Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38673939

ABSTRACT

Polyglutamine (polyQ)-encoding CAG repeat expansions represent a common disease-causing mutation responsible for several dominant spinocerebellar ataxias (SCAs). PolyQ-expanded SCA proteins are toxic for cerebellar neurons, with Purkinje cells (PCs) being the most vulnerable. RNA interference (RNAi) reagents targeting transcripts with expanded CAG reduce the level of various mutant SCA proteins in an allele-selective manner in vitro and represent promising universal tools for treating multiple CAG/polyQ SCAs. However, it remains unclear whether the therapeutic targeting of CAG expansion can be achieved in vivo and if it can ameliorate cerebellar functions. Here, using a mouse model of SCA7 expressing a mutant Atxn7 allele with 140 CAGs, we examined the efficacy of short hairpin RNAs (shRNAs) targeting CAG repeats expressed from PHP.eB adeno-associated virus vectors (AAVs), which were introduced into the brain via intravascular injection. We demonstrated that shRNAs carrying various mismatches with the CAG target sequence reduced the level of polyQ-expanded ATXN7 in the cerebellum, albeit with varying degrees of allele selectivity and safety profile. An shRNA named A4 potently reduced the level of polyQ-expanded ATXN7, with no effect on normal ATXN7 levels and no adverse side effects. Furthermore, A4 shRNA treatment improved a range of motor and behavioral parameters 23 weeks after AAV injection and attenuated the disease burden of PCs by preventing the downregulation of several PC-type-specific genes. Our results show the feasibility of the selective targeting of CAG expansion in the cerebellum using a blood-brain barrier-permeable vector to attenuate the disease phenotype in an SCA mouse model. Our study represents a significant advancement in developing CAG-targeting strategies as a potential therapy for SCA7 and possibly other CAG/polyQ SCAs.


Subject(s)
Ataxin-7 , Dependovirus , Disease Models, Animal , Peptides , Phenotype , RNA, Small Interfering , Spinocerebellar Ataxias , Trinucleotide Repeat Expansion , Animals , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/therapy , Spinocerebellar Ataxias/metabolism , Peptides/genetics , Dependovirus/genetics , Mice , Ataxin-7/genetics , Ataxin-7/metabolism , Trinucleotide Repeat Expansion/genetics , RNA, Small Interfering/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Purkinje Cells/metabolism , Purkinje Cells/pathology , Mice, Transgenic , Cerebellum/metabolism , Cerebellum/pathology , Humans , Genetic Therapy/methods , Alleles
2.
Cells ; 12(23)2023 11 30.
Article in English | MEDLINE | ID: mdl-38067163

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is an autosomal-dominant inherited disease characterized by progressive ataxia and retinal degeneration. SCA7 belongs to a group of neurodegenerative diseases caused by an expanded CAG repeat in the disease-causing gene, resulting in aberrant polyglutamine (polyQ) protein synthesis. PolyQ ataxin-7 is prone to aggregate in intracellular inclusions, perturbing cellular processes leading to neuronal death in specific regions of the central nervous system (CNS). Currently, there is no treatment for SCA7; however, a promising approach successfully applied to other polyQ diseases involves the clearance of polyQ protein aggregates through pharmacological activation of autophagy. Nonetheless, the blood-brain barrier (BBB) poses a challenge for delivering drugs to the CNS, limiting treatment effectiveness. This study aimed to develop a polymeric nanocarrier system to deliver therapeutic agents across the BBB into the CNS. We prepared poly(lactic-co-glycolic acid) nanoparticles (NPs) modified with Poloxamer188 and loaded with rapamycin to enable NPs to activate autophagy. We demonstrated that these rapamycin-loaded NPs were successfully taken up by neuronal and glial cells, demonstrating high biocompatibility without adverse effects. Remarkably, rapamycin-loaded NPs effectively cleared mutant ataxin-7 aggregates in a SCA7 glial cell model, highlighting their potential as a therapeutic approach to fight SCA7 and other polyQ diseases.


Subject(s)
Spinocerebellar Ataxias , Humans , Ataxin-7/genetics , Ataxin-7/metabolism , Spinocerebellar Ataxias/drug therapy , Spinocerebellar Ataxias/genetics , Neurons/metabolism , Neuroglia/metabolism , Sirolimus
3.
Genetics ; 224(3)2023 07 06.
Article in English | MEDLINE | ID: mdl-37075097

ABSTRACT

Ataxin-7 maintains the integrity of Spt-Ada-Gcn5-Acetyltransferase (SAGA), an evolutionarily conserved coactivator in stimulating preinitiation complex (PIC) formation for transcription initiation, and thus, its upregulation or downregulation is associated with various diseases. However, it remains unknown how ataxin-7 is regulated that could provide new insights into disease pathogenesis and therapeutic interventions. Here, we show that ataxin-7's yeast homologue, Sgf73, undergoes ubiquitylation and proteasomal degradation. Impairment of such regulation increases Sgf73's abundance, which enhances recruitment of TATA box-binding protein (TBP) (that nucleates PIC formation) to the promoter but impairs transcription elongation. Further, decreased Sgf73 level reduces PIC formation and transcription. Thus, Sgf73 is fine-tuned by ubiquitin-proteasome system (UPS) in orchestrating transcription. Likewise, ataxin-7 undergoes ubiquitylation and proteasomal degradation, alteration of which changes ataxin-7's abundance that is associated with altered transcription and cellular pathologies/diseases. Collectively, our results unveil a novel UPS regulation of Sgf73/ataxin-7 for normal cellular health and implicate alteration of such regulation in diseases.


Subject(s)
Saccharomyces cerevisiae Proteins , Ubiquitin , Ataxin-7/genetics , Ataxin-7/metabolism , Ubiquitin/metabolism , Proteasome Endopeptidase Complex/metabolism , Histone Acetyltransferases/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
4.
Mol Neurobiol ; 59(8): 5236-5251, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35689166

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) and other polyglutamine (polyQ) diseases are caused by expansions of polyQ repeats in disease-specific proteins. Aggregation of the polyQ proteins resulting in various forms of cellular stress, that could induce the stress granule (SG) response, is believed to be a common pathological mechanism in these disorders. SGs can contribute to cell survival but have also been suggested to exacerbate disease pathology by seeding protein aggregation. In this study, we show that two SG-related proteins, TDP-43 and TIA1, are sequestered into the aggregates formed by polyQ-expanded ATXN7 in SCA7 cells. Interestingly, mutant ATXN7 also localises to induced SGs, and this association altered the shape of the SGs. In spite of this, neither the ability to induce nor to disassemble SGs, in response to arsenite stress induction or relief, was affected in SCA7 cells. Moreover, we could not observe any change in the number of ATXN7 aggregates per cell following SG induction, although a small, non-significant, increase in total aggregated ATXN7 material could be detected using filter trap. However, mutant ATXN7 expression in itself increased the speckling of the SG-nucleating protein G3BP1 and the SG response. Taken together, our results indicate that the SG response is induced, and although some key modulators of SGs show altered behaviour, the dynamics of SGs appear normal in the presence of mutant ATXN7.


Subject(s)
DNA Helicases , Spinocerebellar Ataxias , Ataxin-7/metabolism , Cytoplasmic Granules/metabolism , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Humans , Peptides , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , Spinocerebellar Ataxias/genetics , Stress Granules , T-Cell Intracellular Antigen-1/metabolism
5.
Thorac Cancer ; 13(11): 1597-1610, 2022 06.
Article in English | MEDLINE | ID: mdl-35445786

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) participate in the occurrence and progression of many cancers. CircRNA ataxin 7 (circATXN7) (circBase ID: hsa_circ_0066436) plays a promoting influence on gastric cancer progression. However, the biological role of circATXN7 in non-small cell lung cancer (NSCLC) is indistinct. METHODS: Levels of circATXN7, microRNA (miR)-7-5p, and profilin 2 (PFN2) mRNA were detected using quantitative real-time polymerase chain reaction (RT-qPCR). Proliferation, apoptosis, metastasis, and invasion were analyzed using cell counting kit-8 (CCK-8), colony formation, 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, and transwell assays. Protein levels were analyzed using western blotting (WB) and immunohistochemistry (IHC). The relationship between circATXN7 or PFN2 and miR-7-5p was analyzed by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. The biological function of circATXN7 was verified by xenograft assay. RESULTS: CircATXN7 and PFN2 were highly expressed in NSCLC, whereas miR-7-5p expression had the opposite trend. CircATXN7 overexpression constrained apoptosis and promoted proliferation, metastasis, invasion, and epithelial-mesenchymal transition of NSCLC cells, but circATXN7 silencing played the opposing influence and repressed xenograft tumor growth in vivo. CircATXN7 served as a miR-7-5p sponge, and circATXN7 regulated malignant behaviors of NSCLC cells through sponging miR-7-5p. PFN2 acted as a miR-7-5p target. PFN2 silencing overturned the promoting effect of miR-7-5p inhibitor on NSCLC cell malignancy, while PFN2 overexpression reversed the inhibitory impact of miR-7-5p mimic on NSCLC cell malignancy. CONCLUSION: CircATXN7 accelerated the malignancy of NSCLC cells through adsorbing miR-7-5p and upregulating PFN2, offering evidence to support circATXN7 as a target for NSCLC treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , Ataxin-7/genetics , Ataxin-7/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/genetics , Down-Regulation , Humans , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Profilins/genetics , Profilins/metabolism , RNA, Circular/genetics
6.
Cell Rep ; 37(9): 110062, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34852229

ABSTRACT

A common mechanism in inherited ataxia is a vulnerability of DNA damage. Spinocerebellar ataxia type 7 (SCA7) is a CAG-polyglutamine-repeat disorder characterized by cerebellar and retinal degeneration. Polyglutamine-expanded ataxin-7 protein incorporates into STAGA co-activator complex and interferes with transcription by altering histone acetylation. We performed chromatic immunoprecipitation sequencing ChIP-seq on cerebellum from SCA7 mice and observed increased H3K9-promoter acetylation in DNA repair genes, resulting in increased expression. After detecting increased DNA damage in SCA7 cells, mouse primary cerebellar neurons, and patient stem-cell-derived neurons, we documented reduced homology-directed repair (HDR) and single-strand annealing (SSA). To evaluate repair at endogenous DNA in native chromosome context, we modified linear amplification-mediated high-throughput genome-wide translocation sequencing and found that DNA translocations are less frequent in SCA7 models, consistent with decreased HDR and SSA. Altered DNA repair function in SCA7 may predispose the subject to excessive DNA damage, leading to neuron demise and highlights DNA repair as a therapy target.


Subject(s)
Ataxin-7/metabolism , Cerebellar Diseases/pathology , DNA Repair , Histones/metabolism , Neurons/pathology , Peptides/genetics , Spinocerebellar Ataxias/complications , Acetylation , Animals , Ataxin-7/genetics , Cerebellar Diseases/etiology , Cerebellar Diseases/metabolism , Female , Histones/genetics , Humans , Male , Mice , Neurons/metabolism
7.
Mol Cell Neurosci ; 110: 103584, 2021 01.
Article in English | MEDLINE | ID: mdl-33338633

ABSTRACT

Polyglutamine (polyQ) diseases, such as Spinocerebellar ataxia type 7 (SCA7), are caused by expansions of polyQ repeats in disease specific proteins. The sequestration of vital proteins into aggregates formed by polyQ proteins is believed to be a common pathological mechanism in these disorders. The RNA-binding protein FUS has been observed in polyQ aggregates, though if disruption of this protein plays a role in the neuronal dysfunction in SCA7 or other polyQ diseases remains unclear. We therefore analysed FUS localisation and function in a stable inducible PC12 cell model expressing the SCA7 polyQ protein ATXN7. We found that there was a high degree of FUS sequestration, which was associated with a more cytoplasmic FUS localisation, as well as a decreased expression of FUS regulated mRNAs. In contrast, the role of FUS in the formation of γH2AX positive DNA damage foci was unaffected. In fact, a statistical increase in the number of γH2AX foci, as well as an increased trend of single and double strand DNA breaks, detected by comet assay, could be observed in mutant ATXN7 cells. These results were further corroborated by a clear trend towards increased DNA damage in SCA7 patient fibroblasts. Our findings suggest that both alterations in the RNA regulatory functions of FUS, and increased DNA damage, may contribute to the pathology of SCA7.


Subject(s)
Ataxin-7/genetics , DNA Damage , RNA-Binding Protein FUS/metabolism , Spinocerebellar Ataxias/metabolism , Animals , Ataxin-7/metabolism , Cells, Cultured , Fibroblasts/metabolism , Histones/metabolism , Humans , PC12 Cells , Peptides/chemistry , Peptides/genetics , Protein Transport , Rats , Spinocerebellar Ataxias/genetics
8.
Neurotherapeutics ; 16(4): 1074-1096, 2019 10.
Article in English | MEDLINE | ID: mdl-31432449

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is a rare autosomal dominant neurodegenerative disorder characterized by progressive neuronal loss in the cerebellum, brainstem, and retina, leading to cerebellar ataxia and blindness as major symptoms. SCA7 is due to the expansion of a CAG triplet repeat that is translated into a polyglutamine tract in ATXN7. Larger SCA7 expansions are associated with earlier onset of symptoms and more severe and rapid disease progression. Here, we summarize the pathological and genetic aspects of SCA7, compile the current knowledge about ATXN7 functions, and then focus on recent advances in understanding the pathogenesis and in developing biomarkers and therapeutic strategies. ATXN7 is a bona fide subunit of the multiprotein SAGA complex, a transcriptional coactivator harboring chromatin remodeling activities, and plays a role in the differentiation of photoreceptors and Purkinje neurons, two highly vulnerable neuronal cell types in SCA7. Polyglutamine expansion in ATXN7 causes its misfolding and intranuclear accumulation, leading to changes in interactions with native partners and/or partners sequestration in insoluble nuclear inclusions. Studies of cellular and animal models of SCA7 have been crucial to unveil pathomechanistic aspects of the disease, including gene deregulation, mitochondrial and metabolic dysfunctions, cell and non-cell autonomous protein toxicity, loss of neuronal identity, and cell death mechanisms. However, a better understanding of the principal molecular mechanisms by which mutant ATXN7 elicits neurotoxicity, and how interconnected pathogenic cascades lead to neurodegeneration is needed for the development of effective therapies. At present, therapeutic strategies using nucleic acid-based molecules to silence mutant ATXN7 gene expression are under development for SCA7.


Subject(s)
Ataxin-7/genetics , Disease Models, Animal , Drug Delivery Systems/trends , Gene Targeting/trends , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/therapy , Animals , Ataxin-7/metabolism , Autophagy/physiology , Brain/metabolism , Brain/pathology , Drug Delivery Systems/methods , Gene Targeting/methods , Genetic Therapy/methods , Genetic Therapy/trends , Humans , Neurons/metabolism , Neurons/pathology , Peptides/genetics , Peptides/metabolism , Spinocerebellar Ataxias/metabolism
9.
Elife ; 82019 07 26.
Article in English | MEDLINE | ID: mdl-31348003

ABSTRACT

Atxn7, a subunit of SAGA chromatin remodeling complex, is subject to polyglutamine expansion at the amino terminus, causing spinocerebellar ataxia type 7 (SCA7), a progressive retinal and neurodegenerative disease. Within SAGA, the Atxn7 amino terminus anchors Non-stop, a deubiquitinase, to the complex. To understand the scope of Atxn7-dependent regulation of Non-stop, substrates of the deubiquitinase were sought. This revealed Non-stop, dissociated from Atxn7, interacts with Arp2/3 and WAVE regulatory complexes (WRC), which control actin cytoskeleton assembly. There, Non-stop countered polyubiquitination and proteasomal degradation of WRC subunit SCAR. Dependent on conserved WRC interacting receptor sequences (WIRS), Non-stop augmentation increased protein levels, and directed subcellular localization, of SCAR, decreasing cell area and number of protrusions. In vivo, heterozygous mutation of SCAR did not significantly rescue knockdown of Atxn7, but heterozygous mutation of Atxn7 rescued haploinsufficiency of SCAR.


Subject(s)
Actin Cytoskeleton/metabolism , Ataxin-7/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/physiology , Endopeptidases/metabolism , Microfilament Proteins/metabolism , Animals , Gene Expression Regulation , Protein Interaction Mapping , Protein Interaction Maps
10.
Sci Rep ; 9(1): 7481, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097749

ABSTRACT

Ataxin-7 (Atx7) is a disease-related protein associated with the pathogenesis of spinocerebellar ataxia 7, while its polyglutamine (polyQ) tract in N-terminus is the causative source of aggregation and proteinopathy. We investigated the structure, dynamics and aggregation properties of the N-terminal 62-residue fragment of Atx7 (Atx7-N) by biochemical and biophysical approaches. The results showed that the normal Atx7-N with a tract of 10 glutamines (10Q) overall adopts a flexible and disordered structure, but it may contain a short or small population of helical structure in solution. PolyQ expansion increases the α-helical propensity of the polyQ tract and consequently enhances its transformation into ß-sheet structures during amyloid aggregation. An alanine-rich region (ARR) just ahead of the polyQ tract forms a local and relatively stable α-helix. The ARR α-helix can initiate and stabilize helical formation of the following polyQ tract, but it may suppress aggregation of the polyQ-expanded Atx7-N both in vitro and in cell. Thus, the preceding ARR segment in Atx7-N may influence the dynamic structure and aggregation property of the polyQ tract and even determine the threshold of the pathogenic polyQ lengths. This study may gain structural and dynamic insights into amyloid aggregation of Atx7 and help us further understand the Atx7 proteinopathy based on polyQ expansion.


Subject(s)
Amyloid/chemistry , Ataxin-7/chemistry , Molecular Dynamics Simulation , Protein Multimerization , Amyloid/metabolism , Ataxin-7/metabolism , HEK293 Cells , Humans , Peptides/chemistry , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand
11.
Neuroscience ; 400: 72-84, 2019 02 21.
Article in English | MEDLINE | ID: mdl-30625334

ABSTRACT

Spino-cerebellar ataxia type 7 (SCA7) is a polyglutamine (polyQ) disorder characterized by neurodegeneration of the brain, cerebellum, and retina caused by a polyglutamine expansion in ataxin7. The presence of an expanded polyQ tract in a mutant protein is known to induce protein aggregation, cellular stress, toxicity, and finally cell death. However, the consequences of the presence of mutant ataxin7 in the retina and the mechanisms underlying photoreceptor degeneration remain poorly understood. In this study, we show that in a retinal SCA7 mouse model, polyQ ataxin7 induces stress within the retina and activates Muller cells. Moreover, unfolded protein response and autophagy are activated in SCA7 photoreceptors. We have also shown that the photoreceptor death does not involve a caspase-dependent apoptosis but instead involves apoptosis inducing factor (AIF) and Leukocyte Elastase Inhibitor (LEI/L-DNase II). When these two cell death effectors are downregulated by their siRNA, a significant reduction in photoreceptor death is observed. These results highlight the consequences of polyQ protein expression in the retina and the role of caspase-independent pathways involved in photoreceptor cell death.


Subject(s)
Ataxin-7/metabolism , Cell Death , Peptides/metabolism , Retinal Degeneration/metabolism , Spinocerebellar Ataxias/metabolism , Animals , Apoptosis Inducing Factor/metabolism , Ataxin-7/genetics , Calpain/metabolism , Caspases/metabolism , Cathepsins/metabolism , Disease Models, Animal , Endodeoxyribonucleases/metabolism , HEK293 Cells , Humans , Mice, Inbred C57BL , Mice, Transgenic , Photoreceptor Cells/metabolism , Retinal Degeneration/etiology , Signal Transduction , Spinocerebellar Ataxias/complications , Stress, Physiological
12.
Article in English | MEDLINE | ID: mdl-30125623

ABSTRACT

Attention-deficit/hyperactivity disorder (ADHD) is a heterogeneous neurodevelopmental disorder characterized by varying levels of hyperactivity, inattention, and impulsivity. Patients with ADHD are often classified as (1) predominantly hyperactive-impulsive, (2) predominantly inattentive, and (3) combined type. There is a growing interest in developing specific animal models that would recapitulate specific clinical forms of ADHD, with the goal of developing specific therapeutic strategies. In our previous study, we have identified Ataxin-7 (Atxn7) as a hyperactivity-associated gene. Here, we generated Atxn7 overexpressing (Atxn7 OE) mice to investigate whether the increased Atxn7 expression in the brain correlates with ADHD-like behaviors. Quantitative real-time polymerase chain reaction and immunofluorescence confirmed overexpression of the Atxn7 gene and protein in the prefrontal cortex (PFC) and striatum (STR) of the Atxn7 OE mice. The Atxn7 OE mice displayed hyperactivity and impulsivity, but not inattention. Interestingly, treatment with the ADHD drug, atomoxetine (3 mg/kg, intraperitoneal), attenuated ADHD-like behaviors and reduced Atxn7 gene expression in the PFC and STR of these mice. These findings suggest that Atxn7 plays a role in the pathophysiology of ADHD, and that the Atxn7 OE mice can be used as an animal model of the hyperactive-impulsive phenotype of this disorder. Although confirmatory studies are warranted, the present study provides valuable information regarding the potential genetic underpinnings of ADHD.


Subject(s)
Adrenergic Uptake Inhibitors/therapeutic use , Ataxin-7/metabolism , Atomoxetine Hydrochloride/therapeutic use , Hyperkinesis/drug therapy , Hyperkinesis/genetics , Impulsive Behavior/drug effects , Animals , Ataxin-7/genetics , Delay Discounting/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Impulsive Behavior/physiology , Locomotion/drug effects , Locomotion/genetics , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger , Receptors, Dopamine D1/genetics , Receptors, Dopamine D1/metabolism , Recognition, Psychology/drug effects , Rotarod Performance Test , Statistics, Nonparametric
13.
Dis Model Mech ; 12(1)2019 01 11.
Article in English | MEDLINE | ID: mdl-30559154

ABSTRACT

Perturbation of protein homeostasis and aggregation of misfolded proteins is a major cause of many human diseases. A hallmark of the neurodegenerative disease spinocerebellar ataxia type 7 (SCA7) is the intranuclear accumulation of mutant, misfolded ataxin-7 (polyQ-ATXN7). Here, we show that endogenous ATXN7 is modified by SUMO proteins, thus also suggesting a physiological role for this modification under conditions of proteotoxic stress caused by the accumulation of polyQ-ATXN7. Co-immunoprecipitation experiments, immunofluorescence microscopy and proximity ligation assays confirmed the colocalization and interaction of polyQ-ATXN7 with SUMO2 in cells. Moreover, upon inhibition of the proteasome, both endogenous SUMO2/3 and the RNF4 ubiquitin ligase surround large polyQ-ATXN7 intranuclear inclusions. Overexpression of RNF4 and/or SUMO2 significantly decreased levels of polyQ-ATXN7 and, upon proteasomal inhibition, led to a marked increase in the polyubiquitination of polyQ-ATXN7. This provides a mechanism for the clearance of polyQ-ATXN7 from affected cells that involves the recruitment of RNF4 by SUMO2/3-modified polyQ-ATXN7, thus leading to its ubiquitination and proteasomal degradation. In a SCA7 knock-in mouse model, we similarly observed colocalization of SUMO2/3 with polyQ-ATXN7 inclusions in the cerebellum and retina. Furthermore, we detected accumulation of SUMO2/3 high-molecular-mass species in the cerebellum of SCA7 knock-in mice, compared with their wild-type littermates, and changes in SUMO-related transcripts. Immunohistochemical analysis showed the accumulation of SUMO proteins and RNF4 in the cerebellum of SCA7 patients. Taken together, our results show that the SUMO pathway contributes to the clearance of aggregated ATXN7 and suggest that its deregulation might be associated with SCA7 disease progression.


Subject(s)
Ataxin-7/metabolism , Nuclear Proteins/metabolism , Protein Folding , Proteolysis , Small Ubiquitin-Related Modifier Proteins/metabolism , Spinocerebellar Ataxias/metabolism , Sumoylation , Transcription Factors/metabolism , Animals , Cerebellum/metabolism , Child , Disease Models, Animal , HEK293 Cells , HeLa Cells , Humans , Inclusion Bodies/metabolism , MCF-7 Cells , Mice , Middle Aged , Mutation/genetics , Promyelocytic Leukemia Protein/metabolism , Proteasome Inhibitors/pharmacology , Protein Aggregates/drug effects , Protein Folding/drug effects , Proteolysis/drug effects , Spinocerebellar Ataxias/pathology , Sumoylation/drug effects , Ubiquitin/metabolism
14.
Sci Transl Med ; 10(465)2018 10 31.
Article in English | MEDLINE | ID: mdl-30381411

ABSTRACT

Spinocerebellar ataxia type 7 (SCA7) is an autosomal dominant neurodegenerative disorder characterized by cerebellar and retinal degeneration, and is caused by a CAG-polyglutamine repeat expansion in the ATAXIN-7 gene. Patients with SCA7 develop progressive cone-rod dystrophy, typically resulting in blindness. Antisense oligonucleotides (ASOs) are single-stranded chemically modified nucleic acids designed to mediate the destruction, prevent the translation, or modify the processing of targeted RNAs. Here, we evaluated ASOs as treatments for SCA7 retinal degeneration in representative mouse models of the disease after injection into the vitreous humor of the eye. Using Ataxin-7 aggregation, visual function, retinal histopathology, gene expression, and epigenetic dysregulation as outcome measures, we found that ASO-mediated Ataxin-7 knockdown yielded improvements in treated SCA7 mice. In SCA7 mice with retinal disease, intravitreal injection of Ataxin-7 ASOs also improved visual function despite initiating treatment after symptom onset. Using color fundus photography and autofluorescence imaging, we also determined the nature of retinal degeneration in human SCA7 patients. We observed variable disease severity and cataloged rapidly progressive retinal degeneration. Given the accessibility of neural retina, availability of objective, quantitative readouts for monitoring therapeutic response, and the rapid disease progression in SCA7, ASOs targeting ATAXIN-7 might represent a viable treatment for SCA7 retinal degeneration.


Subject(s)
Ataxin-7/metabolism , Mutant Proteins/metabolism , Oligonucleotides, Antisense/pharmacology , Spinocerebellar Ataxias/physiopathology , Vision, Ocular/drug effects , Animals , Ataxin-7/genetics , Chromatin Assembly and Disassembly/drug effects , Disease Models, Animal , Disease Progression , Epigenesis, Genetic/drug effects , Gene Expression Regulation/drug effects , Humans , Intravitreal Injections , Mice , Oligonucleotides, Antisense/administration & dosage , Peptides/metabolism , Phenotype , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/metabolism , Protein Aggregates/drug effects , Retina/drug effects , Retina/metabolism , Retinal Degeneration/complications , Retinal Degeneration/pathology , Retinal Degeneration/physiopathology , Spinocerebellar Ataxias/complications , Spinocerebellar Ataxias/pathology
15.
Cell Rep ; 21(5): 1240-1252, 2017 Oct 31.
Article in English | MEDLINE | ID: mdl-29091763

ABSTRACT

Myotonic dystrophy types 1 (DM1) and 2 (DM2) are dominantly inherited neuromuscular disorders caused by a toxic gain of function of expanded CUG and CCUG repeats, respectively. Although both disorders are clinically similar, congenital myotonic dystrophy (CDM), a severe DM form, is found only in DM1. CDM is also characterized by muscle fiber immaturity not observed in adult DM, suggesting specific pathological mechanisms. Here, we revealed upregulation of the interleukin-6 (IL-6) myokine signaling pathway in CDM muscles. We also found a correlation between muscle immaturity and not only IL-6 expression but also expanded CTG repeat length and CpG methylation status upstream of the repeats. Aberrant CpG methylation was associated with transcriptional dysregulation at the repeat locus, increasing the toxic RNA burden that upregulates IL-6. Because the IL-6 pathway is involved in myocyte maturation and muscle atrophy, our results indicate that enhanced RNA toxicity contributes to severe CDM phenotypes through aberrant IL-6 signaling.


Subject(s)
Myotonic Dystrophy/pathology , Signal Transduction/physiology , Alternative Splicing , Ataxin-7/genetics , Ataxin-7/metabolism , Binding Sites , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , CpG Islands , DNA Methylation , Humans , Infant , Interleukin-6/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscles/metabolism , Myotonic Dystrophy/genetics , Myotonic Dystrophy/metabolism , Myotonin-Protein Kinase/genetics , Protein Binding , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , STAT3 Transcription Factor/metabolism , Transcription, Genetic , Trinucleotide Repeat Expansion , Up-Regulation
16.
Genes Brain Behav ; 16(7): 664-674, 2017 09.
Article in English | MEDLINE | ID: mdl-28422445

ABSTRACT

Several studies suggest a strong genetic component of attention-deficit/hyperactivity disorder (ADHD), a complex neurodevelopmental disorder characterized by inappropriate levels of hyperactivity, impulsivity and inattention. Determining specific genetic risk variants for each symptom dimension of ADHD may aid in the identification of the biological risk factors of the disorder. In this study, we explored the potential genetic underpinnings of the hyperactive phenotype of ADHD. To this end, we examined differentially expressed genes (DEGs) in the prefrontal cortex (PFC) of SHR/NCrl, an animal model of ADHD, compared with its genetic control, the Wistar Kyoto (WKY/NCrl) rat and the Wistar rat, strain used to represent the 'normal' heterogeneous population. Relative to WKY/NCrl and Wistar controls, SHR/NCrl showed hyperactivity in the open-field test. Treatment with the ADHD drug, amphetamine (AMPH) reduced hyperactivity in SHR/NCrl. Meanwhile, AMPH increased locomotor activity in WKY/NCrl and Wistar rats. Gene expression analysis found 21 common upregulated and 36 downregulated genes in the PFC of drug-naive SHR/NCrl when compared with WKY/NCrl and Wistar rats. Of these DEGs, expression levels of two genes, Atxn7 and Per2, which are involved in transcription and circadian rhythm, respectively, were downregulated following AMPH treatment in SHR/NCrl. Quantitative real-time-polymerase chain reaction analyses verified expression patterns of these genes in the PFC of drug-naïve and AMPH-treated SHR/NCrl. The present findings indicate genetic risk variants that may be associated with the hyperactive phenotype in ADHD. Further studies are warranted to establish the roles of Atxn7 and Per2 in mediating hyperactivity.


Subject(s)
Amphetamine/pharmacology , Attention Deficit Disorder with Hyperactivity/genetics , Central Nervous System Stimulants/pharmacology , Prefrontal Cortex/metabolism , Transcriptome , Amphetamine/therapeutic use , Animals , Ataxin-7/genetics , Ataxin-7/metabolism , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/metabolism , Central Nervous System Stimulants/therapeutic use , Down-Regulation , Locomotion , Male , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Prefrontal Cortex/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Rats, Wistar
17.
Cell Physiol Biochem ; 39(6): 2427-2438, 2016.
Article in English | MEDLINE | ID: mdl-27855399

ABSTRACT

BACKGROUND/AIMS: Hepatocellular carcinoma (HCC) is a lethal disease with nearly equal morbidity and mortality. Thus, the discovery and application of more useful predictive biomarkers for improving therapeutic effects and prediction of clinical outcomes is of crucial significance. METHODS: A total of 475 HBV-related HCC patients were enrolled. Ataxin 7 (ATXN7) single nucleotide polymorphisms (SNPs) were genotyped by Sanger DNA sequencing after PCR amplification. The associations between ATXN7 SNPs and mRNA expression with the prognosis of HBV-related HCC were analyzed. RESULTS: In all, rs3774729 was significantly associated with overall survival (OS) of HBV-related HCC (P = 0.013, HR = 0.66, 95% CI: 0.48-0.94). And patients with the AA genotype and a high level of serum alpha fetoprotein (AFP) had significantly worse OS when compared to patients with AG/GG genotypes and a low level of AFP (adjusted P = 0.007, adjusted HR = 1.83, 95% CI = 1.18-2.82). Furthermore, low expression of ATXN7 was significantly associated with poor recurrence-free survival (RFS) and OS (P = 0.007, HR = 2.38, 95% CI = 1.27-4.45 and P = 0.025, HR = 1.75, 95% CI = 1.18-2.62). CONCLUSION: ATXN7 may be a potential predictor of post-operative prognosis of HBV-related HCC.


Subject(s)
Ataxin-7/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Hepatitis B virus/physiology , Liver Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Ataxin-7/metabolism , Carcinoma, Hepatocellular/blood , Female , Genetic Association Studies , Humans , Kaplan-Meier Estimate , Liver Neoplasms/blood , Liver Neoplasms/virology , Male , Middle Aged , Multivariate Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Treatment Outcome , alpha-Fetoproteins/metabolism
18.
Mol Neurodegener ; 11(1): 58, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27465358

ABSTRACT

BACKGROUND: We used lentiviral vectors (LVs) to generate a new SCA7 animal model overexpressing a truncated mutant ataxin-7 (MUT ATXN7) fragment in the mouse cerebellum, in order to characterize the specific neuropathological and behavioral consequences of the genetic defect in this brain structure. RESULTS: LV-mediated overexpression of MUT ATXN7 into the cerebellum of C57/BL6 adult mice induced neuropathological features similar to that observed in patients, such as intranuclear aggregates in Purkinje cells (PC), loss of synaptic markers, neuroinflammation, and neuronal death. No neuropathological changes were observed when truncated wild-type ataxin-7 (WT ATXN7) was injected. Interestingly, the local delivery of LV-expressing mutant ataxin-7 (LV-MUT-ATXN7) into the cerebellum of wild-type mice also mediated the development of an ataxic phenotype at 8 to 12 weeks post-injection. Importantly, our data revealed abnormal levels of the FUS/TLS, MBNL1, and TDP-43 RNA-binding proteins in the cerebellum of the LV-MUT-ATXN7 injected mice. MUT ATXN7 overexpression induced an increase in the levels of the pathological phosphorylated TDP-43, and a decrease in the levels of soluble FUS/TLS, with both proteins accumulating within ATXN7-positive intranuclear inclusions. MBNL1 also co-aggregated with MUT ATXN7 in most PC nuclear inclusions. Interestingly, no MBNL2 aggregation was observed in cerebellar MUT ATXN7 aggregates. Immunohistochemical studies in postmortem tissue from SCA7 patients and SCA7 knock-in mice confirmed SCA7-induced nuclear accumulation of FUS/TLS and MBNL1, strongly suggesting that these proteins play a physiopathological role in SCA7. CONCLUSIONS: This study validates a novel SCA7 mouse model based on lentiviral vectors, in which strong and sustained expression of MUT ATXN7 in the cerebellum was found sufficient to generate motor defects.


Subject(s)
Ataxin-7/metabolism , DNA-Binding Proteins/metabolism , RNA-Binding Proteins/metabolism , Spinocerebellar Ataxias/genetics , Animals , Ataxin-7/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Female , Humans , Lentivirus/genetics , Mice, Inbred C57BL , Neurons/metabolism , Phenotype
19.
Mol Cancer ; 15(1): 47, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27296891

ABSTRACT

BACKGROUND: Fusion proteins have unique oncogenic properties and their identification can be useful either as diagnostic or therapeutic targets. Next generation sequencing data have previously shown a fusion gene formed between Rad51C and ATXN7 genes in the MCF7 breast cancer cell line. However, the existence of this fusion gene in colorectal patient tumor tissues is largely still unknown. METHODS: We evaluated for the presence of Rad51C-ATXN7 fusion gene in colorectal tumors and cells by RT-PCR, PCR, Topo TA cloning, Real time PCR, immunoprecipitation and immunoblotting techniques. RESULTS: We identified two forms of fusion mRNAs between Rad51C and ATXN7 in the colorectal tumors, including a Variant 1 (fusion transcript between Rad51C exons 1-7 and ATXN7 exons 6-13), and a Variant 2 (between Rad51C exons 1-6 and ATXN7 exons 6-13). In silico analysis showed that the Variant 1 produces a truncated protein, whereas the Variant 2 was predicted to produce a fusion protein with molecular weight of 110 KDa. Immunoprecipitation and Western blot analysis further showed a 110 KDa protein in colorectal tumors. 5-Azacytidine treatment of LS-174 T cells caused a 3.51-fold increase in expression of the fusion gene (Variant 2) as compared to no treatment controls evaluated by real time PCR. CONCLUSION: In conclusion we found a fusion gene between DNA repair gene Rad51C and neuro-cerebral ataxia Ataxin-7 gene in colorectal tumors. The in-frame fusion transcript of Variant 2 results in a fusion protein with molecular weight of 110 KDa. In addition, we found that expression of fusion gene is associated with functional impairment of Fanconi Anemia (FA) DNA repair pathway in colorectal tumors. The expression of Rad51C-ATXN7 in tumors warrants further investigation, as it suggests the potential of the fusion gene in treatment and predictive value in colorectal cancers.


Subject(s)
Ataxin-7/genetics , Cloning, Molecular/methods , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Oncogene Proteins, Fusion/genetics , Ataxin-7/metabolism , Azacitidine/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Computer Simulation , DNA Methylation/drug effects , DNA Repair , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Genetic Variation , Humans , Molecular Weight , Oncogene Proteins, Fusion/drug effects , Oncogene Proteins, Fusion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...