Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Brain ; 140(5): 1267-1279, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28335020

RESUMO

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.


Assuntos
Edema Encefálico/genética , Edema Encefálico/patologia , Cerebelo/patologia , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Neurônios/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Atrofia Óptica/genética , Atrofia Óptica/patologia , Espasmos Infantis/genética , Espasmos Infantis/patologia , Animais , Complexo do Signalossomo COP9 , Movimento Celular/genética , Movimento Celular/fisiologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Cerebelo/metabolismo , Edema/complicações , Edema/genética , Exoma/genética , Edição de Genes , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Microcefalia/complicações , Microcefalia/genética , Mutação de Sentido Incorreto/genética , Mutação de Sentido Incorreto/fisiologia , Neurônios/metabolismo , Proteínas Nucleares/biossíntese , Análise de Sequência de DNA , Fatores de Transcrição/biossíntese , Peixe-Zebra
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.
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
4.
Nat Genet ; 37(12): 1309-11, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16282978

RESUMO

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.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/genética , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas/metabolismo , Degenerações Espinocerebelares/genética , Degenerações Espinocerebelares/metabolismo , Chaperona BiP do Retículo Endoplasmático , Finlândia , Deleção de Genes , Fatores de Troca do Nucleotídeo Guanina/análise , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Músculo Esquelético/química , Mutação , Dobramento de Proteína
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.
Neurobiol Dis ; 47(3): 444-57, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22569358

RESUMO

The neuronal ceroid lipofuscinoses constitute the most common group of childhood neurodegenerative disorders. These devastating disorders still remain without effective treatment. The use of animal models has provided significant information about NCL pathogenesis, highlighting early glial activation and neuron loss in specific brain regions of affected animals. Here, we have characterized the timing and regional-specificity of the pathological events of CLN8 disease utilizing the Cln8 deficient mouse model, Cln8(mnd). We have studied the progression of neuron loss, astrocytosis and microglial activation from early to moderately symptomatic (1, 3 and 5 months) and late symptomatic (8 months) mice. In Cln8 deficiency, the somatosensory pathway comprising the thalamic ventral posterior nucleus (VPM/VPL) and the primary somatosensory cortex (S1BF) was found to be the most affected relay system. Scattered microglia that appeared partially activated were already present at 3 months of age, followed by astrocytosis and the loss of thalamic relay neurons at 5 months of age, with all these phenotypes and glial activation becoming more pronounced with disease progression. Reactive changes followed a similar pattern in the corresponding cortical target regions, but only moderate neuron loss was detected. Compared to the somatosensory system, in the visual thalamocortical pathway, neuron loss appeared relatively late in the disease, at 8 months. Neuron loss was preceded by glial activation in the dorsal lateral geniculate nucleus (LGNd) and in the primary visual cortex (V1). Taken together these data highlight the pathological targeting of the somatosensory thalamocortical pathway in Cln8 deficiency, in common with other forms of NCL. However, in contrast to other previously characterized NCL models, the Cln8(mnd) mouse shows relatively mild and late appearing pathology within the thalamocortical visual pathway.


Assuntos
Neuroglia/patologia , Lipofuscinoses Ceroides Neuronais/patologia , Neurônios/patologia , Córtex Somatossensorial/patologia , Tálamo/patologia , Vias Aferentes/fisiologia , Fatores Etários , Análise de Variância , Animais , Contagem de Células , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Proteínas de Membrana/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neuroglia/metabolismo , Neuroglia/ultraestrutura , Lipofuscinoses Ceroides Neuronais/genética , Neurônios/metabolismo , Neurônios/ultraestrutura
8.
Neurobiol Dis ; 46(1): 19-29, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22182690

RESUMO

CLN5 disease, late infantile variant phenotype neuronal ceroid lipofuscinosis, is a severe neurodegenerative disease caused by mutations in the CLN5 gene, which encodes a lysosomal protein of unknown function. Cln5-deficiency in mice leads to loss of thalamocortical neurons, and glial activation, but the underlying mechanisms are poorly understood. We have now studied the gene expression of Cln5 in the mouse brain and show that it increases gradually with age and differs between neurons and glia, with the highest expression in microglia. In Cln5(-/-) mice, we documented early and significant microglial activation that was already evident at 3 months of age. Loss of Cln5 also leads to defective myelination in vitro and in the developing mouse brain. This was accompanied by early alterations in serum lipid profiles, dysfunctional cellular metabolism and lipid transport in Cln5(-/-) mice. Taken together, these data provide significant new information about events associated with Cln5-deficiency, revealing altered myelination and disturbances in lipid metabolism, together with an early neuroimmune response.


Assuntos
Doenças Desmielinizantes/fisiopatologia , Metabolismo dos Lipídeos/fisiologia , Glicoproteínas de Membrana/deficiência , Microglia/metabolismo , Animais , Células Cultivadas , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Metabolismo dos Lipídeos/genética , Transtornos do Metabolismo dos Lipídeos/genética , Transtornos do Metabolismo dos Lipídeos/metabolismo , Transtornos do Metabolismo dos Lipídeos/patologia , Proteínas de Membrana Lisossomal , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/metabolismo , Lipofuscinoses Ceroides Neuronais/patologia , Neurônios/metabolismo , Neurônios/patologia
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.
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
11.
Neurobiol Dis ; 34(2): 308-19, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19385065

RESUMO

Finnish variant LINCL (vLINCL(Fin)) is the result of mutations in the CLN5 gene. To gain insights into the pathological staging of this fatal pediatric disorder, we have undertaken a stereological analysis of the CNS of Cln5 deficient mice (Cln5-/-) at different stages of disease progression. Consistent with human vLINCL(Fin), these Cln5-/- mice displayed a relatively late onset regional atrophy and generalized cortical thinning and synaptic pathology, preceded by early and localized glial responses within the thalamocortical system. However, in marked contrast to other forms of NCL, neuron loss in Cln5-/- mice began in the cortex and only subsequently occurred within thalamic relay nuclei. Nevertheless, as in other NCL mouse models, this progressive thalamocortical neuron loss was still most pronounced within the visual system. These data provide unexpected evidence for a distinctive sequence of neuron loss in the thalamocortical system of Cln5-/- mice, diametrically opposed to that seen in other forms of NCL.


Assuntos
Córtex Cerebral/patologia , Predisposição Genética para Doença/genética , Glicoproteínas de Membrana/genética , Degeneração Neural/patologia , Lipofuscinoses Ceroides Neuronais/patologia , Tálamo/patologia , Idade de Início , Animais , Atrofia/genética , Atrofia/patologia , Atrofia/fisiopatologia , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Modelos Animais de Doenças , Progressão da Doença , Finlândia , Proteínas de Membrana Lisossomal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Degeneração Neural/genética , Degeneração Neural/fisiopatologia , Vias Neurais/metabolismo , Vias Neurais/patologia , Vias Neurais/fisiopatologia , Lipofuscinoses Ceroides Neuronais/genética , Lipofuscinoses Ceroides Neuronais/fisiopatologia , Tálamo/metabolismo , Tálamo/fisiopatologia , Vias Visuais/metabolismo , Vias Visuais/patologia , Vias Visuais/fisiopatologia
12.
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
13.
BMC Genomics ; 9: 146, 2008 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-18371231

RESUMO

BACKGROUND: The neuronal ceroid lipofuscinoses (NCL) are a group of children's inherited neurodegenerative disorders, characterized by blindness, early dementia and pronounced cortical atrophy. The similar pathological and clinical profiles of the different forms of NCL suggest that common disease mechanisms may be involved. To explore the NCL-associated disease pathology and molecular pathways, we have previously produced targeted knock-out mice for Cln1 and Cln5. Both mouse-models replicate the NCL phenotype and neuropathology; the Cln1-/- model presents with early onset, severe neurodegenerative disease, whereas the Cln5-/- model produces a milder disease with a later onset. RESULTS: Here we have performed quantitative gene expression profiling of the cortex from 1 and 4 month old Cln1-/- and Cln5-/- mice. Combined microarray datasets from both mouse models exposed a common affected pathway: genes regulating neuronal growth cone stabilization display similar aberrations in both models. We analyzed locus specific gene expression and showed regional clustering of Cln1 and three major genes of this pathway, further supporting a close functional relationship between the corresponding gene products; adenylate cyclase-associated protein 1 (Cap1), protein tyrosine phosphatase receptor type F (Ptprf) and protein tyrosine phosphatase 4a2 (Ptp4a2). The evidence from the gene expression data, indicating changes in the growth cone assembly, was substantiated by the immunofluorescence staining patterns of Cln1-/- and Cln5-/- cortical neurons. These primary neurons displayed abnormalities in cytoskeleton-associated proteins actin and beta-tubulin as well as abnormal intracellular distribution of growth cone associated proteins GAP-43, synapsin and Rab3. CONCLUSION: Our data provide the first evidence for a common molecular pathogenesis behind neuronal degeneration in INCL and vLINCL. Since CLN1 and CLN5 code for proteins with distinct functional roles these data may have implications for other forms of NCLs as well.


Assuntos
Encéfalo/metabolismo , Regulação da Expressão Gênica/genética , Cones de Crescimento/patologia , Glicoproteínas de Membrana/genética , Lipofuscinoses Ceroides Neuronais/genética , Tioléster Hidrolases/genética , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Western Blotting , Células Cultivadas , Proteína GAP-43/metabolismo , Perfilação da Expressão Gênica , Genótipo , Cones de Crescimento/metabolismo , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Imuno-Histoquímica , Proteínas de Membrana Lisossomal , Camundongos , Camundongos Knockout , Lipofuscinoses Ceroides Neuronais/patologia , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Sinapsinas/metabolismo , Proteínas rab3 de Ligação ao GTP/metabolismo
14.
Eur J Hum Genet ; 16(8): 961-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18285827

RESUMO

Marinesco-Sjögren syndrome (MSS) is a rare autosomal recessively inherited neurodegenerative disorder characterized by cerebellar ataxia, cataracts, mental retardation, and progressive myopathy. Recently, mutations in the SIL1 gene, which encodes an endoplasmic reticulum (ER) resident cochaperone, were identified as a major cause of MSS. We here report four novel mutations in SIL1, including the first missense substitution p.Leu457Pro described in MSS. In addition, we excluded three functional candidate genes, HSPA5, HYOU1, and AARS, as causative genes in SIL1 mutation-negative patients. To understand the mechanisms of disturbed SIL1 function, we studied the subcellular localization of the missense mutant Leu457Pro protein in COS-1 cells. Moreover, we studied a mutant protein lacking the putative C-terminal ER retrieval signal. In contrast to the wild-type protein's localization to ER and Golgi apparatus, both mutant proteins formed aggregates within the ER depending on the expression level. These data imply that aggregation of mutant proteins may contribute to MSS pathogenesis. The genetic background of a subgroup of patients with MSS remains uncovered.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação de Sentido Incorreto/genética , Mutação/genética , Degenerações Espinocerebelares/genética , Adulto , Animais , Células COS , Pré-Escolar , Chlorocebus aethiops , Embrião de Mamíferos , Chaperona BiP do Retículo Endoplasmático , Feminino , Imunofluorescência , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Mutagênese Sítio-Dirigida , Neurônios/metabolismo , Neurônios/patologia , Linhagem , Degenerações Espinocerebelares/metabolismo , Degenerações Espinocerebelares/patologia , Frações Subcelulares
15.
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
16.
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
17.
BMC Neurosci ; 6: 27, 2005 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-15826318

RESUMO

BACKGROUND: The neuronal ceroid lipofuscinoses (NCLs) are a group of inherited neurodegenerative disorders characterized by accumulation of autofluorescent material in many tissues, especially in neurons. Mutations in the CLN8 gene, encoding an endoplasmic reticulum (ER) transmembrane protein of unknown function, underlie NCL phenotypes in humans and mice. The human phenotype is characterized by epilepsy, progressive psychomotor deterioration and visual loss, while motor neuron degeneration (mnd) mice with a Cln8 mutation show progressive motor neuron dysfunction and retinal degeneration. RESULTS: We investigated spatial and temporal expression of Cln8 messenger ribonucleic acid (mRNA) using in situ hybridization, reverse transcriptase polymerase chain reaction (RT-PCR) and northern blotting. Cln8 is ubiquitously expressed at low levels in embryonic and adult tissues. In prenatal embryos Cln8 is most prominently expressed in the developing gastrointestinal tract, dorsal root ganglia (DRG) and brain. In postnatal brain the highest expression is in the cortex and hippocampus. Expression of Cln8 mRNA in the central nervous system (CNS) was also analyzed in the hippocampal electrical kindling model of epilepsy, in which Cln8 expression was rapidly up-regulated in hippocampal pyramidal and granular neurons. CONCLUSION: Expression of Cln8 in the developing and mature brain suggests roles for Cln8 in maturation, differentiation and supporting the survival of different neuronal populations. The relevance of Cln8 up-regulation in hippocampal neurons of kindled mice should be further explored.


Assuntos
Encéfalo/metabolismo , Epilepsia/metabolismo , Hipocampo/metabolismo , Excitação Neurológica/metabolismo , Proteínas de Membrana/biossíntese , Neurônios/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Diferenciação Celular/genética , Sobrevivência Celular/genética , Modelos Animais de Doenças , Epilepsia/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Excitação Neurológica/genética , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/citologia , Regulação para Cima/fisiologia
18.
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.

19.
J Comp Neurol ; 455(3): 368-77, 2003 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-12483688

RESUMO

Palmitoyl protein thioesterase 1 (PPT1) is a depalmitoylating enzyme whose deficiency leads to infantile neuronal ceroid lipofuscinosis. The disease is characterized by early loss of vision and massive neuronal death. Although PPT1 is expressed in many tissues, a deficiency of PPT1 damages neurons only in the cerebral and cerebellar cortexes and retina; other cell types remain relatively unaffected. We previously demonstrated that PPT1 is present in the synaptosomes and synaptic vesicles of neurons. To understand the crucial role of PPT1 for neuronal cells, we further investigated the expression and targeting of PPT1 in retinal, hippocampal, and cortical neurons during their maturation in culture. We found that PPT1 activity increases by neuronal maturation and is highest in retinal neuron cultures. In retinal neurons the expression of PPT1 precedes that of the synaptic vesicle protein 2 and synaptophysin, indicating a significant role for PPT1 in the early development of neuronal cells. We also found by quantitative confocal immunofluorescence microscopy that PPT1 is targeted preferably to axons in mature neurons, as indicated by its colocalization with the axonal marker microtubule-associated protein 1. In axons PPT1 is targeted specifically to axonal varicosities and presynaptic terminals, as indicated by its significant colocalization with growth-associated protein 43 and synaptophysin. Axonal localization of PPT1 was confirmed by double labeling with synaptophysin and postembedding immunoelectron microscopy. The polarized axonal targeting of PPT1 may well indicate a role for PPT1 in the exocytotic pathway of neurons.


Assuntos
Axônios/enzimologia , Córtex Cerebral/crescimento & desenvolvimento , Hipocampo/crescimento & desenvolvimento , Neurônios/enzimologia , Retina/crescimento & desenvolvimento , Tioléster Hidrolases/metabolismo , Animais , Axônios/metabolismo , Axônios/ultraestrutura , Western Blotting , Técnicas de Cultura de Células , Córtex Cerebral/química , Córtex Cerebral/ultraestrutura , Hipocampo/química , Hipocampo/ultraestrutura , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica , Microscopia Imunoeletrônica , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/ultraestrutura , Terminações Pré-Sinápticas/metabolismo , Retina/química , Retina/ultraestrutura , Sinaptofisina/metabolismo
20.
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
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa