Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Int J Mol Sci ; 24(2)2023 Jan 12.
Article in English | MEDLINE | ID: mdl-36675067

ABSTRACT

Metabotropic glutamate receptor 1 (mGluR1) plays a crucial role in slow excitatory postsynaptic conductance, synapse formation, synaptic plasticity, and motor control. The GRM1 gene is expressed mainly in the brain, with the highest expression in the cerebellum. Mutations in the GRM1 gene have previously been known to cause autosomal recessive and autosomal dominant spinocerebellar ataxias. In this study, whole-exome sequencing of a patient from a family of Azerbaijani origin with a diagnosis of congenital cerebellar ataxia was performed, and a new homozygous missense mutation in the GRM1 gene was identified. The mutation leads to the homozygous amino acid substitution of p.Thr824Arg in an evolutionarily highly conserved region encoding the transmembrane domain 7, which is critical for ligand binding and modulating of receptor activity. This is the first report in which a mutation has been identified in the last transmembrane domain of the mGluR1, causing a congenital autosomal recessive form of cerebellar ataxia with no obvious intellectual disability. Additionally, we summarized all known presumable pathogenic genetic variants in the GRM1 gene to date. We demonstrated that multiple rare variants in the GRM1 underlie a broad diversity of clinical neurological and behavioral phenotypes depending on the nature and protein topology of the mutation.


Subject(s)
Cerebellar Ataxia , Intellectual Disability , Receptors, Metabotropic Glutamate , Spinocerebellar Degenerations , Humans , Cerebellar Ataxia/congenital , Cerebellar Ataxia/diagnosis , Cerebellar Ataxia/genetics , Intellectual Disability/genetics , Mutation , Pedigree , Receptors, Metabotropic Glutamate/genetics , Spinocerebellar Degenerations/congenital , Spinocerebellar Degenerations/genetics
2.
Int J Mol Sci ; 22(18)2021 Sep 14.
Article in English | MEDLINE | ID: mdl-34576077

ABSTRACT

Kv1.2 channels, encoded by the KCNA2 gene, are localized in the central and peripheral nervous system, where they regulate neuronal excitability. Recently, heterozygous mutations in KCNA2 have been associated with a spectrum of symptoms extending from epileptic encephalopathy, intellectual disability, and cerebellar ataxia. Patients are treated with a combination of antiepileptic drugs and 4-aminopyridine (4-AP) has been recently trialed in specific cases. We identified a novel variant in KCNA2, E236K, in a Serbian proband with non-progressive congenital ataxia and early onset epilepsy, treated with sodium valproate. To ascertain the pathogenicity of E236K mutation and to verify its sensitivity to 4-AP, we transfected HEK 293 cells with Kv1.2 WT or E236K cDNAs and recorded potassium currents through the whole-cell patch-clamp. In silico analysis supported the electrophysiological data. E236K channels showed voltage-dependent activation shifted towards negative potentials and slower kinetics of deactivation and activation compared with Kv1.2 WT. Heteromeric Kv1.2 WT+E236K channels, resembling the condition of the heterozygous patient, confirmed a mixed gain- and loss-of-function (GoF/LoF) biophysical phenotype. 4-AP inhibited both Kv1.2 and E236K channels with similar potency. Homology modeling studies of mutant channels suggested a reduced interaction between the residue K236 in the S2 segment and the gating charges at S4. Overall, the biophysical phenotype of E236K channels correlates with the mild end of the clinical spectrum reported in patients with GoF/LoF defects. The response to 4-AP corroborates existing evidence that KCNA2-disorders could benefit from variant-tailored therapeutic approaches, based on functional studies.


Subject(s)
4-Aminopyridine/therapeutic use , Cerebellar Ataxia/congenital , Cerebellar Ataxia/genetics , Epilepsy/drug therapy , Epilepsy/genetics , Kv1.2 Potassium Channel/genetics , Amino Acid Sequence , Brain/diagnostic imaging , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/drug therapy , Child , Child, Preschool , Epilepsy/diagnostic imaging , Humans , Infant , Kv1.2 Potassium Channel/chemistry , Magnetic Resonance Imaging , Male , Molecular Dynamics Simulation , Young Adult
3.
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
4.
Int J Mol Sci ; 22(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801522

ABSTRACT

(1) Background: A non-progressive congenital ataxia (NPCA) phenotype caused by ß-III spectrin (SPTBN2) mutations has emerged, mimicking spinocerebellar ataxia, autosomal recessive type 14 (SCAR14). The pattern of inheritance, however, resembles that of autosomal dominant classical spinocerebellar ataxia type 5 (SCA5). (2) Methods: In-depth phenotyping of two boys studied by a customized gene panel. Candidate variants were sought by structural modeling and protein expression. An extensive review of the literature was conducted in order to better characterize the SPTBN2-associated NPCA. (3) Results: Patients exhibited an NPCA with hypotonia, developmental delay, cerebellar syndrome, and cognitive deficits. Both probands presented with progressive global cerebellar volume loss in consecutive cerebral magnetic resonance imaging studies, characterized by decreasing midsagittal vermis relative diameter measurements. Cortical hyperintensities were observed on fluid-attenuated inversion recovery (FLAIR) images, suggesting a neurodegenerative process. Each patient carried a novel de novo SPTBN2 substitution: c.193A > G (p.K65E) or c.764A > G (p.D255G). Modeling and protein expression revealed that both mutations might be deleterious. (4) Conclusions: The reported findings contribute to a better understanding of the SPTBN2-associated phenotype. The mutations may preclude proper structural organization of the actin spectrin-based membrane skeleton, which, in turn, is responsible for the underlying disease mechanism.


Subject(s)
Cerebellar Ataxia/pathology , Mutation , Neurodegenerative Diseases/pathology , Spectrin/genetics , Age of Onset , Amino Acid Sequence , Cerebellar Ataxia/complications , Cerebellar Ataxia/congenital , Cerebellar Ataxia/genetics , Child , Cohort Studies , Genetic Association Studies , Humans , Male , Neurodegenerative Diseases/complications , Neurodegenerative Diseases/genetics , Neuroimaging , Phenotype , Protein Conformation , Sequence Homology , Spectrin/chemistry , Spectrin/metabolism , Syndrome
5.
Article in English | MEDLINE | ID: mdl-33101765

ABSTRACT

Background: Ataxia with oculomotor apraxia (AOA1) is characterized by early-onset progressive cerebellar ataxia with peripheral neuropathy, oculomotor apraxia and hypoalbuminemia and hypercholesterolemia. Case Report: A 23-year-old previously healthy woman presented with slowly-progressive gait impairment since the age of six years. Neurological examination revealed profound areflexia, chorea, generalized dystonia and oculomotor apraxia. Brain MRI revealed mild cerebellar atrophy and needle EMG showed axonal sensorimotor neuropathy. Whole exome sequencing revealed a mutation in the aprataxin gene. Discussion: AOA1 can present with choreoathetosis mixed with dystonic features, resembling ataxia-telangiectasia. This case is instructive since mixed and complex movement disorders is not very common in AOA1. Highlights: Ataxia with oculomotor apraxia type 1 (AOA1) is characterized by early-onset ataxia and oculomotor apraxia caused by variants in the APTX gene.Ataxia is usually not the sole movement abnormality in AOA1.Hyperkinetic movement disorders, especially chorea and dystonia, may occur.Mixed and complex movement disorders is not very common in AOA1.Patients with early-onset ataxia associated with mixed movement disorders should also be investigated for AOA1.


Subject(s)
Apraxias/physiopathology , Cerebellar Ataxia/congenital , Cerebellum/diagnostic imaging , Chorea/physiopathology , Dystonia/physiopathology , Hypoalbuminemia/physiopathology , Reflex, Abnormal/physiology , Apraxias/diagnostic imaging , Apraxias/genetics , Atrophy , Cerebellar Ataxia/diagnostic imaging , Cerebellar Ataxia/genetics , Cerebellar Ataxia/physiopathology , Cerebellar Diseases/physiopathology , Cerebellum/pathology , DNA-Binding Proteins/genetics , Electromyography , Female , Humans , Hypoalbuminemia/diagnostic imaging , Hypoalbuminemia/genetics , Nuclear Proteins/genetics , Young Adult
6.
Pediatr Neurol ; 104: 40-45, 2020 03.
Article in English | MEDLINE | ID: mdl-31836334

ABSTRACT

BACKGROUND: Congenital ataxias associated with cerebellar atrophy are clinically heterogeneous conditions with a variable age of onset and a diverse molecular basis. The hypothesis-free approach of genomic sequencing has led to the discovery of new genes implicated in these disorders and the identification of unexpected genotype-phenotype correlations. Although a recurrent heterozygous mutation (p.Arg1715His) in CACNA1G is known to cause adult-onset spinocerebellar ataxia 42 (SCA42*616795), gain-of-function mutations in this gene have recently been identified by whole exome sequencing (WES) in four children with cerebellar atrophy and ataxia, psychomotor delay, and other variable features. METHODS: We describe four children from unrelated families with cerebellar anomalies on magnetic resonance imaging (atrophy or hypoplasia of the cerebellar vermis), hypertonia, psychomotor and speech delay, severe intellectual disability, ophthalmologic features and peculiar dysmorphic traits. All patients underwent a trio-based WES analysis. Clinical records were used to characterize the clinical profile of this newly recognized disorder. RESULTS: Two previously reported de novo disease-causing mutations in CACNA1G (c.2881G>A, p.Ala961Thr and c.4591A>G, p.Met1531Val) were identified in these patients, providing further evidence of the specific impact of these variants. All four patients exhibit distinctive dysmorphic and ectodermal features which overlap those of the previously reported patients, allowing us to define the major features characterizing this homogeneous neurodevelopmental syndromic disorder associated with upregulated CACNA1G function. CONCLUSION: Our findings confirm the specific association between a narrow spectrum of missense mutations in CACNA1G and a novel syndrome with infantile-onset cerebellar ataxiaand provide a dysmorphologic delineation of this novel neurodevelopmental trait.


Subject(s)
Calcium Channels, T-Type/genetics , Cerebellar Diseases/genetics , Cerebellar Diseases/pathology , Cerebellar Diseases/physiopathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Age of Onset , Atrophy , Cerebellar Ataxia/congenital , Cerebellar Ataxia/genetics , Cerebellar Ataxia/pathology , Cerebellar Ataxia/physiopathology , Cerebellar Diseases/complications , Child , Child, Preschool , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/pathology , Female , Humans , Infant , Magnetic Resonance Imaging , Male , Mutation, Missense , Neurodevelopmental Disorders/etiology , Exome Sequencing
7.
Article in English | MEDLINE | ID: mdl-28652255

ABSTRACT

Hereditary ataxias are a clinically and genetically heterogeneous family of disorders defined by the inability to control gait and muscle coordination. Given the nonspecific symptoms of many hereditary ataxias, precise diagnosis relies on molecular genetic testing. To this end, we conducted whole-exome sequencing (WES) on a large consanguineous Iranian family with hereditary ataxia and oculomotor apraxia. WES in five affected and six unaffected individuals resulted in the identification of a homozygous novel stop-gain mutation in the APTX gene (c.739A>T; p.Lys247*) that segregates with the phenotype. Mutations in the APTX (OMIM 606350) gene are associated with ataxia with oculomotor apraxia type 1 (OMIM 208920).


Subject(s)
Apraxias/genetics , Cerebellar Ataxia/congenital , DNA-Binding Proteins/genetics , Hypoalbuminemia/genetics , Nuclear Proteins/genetics , Adult , Ataxia/complications , Ataxia/genetics , Cerebellar Ataxia/complications , Cerebellar Ataxia/genetics , Child , Child, Preschool , Consanguinity , DNA-Binding Proteins/metabolism , Female , Humans , Iran , Male , Middle Aged , Mutation/genetics , Nuclear Proteins/metabolism , Pedigree , Phenotype , Spinocerebellar Degenerations/complications , Spinocerebellar Degenerations/genetics
8.
Neurology ; 84(17): 1751-9, 2015 Apr 28.
Article in English | MEDLINE | ID: mdl-25841024

ABSTRACT

OBJECTIVES: In a large family of Algerian origin, we aimed to identify the genetic mutation segregating with simultaneous presence of adult-onset, paucisymptomatic, slowly progressive, cerebellar ataxia in 7 adults and congenital ataxia in 1 child, and then to assess the involvement of GRID2 mutations in 144 patients with congenital cerebellar ataxia. METHODS: We used a combined approach of linkage analysis and whole-exome sequencing in one family, and a targeted gene panel sequencing approach in 144 congenital ataxias. RESULTS: In the large family with spinocerebellar ataxia, we identified a missense mutation (c.1966C>G/p.Leu656Val) in the GRID2 gene, in a heterozygous state in adults, and in a homozygous state in one child with congenital ataxia, compatible with a semidominant transmission pattern. In 144 patients affected with congenital ataxia, we identified 2 missense de novo GRID2 mutations in 2 children (c.1960G>A/p.Ala654Thr, c.1961C>A/p.Ala654Asp). They affect the same amino acid as the previously described Lurcher mutation in mice; the variant in the large family concerns a nearby amino acid. CONCLUSIONS: In humans, GRID2 had only been involved in ataxia through complete loss-of-function mutations due to exon deletions. We report the first point mutations in this gene, with putative gain-of-function mechanisms, and a semidominant transmission as was observed in the Lurcher mice model. Of note, cerebellar ataxia is the core phenotype, but with variable severity ranging from very mild adult-onset to congenital-onset ataxias linked to both the heterozygous and homozygous state of the variant, and the position of the mutation.


Subject(s)
Cerebellar Ataxia/genetics , Point Mutation/genetics , Receptors, Glutamate/genetics , Adolescent , Adult , Aged , Algeria , Cerebellar Ataxia/congenital , Child , Child, Preschool , Exome , Female , Genetic Linkage , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Pedigree , Sequence Analysis, DNA
9.
Nature ; 506(7486): 111-5, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24362567

ABSTRACT

Faithful maintenance and propagation of eukaryotic genomes is ensured by three-step DNA ligation reactions used by ATP-dependent DNA ligases. Paradoxically, when DNA ligases encounter nicked DNA structures with abnormal DNA termini, DNA ligase catalytic activity can generate and/or exacerbate DNA damage through abortive ligation that produces chemically adducted, toxic 5'-adenylated (5'-AMP) DNA lesions. Aprataxin (APTX) reverses DNA adenylation but the context for deadenylation repair is unclear. Here we examine the importance of APTX to RNase-H2-dependent excision repair (RER) of a lesion that is very frequently introduced into DNA, a ribonucleotide. We show that ligases generate adenylated 5' ends containing a ribose characteristic of RNase H2 incision. APTX efficiently repairs adenylated RNA-DNA, and acting in an RNA-DNA damage response (RDDR), promotes cellular survival and prevents S-phase checkpoint activation in budding yeast undergoing RER. Structure-function studies of human APTX-RNA-DNA-AMP-Zn complexes define a mechanism for detecting and reversing adenylation at RNA-DNA junctions. This involves A-form RNA binding, proper protein folding and conformational changes, all of which are affected by heritable APTX mutations in ataxia with oculomotor apraxia 1. Together, these results indicate that accumulation of adenylated RNA-DNA may contribute to neurological disease.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/metabolism , Genome, Human/genetics , Nuclear Proteins/metabolism , RNA/metabolism , Adenosine Monophosphate/metabolism , Apraxias/genetics , Ataxia Telangiectasia/genetics , Cell Survival , Cerebellar Ataxia/congenital , DNA/chemistry , DNA Repair , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Exodeoxyribonucleases/chemistry , Exodeoxyribonucleases/metabolism , Humans , Hypoalbuminemia/genetics , Models, Molecular , Mutation/genetics , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleic Acid Conformation , Protein Conformation , Protein Folding , RNA/chemistry , Ribonuclease H/metabolism , S Phase Cell Cycle Checkpoints , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Structure-Activity Relationship , Zinc/metabolism
12.
AJNR Am J Neuroradiol ; 34(5): 925-34, S1-2, 2013 May.
Article in English | MEDLINE | ID: mdl-22595899

ABSTRACT

Ataxia is the principal symptom of many common neurologic diseases in childhood. Ataxias caused by dysfunction of the cerebellum occur in acute, intermittent, and progressive disorders. Most of the chronic progressive processes are secondary to degenerative and metabolic diseases. In addition, congenital malformation of the midbrain and hindbrain can also be present, with posterior fossa symptoms related to ataxia. Brain MR imaging is the most accurate imaging technique to investigate these patients, and imaging abnormalities include size, shape, and/or signal of the brain stem and/or cerebellum. Supratentorial and cord lesions are also common. This review will discuss a pattern-recognition approach to inherited cerebellar ataxia in childhood. The purpose is to provide a comprehensive discussion that ultimately could help neuroradiologists better manage this important topic in pediatric neurology.


Subject(s)
Brain/pathology , Cerebellar Ataxia/congenital , Cerebellar Ataxia/pathology , Image Enhancement/methods , Magnetic Resonance Imaging/methods , Pattern Recognition, Automated/methods , Child , Humans
13.
Article in Russian | MEDLINE | ID: mdl-23250602

ABSTRACT

AOA are autosomal recessive ataxias with a common feature of oculomotor apraxia (OA) - inability to coordinate eye movements. The group includes AOA1 (APTX gene), relatively common AOA2 (SETX gene) and AOA3 (PIK 3R5 gene) described in 2012 in a Saudi family. OA is typical also for Louis-Bar ataxia-telangiectasia and its variants. А first Russian AOA2 case confirmed by DNA test is presented. The disease in a 25-year-old male started in 18 years, in 23 years he lost independent walking due to incoordination and weakness. OA produced few symptoms and was not recorded previously. Sensorimotor axonal polyneuropathy was confirmed by EMG. MRI showed cerebellar atrophy. Alpha-fetoprotein level was tenfold raised. A hereditary ataxia was considered from the disease onset, and a number of genetic tests were performed, but AOA2 was recognized only seven years later. On direct sequencing of SETX exons 6-8 a novel frame-shift mutation с.2623-2626 del 4 in heterozygous state was detected which is sufficient for AOA2 confirmation; the allelic mutation is in search. Recently a first Russian AOA1 case in a 15-year-old girl was also confirmed in our laboratory: compound-heterozygosity for two novel APTX mutations was detected. Evidently AOA are underestimated in clinical diagnostics while DNA testing permits genetic prophylaxis in families. OA should be purposefully searched for in children and young adults suspicious of autosomal recessive ataxias.


Subject(s)
Apraxias/diagnosis , Apraxias/genetics , Ataxia Telangiectasia/diagnosis , Ataxia Telangiectasia/genetics , Hypoalbuminemia/diagnosis , Hypoalbuminemia/genetics , RNA Helicases/genetics , Adolescent , Adult , Cerebellar Ataxia/congenital , DNA/genetics , DNA Helicases , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Exons/genetics , Female , Genetic Carrier Screening , Humans , Magnetic Resonance Imaging , Male , Multifunctional Enzymes , Mutation , Nuclear Proteins/genetics
14.
PLoS One ; 7(11): e50570, 2012.
Article in English | MEDLINE | ID: mdl-23226316

ABSTRACT

Caytaxin is a highly-conserved protein, which is encoded by the Atcay/ATCAY gene. Mutations in Atcay/ATCAY have been identified as causative of cerebellar disorders such as the rare hereditary disease Cayman ataxia in humans, generalized dystonia in the dystonic (dt) rat, and marked motor defects in three ataxic mouse lines. While several lines of evidence suggest that Caytaxin plays a critical role in maintaining nervous system processes, the physiological function of Caytaxin has not been fully characterized. In the study presented here, we generated novel specific monoclonal antibodies against full-length Caytaxin to examine endogenous Caytaxin expression in wild type and Atcay mutant mouse lines. Caytaxin protein is absent from brain tissues in the two severely ataxic Atcay(jit) (jittery) and Atcay(swd) (sidewinder) mutant lines, and markedly decreased in the mildly ataxic/dystonic Atcay(ji-hes) (hesitant) line, indicating a correlation between Caytaxin expression and disease severity. As the expression of wild type human Caytaxin in mutant sidewinder and jittery mice rescues the ataxic phenotype, Caytaxin's physiological function appears to be conserved between the human and mouse orthologs. Across multiple species and in several neuronal cell lines Caytaxin is expressed as several protein isoforms, the two largest of which are caused by the usage of conserved methionine translation start sites. The work described in this manuscript presents an initial characterization of the Caytaxin protein and its expression in wild type and several mutant mouse models. Utilizing these animal models of human Cayman Ataxia will now allow an in-depth analysis to elucidate Caytaxin's role in maintaining normal neuronal function.


Subject(s)
Cerebellar Ataxia/congenital , Cerebellar Ataxia/genetics , Gene Expression Regulation , Nerve Tissue Proteins/genetics , Phenotype , Amino Acid Sequence , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity , Base Sequence , Brain/metabolism , Cell Line, Tumor , Cerebellar Ataxia/metabolism , Disease Models, Animal , Homozygote , Humans , Mice , Molecular Sequence Data , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Isoforms/metabolism , Species Specificity , Time Factors , Transcription Initiation Site
15.
Iran Biomed J ; 16(4): 223-5, 2012.
Article in English | MEDLINE | ID: mdl-23183622

ABSTRACT

BACKGROUND: Ataxia with oculomotor apraxia type 1 (AOA1) shows early onset with autosomal recessive inheritance and is caused by a mutation in the aprataxin (APTX) gene encoding for the APTX protein. METHODS: In this study, a 7-year-old girl born of a first-cousin consanguineous marriage was described with early-onset progressive ataxia and AOA, with increased cholesterol concentration and decreased albumin concentration in serum. PCR and direct DNA sequencing was performed after DNA extraction. RESULTS: Sequencing analysis revealed a novel homozygous deletion in c.643 and A>T single nucleotide polymorphism in c.641 in exon 6 of the APTX gene [ENST00000379825]. CONCLUSION: It seems that this region of exon 6 is probably a hot spot; however, no deletions have been reported in exon 6 yet.


Subject(s)
Apraxias/genetics , Ataxia Telangiectasia/genetics , DNA-Binding Proteins/genetics , Hypoalbuminemia/genetics , Mutation , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Cerebellar Ataxia/congenital , Child , Female , Humans
16.
Hum Mutat ; 33(2): 351-4, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22065524

ABSTRACT

Autosomal recessive ataxias are heterogeneous group of disorders characterized by cerebellar atrophy and peripheral sensorimotor neuropathy. Molecular characterization of this group of disorders identified a number of genes contributing to these overlapping phenotypes. Ataxia with oculomotor apraxia type 2 (AOA2) is an autosomal recessive form of ataxia caused by mutations in the SETX gene. We report on a consanguineous family with autosomal recessive inheritance and clinical characteristics of AOA2, and no mutations in the SETX gene. We mapped the AOA locus in this family to chromosome 17p12-p13. Sequencing of all genes in the refined region identified a homozygous missense mutation in PIK3R5 that was absent in 477 normal controls. Our characterization of the PIK3R5 protein and findings suggest that it may play a role in the development of the cerebellum and vermis.


Subject(s)
Apraxias/genetics , Ataxia Telangiectasia/genetics , Ataxia/genetics , Class Ib Phosphatidylinositol 3-Kinase/genetics , Hypoalbuminemia/genetics , Mutation, Missense , Phosphatidylinositol 3-Kinases/genetics , Adolescent , Adult , Animals , Apraxias/diagnosis , Ataxia/diagnosis , Ataxia Telangiectasia/diagnosis , Brain/pathology , Cerebellar Ataxia/congenital , Consanguinity , DNA Helicases , Female , Gene Order , Genetic Linkage , Homozygote , Humans , Hypoalbuminemia/diagnosis , Magnetic Resonance Imaging , Male , Mice , Multifunctional Enzymes , Pedigree , Phenotype , RNA Helicases/genetics , Sibling Relations , Young Adult
17.
Cerebellum ; 11(1): 194-211, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21717229

ABSTRACT

Our aim was to perform a comprehensive analysis of the global and segmental features of gait in patients with genetically confirmed inherited ataxias. Sixteen patients with autosomal dominant (spinocerebellar ataxia, SCA1 or 2) or recessive (Friedreich's ataxia, FRDA) ataxia were studied. We used a motion analysis system to record gait kinematic and kinetic data. We measured the mean values of global (time-distance parameters, COM displacement, support moment) and segmental gait parameters (joint displacement and inter-joint coordination), as both discrete and continuous variables, and their variability and correlations with International Cooperative Ataxia Rating Scale (ICARS) scores. We found a marked difference in all global gait parameters between the ataxic patients and the controls and close correlations between longer stride and stance duration and lower gait, posture and total ICARS scores. The only difference between the two patient groups was a shorter step length in the FRDA patients. As regards the segmental features, we found a significantly different waveform shape for all continuous kinematic and kinetic measures between the ataxic patients and the healthy controls, but only minor differences for the discrete measures. Intersegmental coordination evaluated using the continuous relative phase method revealed an irregular alternating joint behaviour without clear evidence of the synchronous pattern of alternating proximal/distal joint seen in healthy subjects. For almost all gait parameters we observed a markedly higher intra-subject variability in the ataxic patients versus the controls, which was strongly related to the clinical ICARS scores. Patients with chronic, progressive inherited ataxias lose the ability to "stabilize" a walking pattern that can be repeated over time. The most peculiar aspect of the gait of inherited ataxia patients, regardless the different genetic forms, seems to be the presence of increased variability of all global and segmental parameters rather than an invariant abnormal gait pattern.


Subject(s)
Cerebellar Ataxia/diagnosis , Friedreich Ataxia/diagnosis , Gait Disorders, Neurologic/diagnosis , Gait/genetics , Adolescent , Adult , Aged , Cerebellar Ataxia/congenital , Cerebellar Ataxia/physiopathology , Chronic Disease , Female , Friedreich Ataxia/genetics , Friedreich Ataxia/physiopathology , Gait Disorders, Neurologic/congenital , Gait Disorders, Neurologic/physiopathology , Humans , Male , Middle Aged , Young Adult
18.
Biofactors ; 37(5): 361-5, 2011.
Article in English | MEDLINE | ID: mdl-21990098

ABSTRACT

CoQ(10) deficiencies are clinically and genetically heterogeneous. This syndrome has been associated with five major clinical phenotypes: (1) encephalomyopathy, (2) severe infantile multisystemic disease, (3) cerebellar ataxia, (4) isolated myopathy, and (5) nephrotic syndrome. In a few patients, pathogenic mutations have been identified in genes involved in the biosynthesis of CoQ(10) (primary CoQ(10) deficiencies) or in genes not directly related to CoQ(10) biosynthesis (secondary CoQ(10) deficiencies). Respiratory chain defects, ROS production, and apoptosis variably contribute to the pathogenesis of primary CoQ(10) deficiencies.


Subject(s)
Ubiquinone/analogs & derivatives , Apraxias/genetics , Apraxias/metabolism , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Cells, Cultured , Cerebellar Ataxia/congenital , DNA-Binding Proteins/genetics , Electron-Transferring Flavoproteins/genetics , Fibroblasts/metabolism , Humans , Hypoalbuminemia/genetics , Hypoalbuminemia/metabolism , Iron-Sulfur Proteins/genetics , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Myopathies/genetics , Mitochondrial Myopathies/metabolism , Mutation , Nuclear Proteins/genetics , Oxidoreductases Acting on CH-NH Group Donors/genetics , Ubiquinone/deficiency , Ubiquinone/metabolism
19.
Nat Struct Mol Biol ; 18(11): 1189-95, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21984210

ABSTRACT

DNA ligases finalize DNA replication and repair through DNA nick-sealing reactions that can abort to generate cytotoxic 5'-adenylation DNA damage. Aprataxin (Aptx) catalyzes direct reversal of 5'-adenylate adducts to protect genome integrity. Here the structure of a Schizosaccharomyces pombe Aptx-DNA-AMP-Zn(2+) complex reveals active site and DNA interaction clefts formed by fusing a histidine triad (HIT) nucleotide hydrolase with a DNA minor groove-binding C(2)HE zinc finger (Znf). An Aptx helical 'wedge' interrogates the base stack for sensing DNA ends or DNA nicks. The HIT-Znf, the wedge and an '[F/Y]PK' pivot motif cooperate to distort terminal DNA base-pairing and direct 5'-adenylate into the active site pocket. Structural and mutational data support a wedge-pivot-cut HIT-Znf catalytic mechanism for 5'-adenylate adduct recognition and removal and suggest that mutations affecting protein folding, the active site pocket and the pivot motif underlie Aptx dysfunction in the neurodegenerative disorder ataxia with oculomotor apraxia 1 (AOA1).


Subject(s)
Apraxias/genetics , Apraxias/physiopathology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/physiopathology , DNA-Binding Proteins/chemistry , DNA/chemistry , Hypoalbuminemia/genetics , Hypoalbuminemia/physiopathology , Nuclear Proteins/chemistry , Amino Acid Motifs , Binding Sites , Cerebellar Ataxia/congenital , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , DNA Breaks, Single-Stranded , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleic Acid Conformation , Protein Structure, Tertiary , Zinc Fingers
20.
J Vet Intern Med ; 25(3): 490-6, 2011.
Article in English | MEDLINE | ID: mdl-21488963

ABSTRACT

BACKGROUND: There is a high incidence of hereditary degenerative diseases of the central nervous system in purebred dogs. HYPOTHESIS: Cerebellar ataxia in Malinois puppies, caused by degenerative changes that predominate in cerebellar nuclei and the granular cell layer, is a hereditary disorder that is distinct from cerebellar cortical abiotrophies. ANIMALS: Thirteen Malinois puppies with cerebellar ataxia. METHODS: Retrospective study. Records of Malinois puppies with spongy degeneration of the cerebellar nuclei were analyzed including clinical signs, histopathological changes, and pedigree data. RESULTS: Signs of cerebellar dysfunction were observed in puppies of both sexes from 5 different litters (1995-2009) of phenotypically normal parents. Clinical signs started before the age of 2 months and resulted in euthanasia of all puppies by the age of 13 weeks. Histopathology disclosed marked bilateral spongy degeneration of the cerebellar nuclei and vacuoles in the granular cell layer and foliate white matter of the cerebellum. In some puppies, discrete vacuoles in gray and white matter were present in other parts of the brain. Furthermore, spheroids and dilated myelin sheaths were observed. Pedigree data and segregation frequency support an autosomal recessive hereditary disorder. CONCLUSIONS AND CLINICAL IMPORTANCE: Malinois suffer from a hereditary spongiform degeneration that predominates in the cerebellum and causes an early onset of clinical signs with unfavorable prognosis. Future efforts should increase awareness among veterinarians and breeders and aim to identify underlying metabolic mechanisms and the affected genes.


Subject(s)
Cerebellar Ataxia/veterinary , Dog Diseases/congenital , Spinocerebellar Degenerations/veterinary , Animals , Cerebellar Ataxia/congenital , Cerebellar Ataxia/genetics , Dog Diseases/genetics , Dogs , Female , Male , Pedigree , Spinocerebellar Degenerations/congenital , Spinocerebellar Degenerations/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...