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










Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 15: 1069122, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36533126

RESUMEN

Cystatin B (CSTB) is a cysteine cathepsin inhibitor whose biallelic loss-of-function mutations in human result in defects in brain development and in neurodegeneration. The physiological function of CSTB is largely unknown, and the mechanisms underlying the human brain diseases remain poorly understood. We previously showed that CSTB modulates the proteolysis of the N-terminal tail of histone H3 (H3cs1) during in vitro neurogenesis. Here we investigated the significance of this mechanism in postnatal mouse brain. Spatiotemporal analysis of H3cs1 intensity showed that while H3cs1 in wild-type (wt) mice was found at varying levels during the first postnatal month, it was virtually absent in adult brain. We further showed that the high level of H3cs1 coincides with chromatin association of de novo synthesized cathepsin L suggesting a role for nuclear cathepsin L in brain development and maturation. On the contrary, the brains of Cstb -/- mice showed sustained H3cs1 proteolysis to adulthood with increased chromatin-associated cathepsin L activity, implying that CSTB regulates chromatin-associated cathepsin L activity in the postnatal mouse brain. As H3 tail proteolysis has been linked to cellular senescence in vitro, we explored the presence of several cellular senescence markers in the maturing Cstb -/- cerebellum, where we see increased levels of H3cs1. While several markers showed alterations in Cstb -/- mice, the results remained inconclusive regarding the association of deficient CSTB function with H3cs1-induced senescence. Together, we identify a molecular role for CSTB in brain with implications for brain development and disease.

2.
Neurobiol Dis ; 156: 105418, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34102276

RESUMEN

Cystatin B (CSTB) acts as an inhibitor of cysteine proteases of the cathepsin family and loss-of-function mutations result in human brain diseases with a genotype-phenotype correlation. In the most severe case, CSTB-deficiency disrupts brain development, and yet the molecular basis of this mechanism is missing. Here, we establish CSTB as a regulator of chromatin structure during neural stem cell renewal and differentiation. Murine neural precursor cells (NPCs) undergo transient proteolytic cleavage of the N-terminal histone H3 tail by cathepsins B and L upon induction of differentiation into neurons and glia. In contrast, CSTB-deficiency triggers premature H3 tail cleavage in undifferentiated self-renewing NPCs and sustained H3 tail proteolysis in differentiating neural cells. This leads to significant transcriptional changes in NPCs, particularly of nuclear-encoded mitochondrial genes. In turn, these transcriptional alterations impair the enhanced mitochondrial respiration that is induced upon neural stem cell differentiation. Collectively, our findings reveal the basis of epigenetic regulation in the molecular pathogenesis of CSTB deficiency.


Asunto(s)
Cistatina B/deficiencia , Histonas/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Animales , Células Cultivadas , Cistatina B/genética , Epigénesis Genética/fisiología , Histonas/genética , Ratones , Ratones de la Cepa 129 , Ratones Noqueados
3.
Am J Hum Genet ; 108(4): 722-738, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33798445

RESUMEN

Progressive myoclonus epilepsies (PMEs) comprise a group of clinically and genetically heterogeneous rare diseases. Over 70% of PME cases can now be molecularly solved. Known PME genes encode a variety of proteins, many involved in lysosomal and endosomal function. We performed whole-exome sequencing (WES) in 84 (78 unrelated) unsolved PME-affected individuals, with or without additional family members, to discover novel causes. We identified likely disease-causing variants in 24 out of 78 (31%) unrelated individuals, despite previous genetic analyses. The diagnostic yield was significantly higher for individuals studied as trios or families (14/28) versus singletons (10/50) (OR = 3.9, p value = 0.01, Fisher's exact test). The 24 likely solved cases of PME involved 18 genes. First, we found and functionally validated five heterozygous variants in NUS1 and DHDDS and a homozygous variant in ALG10, with no previous disease associations. All three genes are involved in dolichol-dependent protein glycosylation, a pathway not previously implicated in PME. Second, we independently validate SEMA6B as a dominant PME gene in two unrelated individuals. Third, in five families, we identified variants in established PME genes; three with intronic or copy-number changes (CLN6, GBA, NEU1) and two very rare causes (ASAH1, CERS1). Fourth, we found a group of genes usually associated with developmental and epileptic encephalopathies, but here, remarkably, presenting as PME, with or without prior developmental delay. Our systematic analysis of these cases suggests that the small residuum of unsolved cases will most likely be a collection of very rare, genetically heterogeneous etiologies.


Asunto(s)
Dolicoles/metabolismo , Mutación/genética , Epilepsias Mioclónicas Progresivas/genética , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Estudios de Cohortes , Variaciones en el Número de Copia de ADN/genética , Femenino , Glicosilación , Humanos , Intrones/genética , Masculino , Persona de Mediana Edad , Epilepsias Mioclónicas Progresivas/clasificación , Secuenciación del Exoma , Adulto Joven
4.
Ann Neurol ; 89(2): 402-407, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33085104

RESUMEN

Exome sequencing was performed in 2 unrelated families with progressive myoclonus epilepsy. Affected individuals from both families shared a rare, homozygous c.191A > G variant affecting a splice site in SLC7A6OS. Analysis of cDNA from lymphoblastoid cells demonstrated partial splice site abolition and the creation of an abnormal isoform. Quantitative reverse transcriptase polymerase chain reaction and Western blot showed a marked reduction of protein expression. Haplotype analysis identified a ~0.85cM shared genomic region on chromosome 16q encompassing the c.191A > G variant, consistent with a distant ancestor common to both families. Our results suggest that biallelic loss-of-function variants in SLC7A6OS are a novel genetic cause of progressive myoclonus epilepsy. ANN NEUROL 2021;89:402-407.


Asunto(s)
Epilepsias Mioclónicas Progresivas/genética , Péptido Hidrolasas/genética , Sitios de Empalme de ARN/genética , Adolescente , Ataxia/genética , Ataxia/fisiopatología , Atrofia , Western Blotting , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Disfunción Cognitiva/genética , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/psicología , ADN Complementario , Electroencefalografía , Femenino , Homocigoto , Humanos , Mutación con Pérdida de Función , Imagen por Resonancia Magnética , Masculino , Epilepsias Mioclónicas Progresivas/diagnóstico por imagen , Epilepsias Mioclónicas Progresivas/fisiopatología , Epilepsias Mioclónicas Progresivas/psicología , Linaje , Péptido Hidrolasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
5.
Neuropediatrics ; 49(4): 256-261, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29801191

RESUMEN

Alexander disease (AxD) is a genetic leukodystrophy caused by GFAP mutations leading to astrocyte dysfunction. Neonatal AxD is a rare phenotype with onset in the first month of life. The proband, belonging to a large pedigree with dominantly inherited benign familial neonatal epilepsy (BFNE), had a phenotype distinct from the rest of the family, with hypotonia and macrocephaly in addition to drug-resistant neonatal seizures. The patient deteriorated and passed away at 6 weeks of age. The pathological and neuroimaging data were consistent with the diagnosis of AxD. Genetic analysis of the proband identified a novel de novo GFAP missense mutation and a KCNQ2 splice site mutation segregating with the BFNE phenotype in the family. The GFAP mutation was located in the coil 2B region of GFAP protein, similar to most neonatal-onset AxD cases with an early death. The clinical and neuroradiological features of the previously published neonatal AxD patients are presented. This study further supports the classification of neonatal-onset AxD as a distinct phenotype based on the age of onset.


Asunto(s)
Enfermedad de Alexander/genética , Proteína Ácida Fibrilar de la Glía/genética , Mutación , Enfermedad de Alexander/diagnóstico por imagen , Enfermedad de Alexander/patología , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Resultado Fatal , Humanos , Lactante , Masculino , Fenotipo
6.
Brain ; 140(5): 1267-1279, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28335020

RESUMEN

Progressive encephalopathy with oedema, hypsarrhythmia, and optic atrophy (PEHO) syndrome is an early childhood onset, severe autosomal recessive encephalopathy characterized by extreme cerebellar atrophy due to almost total granule neuron loss. By combining homozygosity mapping in Finnish families with Sanger sequencing of positional candidate genes and with exome sequencing a homozygous missense substitution of leucine for serine at codon 31 in ZNHIT3 was identified as the primary cause of PEHO syndrome. ZNHIT3 encodes a nuclear zinc finger protein previously implicated in transcriptional regulation and in small nucleolar ribonucleoprotein particle assembly and thus possibly to pre-ribosomal RNA processing. The identified mutation affects a highly conserved amino acid residue in the zinc finger domain of ZNHIT3. Both knockdown and genome editing of znhit3 in zebrafish embryos recapitulate the patients' cerebellar defects, microcephaly and oedema. These phenotypes are rescued by wild-type, but not mutant human ZNHIT3 mRNA, suggesting that the patient missense substitution causes disease through a loss-of-function mechanism. Transfection of cell lines with ZNHIT3 expression vectors showed that the PEHO syndrome mutant protein is unstable. Immunohistochemical analysis of mouse cerebellar tissue demonstrated ZNHIT3 to be expressed in proliferating granule cell precursors, in proliferating and post-mitotic granule cells, and in Purkinje cells. Knockdown of Znhit3 in cultured mouse granule neurons and ex vivo cerebellar slices indicate that ZNHIT3 is indispensable for granule neuron survival and migration, consistent with the zebrafish findings and patient neuropathology. These results suggest that loss-of-function of a nuclear regulator protein underlies PEHO syndrome and imply that establishment of its spatiotemporal interaction targets will be the basis for developing therapeutic approaches and for improved understanding of cerebellar development.


Asunto(s)
Edema Encefálico/genética , Edema Encefálico/patología , Cerebelo/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Atrofia Óptica/genética , Atrofia Óptica/patología , Espasmos Infantiles/genética , Espasmos Infantiles/patología , Animales , Complejo del Señalosoma COP9 , Movimiento Celular/genética , Movimiento Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Cerebelo/metabolismo , Edema/complicaciones , Edema/genética , Exoma/genética , Edición Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Microcefalia/complicaciones , Microcefalia/genética , Mutación Missense/genética , Mutación Missense/fisiología , Neuronas/metabolismo , Proteínas Nucleares/biosíntesis , Análisis de Secuencia de ADN , Factores de Transcripción/biosíntesis , Pez Cebra
7.
J Neuroinflammation ; 13(1): 298, 2016 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-27894304

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited childhood-onset neurodegenerative disorder, characterized by myoclonus, seizures, and ataxia. Mutations in the cystatin B gene (CSTB) underlie EPM1. The CSTB-deficient (Cstb -/- ) mouse model recapitulates key features of EPM1, including myoclonic seizures. The mice show early microglial activation that precedes seizure onset and neuronal loss and leads to neuroinflammation. We here characterized the inflammatory phenotype of Cstb -/- mice in more detail. We found higher concentrations of chemokines and pro-inflammatory cytokines in the serum of Cstb -/- mice and higher CXCL13 expression in activated microglia in Cstb -/- compared to control mouse brains. The elevated chemokine levels were not accompanied by blood-brain barrier disruption, despite increased brain vascularization. Macrophages in the spleen and brain of Cstb -/- mice were predominantly pro-inflammatory. Taken together, these data show that CXCL13 expression is a hallmark of microglial activation in Cstb -/- mice and that the brain inflammation is linked to peripheral inflammatory changes, which might contribute to the disease pathology of EPM1.


Asunto(s)
Cistatina B/deficiencia , Encefalitis/etiología , Regulación de la Expresión Génica/genética , Inflamación/etiología , Epilepsias Mioclónicas Progresivas/complicaciones , Epilepsias Mioclónicas Progresivas/genética , Animales , Encéfalo/patología , Cistatina B/genética , Citocinas/sangre , Modelos Animales de Enfermedad , Encefalitis/sangre , Inflamación/sangre , Ratones , Ratones Noqueados , Microglía/metabolismo
8.
PLoS One ; 11(6): e0158195, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27355630

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1, OMIM254800) is an autosomal recessive neurodegenerative disorder characterized by stimulus-sensitive and action-activated myoclonus, tonic-clonic epileptic seizures, and ataxia. Loss-of-function mutations in the gene encoding the cysteine protease inhibitor cystatin B (CSTB) underlie EPM1. The deficiency of CSTB in mice (Cstb-/- mice) generates a phenotype resembling the symptoms of EPM1 patients and is accompanied by microglial activation at two weeks of age and an upregulation of immune system-associated genes in the cerebellum at one month of age. To shed light on molecular pathways and processes linked to CSTB deficiency in microglia we characterized the transcriptome of cultured Cstb-/- mouse microglia using microarray hybridization and RNA sequencing (RNA-seq). The gene expression profiles obtained with these two techniques were in good accordance and not polarized to either pro- or anti-inflammatory status. In Cstb-/- microglia, altogether 184 genes were differentially expressed. Of these, 33 genes were identified by both methods. Several interferon-regulated genes were weaker expressed in Cstb-/- microglia compared to control. This was confirmed by quantitative real-time PCR of the transcripts Irf7 and Stat1. Subsequently, we explored the biological context of CSTB deficiency in microglia more deeply by functional enrichment and canonical pathway analysis. This uncovered a potential role for CSTB in chemotaxis, antigen-presentation, and in immune- and defense response-associated processes by altering JAK-STAT pathway signaling. These data support and expand the previously suggested involvement of inflammatory processes to the disease pathogenesis of EPM1 and connect CSTB deficiency in microglia to altered expression of interferon-regulated genes.


Asunto(s)
Cistatina B/genética , Perfilación de la Expresión Génica , Interferones/metabolismo , Transducción de Señal , Síndrome de Unverricht-Lundborg/genética , Animales , Antiinflamatorios/química , Janus Quinasa 1/metabolismo , Ratones , Ratones Noqueados , Microglía/metabolismo , Mutación , Fenotipo , Factor de Transcripción STAT1/metabolismo , Análisis de Secuencia de ARN , Síndrome de Unverricht-Lundborg/patología
9.
Neurology ; 84(15): 1529-36, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25770194

RESUMEN

OBJECTIVE: This Finnish nationwide study aimed to refine the clinical phenotype variability and to identify factors that could explain the extensive variability in the clinical severity of the symptoms observed among patients with Unverricht-Lundborg disease (progressive myoclonus epilepsy type 1 [EPM1]) homozygous for the dodecamer expansion mutation in the cystatin B (CSTB) gene. METHODS: The study population consisted of 66 (33 men and 33 women) patients with genetically confirmed EPM1 homozygous for the CSTB expansion mutation for whom the sizes of the expanded alleles were determined. The clinical evaluation included videorecorded Unified Myoclonus Rating Scale and retrospectively collected medical history. The navigated transcranial magnetic stimulation test was used to determine motor threshold (MT) and silent period (SP) of the motor cortex. RESULTS: An earlier age at onset for EPM1 and longer disease duration were associated with more severe action myoclonus, lower performance IQ, increased MT, and prolonged SP. The number of dodecamer repeats in CSTB alleles varied between 38 and 77. On average, the size of the longer expanded alleles of patients was independently associated with MT, but exerted only a modulating effect on age at onset, myoclonus severity, and SP. CONCLUSIONS: As a group, earlier disease onset and longer duration are associated with more severe phenotype. Even though the vast majority of patients with EPM1 have a uniform genetic mutation, the actual size of the longer CSTB expansion mutation allele is likely to have a modulating effect on the age at disease onset, myoclonus severity, and cortical neurophysiology.


Asunto(s)
Cistatina B/genética , Corteza Motora/fisiopatología , Mioclonía/fisiopatología , Síndrome de Unverricht-Lundborg/fisiopatología , Adolescente , Adulto , Edad de Inicio , Niño , Femenino , Finlandia/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Fenotipo , Índice de Severidad de la Enfermedad , Factores de Tiempo , Estimulación Magnética Transcraneal , Síndrome de Unverricht-Lundborg/epidemiología , Síndrome de Unverricht-Lundborg/genética , Adulto Joven
10.
Nat Genet ; 47(1): 39-46, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25401298

RESUMEN

Progressive myoclonus epilepsies (PMEs) are a group of rare, inherited disorders manifesting with action myoclonus, tonic-clonic seizures and ataxia. We sequenced the exomes of 84 unrelated individuals with PME of unknown cause and molecularly solved 26 cases (31%). Remarkably, a recurrent de novo mutation, c.959G>A (p.Arg320His), in KCNC1 was identified as a new major cause for PME. Eleven unrelated exome-sequenced (13%) and two affected individuals in a secondary cohort (7%) had this mutation. KCNC1 encodes KV3.1, a subunit of the KV3 voltage-gated potassium ion channels, which are major determinants of high-frequency neuronal firing. Functional analysis of the Arg320His mutant channel showed a dominant-negative loss-of-function effect. Ten cases had pathogenic mutations in known PME-associated genes (NEU1, NHLRC1, AFG3L2, EPM2A, CLN6 and SERPINI1). Identification of mutations in PRNP, SACS and TBC1D24 expand their phenotypic spectra to PME. These findings provide insights into the molecular genetic basis of PME and show the role of de novo mutations in this disease entity.


Asunto(s)
Mutación Missense , Epilepsias Mioclónicas Progresivas/genética , Mutación Puntual , Canales de Potasio Shaw/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Secuencia de Bases , Proteínas Portadoras/genética , Secuencia Conservada , Exoma , Femenino , Proteínas Activadoras de GTPasa , Genes Dominantes , Proteínas de Choque Térmico/genética , Humanos , Masculino , Proteínas de la Membrana , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso , Linaje , Proteínas Priónicas , Priones/genética , Conformación Proteica , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Canales de Potasio Shaw/fisiología , Especificidad de la Especie
11.
Glia ; 63(3): 400-11, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25327891

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal-recessively inherited neurodegenerative disorder characterized by severely incapacitating myoclonus, seizures, and ataxia, and caused by loss-of-function mutations in the cystatin B gene (CSTB). A central neuropathological finding in the Cstb(-/-) mouse, an animal model for EPM1, is early microglial activation, which precedes astroglial activation, neuronal loss, and onset of myoclonus, thus implying a critical role for microglia in EPM1 pathogenesis. Here, we characterized phenotypic and functional properties of microglia from Cstb(-/-) mice utilizing brain tissue, microglia directly isolated from the brain, and primary microglial cultures. Our results show significantly higher Cstb mRNA expression in microglia than in neurons and astrocytes. In Cstb(-/-) mouse brain, expression of the inflammatory marker p-p38 MAPK and the proportion of both pro-inflammatory M1 and anti-inflammatory M2 microglia is higher than in control mice. Moreover, M1/M2 polarization of microglia in presymptomatic Cstb(-/-) mice is, compared to control mice, skewed towards M2 type at postnatal day 14 (P14), but towards M1 type at P30, a time point associated with onset of myoclonus. At this age, the high expression of both pro-inflammatory inducible nitric oxide synthase (iNOS) and anti-inflammatory arginase 1 (ARG1) in Cstb(-/-) mouse cortex is accompanied by the presence of peripheral immune cells. Consistently, activated Cstb(-/-) microglia show elevated chemokine release and chemotaxis. However, their MHCII surface expression is suppressed. Taken together, our results link CSTB deficiency to neuroinflammation with early activation and dysfunction of microglia and will open new avenues for therapeutic interventions for EPM1.


Asunto(s)
Encéfalo/inmunología , Cistatina B/deficiencia , Microglía/fisiología , Síndrome de Unverricht-Lundborg/inmunología , Animales , Arginasa/metabolismo , Astrocitos/metabolismo , Células Cultivadas , Cistatina B/genética , Modelos Animales de Enfermedad , Genes MHC Clase II/fisiología , Granulocitos/fisiología , Macrófagos/fisiología , Ratones de la Cepa 129 , Neuroinmunomodulación/fisiología , Neuronas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , ARN Mensajero/metabolismo , Linfocitos T/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
PLoS One ; 9(2): e89321, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24586687

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited neurodegenerative disease, manifesting with myoclonus, seizures and ataxia, caused by mutations in the cystatin B (CSTB) gene. With the aim of understanding the molecular basis of pathogenetic events in EPM1 we characterized gene expression changes in the cerebella of pre-symptomatic postnatal day 7 (P7) and symptomatic P30 cystatin B -deficient (Cstb(-/-) ) mice, a model for the disease, and in cultured Cstb(-/-) cerebellar granule cells using a pathway-based approach. Differentially expressed genes in P7 cerebella were connected to synaptic function and plasticity, and in cultured cerebellar granule cells, to cell cycle, cytoskeleton, and intracellular transport. In particular, the gene expression data pinpointed alterations in GABAergic pathway. Electrophysiological recordings from Cstb(-/-) cerebellar Purkinje cells revealed a shift of the balance towards decreased inhibition, yet the amount of inhibitory interneurons was not declined in young animals. Instead, we found diminished number of GABAergic terminals and reduced ligand binding to GABAA receptors in Cstb(-/-) cerebellum. These results suggest that alterations in GABAergic signaling could result in reduced inhibition in Cstb(-/-) cerebellum leading to the hyperexcitable phenotype of Cstb(-/-) mice. At P30, the microarray data revealed a marked upregulation of immune and defense response genes, compatible with the previously reported early glial activation that precedes neuronal degeneration. This further implies the role of early-onset neuroinflammation in the pathogenesis of EPM1.


Asunto(s)
Cerebelo/metabolismo , Cistatina B/genética , Regulación de la Expresión Génica , Epilepsias Mioclónicas Progresivas/genética , Neuronas/metabolismo , Animales , Animales Recién Nacidos , Cerebelo/inmunología , Modelos Animales de Enfermedad , Femenino , Neuronas GABAérgicas/metabolismo , Ligandos , Masculino , Ratones , Ratones Noqueados , Unión Proteica , Células de Purkinje/metabolismo , Receptores de GABA-A/metabolismo , Reproducibilidad de los Resultados , Potenciales Sinápticos , Factores de Tiempo
13.
J Neuropathol Exp Neurol ; 71(1): 40-53, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22157618

RESUMEN

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.


Asunto(s)
Cistatina B/deficiencia , Modelos Animales de Enfermedad , Microglía/metabolismo , Microglía/patología , Neuronas/patología , Síndrome de Unverricht-Lundborg/patología , Animales , Encéfalo , Muerte Celular/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Cistatina B/genética , Masculino , Ratones , Ratones Noqueados , Neuronas/metabolismo , Factores de Tiempo , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/metabolismo
14.
J Neurosci ; 29(18): 5910-5, 2009 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-19420257

RESUMEN

The progressive myoclonus epilepsies, featuring the triad of myoclonus, seizures, and ataxia, comprise a large group of inherited neurodegenerative diseases that remain poorly understood and refractory to treatment. The Cystatin B gene is mutated in one of the most common forms of progressive myoclonus epilepsy, Unverricht-Lundborg disease (EPM1). Cystatin B knockout in a mouse model of EPM1 triggers progressive degeneration of cerebellar granule neurons. Here, we report impaired redox homeostasis as a key mechanism by which Cystatin B deficiency triggers neurodegeneration. Oxidative stress induces the expression of Cystatin B in cerebellar granule neurons, and EPM1 patient-linked mutation of the Cystatin B gene promoter impairs oxidative stress induction of Cystatin B transcription. Importantly, Cystatin B knockout or knockdown sensitizes cerebellar granule neurons to oxidative stress-induced cell death. The Cystatin B deficiency-induced predisposition to oxidative stress in neurons is mediated by the lysosomal protease Cathepsin B. We uncover evidence of oxidative damage, reflected by depletion of antioxidants and increased lipid peroxidation, in the cerebellum of Cystatin B knock-out mice in vivo. Collectively, our findings define a pathophysiological mechanism in EPM1, whereby Cystatin B deficiency couples oxidative stress to neuronal death and degeneration, and may thus provide the basis for novel treatment approaches for the progressive myoclonus epilepsies.


Asunto(s)
Cistationina gamma-Liasa/deficiencia , Neuronas/fisiología , Estrés Oxidativo/genética , Síndrome de Unverricht-Lundborg/fisiopatología , Análisis de Varianza , Animales , Animales Recién Nacidos , Catepsina B , Muerte Celular/genética , Células Cultivadas , Cerebelo/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Galactósidos/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Ácido Glutámico/farmacología , Proteínas Fluorescentes Verdes/genética , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Noqueados , Neuronas/efectos de los fármacos , Oxidantes/farmacología , Oxidación-Reducción/efectos de los fármacos , ARN Interferente Pequeño/farmacología , Ratas , Transfección/métodos , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patología
15.
Epilepsia ; 49(4): 557-63, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18028412

RESUMEN

Unverricht-Lundborg disease (EPM1) is an autosomal recessively inherited neurodegenerative disorder and the most common single cause of progressive myoclonus epilepsy worldwide. Mutations in the gene encoding cystatin B (CSTB), a cysteine protease inhibitor, are responsible for the primary defect underlying EPM1. Here, progress toward understanding the molecular mechanisms in EPM1 is reviewed. We summarize the current knowledge about the CSTB gene and mutations as well as the cellular biology of the CSTB protein with emphasis on data emerging from analysis of EPM1 patients. We shed light on the disease mechanisms of EPM1 based on characterization of the CSTB-deficient mouse model.


Asunto(s)
Cistatinas/genética , Síndrome de Unverricht-Lundborg/genética , Animales , Cistatina B , Cistatinas/deficiencia , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación Missense/genética , Regiones Promotoras Genéticas/genética
16.
J Med Genet ; 44(2): 153-60, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16963483

RESUMEN

BACKGROUND: Usher syndrome, a combination of retinitis pigmentosa (RP) and sensorineural hearing loss with or without vestibular dysfunction, displays a high degree of clinical and genetic heterogeneity. Three clinical subtypes can be distinguished, based on the age of onset and severity of the hearing impairment, and the presence or absence of vestibular abnormalities. Thus far, eight genes have been implicated in the syndrome, together comprising 347 protein-coding exons. METHODS: To improve DNA diagnostics for patients with Usher syndrome, we developed a genotyping microarray based on the arrayed primer extension (APEX) method. Allele-specific oligonucleotides corresponding to all 298 Usher syndrome-associated sequence variants known to date, 76 of which are novel, were arrayed. RESULTS: Approximately half of these variants were validated using original patient DNAs, which yielded an accuracy of >98%. The efficiency of the Usher genotyping microarray was tested using DNAs from 370 unrelated European and American patients with Usher syndrome. Sequence variants were identified in 64/140 (46%) patients with Usher syndrome type I, 45/189 (24%) patients with Usher syndrome type II, 6/21 (29%) patients with Usher syndrome type III and 6/20 (30%) patients with atypical Usher syndrome. The chip also identified two novel sequence variants, c.400C>T (p.R134X) in PCDH15 and c.1606T>C (p.C536S) in USH2A. CONCLUSION: The Usher genotyping microarray is a versatile and affordable screening tool for Usher syndrome. Its efficiency will improve with the addition of novel sequence variants with minimal extra costs, making it a very useful first-pass screening tool.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos , Síndromes de Usher/genética , ADN/genética , Cartilla de ADN , Europa (Continente) , Variación Genética , Genotipo , Humanos
17.
Eur J Hum Genet ; 15(2): 185-93, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17003839

RESUMEN

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessive neurodegenerative disorder caused by mutations in the cystatin B gene (CSTB) that encodes an inhibitor of several lysosomal cathepsins. An unstable expansion of a dodecamer repeat in the CSTB promoter accounts for the majority of EPM1 disease alleles worldwide. We here describe a novel PCR protocol for detection of the dodecamer repeat expansion. We describe two novel EPM1-associated mutations, c.149G > A leading to the p.G50E missense change and an intronic 18-bp deletion (c.168+1_18del), which affects splicing of CSTB. The p.G50E mutation that affects the conserved QVVAG amino acid sequence critical for cathepsin binding fails to associate with lysosomes. This further supports the previously implicated physiological importance of the CSTB-lysosome association. Expression of CSTB mRNA and protein was markedly reduced in lymphoblastoid cells of the patients irrespective of the mutation type. Patients homozygous for the dodecamer expansion mutation showed 5-10% expression compared to controls. By combining database searches with RT-PCR we identified several alternatively spliced CSTB isoforms. One of these, CSTB2, was also present in mouse and was analyzed in more detail. In real-time PCR quantification, CSTB2 expression was less than 5% of total CSTB expression in all human adult and fetal tissues analyzed. In patients homozygous for the minisatellite mutation, the level of CSTB2 was reduced similarly to that of CSTB implicating regulation from the same promoter. The physiological significance of CSTB2 remains to be determined.


Asunto(s)
Cistatinas/genética , Epilepsias Mioclónicas Progresivas/genética , Síndrome de Unverricht-Lundborg/genética , Empalme Alternativo/genética , Cistatina B , Cistatinas/análisis , Cistatinas/metabolismo , Análisis Mutacional de ADN , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Repeticiones de Microsatélite , Mutación , Reacción en Cadena de la Polimerasa/métodos , Isoformas de Proteínas/genética , ARN Mensajero/análisis
18.
Gene ; 366(1): 180-8, 2006 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-16310976

RESUMEN

The TRIM37 gene encodes a peroxisomal protein of unknown function. Mutations in TRIM37 underlie mulibrey nanism, a rare autosomal recessively inherited disorder with severe growth failure of prenatal onset, constrictive pericardium, hepatomegaly and characteristic dysmorphic features. Eleven mulibrey nanism-associated mutations have been identified. We here characterised TRIM37 further by mapping the transcription initiation site and promoter region as well as by analysing splice variants. By primer extension analysis, several transcription initiation sites were localised to a region between -246 and -373 relative to the ATG codon for translation initiation. Basal promoter activity was mapped within 600 nucleotides upstream from the translation initiation site using promoter-luciferase reporter constructs. Several alternative splice variants of TRIM37 exist in databases. Most of these predict non-functional protein products, are expressed at low levels and are thus likely to be targets for nonsense-mediated mRNA decay. A novel splice variant, TRIM37b, with an alternative termination codon and 3'untranslated region (UTR) transcribed from an exon 16 kb downstream from exon 24, predicts an identical protein product with the previously identified transcript, TRIM37a. As seen by Northern blot analysis and quantitative real-time PCR, both transcripts are highly expressed in testis, whereas in other tissues TRIM37a is prominent. The 3'UTR of the PPM1E gene in the opposite strand overlaps TRIM37b. These data suggest that TRIM37 expression is regulated by several mechanisms: through nonsense surveillance of non-functional transcripts, as well as through 3'UTR regulatory sequences and/or naturally occurring antisense RNAs especially in testis.


Asunto(s)
Empalme Alternativo/genética , Regulación de la Expresión Génica/genética , Enanismo Mulibrey/genética , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , Transcripción Genética/genética , Regiones no Traducidas 3'/genética , Regiones no Traducidas 3'/metabolismo , Codón Iniciador/genética , Codón Iniciador/metabolismo , Codón de Terminación/genética , Codón de Terminación/metabolismo , Humanos , Masculino , Enanismo Mulibrey/metabolismo , Proteínas Nucleares/biosíntesis , Especificidad de Órganos , ARN sin Sentido/genética , ARN sin Sentido/metabolismo , Testículo/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
19.
Nat Genet ; 37(12): 1309-11, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16282978

RESUMEN

We identified the gene underlying Marinesco-Sjögren syndrome, which is characterized by cerebellar ataxia, progressive myopathy and cataracts. We identified four disease-associated, predicted loss-of-function mutations in SIL1, which encodes a nucleotide exchange factor for the heat-shock protein 70 (HSP70) chaperone HSPA5. These data, together with the similar spatial and temporal patterns of tissue expression of Sil1 and Hspa5, suggest that disturbed SIL1-HSPA5 interaction and protein folding is the primary pathology in Marinesco-Sjögren syndrome.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas/metabolismo , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/metabolismo , Chaperón BiP del Retículo Endoplásmico , Finlandia , Eliminación de Gen , Factores de Intercambio de Guanina Nucleótido/análisis , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Músculo Esquelético/química , Mutación , Pliegue de Proteína
20.
Eur J Hum Genet ; 10(6): 339-50, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12080385

RESUMEN

Usher syndrome type 3 (USH3) is an autosomal recessive disorder characterised by the association of post-lingual progressive hearing loss, progressive visual loss due to retinitis pigmentosa and variable presence of vestibular dysfunction. Because the previously defined transcripts do not account for all USH3 cases, we performed further analysis and revealed the presence of additional exons embedded in longer human and mouse USH3A transcripts and three novel USH3A mutations. Expression of Ush3a transcripts was localised by whole mount in situ hybridisation to cochlear hair cells and spiral ganglion cells. The full length USH3A transcript encodes clarin-1, a four-transmembrane-domain protein, which defines a novel vertebrate-specific family of three paralogues. Limited sequence homology to stargazin, a cerebellar synapse four-transmembrane-domain protein, suggests a role for clarin-1 in hair cell and photoreceptor cell synapses, as well as a common pathophysiological pathway for different Usher syndromes.


Asunto(s)
Células Ciliadas Auditivas/fisiología , Proteínas de la Membrana/genética , Sinapsis/fisiología , Secuencia de Aminoácidos , Animales , Canales de Calcio/genética , Mapeo Cromosómico , Femenino , Perfilación de la Expresión Génica , Humanos , Hibridación in Situ , Masculino , Proteínas de la Membrana/fisiología , Ratones , Datos de Secuencia Molecular , Mutación , Linaje , Filogenia , Alineación de Secuencia , Análisis de Secuencia de ADN , Análisis de Secuencia de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...