galanin have been demonstrated in preclinical studies to have a positive effect on seizure activity. For a successful gene therapy-based treatment, efficient delivery of a transgene to target neurons is also essential. To this end, advances have been made in the areas of cell transplantation and in the development of recombinant viral vectors for gene delivery. Recombinant adeno-associated viral (rAAV) vectors in particular show promise for gene therapy of neurological disorders due to their neuronal tropism, lack of toxicity, and stable persistence in neurons, which results in robust, long-term expression of the transgene. rAAV vectors have been recently used in phase I clinical trials of Parkinson's disease with an excellent safety profile. Prior to commencement of phase I trials for gene therapy of epilepsy, further preclinical studies are ongoing including evaluation of the therapeutic benefit in chronic models of epileptogenesis, as well as assessment of safety in toxicological studies.


Subject(s)
Epilepsy/genetics , Epilepsy/therapy , Genetic Therapy/methods , Cell Transplantation/methods , Embryonic Stem Cells/transplantation , Feasibility Studies , Galanin/genetics , Gene Transfer Techniques , Genes, Viral/genetics , Genetic Vectors/genetics , Humans , Neuropeptide Y/genetics
17.
Mol Ther ; 16(5): 947-56, 2008 May.
Article in English | MEDLINE | ID: mdl-18388917

ABSTRACT

We report the characterization of a new rapid-onset model of Huntington's disease (HD) generated by adeno-associated virus (AAV) vector-mediated gene transfer of N-terminal huntingtin (htt) constructs into the rat striatum. Expression of exon 1 of mutant htt containing 70 CAG repeats rapidly led to neuropathological features associated with HD. In addition, we report novel data relating to neuronal transduction of AAV vectors that modulated the phenotype observed in this model. Quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) revealed that AAV vector-mediated expression in the striatum increased by >100-fold as compared to the endogenous htt level. Moreover, AAV vectors exhibited nonuniform transduction patterns in striatal neuronal populations, as well as axonal transport leading to transduction and neuronal cell death in the globus pallidus and substantia nigra (SN). These findings may inform future studies that utilize AAV vectors for neurodegenerative disease modeling. Further, RNA interference (RNAi) of mutant htt expression mediated by virus vector delivery of short hairpin RNAs (shRNAs) ameliorates early-stage disease phenotypes in transgenic mouse models of HD. However, it has not been reported whether shRNA-mediated knockdown of mutant htt expression is neuroprotective. AAV-shRNA was shown to mediate a dramatic knockdown of HD70 expression, preventing striatal neurodegeneration and concomitant motor behavioral impairment. These results provide further support for the use of AAV vector-mediated RNAi as a therapeutic strategy for HD.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Huntington Disease/genetics , Huntington Disease/therapy , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuroprotective Agents/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , RNA Interference , Animals , Corpus Striatum/metabolism , Exons , Genetic Vectors , Humans , Huntingtin Protein , Neurons/metabolism , Phenotype , Rats , Reverse Transcriptase Polymerase Chain Reaction
18.
Mol Ther ; 16(6): 1048-55, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18398426

ABSTRACT

Defects in cellular quality control mechanisms are thought to contribute to the neuropathology of Parkinson's disease (PD). Overexpressing heat shock proteins (HSPs) may constitute a powerful therapeutic strategy for PD, because they boost the ability of the cell to eliminate unwanted proteins. We investigated the neuroprotective potential of HSP70, HSP40, and H-BH, a constitutively active form of heat shock factor 1, in a rat model of PD based on adeno-associated virus (AAV) vector-mediated overexpression of CDCrel-1, a parkin substrate known to be toxic to dopaminergic neurons. AAV vector-mediated overexpression of H-BH and of HSP70 afforded similar levels of protection against CDCrel-1 toxicity, with approximately 20% improvement in survival of dopaminergic neurons as compared to the controls. The assessment of protection conferred was made using tyrosine hydroxylase (TH) and HuC/D immunohistochemistry and Fluoro-Gold retrograde tracing, and by observing the extent of preservation of spontaneous function and also the extent of drug-induced motor function. In contrast to H-BH and HSP70, HSP40 overexpression exacerbated CDCrel-1-mediated cell death. Real-time reverse transcriptase (RT)-PCR analysis showed that H-BH had the effect of upregulating endogenous HSP70 and HSP40 mRNA levels 10-fold and 4-fold over basal levels, respectively, whereas AAV vector-mediated HSP70 and HSP40 mRNA levels were over 100-fold higher. Our results suggest that a comparatively modest upregulation of multiple HSPs may be an effective approach for achieving significant neuroprotection in PD.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , Transcription Factors/metabolism , Animals , Behavior, Animal , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Dependovirus/metabolism , Dopamine/metabolism , HSP40 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Humans , Models, Biological , Neurons/metabolism , Septins
19.
Lancet ; 369(9579): 2097-105, 2007 Jun 23.
Article in English | MEDLINE | ID: mdl-17586305

ABSTRACT

BACKGROUND: Dopaminergic neuronal loss in Parkinson's disease leads to changes in the circuitry of the basal ganglia, such as decreased inhibitory GABAergic input to the subthalamic nucleus. We aimed to measure the safety, tolerability, and potential efficacy of transfer of glutamic acid decarboxylase (GAD) gene with adeno-associated virus (AAV) into the subthalamic nucleus of patients with Parkinson's disease. METHODS: We did an open label, safety and tolerability trial of unilateral subthalamic viral vector (AAV-GAD) injection in 11 men and 1 woman with Parkinson's disease (mean age 58.2, SD=5.7 years). Four patients received low-dose, four medium-dose, and four high-dose AAV-GAD at New York Presbyterian Hospital. Inclusion criteria consisted of Hoehn and Yahr stage 3 or greater, motor fluctuations with substantial off time, and age 70 years or less. Patients were assessed clinically both off and on medication at baseline and after 1, 3, 6, and 12 months at North Shore Hospital. Efficacy measures included the Unified Parkinson's Disease Rating Scale (UPDRS), scales of activities of daily living (ADL), neuropsychological testing, and PET imaging with 18F-fluorodeoxyglucose. The trial is registered with the ClinicalTrials.gov registry, number NCT00195143. FINDINGS: All patients who enrolled had surgery, and there were no dropouts or patients lost to follow-up. There were no adverse events related to gene therapy. Significant improvements in motor UPDRS scores (p=0.0015), predominantly on the side of the body that was contralateral to surgery, were seen 3 months after gene therapy and persisted up to 12 months. PET scans revealed a substantial reduction in thalamic metabolism that was restricted to the treated hemisphere, and a correlation between clinical motor scores and brain metabolism in the supplementary motor area. INTERPRETATION: AAV-GAD gene therapy of the subthalamic nucleus is safe and well tolerated by patients with advanced Parkinson's disease, suggesting that in-vivo gene therapy in the adult brain might be safe for various neurodegenerative diseases.


Subject(s)
Activities of Daily Living , Dependovirus , Genetic Therapy/methods , Glutamate Decarboxylase/genetics , Parkinson Disease/therapy , Aged , Dose-Response Relationship, Drug , Female , Genetic Therapy/adverse effects , Humans , Male , Middle Aged , Neuropsychological Tests , Parkinson Disease/classification , Severity of Illness Index , Treatment Outcome
20.
Nat Med ; 9(9): 1173-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12925848

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a gut peptide that, together with its receptor, GLP-1R, is expressed in the brain. Here we show that intracerebroventricular (i.c.v.) GLP-1 and [Ser(2)]exendin(1-9) (HSEGTFTSD; homologous to a conserved domain in the glucagon/GLP-1 family) enhance associative and spatial learning through GLP-1R. [Ser(2)]exendin(1-9), but not GLP-1, is also active when administered peripherally. GLP-1R-deficient mice have a phenotype characterized by a learning deficit that is restored after hippocampal Glp1r gene transfer. In addition, rats overexpressing GLP-1R in the hippocampus show improved learning and memory. GLP-1R-deficient mice also have enhanced seizure severity and neuronal injury after kainate administration, with an intermediate phenotype in heterozygotes and phenotypic correction after Glp1r gene transfer in hippocampal somatic cells. Systemic administration of [Ser(2)]exendin(1-9) in wild-type animals prevents kainate-induced apoptosis of hippocampal neurons. Brain GLP-1R represents a promising new target for both cognitive-enhancing and neuroprotective agents.


Subject(s)
Glucagon/pharmacology , Hippocampus/physiology , Learning/physiology , Peptide Fragments/pharmacology , Protein Precursors/pharmacology , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Administration, Intranasal , Amino Acid Sequence , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Cognition/drug effects , Gene Expression , Gene Transfer Techniques , Glucagon/administration & dosage , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Hippocampus/cytology , Hippocampus/drug effects , Kainic Acid/adverse effects , Learning/drug effects , Male , Mice , Mice, Knockout , Molecular Sequence Data , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/pharmacology , Peptide Fragments/administration & dosage , Protein Precursors/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Glucagon/drug effects , Seizures/chemically induced , Seizures/genetics , Sequence Homology, Amino Acid , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...