Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Neuroimage Clin ; 39: 103459, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37541097

RESUMO

PURPOSE: Progressive myoclonic epilepsy, type 1A (EPM1, Unverricht-Lundborg disease), is a rare neurodegenerative autosomal recessive disorder characterized by stimulus-sensitive and action myoclonus and tonic-clonic epileptic seizures. Patients develop neurological symptoms, including ataxia, intention tremor, and dysarthria, over time, with relatively limited and nonspecific MRI atrophy findings. The effects of the disease on brain metabolism are largely unknown. METHOD: Eighteen EPM1 patients (9 M, 9F) underwent clinical evaluation and neuropsychological testing, which included the assessment of intellectual ability, verbal memory, and psychomotor and executive functions. Magnetic resonance spectroscopy (MRS) and imaging (MRI) were performed on a 1.5 T MRI system. 2D MRS chemical shift imaging (CSI) maps (TE = 270) were obtained from the following regions of the brain: basal ganglia, thalamus, insula, splenium, and occipital white and gray matter, and N-acetyl-aspartate (NAA)-, choline (Cho)-, and lactate (Lac)-to-creatine (Cr) ratios were analyzed. Ten healthy age-and sex-matched subjects (5M, 5F) were used as controls for MRS. RESULTS: We found significant brain metabolic changes involving lactate, NAA, and choline, which are widespread in the basal ganglia, thalamic nuclei, insula, and occipital areas of EPM1 patients. Changes, especially in the right insula, basal ganglia, and thalamus, were associated with intellectual abilities and impairment of the psychomotor and executive functions of EPM1 patients. CONCLUSION: Multiple brain metabolic alterations suggest the presence of neurodegeneration associated with EPM1 progression. The changes in metabolite ratios are associated with the neurocognitive dysfunction caused by the disease. However, the role of MRS findings in understanding pathophysiology of EPM1 warrants further studies.


Assuntos
Epilepsias Mioclônicas Progressivas , Síndrome de Unverricht-Lundborg , Humanos , Síndrome de Unverricht-Lundborg/metabolismo , Espectroscopia de Prótons por Ressonância Magnética , Encéfalo , Epilepsias Mioclônicas Progressivas/metabolismo , Espectroscopia de Ressonância Magnética , Imageamento por Ressonância Magnética , Cognição , Metaboloma , Colina/metabolismo , Ácido Aspártico , Creatina/metabolismo
2.
Epilepsia ; 61(11): 2593-2608, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32940364

RESUMO

OBJECTIVE: Microglial phagocytosis of apoptotic cells is an essential component of the brain regenerative response during neurodegeneration. Whereas it is very efficient in physiological conditions, it is impaired in mouse and human mesial temporal lobe epilepsy, and now we extend our studies to a model of progressive myoclonus epilepsy type 1 in mice lacking cystatin B (CSTB). METHODS: We used confocal imaging and stereology-based quantification of apoptosis and phagocytosis of the hippocampus of Cstb knockout (KO) mice, an in vitro model of phagocytosis and siRNAs to acutely reduce Cstb expression, and a virtual three-dimensional (3D) model to analyze the physical relationship between apoptosis, phagocytosis, and active hippocampal neurons. RESULTS: Microglial phagocytosis was impaired in the hippocampus of Cstb KO mice at 1 month of age, when seizures arise and hippocampal atrophy begins. This impairment was not related to the lack of Cstb in microglia alone, as shown by in vitro experiments with microglial Cstb depletion. The phagocytosis impairment was also unrelated to seizures, as it was also present in Cstb KO mice at postnatal day 14, before seizures begin. Importantly, phagocytosis impairment was restricted to the granule cell layer and spared the subgranular zone, where there are no active neurons. Furthermore, apoptotic cells (both phagocytosed and not phagocytosed) in Cstb-deficient mice were at close proximity to active cFos+ neurons, and a virtual 3D model demonstrated that the physical relationship between apoptotic cells and cFos+ neurons was specific for Cstb KO mice. SIGNIFICANCE: These results suggest a complex crosstalk between apoptosis, phagocytosis, and neuronal activity, hinting that local neuronal activity could be related to phagocytosis dysfunction in Cstb KO mice. Overall, these data suggest that phagocytosis impairment is an early feature of hippocampal damage in epilepsy and opens novel therapeutic approaches for epileptic patients based on targeting microglial phagocytosis.


Assuntos
Giro Denteado/metabolismo , Modelos Animais de Doenças , Microglia/metabolismo , Neurônios/metabolismo , Fagocitose/fisiologia , Síndrome de Unverricht-Lundborg/metabolismo , Animais , Cistatina B/deficiência , Cistatina B/genética , Giro Denteado/patologia , Camundongos , Camundongos Knockout , Microglia/patologia , Neurônios/patologia , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patologia
3.
Radiology ; 269(1): 232-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23788720

RESUMO

PURPOSE: To study white matter (WM) changes in patients with Unverricht-Lundborg progressive myoclonus epilepsy (EPM1) caused by mutations in the cystatin B gene and in the cystatin B-deficient (Cstb-/-) mouse model and to validate imaging findings with histopathologic analysis of mice. MATERIALS AND METHODS: Informed consent was obtained and the study was approved by an institutional ethics committee. Animal work was approved by the Animal Experiment Board of Finland. Diffusion-tensor imaging and tract-based spatial statistics (TBSS) were used to compare fractional anisotropic (FA) results and axial, radial, and mean diffusion among patients with EPM1 (n = 19) and control subjects (n = 18). Ex vivo diffusion-tensor imaging and TBSS were used to compare Cstb-/- mice (n = 9) with wild controls (n = 4). Areas of FA decrease in mice were characterized by means of immunohistochemical analysis and transmission electron microscopy. Student t test statistics were applied to report significant findings (threshold-free cluster enhancement, P < .05). RESULTS: Patients with EPM1 showed significantly (P < .05) reduced FA and increased radial and mean diffusion in all major WM tracts compared with those of control subjects, shown as global FA decrease along the TBSS skeleton (0.41 ± 0.03 vs 0.45 ± 0.02, respectively; P < 5 × 10(-6)). Cstb-/- mice exhibited significantly reduced FA (P < .05) and antimyelin basic protein staining. Transmission electron microscopy revealed degenerating axons in Cstb-/- mice vs controls (979 axons counted, 51 degenerating axons; 2.09 ± 0.29 per field vs 1072 axons counted, nine degenerating axons; 0.48 ± 0.19 per field; P = .002). CONCLUSION: EPM1 is characterized by widespread alterations in subcortical WM, the thalamocortical system, and the cerebellum, which result in axonal degeneration and WM loss. These data suggest that motor disturbances and other symptoms in patients with EPM1 involve not only the cortical system but also the thalamocortical system and cerebellum.


Assuntos
Cistatina B/deficiência , Imageamento por Ressonância Magnética/métodos , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Síndrome de Unverricht-Lundborg/metabolismo , Síndrome de Unverricht-Lundborg/patologia , Adolescente , Adulto , Animais , Criança , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Pesquisa Translacional Biomédica , Adulto Jovem
4.
Biol Chem ; 394(6): 783-90, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23362198

RESUMO

Human stefins and cystatins are physiologically important cysteine proteinase inhibitors, acting as a first line of defense against undesirable proteolysis. Mutations in the cystatin B gene cause a rare form of epilepsy EPM1. Its two missense mutants, G50E and Q71P, lack the inhibitory activity and are partially unfolded, which leads to changes in their aggregation behavior, both in vitro and in the cell. SDS-PAGE and MALDI-TOF mass spectrometry were used to follow the hydrolysis of human stefin B wild type, G50E and Q71P, by cathepsins B and S in vitro. Cathepsin S was found to degrade both mutants, with Q71P being degraded faster. This correlates with the openness of the protein structure, Q71P having more exposed hydrophobic surfaces. Cathepsin B acted more selectively, degrading G50E into smaller fragments, while still leaving a portion of the full-length protein intact. Q71P was cleaved only at the exposed N-terminal end. The co-localization of stefin B wild type and EPM1 mutants with cathepsins showed that cathepsins accumulate around the aggregates formed by the EPM1 mutants. We hypothesize that the aggregation of both full-length mutants prevents the cathepsin molecule from accessing the substrate protein's core, whereas the cleaved fragments would be expected to aggregate stronger.


Assuntos
Catepsina B/metabolismo , Catepsinas/metabolismo , Cistatina B/química , Cistatina B/metabolismo , Proteínas Mutantes/metabolismo , Desdobramento de Proteína , Síndrome de Unverricht-Lundborg/metabolismo , Catepsinas/química , Eletroforese em Gel de Poliacrilamida , Imunofluorescência , Humanos , Proteínas Mutantes/química , Estabilidade Proteica , Estrutura Quaternária de Proteína , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
5.
Bone ; 51(6): 1016-24, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23010349

RESUMO

PURPOSE: Unverricht-Lundborg disease (EPM1) is a rare type of inherited progressive myoclonic epilepsy resulting from mutations in the cystatin B gene, CSTB, which encodes a cysteine cathepsin inhibitor. Cystatin B, cathepsin K, and altered osteoclast bone resorption activity are interconnected in vitro. This study evaluated the skeletal characteristics of patients with EPM1. METHODS: Sixty-six genetically verified EPM1 patients and 50 healthy controls underwent head MRI. Skull dimensions and regional calvarial thickness was measured perpendicular to each calvarial bone from T1-weighted 3-dimensional images using multiple planar reconstruction tools. All clinical X-ray files of EPM1 patients were collected and reviewed by an experienced radiologist. A total of 337 X-ray studies were analyzed, and non-traumatic structural anomalies, dysplasias and deformities were registered. RESULTS: EPM1 patients exhibited significant thickening in all measured cranial bones compared to healthy controls. The mean skull thickness was 10.0±2.0mm in EPM1 patients and 7.6±1.2mm in healthy controls (p<0.001). The difference was evident in all age groups and was not explained by former phenytoin use. Observed abnormalities in other skeletal structures in EPM1 patients included thoracic scoliosis (35% of EPM1 patients) and lumbar spine scoliosis (35%), large paranasal sinuses (27%), accessory ossicles of the foot, and arachnodactyly (18%). CONCLUSIONS: Skull thickening and an increased prevalence of abnormal findings in skeletal radiographs of patients with EPM1 suggest that this condition is connected to defective cystatin B function. These findings further emphasize the role of cystatin B in bone metabolism in humans.


Assuntos
Cistatina B/metabolismo , Escoliose/metabolismo , Escoliose/patologia , Crânio/metabolismo , Crânio/patologia , Síndrome de Unverricht-Lundborg/metabolismo , Síndrome de Unverricht-Lundborg/patologia , Adolescente , Adulto , Idoso , Criança , Feminino , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Adulto Jovem
6.
Neurobiol Dis ; 47(2): 216-24, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22538221

RESUMO

Unverricht-Lundborg disease (ULD) is the most common progressive myoclonic epilepsy. Its etiology has been identified in a defect of a protease inhibitor, cystatin B (CSTB), but the mechanism(s) by which this defect translates in the clinical manifestations of the disease are still obscure. We tested the hypothesis that ULD is accompanied by a loss of cortical GABA inhibition in a murine model (the CSTB knockout mouse) and in a human case. Cortical GABA signaling has been investigated measuring VGAT immunohistochemistry (a histological marker of the density of GABA terminals), GABA release from synaptosomes and paired-pulse stimulation. In CSTB knockout mice, a progressive decrease in neocortex thickness was found, associated with a prevalent loss of GABA interneurons. A marked reduction in VGAT labeling was found in the cortex of both CSTB knockout mice and an ULD patient. This implicates a reduction in GABA synaptic transmission, which was confirmed in the mouse model as reduction in GABA release from isolated nerve terminals and as loss of electrophysiologically measured GABA inhibition. The alterations in VGAT immunolabeling progressed in time, paralleling the worsening of myoclonus. These results provide direct evidence that loss of cortical GABA input occurs in a relevant animal model and in a case of human ULD, leading to a condition of latent hyperexcitability that favors myoclonus and seizures. These findings contribute to the understanding of the pathogenic mechanism of ULD and of the neurobiological basis of the effect of currently employed drugs.


Assuntos
Córtex Cerebral/patologia , Terminações Pré-Sinápticas/patologia , Síndrome de Unverricht-Lundborg/patologia , Ácido gama-Aminobutírico/deficiência , Adulto , Animais , Córtex Cerebral/metabolismo , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Pessoa de Meia-Idade , Terminações Pré-Sinápticas/metabolismo , Síndrome de Unverricht-Lundborg/metabolismo
7.
J Neuropathol Exp Neurol ; 71(1): 40-53, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22157618

RESUMO

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is a hereditary neurodegenerative disorder caused by mutations in the cystatin B (CSTB) gene encoding an inhibitor of cysteine proteases. Here, we provide the first detailed description of the onset and progression of pathologic changes in the CNS of Cstb-deficient (Cstb) mice. Our data reveal early and localized glial activation in brain regions where neuron loss subsequently occurs. These changes are most pronounced in the thalamocortical system, with neuron loss occurring first within the cortex and only subsequently in the corresponding thalamic relay nucleus. Microglial activation precedes the emergence of myoclonia and is followed by successive astrocytosis and selective neuron loss. Neuron loss was not detected in thalamic relay nuclei that displayed no glial activation. Microglia showed morphologic changes during disease progression from that of phagocytic brain macrophages in young animals to having thickened branched processes in older animals. These novel data on the timing of pathologic events in the CSTB-deficient brain highlight the potential role of glial activation at the initial stages of the disease. Determining the precise sequence of the neurodegenerative events in Cstb mouse brains will lay the basis for understanding the pathophysiology of EPM1.


Assuntos
Cistatina B/deficiência , Modelos Animais de Doenças , Microglia/metabolismo , Microglia/patologia , Neurônios/patologia , Síndrome de Unverricht-Lundborg/patologia , Animais , Encéfalo , Morte Celular/genética , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Cistatina B/genética , Masculino , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Fatores de Tempo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo
8.
Acta Neuropathol ; 121(3): 421-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20721566

RESUMO

Unverricht-Lundborg disease (EPM1A), also known as Baltic myoclonus, is the most common form of progressive myoclonic epilepsy. It is inherited as an autosomal recessive trait, due to mutations in the Cystatin-B gene promoter region. Although there is much work on rodent models of this disease, there is very little published neuropathology in patients with EPM1A. Here, we present the neuropathology of a patient with genetically confirmed EPM1A, who died at the age of 76. There was atrophy and gliosis affecting predominantly the cerebellum, frontotemporal cortex, hippocampus and thalamus. We have identified neuronal cytoplasmic inclusions containing the lysosomal proteins, Cathepsin-B and CD68. These inclusions also showed immunopositivity to both TDP-43 and FUS, in some cases associated with an absence of normal neuronal nuclear TDP-43 staining. There were also occasional ubiquitinylated neuronal intranuclear inclusions, some of which were FUS immunopositive. This finding is consistent with neurodegeneration in EPM1A as at least a partial consequence of lysosomal damage to neurons, which have reduced Cystatin-B-related neuroprotection. It also reveals a genetically defined neurodegenerative disease with both FUS and TDP-43 related pathology.


Assuntos
Corpos de Inclusão/patologia , Corpos de Inclusão Intranuclear/patologia , Neurônios/patologia , Síndrome de Unverricht-Lundborg/patologia , Idoso , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Atrofia/patologia , Cistatina B/genética , Cistatina B/metabolismo , Proteínas de Ligação a DNA/metabolismo , Evolução Fatal , Feminino , Humanos , Corpos de Inclusão/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , Mutação/genética , Neurônios/metabolismo , Proteína FUS de Ligação a RNA/metabolismo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo
9.
Epilepsia ; 47(10): 1650-4, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17054687

RESUMO

PURPOSE: Progressive myoclonus epilepsy of the Unverricht-Lundborg type (EPM1) is a rare neurologic disorder, associated with mutations in the Cystatin B (Cstb) gene. Mice lacking Cstb, a cysteine protease inhibitor of the cathepsine family of proteases, provide a mammalian model for EPM1 by displaying similarly progressive ataxia, myoclonic seizures, and neurodegeneration. However, the linkage of Cstb deficit on the molecular level to pathologic features like myoclonic jerks or tonic-clonic seizures has remained unclear. We examined the tryptophan (TRP) metabolism, along the serotonin (5HT) and kynurenine (KYN) pathway in the brain of Cstb-deficient mice, in relation to their possible involvement in the seizure phenotype. METHODS: TRP and its metabolites, along the 5HT and KYN pathways, were assayed in brain tissue by high-pressure liquid chromatography (HPLC) with electrochemical detection. The inverted wire grid and mild handling tests were used for evaluation of ataxia and myoclonic activity. RESULTS: The Cstb-deficient mice had constitutively increased TRP, 5HT, and 5-hydroxyindole acetic acid (5HIAA) levels in the cerebral cortex and cerebellum and increased levels of KYN in the cerebellum. These neurochemical changes were accompanied with ataxia and an apparent myoclonic phenotype among the Cstb-deficient mice. CONCLUSIONS: Our findings suggest that secondary processes (i.e., overstimulation of serotoninergic transmission) on the cellular level, initiated by Cstb deficiency in specific brain regions, may be responsible for the myoclonic/seizure phenotype in EPM1.


Assuntos
Encéfalo/metabolismo , Cistatinas/deficiência , Modelos Animais de Doenças , Mutação/genética , Triptofano/metabolismo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo , Animais , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Cromatografia Líquida de Alta Pressão , Cistatina B , Cistatinas/metabolismo , Feminino , Ácido Hidroxi-Indolacético/metabolismo , Cinurenina/metabolismo , Masculino , Camundongos , Fenótipo , Serotonina/metabolismo , Serotonina/fisiologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia , Triptofano/genética
11.
Med Hypotheses ; 64(5): 955-9, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15780491

RESUMO

Loss of function mutations in the gene (CSTB) encoding human cystatin B, a widely expressed cysteine protease inhibitor, are responsible for a severe neurological disorder known as an Unverricht-Lundborg disease (EPM1). EPM1 had been linked to chromosome 21q22.3 in Finnish families and it is an autosomal recessive inherited disorder with a homozygous minisatellite expansion in the cystatin B gene (stefin B gene). This disease is difficult to treat because it is refractory to most antiepileptic drugs and using multiple medications had been unsuccessful so far. To come a step closer to understanding of the nature of this disease, especially about the events on the molecular level, in vitro properties of missense EPM1 mutant G4R were determined. It was observed that the mutant has a prolonged lag phase of fibrillation at the same protein stability, which could indicate it were more toxic to the cells. Similar experiments with the N-terminal fragment of 67 aminoacid residues are ongoing, showing higher propensity to aggregate. Therefore, a hypothesis is launched that at least in a subset of Unverricht-Lundborg disease patients, cystatin B protein may aggregate in the cell. Protein aggregation can be secondary to external insults or aging, however, inherited forms of neurodegenerative diseases, such as familial Parkinson's, Huntington's or familial Alzheimer's disease, are directly linked to the mutant proteins aggregation. Protein aggregates in the form of amyloid plaques, neurofibrilary tangles, intra-cytoplasmic or intra-nuclear inclusions lead to increased production of the reactive oxygen species and dysfunction of the ubiquitine/proteasome system. Finally, mitochondrial dysfunction and cell death are observed. Certainly, it remains to be checked by experiments whether overexpression in cell culture of the missense mutants G4R and N-terminal fragment to residue 68 lead to cellular inclusions and the accompanying changes characteristic for the conformational disorders.


Assuntos
Cistatinas/metabolismo , Síndrome de Unverricht-Lundborg/metabolismo , Anticonvulsivantes/uso terapêutico , Cistatina B , Cistatinas/genética , Humanos , Mutação , Síndrome de Unverricht-Lundborg/tratamento farmacológico , Síndrome de Unverricht-Lundborg/patologia
12.
J Neurobiol ; 56(4): 315-27, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12918016

RESUMO

The inherited epilepsy Unverricht-Lundborg disease (EPM1) is caused by loss-of-function mutations in the cysteine protease inhibitor, cystatin B. Because cystatin B inhibits a class of lysosomal cysteine proteases called cathepsins, we hypothesized that increased proteolysis by one or more of these cathepsins is likely to be responsible for the seizure, ataxia, and neuronal apoptosis phenotypes characteristic of EPM1. To test this hypothesis and to identify which cysteine cathepsins contribute to EPM1, we have genetically removed three candidate cathepsins from cystatin B-deficient mice and tested for rescue of their EPM1 phenotypes. Whereas removal of cathepsins L or S from cystatin B-deficient mice did not ameliorate any aspect of the EPM1 phenotype, removal of cathepsin B resulted in a 36-89% reduction in the amount of cerebellar granule cell apoptosis depending on mouse age. The incidence of an incompletely penetrant eye phenotype was also reduced upon removal of cathepsin B. Because the apoptosis and eye phenotypes were not abolished completely and the ataxia and seizure phenotypes experienced by cystatin B-deficient animals were not diminished, this suggests that another molecule besides cathepsin B is also responsible for the pathogenesis, or that another molecule can partially compensate for cathepsin B function. These findings establish cathepsin B as a contributor to the apoptotic phenotype of cystatin B-deficient mice and humans with EPM1. They also suggest that the identification of cathepsin B substrates may further reveal the molecular basis for EPM1.


Assuntos
Catepsina B/fisiologia , Catepsinas , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo , Animais , Catepsina B/biossíntese , Catepsina B/deficiência , Catepsina B/genética , Catepsina L , Catepsinas/biossíntese , Catepsinas/deficiência , Catepsinas/genética , Catepsinas/fisiologia , Cisteína Endopeptidases , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Síndrome de Unverricht-Lundborg/patologia
13.
Ann Med ; 34(5): 380-5, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12452481

RESUMO

BACKGROUND: Loss-of-function mutations in the gene encoding cystatin B (CSTB) underlie an inherited neurodegenerative disorder, progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1). CSTB is an inhibitor of several papain-family cysteine proteases, the lysosomal cathepsins. Its physiological function and the molecular pathways leading to the clinical EPM1 phenotype are unknown. AIM: To elucidate the role of CSTB and different cathepsins in pathogenesis of EPM1. METHOD: We determined the total papain inhibitory (cystatin) and papain-like (cathepsin) activity as well as specific activities of cathepsins B, H, L and S in lymphoblastoid cells of EPM1 patients, carriers and controls. RESULTS: In EPM1 patients, who express reduced levels of CSTB mRNA, the papain inhibitory activity was significantly decreased or absent. This reduction was correlated with significant increase in general cathepsin activity. The increase in cathepsin B, L and S activities was highly significant, whereas the increase in cathepsin H activity was not. CONCLUSIONS: This is the first demonstration of cysteine protease activity being regulated by CSTB activity in a biological context. The effects of decreased CSTB activity in EPM1 pathogenesis may, at least in part, be mediated by cathepsins through increased activity of cathepsins S and L.


Assuntos
Catepsinas/fisiologia , Cistatinas/metabolismo , Inibidores de Cisteína Proteinase/metabolismo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo , Apoptose/fisiologia , Catepsina B/fisiologia , Catepsina H , Catepsina L , Cistatina B , Cisteína Endopeptidases , Humanos , Síndrome de Unverricht-Lundborg/fisiopatologia
14.
Neurology ; 58(11): 1686-9, 2002 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-12058102

RESUMO

MRI of the brain and proton MRS ((1)H MRS) of the pons and dentate were obtained in 10 patients with genetically confirmed Unverricht-Lundborg disease (EPM1) and 20 control subjects. Patients with EPM1 showed (p < or = 0.01) loss of bulk of the basis pontis, medulla, and cerebellar hemispheres. Cerebral atrophy was present in six patients. The N-acetylaspartate/creatine and choline/creatine ratios were reduced in the pons but not in the dentate (p < or = 0.005). Brainstem involvement could play a role in pathophysiology of EPM1.


Assuntos
Ácido Aspártico/análogos & derivados , Tronco Encefálico/patologia , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Síndrome de Unverricht-Lundborg/patologia , Adolescente , Adulto , Ácido Aspártico/metabolismo , Tronco Encefálico/metabolismo , Cerebelo/metabolismo , Cerebelo/patologia , Colina/metabolismo , Creatina/metabolismo , Feminino , Humanos , Masculino , Bulbo/metabolismo , Bulbo/patologia , Ponte/metabolismo , Ponte/patologia , Prótons , Síndrome de Unverricht-Lundborg/metabolismo
15.
Epilepsy Res ; 40(1): 33-9, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10771256

RESUMO

Progressive myoclonic epilepsies (EPM) are difficult to treat and refractory to most antiepileptic drugs. Besides epilepsy, EPMs also involve continuous neurological deterioration. Oxidative stress is thought to be an important factor in this process. We therefore analyzed a series of antioxidant enzymes in the blood of patients and compared with healthy age matched controls. In addition patients were given high doses of N-acetylcysteine (NAC), a glutathione percursor to determine if symptoms of EPM would improve. Five patients, four with EPM 1 (Unverricht-Lundborg disease) and one patient with EPM2 (Lafora body disease) were treated with 6 g/day of NAC. Before treatment, plasma samples were analyzed for glutathione peroxidase activity, catalase activity, extracellular superoxide dismutase (SOD) and CuZn-SOD and compared with the controls. Erythrocyte CuZn-SOD was significantly lower in the EPM patients compared to controls. NAC improved markedly and stabilized the neurological symptoms in patients with EPM 1 but had a doubtful effect in the patient with EPM 2.


Assuntos
Epilepsias Mioclônicas/enzimologia , Glutationa Peroxidase/metabolismo , Superóxido Dismutase/metabolismo , Acetilcisteína/uso terapêutico , Adulto , Epilepsias Mioclônicas/sangue , Epilepsias Mioclônicas/tratamento farmacológico , Epilepsias Mioclônicas/fisiopatologia , Eritrócitos/enzimologia , Feminino , Humanos , Doença de Lafora/sangue , Doença de Lafora/tratamento farmacológico , Doença de Lafora/enzimologia , Doença de Lafora/fisiopatologia , Masculino , Valores de Referência , Síndrome de Unverricht-Lundborg/tratamento farmacológico , Síndrome de Unverricht-Lundborg/metabolismo , Síndrome de Unverricht-Lundborg/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...