Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38585782

ABSTRACT

Mitochondrial dysfunction has been linked to both idiopathic and familial forms of Parkinson's disease (PD). We have previously identified RCC1-like (RCC1L) as a protein of the inner mitochondrial membrane important to mitochondrial fusion. Herein, to test whether deficits in RCC1L mitochondrial function might be involved in PD pathology, we have selectively ablated the Rcc1l gene in the dopaminergic (DA) neurons of mice. A PD-like phenotype resulted that includes progressive movement abnormalities, paralleled by progressive degeneration of the nigrostriatal tract. Experimental and control groups were examined at 2, 3-4, and 5-6 months of age. Animals were tested in the open field task to quantify anxiety, exploratory drive, locomotion, and immobility; and in the cylinder test to quantify rearing behavior. Beginning at 3-4 months, both female and male Rcc1l knockout mice show rigid muscles and resting tremor, kyphosis and a growth deficit compared with heterozygous or wild type littermate controls. Rcc1l knockout mice begin showing locomotor impairments at 3-4 months, which progress until 5-6 months of age, at which age the Rcc1l knockout mice die. The progressive motor impairments were associated with progressive and significantly reduced tyrosine hydroxylase immunoreactivity in the substantia nigra pars compacta (SNc), and dramatic loss of nigral DA projections in the striatum. Dystrophic spherical mitochondria are apparent in the soma of SNc neurons in Rcc1l knockout mice as early as 1.5-2.5 months of age and become progressively more pronounced until 5-6 months. Together, the results reveal the RCC1L protein to be essential to in vivo mitochondrial function in DA neurons. Further characterization of this mouse model will determine whether it represents a new model for in vivo study of PD, and the putative role of the human RCC1L gene as a risk factor that might increase PD occurrence and severity in humans.

2.
J Neurochem ; 154(4): 404-423, 2020 08.
Article in English | MEDLINE | ID: mdl-31945187

ABSTRACT

Nε-lysine acetylation of nascent glycoproteins within the endoplasmic reticulum (ER) lumen regulates the efficiency of the secretory pathway. The ER acetylation machinery consists of the membrane transporter, acetyl-CoA transporter 1 (AT-1/SLC33A1), and two acetyltransferases, ATase1/NAT8B and ATase2/NAT8. Dysfunctional ER acetylation is associated with severe neurological diseases with duplication of AT-1/SLC33A1 being associated with autism spectrum disorder, intellectual disability, and dysmorphism. Neuron-specific AT-1 over-expression in the mouse alters neuron morphology and function, causing an autism-like phenotype, indicating that ER acetylation plays a key role in neurophysiology. As such, characterizing the molecular mechanisms that regulate the acetylation machinery could reveal critical information about its biology. By using structure-biochemistry approaches, we discovered that ATase1 and ATase2 share enzymatic properties but differ in that ATase1 is post-translationally regulated via acetylation. Furthermore, gene expression studies revealed that the promoters of AT-1, ATase1, and ATase2 contain functional binding sites for the neuron-related transcription factors cAMP response element-binding protein and the immediate-early genes c-FOS and c-JUN, and that ATase1 and ATase2 exhibit additional modes of transcriptional regulation relevant to aging and Alzheimer's disease. In vivo rodent gene expression experiments revealed that Atase2 is specifically induced following activity-dependent events. Finally, over-expression of either ATase1 or ATase2 was sufficient to increase the engagement of the secretory pathway in PC12 cells. Our results indicate important regulatory roles for ATase1 and ATase2 in neuron function with induction of ATase2 expression potentially serving as a critical event that adjusts the efficiency of the secretory pathway for activity-dependent neuronal functions.


Subject(s)
Acetyltransferases/metabolism , Endoplasmic Reticulum/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism , Secretory Pathway/physiology , Acetylation , Animals , Humans , Male , Mice , Mice, Inbred C57BL , PC12 Cells , Protein Processing, Post-Translational , Rats , Rats, Inbred F344 , Transcription, Genetic
3.
Methods Mol Biol ; 1941: 93-105, 2019.
Article in English | MEDLINE | ID: mdl-30707430

ABSTRACT

Scientific progress in the understanding of the molecular mechanisms of aging in the brain is essential for the identification of novel targets for the treatment and prevention of age-associated cognitive disorders. Electrophysiological analysis of synaptic plasticity using extracellular field recordings in rodent hippocampal slices is a well-established method for investigating molecular mechanisms of learning and memory. These methods can be applied to the study of aging in the brain by utilizing hippocampal slices from aged animals. However, this application can be challenging as it is difficult to ensure and maintain the health of slices originating from aged animals. The technique described in this chapter outlines the procedure for measuring metabotropic glutamate receptor-mediated long-term depression in hippocampal slices using extracellular field recordings and includes specific details for the application of this technique in the study of neuronal aging.


Subject(s)
Aging , Hippocampus/metabolism , Long-Term Synaptic Depression , Receptors, Metabotropic Glutamate/metabolism , Animals , Electrophysiology/methods , Glutamic Acid/metabolism , Hippocampus/cytology , Rats
4.
Neurobiol Learn Mem ; 156: 17-23, 2018 12.
Article in English | MEDLINE | ID: mdl-30336208

ABSTRACT

Coiled-coil forms of Homer1, including Homer1b and c (Homer1b/c) have been shown to play a role in hippocampal learning and memory and synaptic plasticity. We have previously found that overexpression of hippocampal Homer1c is sufficient to rescue learning and memory ability in aged learning impaired rats and in Homer1 knockout (KO) mice, and to rescue group I metabotropic glutamate receptor (mGluR1/5) mediated long-term potentiation in KO mice. Here, to determine if Homer1b/c is necessary for successful learning and memory we have utilized a rAAV5 vector expressing a Homer1b/c-targeting short hairpin RNA to knock down the expression of hippocampal Homer1b/c in adult 4-6-month old male Sprague Dawley rats. We have found that reduced hippocampal Homer1b/c expression elicits significant learning deficits in contextual fear conditioning, but not in the Morris water maze or novel object recognition tasks. Furthermore, we demonstrate that reduced hippocampal Homer1b/c is sufficient to completely block mGluR1/5 mediated long-term depression in the Schaffer collateral pathway. These results support a significant role for Homer1b/c in learning and synaptic plasticity; however, the exact role of each of these two protein isoforms in learning and memory remains elusive.


Subject(s)
Conditioning, Classical/physiology , Fear/physiology , Hippocampus/metabolism , Homer Scaffolding Proteins/metabolism , Maze Learning/physiology , Neuronal Plasticity/physiology , Receptors, Metabotropic Glutamate/metabolism , Recognition, Psychology/physiology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Gene Knockdown Techniques , Homer Scaffolding Proteins/genetics , Male , Rats , Rats, Sprague-Dawley
5.
Neurobiol Aging ; 63: 1-11, 2018 03.
Article in English | MEDLINE | ID: mdl-29207276

ABSTRACT

Previous studies from our laboratory have shown that environmental enrichment (EE) in young rats results in improved learning ability and enhanced metabotropic glutamate receptor-dependent long-term potentiation (mGluR-dependent LTP) resulting from sustained activation of p70S6 kinase. Here, we investigated whether 1-month EE is sufficient to improve hippocampus-dependent learning and memory and enhance hippocampal LTP in 23-24 month-old Fischer 344 male rats. Aged rats were housed in environmentally enriched, socially enriched, or standard housing conditions. We find that aged rats exposed to 1-month of EE demonstrate enhanced learning and memory relative to standard housed controls when tested in the Morris water maze and novel object recognition behavioral tasks. Furthermore, we find that environmentally enriched rats perform significantly better than socially enriched or standard housed rats in the radial-arm water maze and display enhanced mGluR5-dependent hippocampal LTP. Enhanced hippocampal function results from activity-dependent increases in the levels of mGluR5, Homer1c, and phospho-p70S6 kinase. These findings demonstrate that a short exposure of EE to aged rats can have significant effects on hippocampal function.


Subject(s)
Aging/physiology , Aging/psychology , Environment , Hippocampus/physiology , Homer Scaffolding Proteins/metabolism , Learning/physiology , Long-Term Potentiation/physiology , Maze Learning/physiology , Memory/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Metabotropic Glutamate/physiology , Animals , Behavior, Animal/physiology , Housing, Animal , Male , Rats, Inbred F344 , Recognition, Psychology/physiology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Social Environment
6.
Behav Brain Res ; 322(Pt B): 269-279, 2017 03 30.
Article in English | MEDLINE | ID: mdl-27544872

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease that targets memory and cognition, and is the most common form of dementia among the elderly. Although AD itself has been extensively studied, very little is known about early-stage preclinical events and/or mechanisms that may underlie AD pathogenesis. Since the majority of AD cases are sporadic in nature, advancing age remains the greatest known risk factor for AD. However, additional environmental and epigenetic factors are thought to accompany increasing age to play a significant role in the pathogenesis of AD. Postoperative cognitive delirium (POD) is a behavioral syndrome that primarily occurs in elderly patients following a surgical procedure or injury and is characterized by disruptions in cognition. Individuals that experience POD are at an increased risk for developing dementia and AD compared to normal aging individuals. One way in which cognitive function is affected in cases of POD is through activation of the inflammatory cascade following surgery or injury. There is compelling evidence that immune challenges (surgery and/or injury) associated with POD trigger the release of pro-inflammatory cytokines into both the periphery and central nervous system. Thus, it is possible that cognitive impairments following an inflammatory episode may lead to more severe forms of dementia and AD pathogenesis. Here we will discuss the inflammation associated with POD, and highlight the advantages of using POD as a model to study inflammation-evoked cognitive impairment. We will explore the possibility that advancing age and immune challenges may provide mechanistic evidence correlating early life POD with AD. We will review and propose neural mechanisms by which cognitive impairments occur in cases of POD, and discuss how POD may augment the onset of AD.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , Cognition Disorders/immunology , Delirium/immunology , Inflammation/physiopathology , Postoperative Complications/immunology , Aging/psychology , Animals , Cognition Disorders/etiology , Delirium/etiology , Humans , Inflammation/psychology , Postoperative Complications/psychology
7.
J Exp Med ; 213(7): 1267-84, 2016 06 27.
Article in English | MEDLINE | ID: mdl-27242167

ABSTRACT

The import of acetyl-CoA into the lumen of the endoplasmic reticulum (ER) by AT-1/SLC33A1 regulates Nε-lysine acetylation of ER-resident and -transiting proteins. Specifically, lysine acetylation within the ER appears to influence the efficiency of the secretory pathway by affecting ER-mediated quality control. Mutations or duplications in AT-1/SLC33A1 have been linked to diseases such as familial spastic paraplegia, developmental delay with premature death, and autism spectrum disorder with intellectual disability. In this study, we generated an AT-1 Tg mouse model that selectively overexpresses human AT-1 in neurons. These animals demonstrate cognitive deficits, autistic-like social behavior, aberrations in synaptic plasticity, an increased number of dendritic spines and branches, and widespread proteomic changes. We also found that AT-1 activity regulates acetyl-CoA flux, causing epigenetic modulation of the histone epitope H3K27 and mitochondrial adaptation. In conclusion, our results indicate that increased expression of AT-1 can cause an autistic-like phenotype by affecting key neuronal metabolic pathways.


Subject(s)
Autism Spectrum Disorder/metabolism , Dendritic Spines/metabolism , Epigenesis, Genetic , Membrane Transport Proteins/biosynthesis , Phenotype , Acetyl Coenzyme A/genetics , Acetyl Coenzyme A/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/pathology , Dendritic Spines/genetics , Dendritic Spines/pathology , Histones/genetics , Histones/metabolism , Humans , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
8.
Brain ; 139(Pt 3): 937-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26787453

ABSTRACT

The aberrant accumulation of toxic protein aggregates is a key feature of many neurodegenerative diseases, including Huntington's disease, amyotrophic lateral sclerosis and Alzheimer's disease. As such, improving normal proteostatic mechanisms is an active target for biomedical research. Although they share common pathological features, protein aggregates form in different subcellular locations. Nε-lysine acetylation in the lumen of the endoplasmic reticulum has recently emerged as a new mechanism to regulate the induction of autophagy. The endoplasmic reticulum acetylation machinery includes AT-1/SLC33A1, a membrane transporter that translocates acetyl-CoA from the cytosol into the endoplasmic reticulum lumen, and ATase1 and ATase2, two acetyltransferases that acetylate endoplasmic reticulum cargo proteins. Here, we used a mutant form of α-synuclein to show that inhibition of the endoplasmic reticulum acetylation machinery specifically improves autophagy-mediated disposal of toxic protein aggregates that form within the secretory pathway, but not those that form in the cytosol. Consequently, haploinsufficiency of AT-1/SLC33A1 in the mouse rescued Alzheimer's disease, but not Huntington's disease or amyotrophic lateral sclerosis. In fact, intracellular toxic protein aggregates in Alzheimer's disease form within the secretory pathway while in Huntington's disease and amyotrophic lateral sclerosis they form in different cellular compartments. Furthermore, biochemical inhibition of ATase1 and ATase2 was also able to rescue the Alzheimer's disease phenotype in a mouse model of the disease. Specifically, we observed reduced levels of soluble amyloid-ß aggregates, reduced amyloid-ß pathology, reduced phosphorylation of tau, improved synaptic plasticity, and increased lifespan of the animals. In conclusion, our results indicate that Nε-lysine acetylation in the endoplasmic reticulum lumen regulates normal proteostasis of the secretory pathway; they also support therapies targeting endoplasmic reticulum acetyltransferases, ATase1 and ATase2, for a subset of chronic degenerative diseases.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Membrane Transport Proteins/biosynthesis , Protein Biosynthesis/physiology , Secretory Pathway/physiology , Alzheimer Disease/prevention & control , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Mice , Mice, Transgenic , Protein Processing, Post-Translational/physiology
9.
Methods Mol Biol ; 1382: 95-106, 2016.
Article in English | MEDLINE | ID: mdl-26611581

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors have become increasingly popular in research and clinical trials due to their efficient gene transfer and long-term expression in tissues including brain. In addition, rAAV has demonstrated an impressive safety profile in gene therapy trials. The emergence of rAAV serotypes with different cell tropisms and distribution properties has allowed scientists to tailor serotypes to specific experimental needs. AAV does not have a cytopathic effect; therefore, purification methods require extraction of the viral vector from the cell. This involves gradient ultracentrifugation of the cellular extract sometimes followed by chromatography. This chapter describes a small-scale production method for rAAV purification from ten to twenty 15 cm plates of human embryonic kidney-derived 293B cells (HEK 293) cells that can yield approximately 300 µl of a 5 × 10(12) to 1 × 10(13) genome copies/ml viral preparation final concentration.


Subject(s)
Dependovirus/isolation & purification , Triiodobenzoic Acids/chemistry , Dependovirus/genetics , Genetic Vectors , HEK293 Cells , Humans , Viral Load
10.
Neurobiol Learn Mem ; 125: 126-34, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26341144

ABSTRACT

Previous studies from our lab have demonstrated that mild cognitive impairments identified early in life are predictive of cognitive deficits that develop with age, suggesting that enhancements in cognition at an early age can provide a buffer against age-related cognitive decline. Environmental enrichment has been shown to improve learning and memory in the rodent, but the impact of enrichment on synaptic plasticity and the molecular mechanisms behind enrichment are not completely understood. To address these unresolved issues, we have housed 2-month old rats in environmentally enriched (EE), socially enriched (SE), or standard housing (SC) and conducted tests of learning and memory formation at various time intervals. Here we demonstrate that animals that have been exposed to one month of social or environmental enrichment demonstrate enhanced learning and memory relative to standard housed controls. However, we have found that after 4months EE animals perform better than both SE and SC groups and demonstrate an enhanced hippocampal LTP. Our results demonstrate that this LTP is dependent on mGluR5 signaling, activation of ERK and mTOR signaling cascades, and sustained phosphorylation of p70s6 kinase, thus providing a potential target mechanism for future studies of cognitive enhancement in the rodent.


Subject(s)
Environment , Learning/physiology , Long-Term Potentiation/physiology , Memory/physiology , Receptor, Metabotropic Glutamate 5/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Housing, Animal , Learning/drug effects , Long-Term Potentiation/drug effects , Memory/drug effects , Phosphorylation/drug effects , Pyridines/pharmacology , Rats , Receptor, Metabotropic Glutamate 5/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
11.
Behav Brain Res ; 294: 224-33, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26283528

ABSTRACT

The Morris water maze (MWM) behavioral paradigm is commonly used to measure spatial learning and memory in rodents. It is widely accepted that performance in the MWM declines with age. However, young rats ubiquitously perform very well on established versions of the water maze, suggesting that more challenging tasks may be required to reveal subtle differences in young animals. Therefore, we have used a one-day water maze and novel object recognition to test whether more sensitive paradigms of memory in young animals could identify subtle cognitive impairments early in life that might become accentuated later with senescence. We have found that these two tasks reliably separate young rats into inferior and superior learners, are highly correlated, and that performance on these tasks early in life is predictive of performance at 12 months of age. Furthermore, we have found that repeated training in this task selectively improves the performance of inferior learners, suggesting that behavioral training from an early age may provide a buffer against age-related cognitive decline.


Subject(s)
Aging/psychology , Learning Disabilities , Aging/physiology , Animals , Cognition Disorders/physiopathology , Cognition Disorders/prevention & control , Individuality , Learning/physiology , Learning Disabilities/physiopathology , Male , Maze Learning/physiology , Psychological Tests , Rats, Sprague-Dawley , Recognition, Psychology/physiology
12.
Neurobiol Aging ; 36(10): 2725-36, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26174856

ABSTRACT

p44 is a short isoform of the tumor suppressor protein p53 that is regulated in an age-dependent manner. When overexpressed in the mouse, it causes a progeroid phenotype that includes premature cognitive decline, synaptic defects, and hyperphosphorylation of tau. The hyperphosphorylation of tau has recently been linked to the ability of p44 to regulate transcription of relevant tau kinases. Here, we report that the amyloid precursor protein (APP) intracellular domain (AICD), which results from the processing of the APP, regulates translation of p44 through a cap-independent mechanism that requires direct binding to the second internal ribosome entry site (IRES) of the p53 mRNA. We also report that AICD associates with nucleolin, an already known IRES-specific trans-acting factor that binds with p53 IRES elements and regulates translation of p53 isoforms. The potential biological impact of our findings was assessed in a mouse model of Alzheimer's disease. In conclusion, our study reveals a novel aspect of AICD and p53/p44 biology and provides a possible molecular link between APP, p44, and tau.


Subject(s)
Amyloid beta-Protein Precursor/physiology , Internal Ribosome Entry Sites/genetics , Peptide Fragments/genetics , Protein Biosynthesis/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Aging/genetics , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/chemistry , Animals , Cells, Cultured , Disease Models, Animal , Humans , Mice, Transgenic , Peptide Fragments/physiology , Phosphorylation , Protein Binding , Protein Structure, Tertiary , RNA, Messenger/genetics , Transcription Factors/physiology , Tumor Suppressor Protein p53/physiology , tau Proteins/metabolism
13.
Nat Neurosci ; 17(12): 1736-43, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25383899

ABSTRACT

In Noonan syndrome (NS) 30-50% of subjects show cognitive deficits of unknown etiology and with no known treatment. Here, we report that knock-in mice expressing either of two NS-associated mutations in Ptpn11, which encodes the nonreceptor protein tyrosine phosphatase Shp2, show hippocampal-dependent impairments in spatial learning and deficits in hippocampal long-term potentiation (LTP). In addition, viral overexpression of an NS-associated allele PTPN11(D61G) in adult mouse hippocampus results in increased baseline excitatory synaptic function and deficits in LTP and spatial learning, which can be reversed by a mitogen-activated protein kinase kinase (MEK) inhibitor. Furthermore, brief treatment with lovastatin reduces activation of the GTPase Ras-extracellular signal-related kinase (Erk) pathway in the brain and normalizes deficits in LTP and learning in adult Ptpn11(D61G/+) mice. Our results demonstrate that increased basal Erk activity and corresponding baseline increases in excitatory synaptic function are responsible for the LTP impairments and, consequently, the learning deficits in mouse models of NS. These data also suggest that lovastatin or MEK inhibitors may be useful for treating the cognitive deficits in NS.


Subject(s)
Disease Models, Animal , Learning/physiology , Long-Term Potentiation/physiology , Lovastatin/therapeutic use , Memory Disorders/physiopathology , Noonan Syndrome/physiopathology , Animals , Female , Humans , Learning/drug effects , Long-Term Potentiation/drug effects , Lovastatin/pharmacology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory Disorders/drug therapy , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Noonan Syndrome/drug therapy , Random Allocation , Rats , Treatment Outcome
14.
Hippocampus ; 24(1): 1-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24167026

ABSTRACT

Group I metabotropic glutamate receptors (mGluR1/5) play a role in synaptic plasticity and they demonstrate direct interactions with the neuronal Homer1c protein. We have previously shown that Homer1c can restore the plasticity deficits in Homer1 knockout mice (H1-KO). Here, we investigated the role of Homer1c in mGluR-dependent synaptic plasticity in wild-type mice, H1-KO, and H1-KO mice overexpressing Homer1c (KO+H1c). We used a form of plasticity induced by activation of mGluR1/5 that transforms short-term potentiaion (STP) induced by a subthreshold theta burst stimulation into long-term potentiation (LTP). We have shown that although acute hippocampal slices from wild-type animals can induce LTP using this stimulation protocol, H1-KO only show STP. Gene delivery of Homer1c into the hippocampus of H1-KO mice rescued LTP to wild-type levels. This form of synaptic plasticity was dependent on mGluR5 but not mGluR1 activation both in wild-type mice and in KO+H1c. mGluR1/5-dependent LTP was blocked with inhibitors of the MEK-ERK and PI3K-mTOR pathways in KO+H1c mice. Moreover, blocking Homer1c-mGluR5 interactions prevented the maintenance of LTP in acute hippocampal slices from KO+H1c. These data indicate that Homer1c-mGluR5 interactions are necessary for mGluR-dependent LTP, and that mGluR1/5-dependent LTP involves PI3K and ERK activation.


Subject(s)
Carrier Proteins/metabolism , Hippocampus/metabolism , Long-Term Potentiation/physiology , Receptors, Metabotropic Glutamate/metabolism , Animals , Electrophysiology , Excitatory Postsynaptic Potentials/physiology , Homer Scaffolding Proteins , Mice , Mice, Knockout
15.
Article in English | MEDLINE | ID: mdl-26015943

ABSTRACT

Intraoperative magnetic resonance imaging (MRI) has been proposed as a method to optimize intracerebral targeting and for tracking infusate distribution in gene therapy trials for nervous system disorders. We thus investigated possible effects of two MRI contrast agents, gadoteridol (Gd) and galbumin (Gab), on the distribution and levels of transgene expression in the rat striatum and their effect on integrity and stability of recombinant adeno-associated virus (rAAV) particles. MRI studies showed that contrast agent distribution did not predict rAAV distribution. However, green fluorescent protein (GFP) immunoreactivity revealed an increase in distribution of rAAV5-GFP, but not rAAV2-GFP, in the presence of Gd when compared with viral vector injected alone. In contrast, Gab increased the distribution of rAAV2-GFP not rAAV5-GFP. These observations pointed to a direct effect of infused contrast agent on the rAAV particles. Negative-stain electron microscopy (EM), DNAase treatment, and differential scanning calorimetry (DSC) were used to monitor rAAV2 and rAAV5 particle integrity and stability following contrast agent incubation. EMs of rAAV2-GFP and rAAV5-GFP particles pretreated with Gd appear morphologically similar to the untreated sample; however, Gab treatment resulted in surface morphology changes and aggregation. A compromise of particle integrity was suggested by sensitivity of the packaged genome to DNAase treatment following Gab incubation but not Gd for both vectors. However, neither agent significantly affected particle stability when analyzed by DSC. An increase in T m was observed for AAV2 in lactated Ringer's buffer. These results thus highlight potential interactions between MRI contrast agents and AAV that might affect vector distribution and stability, as well as the stabilizing effect of lactated Ringer's solution on AAV2.

16.
PLoS Biol ; 11(8): e1001627, 2013.
Article in English | MEDLINE | ID: mdl-23966835

ABSTRACT

Tuberous sclerosis complex (TSC) is a multisystem genetic disease that manifests with mental retardation, tumor formation, autism, and epilepsy. Heightened signaling through the mammalian target of rapamycin (mTOR) pathway is involved in TSC pathology, however it remains unclear how other signaling pathways are perturbed and contribute to disease symptoms. Reduced long-term depression (LTD) was recently reported in TSC mutant mice. We find that although reduced LTD is a feature of the juvenile mutant hippocampus, heightened expression of metabotropic glutamate receptor 5 and constitutively activated Erk signaling in the adult hippocampus drives wild-type levels of LTD. Increased mGluR5 and Erk results in a novel mTOR-independent LTD in CA1 hippocampus of adult mice, and contributes to the development of epileptiform bursting activity in the TSC2(+/-) CA3 region of the hippocampus. Inhibition of mGluR5 or Erk signaling restores appropriate mTOR-dependence to LTD, and significantly reduces epileptiform bursting in TSC2(+/-) hippocampal slices. We also report that adult TSC2(+/-) mice exhibit a subtle perseverative behavioral phenotype that is eliminated by mGluR5 antagonism. These findings highlight the potential of modulating the mGluR5-Erk pathway in a developmental stage-specific manner to treat TSC.


Subject(s)
Depression/physiopathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/physiopathology , Tuberous Sclerosis/psychology , Animals , Blotting, Western , Electrophysiology , Extracellular Signal-Regulated MAP Kinases/genetics , Male , Mice , Receptor, Metabotropic Glutamate 5/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
17.
PLoS One ; 8(5): e62360, 2013.
Article in English | MEDLINE | ID: mdl-23667471

ABSTRACT

Aged-related cognitive ability is highly variable, ranging from unimpaired to severe impairments. The Morris water maze (a reliable tool for assessing memory) has been used to distinguish aged rodents that are superior learners from those that are learning impaired. This task, however, is not practical for pre- and post-pharmacological treatment, as the memory of the task is long lasting. In contrast, the object location memory task, also a spatial learning paradigm, results in a less robust memory that decays quickly. We demonstrate for the first time how these two paradigms can be used together to assess hippocampal cognitive impairments before and after pharmacological or genetic manipulations in rodents. Rats were first segregated into superior learning and learning impaired groups using the object location memory task, and their performance was correlated with future outcome on this task and on the Morris water maze. This method provides a tool to evaluate the effect of treatments on cognitive impairment associated with aging and neurodegenerative disorders.


Subject(s)
Aging/physiology , Behavior, Animal/drug effects , Genetic Techniques , Hippocampus/drug effects , Hippocampus/physiology , Animals , Cognition Disorders/drug therapy , Cognition Disorders/genetics , Cognition Disorders/physiopathology , Maze Learning/drug effects , Memory/drug effects , Rats , Reproducibility of Results , Spatial Behavior/drug effects , Spatial Behavior/physiology
18.
Neurobiol Aging ; 34(8): 1963-70, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23523268

ABSTRACT

Homer1c has been shown to play a role in learning and memory. Overexpression of Homer1c in the hippocampus can improve memory in normal rats and can also rescue spatial learning deficits in Homer1 knockout mice. In a previous study, we found that Homer1c mRNA is upregulated after a spatial learning paradigm in aged rats that successfully learn the task, when compared to aged rats that are learning-impaired (AI). This study was designed to validate the role of Homer1c in successful cognitive aging. In this article, we report that gene delivery of Homer1c into the hippocampus of aged learning-impaired rats significantly improves individual performance on an object location memory task. The learning ability of these rats on the Morris Water Maze was also superior to that of AI control rats. In summary, using 2 independent spatial memory tasks, we demonstrate that Homer1c is sufficient to improve the spatial learning deficits in a rodent model of cognitive aging. These results point to Homer1c as a potential therapeutic target for improving age-related cognitive impairment.


Subject(s)
Aging/genetics , Aging/psychology , Carrier Proteins/genetics , Carrier Proteins/physiology , Maze Learning/physiology , Memory Disorders/genetics , Memory Disorders/psychology , Spatial Behavior/physiology , Animals , Disease Models, Animal , Gene Expression , Gene Transfer Techniques , Genetic Therapy/methods , Hippocampus/physiology , Homer Scaffolding Proteins , Male , Memory Disorders/therapy , Mice , Molecular Targeted Therapy , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Up-Regulation
19.
Neurobiol Learn Mem ; 97(1): 17-29, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21945599

ABSTRACT

Homer1 belongs to a family of scaffolding proteins that interact with various post-synaptic density proteins including group I metabotropic glutamate receptors (mGluR1/5). Previous research in our laboratory implicates the Homer1c isoform in spatial learning. Homer1 knockout mice (H1-KO) display cognitive impairments, but their synaptic plasticity properties have not been described. Here, we investigated the role of Homer1 in long-term potentiation (LTP) in the hippocampal CA1 region of H1-KO mice in vitro. We found that late-phase LTP elicited by high frequency stimulation (HFS) was impaired, and that the induction and maintenance of theta burst stimulation (TBS) LTP were reduced in H1-KO. To test the hypothesis that Homer1c was sufficient to rescue these LTP deficits, we delivered Homer1c to the hippocampus of H1-KO using recombinant adeno-associated virus (rAAV). We found that rAAV-Homer1c rescued HFS and TBS-LTP in H1-KO animals. Next, we tested whether the LTP rescue by Homer1c was occurring via mGluR1/5. A selective mGluR5 antagonist, but not an mGluR1 antagonist, blocked the Homer1c-induced recovery of late-LTP, suggesting that Homer1c mediates functional effects on plasticity via mGluR5. To investigate the role of Homer1c in spatial learning, we injected rAAV-Homer1c to the hippocampus of H1-KO. We found that rAAV-Homer1c significantly improved H1-KO performance in the Radial Arm Water Maze. These results point to a significant role for Homer1c in synaptic plasticity and learning.


Subject(s)
Carrier Proteins/genetics , Hippocampus/metabolism , Maze Learning/physiology , Neuronal Plasticity/physiology , Recognition, Psychology/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzoates/pharmacology , Carrier Proteins/metabolism , Gene Transfer Techniques , Glycine/analogs & derivatives , Glycine/pharmacology , Hippocampus/drug effects , Homer Scaffolding Proteins , Maze Learning/drug effects , Mice , Mice, Knockout , Neuronal Plasticity/drug effects , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Recognition, Psychology/drug effects , Social Behavior , Synapses/drug effects , Synapses/metabolism
20.
PLoS One ; 6(10): e26549, 2011.
Article in English | MEDLINE | ID: mdl-22046307

ABSTRACT

Fragile X syndrome (FXS) is the most common form of inherited intellectual disability and the leading known genetic cause of autism. Fragile X mental retardation protein (FMRP), which is absent or expressed at substantially reduced levels in FXS, binds to and controls the postsynaptic translation of amyloid ß-protein precursor (AßPP) mRNA. Cleavage of AßPP can produce ß-amyloid (Aß), a 39-43 amino acid peptide mis-expressed in Alzheimer's disease (AD) and Down syndrome (DS). Aß is over-expressed in the brain of Fmr1(KO) mice, suggesting a pathogenic role in FXS. To determine if genetic reduction of AßPP/Aß rescues characteristic FXS phenotypes, we assessed audiogenic seizures (AGS), anxiety, the ratio of mature versus immature dendritic spines and metabotropic glutamate receptor (mGluR)-mediated long-term depression (LTD) in Fmr1(KO) mice after removal of one App allele. All of these phenotypes were partially or completely reverted to normal. Plasma Aß(1-42) was significantly reduced in full-mutation FXS males compared to age-matched controls while cortical and hippocampal levels were somewhat increased, suggesting that Aß is sequestered in the brain. Evolving therapies directed at reducing Aß in AD may be applicable to FXS and Aß may serve as a plasma-based biomarker to facilitate disease diagnosis or assess therapeutic efficacy.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/prevention & control , Fragile X Syndrome/therapy , Peptide Fragments/metabolism , Amyloid beta-Peptides/blood , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/blood , Amyloid beta-Protein Precursor/genetics , Animals , Brain Chemistry , Dendritic Spines , Down-Regulation , Female , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , Genetic Therapy , Male , Mice , Mice, Knockout , Neurons/ultrastructure , Peptide Fragments/blood , Peptide Fragments/genetics , Phenotype , Receptors, Metabotropic Glutamate/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...