Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neurobiol Dis ; 50: 127-34, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23064434

ABSTRACT

Deposition of amyloid ß (Aß) containing plaques in the brain is one of the neuropathological hallmarks of Alzheimer's disease (AD). It has been suggested that modulation of neuronal activity may alter Aß production in the brain. We postulate that these changes in Aß production are due to changes in the rate-limiting step of Aß generation, APP cleavage by γ-secretase. By combining biochemical approaches with fluorescence lifetime imaging microscopy, we found that neuronal inhibition decreases endogenous APP and PS1 interactions, which correlates with reduced Aß production. By contrast, neuronal activation had a two-phase effect: it initially enhanced APP-PS1 interaction leading to increased Aß production, which followed by a decrease in the APP and PS1 proximity/interaction. Accordingly, treatment of neurons with naturally secreted Aß isolated from AD brain or with synthetic Aß resulted in reduced APP and PS1 proximity. Moreover, applying low concentration of Aß(42) to cultured neurons inhibited de novo Aß synthesis. These data provide evidence that neuronal activity regulates endogenous APP-PS1 interactions, and suggest a model of a product-enzyme negative feedback. Thus, under normal physiological conditions Aß may impact its own production by modifying γ-secretase cleavage of APP. Disruption of this negative modulation may cause Aß overproduction leading to neurotoxicity.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Neurons/metabolism , Presenilin-1/metabolism , Alzheimer Disease/pathology , Animals , Blotting, Western , Feedback, Physiological , Humans , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Transgenic , Neurons/pathology
2.
J Clin Invest ; 121(10): 4070-81, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21926467

ABSTRACT

Aging is a major risk factor for the progression of neurodegenerative diseases, including Huntington disease (HD). Reduced neuronal IGF1 or Irs2 signaling have been shown to extend life span in mice. To determine whether Irs2 signaling modulates neurodegeneration in HD, we genetically modulated Irs2 concentrations in the R6/2 mouse model of HD. Increasing Irs2 levels in the brains of R6/2 mice significantly reduced life span and increased neuronal oxidative stress and mitochondrial dysfunction. In contrast, reducing Irs2 levels throughout the body (except in ß cells, where Irs2 expression is needed to prevent diabetes onset; R6/2•Irs2+/-•Irs2ßtg mice) improved motor performance and extended life span. The slower progression of HD-like symptoms was associated with increased nuclear localization of the transcription factor FoxO1 and increased expression of FoxO1-dependent genes that promote autophagy, mitochondrial function, and resistance to oxidative stress. Mitochondrial function improved and the number of autophagosomes increased in R6/2•Irs2+/-•Irs2ßtg mice, whereas aggregate formation and oxidative stress decreased. Thus, our study suggests that Irs2 signaling can modulate HD progression. Since we found the expression of Irs2 to be normal in grade II HD patients, our results suggest that decreasing IRS2 signaling could be part of a therapeutic approach to slow the progression of HD.


Subject(s)
Huntington Disease/physiopathology , Insulin Receptor Substrate Proteins/physiology , Mitochondria/physiology , Aging/genetics , Aging/physiology , Animals , Brain/pathology , Brain/physiopathology , Disease Models, Animal , Disease Progression , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Gene Expression , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Insulin Receptor Substrate Proteins/deficiency , Insulin Receptor Substrate Proteins/genetics , Longevity/genetics , Longevity/physiology , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , Oxidative Stress , Signal Transduction
3.
Neuron ; 67(6): 936-52, 2010 Sep 23.
Article in English | MEDLINE | ID: mdl-20869592

ABSTRACT

Spinobulbar muscular atrophy (SBMA) is a neurodegenerative disease caused by expansion of a polyglutamine tract in the androgen receptor (AR). This mutation confers toxic function to AR through unknown mechanisms. Mutant AR toxicity requires binding of its hormone ligand, suggesting that pathogenesis involves ligand-induced changes in AR. However, whether toxicity is mediated by native AR function or a novel AR function is unknown. We systematically investigated events downstream of ligand-dependent AR activation in a Drosophila model of SBMA. We show that nuclear translocation of AR is necessary, but not sufficient, for toxicity and that DNA binding by AR is necessary for toxicity. Mutagenesis studies demonstrated that a functional AF-2 domain is essential for toxicity, a finding corroborated by a genetic screen that identified AF-2 interactors as dominant modifiers of degeneration. These findings indicate that SBMA pathogenesis is mediated by misappropriation of native protein function, a mechanism that may apply broadly to polyglutamine diseases.


Subject(s)
Muscular Disorders, Atrophic/etiology , Muscular Disorders, Atrophic/genetics , Mutation/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Blindness/genetics , Blindness/pathology , Cell Line, Transformed , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Eye/metabolism , Eye/pathology , Female , Furylfuramide/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Humans , Larva/physiology , Locomotion/genetics , Motor Neurons/metabolism , Muscular Disorders, Atrophic/pathology , Mutagenesis/physiology , Neuromuscular Junction/pathology , Oligonucleotide Array Sequence Analysis/methods , Phenotype , Principal Component Analysis , Protein Transport/genetics , RNA Interference/physiology , Receptors, Androgen/chemistry , Salivary Glands/metabolism , Salivary Glands/pathology , Statistics, Nonparametric , Transfection/methods , Trinucleotide Repeat Expansion , Tubulin/metabolism
4.
J Neurosci Res ; 88(10): 2207-16, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20336775

ABSTRACT

Expanded polyglutamine tracts cause neurodegeneration through a toxic gain-of-function mechanism. Generation of inclusions is a common feature of polyglutamine diseases and other protein misfolding disorders. Inclusion formation is likely to be a defensive response of the cell to the presence of unfolded protein. Recently, the compound B2 has been shown to increase inclusion formation and decrease toxicity of polyglutamine-expanded huntingtin in cultured cells. We explored the effect of B2 on spinal and bulbar muscular atrophy (SBMA). SBMA is caused by expansion of polyglutamine in the androgen receptor (AR) and is characterized by the loss of motor neurons in the brainstem and spinal cord. We found that B2 increases the deposition of mutant AR into nuclear inclusions, without altering the ligand-induced aggregation, expression, or subcellular distribution of the mutant protein. The effect of B2 on inclusions was associated with a decrease in AR transactivation function. We show that B2 reduces mutant AR toxicity in cell and fly models of SBMA, further supporting the idea that accumulation of polyglutamine-expanded protein into inclusions is protective. Our findings suggest B2 as a novel approach to therapy for SBMA.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked/drug therapy , Bulbo-Spinal Atrophy, X-Linked/metabolism , Neuroprotective Agents/pharmacology , Nitroquinolines/pharmacology , Peptides/metabolism , Piperazines/pharmacology , Receptors, Androgen/metabolism , Animals , Animals, Genetically Modified , Cell Line , Disease Models, Animal , Drosophila melanogaster , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Intranuclear Inclusion Bodies/drug effects , Intranuclear Inclusion Bodies/metabolism , Ligands , Mutation , Protein Multimerization , Rats , Receptors, Androgen/genetics
5.
Neuron ; 63(3): 316-28, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19679072

ABSTRACT

Expansion of a polyglutamine tract in the androgen receptor (AR) causes spinal and bulbar muscular atrophy (SBMA). We previously showed that Akt-mediated phosphorylation of AR reduces ligand binding and attenuates the mutant AR toxicity. Here, we show that in culture insulin-like growth factor 1 (IGF-1) reduces AR aggregation and increases AR clearance via the ubiquitin-proteasome system through phosphorylation of AR by Akt. In vivo, SBMA transgenic mice overexpressing a muscle-specific isoform of IGF-1 selectively in skeletal muscle show evidence of increased Akt activation and AR phosphorylation and decreased AR aggregation. Augmentation of IGF-1/Akt signaling rescues behavioral and histopathological abnormalities, extends the life span, and reduces both muscle and spinal cord pathology of SBMA mice. This study establishes IGF-1/Akt-mediated inactivation of mutant AR as a strategy to counteract disease in vivo and demonstrates that skeletal muscle is a viable target tissue for therapeutic intervention in SBMA.


Subject(s)
Gene Expression Regulation/genetics , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Cell Line, Transformed , Chlorocebus aethiops , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscular Atrophy, Spinal/mortality , Muscular Atrophy, Spinal/therapy , Mutation/genetics , Oncogene Protein v-akt/metabolism , Peptides/genetics , Peptides/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Serine/metabolism , Time Factors , Transfection/methods , Trinucleotide Repeat Expansion/drug effects , Trinucleotide Repeat Expansion/physiology , Ubiquitin/metabolism
6.
Hum Mol Genet ; 18(R1): R40-7, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19297400

ABSTRACT

Polyglutamine diseases are a family of nine neurodegenerative disorders caused by expansion in different genes of a CAG triplet repeat stretch, which encodes an elongated polyglutamine tract. This polyglutamine tract is thought to confer a toxic gain of function to the bearing proteins, which leads to late onset and progressive loss of neurons in specific regions of the central nervous system. The mechanisms underlying specificity for neuronal vulnerability remain enigmatic. One explanation is that the polyglutamine tract is not the only determinant of neurodegeneration and that protein context and post-translational events may also be crucial for pathogenesis. Here, we review how post-translational modifications of the polyglutamine proteins contribute to modulate neurotoxicity.


Subject(s)
Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Peptides/metabolism , Protein Processing, Post-Translational , Animals , Humans
7.
J Steroid Biochem Mol Biol ; 108(3-5): 245-53, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17945479

ABSTRACT

The androgen receptor (AR) is a ligand-activated transcription factor which is responsible for the androgen responsiveness of target cells. Several types of mutations have been found in the AR and linked to endocrine dysfunctions. Surprisingly, the polymorphism involving the CAG triplet repeat expansion of the AR gene, coding for a polyglutamine (PolyGln) tract in the N-terminal transactivation domain of the AR protein, has been involved either in endocrine or neurological disorders. For example, among endocrine-related-diseases, the PolyGln size has been proposed to be associated to prostate cancer susceptibility, hirsutism, male infertility, cryptorchidism (in conjunction with polyglycine stretches polymorphism), etc.; the molecular mechanisms of these alterations are thought to involve a modulation of AR transcriptional competence, which inversely correlates with the PolyGln length. Among neurological alterations, a decreased AR function seems to be also involved in depression. Moreover, when the polymorphic PolyGln becomes longer than 35-40 contiguous glutamines (ARPolyGln), the ARPolyGln acquires neurotoxicity, because of an unknown gain-of-function. This mutation has been linked to a rare inherited X-linked motor neuronal disorder, the Spinal and Bulbar Muscular Atrophy, or Kennedy's disease. The disorder is characterized by death of motor neurons expressing high levels of AR. The degenerating motor neurons are mainly located in the anterior horns of the spinal cord and in the bulbar region; some neurons of the dorsal root ganglia may also be involved. Interestingly, the same type of PolyGln elongation has been found in other totally unrelated proteins responsible for different neurodegenerative diseases. A common feature of all these disorders is the formation of intracellular aggregates containing the mutated proteins; at present, but their role in the disease is largely debated. This review will discuss how the PolyGln neurotoxicity of SBMA AR may be either mediated or decreased by aggregates, and will present data on the dual role played by testosterone on motor neuronal functions and dysfunctions.


Subject(s)
Peptides/physiology , Proteasome Endopeptidase Complex/physiology , Receptors, Androgen/chemistry , Humans , Motor Neurons/pathology , Muscular Disorders, Atrophic/genetics , Polymorphism, Genetic , Protein Structure, Quaternary , Receptors, Androgen/genetics , Trinucleotide Repeat Expansion
8.
Hum Mol Genet ; 16(13): 1604-18, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17504823

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease caused by motoneuron loss. Some familial cases (fALS) are linked to mutations of superoxide dismutase type-1 (SOD1), an antioxidant enzyme whose activity is preserved in most mutant forms. Owing to the similarities in sporadic and fALS forms, mutant SOD1 animal and cellular models are a useful tool to study the disease. In transgenic mice expressing either wild-type (wt) human SOD1 or mutant G93A-SOD1, we found that wtSOD1 was present in cytoplasm and in nuclei of motoneurons, whereas mutant SOD1 was mainly cytoplasmic. Similar results were obtained in immortalized motoneurons (NSC34 cells) expressing either wtSOD1 or G93A-SOD1. Analyzing the proteasome activity, responsible for misfolded protein clearance, in the two subcellular compartments, we found proteasome impairment only in the cytoplasm. The effect of G93A-SOD1 exclusion from nuclei was then analyzed after oxidative stress. Cells expressing G93A-SOD1 showed a higher DNA damage compared with those expressing wtSOD1, possibly because of a loss of nuclear protection. The toxicity of mutant SOD1 might, therefore, arise from an initial misfolding (gain of function) reducing nuclear protection from the active enzyme (loss of function in the nuclei), a process that may be involved in ALS pathogenesis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Mutation , Superoxide Dismutase/genetics , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Damage , Gene Expression Regulation, Enzymologic , Mice , Mice, Transgenic , Microscopy, Fluorescence , Oxidative Stress , Oxygen/metabolism , Proteasome Endopeptidase Complex/metabolism , Spinal Cord/metabolism
9.
Hum Mol Genet ; 16(13): 1593-603, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17470458

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is a progressive neurodegenerative disease caused by an expansion of the polyglutamine tract in the androgen receptor (AR). Here, we investigated the regulation of AR phosphorylation in order to understand factors that may modify SBMA disease progression. We show that expanded polyglutamine AR is phosphorylated by Akt. Substitution of the AR at two Akt consensus sites, S215 and S792, with aspartate, which mimics phosphorylation, reduces ligand binding, ligand-dependent nuclear translocation, transcriptional activation and toxicity of expanded polyglutamine AR. Co-expression of constitutively active Akt and the AR has similar consequences, which are blocked by alanine substitutions at residues 215 and 792. Furthermore, in motor neuron-derived MN-1 cells toxicity associated with polyglutamine-expanded AR is rescued by co-expression with Akt. Insulin-like growth factor-1 (IGF-1) stimulation, which activates several cell survival promoting pathways, also reduces toxicity of the expanded polyglutamine AR in MN-1 cells, in a manner dependent upon phospho-inositol-3-kinase. IGF-1 rescue of AR toxicity is diminished by alanine substitutions at the Akt consensus sites. These results highlight potential targets for therapeutic intervention in SBMA.


Subject(s)
DNA Repeat Expansion/genetics , Peptides/metabolism , Proto-Oncogene Proteins c-akt/genetics , Receptors, Androgen/metabolism , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Humans , Insulin-Like Growth Factor I/metabolism , Ligands , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Sequence Homology, Amino Acid , Transcriptional Activation
10.
Neurobiol Aging ; 28(7): 1099-111, 2007 Jul.
Article in English | MEDLINE | ID: mdl-16781019

ABSTRACT

Aggregates, a hallmark of most neurodegenerative diseases, may have different properties, and possibly different roles in neurodegeneration. We analysed ubiquitin-proteasome pathway functions during cytoplasmic aggregation in polyglutamine (polyQ) diseases, using a unique model of motor neuron disease, the SpinoBulbar Muscular Atrophy. The disease, which is linked to a polyQ tract elongation in the androgen receptor (ARpolyQ), has the interesting feature that ARpolyQ aggregation is triggered by the AR ligand, testosterone. Using immortalized motor neurons expressing ARpolyQ, we found that a proteasome reporter, YFPu, accumulated in absence of aggregates; testosterone treatment, which induced ARpolyQ aggregation, allowed the normal clearance of YFPu, suggesting that aggregation contributed to proteasome de-saturation, an effect not related to AR nuclear translocation. Using AR antagonists to modulate the kinetic of ARpolyQ aggregation, we demonstrated that aggregation, by removing the neurotoxic protein from the soluble compartment, protected the proteasome from an excess of misfolded protein to be processed.


Subject(s)
Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptors, Androgen/genetics , Trinucleotide Repeat Expansion/genetics , Animals , Cell Line, Transformed , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Motor Neurons/drug effects , Mutation , Peptides/genetics , Protein Transport/drug effects , Protein Transport/genetics , RNA, Messenger/biosynthesis , Receptors, Androgen/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Testosterone/pharmacology , Transfection/methods , Ubiquitin-Protein Ligase Complexes
SELECTION OF CITATIONS
SEARCH DETAIL
...