Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Hum Mol Genet ; 32(23): 3263-3275, 2023 Nov 17.
Article in English | MEDLINE | ID: mdl-37658769

ABSTRACT

The COPI coatomer subunit α-COP has been shown to co-precipitate mRNA in multiple settings, but it was unclear whether the interaction with mRNA was direct or mediated by interaction with an adapter protein. The COPI complex often interacts with proteins via C-terminal dilysine domains. A search for candidate RNA binding proteins with C-terminal dilysine motifs yielded Nucleolin, which terminates in a KKxKxx sequence. This protein was an especially intriguing candidate as it has been identified as an interacting partner for Survival Motor Neuron protein (SMN). Loss of SMN causes the neurodegenerative disease Spinal Muscular Atrophy. We have previously shown that SMN and α-COP interact and co-migrate in axons, and that overexpression of α-COP reduced phenotypic severity in cell culture and animal models of SMA. We show here that in an mRNA independent manner, endogenous Nucleolin co-precipitates endogenous α-COP and ε-COP but not ß-COP which may reflect an interaction with the so-called B-subcomplex rather a complete COPI heptamer. The ability of Nucleolin to bind to α-COP requires the presence of the C-terminal KKxKxx domain of Nucleolin. Furthermore, we have generated a point mutant in the WD40 domain of α-COP which eliminates its ability to co-precipitate Nucleolin but does not interfere with precipitation of partners mediated by non-KKxKxx motifs such as the kainate receptor subunit 2. We propose that via interaction between the C-terminal dilysine motif of Nucleolin and the WD40 domain of α-COP, Nucleolin acts an adaptor to allow α-COP to interact with a population of mRNA.


Subject(s)
Muscular Atrophy, Spinal , Neurodegenerative Diseases , Animals , Coatomer Protein/genetics , Protein Binding , Phosphoproteins/genetics , Phosphoproteins/metabolism , Muscular Atrophy, Spinal/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleolin
2.
Neurobiol Aging ; 101: 57-69, 2021 05.
Article in English | MEDLINE | ID: mdl-33582567

ABSTRACT

Understanding the cellular processes that lead to Alzheimer's disease (AD) is critical, and one key lies in the genetics of families with histories of AD. Mutations a complex known as COPI were found in families with AD. The COPI complex is involved in protein processing and trafficking. Intriguingly, several recent publications have found components of the COPI complex can affect the metabolism of pathogenic AD proteins. We reduced levels of the COPI subunit α-COP, altering maturation and cleavage of amyloid precursor protein (APP), resulting in decreased release of Aß-42 and decreased accumulation of the AICD. Depletion of α-COP reduced uptake of proteopathic Tau seeds and reduces intracellular Tau self-association. Expression of AD-associated mutant α-COP altered APP processing, resulting in increased release of Aß-42 and increased intracellular Tau aggregation and release of Tau oligomers. These results show that COPI coatomer function modulates processing of both APP and Tau, and expression of AD-associated α-COP confers a toxic gain of function, resulting in potentially pathogenic changes in both APP and Tau.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Coat Protein Complex I/genetics , Coat Protein Complex I/physiology , Coatomer Protein/genetics , Coatomer Protein/physiology , Mutation/genetics , Mutation/physiology , Peptide Fragments/metabolism , tau Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Cells, Cultured , Mice
3.
Biochem Biophys Res Commun ; 514(2): 530-537, 2019 06 25.
Article in English | MEDLINE | ID: mdl-31060774

ABSTRACT

We report that expression of the α-COP protein rescues disease phenotype in a severe mouse model of Spinal Muscular Atrophy (SMA). Lentiviral particles expressing α-COP were injected directly into the testes of genetically pure mouse strain of interest resulting in infection of the spermatagonial stem cells. α-COP was stably expressed in brain, skeletal muscle, and spinal cord without altering SMN protein levels. SMA mice transgenic for α-COP live significantly longer than their non-transgenic littermates, and showed increased body mass and normal muscle morphology at postnatal day 15. We previously reported that binding between SMN and α-COP is required for restoration of neurite outgrowth in cells lacking SMN, and we report similar finding here. Lentiviral-mediated transgenic expression of SMN where the dilysine domain in exon 2b was mutated was not able to rescue the SMA phenotype despite robust expression of the mutant SMN protein in brain, muscle and spinal cord. These results demonstrate that α-COP is a validated modifier of SMA disease phenotype in a mammalian, vertebrate model and is a potential target for development of future SMN-independent therapeutic interventions.


Subject(s)
Coatomer Protein/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Spinal Cord/metabolism , Survival of Motor Neuron 1 Protein/genetics , Amino Acid Sequence , Animals , Brain/metabolism , Brain/pathology , Coatomer Protein/metabolism , Disease Models, Animal , Exons , Female , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Motor Neurons/pathology , Muscle, Skeletal/pathology , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/therapy , Mutation , Phenotype , Protein Binding , Signal Transduction , Spinal Cord/pathology , Survival Analysis , Survival of Motor Neuron 1 Protein/metabolism
4.
Restor Neurol Neurosci ; 36(3): 417-422, 2018.
Article in English | MEDLINE | ID: mdl-29614705

ABSTRACT

BACKGROUND: When nerve transection is performed on adult rodents, a substantial population of neurons survives short-term disconnection from target, and the immune system supports this neuronal survival, however long-term survival remains unknown. Understanding the effects of permanent axotomy on cell body survival is important as target disconnection is the first pathological occurrence in fatal motoneuron diseases such as amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA). OBJECTIVE: The goal of this study was to determine if facial motoneurons (FMN) could survive permanent target disconnection up to 26 weeks post-operation (wpo) after facial nerve axotomy (FNA). In addition, the potentially additive effects of immunodeficiency and motoneuron disease on post-axotomy FMN survival were examined. METHODS: This study included three wild type (WT) mouse strains (C57BL/6J, B6SJL, and FVB/NJ) and three experimental models (RAG-2-/-: immunodeficiency; mSOD1: ALS; Smn-/-/SMN2+/+: SMA). All animals received a unilateral FNA, and FMN survival was quantified at early and extended post-operative timepoints. RESULTS: In the C57BL/6J WT group, FMN survival significantly decreased at 10 wpo (55±6%), and then remained stable out to 26 wpo (47±6%). In the RAG-2-/- and mSOD1 groups, FMN death occurred much earlier at 4 wpo, and survival plateaued at approximately 50% at 10 wpo. The SMA model and other WT strains also exhibited approximately 50% FMN survival after FNA. CONCLUSION: These results indicate that immunodeficiency and motoneuron disease accelerate axotomy-induced neuron death, but do not increase total neuron death in the context of permanent target disconnection. This consistent finding of a target disconnection-resilient motoneuron population is prevalent in other peripheral nerve injury models and in neurodegenerative disease models as well. Characterization of the distinct populations of vulnerable and resilient motoneurons may reveal new therapeutic approaches for injury and disease.


Subject(s)
Central Nervous System Diseases/pathology , Facial Nerve Injuries/pathology , Facial Nerve/pathology , Motor Neurons/pathology , Animals , Axotomy/methods , Cell Death/physiology , Cell Survival/physiology , Mice, Inbred C57BL
5.
PLoS One ; 12(9): e0185079, 2017.
Article in English | MEDLINE | ID: mdl-28945765

ABSTRACT

C5-substituted 2,4-diaminoquinazoline inhibitors of the decapping scavenger enzyme DcpS (DAQ-DcpSi) have been developed for the treatment of spinal muscular atrophy (SMA), which is caused by genetic deficiency in the Survival Motor Neuron (SMN) protein. These compounds are claimed to act as SMN2 transcriptional activators but data underlying that claim are equivocal. In addition it is unclear whether the claimed effects on SMN2 are a direct consequence of DcpS inhibitor or might be a consequence of lysosomotropism, which is known to be neuroprotective. DAQ-DcpSi effects were characterized in cells in vitro utilizing DcpS knockdown and 7-methyl analogues as probes for DcpS vs non-DcpS-mediated effects. We also performed analysis of Smn transcript levels, RNA-Seq analysis of the transcriptome and SMN protein in order to identify affected pathways underlying the therapeutic effect, and studied lysosomotropic and non-lysosomotropic DAQ-DCpSi effects in 2B/- SMA mice. Treatment of cells caused modest and transient SMN2 mRNA increases with either no change or a decrease in SMNΔ7 and no change in SMN1 transcripts or SMN protein. RNA-Seq analysis of DAQ-DcpSi-treated N2a cells revealed significant changes in expression (both up and down) of approximately 2,000 genes across a broad range of pathways. Treatment of 2B/- SMA mice with both lysomotropic and non-lysosomotropic DAQ-DcpSi compounds had similar effects on disease phenotype indicating that the therapeutic mechanism of action is not a consequence of lysosomotropism. In striking contrast to the findings in vitro, Smn transcripts were robustly changed in tissues but there was no increase in SMN protein levels in spinal cord. We conclude that DAQ-DcpSi have reproducible benefit in SMA mice and a broad spectrum of biological effects in vitro and in vivo, but these are complex, context specific, and not the result of simple SMN2 transcriptional activation.


Subject(s)
Endoribonucleases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Muscular Atrophy, Spinal/drug therapy , Muscular Atrophy, Spinal/enzymology , Quinazolines/pharmacology , Animals , Cell Line , Disease Models, Animal , Enzyme Inhibitors/chemistry , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Muscular Atrophy, Spinal/genetics , Promoter Regions, Genetic , Quinazolines/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival of Motor Neuron 2 Protein/deficiency , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
6.
PLoS One ; 11(10): e0163954, 2016.
Article in English | MEDLINE | ID: mdl-27736905

ABSTRACT

Spinal muscular atrophy (SMA) is an intractable neurodegenerative disease afflicting 1 in 6-10,000 live births. One of the key functions of the SMN protein is regulation of spliceosome assembly. Reduced levels of the SMN protein that are observed in SMA have been shown to result in aberrant mRNA splicing. SMN-dependent mis-spliced transcripts in motor neurons may cause stresses that are particularly harmful and may serve as potential targets for the treatment of motor neuron disease or as biomarkers in the SMA patient population. We performed deep RNA sequencing using motor neuron-like NSC-34 cells to screen for SMN-dependent mRNA processing changes that occur following acute depletion of SMN. We identified SMN-dependent splicing changes, including an intron retention event that results in the production of a truncated Rit1 transcript. This intron-retained transcript is stable and is mis-spliced in spinal cord from symptomatic SMA mice. Constitutively active Rit1 ameliorated the neurite outgrowth defect in SMN depleted NSC-34 cells, while expression of the truncated protein product of the mis-spliced Rit1 transcript inhibited neurite extension. These results reveal new insights into the biological consequence of SMN-dependent splicing in motor neuron-like cells.


Subject(s)
Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , RNA Splicing , RNA, Messenger/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Animals , Cell Line , Disease Models, Animal , Exons , Female , Gene Expression Regulation , Introns , Male , Mice , Mice, Transgenic , Motor Neurons/metabolism , Muscular Atrophy, Spinal/pathology , Spinal Cord/metabolism , Spinal Cord/pathology , Transcriptome
7.
Hum Mol Genet ; 24(25): 7295-307, 2015 Dec 20.
Article in English | MEDLINE | ID: mdl-26464491

ABSTRACT

Spinal muscular atrophy (SMA), a heritable neurodegenerative disease, results from insufficient levels of the survival motor neuron (SMN) protein. α-COP binds to SMN, linking the COPI vesicular transport pathway to SMA. Reduced levels of α-COP restricted development of neuronal processes in NSC-34 cells and primary cortical neurons. Remarkably, heterologous expression of human α-COP restored normal neurite length and morphology in SMN-depleted NSC-34 cells in vitro and zebrafish motor neurons in vivo. We identified single amino acid mutants of α-COP that selectively abrogate SMN binding, retain COPI-mediated Golgi-ER trafficking functionality, but were unable to support neurite outgrowth in cellular and zebrafish models of SMA. Taken together, these demonstrate the functional role of COPI association with the SMN protein in neuronal development.


Subject(s)
Coatomer Protein/metabolism , Motor Neurons/metabolism , Muscular Atrophy, Spinal/metabolism , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Line , Cells, Cultured , Coatomer Protein/genetics , Fluorescent Antibody Technique , Humans , Immunoprecipitation , Neurites/metabolism , Protein Binding , Survival of Motor Neuron 1 Protein/genetics , Zebrafish
8.
Hum Mol Genet ; 24(14): 3908-17, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25859008

ABSTRACT

The neurodegenerative disorder spinocerebellar ataxia type 7 (SCA7) is caused by a polyglutamine (polyQ) expansion in the ataxin-7 protein, categorizing SCA7 as one member of a large class of heritable neurodegenerative proteinopathies. Cleavage of ataxin-7 by the protease caspase-7 has been demonstrated in vitro, and the accumulation of proteolytic cleavage products in SCA7 patients and mouse models has been identified as an early pathological change. However, it remains unknown whether a causal relationship exists between ataxin-7 proteolysis and in vivo SCA7 disease progression. To determine whether caspase cleavage is a critical event in SCA7 disease pathogenesis, we generated transgenic mice expressing polyQ-expanded ataxin-7 with a second-site mutation (D266N) to prevent caspase-7 proteolysis. When we compared SCA7-D266N mice with SCA7 mice lacking the D266N mutation, we found that SCA7-D266N mice exhibited improved motor performance, reduced neurodegeneration and substantial lifespan extension. Our findings indicate that proteolysis at the D266 caspase-7 cleavage site is an important mediator of ataxin-7 neurotoxicity, suggesting that inhibition of caspase-7 cleavage of polyQ-ataxin-7 may be a promising therapeutic strategy for this untreatable disorder.


Subject(s)
Ataxin-7/metabolism , Neurodegenerative Diseases/genetics , Peptides/metabolism , Promoter Regions, Genetic , Proteolysis , Retinal Degeneration/genetics , Animals , Aspartic Acid/metabolism , Ataxin-7/genetics , Caspase 7/genetics , Caspase 7/metabolism , Disease Models, Animal , Genetic Therapy , Humans , Mice , Mice, Transgenic , Neurodegenerative Diseases/therapy , Phenotype , Purkinje Cells/metabolism , Retinal Degeneration/therapy
9.
Mol Cell Neurosci ; 61: 133-40, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24983518

ABSTRACT

Abnormal autophagy has become a central thread linking neurodegenerative diseases, particularly of the motor neuron. One such disease is spinal muscular atrophy (SMA), a genetic neuromuscular disorder caused by mutations in the SMN1 gene resulting in low levels of Survival Motor Neuron (SMN) protein. Despite knowing the causal protein, the exact intracellular processes that are involved in the selective loss of motor neurons remain unclear. Autophagy induction can be helpful or harmful depending on the situation, and we sought to understand the state of the autophagic response in SMA. We show that cell culture and animal models demonstrate induction of autophagy accompanied by attenuated autophagic flux, resulting in the accumulation of autophagosomes and their associated cargo. Expression of the SMN-binding protein a-COP, a known modulator of autophagic flux, can ameliorate this autophagic traffic jam.


Subject(s)
Autophagy/physiology , Muscular Atrophy, Spinal/physiopathology , Survival of Motor Neuron 1 Protein/metabolism , Animals , Cell Line, Transformed , Disease Models, Animal , Doxycycline/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Motor Neurons/physiology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , RNA, Small Interfering/metabolism , RNA, Small Interfering/pharmacology , Survival of Motor Neuron 1 Protein/genetics , Time Factors , Transfection , Red Fluorescent Protein
10.
Hum Mol Genet ; 22(20): 4043-52, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23727837

ABSTRACT

Spinal muscular atrophy (SMA) is a devastating neuromuscular disorder that stems from low levels of survival of motor neuron (SMN) protein. The processes that cause motor neurons and muscle cells to become dysfunctional are incompletely understood. We are interested in neuromuscular homeostasis and the stresses put upon that system by loss of SMN. We recently reported that α-COP, a member of the coatomer complex of coat protein I (COPI) vesicles, is an SMN-binding partner, implicating this protein complex in normal SMN function. To investigate the functional significance of the interaction between α-COP and SMN, we constructed an inducible NSC-34 cell culture system to model the consequences of SMN depletion and find that depletion of SMN protein results in shortened neurites. Heterologous expression of human SMN, and interestingly over-expression of α-COP, restores normal neurite length and morphology. Mutagenesis of the canonical COPI dilysine motifs in exon 2b results in failure to bind to α-COP and abrogates the ability of human SMN to restore neurite outgrowth in SMN-depleted motor neuron-like NSC-34 cells. We conclude that the interaction between SMN and α-COP serves an important function in the growth and maintenance of motor neuron processes and may play a significant role in the pathogenesis of SMA.


Subject(s)
Coatomer Protein/metabolism , Dipeptides/metabolism , Exons , Models, Biological , Muscular Atrophy, Spinal/pathology , Neurites/physiology , Protein Interaction Domains and Motifs/physiology , Survival of Motor Neuron 1 Protein/chemistry , Survival of Motor Neuron 1 Protein/metabolism , Amino Acid Sequence , Cell Culture Techniques/methods , Cell Line , Coat Protein Complex I/metabolism , Dipeptides/chemistry , Dipeptides/genetics , Humans , Molecular Sequence Data , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Mutation , Neurites/pathology , Survival of Motor Neuron 1 Protein/genetics
11.
J Neurosci ; 30(22): 7729-39, 2010 Jun 02.
Article in English | MEDLINE | ID: mdl-20519548

ABSTRACT

Inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia (IBMPFD) is a dominantly inherited degenerative disorder caused by mutations in the valosin-containing protein (VCP7) gene. VCP (p97 in mouse, TER94 in Drosophila melanogaster, and CDC48 in Saccharomyces cerevisiae) is a highly conserved AAA(+) (ATPases associated with multiple cellular activities) ATPase that regulates a wide array of cellular processes. The mechanism of IBMPFD pathogenesis is unknown. To elucidate the pathogenic mechanism, we developed and characterized a Drosophila model of IBMPFD (mutant-VCP-related degeneration). Based on genetic screening of this model, we identified three RNA-binding proteins that dominantly suppressed degeneration; one of these was TBPH, the Drosophila homolog of TAR (trans-activating response region) DNA-binding protein 43 (TDP-43). Here we demonstrate that VCP and TDP-43 interact genetically and that disease-causing mutations in VCP lead to redistribution of TDP-43 to the cytoplasm in vitro and in vivo, replicating the major pathology observed in IBMPFD and other TDP-43 proteinopathies. We also demonstrate that TDP-43 redistribution from the nucleus to the cytoplasm is sufficient to induce cytotoxicity. Furthermore, we determined that a pathogenic mutation in TDP-43 promotes redistribution to the cytoplasm and enhances the genetic interaction with VCP. Together, our results show that degeneration associated with VCP mutations is mediated in part by toxic gain of function of TDP-43 in the cytoplasm. We suggest that these findings are likely relevant to the pathogenic mechanism of a broad array of TDP-43 proteinopathies, including frontotemporal lobar degeneration and amyotrophic lateral sclerosis.


Subject(s)
Cell Cycle Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Frontotemporal Dementia/genetics , Mutation/genetics , Osteitis Deformans/genetics , Aminopeptidases/metabolism , Animals , Animals, Genetically Modified , Binding Sites/genetics , Cell Line, Transformed , Central Nervous System/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA-Binding Proteins/genetics , Disease Models, Animal , Drosophila , Drosophila Proteins/metabolism , Frontotemporal Dementia/complications , Frontotemporal Dementia/pathology , Gene Expression Regulation/genetics , Glycoproteins/metabolism , Humans , Indoles , Models, Biological , Osteitis Deformans/complications , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Saccharomyces cerevisiae/genetics , Transfection/methods , Valosin Containing Protein
12.
Hum Mol Genet ; 19(9): 1741-55, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20147319

ABSTRACT

Inclusion body myopathy associated with Paget's disease of bone and frontotemporal dementia (IBMPFD) is a dominantly inherited degenerative disorder caused by mutations in the valosin-containing protein (VCP) gene. VCP (p97 in mouse, TER94 in Drosophila melanogaster and CDC48 in Saccharomyces cerevisiae) is a highly conserved AAA(+)-ATPase that regulates a wide array of cellular processes. The mechanism of IBMPFD pathogenesis is unknown. Towards elucidating the pathogenic mechanism we have developed and characterized transgenic mice with ubiquitous expression of wild-type and disease-causing versions of human VCP/p97. Here, we report that mice expressing VCP/p97 harboring the mutations R155H or A232E develop pathology that is limited to muscle, brain and bone, recapitulating the spectrum of disease in humans with IBMPFD. The mice exhibit progressive muscle weakness and pathological examination of muscle shows classic characteristics of inclusion body myopathy including rimmed vacuoles and TDP-43 pathology. The mice exhibit abnormalities in behavioral testing and pathological examination of the brain shows widespread TDP-43 pathology. Furthermore, radiological examination of the skeleton reveals that mutant mice develop severe osteopenia accompanied by focal lytic and sclerotic lesions in vertebrae and femur. In vitro studies indicate that mutant VCP causes inappropriate activation of the NF-kappaB signaling cascade, which could contribute to the mechanism of pathogenesis in multiple tissues including muscle, bone and brain.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Disease Models, Animal , Frontotemporal Dementia/genetics , Myositis, Inclusion Body/genetics , Osteitis Deformans/genetics , Animals , Blotting, Western , Brain/pathology , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/complications , Frontotemporal Dementia/pathology , Immunohistochemistry , Maze Learning , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Muscle, Skeletal/ultrastructure , Mutation, Missense/genetics , Myositis, Inclusion Body/complications , Myositis, Inclusion Body/pathology , Osteitis Deformans/complications , Osteitis Deformans/pathology , Saccharomyces cerevisiae Proteins , Valosin Containing Protein
13.
Nat Neurosci ; 9(10): 1302-11, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16936724

ABSTRACT

Non-neuronal cells may be pivotal in neurodegenerative disease, but the mechanistic basis of this effect remains ill-defined. In the polyglutamine disease spinocerebellar ataxia type 7 (SCA7), Purkinje cells undergo non-cell-autonomous degeneration in transgenic mice. We considered the possibility that glial dysfunction leads to Purkinje cell degeneration, and generated mice that express ataxin-7 in Bergmann glia of the cerebellum with the Gfa2 promoter. Bergmann glia-specific expression of mutant ataxin-7 was sufficient to produce ataxia and neurodegeneration. Expression of the Bergmann glia-specific glutamate transporter GLAST was reduced in Gfa2-SCA7 mice and was associated with impaired glutamate transport in cultured Bergmann glia, cerebellar slices and cerebellar synaptosomes. Ultrastructural analysis of Purkinje cells revealed findings of dark cell degeneration consistent with excitotoxic injury. Our studies indicate that impairment of glutamate transport secondary to glial dysfunction contributes to SCA7 neurodegeneration, and suggest a similar role for glial dysfunction in other polyglutamine diseases and SCAs.


Subject(s)
Amino Acid Transport System X-AG/metabolism , Gene Expression/physiology , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neuroglia/metabolism , Age Factors , Aged , Animals , Animals, Newborn , Ataxin-7 , Behavior, Animal , Blotting, Western/methods , Brain/pathology , Cells, Cultured , Female , Glial Fibrillary Acidic Protein/metabolism , Glutamic Acid/metabolism , Humans , Immunohistochemistry/methods , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Nerve Tissue Proteins/genetics , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Neuroglia/ultrastructure , Transfection/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...