Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
J Neurosci Res ; 102(1): e25278, 2024 01.
Article in English | MEDLINE | ID: mdl-38284836

ABSTRACT

Spinal and bulbar muscular atrophy (SBMA) is an X-linked disorder that affects males who inherit the androgen receptor (AR) gene with an abnormal CAG triplet repeat expansion. The resulting protein contains an elongated polyglutamine (polyQ) tract and causes motor neuron degeneration in an androgen-dependent manner. The precise molecular sequelae of SBMA are unclear. To assist with its investigation and the identification of therapeutic options, we report here a new model of SBMA in Drosophila melanogaster. We generated transgenic flies that express the full-length, human AR with a wild-type or pathogenic polyQ repeat. Each transgene is inserted into the same safe harbor site on the third chromosome of the fly as a single copy and in the same orientation. Expression of pathogenic AR, but not of its wild-type variant, in neurons or muscles leads to consistent, progressive defects in longevity and motility that are concomitant with polyQ-expanded AR protein aggregation and reduced complexity in neuromuscular junctions. Additional assays show adult fly eye abnormalities associated with the pathogenic AR species. The detrimental effects of pathogenic AR are accentuated by feeding flies the androgen, dihydrotestosterone. This new, robust SBMA model can be a valuable tool toward future investigations of this incurable disease.


Subject(s)
Bulbo-Spinal Atrophy, X-Linked , Drosophila , Adult , Humans , Male , Animals , Drosophila melanogaster , Androgens , Bulbo-Spinal Atrophy, X-Linked/genetics , Muscular Atrophy
2.
PLoS Genet ; 16(6): e1008778, 2020 06.
Article in English | MEDLINE | ID: mdl-32579604

ABSTRACT

Endurance exercise has broadly protective effects across organisms, increasing metabolic fitness and reducing incidence of several age-related diseases. Drosophila has emerged as a useful model for studying changes induced by chronic endurance exercise, as exercising flies experience improvements to various aspects of fitness at the cellular, organ and organismal level. The activity of octopaminergic neurons is sufficient to induce the conserved cellular and physiological changes seen following endurance training. All 4 octopamine receptors are required in at least one target tissue, but only one, Octß1R, is required for all of them. Here, we perform tissue- and adult-specific knockdown of alpha- and beta-adrenergic octopamine receptors in several target tissues. We find that reduced expression of Octß1R in adult muscles abolishes exercise-induced improvements in endurance, climbing speed, flight, cardiac performance and fat-body catabolism in male Drosophila. Importantly, Octß1R and OAMB expression in the heart is also required cell-nonautonomously for adaptations in other tissues, such as skeletal muscles in legs and adult fat body. These findings indicate that activation of distinct octopamine receptors in skeletal and cardiac muscle are required for Drosophila exercise adaptations, and suggest that cell non-autonomous factors downstream of octopaminergic activation play a key role.


Subject(s)
Adaptation, Physiological , Drosophila Proteins/metabolism , Flight, Animal , Heart/physiology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neurotransmitter/metabolism , Animals , Drosophila , Drosophila Proteins/genetics , Fat Body/metabolism , Muscle, Skeletal/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, Neurotransmitter/genetics
3.
Neurobiol Dis ; 160: 105516, 2021 12.
Article in English | MEDLINE | ID: mdl-34563642

ABSTRACT

Of the family of polyglutamine (polyQ) neurodegenerative diseases, Spinocerebellar Ataxia Type 3 (SCA3) is the most common. Like other polyQ diseases, SCA3 stems from abnormal expansions in the CAG triplet repeat of its disease gene resulting in elongated polyQ repeats within its protein, ataxin-3. Various ataxin-3 protein domains contribute to its toxicity, including the valosin-containing protein (VCP)-binding motif (VBM). We previously reported that VCP, a homo-hexameric protein, enhances pathogenic ataxin-3 aggregation and exacerbates its toxicity. These findings led us to explore the impact of targeting the SCA3 protein by utilizing a decoy protein comprising the N-terminus of VCP (N-VCP) that binds ataxin-3's VBM. The notion was that N-VCP would reduce binding of ataxin-3 to VCP, decreasing its aggregation and toxicity. We found that expression of N-VCP in Drosophila melanogaster models of SCA3 ameliorated various phenotypes, coincident with reduced ataxin-3 aggregation. This protective effect was specific to pathogenic ataxin-3 and depended on its VBM. Increasing the amount of N-VCP resulted in further phenotype improvement. Our work highlights the protective potential of targeting the VCP-ataxin-3 interaction in SCA3, a key finding in the search for therapeutic opportunities for this incurable disorder.


Subject(s)
Ataxin-3/metabolism , Machado-Joseph Disease/metabolism , Valosin Containing Protein/metabolism , Animals , Ataxin-3/genetics , Disease Models, Animal , Drosophila melanogaster , Machado-Joseph Disease/genetics , Phenotype , Protein Binding
4.
Neurobiol Dis ; 137: 104697, 2020 04.
Article in English | MEDLINE | ID: mdl-31783119

ABSTRACT

Spinocerebellar Ataxia type 3 (SCA3, also known as Machado-Joseph disease) is a neurodegenerative disorder caused by a CAG repeat expansion encoding an abnormally long polyglutamine (polyQ) tract in the disease protein, ataxin-3 (ATXN3). No preventive treatment is yet available for SCA3. Because SCA3 is likely caused by a toxic gain of ATXN3 function, a rational therapeutic strategy is to reduce mutant ATXN3 levels by targeting pathways that control its production or stability. Here, we sought to identify genes that modulate ATXN3 levels as potential therapeutic targets in this fatal disorder. We screened a collection of siRNAs targeting 2742 druggable human genes using a cell-based assay based on luminescence readout of polyQ-expanded ATXN3. From 317 candidate genes identified in the primary screen, 100 genes were selected for validation. Among the 33 genes confirmed in secondary assays, 15 were validated in an independent cell model as modulators of pathogenic ATXN3 protein levels. Ten of these genes were then assessed in a Drosophila model of SCA3, and one was confirmed as a key modulator of physiological ATXN3 abundance in SCA3 neuronal progenitor cells. Among the 15 genes shown to modulate ATXN3 in mammalian cells, orthologs of CHD4, FBXL3, HR and MC3R regulate mutant ATXN3-mediated toxicity in fly eyes. Further mechanistic studies of one of these genes, FBXL3, encoding a F-box protein that is a component of the SKP1-Cullin-F-box (SCF) ubiquitin ligase complex, showed that it reduces levels of normal and pathogenic ATXN3 in SCA3 neuronal progenitor cells, primarily via a SCF complex-dependent manner. Bioinformatic analysis of the 15 genes revealed a potential molecular network with connections to tumor necrosis factor-α/nuclear factor-kappa B (TNF/NF-kB) and extracellular signal-regulated kinases 1 and 2 (ERK1/2) pathways. Overall, we identified 15 druggable genes with diverse functions to be suppressors or enhancers of pathogenic ATXN3 abundance. Among identified pathways highlighted by this screen, the FBXL3/SCF axis represents a novel molecular pathway that regulates physiological levels of ATXN3 protein.


Subject(s)
Ataxin-3/genetics , Machado-Joseph Disease/genetics , Neurons/metabolism , Repressor Proteins/genetics , Humans , Machado-Joseph Disease/pathology , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/genetics
5.
J Neurosci Res ; 98(10): 2096-2108, 2020 10.
Article in English | MEDLINE | ID: mdl-32643791

ABSTRACT

Ataxin-3 is a deubiquitinase and polyglutamine disease protein whose cellular properties and functions are not entirely understood. Mutations in ataxin-3 cause spinocerebellar ataxia type 3 (SCA3), a neurodegenerative disorder that is a member of the polyglutamine family of diseases. Two major isoforms arise from alternative splicing of ATXN3 and are differently toxic in vivo as a result of faster proteasomal degradation of one isoform compared to the other. The isoforms vary only at their C-termini, suggesting that the hydrophobic C-terminus of the more quickly degraded form of ataxin-3 (here referred to as isoform 2) functions as a degron-that is, a peptide sequence that expedites the degradation of its host protein. We explored this notion in this study and present evidence that: (a) the C-terminus of ataxin-3 isoform 2 signals its degradation in a proteasome-dependent manner, (b) this effect from the C-terminus of isoform 2 does not require the ubiquitination of ataxin-3, and (c) the isolated C-terminus of isoform 2 can enhance the degradation of an unrelated protein. According to our data, the C-terminus of ataxin-3 isoform 2 is a degron, increasing overall understanding of the cellular properties of the SCA3 protein.


Subject(s)
Ataxin-3/genetics , Computer Simulation , Peptides/genetics , Repressor Proteins/genetics , Amino Acid Sequence , Ataxin-3/metabolism , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Peptides/metabolism , Repressor Proteins/metabolism , Ubiquitination/physiology
6.
Neurobiol Dis ; 132: 104535, 2019 12.
Article in English | MEDLINE | ID: mdl-31310802

ABSTRACT

The most commonly inherited dominant ataxia, Spinocerebellar Ataxia Type 3 (SCA3), is caused by a CAG repeat expansion that encodes an abnormally long polyglutamine (polyQ) repeat in the disease protein ataxin-3, a deubiquitinase. Two major full-length isoforms of ataxin-3 exist, both of which contain the same N-terminal portion and polyQ repeat, but differ in their C-termini; one (denoted here as isoform 1) contains a motif that binds ataxin-3's substrate, ubiquitin, whereas the other (denoted here as isoform 2) has a hydrophobic tail. Most SCA3 studies have focused on isoform 1, the predominant version in mammalian brain, yet both isoforms are present in brain and a better understanding of their relative pathogenicity in vivo is needed. We took advantage of the fruit fly, Drosophila melanogaster to model SCA3 and to examine the toxicity of each ataxin-3 isoform. Our assays reveal isoform 1 to be markedly more toxic than isoform 2 in all fly tissues. Reduced toxicity from isoform 2 is due to much lower protein levels as a result of its expedited degradation. Additional studies indicate that isoform 1 is more aggregation-prone than isoform 2 and that the C-terminus of isoform 2 is critical for its enhanced proteasomal degradation. According to our results, although both full-length, pathogenic ataxin-3 isoforms are toxic, isoform 1 is likely the primary contributor to SCA3 due to its presence at higher levels. Isoform 2, as a result of rapid degradation that is dictated by its tail, is unlikely to be a key player in this disease. Our findings provide new insight into the biology of this ataxia and the cellular processing of the underlying disease protein.


Subject(s)
Ataxin-3/genetics , Ataxin-3/toxicity , Drosophila Proteins/genetics , Drosophila Proteins/toxicity , Machado-Joseph Disease/genetics , Repressor Proteins/genetics , Repressor Proteins/toxicity , Amino Acid Sequence , Animals , Animals, Genetically Modified , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Humans , Machado-Joseph Disease/physiopathology , Protein Isoforms/genetics , Protein Isoforms/toxicity
7.
Hum Mol Genet ; 26(8): 1419-1431, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28158474

ABSTRACT

Polyglutamine (polyQ) repeat expansion in the deubiquitinase ataxin-3 causes neurodegeneration in Spinocerebellar Ataxia Type 3 (SCA3), one of nine inherited, incurable diseases caused by similar mutations. Ataxin-3's degradation is inhibited by its binding to the proteasome shuttle Rad23 through ubiquitin-binding site 2 (UbS2). Disrupting this interaction decreases levels of ataxin-3. Since reducing levels of polyQ proteins can decrease their toxicity, we tested whether genetically modulating the ataxin-3-Rad23 interaction regulates its toxicity in Drosophila. We found that exogenous Rad23 increases the toxicity of pathogenic ataxin-3, coincident with increased levels of the disease protein. Conversely, reducing Rad23 levels alleviates toxicity in this SCA3 model. Unexpectedly, pathogenic ataxin-3 with a mutated Rad23-binding site at UbS2, despite being present at markedly lower levels, proved to be more pathogenic than a disease-causing counterpart with intact UbS2. Additional studies established that the increased toxicity upon mutating UbS2 stems from disrupting the autoprotective role that pathogenic ataxin-3 has against itself, which depends on the co-chaperone, DnaJ-1. Our data reveal a previously unrecognized balance between pathogenic and potentially therapeutic properties of the ataxin-3-Rad23 interaction; they highlight this interaction as critical for the toxicity of the SCA3 protein, and emphasize the importance of considering protein context when pursuing suppressive avenues.


Subject(s)
Ataxin-3/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Machado-Joseph Disease/genetics , Nerve Degeneration/genetics , Repressor Proteins/genetics , Animals , Ataxin-3/metabolism , Binding Sites , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Disease Models, Animal , Drosophila melanogaster/genetics , Humans , Machado-Joseph Disease/metabolism , Machado-Joseph Disease/pathology , Molecular Chaperones/genetics , Nerve Degeneration/pathology , Peptides/genetics , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Repressor Proteins/metabolism , Ubiquitin/genetics
8.
Mol Cell ; 43(4): 599-612, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21855799

ABSTRACT

The mechanisms by which ubiquitin ligases are regulated remain poorly understood. Here we describe a series of molecular events that coordinately regulate CHIP, a neuroprotective E3 implicated in protein quality control. Through their opposing activities, the initiator E2, Ube2w, and the specialized deubiquitinating enzyme (DUB), ataxin-3, participate in initiating, regulating, and terminating the CHIP ubiquitination cycle. Monoubiquitination of CHIP by Ube2w stabilizes the interaction between CHIP and ataxin-3, which through its DUB activity limits the length of chains attached to CHIP substrates. Upon completion of substrate ubiquitination, ataxin-3 deubiquitinates CHIP, effectively terminating the reaction. Our results suggest that functional pairing of E3s with ataxin-3 or similar DUBs represents an important point of regulation in ubiquitin-dependent protein quality control. In addition, the results shed light on disease pathogenesis in SCA3, a neurodegenerative disorder caused by polyglutamine expansion in ataxin-3.


Subject(s)
Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Ubiquitin-Conjugating Enzymes/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Ataxin-3 , Humans , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stress, Physiological , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitination
9.
J Biol Chem ; 292(36): 14867-14884, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28710277

ABSTRACT

TMPRSS13 is a member of the type II transmembrane serine protease (TTSP) family. Although various TTSPs have been characterized in detail biochemically and functionally, the basic properties of TMPRSS13 remain unclear. Here, we investigate the activation, inhibition, post-translational modification, and localization of TMPRSS13. We show that TMPRSS13 is a glycosylated, active protease and that its own proteolytic activity mediates zymogen cleavage. Full-length, active TMPRSS13 exhibits impaired cell-surface expression in the absence of the cognate Kunitz-type serine protease inhibitors, hepatocyte growth factor activator inhibitor (HAI)-1 or HAI-2. Concomitant presence of TMPRSS13 with either HAI-1 or -2 mediates phosphorylation of residues in the intracellular domain of the protease, and it coincides with efficient transport of the protease to the cell surface and its subsequent shedding. Cell-surface labeling experiments indicate that the dominant form of TMPRSS13 on the cell surface is phosphorylated, whereas intracellular TMPRSS13 is predominantly non-phosphorylated. These data provide novel insight into the cellular properties of TMPRSS13 and highlight phosphorylation of TMPRSS13 as a novel post-translational modification of this TTSP family member and potentially other members of this family of proteases.


Subject(s)
Membrane Glycoproteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Proteinase Inhibitory Proteins, Secretory/metabolism , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Phosphorylation , Serine Endopeptidases/genetics
10.
Neurobiol Dis ; 116: 78-92, 2018 08.
Article in English | MEDLINE | ID: mdl-29704548

ABSTRACT

Among the nine dominantly inherited, age-dependent neurodegenerative diseases caused by abnormal expansion in the polyglutamine (polyQ) repeat of otherwise unrelated proteins is Spinocerebellar Ataxia Type 3 (SCA3). SCA3 is caused by polyQ expansion in the deubiquitinase (DUB), ataxin-3. Molecular sequelae related to SCA3 remain unclear. Here, we sought to understand the role of protein context in SCA3 by focusing on the interaction between this DUB and Valosin-Containing Protein (VCP). VCP is bound directly by ataxin-3 through an arginine-rich area preceding the polyQ repeat. We examined the importance of this interaction in ataxin-3-dependent degeneration in Drosophila melanogaster. Our assays with new isogenic fly lines expressing pathogenic ataxin-3 with an intact or mutated VCP-binding site show that disrupting the ataxin-3-VCP interaction delays the aggregation of the toxic protein in vivo. Importantly, early on flies that express pathogenic ataxin-3 with a mutated VCP-binding site are indistinguishable from flies that do not express any SCA3 protein. Also, reducing levels of VCP through RNA-interference has a similar, protective effect to mutating the VCP-binding site of pathogenic ataxin-3. Based on in vivo pulse-chases, aggregated species of ataxin-3 are highly stable, in a manner independent of VCP-binding. Collectively, our results highlight an important role for the ataxin-3-VCP interaction in SCA3, based on a model that posits a seeding effect from VCP on pathogenic ataxin-3 aggregation and subsequent toxicity.


Subject(s)
Ataxin-3/metabolism , Drosophila Proteins/metabolism , Peptides/metabolism , Protein Aggregates/physiology , Valosin Containing Protein/metabolism , Animals , Animals, Genetically Modified , Ataxin-3/genetics , Drosophila melanogaster , HEK293 Cells , Humans , Peptides/genetics , Protein Binding/physiology
11.
J Biol Chem ; 291(22): 11566-71, 2016 May 27.
Article in English | MEDLINE | ID: mdl-27129248

ABSTRACT

SIN3 is a transcriptional corepressor that acts as a scaffold for a histone deacetylase (HDAC) complex. The SIN3 complex regulates various biological processes, including organ development, cell proliferation, and energy metabolism. Little is known, however, about the regulation of SIN3 itself. There are two major isoforms of Drosophila SIN3, 187 and 220, which are differentially expressed. Intrigued by the developmentally timed exchange of SIN3 isoforms, we examined whether SIN3 187 controls the fate of the 220 counterpart. Here, we show that in developing tissue, there is interplay between SIN3 isoforms: when SIN3 187 protein levels increase, SIN3 220 protein decreases concomitantly. SIN3 187 has a dual effect on SIN3 220. Expression of 187 leads to reduced 220 transcript, while also increasing the turnover of SIN3 220 protein by the proteasome. These data support the presence of a novel, inter-isoform-dependent mechanism that regulates the amount of SIN3 protein, and potentially the level of specific SIN3 complexes, during distinct developmental stages.


Subject(s)
Drosophila/metabolism , Gene Expression Regulation, Developmental , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/metabolism , Animals , Blotting, Western , Cells, Cultured , Drosophila/growth & development , Immunoenzyme Techniques , Protein Isoforms , Proteolysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sin3 Histone Deacetylase and Corepressor Complex
12.
J Biol Chem ; 291(17): 9161-72, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-26917723

ABSTRACT

Ubiquitination is a post-translational modification that regulates most cellular pathways and processes, including degradation of proteins by the proteasome. Substrate ubiquitination is controlled at various stages, including through its reversal by deubiquitinases (DUBs). A critical outcome of this process is the recycling of monoubiquitin. One DUB whose function has been proposed to include monoubiquitin recycling is USP5. Here, we investigated whether Drosophila USP5 is important for maintaining monoubiquitin in vivo We found that the fruit fly orthologue of USP5 has catalytic preferences similar to its human counterpart and that this DUB is necessary during fly development. Our biochemical and genetic experiments indicate that reduction of USP5 does not lead to monoubiquitin depletion in developing flies. Also, introduction of exogenous ubiquitin does not suppress developmental lethality caused by loss of endogenous USP5. Our work indicates that a primary physiological role of USP5 is not to recycle monoubiquitin for reutilization, but that it may involve disassembly of conjugated ubiquitin to maintain proteasome function.


Subject(s)
Drosophila Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin/metabolism , Ubiquitination/physiology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Proteasome Endopeptidase Complex/genetics , Ubiquitin/genetics , Ubiquitin-Specific Proteases/genetics
13.
Hum Mol Genet ; 24(15): 4385-96, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25954029

ABSTRACT

Spinocerebellar ataxia type 6 (SCA6) belongs to the family of CAG/polyglutamine (polyQ)-dependent neurodegenerative disorders. SCA6 is caused by abnormal expansion in a CAG trinucleotide repeat within exon 47 of CACNA1A, a bicistronic gene that encodes α1A, a P/Q-type calcium channel subunit and a C-terminal protein, termed α1ACT. Expansion of the CAG/polyQ region of CACNA1A occurs within α1ACT and leads to ataxia. There are few animal models of SCA6. Here, we describe the generation and characterization of the first Drosophila melanogaster models of SCA6, which express the entire human α1ACT protein with a normal or expanded polyQ. The polyQ-expanded version of α1ACT recapitulates the progressively degenerative nature of SCA6 when expressed in various fly tissues and the presence of densely staining aggregates. Additional studies identify the co-chaperone DnaJ-1 as a potential therapeutic target for SCA6. Expression of DnaJ-1 potently suppresses α1ACT-dependent degeneration and lethality, concomitant with decreased aggregation and reduced nuclear localization of the pathogenic protein. Mutating the nuclear importer karyopherin α3 also leads to reduced toxicity from pathogenic α1ACT. Little is known about the steps leading to degeneration in SCA6 and the means to protect neurons in this disease are lacking. Invertebrate animal models of SCA6 can expand our understanding of molecular sequelae related to degeneration in this disorder and lead to the rapid identification of cellular components that can be targeted to treat it.


Subject(s)
Calcium Channels/genetics , Drosophila Proteins/genetics , HSP40 Heat-Shock Proteins/genetics , Spinocerebellar Ataxias/genetics , alpha Karyopherins/genetics , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Gene Expression Regulation , HSP40 Heat-Shock Proteins/biosynthesis , Humans , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Neurons/pathology , Spinocerebellar Ataxias/pathology , Trinucleotide Repeat Expansion/genetics , alpha Karyopherins/biosynthesis
14.
Brain ; 139(11): 2891-2908, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27645800

ABSTRACT

No disease-modifying treatment exists for the fatal neurodegenerative polyglutamine disease known both as Machado-Joseph disease and spinocerebellar ataxia type 3. As a potential route to therapy, we identified small molecules that reduce levels of the mutant disease protein, ATXN3. Screens of a small molecule collection, including 1250 Food and Drug Administration-approved drugs, in a novel cell-based assay, followed by secondary screens in brain slice cultures from transgenic mice expressing the human disease gene, identified the atypical antipsychotic aripiprazole as one of the hits. Aripiprazole increased longevity in a Drosophila model of Machado-Joseph disease and effectively reduced aggregated ATXN3 species in flies and in brains of transgenic mice treated for 10 days. The aripiprazole-mediated decrease in ATXN3 abundance may reflect a complex response culminating in the modulation of specific components of cellular protein homeostasis. Aripiprazole represents a potentially promising therapeutic drug for Machado-Joseph disease and possibly other neurological proteinopathies.


Subject(s)
Antipsychotic Agents/therapeutic use , Aripiprazole/therapeutic use , Ataxin-3/metabolism , Machado-Joseph Disease/drug therapy , Machado-Joseph Disease/metabolism , Mutant Proteins/drug effects , Animals , Animals, Genetically Modified , Ataxin-3/genetics , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Disease Models, Animal , Drosophila , Drug Evaluation, Preclinical , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , HEK293 Cells/drug effects , HEK293 Cells/metabolism , HEK293 Cells/ultrastructure , Humans , Machado-Joseph Disease/genetics , Mice , Mutant Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Peptides/genetics , Piperidines/pharmacology , Pyrans/pharmacology , Pyrazoles/pharmacology
15.
Neurobiol Dis ; 82: 12-21, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26007638

ABSTRACT

Ataxin-3 is a deubiquitinase and polyglutamine (polyQ) disease protein with a protective role in Drosophila melanogaster models of neurodegeneration. In the fruit fly, wild-type ataxin-3 suppresses toxicity from several polyQ disease proteins, including a pathogenic version of itself that causes spinocerebellar ataxia type 3 and pathogenic huntingtin, which causes Huntington's disease. The molecular partners of ataxin-3 in this protective function are unclear. Here, we report that ataxin-3 requires its direct interaction with the ubiquitin-binding and proteasome-associated protein, Rad23 (known as hHR23A/B in mammals) in order to suppress toxicity from polyQ species in Drosophila. According to additional studies, ataxin-3 does not rely on autophagy or the proteasome to suppress polyQ-dependent toxicity in fly eyes. Instead this deubiquitinase, through its interaction with Rad23, leads to increased protein levels of the co-chaperone DnaJ-1 and depends on it to protect against degeneration. Through DnaJ-1, our data connect ataxin-3 and Rad23 to protective processes involved with protein folding rather than increased turnover of toxic polyQ species.


Subject(s)
Ataxin-3/metabolism , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , HSP40 Heat-Shock Proteins/metabolism , Neurodegenerative Diseases/metabolism , Neuroprotection/physiology , Animals , Animals, Genetically Modified , Ataxin-3/genetics , Autophagy/genetics , DNA Repair Enzymes/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , HSP40 Heat-Shock Proteins/genetics , Neurodegenerative Diseases/genetics , Peptides , Protein Folding
16.
J Biol Chem ; 288(24): 17145-55, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23625928

ABSTRACT

The functional diversity of deubiquitinating enzymes (DUBs) is not well understood. The MJD family of DUBs consists of four cysteine proteases that share a catalytic "Josephin" domain. The family is named after the DUB ATXN3, which causes the neurodegenerative disease Machado-Joseph disease. The two closely related Josephin domain-containing (JosD) proteins 1 and 2 consist of little more than the Josephin domain. To gain insight into the properties of Josephin domains, we investigated JosD1 and JosD2. JosD1 and JosD2 were found to differ fundamentally in many respects. In vitro, only JosD2 can cleave ubiquitin chains. In contrast, JosD1 cleaves ubiquitin chains only after it is monoubiquitinated, a form of posttranslational-dependent regulation shared with ATXN3. A significant fraction of JosD1 is monoubiquitinated in diverse mouse tissues. In cell-based studies, JosD2 localizes to the cytoplasm whereas JosD1 preferentially localizes to the plasma membrane, particularly when ubiquitinated. The membrane occupancy by JosD1 suggests that it could participate in membrane-dependent events such as cell motility and endocytosis. Indeed, time-lapse imaging revealed that JosD1 enhances membrane dynamics and cell motility. JosD1 also influences endocytosis in cultured cells by increasing the uptake of endocytic markers of macropinocytosis while decreasing those for clathrin- and caveolae-mediated endocytosis. Our results establish that two closely related DUBs differ markedly in activity and function and that JosD1, a membrane-associated DUB whose activity is regulated by ubiquitination, helps regulate membrane dynamics, cell motility, and endocytosis.


Subject(s)
Cell Membrane/enzymology , Cell Movement , Endocytosis , Endopeptidases/metabolism , Ubiquitination , Animals , COS Cells , Caveolae/metabolism , Chlorocebus aethiops , Clathrin/metabolism , Endopeptidases/genetics , Gene Expression , HEK293 Cells , Humans , Mice , Organ Specificity , Protein Transport , Single-Cell Analysis , Time-Lapse Imaging
17.
J Biol Chem ; 288(48): 34460-9, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24106274

ABSTRACT

Deubiquitinases (DUBs) are proteases that regulate various cellular processes by controlling protein ubiquitination. Cell-based studies indicate that the regulation of the activity of DUBs is important for homeostasis and is achieved by multiple mechanisms, including through their own ubiquitination. However, the physiological significance of the ubiquitination of DUBs to their functions in vivo is unclear. Here, we report that ubiquitination of the DUB ataxin-3 at lysine residue 117, which markedly enhances its protease activity in vitro, is critical for its ability to suppress toxic protein-dependent degeneration in Drosophila melanogaster. Compared with ataxin-3 with only Lys-117 present, ataxin-3 that does not become ubiquitinated performs significantly less efficiently in suppressing or delaying the onset of toxic protein-dependent degeneration in flies. According to further studies, the C terminus of Hsc70-interacting protein (CHIP), an E3 ubiquitin ligase that ubiquitinates ataxin-3 in vitro, is dispensable for its ubiquitination in vivo and is not required for the neuroprotective function of this DUB in Drosophila. Our work also suggests that ataxin-3 suppresses degeneration by regulating toxic protein aggregation rather than stability.


Subject(s)
Drosophila melanogaster/enzymology , Lysine/genetics , Ubiquitin-Specific Proteases/metabolism , Ubiquitination , Animals , Ataxin-3 , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Lysine/metabolism , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Pigmentation/genetics , Proteolysis , Repressor Proteins/genetics , Retina/growth & development , Retina/metabolism , Ubiquitin/chemistry , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Specific Proteases/genetics
18.
J Biol Chem ; 288(26): 18784-8, 2013 Jun 28.
Article in English | MEDLINE | ID: mdl-23696636

ABSTRACT

Attachment of ubiquitin to substrate is typically thought to occur via formation of an isopeptide bond between the C-terminal glycine residue of ubiquitin and a lysine residue in the substrate. In vitro, Ube2w is nonreactive with free lysine yet readily ubiquitinates substrate. Ube2w also contains novel residues within its active site that are important for its ability to ubiquitinate substrate. To identify the site of modification, we analyzed ubiquitinated substrates by mass spectrometry and found the N-terminal -NH2 group as the site of conjugation. To confirm N-terminal ubiquitination, we generated lysine-less and N-terminally blocked versions of one substrate, the polyglutamine disease protein ataxin-3, and showed that Ube2w can ubiquitinate a lysine-less, but not N-terminally blocked, ataxin-3. This was confirmed with a second substrate, the neurodegenerative disease protein Tau. Finally, we directly sequenced the N terminus of unmodified and ubiquitinated ataxin-3, demonstrating that Ube2w attaches ubiquitin to the N terminus of its substrates. Together these data demonstrate that Ube2w has novel enzymatic properties that direct ubiquitination of the N terminus of substrates.


Subject(s)
Lysine/chemistry , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin/chemistry , Amino Acid Sequence , Ataxin-3 , Catalytic Domain , Chromatography, Liquid , Humans , Mass Spectrometry , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Peptides/chemistry , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Repressor Proteins/chemistry , Sequence Homology, Amino Acid , tau Proteins/chemistry
19.
J Neurosci Res ; 92(9): 1100-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24798551

ABSTRACT

Age-related neurodegeneration has been studied extensively through the use of model organisms, including the genetically versatile Drosophila melanogaster. Various neurotoxic proteins have been expressed in fly eyes to approximate degeneration occurring in humans, and much has been learned from this heterologous system. Although Drosophila expedites scientific research through rapid generational times and relative inexpensiveness, one factor that can hinder analyses is the examination of milder forms of degeneration caused by some toxic proteins in fly eyes. Whereas several disease proteins cause massive degeneration that is easily observed by examining the external structure of the fly eye, others cause mild degeneration that is difficult to observe externally and requires laborious histological preparation to assess and monitor. Here, we describe a sensitive fluorescence-based method to observe, monitor, and quantify mild Drosophila eye degeneration caused by various proteins, including the polyglutamine disease proteins ataxin-3 (spinocerebellar ataxia type 3) and huntingtin (Huntington's disease), mutant α-synuclein (Parkinson's disease), and Aß42 (Alzheimer's disease). We show that membrane-targeted green fluorescent protein reports degeneration robustly and quantitatively. This simple yet powerful technique, which is amenable to large-scale screens, can help accelerate studies to understand age-related degeneration and to find factors that suppress it for therapeutic purposes.


Subject(s)
Amyloid beta-Peptides/metabolism , Green Fluorescent Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Repressor Proteins/metabolism , Retinal Degeneration/diagnosis , Retinal Degeneration/genetics , Amyloid beta-Peptides/genetics , Animals , Ataxin-3 , CD8 Antigens/genetics , CD8 Antigens/metabolism , Disease Models, Animal , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Green Fluorescent Proteins/genetics , Humans , Huntingtin Protein , Mice, Transgenic , Microtubule-Associated Proteins/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptide Fragments/genetics , Peptides/genetics , Peptides/metabolism , Repressor Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , alpha-Synuclein/genetics
20.
Sci Rep ; 14(1): 14332, 2024 06 21.
Article in English | MEDLINE | ID: mdl-38906973

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of an uninterrupted polyglutamine (polyQ) repeat in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in fruit fly survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.


Subject(s)
Ataxin-7 , Disease Models, Animal , Peptides , Spinocerebellar Ataxias , Animals , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology , Spinocerebellar Ataxias/metabolism , Ataxin-7/genetics , Ataxin-7/metabolism , Humans , Peptides/metabolism , Peptides/genetics , Drosophila/genetics , Animals, Genetically Modified , Disease Progression , Drosophila melanogaster/genetics , Retina/metabolism , Retina/pathology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL