Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 255
Filter
1.
J Biol Chem ; 300(4): 107138, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447794

ABSTRACT

Short tandem repeats are inherently unstable during DNA replication depending on repeat length, and the expansion of the repeat length in the human genome is responsible for repeat expansion disorders. Pentanucleotide AAGGG and ACAGG repeat expansions in intron 2 of the gene encoding replication factor C subunit 1 (RFC1) cause cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS) and other phenotypes of late-onset cerebellar ataxia. Herein, we reveal the structural polymorphism of the RFC1 repeats associated with CANVAS in vitro. Single-stranded AAGGG repeat DNA formed a hybrid-type G-quadruplex, whereas its RNA formed a parallel-type G-quadruplex with three layers. The RNA of the ACAGG repeat formed hairpin structure comprising C-G and G-C base pairs with A:A and GA:AG mismatched repeats. Furthermore, both pathogenic repeat RNAs formed more rigid structures than those of the nonpathogenic repeat RNAs. These findings provide novel insights into the structural polymorphism of the RFC1 repeats, which may be closely related to the disease mechanism of CANVAS.


Subject(s)
Cerebellar Ataxia , DNA Repeat Expansion , Peripheral Nervous System Diseases , Replication Protein C , Vestibular Diseases , Humans , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , G-Quadruplexes , Microsatellite Repeats , Polymorphism, Genetic , Replication Protein C/genetics , Replication Protein C/metabolism , Replication Protein C/chemistry , RNA/chemistry , RNA/genetics , RNA/metabolism , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Vestibular Diseases/genetics , Vestibular Diseases/metabolism
2.
J Chem Neuroanat ; 137: 102399, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38401660

ABSTRACT

Cerebellar ataxia (CA) is a condition in which cerebellar dysfunction results in movement disorders such as dysmetria, synergy and dysdiadochokinesia. This study investigates the therapeutic effects of elderberry (EB) diet on the 3-acetylpyridine-induced (3-AP) CA rat model. First, CA rat models were generated by 3-AP administration followed by elderberry diet treatment containing 2 % EB for 8 consecutive weeks. Motor performance, electromyographic activity and gene expression were then evaluated. The number of Purkinje neurons were evaluated by stereological methods. Immunohistochemistry for the microgliosis, astrogliosis and apoptosis marker caspase-3 was also performed. In addition, the morphology of microglia and astrocytes was assessed using the Sholl analysis method. The results showed that EB diet administration in a 3-AP ataxia model improved motor coordination, locomotor activity and neuro-muscular function, prevented Purkinje neurons degeneration, increased microglia and astrocyte complexity and reduced cell soma size. Moreover, EB diet administration decreased apoptosis in cerebellum of 3-AP ataxic model. In addition, elderberry diet treatment decreased the expression of inflammatory, apoptotic and necroptotic genes and increased the expression of antioxidant-related genes. The results suggest that the EB diet attenuates 3-AP-induced neuroinflammation leading to cell death and improves motor performance. Thus, the EB diet could be used as a therapeutic procedure for CA due to its neuroprotective effects.


Subject(s)
Cerebellar Ataxia , Disease Models, Animal , Pyridines , Animals , Rats , Cerebellar Ataxia/pathology , Cerebellar Ataxia/metabolism , Male , Cell Death , Neuroinflammatory Diseases/pathology , Neuroinflammatory Diseases/metabolism , Purkinje Cells/pathology , Purkinje Cells/metabolism , Motor Activity/physiology , Diet , Rats, Wistar , Microglia/metabolism , Microglia/pathology , Cerebellum/pathology , Cerebellum/metabolism
3.
Mol Neurobiol ; 60(9): 5395-5410, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37314654

ABSTRACT

Niemann-Pick type C1 (NPC1) disease is a lysosomal lipid storage disorder due to mutations in the NPC1 gene resulting in the accumulation of cholesterol within the endosomal/lysosomal compartments. The prominent feature of the disorder is the progressive Purkinje cell degeneration leading to ataxia.In a mouse model of NPC1 disease, we have previously demonstrated that impaired Sonic hedgehog signaling causes defective proliferation of granule cells (GCs) and abnormal cerebellar morphogenesis. Studies conducted on cortical and hippocampal neurons indicate a functional interaction between Sonic hedgehog and brain-derived neurotrophic factor (BDNF) expression, leading us to hypothesize that BDNF signaling may be altered in Npc1 mutant mice, contributing to the onset of cerebellar alterations present in NPC1 disease before the appearance of signs of ataxia.We characterized the expression/localization patterns of the BDNF and its receptor, tropomyosin-related kinase B (TrkB), in the early postnatal and young adult cerebellum of the Npc1nmf164 mutant mouse strain.In Npc1nmf164 mice, our results show (i) a reduced expression of cerebellar BDNF and pTrkB in the first 2 weeks postpartum, phases in which most GCs complete the proliferative/migrative program and begin differentiation; (ii) an altered subcellular localization of the pTrkB receptor in GCs, both in vivo and in vitro; (iii) reduced chemotactic response to BDNF in GCs cultured in vitro, associated with impaired internalization of the activated TrkB receptor; (iv) an overall increase in dendritic branching in mature GCs, resulting in impaired differentiation of the cerebellar glomeruli, the major synaptic complex between GCs and mossy fibers.


Subject(s)
Brain-Derived Neurotrophic Factor , Cerebellar Ataxia , Female , Mice , Animals , Brain-Derived Neurotrophic Factor/metabolism , Hedgehog Proteins/metabolism , Neurons/metabolism , Cerebellum/metabolism , Purkinje Cells/metabolism , Cell Differentiation , Cerebellar Ataxia/metabolism , Cholesterol/metabolism
4.
Neurobiol Dis ; 183: 106157, 2023 07.
Article in English | MEDLINE | ID: mdl-37209925

ABSTRACT

Mitochondrial deficits have been observed in animal models of Autosomal-recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) and in patient-derived fibroblasts. We investigated whether mitochondrial function could be restored in Sacs-/- mice, a mouse model of ARSACS, using the mitochondrial-targeted antioxidant ubiquinone MitoQ. After 10weeks of chronic MitoQ administration in drinking water, we partially reversed motor coordination deficits in Sacs-/- mice but did not affect litter-matched wild-type control mice. MitoQ administration led to a restoration of superoxide dismutase 2 (SOD2) in cerebellar Purkinje cell somata without altering Purkinje cell firing deficits. Purkinje cells in anterior vermis of Sacs-/- mice normally undergo cell death in ARSACS; however, Purkinje cells numbers were elevated after chronic MitoQ treatment. Furthermore, Purkinje cell innervation of target neurons in the cerebellar nuclei of Sacs-/- mice was also partially restored with MitoQ treatment. Our data suggest that MitoQ is a potential therapeutic treatment for ARSACS and that it improves motor coordination via increasing cerebellar Purkinje cell mitochondria function and reducing Purkinje cell death.


Subject(s)
Cerebellar Ataxia , Purkinje Cells , Animals , Mice , Purkinje Cells/metabolism , Antioxidants/pharmacology , Ataxia/drug therapy , Ataxia/metabolism , Cerebellar Ataxia/metabolism , Mitochondria , Disease Models, Animal
5.
Int J Mol Sci ; 24(7)2023 Mar 23.
Article in English | MEDLINE | ID: mdl-37047062

ABSTRACT

Cerebellar ataxia is a neurodegenerative disorder with no definitive treatment. Although previous study demonstrated the neuroprotective effects of Hericium erinaceus (H.E.), the mechanisms of H.E. treatment on the neuroinflammatory response, neurotransmission, and related metabolites remain largely unknown. We demonstrated that 3-AP rats treated with 25 mg/kg H.E. extracts had improved motor coordination and balance in the accelerated rotarod and rod tests. We showed that the H.E. treatment upregulated the expression of Tgfb1, Tgfb2, and Smad3 genes to levels comparable to those in the non-3-AP control group. Interestingly, we also observed a significant correlation between Tgfb2 gene expression and rod test performance in the 3-AP saline group, but not in the non-3-AP control or H.E.+3-AP groups, indicating a relationship between Tgfb2 gene expression and motor balance in the 3-AP rat model. Additionally, we also found that the H.E. treatment increased mitochondrial COX-IV protein expression and normalized dopamine-serotonin neurotransmission and metabolite levels in the cerebellum of the H.E.+3-AP group compared to the 3-AP saline group. In conclusion, our findings suggest that the H.E. treatment improved motor function in the 3-AP rat model, which was potentially mediated through neuroprotective mechanisms involving TGFB2-Smad3 signaling via normalization of neurotransmission and metabolic pathways.


Subject(s)
Cerebellar Ataxia , Rats , Animals , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , Hericium , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use
6.
Brain ; 146(9): 3836-3850, 2023 09 01.
Article in English | MEDLINE | ID: mdl-36960552

ABSTRACT

COQ8A-ataxia is a rare form of neurodegenerative disorder due to mutations in the COQ8A gene. The encoded mitochondrial protein is involved in the regulation of coenzyme Q10 biosynthesis. Previous studies on the constitutive Coq8a-/- mice indicated specific alterations of cerebellar Purkinje neurons involving altered electrophysiological function and dark cell degeneration. In the present manuscript, we extend our understanding of the contribution of Purkinje neuron dysfunction to the pathology. By generating a Purkinje-specific conditional COQ8A knockout, we demonstrate that loss of COQ8A in Purkinje neurons is the main cause of cerebellar ataxia. Furthermore, through in vivo and in vitro approaches, we show that COQ8A-depleted Purkinje neurons have abnormal dendritic arborizations, altered mitochondria function and intracellular calcium dysregulation. Furthermore, we demonstrate that oxidative phosphorylation, in particular Complex IV, is primarily altered at presymptomatic stages of the disease. Finally, the morphology of primary Purkinje neurons as well as the mitochondrial dysfunction and calcium dysregulation could be rescued by CoQ10 treatment, suggesting that CoQ10 could be a beneficial treatment for COQ8A-ataxia.


Subject(s)
Cerebellar Ataxia , Mice , Animals , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , Purkinje Cells/pathology , Calcium/metabolism , Ataxia/drug therapy , Ataxia/genetics , Ataxia/metabolism , Mitochondria/metabolism
7.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982413

ABSTRACT

The cytosolic carboxypeptidase (CCP) 1 protein, encoded by CCP1, is expressed in cerebellar Purkinje cells (PCs). The dysfunction of CCP1 protein (caused by CCP1 point mutation) and the deletion of CCP1 protein (caused by CCP1 gene knockout) all lead to the degeneration of cerebellar PCs, which leads to cerebellar ataxia. Thus, two CCP1 mutants (i.e., Ataxia and Male Sterility [AMS] mice and Nna1 knockout [KO] mice) are used as disease models. We investigated the cerebellar CCP1 distribution in wild-type (WT), AMS and Nna1 KO mice on postnatal days (P) 7-28 to investigate the differential effects of CCP protein deficiency and disorder on cerebellar development. Immunohistochemical and immunofluorescence studies revealed significant differences in the cerebellar CCP1 expression in WT and mutant mice of P7 and P15, but no significant difference between AMS and Nna1 KO mice. Electron microscopy showed slight abnormality in the nuclear membrane structure of PCs in the AMS and Nna1 KO mice at P15 and significant abnormality with depolymerization and fragmentation of microtubule structure at P21. Using two CCP1 mutant mice strains, we revealed the morphological changes of PCs at postnatal stages and indicated that CCP1 played an important role in cerebellar development, most likely via polyglutamylation.


Subject(s)
Cerebellar Ataxia , Serine-Type D-Ala-D-Ala Carboxypeptidase , Animals , Male , Mice , Ataxia/genetics , Cerebellar Ataxia/metabolism , GTP-Binding Proteins/metabolism , Mice, Knockout , Protein Processing, Post-Translational , Purkinje Cells/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/genetics , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Tubulin/genetics , Tubulin/metabolism
8.
Front Immunol ; 14: 1289175, 2023.
Article in English | MEDLINE | ID: mdl-38332912

ABSTRACT

Background: Autoantibodies are useful biomarkers for the early detection and diagnosis of autoimmune cerebellar ataxia (ACA). Objective: To identify novel autoantibody candidates in ACA patients. Methods: Patients with cerebellar ataxia of unknown cause were recruited from July 2018 to February 2023. Anti-neural autoantibodies in patient samples were detected by tissue-based indirect immunofluorescence assay (TBA) on rat cerebellum sections. TBA-positive samples were further screened for well-established anti-neural autoantibodies using commercial kits. Tissue-immunoprecipitation (TIP) and subsequent mass spectrometric (MS) analysis were used to explore the target antigens of autoantibodies in samples that were TBA-positive but negative for known autoantibodies. The specific binding between autoantibodies and the identified target antigen was confirmed by neutralization experiments, recombinant cell-based indirect immunofluorescence assay (CBA), and western blotting experiments. Results: The eukaryotic translation elongation factor 1 delta (EEF1D) protein was identified as a target antigen of autoantibodies in samples from a 43-year-old female ACA patient, while the specific binding of autoantibodies and EEF1D was confirmed by subsequent experiments. A second anti-EEF1D autoantibody-positive ACA patient, a 59-year-old female, was detected in simultaneous screening. The main clinical manifestations in each of the two patients were cerebellar syndrome, such as unsteady walking and limb ataxia. Both patients received immunotherapy, including corticosteroids, intravenous immunoglobulin, and mycophenolate mofetil. Their outcomes provided evidence to support the effectiveness of immunotherapy, but the cerebellar atrophy that occurred before treatment may be irreversible. Conclusion: In the current study, we identified anti-EEF1D autoantibody as a novel autoantibody candidate in ACA. Its pathological roles and diagnostic value need to be further verified in larger-scale studies.


Subject(s)
Autoantibodies , Cerebellar Ataxia , Female , Humans , Adult , Middle Aged , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Peptide Elongation Factor 1 , Biomarkers , Immunotherapy
9.
Cell Death Dis ; 13(10): 855, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36207321

ABSTRACT

Calcium concentration must be finely tuned in all eukaryotic cells to ensure the correct performance of its signalling function. Neuronal activity is exquisitely dependent on the control of Ca2+ homeostasis: its alterations ultimately play a pivotal role in the origin and progression of many neurodegenerative processes. A complex toolkit of Ca2+ pumps and exchangers maintains the fluctuation of cytosolic Ca2+ concentration within the appropriate threshold. Two ubiquitous (isoforms 1 and 4) and two neuronally enriched (isoforms 2 and 3) of the plasma membrane Ca2+ATPase (PMCA pump) selectively regulate cytosolic Ca2+ transients by shaping the sub-plasma membrane (PM) microdomains. In humans, genetic mutations in ATP2B1, ATP2B2 and ATP2B3 gene have been linked with hearing loss, cerebellar ataxia and global neurodevelopmental delay: all of them were found to impair pump activity. Here we report three additional mutations in ATP2B3 gene corresponding to E1081Q, R1133Q and R696H amino acids substitution, respectively. Among them, the novel missense mutation (E1081Q) immediately upstream the C-terminal calmodulin-binding domain (CaM-BD) of the PMCA3 protein was present in two patients originating from two distinct families. Our biochemical and molecular studies on PMCA3 E1081Q mutant have revealed a splicing variant-dependent effect of the mutation in shaping the sub-PM [Ca2+]. The E1081Q substitution in the full-length b variant abolished the capacity of the pump to reduce [Ca2+] in the sub-PM microdomain (in line with the previously described ataxia-related PMCA mutations negatively affecting Ca2+ pumping activity), while, surprisingly, its introduction in the truncated a variant selectively increased Ca2+ extrusion activity in the sub-PM Ca2+ microdomains. These results highlight the importance to set a precise threshold of [Ca2+] by fine-tuning the sub-PM microdomains and the different contribution of the PMCA splice variants in this regulation.


Subject(s)
Cerebellar Ataxia , Plasma Membrane Calcium-Transporting ATPases/metabolism , Amino Acids , Ataxia/genetics , Ataxia/metabolism , Calcium/metabolism , Calmodulin/genetics , Cell Membrane/metabolism , Cerebellar Ataxia/genetics , Cerebellar Ataxia/metabolism , Humans , Mutation/genetics , Plasma Membrane Calcium-Transporting ATPases/chemistry , Plasma Membrane Calcium-Transporting ATPases/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
10.
Neuropeptides ; 96: 102295, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36280441

ABSTRACT

BACKGROUND: Cerebellar ataxia (CA) is a form of ataxia that adversely affects the cerebellum. This study aims to investigate the therapeutic effects of melittin (MEL) on a 3-acetylpyridine-induced (3-AP) cerebellar ataxia (CA) rat model. METHODS: Initially, CA rat models were generated by 3-AP administration followed by the subcutaneous injection of MEL. The open-field test was used for the evaluation of locomotion and anxiety. Immunohistochemistry was also conducted for the autophagy markers of LC3 and Beclin1. In the next step, the morphology of the astrocyte, the cell responsible for maintaining homeostasis in the CNS, was evaluated by the Sholl analysis. RESULTS: The findings suggested that the administration of MEL in a 3-AP model of ataxia improved locomotion and anxiety (P < 0.001), decreased the expression of LC3 (P < 0.01) and Beclin1 (P < 0.05), increased astrocyte complexity (P < 0.05) and reduced astrocyte cell soma size (P < 0.001). CONCLUSIONS: Overall, the findings imply that the MEL attenuates the 3-AP-induced autophagy, causes cell death and improves motor function. As such, it could be used as a therapeutic procedure for CA due to its neuroprotective effects.


Subject(s)
Cerebellar Ataxia , Melitten , Animals , Rats , Ataxia/metabolism , Autophagy , Beclin-1/metabolism , Cell Death , Cerebellar Ataxia/chemically induced , Cerebellar Ataxia/drug therapy , Cerebellar Ataxia/metabolism , Gliosis/metabolism , Melitten/pharmacology , Purkinje Cells , Rats, Sprague-Dawley
11.
Stem Cell Res ; 61: 102782, 2022 05.
Article in English | MEDLINE | ID: mdl-35421843

ABSTRACT

Spinal cerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease, is the result of abnormal repeat amplification of CAG of the ATXN3 gene. It is one of the main types of autosomal dominant ataxia, with motor symptoms of cerebellar ataxia, mainly accompanied by non-motor symptoms, such as ocular symptoms, psychiatric symptoms, and nutritional disorders. Currently, no effective treatment is available for patients with SCA3. The construction of induced pluripotent stem cells (iPSCs) from two SCA3 patients (14/74 CAG repeats) will be an excellent tool for studying SCA3 disease mechanisms and for drug screening.


Subject(s)
Cerebellar Ataxia , Induced Pluripotent Stem Cells , Ataxin-3/genetics , Ataxin-3/metabolism , Cerebellar Ataxia/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Mutation/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism
12.
Alcohol Clin Exp Res ; 45(10): 2006-2016, 2021 10.
Article in English | MEDLINE | ID: mdl-34453331

ABSTRACT

BACKGROUND: Alcohol intoxication produces ataxia by affecting the cerebellum, which coordinates movements. Fragile X mental retardation (FMR) protein is a complex regulator of RNA and synaptic plasticity implicated in fragile X-associated tremor/ataxia syndrome, which features ataxia and increased Fmr1 mRNA expression resulting from epigenetic dysregulation of FMRP. We recently demonstrated that acute ethanol-induced ataxia is associated with increased cerebellar Fmr1 gene expression via histone modifications in rats, but it is unknown whether similar behavioral and molecular changes occur following chronic ethanol exposure. Here, we investigated the effects of chronic ethanol exposure on ataxia and epigenetically regulated changes in Fmr1 expression in the cerebellum. METHODS: Male adult Sprague-Dawley rats were trained on the accelerating rotarod and then fed with chronic ethanol or a control Lieber-DeCarli diet while undergoing periodic behavioral testing for ataxia during ethanol exposure and withdrawal. Cerebellar tissues were analyzed for expression of the Fmr1 gene and its targets using a real-time quantitative polymerase chain reaction assay. The epigenetic regulation of Fmr1 was also investigated using a chromatin immunoprecipitation assay. RESULTS: Ataxic behavior measured by the accelerating rotarod behavioral test developed during chronic ethanol treatment and persisted at both the 8-h and 24-h withdrawal time points compared to control diet-fed rats. In addition, chronic ethanol treatment resulted in up-regulated expression of Fmr1 mRNA and increased activating epigenetic marks H3K27 acetylation and H3K4 trimethylation at 2 sites within the Fmr1 promoter. Finally, measurement of the expression of relevant FMRP mRNA targets in the cerebellum showed that chronic ethanol up-regulated cAMP response element binding (CREB) Creb1, Psd95, Grm5, and Grin2b mRNA expression without altering Grin2a, Eaa1, or histone acetyltransferases CREB binding protein (Cbp) or p300 mRNA transcripts. CONCLUSIONS: These results suggest that epigenetic regulation of Fmr1 and subsequent FMRP regulation of target mRNA transcripts constitute neuroadaptations in the cerebellum that may underlie the persistence of ataxic behavior during chronic ethanol exposure and withdrawal.


Subject(s)
Central Nervous System Depressants/adverse effects , Cerebellar Ataxia/chemically induced , Cerebellum/drug effects , Ethanol/adverse effects , Fragile X Mental Retardation Protein/metabolism , Alcoholic Intoxication/etiology , Alcoholic Intoxication/metabolism , Animals , Central Nervous System Depressants/administration & dosage , Cerebellar Ataxia/metabolism , Cerebellum/metabolism , Epigenesis, Genetic/drug effects , Ethanol/administration & dosage , Histone Code/drug effects , Male , Rats, Sprague-Dawley
13.
PLoS Genet ; 17(8): e1009716, 2021 08.
Article in English | MEDLINE | ID: mdl-34339417

ABSTRACT

We investigated a hereditary cerebellar ataxia in Belgian Shepherd dogs. Affected dogs developed uncoordinated movements and intention tremor at two weeks of age. The severity of clinical signs was highly variable. Histopathology demonstrated atrophy of the CNS, particularly in the cerebellum. Combined linkage and homozygosity mapping in a family with four affected puppies delineated a 52 Mb critical interval. The comparison of whole genome sequence data of one affected dog to 735 control genomes revealed a private homozygous structural variant in the critical interval, Chr4:66,946,539_66,963,863del17,325. This deletion includes the entire protein coding sequence of SELENOP and is predicted to result in complete absence of the encoded selenoprotein P required for selenium transport into the CNS. Genotypes at the deletion showed the expected co-segregation with the phenotype in the investigated family. Total selenium levels in the blood of homozygous mutant puppies of the investigated litter were reduced to about 30% of the value of a homozygous wildtype littermate. Genotyping >600 Belgian Shepherd dogs revealed an additional homozygous mutant dog. This dog also suffered from pronounced ataxia, but reached an age of 10 years. Selenop-/- knock-out mice were reported to develop ataxia, but their histopathological changes were less severe than in the investigated dogs. Our results demonstrate that deletion of the SELENOP gene in dogs cause a defect in selenium transport associated with CNS atrophy and cerebellar ataxia (CACA). The affected dogs represent a valuable spontaneous animal model to gain further insights into the pathophysiological consequences of CNS selenium deficiency.


Subject(s)
Cerebellar Ataxia/genetics , Selenoprotein P/genetics , Selenoprotein P/metabolism , Animals , Atrophy/physiopathology , Central Nervous System/physiology , Cerebellar Ataxia/metabolism , Dog Diseases/genetics , Dogs , Female , Genetic Linkage/genetics , Genome/genetics , Genotype , Homozygote , Male , Phenotype , Whole Genome Sequencing/methods
14.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445111

ABSTRACT

Autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS) is a multisystem hereditary ataxia associated with mutations in SACS, which encodes sacsin, a protein of still only partially understood function. Although mouse models of ARSACS mimic largely the disease progression seen in humans, their use in the validation of effective therapies has not yet been proposed. Recently, the teleost Danio rerio has attracted increasing attention as a vertebrate model that allows rapid and economical screening, of candidate molecules, and thus combines the advantages of whole-organism phenotypic assays and in vitro high-throughput screening assays. Through CRISPR/Cas9-based mutagenesis, we generated and characterized a zebrafish sacs-null mutant line that replicates the main features of ARSACS. The sacs-null fish showed motor impairment, hindbrain atrophy, mitochondrial dysfunction, and reactive oxygen species accumulation. As proof of principle for using these mutant fish in high-throughput screening studies, we showed that both acetyl-DL-leucine and tauroursodeoxycholic acid improved locomotor and biochemical phenotypes in sacs-/- larvae treated with these neuroprotective agents, by mediating significant rescue of the molecular functions altered by sacsin loss. Taken together, the evidence here reported shows the zebrafish to be a valuable model organism for the identification of novel molecular mechanisms and for efficient and rapid in vivo optimization and screening of potential therapeutic compounds. These findings may pave the way for new interventions targeting the earliest phases of Purkinje cell degeneration in ARSACS.


Subject(s)
Heat-Shock Proteins/metabolism , Neuroprotective Agents/metabolism , Zebrafish/metabolism , Animals , Animals, Genetically Modified/metabolism , Ataxia/metabolism , Cerebellar Ataxia/metabolism , Disease Models, Animal , Disease Progression , Muscle Spasticity/metabolism , Mutation/genetics , Phenotype , Purkinje Cells/metabolism , Spinocerebellar Ataxias/congenital , Spinocerebellar Ataxias/metabolism
15.
J Biol Chem ; 297(3): 101089, 2021 09.
Article in English | MEDLINE | ID: mdl-34416235

ABSTRACT

Familial British dementia and familial Danish dementia are neurodegenerative disorders caused by mutations in the gene integral membrane protein 2B (ITM2b) encoding BRI2, which tunes excitatory synaptic transmission at both presynaptic and postsynaptic termini. In addition, BRI2 interacts with and modulates proteolytic processing of amyloid-ß precursor protein (APP), whose mutations cause familial forms of Alzheimer's disease (AD) (familial AD). To study the pathogenic mechanisms triggered by the Danish mutation, we generated rats carrying the Danish mutation in the rat Itm2b gene (Itm2bD rats). Given the BRI2/APP interaction and the widely accepted relevance of human amyloid ß (Aß), a proteolytic product of APP, to AD, Itm2bD rats were engineered to express two humanized App alleles and produce human Aß. Here, we studied young Itm2bD rats to investigate early pathogenic changes in these diseases. We found that periadolescent Itm2bD rats not only present subtle changes in human Aß levels along with decreased spontaneous glutamate release and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor-mediated responses but also had increased short-term synaptic facilitation in the hippocampal Schaeffer-collateral pathway. These alterations in excitatory interneuronal communication can impair learning and memory processes and were akin to those observed in adult mice producing rodent Aß and carrying either the Danish or British mutations in the mouse Itm2b gene. Collectively, the data show that the pathogenic Danish mutation alters the physiological function of BRI2 at glutamatergic synapses across species and early in life. Future studies will determine whether this phenomenon represents an early pathogenic event in human dementia.


Subject(s)
Cataract/physiopathology , Cerebellar Ataxia/physiopathology , Deafness/physiopathology , Dementia/physiopathology , Membrane Proteins/genetics , Synaptic Transmission/physiology , Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Cataract/metabolism , Cerebellar Ataxia/metabolism , Deafness/metabolism , Dementia/genetics , Dementia/metabolism , Disease Models, Animal , Excitatory Amino Acid Agents/metabolism , Female , Male , Membrane Proteins/metabolism , Memory , Presynaptic Terminals/metabolism , Rats , Receptors, Glutamate/metabolism , Synapses/metabolism
16.
Clin Immunol ; 229: 108776, 2021 08.
Article in English | MEDLINE | ID: mdl-34118401

ABSTRACT

Early-onset ataxia with ocular motor apraxia and hypoalbuminemia (EAOH) is a neurodegenerative disorder caused by mutation in the aprataxin (APTX)-coding gene APTX, which is involved in DNA single-strand break repair (SSBR). The neurological abnormalities associated with EAOH are similar to those observed in patients with ataxia-telangiectasia. However, the immunological abnormalities in patients with EAOH have not been described. In this study, we report that EAOH patients have immunological abnormalities, including lymphopenia; decreased levels of CD4+ T-cells, CD8+ T-cells, and B-cells; hypogammaglobulinemia; low T-cell recombination excision circles and kappa-deleting element recombination circles; and oligoclonality of T-cell receptor ß-chain variable repertoire. These immunological abnormalities vary among the EAOH patients. Additionally, mild radiosensitivity in the lymphocytes obtained from the patients with EAOH was demonstrated. These findings suggested that the immunological abnormalities and mild radiosensitivity evident in patients with EAOH could be probably caused by the DNA repair defects.


Subject(s)
Apraxias/immunology , Cerebellar Ataxia/congenital , Hypoalbuminemia/immunology , Adolescent , Adult , Apraxias/genetics , Apraxias/metabolism , Case-Control Studies , Cerebellar Ataxia/genetics , Cerebellar Ataxia/immunology , Cerebellar Ataxia/metabolism , Child , DNA Breaks, Single-Stranded , DNA Repair/genetics , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , Female , Genes, T-Cell Receptor , Genetic Variation , Humans , Hypoalbuminemia/genetics , Hypoalbuminemia/metabolism , Male , Middle Aged , Mutation , Nuclear Proteins/genetics , Radiation Tolerance/genetics , Radiation Tolerance/immunology , T-Lymphocytes/immunology , Young Adult
17.
Neurobiol Dis ; 156: 105424, 2021 08.
Article in English | MEDLINE | ID: mdl-34118418

ABSTRACT

Neuroinflammatory changes involving neuronal HMGB1 release and astrocytic NF-κB nuclear translocation occur following cortical spreading depolarization (CSD) in wildtype (WT) mice but it is unknown to what extent this occurs in the migraine brain. We therefore investigated in familial hemiplegic migraine type 1 (FHM1) knock-in mice, which express an intrinsic hyperexcitability phenotype, the extent of neuroinflammation without and after CSD. CSD was evoked in one hemisphere by pinprick (single CSD) or topical KCl application (multiple CSDs). Neuroinflammatory (HMGB1, NF-κB) and neuronal activation (pERK) markers were investigated by immunohistochemistry in the brains of WT and FHM1 mutant mice without and after CSD. Effects of NMDA receptor antagonism on basal and CSD-induced neuroinflammatory changes were examined by, respectively, systemically administered MK801 and ifenprodil or topical MK801 application. In FHM1 mutant mice, CSD caused enhanced neuronal HMGB1 release and astrocytic NF-κB nuclear translocation in the cortex and subcortical areas that were equally high in both hemispheres. In WT mice such effects were only pronounced in the hemisphere in which CSD was induced. Neuroinflammatory responses were associated with pERK expression indicating neuronal activation. Upon CSD, contralateral cortical and striatal HMGB1 release was reduced by topical application of MK801 in the hemisphere contralateral to the one in which CSD was induced. This study reveals that neuroinflammatory activation after CSD is widespread and extends to the contralateral hemisphere, particularly in brains of FHM1 mutant mice. Effective blockade of CSD-induced neuroinflammatory responses in the contralateral hemisphere in FHM1 mice by local NMDA receptor antagonism suggests that neuronal hyperexcitability-related neuroinflammation is relevant in migraine pathophysiology, but possibly also other neurological disorders in which spreading depolarization is involved.


Subject(s)
Brain/metabolism , Cerebellar Ataxia/metabolism , Cortical Spreading Depression/physiology , HMGB1 Protein/metabolism , Migraine Disorders/metabolism , NF-kappa B/metabolism , Parenchymal Tissue/metabolism , Animals , Astrocytes/metabolism , Brain/physiopathology , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Female , HMGB1 Protein/genetics , Humans , Mice , Mice, Transgenic , Migraine Disorders/genetics , Migraine Disorders/physiopathology , NF-kappa B/genetics , Parenchymal Tissue/physiopathology
18.
Brain ; 144(5): 1467-1481, 2021 06 22.
Article in English | MEDLINE | ID: mdl-33889951

ABSTRACT

Peroxiredoxin 3 (PRDX3) belongs to a superfamily of peroxidases that function as protective antioxidant enzymes. Among the six isoforms (PRDX1-PRDX6), PRDX3 is the only protein exclusively localized to the mitochondria, which are the main source of reactive oxygen species. Excessive levels of reactive oxygen species are harmful to cells, inducing mitochondrial dysfunction, DNA damage, lipid and protein oxidation and ultimately apoptosis. Neuronal cell damage induced by oxidative stress has been associated with numerous neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Leveraging the large aggregation of genomic ataxia datasets from the PREPARE (Preparing for Therapies in Autosomal Recessive Ataxias) network, we identified recessive mutations in PRDX3 as the genetic cause of cerebellar ataxia in five unrelated families, providing further evidence for oxidative stress in the pathogenesis of neurodegeneration. The clinical presentation of individuals with PRDX3 mutations consists of mild-to-moderate progressive cerebellar ataxia with concomitant hyper- and hypokinetic movement disorders, severe early-onset cerebellar atrophy, and in part olivary and brainstem degeneration. Patient fibroblasts showed a lack of PRDX3 protein, resulting in decreased glutathione peroxidase activity and decreased mitochondrial maximal respiratory capacity. Moreover, PRDX3 knockdown in cerebellar medulloblastoma cells resulted in significantly decreased cell viability, increased H2O2 levels and increased susceptibility to apoptosis triggered by reactive oxygen species. Pan-neuronal and pan-glial in vivo models of Drosophila revealed aberrant locomotor phenotypes and reduced survival times upon exposure to oxidative stress. Our findings reveal a central role for mitochondria and the implication of oxidative stress in PRDX3 disease pathogenesis and cerebellar vulnerability and suggest targets for future therapeutic approaches.


Subject(s)
Cerebellar Ataxia/genetics , Oxidative Stress/genetics , Peroxiredoxin III/genetics , Adult , Animals , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Drosophila , Female , Humans , Loss of Function Mutation , Male , Middle Aged , Pedigree
19.
Int J Mol Sci ; 22(6)2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33804256

ABSTRACT

Lysosomal acid phosphatase 2 (Acp2) mutant mice (naked-ataxia, nax) have a severe cerebellar cortex defect with a striking reduction in the number of granule cells. Using a combination of in vivo and in vitro immunohistochemistry, Western blotting, BrdU assays, and RT-qPCR, we show downregulation of MYCN and dysregulation of the SHH signaling pathway in the nax cerebellum. MYCN protein expression is significantly reduced at P10, but not at the peak of proliferation at around P6 when the number of granule cells is strikingly reduced in the nax cerebellum. Despite the significant role of the SHH-MycN pathway in granule cell proliferation, our study suggests that a broader molecular pathway and additional mechanisms regulating granule cell development during the clonal expansion period are impaired in the nax cerebellum. In particular, our results indicate that downregulation of the protein synthesis machinery may contribute to the reduced number of granule cells in the nax cerebellum.


Subject(s)
Acid Phosphatase/genetics , Cerebellar Ataxia/genetics , Cerebellar Cortex/metabolism , Hedgehog Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cerebellar Ataxia/metabolism , Cerebellar Ataxia/pathology , Cerebellar Cortex/abnormalities , Cerebellar Cortex/pathology , Cytoplasmic Granules/genetics , Cytoplasmic Granules/pathology , Disease Models, Animal , Gene Expression Regulation, Developmental , Humans , Lysosomes/genetics , Lysosomes/pathology , Mice , Mutation , Neurons/metabolism , Neurons/pathology , Purkinje Cells/metabolism , Purkinje Cells/pathology , Signal Transduction/genetics
20.
Int J Mol Sci ; 22(4)2021 Feb 12.
Article in English | MEDLINE | ID: mdl-33673348

ABSTRACT

The cerebellum, the region of the brain primarily responsible for motor coordination and balance, also contributes to non-motor functions, such as cognition, speech, and language comprehension. Maldevelopment and dysfunction of the cerebellum lead to cerebellar ataxia and may even be associated with autism, depression, and cognitive deficits. Hence, normal development of the cerebellum and its neuronal circuitry is critical for the cerebellum to function properly. Although nine major types of cerebellar neurons have been identified in the cerebellar cortex to date, the exact functions of each type are not fully understood due to a lack of cell-specific markers in neurons that renders cell-specific labeling and functional study by genetic manipulation unfeasible. The availability of cell-specific markers is thus vital for understanding the role of each neuronal type in the cerebellum and for elucidating the interactions between cell types within both the developing and mature cerebellum. This review discusses various technical approaches and recent progress in the search for cell-specific markers for cerebellar neurons.


Subject(s)
Cerebellar Ataxia/metabolism , Cerebellar Cortex/metabolism , Neurons/metabolism , Animals , Biomarkers/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...