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










Publication year range
1.
J Pharm Sci ; 113(1): 131-140, 2024 01.
Article in English | MEDLINE | ID: mdl-37659717

ABSTRACT

INTRODUCTION: Recombinant adeno-associated viruses(rAAVs) are an attractive tool to ensure long-term expression monoclonal antibody(mAb) in the central nervous system(CNS). It is still unclear whether systemic injection or local CNS administration of AAV9 is more beneficial for the exposure of the expressed mAb in the brain. Hence, we compared the biodistribution and transgene expression following AAV9-Trastuzumab administration through different routes. METHODS AND RESULT: In-house generated AAV9-Trastuzumab vectors were administered at 5E+11 Vgs/rat through intravenous(IV), intracerebroventricular(ICV), intra-cisterna magna(ICM) and intrastriatal(IST) routes. Vector and trastuzumab blood/plasma concentrations were assessed at different time points up to the terminal time point of 21 days. Different brain regions in addition to the spinal cord, cerebrospinal fluid(CSF) and interstitial fluid(ISF), were also analyzed at the terminal time point. Our results show that vector biodistribution and Trastuzumab expression in the brain could the ranked as follows: IST>ICM>ICV>IV. Rapid clearance of vector was observed after administration via the ICM and ICV routes. The ICV route produced similar expression levels across different brain regions, while the ICM route had better expression in the hindbrain and spinal cord region. The IST route had higher expression in the forebrain region compared to the hindbrain region. A sharp decline in trastuzumab plasma concentration was observed across all routes of administration due to anti-trastuzumab antibody response. CONCLUSION: In this study we have characterized vector biodistribution and transgene mAb expression after AAV9 vector administration through different routes in rats. IST and ICM represent the best administration routes to deliver antibody genes to the brain.


Subject(s)
Brain , Genetic Therapy , Rats , Animals , Transduction, Genetic , Genetic Therapy/methods , Tissue Distribution , Trastuzumab , Brain/metabolism , Genetic Vectors
2.
Gene Ther ; 30(1-2): 132-141, 2023 02.
Article in English | MEDLINE | ID: mdl-35637286

ABSTRACT

Challenges in obtaining efficient transduction of brain and spinal cord following systemic AAV delivery have led to alternative administration routes being used in clinical trials that directly infuse the virus into the CNS. However, data comparing different direct AAV injections into the brain remain limited making it difficult to choose optimal routes. Here we tested both AAV9-egfp and AAV9-fLuc delivery via intrastriatal (IST), intracisterna magna (ICM) and lumbar intrathecal (LIT) routes in adult rats and assessed vector distribution and transduction in brain, spinal cord and peripheral tissues. We find that IST infusion leads to robust transgene expression in the striatum, thalamus and cortex with lower peripheral tissue transduction and anti-AAV9 capsid titers compared to ICM or LIT. ICM delivery provided strong GFP and luciferase expression across more brain regions than the other routes and similar expression in the spinal cord to LIT injections, which itself largely failed to transduce the rat brain. Our data highlight the strengths and weaknesses of each direct CNS delivery route which will help with future clinical targeting.


Subject(s)
Gene Transfer Techniques , Spinal Cord , Rats , Animals , Transduction, Genetic , Spinal Cord/metabolism , Brain/metabolism , Transgenes , Genetic Vectors/genetics , Dependovirus/genetics , Dependovirus/metabolism
3.
Adv Drug Deliv Rev ; 170: 214-237, 2021 03.
Article in English | MEDLINE | ID: mdl-33486008

ABSTRACT

While protein therapeutics are one of the most successful class of drug molecules, they are expensive and not suited for treating chronic disorders that require long-term dosing. Adeno-associated virus (AAV) mediated in vivo gene therapy represents a viable alternative, which can deliver the genes of protein therapeutics to produce long-term expression of proteins in target tissues. Ongoing clinical trials and recent regulatory approvals demonstrate great interest in these therapeutics, however, there is a lack of understanding regarding their cellular disposition, whole-body disposition, dose-exposure relationship, exposure-response relationship, and how product quality and immunogenicity affects these important properties. In addition, there is a lack of quantitative studies to support the development of pharmacokinetic-pharmacodynamic models, which can support the discovery, development, and clinical translation of this delivery system. In this review, we have provided a state-of-the-art overview of current progress and limitations related to AAV mediated delivery of protein therapeutic genes, along with our perspective on the steps that need to be taken to improve clinical translation of this therapeutic modality.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Proteins/genetics , Humans , Models, Molecular , Proteins/chemistry , Proteins/pharmacokinetics
4.
Sci Rep ; 7(1): 14766, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116194

ABSTRACT

Adeno-associated viruses (AAVs) are attractive gene therapy vectors due to their low toxicity, high stability, and rare integration into the host genome. Expressing ligands on the viral capsid can re-target AAVs to new cell types, but limited sites have been identified on the capsid that tolerate a peptide insertion. Here, we incorporated a site-specific tetracysteine sequence into the AAV serotype 9 (AAV9) capsid, to permit labelling of viral particles with either a fluorescent dye or biotin. We demonstrate that fluorescently labelled particles are detectable in vitro, and explore the utility of the method in vivo in mice with time-lapse imaging. We exploit the biotinylated viral particles to generate two distinct AAV interactomes, and identify several functional classes of proteins that are highly represented: actin/cytoskeletal protein binding, RNA binding, RNA splicing/processing, chromatin modifying, intracellular trafficking and RNA transport proteins. To examine the biological relevance of the capsid interactome, we modulated the expression of two proteins from the interactomes prior to AAV transduction. Blocking integrin αVß6 receptor function reduced AAV9 transduction, while reducing histone deacetylase 4 (HDAC4) expression enhanced AAV transduction. Our method demonstrates a strategy for inserting motifs into the AAV capsid without compromising viral titer or infectivity.


Subject(s)
Capsid/metabolism , Dependovirus/genetics , Mutation , Optical Imaging/methods , Virion/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm , Cell Line, Tumor , Cysteine/metabolism , Genetic Vectors , HEK293 Cells , Histone Deacetylases , Humans , Integrins/antagonists & inhibitors , Maleimides/chemistry , Mice , Repressor Proteins/antagonists & inhibitors , Sequence Homology, Amino Acid , Transduction, Genetic , Viral Proteins/chemistry , Viral Proteins/metabolism
5.
Adv Exp Med Biol ; 1007: 241-257, 2017.
Article in English | MEDLINE | ID: mdl-28840561

ABSTRACT

Genetic disorders, caused by deleterious changes in the DNA sequence away from the normal genomic sequence, affect millions of people worldwide. Gene therapy as a treatment option for patients is an attractive proposition due to its conceptual simplicity. In principle, gene therapy involves correcting the genetic disorder by either restoring a normal functioning copy of a gene or reducing the toxicity arising from a mutated gene. In this way specific genetic function can be restored without altering the expression of other genes and the proteins they encode. The reality however is much more complex, and as a result the vector systems used to deliver gene therapies have by necessity continued to evolve and improve over time with respect to safety profile, efficiency, and long-term expression. In this chapter we examine the current approaches to gene therapy, assess the different gene delivery systems utilized, and highlight the failures and successes of relevant clinical trials. We do not intend for this chapter to be a comprehensive and exhaustive assessment of all clinical trials that have been conducted in the CNS, but instead will focus on specific diseases that have seen successes and failures with different gene therapy vehicles to gauge how preclinical models have informed the design of clinical trials.


Subject(s)
Gene Transfer Techniques , Genetic Diseases, Inborn/therapy , Genetic Therapy/methods , Animals , Genetic Diseases, Inborn/genetics , Humans
6.
Nat Neurosci ; 20(9): 1225-1235, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28714954

ABSTRACT

Hexanucleotide repeat expansions represent the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia, though the mechanisms by which such expansions cause neurodegeneration are poorly understood. We report elevated levels of DNA-RNA hybrids (R-loops) and double strand breaks in rat neurons, human cells and C9orf72 ALS patient spinal cord tissues. Accumulation of endogenous DNA damage is concomitant with defective ATM-mediated DNA repair signaling and accumulation of protein-linked DNA breaks. We reveal that defective ATM-mediated DNA repair is a consequence of P62 accumulation, which impairs H2A ubiquitylation and perturbs ATM signaling. Virus-mediated expression of C9orf72-related RNA and dipeptide repeats in the mouse central nervous system increases double strand breaks and ATM defects and triggers neurodegeneration. These findings identify R-loops, double strand breaks and defective ATM-mediated repair as pathological consequences of C9orf72 expansions and suggest that C9orf72-linked neurodegeneration is driven at least partly by genomic instability.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Chromosome Breakage , DNA Repair/physiology , DNA Repeat Expansion/physiology , Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , C9orf72 Protein , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Proteins/metabolism , Random Allocation , Rats , Spinal Cord/metabolism , Spinal Cord/pathology
7.
Nat Commun ; 8: 16063, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28677678

ABSTRACT

Hexanucleotide repeat expansions in the C9ORF72 gene are the commonest known genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia. Expression of repeat transcripts and dipeptide repeat proteins trigger multiple mechanisms of neurotoxicity. How repeat transcripts get exported from the nucleus is unknown. Here, we show that depletion of the nuclear export adaptor SRSF1 prevents neurodegeneration and locomotor deficits in a Drosophila model of C9ORF72-related disease. This intervention suppresses cell death of patient-derived motor neuron and astrocytic-mediated neurotoxicity in co-culture assays. We further demonstrate that either depleting SRSF1 or preventing its interaction with NXF1 specifically inhibits the nuclear export of pathological C9ORF72 transcripts, the production of dipeptide-repeat proteins and alleviates neurotoxicity in Drosophila, patient-derived neurons and neuronal cell models. Taken together, we show that repeat RNA-sequestration of SRSF1 triggers the NXF1-dependent nuclear export of C9ORF72 transcripts retaining expanded hexanucleotide repeats and reveal a novel promising therapeutic target for neuroprotection.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , C9orf72 Protein/metabolism , Frontotemporal Dementia/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism , Adult , Aged , Amyotrophic Lateral Sclerosis/etiology , Animals , Astrocytes/physiology , Cell Line , Coculture Techniques , Disease Models, Animal , Drosophila , Female , Frontotemporal Dementia/etiology , Humans , Male , Mice , Middle Aged , Nuclear Proteins/metabolism , Rats , Transcription Factors/metabolism
8.
Dis Model Mech ; 10(7): 859-868, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28550099

ABSTRACT

Intronic GGGGCC repeat expansions in C9orf72 are the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Two major pathologies stemming from the hexanucleotide RNA expansions (HREs) have been identified in postmortem tissue: intracellular RNA foci and repeat-associated non-ATG dependent (RAN) dipeptides, although it is unclear how these and other hallmarks of disease contribute to the pathophysiology of neuronal injury. Here, we describe two novel lines of mice that overexpress either 10 pure or 102 interrupted GGGGCC repeats mediated by adeno-associated virus (AAV) and recapitulate the relevant human pathology and disease-related behavioural phenotypes. Similar levels of intracellular RNA foci developed in both lines of mice, but only mice expressing 102 repeats generated C9orf72 RAN pathology, neuromuscular junction (NMJ) abnormalities, dispersal of the hippocampal CA1, enhanced apoptosis, and deficits in gait and cognition. Neither line of mice, however, showed extensive TAR DNA-binding protein 43 (TDP-43) pathology or neurodegeneration. Our data suggest that RNA foci pathology is not a good predictor of C9orf72 RAN dipeptide formation, and that RAN dipeptides and NMJ dysfunction are drivers of C9orf72 disease pathogenesis. These AAV-mediated models of C9orf72-associated ALS/FTD will be useful tools for studying disease pathophysiology and developing new therapeutic approaches.


Subject(s)
Behavior, Animal , Brain/pathology , C9orf72 Protein/genetics , DNA Repeat Expansion/genetics , Dependovirus/metabolism , Gene Transfer Techniques , Animals , Biomarkers/metabolism , Brain/physiopathology , CA1 Region, Hippocampal/pathology , Cell Death , Cell Nucleus/metabolism , Cognition , Gait , HEK293 Cells , Humans , Mice , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Neurons/metabolism , Neurons/pathology , RNA/metabolism , Sequestosome-1 Protein/metabolism , Up-Regulation
9.
Neural Regen Res ; 10(4): 555-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26170811
10.
PLoS One ; 9(10): e108088, 2014.
Article in English | MEDLINE | ID: mdl-25272038

ABSTRACT

Disrupted in schizophrenia 1 (DISC1) is a risk factor for a spectrum of neuropsychiatric illnesses including schizophrenia, bipolar disorder, and major depressive disorder. Here we use two missense Disc1 mouse mutants, described previously with distinct behavioural phenotypes, to demonstrate that Disc1 variation exerts differing effects on the formation of newly generated neurons in the adult hippocampus. Disc1 mice carrying a homozygous Q31L mutation, and displaying depressive-like phenotypes, have fewer proliferating cells while Disc1 mice with a homozygous L100P mutation that induces schizophrenia-like phenotypes, show changes in the generation, placement and maturation of newly generated neurons in the hippocampal dentate gyrus. Our results demonstrate Disc1 allele specific effects in the adult hippocampus, and suggest that the divergence in behavioural phenotypes may in part stem from changes in specific cell populations in the brain.


Subject(s)
Genetic Variation , Nerve Tissue Proteins/genetics , Neurogenesis/genetics , Animals , Cell Movement/genetics , Disease Models, Animal , Gene Expression , Male , Mice , Mice, Transgenic , Mutation, Missense , Neural Stem Cells/metabolism , Phenotype , Schizophrenia/genetics
11.
Mol Brain ; 2: 23, 2009 Jul 24.
Article in English | MEDLINE | ID: mdl-19630956

ABSTRACT

Dysfunction of alsin, particularly its putative Rab5 guanine-nucleotide-exchange factor activity, has been linked to one form of juvenile onset recessive familial amyotrophic lateral sclerosis (ALS2). Multiple lines of alsin knockout (ALS2(-/-)) mice have been generated to model this disease. However, it remains elusive whether the Rab5-dependent endocytosis is altered in ALS2(-/-) neurons. To directly examine the Rab5-mediated endosomal trafficking in ALS2(-/-) neurons, we introduced green fluorescent protein (GFP)-tagged Rab5 into cultured hippocampal neurons to monitor the morphology and motility of Rab5-associated early endosomes. Here we report that Rab5-mediated endocytosis was severely altered in ALS2(-/-) neurons. Excessive accumulation of Rab5-positive vesicles was observed in ALS2(-/-) neurons, which correlated with a significant reduction in endosomal motility and augmentation in endosomal conversion to lysosomes. Consequently, a significant increase in endosome/lysosome-dependent degradation of internalized glutamate receptors was observed in ALS2(-/-) neurons. These phenotypes closely resembled the endosomal trafficking abnormalities induced by a constitutively active form of Rab5 in wild-type neurons. Therefore, our findings reveal a negatively regulatory mechanism of alsin in Rab5-mediated endosomal trafficking, suggesting that enhanced endosomal degradation in ALS2(-/-) neurons may underlie the pathogenesis of motor neuron degeneration in ALS2 and related motor neuron diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Endosomes/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Protein Processing, Post-Translational , Amyotrophic Lateral Sclerosis/pathology , Animals , Endocytosis/drug effects , Endosomes/drug effects , Guanine Nucleotide Exchange Factors/deficiency , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Lysosomal Membrane Proteins/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Protein Processing, Post-Translational/drug effects , Protein Transport/drug effects , Receptors, Glutamate/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology , rab5 GTP-Binding Proteins/metabolism
12.
Neurodegener Dis ; 5(6): 359-66, 2008.
Article in English | MEDLINE | ID: mdl-18714162

ABSTRACT

Autosomal recessive mutations in the ALS2 gene have been linked to juvenile-onset amyotrophic lateral sclerosis (ALS2), primary lateral sclerosis and juvenile-onset ascending hereditary spastic paraplegia. Except for two recently identified missense mutations, all other mutations in the ALS2 gene lead to a premature stop codon and likely abrogate all the potential functions of alsin, the protein encoded by the ALS2 gene. To study the pathologic mechanisms of ALS2 deficiency, four different lines of ALS2 knockout (ALS2(-/-)) mice have been generated by independent groups. The loss of ALS2/alsin does not have a drastic effect on the survival or function of motor neurons in mice. However, subtle deficits observed in the behavior and pathology of these mice have aided in our understanding of the relationship between alsin and motor neuron dysfunction. In this review, we summarize and reconcile major findings of ALS2(-/-) mice and attempt to place these results within the larger context of modeling recessive movement disorders in mice.


Subject(s)
Disease Models, Animal , Guanine Nucleotide Exchange Factors/genetics , Motor Neuron Disease , Motor Neurons/physiology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/physiopathology , Animals , DNA Mutational Analysis , Endocytosis/physiology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Mice , Mice, Knockout , Motor Activity , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Motor Neuron Disease/pathology , Motor Neuron Disease/physiopathology , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism , Spastic Paraplegia, Hereditary/pathology , Spastic Paraplegia, Hereditary/physiopathology , rab5 GTP-Binding Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 105(29): 10244-9, 2008 Jul 22.
Article in English | MEDLINE | ID: mdl-18626009

ABSTRACT

Parkinson's disease (PD) is a major neurodegenerative condition with several rare Mendelian forms. Oxidative stress and mitochondrial function have been implicated in the pathogenesis of PD but the molecular mechanisms involved in the degeneration of neurons remain unclear. DJ-1 mutations are one cause of recessive parkinsonism, but this gene is also reported to be involved in cancer by promoting Ras signaling and suppressing PTEN-induced apoptosis. The specific function of DJ-1 is unknown, although it is responsive to oxidative stress and may play a role in the maintenance of mitochondria. Here, we show, using four independent methods, that DJ-1 associates with RNA targets in cells and the brain, including mitochondrial genes, genes involved in glutathione metabolism, and members of the PTEN/PI3K cascade. Pathogenic recessive mutants are deficient in this activity. We show that DJ-1 is sufficient for RNA binding at nanomolar concentrations. Further, we show that DJ-1 binds RNA but dissociates after oxidative stress. These data implicate a single mechanism for the pleiotropic effects of DJ-1 in different model systems, namely that the protein binds multiple RNA targets in an oxidation-dependent manner.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Oncogene Proteins/metabolism , Parkinsonian Disorders/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , Brain/metabolism , Cell Line , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Genes, Mitochondrial , Genes, Recessive , Glutathione/metabolism , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Molecular Sequence Data , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Oxidative Stress , PTEN Phosphohydrolase/metabolism , Parkinsonian Disorders/genetics , Peroxiredoxins , Phosphatidylinositol 3-Kinases/metabolism , Protein Deglycase DJ-1 , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
14.
J Neurosci ; 28(13): 3384-91, 2008 Mar 26.
Article in English | MEDLINE | ID: mdl-18367605

ABSTRACT

Parkinson's disease (PD), a progressive neurodegenerative disease characterized by bradykinesia, rigidity, and resting tremor, is the most common neurodegenerative movement disorder. Although the majority of PD cases are sporadic, some are inherited, including those caused by leucine-rich repeat kinase 2 (LRRK2) mutations. The substitution of serine for glycine at position 2019 (G2019S) in the kinase domain of LRRK2 represents the most prevalent genetic mutation in both familial and apparently sporadic cases of PD. Because mutations in LRRK2 are likely associated with a toxic gain of function, destabilization of LRRK2 may be a novel way to limit its detrimental effects. Here we show that LRRK2 forms a complex with heat shock protein 90 (Hsp90) in vivo and that inhibition of Hsp90 disrupts the association of Hsp90 with LRRK2 and leads to proteasomal degradation of LRRK2. Hsp90 inhibitors may therefore limit the mutant LRRK2-elicited toxicity to neurons. As a proof of principle, we show that Hsp90 inhibitors rescue the axon growth retardation caused by overexpression of the LRRK2 G2019S mutation in neurons. Therefore, inhibition of LRRK2 kinase activity can be achieved by blocking Hsp90-mediated chaperone activity and Hsp90 inhibitors may serve as potential anti-PD drugs.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Analysis of Variance , Animals , Animals, Newborn , Benzodioxoles/pharmacology , Brain/cytology , Cell Cycle Proteins/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Glycine/genetics , Humans , Immunoprecipitation/methods , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mass Spectrometry/methods , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/metabolism , Mutation/physiology , Neurons/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Purines/pharmacology , Serine/genetics , Silver Staining/methods , Transfection/methods
15.
Neuromolecular Med ; 10(1): 40-5, 2008.
Article in English | MEDLINE | ID: mdl-18246449

ABSTRACT

Mutations in DJ-1 cause inherited Parkinson's disease (PD) in several families. The normal function of DJ-1 is unknown, but mice lacking DJ-1 exhibit a deficit in dopaminergic signaling in the striatum. Since the hippocampus contains relatively high levels of DJ-1, and PD patients are often cognitively impaired, we evaluated the effects of DJ-1 deficiency on the plasticity of hippocampal CA1 synapses. LTP was slightly impaired and LTD was abolished in DJ-1-/- mice, whereas DJ-1+/- mice exhibited no alterations in synaptic plasticity. The dopamine receptor D2/3 agonist quinpirole rescued LTD in DJ-1-/- mice, suggesting a role for impaired dopaminergic signaling in the hippocampal LTD deficit.


Subject(s)
Hippocampus/physiology , Long-Term Synaptic Depression/physiology , Oncogene Proteins/physiology , Synapses/physiology , Animals , Dopamine D2 Receptor Antagonists , Humans , Long-Term Synaptic Depression/drug effects , Mice , Mice, Knockout , Oncogene Proteins/genetics , Peroxiredoxins , Protein Deglycase DJ-1 , Quinpirole/pharmacology , Receptors, Dopamine D3/drug effects , Synaptic Transmission
16.
Neurobiol Dis ; 29(3): 505-14, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18187333

ABSTRACT

Loss-of-function mutations in the DJ-1 gene account for an autosomal recessive form of Parkinson's disease (PD). To investigate the physiological functions of DJ-1 in vivo, we generated DJ-1 knockout (DJ-1(-/-)) mice. Younger (<1 year) DJ-1(-/-) mice were hypoactive and had mild gait abnormalities. Older DJ-1(-/-), however, showed decreased body weight and grip strength and more severe gait irregularities compared to wild-type littermates. The basal level of extracellular dopamine, evoked dopamine release and dopamine receptor D2 sensitivity appeared normal in the striatum of DJ-1(-/-) mice, which was consistent with similar results between DJ-1(-/-) and controls in behavioral paradigms specific for the dopaminergic system. An examination of spinal cord, nerve and muscle tissues failed to identify any pathological changes that were consistent with the noted motor deficits. Taken together, our findings suggest that loss of DJ-1 leads to progressive behavioral changes without significant alterations in nigrostriatal dopaminergic and spinal motor systems.


Subject(s)
Behavior, Animal/physiology , Corpus Striatum/physiology , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Substantia Nigra/physiology , Animals , Disease Progression , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology
17.
J Neurosci ; 27(51): 13982-90, 2007 Dec 19.
Article in English | MEDLINE | ID: mdl-18094236

ABSTRACT

The G59S missense mutation at the conserved microtubule-binding domain of p150(glued), a major component of dynein/dynactin complex, has been linked to an autosomal dominant form of motor neuron disease (MND). To study how this mutation affects the function of the dynein/dynactin complex and contributes to motor neuron degeneration, we generated p150(glued) G59S knock-in mice. We found that the G59S mutation destabilizes p150(glued) and disrupts the function of dynein/dynactin complex, resulting in early embryonic lethality of homozygous knock-in mice. Heterozygous knock-in mice, which developed normally, displayed MND-like phenotypes after 10 months of age, including excessive accumulation of cytoskeletal and synaptic vesicle proteins at neuromuscular junctions, loss of spinal motor neurons, increase of reactive astrogliosis, and shortening of gait compared with wild-type littermates and age-matched p150(glued) heterozygous knock-out mice. Our findings indicate that the G59S mutation in p150(glued) abrogates the normal function of p150(glued) and accelerates motor neuron degeneration.


Subject(s)
Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/physiology , Motor Neuron Disease/genetics , Mutation, Missense , Animals , Dynactin Complex , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Motor Neuron Disease/metabolism , Pregnancy
18.
Mol Neurobiol ; 36(3): 224-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17955197

ABSTRACT

Autosomal recessive mutations in the ALS2 gene lead to a clinical spectrum of motor dysfunction including juvenile onset amyotrophic lateral sclerosis (ALS2), primary lateral sclerosis, and hereditary spastic paraplegia. The 184-kDa alsin protein, encoded by the full-length ALS2 gene, contains three different guanine-nucleotide-exchange factor-like domains, which may play a role in the etiology of the disease. Multiple in vitro biochemical and cell biology assays suggest that alsin dysfunction affects endosome trafficking through a Rab5 small GTPase family-mediated mechanism. Four ALS2-deficient mouse models have been generated by different groups and used to study the behavioral and pathological impact of alsin deficiency. These mouse models largely fail to recapitulate hallmarks of motor neuron disease, but the subtle deficits that are observed in behavior and pathology have aided in our understanding of the relationship between alsin and motor dysfunction. In this review, we summarize recent clinical and molecular reports regarding alsin and attempt to place these results within the larger context of motor neuron disease.


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Motor Neuron Disease/genetics , Motor Neurons/physiology , Spastic Paraplegia, Hereditary/genetics , Animals , Guanine Nucleotide Exchange Factors/chemistry , Humans , Mice , Mice, Knockout , Models, Molecular , Mutation , Polymorphism, Single Nucleotide , Protein Conformation , Sequence Deletion
19.
PLoS Genet ; 3(6): e108, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17590087

ABSTRACT

We observed a severe autosomal recessive movement disorder in mice used within our laboratory. We pursued a series of experiments to define the genetic lesion underlying this disorder and to identify a cognate disease in humans with mutation at the same locus. Through linkage and sequence analysis we show here that this disorder is caused by a homozygous in-frame 18-bp deletion in Itpr1 (Itpr1(Delta18/Delta18)), encoding inositol 1,4,5-triphosphate receptor 1. A previously reported spontaneous Itpr1 mutation in mice causes a phenotype identical to that observed here. In both models in-frame deletion within Itpr1 leads to a decrease in the normally high level of Itpr1 expression in cerebellar Purkinje cells. Spinocerebellar ataxia 15 (SCA15), a human autosomal dominant disorder, maps to the genomic region containing ITPR1; however, to date no causal mutations had been identified. Because ataxia is a prominent feature in Itpr1 mutant mice, we performed a series of experiments to test the hypothesis that mutation at ITPR1 may be the cause of SCA15. We show here that heterozygous deletion of the 5' part of the ITPR1 gene, encompassing exons 1-10, 1-40, and 1-44 in three studied families, underlies SCA15 in humans.


Subject(s)
Inositol 1,4,5-Trisphosphate Receptors/genetics , Sequence Deletion , Spinocerebellar Ataxias/genetics , Animals , Base Sequence , Cell Line, Transformed , Female , Humans , Inositol 1,4,5-Trisphosphate Receptors/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...