Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 417
Filtrar
1.
Mol Genet Genomic Med ; 12(6): e2466, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38860480

RESUMEN

BACKGROUND: Spinocerebellar ataxia 29 (SCA29) is a rare genetic disorder characterized by early-onset ataxia, gross motor delay, and infantile hypotonia, and is primarily associated with variants in the ITPR1 gene. Cases of SCA29 in Asia are rarely reported, limiting our understanding of this disease. METHODS: A female Korean infant, demonstrating clinical features of SCA29, underwent evaluation and rehabilitation at our outpatient clinic from the age of 3 months to the current age of 4 years. Trio-based genome sequencing tests were performed on the patient and her biological parents. RESULTS: The infant initially presented with macrocephaly, hypotonia, and nystagmus, with nonspecific findings on initial neuroimaging. Subsequent follow-up revealed gross motor delay, early onset ataxia, strabismus, and cognitive impairment. Further neuroimaging revealed atrophy of the cerebellum and vermis, and genetic analysis revealed a de novo pathogenic heterozygous c.800C>T, p.Thr267Met missense mutation in the ITPR1 gene (NM_001378452.1). CONCLUSION: This is the first reported case of SCA29 in a Korean patient, expanding the genetic and phenotypic spectrum of ITPR1-related ataxias. Our case highlights the importance of recognizing early-onset ataxic symptoms, central hypotonia, and gross motor delays with poor ocular fixation, cognitive deficits, and isolated cerebellar atrophy as crucial clinical indicators of SCA29.


Asunto(s)
Receptores de Inositol 1,4,5-Trifosfato , Mutación Missense , Degeneraciones Espinocerebelosas , Humanos , Femenino , Receptores de Inositol 1,4,5-Trifosfato/genética , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Preescolar , Ataxia Cerebelosa/genética , Ataxia Cerebelosa/patología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/patología , Lactante
2.
Mult Scler ; 29(9): 1195-1198, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37401111

RESUMEN

Cerebellar dysfunction is likely to cause severe and treatment-resistant disability in multiple sclerosis (MS). Certain spinocerebellar ataxia (SCA)-related alleles can increase MS susceptibility, and channel polymorphisms can impact disability measures. Following an index patient with the coexistence of MS and SCA Type-8 (SCA8) in the MS clinic, an institutional engine search for MS and hereditary ataxia coexistence was conducted but did not reveal any other cases. This extremely rare coexistence of MS and SCA8 in our index patient may be incidental; however, a yet-to-be-identified contribution of coexistent hereditary ataxia(s) to the susceptibility of a prominent progressive ataxia MS phenotype cannot be ruled out.


Asunto(s)
Esclerosis Múltiple , Degeneraciones Espinocerebelosas , Esclerosis Múltiple/complicaciones , Esclerosis Múltiple/diagnóstico , Esclerosis Múltiple/genética , Esclerosis Múltiple/patología , Degeneraciones Espinocerebelosas/complicaciones , Degeneraciones Espinocerebelosas/diagnóstico , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Humanos , Masculino , Adulto , Ataxia/genética , Ataxia/patología , Encéfalo/patología , Médula Espinal
3.
Rinsho Shinkeigaku ; 63(4): 201-208, 2023 Apr 25.
Artículo en Japonés | MEDLINE | ID: mdl-36990780

RESUMEN

Ataxia is not only due to cerebellar lesions, but also due to non-cerebellar lesions such as those in the brain, spinal cord, dorsal root (DR), peripheral nerve. In this article, optic ataxia is excluded and 'vestibular ataxia' is briefly referred. Non-cerebellar ataxias are generically called sensory ataxia or posterior column ataxia. However, since non-cerebellar lesions, e.g. frontal lobe lesions, may develop "cerebellar-like ataxia" (Hirayama, 2010). At the same time, non-posterior column lesions, e.g. parietal lobe lesion, can show "posterior column-like ataxia". From these viewpoints, I here describe various non-cerebellar ataxia in some disorders such as tabes dorsalis and sensory neuropathies and emphasize a role of a peripheral sensory input to the cerebellum via the DR ganglia and spinocerebellar tract for sensory ataxia because there is the International Consensus (2016) that the ataxia in Miller Fisher syndrome is suggested cerebellar-like clinicophysiologically.


Asunto(s)
Ataxia Cerebelosa , Degeneraciones Espinocerebelosas , Humanos , Ataxia Cerebelosa/etiología , Degeneraciones Espinocerebelosas/patología , Ataxia/etiología , Cerebelo/patología , Raíces Nerviosas Espinales/patología
4.
Cerebellum ; 22(4): 531-533, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35881322

RESUMEN

Prior to 1982, ovarian and certain other cancers were known to have a rare complication of progressive cerebellar ataxia, a disorder characterized pathologically by severe-often total-obliteration of cerebellar Purkinje cells. However, the cause of cerebellar injury in these patients was unknown. In that year, we began studies in which sera from individuals with this disorder were reacted with frozen sections of human cerebellum. These studies revealed that patients with ovarian cancer and cerebellar degeneration had high titers of antibodies directed against cytoplasmic antigens of Purkinje cells and deep cerebellar nuclei-a previously undescribed pattern of antibody response which was subsequently found not to be present in ovarian cancer patients who remained neurologically normal. This antibody, now known as "anti-Yo" or "anti-PCA1" provides a marker for rapidly progressive cerebellar ataxia and is heavily associated with gynecological and breast malignancies. Although the role of anti-Yo antibody in cerebellar injury has not been established in living animals, in vitro studies have demonstrated that anti-Yo antibody causes Purkinje cell death in the absence of T lymphocytes. In this commentary, we describe our studies leading to initial discovery of anti-Yo antibody, discuss the relationship of this discovery to current knowledge of paraneoplastic neurological disease, and outline some important questions which remain to be resolved before we fully understand the pathogenesis and optimal treatment of this disorder.


Asunto(s)
Ataxia Cerebelosa , Enfermedades Cerebelosas , Neoplasias Ováricas , Degeneración Cerebelosa Paraneoplásica , Degeneraciones Espinocerebelosas , Animales , Femenino , Humanos , Enfermedades Cerebelosas/patología , Cerebelo/patología , Autoanticuerpos , Células de Purkinje/metabolismo , Degeneraciones Espinocerebelosas/patología , Neoplasias Ováricas/complicaciones , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología
5.
Neuropathology ; 42(5): 379-393, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35859519

RESUMEN

Spinocerebellar degenerations (SCDs) are a diverse group of rare and slowly progressive neurological diseases that include spinocerebellar ataxia type 1 (SCA1), SCA2, SCA3, SCA6, SCA7, dentatorubral-pallidoluysian atrophy (DRPLA) and multiple system atrophy (MSA). They are often inherited, and affect the cerebellum and related pathways. The combination of clinical findings and lesion distribution has been the gold-standard for classifying SCDs. This conventional approach has not been very successful in providing a solid framework shared among researchers because their points of views have been quite variable. After identification of genetic abnormalities, classification was overwhelmed by genotyping, replacing the conventional approach far behind. In this review, we describe a stepwise operational approach that we constructed based only on macroscopic findings without microscopy to classify SCDs into three major groups: pure cerebellar type for SCA6 and SCA31; olivopontocerebellar (OPC) type for SCA1, SCA2, SCA7 and MSA; and dentatorubral-pallidoluysian (DRPL) type for SCA1, SCA3, DRPLA and progressive supranuclear palsy (PSP). Spinocerebellar tract involvement distinguishes SCA1 and SCA3 from DRPLA. Degeneration of the internal segment of the pallidum is accentuated in SCA3 and PSP, while degeneration of the external segment is accentuated in SCA1 and DRPLA. These contrasts are helpful in subdividing OPC and DRPL types to predict their genotypes. Lesion distribution represents disease-specific selective vulnerability, which is readily differentiated macroscopically using our stepwise operational approach. Precise prediction of the major genotypes will provide a basis to understand how genetic abnormalities lead to corresponding phenotypes through disease-specific selective vulnerabilities.


Asunto(s)
Ataxias Espinocerebelosas , Degeneraciones Espinocerebelosas , Humanos , Ataxias Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología
6.
Mol Neurobiol ; 59(6): 3800-3828, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35420383

RESUMEN

Cerebellar ataxia is a form of ataxia that originates from dysfunction of the cerebellum, but may involve additional neurological tissues. Its clinical symptoms are mainly characterized by the absence of voluntary muscle coordination and loss of control of movement with varying manifestations due to differences in severity, in the site of cerebellar damage and in the involvement of extracerebellar tissues. Cerebellar ataxia may be sporadic, acquired, and hereditary. Hereditary ataxia accounts for the majority of cases. Hereditary ataxia has been tentatively divided into several subtypes by scientists in the field, and nearly all of them remain incurable. This is mainly because the detailed mechanisms of these cerebellar disorders are incompletely understood. To precisely diagnose and treat these diseases, studies on their molecular mechanisms have been conducted extensively in the past. Accumulating evidence has demonstrated that some common pathogenic mechanisms exist within each subtype of inherited ataxia. However, no reports have indicated whether there is a common mechanism among the different subtypes of inherited cerebellar ataxia. In this review, we summarize the available references and databases on neurological disorders characterized by cerebellar ataxia and show that a subset of genes involved in lipid homeostasis form a new group that may cause ataxic disorders through a common mechanism. This common signaling pathway can provide a valuable reference for future diagnosis and treatment of ataxic disorders.


Asunto(s)
Ataxia Cerebelosa , Degeneraciones Espinocerebelosas , Ataxia/patología , Ataxia Cerebelosa/diagnóstico , Ataxia Cerebelosa/genética , Cerebelo/patología , Humanos , Lípidos , Degeneraciones Espinocerebelosas/patología
7.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830330

RESUMEN

Marinesco-Sjogren syndrome (MSS) is a rare multisystem pediatric disorder, caused by loss-of-function mutations in the gene encoding the endoplasmic reticulum cochaperone SIL1. SIL1 acts as a nucleotide exchange factor for BiP, which plays a central role in secretory protein folding. SIL1 mutant cells have reduced BiP-assisted protein folding, cannot fulfil their protein needs, and experience chronic activation of the unfolded protein response (UPR). Maladaptive UPR may explain the cerebellar and skeletal muscle degeneration responsible for the ataxia and muscle weakness typical of MSS. However, the cause of other more variable, clinical manifestations, such as mild to severe mental retardation, hypogonadism, short stature, and skeletal deformities, is less clear. To gain insights into the pathogenic mechanisms and/or adaptive responses to SIL1 loss, we carried out cell biological and proteomic investigations in skin fibroblasts derived from a young patient carrying the SIL1 R111X mutation. Despite fibroblasts not being overtly affected in MSS, we found morphological and biochemical changes indicative of UPR activation and altered cell metabolism. All the cell machineries involved in RNA splicing and translation were strongly downregulated, while protein degradation via lysosome-based structures was boosted, consistent with an attempt of the cell to reduce the workload of the endoplasmic reticulum and dispose of misfolded proteins. Cell metabolism was extensively affected as we observed a reduction in lipid synthesis, an increase in beta oxidation, and an enhancement of the tricarboxylic acid cycle, with upregulation of eight of its enzymes. Finally, the catabolic pathways of various amino acids, including valine, leucine, isoleucine, tryptophan, lysine, aspartate, and phenylalanine, were enhanced, while the biosynthetic pathways of arginine, serine, glycine, and cysteine were reduced. These results indicate that, in addition to UPR activation and increased protein degradation, MSS fibroblasts have profound metabolic alterations, which may help them cope with the absence of SIL1.


Asunto(s)
Fibroblastos/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Mutación con Pérdida de Función , Empalme del ARN , Degeneraciones Espinocerebelosas/genética , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Aminoácidos/metabolismo , Niño , Ciclo del Ácido Cítrico/genética , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Factor 2 Eucariótico de Iniciación/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Fibroblastos/patología , Expresión Génica , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Factores de Intercambio de Guanina Nucleótido/deficiencia , Humanos , Metabolismo de los Lípidos/genética , Anotación de Secuencia Molecular , Cultivo Primario de Células , Proteolisis , Degeneraciones Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/patología , Proteína 1 de Unión a la X-Box/genética , Proteína 1 de Unión a la X-Box/metabolismo
8.
Brain ; 144(8): 2427-2442, 2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-33792664

RESUMEN

Marinesco-Sjögren syndrome is a rare human disorder caused by biallelic mutations in SIL1 characterized by cataracts in infancy, myopathy and ataxia, symptoms which are also associated with a novel disorder caused by mutations in INPP5K. While these phenotypic similarities may suggest commonalties at a molecular level, an overlapping pathomechanism has not been established yet. In this study, we present six new INPP5K patients and expand the current mutational and phenotypical spectrum of the disease showing the clinical overlap between Marinesco-Sjögren syndrome and the INPP5K phenotype. We applied unbiased proteomic profiling on cells derived from Marinesco-Sjögren syndrome and INPP5K patients and identified alterations in d-3-PHGDH as a common molecular feature. d-3-PHGDH modulates the production of l-serine and mutations in this enzyme were previously associated with a neurological phenotype, which clinically overlaps with Marinesco-Sjögren syndrome and INPP5K disease. As l-serine administration represents a promising therapeutic strategy for d-3-PHGDH patients, we tested the effect of l-serine in generated sil1, phgdh and inpp5k a+b zebrafish models, which showed an improvement in their neuronal phenotype. Thus, our study defines a core phenotypical feature underpinning a key common molecular mechanism in three rare diseases and reveals a common and novel therapeutic target for these patients.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Inositol Polifosfato 5-Fosfatasas/genética , Mutación , Fenotipo , Fosfoglicerato-Deshidrogenasa/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Adulto , Animales , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Músculo Esquelético/patología , Proteómica , Degeneraciones Espinocerebelosas/patología , Pez Cebra
9.
Brain Dev ; 43(3): 464-469, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33272776

RESUMEN

Spinocerebellar ataxia, autosomal recessive 2 (SCAR2) [MIM:213200] is a rare autosomal recessive disease of spinocerebellar ataxia associated with degeneration of the cerebellum with variable involvement of the brainstem and spinal cord. SCAR2 is characterized by onset of impaired motor development and ataxic gait in early childhood. Recently, several PMPCA gene variants have been reported in SCAR2 patients with mild and non-progressive symptoms. PMPCA codes frataxin, which is crucial for iron biosynthesis in cells. We report a case of a 15-year-old Japanese girl with infancy-onset, very severe and progressive developmental delay, cerebellar ataxia, and extrapyramidal symptoms. Brain magnetic resonance imaging showed cerebellar atrophy and excessive brain iron accumulation in the bilateral globus pallidi and substantia nigra. Based on the clinical phenotypes and imaging, neurodegeneration with brain iron accumulation was suspected. Whole-exome sequencing on the proband and her parents revealed novel compound heterozygous variants at c.667C > T (p.Arg223Cys) and c.853del (p.Asp285llefs*16) in PMPCA. Thus, her disease was diagnosed as SCAR2. Phenotype in our case was different from ones previously reported for SCARs in the points of much severer clinical presentations with extrapyramidal signs and imaging suspected iron accumulation, and might overlap neurodegeneration with brain iron accumulation or NBIA subtypes. Our case might provide a new insight into PMPCA gene-related disorders and expand the disease concept.


Asunto(s)
Proteínas de Unión a Hierro/genética , Degeneraciones Espinocerebelosas/genética , Adolescente , Encéfalo/patología , Femenino , Genotipo , Humanos , Mutación , Linaje , Fenotipo , Degeneraciones Espinocerebelosas/patología , Frataxina
10.
Brain Pathol ; 31(2): 239-252, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33043513

RESUMEN

Spinocerebellar ataxia type 23 (SCA23) is a late-onset neurodegenerative disorder characterized by slowly progressive gait and limb ataxia, for which there is no therapy available. It is caused by pathogenic variants in PDYN, which encodes prodynorphin (PDYN). PDYN is processed into the opioid peptides α-neoendorphin and dynorphins (Dyn) A and B; inhibitory neurotransmitters that function in pain signaling, stress-induced responses and addiction. Variants causing SCA23 mostly affect Dyn A, leading to loss of secondary structure and increased peptide stability. PDYNR212W mice express human PDYN containing the SCA23 variant p.R212W. These mice show progressive motor deficits from 3 months of age, climbing fiber (CF) deficits from 3 months of age, and Purkinje cell (PC) loss from 12 months of age. A mouse model for SCA1 showed similar CF deficits, and a recent study found additional developmental abnormalities, namely increased GABAergic interneuron connectivity and non-cell autonomous disruption of PC function. As SCA23 mice show a similar pathology to SCA1 mice in adulthood, we hypothesized that SCA23 may also follow SCA1 pathology during development. Examining PDYNR212W cerebella during development, we uncovered developmental deficits from 2 weeks of age, namely a reduced number of GABAergic synapses on PC soma, possibly leading to the observed delay in early phase CF elimination between 2 and 3 weeks of age. Furthermore, CFs did not reach terminal height, leaving proximal PC dendrites open to be occupied by parallel fibers (PFs). The observed increase in vGlut1 protein-a marker for PF-PC synapses-indicates that PFs indeed take over CF territory and have increased connectivity with PCs. Additionally, we detected altered expression of several critical Ca2+ channel subunits, potentially contributing to altered Ca2+ transients in PDYNR212W cerebella. These findings indicate that developmental abnormalities contribute to the SCA23 pathology and uncover a developmental role for PDYN in the cerebellum.


Asunto(s)
Cerebelo/patología , Encefalinas/genética , Neurogénesis/genética , Precursores de Proteínas/genética , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Animales , Cerebelo/crecimiento & desarrollo , Humanos , Ratones , Ratones Transgénicos
11.
Brain Nerve ; 72(9): 923-930, 2020 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-32934181

RESUMEN

Marie et al. (1922) first proposed a disease entity "late cortical cerebellar atrophy (LCCA)", which is characterized neuropathologically by pure cerebello-olivary degeneration. LCCA was originally described as sporadic, late-onset, pure cerebellar ataxia of unknown etiology; however, it has occasionally been used to denote familial or secondary ataxias, particularly alcoholic cerebellar degeneration. Sporadic ataxia is classified mainly into LCCA or CCA and olivo-ponto-cerebellar atrophy (OPCA) in Japan. OPCA, now multiple system atrophy with predominant cerebellar ataxia, has characteristic brain imaging features and is clearly diagnosed based on the consensus criteria. On the other hand, there is no specific biomarker for LCCA/CCA, and neuropathological examination is required for a definitive diagnosis. Therefore, the clinical diagnosis of LCCA/CCA depends on the exclusion of other diseases manifesting as cerebellar ataxia. However the differential diagnosis for LCCA/CCA is not necessarily made carefully. As a result, the LCCA/CCA category in Japan is a "waste basket," including OPCA, hereditary ataxias, and secondary ataxias, which are unidentified yet. To refine the LCCA/CCA category, we proposed the clinically-defined term "idiopathic cerebellar ataxia (IDCA)" and established its diagnostic criteria. By nationwide screening, we have identified 51 patients with probable IDCA according to the criteria so far. Here we review the clinical characteristics of IDCA patients.


Asunto(s)
Ataxia Cerebelosa , Degeneraciones Espinocerebelosas , Atrofia/patología , Ataxia Cerebelosa/diagnóstico , Cerebelo , Humanos , Japón , Degeneraciones Espinocerebelosas/diagnóstico , Degeneraciones Espinocerebelosas/patología
12.
Brain Nerve ; 72(9): 947-959, 2020 Sep.
Artículo en Japonés | MEDLINE | ID: mdl-32934184

RESUMEN

Cortical cerebellar atrophy (CCA) contains hereditary spinocerebellar degeneration (hSCD) and genetic testing is necessary for an accurate diagnosis. Screening for frequent hSCDs (triplet repeat disease and SCA31) was performed. Panel analysis and whole exome analysis using a next-generation sequencer were also performed. The Japan Consortium for Ataxias, J-CAT, contributes to the elucidation of the genetic epidemiology of CCA. The elucidation of CCA would be promoted by comprehensive gene analysis, including whole genome analysis.


Asunto(s)
Ataxias Espinocerebelosas , Degeneraciones Espinocerebelosas , Atrofia/patología , Cerebelo , Humanos , Japón , Degeneraciones Espinocerebelosas/patología
13.
Ann Clin Transl Neurol ; 7(8): 1436-1442, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32767480

RESUMEN

FBXO7 is implicated in the ubiquitin-proteasome system and parkin-mediated mitophagy. FBXO7defects cause a levodopa-responsive parkinsonian-pyramidal syndrome(PPS). METHODS: We investigated the disease molecular bases in a child with PPS and brain iron accumulation. RESULTS: A novel homozygous c.368C>G (p.S123*) FBXO7 mutation was identified in a child with spastic paraplegia, epilepsy, cerebellar degeneration, levodopa nonresponsive parkinsonism, and brain iron deposition. Patient's fibroblasts assays demonstrated an absence of FBXO7 RNA expression leading to impaired proteasome degradation and accumulation of poly-ubiquitinated proteins. CONCLUSION: This novel FBXO7 phenotype associated with impaired proteasome activity overlaps with neurodegeneration with brain iron accumulation disorders.


Asunto(s)
Proteínas F-Box/genética , Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Trastornos Parkinsonianos , Complejo de la Endopetidasa Proteasomal/metabolismo , Adulto , Consanguinidad , Epilepsia/enzimología , Epilepsia/genética , Epilepsia/patología , Epilepsia/fisiopatología , Femenino , Humanos , Trastornos del Metabolismo del Hierro/enzimología , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Trastornos del Metabolismo del Hierro/fisiopatología , Distrofias Neuroaxonales/enzimología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Distrofias Neuroaxonales/fisiopatología , Paraplejía/enzimología , Paraplejía/genética , Paraplejía/patología , Paraplejía/fisiopatología , Trastornos Parkinsonianos/enzimología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Degeneraciones Espinocerebelosas/enzimología , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Degeneraciones Espinocerebelosas/fisiopatología , Síndrome , Adulto Joven
14.
J Hum Genet ; 65(12): 1143-1147, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32694621

RESUMEN

Recently, the expansion of an intronic AAGGG repeat in the replication factor C subunit 1 (RFC1) gene was reported to cause cerebellar ataxia, neuropathy, vestibular areflexia syndrome (CANVAS). In Europeans, the expansion accounted for 22% of sporadic patients with late-onset ataxia. We genotyped 37 Japanese patients comprising 25 familial (autosomal recessive or undecided transmission) and 12 sporadic ones with late-onset ataxia. We found intronic repeat expansions in RFC1 in three (12%) of the familial patients and one (8.5%) of the sporadic ones. Although our cohort study was small, the disease frequency in Japanese patients with CANVAS might be lower than that in European ones. In addition, we found biallelic ACAGG repeat expansion in one patient, indicating ACAGG repeat expansion might cause CANVAS. Clinically, we found one patient with sleep apnea syndrome, which has not been reported previously. Thus, this study might expand the clinical and genetic spectrum of CANVAS.


Asunto(s)
Expansión de las Repeticiones de ADN/genética , Predisposición Genética a la Enfermedad , Proteína de Replicación C/genética , Degeneraciones Espinocerebelosas/genética , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Intrones/genética , Japón/epidemiología , Masculino , Persona de Mediana Edad , Degeneraciones Espinocerebelosas/epidemiología , Degeneraciones Espinocerebelosas/patología
15.
Eur J Med Genet ; 63(9): 103998, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32622959

RESUMEN

Autosomal-recessive spinocerebellar ataxia type 18 (SCAR18) is a rare neurologic disorder. It is caused by bi-allelic aberrations in the GRID2 gene, encoding an ionotropic glutamate receptor. In total, 20 affected individuals with mainly homozygous/compound heterozygous intragenic deletions/duplications, two different missense variants and one nonsense variant in GRID2 have been reported, so far. SCAR18 is characterized by delayed psychomotor development, intellectual disability, severely impaired gait due to cerebellar ataxia, ocular movement abnormalities, and cerebellar atrophy in brain imaging. By trio exome sequencing, we now identified a novel homozygous nonsense variant (c.568C > T; p.Gln190*) in GRID2 in a four year old female from a consanguineous family who presented with a particularly severe manifestation of SCAR18. The girl was born after an uneventful pregnancy and showed early-onset, profoundly delayed psychomotor development with no achieved psychomotor milestones at age 4 years. Additionally, she presented with severe muscular hypotonia, progressive truncal and appendicular ataxia, binocular vertical nystagmus, central hearing loss and incomplete loss of sight. She was dystrophic, interacted only very little and had behavioral anomalies such as eating hair and bruxism. Brain imaging showed cerebellar hypoplasia, extended cerebrospinal fluid spaces and beginning reduction of cerebral volume. Our findings further delineate the mutational and clinical spectrum of GRID2-associated spinocerebellar ataxia type 18 and indicate that homozygous nonsense variants are possibly associated with the severe end of the SCAR18 phenotypic spectrum.


Asunto(s)
Codón sin Sentido , Receptores de Glutamato/genética , Degeneraciones Espinocerebelosas/genética , Preescolar , Femenino , Humanos , Degeneraciones Espinocerebelosas/patología
16.
Int J Biol Macromol ; 161: 692-703, 2020 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-32535204

RESUMEN

Mutation in TMEM240 is suggested to cause SCA21, but the specific mechanism has not been clarified. The subcellular localization, specific biological function, and corresponding mechanism of action of TMEM240 have also not been delineated. In this study, the mRNA and protein expression of TMEM240 were assessed using qPCR and western blotting, respectively. Live cell imaging was used to establish the sub-cellular location of TMEM240, and electron microscopy was used to determine the morphology and distribution of TMEM240 in the cell. TMEM240 was specifically expressed in the neurons. Exogenous TMEM240 formed a multilayered cell structure, which we refer to as TMEM240-Body (T240-Body). T240-Body was separated and purified by centrifugation and filtration. An anchor protein His-tagged-GFP-BP on Ni-NTA agarose was used to pull down T240-GFP binding proteins. Both the N-terminal and the C-terminal of TMEM240 were confirmed to be inside the T240-Body. Co-localization experiments suggested that peroxisomes might contribute to T240-Body formation, and the two transmembrane regions of TMEM240 appear to be essential for formation of the T240-Body. Emerin protein contributed to formation of T240-Body when combined with TMEM240. Overall, this study provides new insights into TMEM240, which inform future research to further our understanding of its biological function.


Asunto(s)
Encéfalo , Proteínas de la Membrana/metabolismo , Mutación , Neuronas , Peroxisomas , Degeneraciones Espinocerebelosas , Animales , Encéfalo/metabolismo , Encéfalo/ultraestructura , Células Hep G2 , Humanos , Proteínas de la Membrana/genética , Ratones , Neuronas/metabolismo , Neuronas/ultraestructura , Peroxisomas/genética , Peroxisomas/metabolismo , Peroxisomas/ultraestructura , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/metabolismo , Degeneraciones Espinocerebelosas/patología
17.
J Clin Neurosci ; 77: 232-234, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32387255

RESUMEN

Autosomal recessive spinocerebellar ataxia type 18 (SCAR18) is caused by pathogenic variants in the Glutamate Receptor, Ionotropic, Delta-2 (GRID2) gene. We describe the long-term follow-up from 1 to 31 years of an Italian patient with congenital SCAR18 who is compound heterozygous for a maternally-inherited nonsense variant and a de novo microdeletion. To date, this is the longest follow-up in congenital SCAR18.


Asunto(s)
Degeneraciones Espinocerebelosas/patología , Adolescente , Adulto , Niño , Preescolar , Estudios de Seguimiento , Humanos , Lactante , Estudios Longitudinales , Masculino , Mutación , Receptores de Glutamato/genética , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/fisiopatología , Adulto Joven
18.
J Integr Neurosci ; 19(1): 125-129, 2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32259893

RESUMEN

Autosomal recessive cerebellar ataxias comprise many types of diseases. The most frequent autosomal recessive cerebellar ataxias are Friedreich ataxia, but other types are relatively rare. We encountered a consanguineous family with two cases of late-onset cerebellar ataxia with neuropathy. We performed whole-exome sequencing in one patient and confirmed by Sanger sequencing in other family members. Neurological examination revealed cerebellar ataxia, hand tremor, and neck dystonia, distal muscle wasting, and diminished tendon reflexes. The patients had no conjunctival telangiectasia or immunodeficiency. Blood examination revealed slightly elevated α-fetoprotein. Brain MRI demonstrated marked cerebellar atrophy and mild brainstem atrophy. The electrophysiologic study and nerve biopsy showed axonal neuropathy. Whole-exome sequencing revealed a novel homozygous missense variant (NM_000051.3: c.496G > C) in the ataxia-telangiectasia mutated gene. This homozygous variant was found in another patient, co-segregated within the family members-this variant results in aberrant splicing (skipping exon 5) on RT-PCR analysis. We identified the ataxia-telangiectasia mutated variant in an adult, late-onset autosomal recessive cerebellar ataxias family. We should consider ataxia-telangiectasia even in late-onset autosomal recessive cerebellar ataxias without telangiectasia or immunodeficiency.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Adulto , Axones/patología , Encéfalo/patología , Femenino , Humanos , Masculino , Mutación , Linaje , Degeneraciones Espinocerebelosas/fisiopatología , Secuenciación del Exoma
19.
Cell Mol Life Sci ; 77(20): 4015-4029, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32306062

RESUMEN

The family of hereditary cerebellar ataxias is a large group of disorders with heterogenous clinical manifestations and genetic etiologies. Among these, over 30 autosomal dominantly inherited subtypes have been identified, collectively referred to as the spinocerebellar ataxias (SCAs). Generally, the SCAs are characterized by a progressive gait impairment with classical cerebellar features, and in a subset of SCAs, accompanied by extra-cerebellar features. Beyond the common gait impairment and cerebellar atrophy, the wide range of additional clinical features observed across the SCAs is likely explained by the diverse set of mutated genes that encode proteins with seemingly disparate functional roles in nervous system biology. By synthesizing knowledge obtained from studies of the various SCAs over the past several decades, convergence onto a few key cellular changes, namely ion channel dysfunction and transcriptional dysregulation, has become apparent and may represent central mechanisms of cerebellar disease pathogenesis. This review will detail our current understanding of the molecular pathogenesis of the SCAs, focusing primarily on the first described autosomal dominant spinocerebellar ataxia, SCA1, as well as the emerging common core mechanisms across the various SCAs.


Asunto(s)
Ataxias Espinocerebelosas/patología , Animales , Cerebelo/patología , Humanos , Proteínas Nucleares/genética , Degeneraciones Espinocerebelosas/patología
20.
Proc Natl Acad Sci U S A ; 117(11): 6023-6034, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32132200

RESUMEN

Despite a growing number of ion channel genes implicated in hereditary ataxia, it remains unclear how ion channel mutations lead to loss-of-function or death of cerebellar neurons. Mutations in the gene KCNMA1, encoding the α-subunit of the BK channel have emerged as responsible for a variety of neurological phenotypes. We describe a mutation (BKG354S) in KCNMA1, in a child with congenital and progressive cerebellar ataxia with cognitive impairment. The mutation in the BK channel selectivity filter dramatically reduced single-channel conductance and ion selectivity. The BKG354S channel trafficked normally to plasma, nuclear, and mitochondrial membranes, but caused reduced neurite outgrowth, cell viability, and mitochondrial content. Small interfering RNA (siRNA) knockdown of endogenous BK channels had similar effects. The BK activator, NS1619, rescued BKG354S cells but not siRNA-treated cells, by selectively blocking the mutant channels. When expressed in cerebellum via adenoassociated virus (AAV) viral transfection in mice, the mutant BKG354S channel, but not the BKWT channel, caused progressive impairment of several gait parameters consistent with cerebellar dysfunction from 40- to 80-d-old mice. Finally, treatment of the patient with chlorzoxazone, a BK/SK channel activator, partially improved motor function, but ataxia continued to progress. These studies indicate that a loss-of-function BK channel mutation causes ataxia and acts by reducing mitochondrial and subsequently cellular viability.


Asunto(s)
Cerebelo/patología , Clorzoxazona/administración & dosificación , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Mitocondrias/patología , Degeneraciones Espinocerebelosas/genética , Adolescente , Animales , Animales Recién Nacidos , Línea Celular , Cerebelo/citología , Análisis Mutacional de ADN , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Técnicas de Silenciamiento del Gen , Vectores Genéticos/genética , Humanos , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/antagonistas & inhibidores , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Mutación con Pérdida de Función , Ratones , Oocitos , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Degeneraciones Espinocerebelosas/diagnóstico , Degeneraciones Espinocerebelosas/tratamiento farmacológico , Degeneraciones Espinocerebelosas/patología , Transfección , Secuenciación del Exoma , Xenopus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA