Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Bone Rep ; 3: 76-82, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28377970

RESUMO

Progressive myoclonus epilepsy of Unverricht-Lundborg type (EPM1) is an autosomal recessively inherited disorder characterized by incapacitating stimulus-sensitive myoclonus and tonic-clonic epileptic seizures with onset at the age of 6 to 16 years. EPM1 patients also exhibit a range of skeletal changes, e.g., thickened frontal cranial bone, arachnodactyly and scoliosis. Mutations in the gene encoding cystatin B (CSTB) underlie EPM1. CSTB is an inhibitor of cysteine cathepsins, including cathepsin K, a key enzyme in bone resorption by osteoclasts. CSTB has previously been shown to protect osteoclasts from experimentally induced apoptosis and to modulate bone resorption in vitro. Nevertheless, its physiological function in bone and the cause of the bone changes in patients remain unknown. Here we used the CSTB-deficient mouse (Cstb-/-) model of EPM1 to evaluate the contribution of defective CSTB protein function on bone pathology and osteoclast differentiation and function. Micro-computed tomography of hind limbs revealed thicker trabeculae and elevated bone mineral density in the trabecular bone of Cstb-/- mice. Histology from Cstb-/- mouse bones showed lower osteoclast count and thinner growth plates in long bones. Bone marrow-derived osteoclast cultures revealed lower osteoclast number and size in the Cstb-/- group. Cstb-/- osteoclasts formed less and smaller resorption pits in an in vitro assay. This impaired resorptive capacity was likely due to a decrease in osteoclast numbers and size. These data imply that the skeletal changes in Cstb-/- mice and in EPM1 patients are a result of CSTB deficiency leading to impaired osteoclast formation and consequently compromised resorptive capacity. These results suggest that the role of CSTB in osteoclast homeostasis and modulation of bone metabolism extends beyond cathepsin K regulation.

2.
Glia ; 63(3): 400-11, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25327891

RESUMO

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.


Assuntos
Encéfalo/imunologia , Cistatina B/deficiência , Microglia/fisiologia , Síndrome de Unverricht-Lundborg/imunologia , Animais , Arginase/metabolismo , Astrócitos/metabolismo , Células Cultivadas , Cistatina B/genética , Modelos Animais de Doenças , Genes MHC da Classe II/fisiologia , Granulócitos/fisiologia , Macrófagos/fisiologia , Camundongos da Linhagem 129 , Neuroimunomodulação/fisiologia , Neurônios/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Linfócitos T/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
3.
PLoS One ; 9(6): e90709, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24603771

RESUMO

Unverricht-Lundborg type progressive myoclonus epilepsy (EPM1, OMIM 254800) is an autosomal recessive disorder characterized by onset at the age of 6 to 16 years, incapacitating stimulus-sensitive myoclonus and tonic-clonic epileptic seizures. It is caused by mutations in the gene encoding cystatin B. Previously, widespread white matter changes and atrophy has been detected both in adult EPM1 patients and in 6-month-old cystatin B-deficient mice, a mouse model for the EPM1 disease. In order to elucidate the spatiotemporal dynamics of the brain atrophy and white matter changes in EPM1, we conducted longitudinal in vivo magnetic resonance imaging and ex vivo diffusion tensor imaging accompanied with tract-based spatial statistics analysis to compare volumetric changes and fractional anisotropy in the brains of 1 to 6 months of age cystatin B-deficient and control mice. The results reveal progressive but non-uniform volume loss of the cystatin B-deficient mouse brains, indicating that different neuronal populations possess distinct sensitivity to the damage caused by cystatin B deficiency. The diffusion tensor imaging data reveal early and progressive white matter alterations in cystatin B-deficient mice affecting all major tracts. The results also indicate that the white matter damage in the cystatin B-deficient brain is most likely secondary to glial activation and neurodegenerative events rather than a primary result of CSTB deficiency. The data also show that diffusion tensor imaging combined with TBSS analysis provides a feasible approach not only to follow white matter damage in neurodegenerative mouse models but also to detect fractional anisotropy changes related to normal white matter maturation and reorganisation.


Assuntos
Cerebelo/patologia , Cistatina B/deficiência , Tálamo/patologia , Síndrome de Unverricht-Lundborg/patologia , Animais , Imagem de Tensor de Difusão , Progressão da Doença , Feminino , Humanos , Imageamento por Ressonância Magnética , Camundongos Knockout , Tamanho do Órgão
4.
PLoS One ; 9(2): e89321, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586687

RESUMO

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.


Assuntos
Cerebelo/metabolismo , Cistatina B/genética , Regulação da Expressão Gênica , Epilepsias Mioclônicas Progressivas/genética , Neurônios/metabolismo , Animais , Animais Recém-Nascidos , Cerebelo/imunologia , Modelos Animais de Doenças , Feminino , Neurônios GABAérgicos/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica , Células de Purkinje/metabolismo , Receptores de GABA-A/metabolismo , Reprodutibilidade dos Testes , Potenciais Sinápticos , Fatores de Tempo
5.
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
6.
J Med Genet ; 49(6): 391-9, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22693283

RESUMO

BACKGROUND: The progressive myoclonus epilepsies (PMEs) comprise a group of clinically and genetically heterogeneous disorders characterised by myoclonus, epilepsy, and neurological deterioration. This study aimed to identify the underlying gene(s) in childhood onset PME patients with unknown molecular genetic background. METHODS: Homozygosity mapping was applied on genome-wide single nucleotide polymorphism data of 18 Turkish patients. The potassium channel tetramerisation domain-containing 7 (KCTD7) gene, previously associated with PME in a single inbred family, was screened for mutations. The spatiotemporal expression of KCTD7 was assessed in cellular cultures and mouse brain tissue. RESULTS: Overlapping homozygosity in 8/18 patients defined a 1.5 Mb segment on 7q11.21 as the major candidate locus. Screening of the positional candidate gene KCTD7 revealed homozygous missense mutations in two of the eight cases. Screening of KCTD7 in a further 132 PME patients revealed four additional mutations (two missense, one in-frame deletion, and one frameshift-causing) in five families. Eight patients presented with myoclonus and epilepsy and one with ataxia, the mean age of onset being 19 months. Within 2 years after onset, progressive loss of mental and motor skills ensued leading to severe dementia and motor handicap. KCTD7 showed cytosolic localisation and predominant neuronal expression, with widespread expression throughout the brain. None of three polypeptides carrying patient missense mutations affected the subcellular distribution of KCTD7. DISCUSSION: These data confirm the causality of KCTD7 defects in PME, and imply that KCTD7 mutation screening should be considered in PME patients with onset around 2 years of age followed by rapid mental and motor deterioration.


Assuntos
Mutação , Epilepsias Mioclônicas Progressivas/genética , Canais de Potássio/genética , Animais , Western Blotting , Química Encefálica , Células Cultivadas , Mapeamento Cromossômico , Homozigoto , Humanos , Espaço Intracelular , Camundongos , Microscopia de Fluorescência , Linhagem , Fenótipo , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Turquia
7.
Cell Metab ; 15(1): 100-9, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22225879

RESUMO

Somatic stem cell (SSC) dysfunction is typical for different progeroid phenotypes in mice with genomic DNA repair defects. MtDNA mutagenesis in mice with defective Polg exonuclease activity also leads to progeroid symptoms, by an unknown mechanism. We found that Polg-Mutator mice had neural (NSC) and hematopoietic progenitor (HPC) dysfunction already from embryogenesis. NSC self-renewal was decreased in vitro, and quiescent NSC amounts were reduced in vivo. HPCs showed abnormal lineage differentiation leading to anemia and lymphopenia. N-acetyl-L-cysteine treatment rescued both NSC and HPC abnormalities, suggesting that subtle ROS/redox changes, induced by mtDNA mutagenesis, modulate SSC function. Our results show that mtDNA mutagenesis affected SSC function early but manifested as respiratory chain deficiency in nondividing tissues in old age. Deletor mice, having mtDNA deletions in postmitotic cells and no progeria, had normal SSCs. We propose that SSC compartment is sensitive to mtDNA mutagenesis, and that mitochondrial dysfunction in SSCs can underlie progeroid manifestations.


Assuntos
DNA Mitocondrial/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Neurais/citologia , Acetilcisteína/farmacologia , Animais , Diferenciação Celular/genética , DNA Mitocondrial/metabolismo , Transporte de Elétrons , Eritropoese , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Linfopoese , Camundongos , Camundongos Mutantes , Doenças Mitocondriais/patologia , Mutagênese , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Oxirredução , Fenótipo , Espécies Reativas de Oxigênio/metabolismo
8.
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
9.
J Neurosci ; 29(18): 5910-5, 2009 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-19420257

RESUMO

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.


Assuntos
Cistationina gama-Liase/deficiência , Neurônios/fisiologia , Estresse Oxidativo/genética , Síndrome de Unverricht-Lundborg/fisiopatologia , Análise de Variância , Animais , Animais Recém-Nascidos , Catepsina B , Morte Celular/genética , Células Cultivadas , Cerebelo/patologia , Modelos Animais de Doenças , Progressão da Doença , Galactosídeos/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Ácido Glutâmico/farmacologia , Proteínas de Fluorescência Verde/genética , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Knockout , Neurônios/efeitos dos fármacos , Oxidantes/farmacologia , Oxirredução/efeitos dos fármacos , RNA Interferente Pequeno/farmacologia , Ratos , Transfecção/métodos , Síndrome de Unverricht-Lundborg/genética , Síndrome de Unverricht-Lundborg/patologia
10.
Exp Cell Res ; 314(15): 2895-905, 2008 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-18621045

RESUMO

Juvenile neuronal ceroid lipofuscinosis (JNCL, Batten disease) is the most common progressive neurodegenerative disorder of childhood. CLN3, the transmembrane protein underlying JNCL, is proposed to participate in multiple cellular events including membrane trafficking and cytoskeletal functions. We demonstrate here that CLN3 interacts with the plasma membrane-associated cytoskeletal and endocytic fodrin and the associated Na(+), K(+) ATPase. The ion pumping activity of Na(+), K(+) ATPase was unchanged in Cln3(-/-) mouse primary neurons. However, the immunostaining pattern of fodrin appeared abnormal in JNCL fibroblasts and Cln3(-/-) mouse brains suggesting disturbances in the fodrin cytoskeleton. Furthermore, the basal subcellular distribution as well as ouabain-induced endocytosis of neuron-specific Na(+), K(+) ATPase were remarkably affected in Cln3(-/-) mouse primary neurons. These data suggest that CLN3 is involved in the regulation of plasma membrane fodrin cytoskeleton and consequently, the plasma membrane association of Na(+), K(+) ATPase. Most of the processes regulated by multifunctional fodrin and Na(+), K(+) ATPase are also affected in JNCL and Cln3-deficiency implicating that dysregulation of fodrin cytoskeleton and non-pumping functions of Na(+), K(+) ATPase may play a role in the neuronal degeneration in JNCL.


Assuntos
Proteínas de Transporte/metabolismo , Membrana Celular/metabolismo , Citoesqueleto/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Chaperonas Moleculares/metabolismo , Lipofuscinoses Ceroides Neuronais/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Endocitose/fisiologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Íons/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Degeneração Neural/genética , Degeneração Neural/metabolismo , Lipofuscinoses Ceroides Neuronais/genética , Neurônios/metabolismo , Neurônios/patologia
11.
Hum Mol Genet ; 17(10): 1406-17, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18245779

RESUMO

Infantile neuronal ceroid lipofuscinosis (INCL) is a severe neurodegenerative disease caused by deficiency of palmitoyl protein thioesterase 1 (PPT1). INCL results in dramatic loss of thalamocortical neurons, but the disease mechanism has remained elusive. In the present work we describe the first interaction partner of PPT1, the F(1)-complex of the mitochondrial ATP synthase, by co-purification and in vitro-binding assays. In addition to mitochondria, subunits of F(1)-complex have been reported to localize in the plasma membrane, and to be capable of acting as receptors for various ligands such as apolipoprotein A-1. We verified here the plasma membrane localization of F(1)-subunits on mouse primary neurons and fibroblasts by cell surface biotinylation and TIRF-microscopy. To gain further insight into the Ppt1-mediated properties of the F(1)-complex, we utilized the Ppt1-deficient Ppt1(Delta ex4) mice. While no changes in the mitochondrial function could be detected in the brain of the Ppt1(Delta ex4) mice, the levels of F(1)-subunits alpha and beta on the plasma membrane were specifically increased in the Ppt1(Delta ex4) neurons. Significant changes were also detected in the apolipoprotein A-I uptake by the Ppt1(Delta ex4) neurons and the serum lipid composition in the Ppt1(Delta ex4) mice. These data indicate neuron-specific changes for F(1)-complex in the Ppt1-deficient cells and give clues for a possible link between lipid metabolism and neurodegeneration in INCL.


Assuntos
Colesterol/metabolismo , Lipofuscinoses Ceroides Neuronais/metabolismo , ATPases Translocadoras de Prótons/metabolismo , Tioléster Hidrolases/genética , Tioléster Hidrolases/metabolismo , Animais , Apolipoproteína A-I/sangue , Apolipoproteína A-I/metabolismo , Encéfalo/anormalidades , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/fisiopatologia , Membrana Celular/metabolismo , Colesterol/sangue , Complexo II de Transporte de Elétrons/metabolismo , Feminino , Humanos , Metabolismo dos Lipídeos , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Subunidades Proteicas/análise , Subunidades Proteicas/metabolismo , ATPases Translocadoras de Prótons/análise , Tioléster Hidrolases/sangue , Tioléster Hidrolases/isolamento & purificação
12.
Epilepsia ; 49(4): 557-63, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18028412

RESUMO

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.


Assuntos
Cistatinas/genética , Síndrome de Unverricht-Lundborg/genética , Animais , Cistatina B , Cistatinas/deficiência , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação de Sentido Incorreto/genética , Regiões Promotoras Genéticas/genética
13.
BMC Cell Biol ; 8: 22, 2007 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-17565660

RESUMO

BACKGROUND: Neuronal ceroid lipofuscinoses (NCLs) are collectively the most common type of recessively inherited childhood encephalopathies. The most severe form of NCL, infantile neuronal ceroid lipofuscinosis (INCL), is caused by mutations in the CLN1 gene, resulting in a deficiency of the lysosomal enzyme, palmitoyl protein thioesterase 1 (PPT1). The deficiency of PPT1 causes a specific death of neocortical neurons by a mechanism, which is currently unclear. To understand the function of PPT1 in more detail, we have further analyzed the basic properties of the protein, especially focusing on possible differences in non-neuronal and neuronal cells. RESULTS: Our study shows that the N-glycosylation of N197 and N232, but not N212, is essential for PPT1's activity and intracellular transport. Deglycosylation of overexpressed PPT1 produced in neurons and fibroblasts demonstrates differentially modified PPT1 in different cell types. Furthermore, antibody internalization assays showed differences in PPT1 transport when compared with a thoroughly characterized lysosomal enzyme aspartylglucosaminidase (AGA), an important observation potentially influencing therapeutic strategies. PPT1 was also demonstrated to form oligomers by size-exclusion chromatography and co-immunoprecipitation assays. Finally, the consequences of disease mutations were analyzed in the perspective of our new results, suggesting that the mutations increase both the degree of glycosylation of PPT1 and its ability to form complexes. CONCLUSION: Our current study describes novel properties for PPT1. We observe differences in PPT1 processing and trafficking in neuronal and non-neuronal cells, and describe for the first time the ability of PPT1 to form complexes. Understanding the basic characteristics of PPT1 is fundamental in order to clarify the molecular pathogenesis behind neurodegeneration in INCL.


Assuntos
Proteínas de Membrana/metabolismo , Neurônios/fisiologia , Tioléster Hidrolases/metabolismo , Animais , Aspartilglucosilaminase/metabolismo , Células COS , Técnicas de Cultura de Células , Chlorocebus aethiops , Glicosilação , Células HeLa , Humanos , Proteínas de Membrana/genética , Camundongos , Mutação , Neurônios/citologia , Neurônios/enzimologia , Especificidade de Órgãos , Células PC12 , Transporte Proteico , Ratos , Proteínas Recombinantes/metabolismo
14.
J Mol Med (Berl) ; 85(9): 971-83, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17530208

RESUMO

Rare monogenic dementias have repeatedly exposed novel pathways guiding to details of the molecular pathogenesis behind this complex clinical phenotype. In this paper, we have studied polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL), an early onset dementia with bone fractures caused by mutations in TYROBP (DAP12) and TREM2 genes, which encode important signaling molecules in human dendritic cells (DCs). To identify the pathways and biological processes associated with DAP12/TREM2-mediated signaling, we performed genome wide transcript analysis of in vitro differentiated DCs of PLOSL patients representing functional knockouts of either DAP12 or TREM2. Both DAP12- and TREM2-deficient cells differentiated into DCs and responded to pathogenic stimuli. However, the DCs showed morphological differences compared to control cells due to defects in the actin filaments. Not unexpectedly, transcript profiles of the patient DCs showed differential expression of genes involved in immune response. Importantly, significantly diverging transcript levels were also evident for genes earlier associated with other disorders of the central nervous system (CNS) and genes involved in the remodeling of bone, linking these two immunological genes with critical tissue phenotypes of patients. The data underline the functional diversity of the molecules of the innate immune system and implies their significant contribution also in demyelinating CNS disorders, including those resulting in dementia.


Assuntos
Doenças do Sistema Nervoso Central/genética , Doenças Desmielinizantes/genética , Células Dendríticas/metabolismo , Perfilação da Expressão Gênica , Actinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Northern Blotting , Diferenciação Celular/genética , Doenças do Sistema Nervoso Central/sangue , Citoesqueleto/metabolismo , Doenças Desmielinizantes/sangue , Células Dendríticas/citologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Regulação da Expressão Gênica , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Microscopia Confocal , Modelos Biológicos , Análise de Sequência com Séries de Oligonucleotídeos , Osteocondrodisplasias/sangue , Osteocondrodisplasias/genética , Receptores Imunológicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Nat Genet ; 38(2): 155-7, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16415886

RESUMO

Meckel syndrome (MKS) is a severe fetal developmental disorder reported in most populations. The clinical hallmarks are occipital meningoencephalocele, cystic kidney dysplasia, fibrotic changes of the liver and polydactyly. Here we report the identification of a gene, MKS1, mutated in MKS families linked to 17q. Mks1 expression in mouse embryos, as determined by in situ hybridization, agrees well with the tissue phenotype of MKS. Comparative genomics and proteomics data implicate MKS1 in ciliary functions.


Assuntos
Anormalidades Múltiplas/genética , Flagelos/metabolismo , Mutação/genética , Proteínas/genética , Proteoma/metabolismo , Anormalidades Múltiplas/metabolismo , Animais , Embrião de Mamíferos/metabolismo , Etnicidade/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Síndrome
16.
Hum Mol Genet ; 14(11): 1475-88, 2005 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-15843405

RESUMO

Hydrolethalus syndrome (HLS) is an autosomal recessive lethal malformation syndrome characterized by multiple developmental defects of fetus. We have earlier mapped and restricted the HLS region to a critical 1 cM interval on 11q23-25. The linkage disequilibrium (LD) and haplotype analyses of single nucleotide polymorphism (SNP) markers helped to further restrict the HLS locus to 476 kb between genes PKNOX2 and DDX25. An HLS associated mutation was identified in a novel regional transcript (GenBank accession no. FLJ32915), referred to here as the HYLS1 gene. The identified A to G transition results in a D211G change in the 299 amino acid polypeptide with unknown function. The HYLS1 gene shows alternative splicing and the transcript is found in multiple tissues during fetal development. In situ hybridization shows spatial and temporal distributions of transcripts in good agreement with the tissue phenotype of HLS patients. Immunostaining of in vitro expressed polypeptides from wild-type (WT) cDNA revealed cytoplasmic staining, whereas mutant polypeptides became localized in distinct nuclear structures, implying a disturbed cellular localization of the mutant protein. The Drosophila melanogaster model confirmed these findings and provides evidence for the significance of the mutation both in vitro and in vivo.


Assuntos
Anormalidades Múltiplas/genética , Genes Letais , Mutação de Sentido Incorreto , Proteínas/genética , Sequência de Aminoácidos , Animais , Animais Geneticamente Modificados , Sequência de Bases , Células COS , Chlorocebus aethiops , DNA Complementar , Drosophila melanogaster/genética , Desenvolvimento Fetal , Genes Recessivos , Humanos , Hibridização In Situ , Desequilíbrio de Ligação , Camundongos , Dados de Sequência Molecular , Proteínas/química , Proteínas/metabolismo , RNA Mensageiro/genética , Homologia de Sequência de Aminoácidos , Frações Subcelulares/metabolismo
17.
Neurobiol Dis ; 18(2): 314-22, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15686960

RESUMO

Polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL) is a recessively inherited disease characterized by early onset dementia associated with bone cysts. Our group has recently established the molecular background of PLOSL by identifying mutations in DAP12 and TREM2 genes. To understand how loss of function of the immune cell activating DAP12/TREM2 signaling complex leads to dementia and loss of myelin, we have analyzed here Dap12 and Trem2 expression in the mouse CNS. We show that Dap12 and Trem2 are expressed from embryonic stage to adulthood, and demonstrate a highly similar expression pattern. In addition, we identify microglial cells and oligodendrocytes as the major Dap12/Trem2-producing cells in the CNS and, consequently, as the predominant cell types involved in PLOSL pathogenesis. These findings provide a good starting point for the study of the molecular mechanisms of this inherited dementia and new evidence for the involvement of the immune system in neuronal degeneration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Sistema Nervoso Central/imunologia , Imunidade Inata/imunologia , Glicoproteínas de Membrana/imunologia , Doenças Neurodegenerativas/imunologia , Receptores Imunológicos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Animais Recém-Nascidos , Antígenos de Diferenciação/metabolismo , Biomarcadores , Células Cultivadas , Sistema Nervoso Central/embriologia , Sistema Nervoso Central/metabolismo , Demência/imunologia , Demência/metabolismo , Demência/fisiopatologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/fisiopatologia , Oligodendroglia/metabolismo , Ratos , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Síndrome
18.
Neurobiol Dis ; 15(2): 251-61, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15006695

RESUMO

SLC17A5 encodes a lysosomal membrane protein, sialin, which transports sialic acid from lysosomes. Mutations in sialin result in neurodegenerative sialic acid storage disorders, Salla disease (SD) and infantile sialic acid storage disease (ISSD). Here we analyzed sialin in mouse central nervous system (CNS) and primary cortical and hippocampal neurons and glia. In the CNS, sialin was predominantly expressed in neurons, especially in the proliferative zone of the prospective neocortex and the hippocampus in developing brain. In nonneuronal cells and primary glial cell cultures, mouse sialin was localized into lysosomes but interestingly, in primary neuronal cultures sialin was not targeted into lysosomes but rather revealed a punctate staining along the neuronal processes and was also seen in the plasma membrane. These data demonstrate a nonlysosomal localization of sialin in neurons and would imply a role for sialin in the secretory processes of neuronal cells.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Membrana Celular/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Transportadores de Ânions Orgânicos/metabolismo , Simportadores/metabolismo , Animais , Biomarcadores , Encéfalo/ultraestrutura , Diferenciação Celular/fisiologia , Membrana Celular/ultraestrutura , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Feto , Células HeLa , Hipocampo/embriologia , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Humanos , Imuno-Histoquímica , Camundongos , Ácido N-Acetilneuramínico/metabolismo , Neuritos/metabolismo , Neuritos/ultraestrutura , Neuroglia/citologia , Neuroglia/metabolismo , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Terminações Pré-Sinápticas/ultraestrutura , Doença do Armazenamento de Ácido Siálico/genética , Doença do Armazenamento de Ácido Siálico/metabolismo , Doença do Armazenamento de Ácido Siálico/patologia , Doença do Armazenamento de Ácido Siálico/fisiopatologia
19.
Hum Mol Genet ; 12(21): 2837-44, 2003 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12952869

RESUMO

The RECQL4 helicase gene is a member of the RECQL gene family, mutated in some Rothmund-Thomson syndrome (RTS) patients. Other members of this gene family are BLM mutated in Bloom syndrome, WRN mutated in Werner syndrome and RECQL and RECQL5. All polypeptides encoded by RECQL genes share a central region of seven helicase domains. The function of RECQL4 remains unknown, but based on the domain homology it possesses ATP-dependent DNA helicase activity such as BLM and WRN. Rothmund-Thomson, Bloom and Werner syndromes have overlapping clinical features, of which high predisposition to malignancies is the most remarkable feature. Here we report a fourth syndrome resulting in mutations in the RECQL genes. RAPADILINO syndrome is an autosomal recessive disorder characterized by short stature, radial ray defects and other malformations, as well as infantile diarrhoea, but not by a significant cancer risk. Four mutations in the RECQL4 gene were found in the Finnish patients, the most common mutation representing exon 7 in-frame deletion saving the helicase domain and showing dominant effect over other three nonsense mutations. The tissue expression of Recql4 in mouse well agrees with the tissue symptoms of RAPADILINO. The skeletal malformations in RAPADILINO and RTS patients as well as the high osteosarcoma risk in RTS propose a special role for RECQL4 in bone development.


Assuntos
Adenosina Trifosfatases/genética , Síndrome de Bloom/genética , DNA Helicases/genética , Síndrome de Rothmund-Thomson/genética , Síndrome de Werner/genética , Adenosina Trifosfatases/metabolismo , Animais , Sequência de Bases , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Células Cultivadas , DNA Helicases/metabolismo , Éxons , Fibroblastos/metabolismo , Humanos , Hibridização In Situ , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Dados de Sequência Molecular , Mutação , RecQ Helicases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA