Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.203
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 63(4): 608-620, 2016 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-27499294

RESUMO

The UbiB protein kinase-like (PKL) family is widespread, comprising one-quarter of microbial PKLs and five human homologs, yet its biochemical activities remain obscure. COQ8A (ADCK3) is a mammalian UbiB protein associated with ubiquinone (CoQ) biosynthesis and an ataxia (ARCA2) through unclear means. We show that mice lacking COQ8A develop a slowly progressive cerebellar ataxia linked to Purkinje cell dysfunction and mild exercise intolerance, recapitulating ARCA2. Interspecies biochemical analyses show that COQ8A and yeast Coq8p specifically stabilize a CoQ biosynthesis complex through unorthodox PKL functions. Although COQ8 was predicted to be a protein kinase, we demonstrate that it lacks canonical protein kinase activity in trans. Instead, COQ8 has ATPase activity and interacts with lipid CoQ intermediates, functions that are likely conserved across all domains of life. Collectively, our results lend insight into the molecular activities of the ancient UbiB family and elucidate the biochemical underpinnings of a human disease.


Assuntos
Comportamento Animal , Ataxia Cerebelar/enzimologia , Cerebelo/enzimologia , Proteínas Mitocondriais/deficiência , Músculo Esquelético/enzimologia , Ubiquinona/deficiência , Animais , Células COS , Ataxia Cerebelar/genética , Ataxia Cerebelar/fisiopatologia , Ataxia Cerebelar/psicologia , Cerebelo/fisiopatologia , Cerebelo/ultraestrutura , Chlorocebus aethiops , Modelos Animais de Doenças , Tolerância ao Exercício , Feminino , Predisposição Genética para Doença , Células HEK293 , Humanos , Metabolismo dos Lipídeos , Masculino , Aprendizagem em Labirinto , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Mitocondriais/química , Proteínas Mitocondriais/genética , Modelos Moleculares , Atividade Motora , Força Muscular , Músculo Esquelético/fisiopatologia , Fenótipo , Ligação Proteica , Conformação Proteica , Proteômica/métodos , Reconhecimento Psicológico , Teste de Desempenho do Rota-Rod , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Convulsões/enzimologia , Convulsões/genética , Convulsões/fisiopatologia , Relação Estrutura-Atividade , Fatores de Tempo , Transfecção , Ubiquinona/química , Ubiquinona/genética
2.
J Neurosci ; 39(13): 2542-2561, 2019 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-30683682

RESUMO

Maternal immune activation (MIA) is a principal environmental risk factor contributing to autism spectrum disorder (ASD), which compromises fetal brain development at critical periods of pregnancy and might be causally linked to ASD symptoms. We report that endogenous activation of the purinergic ion channel P2X7 (P2rx7) is necessary and sufficient to transduce MIA to autistic phenotype in male offspring. MIA induced by poly(I:C) injections to P2rx7 WT mouse dams elicited an autism-like phenotype in their offspring, and these alterations were not observed in P2rx7-deficient mice, or following maternal treatment with a specific P2rx7 antagonist, JNJ47965567. Genetic deletion and pharmacological inhibition of maternal P2rx7s also counteracted the induction of IL-6 in the maternal plasma and fetal brain, and disrupted brain development, whereas postnatal P2rx7 inhibition alleviated behavioral and morphological alterations in the offspring. Administration of ATP to P2rx7 WT dams also evoked autistic phenotype, but not in KO dams, implying that P2rx7 activation by ATP is sufficient to induce autism-like features in offspring. Our results point to maternal and offspring P2rx7s as potential therapeutic targets for the early prevention and treatment of ASD.SIGNIFICANCE STATEMENT Autism spectrum disorder (ASD) is a neurodevelopmental psychiatric disorder caused by genetic and environmental factors. Recent studies highlighted the importance of perinatal risks, in particular, maternal immune activation (MIA), showing strong association with the later emergence of ASD in the affected children. MIA could be mimicked in animal models via injection of a nonpathogenic agent poly(I:C) during pregnancy. This is the first report showing the key role of a ligand gated ion channel, the purinergic P2X7 receptor in MIA-induced autism-like behavioral and biochemical features. We show that genetic or pharmacological inhibition of both maternal and offspring P2X7 receptors could reverse the compromised brain development and autistic phenotype pointing to new possibilities for prevention and treatment of ASD.


Assuntos
Transtorno do Espectro Autista/imunologia , Receptores Purinérgicos P2X7/imunologia , Animais , Transtorno do Espectro Autista/induzido quimicamente , Transtorno do Espectro Autista/patologia , Cerebelo/ultraestrutura , Citocinas/imunologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli I-C/administração & dosagem , Gravidez , Efeitos Tardios da Exposição Pré-Natal/imunologia , Receptores Purinérgicos P2X7/genética
3.
Development ; 144(22): 4125-4136, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-29061636

RESUMO

During CNS development, interneuron precursors have to migrate extensively before they integrate in specific microcircuits. Known regulators of neuronal motility include classical neurotransmitters, yet the mechanisms that assure interneuron dispersal and interneuron/projection neuron matching during histogenesis remain largely elusive. We combined time-lapse video microscopy and electrophysiological analysis of the nascent cerebellum of transgenic Pax2-EGFP mice to address this issue. We found that cerebellar interneuronal precursors regularly show spontaneous postsynaptic currents, indicative of synaptic innervation, well before settling in the molecular layer. In keeping with the sensitivity of these cells to neurotransmitters, ablation of synaptic communication by blocking vesicular release in acute slices of developing cerebella slows migration. Significantly, abrogation of exocytosis primarily impedes the directional persistence of migratory interneuronal precursors. These results establish an unprecedented function of the early synaptic innervation of migrating neuronal precursors and demonstrate a role for synapses in the regulation of migration and pathfinding.


Assuntos
Movimento Celular , Interneurônios/citologia , Células-Tronco Neurais/citologia , Sinapses/metabolismo , Animais , Forma Celular , Cerebelo/citologia , Cerebelo/ultraestrutura , Fenômenos Eletrofisiológicos , Feminino , Glutamatos/metabolismo , Interneurônios/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Fator de Transcrição PAX2/metabolismo , Ácido gama-Aminobutírico/metabolismo
4.
EMBO Rep ; 19(9)2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29987134

RESUMO

Expansion microscopy is a recently introduced imaging technique that achieves super-resolution through physically expanding the specimen by ~4×, after embedding into a swellable gel. The resolution attained is, correspondingly, approximately fourfold better than the diffraction limit, or ~70 nm. This is a major improvement over conventional microscopy, but still lags behind modern STED or STORM setups, whose resolution can reach 20-30 nm. We addressed this issue here by introducing an improved gel recipe that enables an expansion factor of ~10× in each dimension, which corresponds to an expansion of the sample volume by more than 1,000-fold. Our protocol, which we termed X10 microscopy, achieves a resolution of 25-30 nm on conventional epifluorescence microscopes. X10 provides multi-color images similar or even superior to those produced with more challenging methods, such as STED, STORM, and iterative expansion microscopy (iExM). X10 is therefore the cheapest and easiest option for high-quality super-resolution imaging currently available. X10 should be usable in any laboratory, irrespective of the machinery owned or of the technical knowledge.


Assuntos
Microscopia de Fluorescência/métodos , Acrilamida/química , Animais , Linhagem Celular , Cerebelo/ultraestrutura , Chlorocebus aethiops , Etilenodiaminas/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Neurônios/ultraestrutura , Peroxissomos/ultraestrutura , Polimerização , Compostos de Potássio/química , Ratos , Ratos Wistar , Sulfatos/química , Sinapses/ultraestrutura , Tubulina (Proteína)/ultraestrutura
5.
Neuropathology ; 40(2): 167-179, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31797465

RESUMO

The cerebellar lesions of bovine spongiform encephalopathy (BSE)-infected guinea pigs were characterized as severe atrophy of the cerebellar cortex associated with the loss of granule cells, decrease in the width of the molecular layer, and intense protease-resistant prion protein (PrPSc ) accumulations that are similar to cerebellar lesions in kuru and the VV2 type of sporadic Creutzfeldt-Jakob disease. The aim of this study is to assess the relationships between the distribution and localization of PrPSc and synapses expressing neurotransmitter transporters in order to reveal the pathogenesis of the disease. We used cell-type-specific immunohistochemical makers recognizing glutamatergic and γ-aminobutylic acid (GABA)ergic terminals to identify terminals impaired with PrPSc accumulations. The distribution of PrPSc accumulations and immunoreactivity of synaptic vesicles were studied throughout the neuroanatomical pathways in cerebellar lesions. Time course study demonstrated that PrPSc accumulation showed a tendency to spread from granular layer to molecular layer. The immunoreactivity of vesicular glutamate transporter 1 (VGluT1) was localized in axon terminals of cerebellar granule cells, and decreased in association with the severity of PrPSc accumulations and loss of granule cells. Immunoreactivities of vesicular glutamate transporter 2 (VGluT2) and vesicular GABA transporter (VGAT) that exist in axon terminals of inferior olivary neurons and GABAergic synapses of Purkinje cells, respectively, were preserved well in these lesions. In brainstem, VGluT1 immunoreactivity decreased selectively in pontine nuclei that are a component of the pontocerebellar pathway, although other neurotransmitter immunoreactivities were preserved well. Our findings suggest that the selective loss of VGluT1-immunoreactive synapses subsequent to PrPSc accumulations can contribute to the pathogenesis of cerebellar lesions of BSE-infected guinea pigs.


Assuntos
Cerebelo/patologia , Encefalopatia Espongiforme Bovina/patologia , Neurônios/patologia , Proteínas PrPSc , Animais , Bovinos , Cerebelo/ultraestrutura , Feminino , Cobaias , Imuno-Histoquímica , Microscopia Eletrônica de Transmissão , Neurônios/ultraestrutura
6.
Brain ; 141(1): 85-98, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29244098

RESUMO

Hypoxia can injure brain white matter tracts, comprised of axons and myelinating oligodendrocytes, leading to cerebral palsy in neonates and delayed post-hypoxic leukoencephalopathy (DPHL) in adults. In these conditions, white matter injury can be followed by myelin regeneration, but myelination often fails and is a significant contributor to fixed demyelinated lesions, with ensuing permanent neurological injury. Non-myelinating oligodendrocyte precursor cells are often found in lesions in plentiful numbers, but fail to mature, suggesting oligodendrocyte precursor cell differentiation arrest as a critical contributor to failed myelination in hypoxia. We report a case of an adult patient who developed the rare condition DPHL and made a nearly complete recovery in the setting of treatment with clemastine, a widely available antihistamine that in preclinical models promotes oligodendrocyte precursor cell differentiation. This suggested possible therapeutic benefit in the more clinically prevalent hypoxic injury of newborns, and we demonstrate in murine neonatal hypoxic injury that clemastine dramatically promotes oligodendrocyte precursor cell differentiation, myelination, and improves functional recovery. We show that its effect in hypoxia is oligodendroglial specific via an effect on the M1 muscarinic receptor on oligodendrocyte precursor cells. We propose clemastine as a potential therapy for hypoxic brain injuries associated with white matter injury and oligodendrocyte precursor cell maturation arrest.


Assuntos
Clemastina/uso terapêutico , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/etiologia , Antagonistas dos Receptores Histamínicos H1/uso terapêutico , Hipóxia Encefálica/complicações , Recuperação de Função Fisiológica/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Doenças Desmielinizantes/diagnóstico por imagem , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Hipóxia Encefálica/diagnóstico por imagem , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Bainha de Mielina/efeitos dos fármacos , Bainha de Mielina/ultraestrutura , Células Precursoras de Oligodendrócitos/efeitos dos fármacos , Nervo Óptico/fisiopatologia , Oxigênio/farmacologia , Receptor Muscarínico M1/genética , Receptor Muscarínico M1/metabolismo
7.
Environ Toxicol ; 34(2): 103-111, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30375170

RESUMO

Intake of arsenic (As) via drinking water has been a serious threat to global public health. Though there are numerous reports of As neurotoxicity, its pathogenesis mechanisms remain vague especially its chronic effects on metabolic network. Hippocampus is a renowned area in relation to learning and memory, whilst recently, cerebellum is argued to be involved with process of cognition. Therefore, the study aimed to explore metabolomics alternations in these two areas after chronic As exposure, with the purpose of further illustrating details of As neurotoxicity. Twelve 3-week-old male C57BL/6J mice were divided into two groups, receiving deionized drinking water (control group) or 50 mg/L of sodium arsenite (via drinking water) for 24 weeks. Learning and memory abilities were tested by Morris water maze (MWM) test. Pathological and morphological changes of hippocampus and cerebellum were captured via transmission electron microscopy (TEM). Metabolic alterations were analyzed by gas chromatography-mass spectrometry (GC-MS). MWM test confirmed impairments of learning and memory abilities of mice after chronic As exposure. Metabolomics identifications indicated that tyrosine increased and aspartic acid (Asp) decreased simultaneously in both hippocampus and cerebellum. Intermediates (succinic acid) and indirect involved components of tricarboxylic acid cycle (proline, cysteine, and alanine) were found declined in cerebellum, indicating disordered energy metabolism. Our findings suggest that these metabolite alterations are related to As-induced disorders of amino acids and energy metabolism, which might therefore, play an important part in mechanisms of As neurotoxicity.


Assuntos
Arsênio/toxicidade , Cerebelo/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Redes e Vias Metabólicas/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais , Arsênio/metabolismo , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Cromatografia Gasosa-Espectrometria de Massas , Hipocampo/metabolismo , Hipocampo/ultraestrutura , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Memória/efeitos dos fármacos , Metabolômica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/patologia , Ratos , Poluentes Químicos da Água/metabolismo
8.
Am J Pathol ; 187(4): 864-883, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28342444

RESUMO

Farber disease is a rare autosomal recessive disorder caused by acid ceramidase deficiency that usually presents as early-onset progressive visceral and neurologic disease. To understand the neurologic abnormality, we investigated behavioral, biochemical, and cellular abnormalities in the central nervous system of Asah1P361R/P361R mice, which serve as a model of Farber disease. Behaviorally, the mutant mice had reduced voluntary locomotion and exploration, increased thigmotaxis, abnormal spectra of basic behavioral activities, impaired muscle grip strength, and defects in motor coordination. A few mutant mice developed hydrocephalus. Mass spectrometry revealed elevations of ceramides, hydroxy-ceramides, dihydroceramides, sphingosine, dihexosylceramides, and monosialodihexosylganglioside in the brain. The highest accumulation was in hydroxy-ceramides. Storage compound distribution was analyzed by mass spectrometry imaging and morphologic analyses and revealed involvement of a wide range of central nervous system cell types (eg, neurons, endothelial cells, and choroid plexus cells), most notably microglia and/or macrophages. Coalescing and mostly perivascular granuloma-like accumulations of storage-laden CD68+ microglia and/or macrophages were seen as early as 3 weeks of age and located preferentially in white matter, periventricular zones, and meninges. Neurodegeneration was also evident in specific cerebral areas in late disease. Overall, our central nervous system studies in Asah1P361R/P361R mice substantially extend the understanding of human Farber disease and suggest that this model can be used to advance therapeutic approaches for this currently untreatable disorder.


Assuntos
Sistema Nervoso Central/anormalidades , Lipogranulomatose de Farber/complicações , Lipogranulomatose de Farber/patologia , Malformações do Sistema Nervoso/etiologia , Malformações do Sistema Nervoso/patologia , Ceramidase Ácida/metabolismo , Animais , Comportamento Animal , Sistema Nervoso Central/patologia , Cerebelo/patologia , Cerebelo/ultraestrutura , Cérebro/patologia , Cérebro/ultraestrutura , Homozigoto , Hidrocefalia/patologia , Camundongos , Camundongos Transgênicos , Atividade Motora , Neurônios/patologia , Neurônios/ultraestrutura , Fenótipo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Esfingolipídeos/metabolismo , Fatores de Tempo
9.
PLoS Biol ; 13(3): e1002103, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25811491

RESUMO

Autophagy helps deliver sequestered intracellular cargo to lysosomes for proteolytic degradation and thereby maintains cellular homeostasis by preventing accumulation of toxic substances in cells. In a forward mosaic screen in Drosophila designed to identify genes required for neuronal function and maintenance, we identified multiple cacophony (cac) mutant alleles. They exhibit an age-dependent accumulation of autophagic vacuoles (AVs) in photoreceptor terminals and eventually a degeneration of the terminals and surrounding glia. cac encodes an α1 subunit of a Drosophila voltage-gated calcium channel (VGCC) that is required for synaptic vesicle fusion with the plasma membrane and neurotransmitter release. Here, we show that cac mutant photoreceptor terminals accumulate AV-lysosomal fusion intermediates, suggesting that Cac is necessary for the fusion of AVs with lysosomes, a poorly defined process. Loss of another subunit of the VGCC, α2δ or straightjacket (stj), causes phenotypes very similar to those caused by the loss of cac, indicating that the VGCC is required for AV-lysosomal fusion. The role of VGCC in AV-lysosomal fusion is evolutionarily conserved, as the loss of the mouse homologues, Cacna1a and Cacna2d2, also leads to autophagic defects in mice. Moreover, we find that CACNA1A is localized to the lysosomes and that loss of lysosomal Cacna1a in cerebellar cultured neurons leads to a failure of lysosomes to fuse with endosomes and autophagosomes. Finally, we show that the lysosomal CACNA1A but not the plasma-membrane resident CACNA1A is required for lysosomal fusion. In summary, we present a model in which the VGCC plays a role in autophagy by regulating the fusion of AVs with lysosomes through its calcium channel activity and hence functions in maintaining neuronal homeostasis.


Assuntos
Canais de Cálcio Tipo N/genética , Canais de Cálcio/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/metabolismo , Endossomos/metabolismo , Lisossomos/metabolismo , Neurônios/metabolismo , Fagossomos/metabolismo , Animais , Autofagia/genética , Cálcio/metabolismo , Canais de Cálcio/deficiência , Canais de Cálcio Tipo N/deficiência , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Proteínas de Drosophila/deficiência , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Endossomos/ultraestrutura , Feminino , Regulação da Expressão Gênica , Homeostase/genética , Lisossomos/ultraestrutura , Masculino , Fusão de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/ultraestrutura , Fagossomos/ultraestrutura , Cultura Primária de Células , Transmissão Sináptica , Vesículas Sinápticas/metabolismo , Vesículas Sinápticas/ultraestrutura
10.
Med Mol Morphol ; 51(4): 208-216, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29796936

RESUMO

Impaired nerve conduction, axonal degeneration, and synaptic alterations contribute to neurological disabilities in inflammatory demyelinating diseases. Cerebellar dysfunction is associated with demyelinating disorders, but the alterations of axon terminals in cerebellar gray matter during chronic demyelination are still unclear. We analyzed the morphological and ultrastructural changes of climbing fiber terminals in a mouse model of hereditary chronic demyelination. Three-dimensional ultrastructural analyses using serial block-face scanning electron microscopy and immunostaining for synaptic markers were performed in a demyelination mouse model caused by extra copies of myelin gene (PLP4e). At 1 month old, many myelinated axons were observed in PLP4e and wild-type mice, but demyelinated axons and axons with abnormally thin myelin were prominent in PLP4e mice at 5 months old. The density of climbing fiber terminals was significantly reduced in PLP4e mice at 5 months old. Reconstruction of climbing fiber terminals revealed that PLP4e climbing fibers had increased varicosity volume and enlarged mitochondria in the varicosities at 5-month-old mice. These results suggest that chronic demyelination is associated with alterations and loss of climbing fiber terminals in the cerebellar cortex, and that synaptic changes may contribute to cerebellar phenotypes observed in hereditary demyelinating disorders.


Assuntos
Cerebelo/ultraestrutura , Doenças Desmielinizantes/patologia , Mitocôndrias/ultraestrutura , Terminações Pré-Sinápticas/ultraestrutura , Animais , Cerebelo/patologia , Modelos Animais de Doenças , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Eletrônica de Varredura , Terminações Pré-Sinápticas/patologia
11.
Cell Tissue Res ; 368(3): 441-458, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28191598

RESUMO

Following activation of Gq protein-coupled receptors, phospholipase C yields a pair of second messengers: diacylglycerol (DG) and inositol 1,4,5-trisphosphate. Diacylglycerol kinase (DGK) phosphorylates DG to produce phosphatidic acid, another second messenger. Of the DGK family, DGKε is the only DGK isoform that exhibits substrate specificity for DG with an arachidonoyl acyl chain at the sn-2 position. Recently, we demonstrated that hydrophobic residues in the N-terminus of DGKε play an important role in targeting the endoplasmic reticulum in transfected cells. However, its cellular expression and subcellular localization in the brain remain elusive. In the present study, we investigate this issue using specific DGKε antibody. DGKε was richly expressed in principal neurons of higher brain regions, including pyramidal cells in the hippocampus and neocortex, medium spiny neurons in the striatum and Purkinje cells in the cerebellum. In Purkinje cells, DGKε was localized to the subsurface cisterns and colocalized with inositol 1,4,5-trisphosphate receptor-1 in dendrites and axons. In dendrites of Purkinje cells, DGKε was also distributed in close apposition to DG lipase-α, which catalyzes arachidonoyl-DG to produce 2-arachidonoyl glycerol, a major endocannabinoid in the brain. Behaviorally, DGKε-knockout mice exhibited hyper-locomotive activities and impaired motor coordination and learning. These findings suggest that DGKε plays an important role in neuronal and brain functions through its distinct neuronal expression and subcellular localization and also through coordinated arrangement with other molecules involving the phosphoinositide signaling pathway.


Assuntos
Cerebelo/enzimologia , Diacilglicerol Quinase/metabolismo , Células de Purkinje/enzimologia , Animais , Encéfalo/enzimologia , Cerebelo/citologia , Cerebelo/ultraestrutura , Diacilglicerol Quinase/genética , Células HeLa , Humanos , Immunoblotting , Imuno-Histoquímica , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Aprendizagem , Locomoção , Camundongos , Camundongos Knockout , Células PC12 , Fosfatidilinositóis/metabolismo , Desempenho Psicomotor , Células de Purkinje/ultraestrutura , Ratos , Ratos Wistar , Sistemas do Segundo Mensageiro , Distribuição Tecidual
12.
Bratisl Lek Listy ; 118(10): 575-579, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29198122

RESUMO

BACKGROUND: Mobile phone application may cause structural, functional changes and accumulation of toxic elements in brain. OBJECTIVES: The aim of this study was to investigate iron accumulation in rabbit cerebellum after exposure to RF EMF with light and scanning electron microscopy. MATERIALS AND METHODS: Histochemical analysis of iron distribution by light and electron microscopy with energy-dispersive microanalysis was used. RESULTS: Light microscopy revealed dystrophic changes of Purkinje cells in irradiated groups and iron deposits located in various parts of cerebellum. Deposits consists of C, O, Na, Mg, Al, Si, P, S, Cl, Ca and Fe. CONCLUSION: Our experiment revealed structural changes of Purkinje cells and iron and aluminium accumulations in stratum granulosum of rabbit's cerebellum after exposure to RF EMF (Fig. 6, Ref. 33).


Assuntos
Cerebelo/metabolismo , Campos Eletromagnéticos , Ferro/metabolismo , Ondas de Rádio , Alumínio/metabolismo , Animais , Telefone Celular , Cerebelo/patologia , Cerebelo/ultraestrutura , Microscopia Eletrônica , Células de Purkinje/metabolismo , Células de Purkinje/patologia , Células de Purkinje/ultraestrutura , Coelhos , Espectrometria por Raios X
13.
J Neurosci ; 35(13): 5293-306, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25834054

RESUMO

The demyelinating disease multiple sclerosis (MS) has an early inflammatory phase followed by an incurable progressive phase with subdued inflammation and poorly understood neurodegenerative mechanism. In this study, we identified various parallelisms between progressive MS and the dysmyelinating mouse model Shiverer and then genetically deleted a major neuron-specific mitochondrial anchoring protein Syntaphilin (SNPH) from the mouse. Prevailing evidence suggests that deletion of SNPH is harmful in demyelination. Surprisingly, SNPH deletion produces striking benefits in the Shiverer by prolonging survival, reducing cerebellar damage, suppressing oxidative stress, and improving mitochondrial health. In contrast, SNPH deletion does not benefit clinical symptoms in experimental autoimmune encephalomyelitis (EAE), a model for early-phase MS. We propose that deleting mitochondrial anchoring is a novel, specific treatment for progressive MS.


Assuntos
Modelos Animais de Doenças , Proteínas Associadas aos Microtúbulos/deficiência , Proteínas Associadas aos Microtúbulos/genética , Mitocôndrias/metabolismo , Esclerose Múltipla Crônica Progressiva/genética , Animais , Cerebelo/patologia , Cerebelo/ultraestrutura , Encefalomielite Autoimune Experimental/genética , Substância Cinzenta/patologia , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/genética , Esclerose Múltipla Crônica Progressiva/terapia , Proteínas do Tecido Nervoso , Estresse Oxidativo/genética , Análise de Sobrevida , Substância Branca/patologia
14.
Neurobiol Dis ; 86: 75-85, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26607784

RESUMO

An autosomal recessive disease of Black Russian Terriers was previously described as a juvenile-onset, laryngeal paralysis and polyneuropathy similar to Charcot Marie Tooth disease in humans. We found that in addition to an axonal neuropathy, affected dogs exhibit microphthalmia, cataracts, and miotic pupils. On histopathology, affected dogs exhibit a spongiform encephalopathy characterized by accumulations of abnormal, membrane-bound vacuoles of various sizes in neuronal cell bodies, axons and adrenal cells. DNA from an individual dog with this polyneuropathy with ocular abnormalities and neuronal vacuolation (POANV) was used to generate a whole genome sequence which contained a homozygous RAB3GAP1:c.743delC mutation that was absent from 73 control canine whole genome sequences. An additional 12 Black Russian Terriers with POANV were RAB3GAP1:c.743delC homozygotes. DNA samples from 249 Black Russian Terriers with no known signs of POANV were either heterozygotes or homozygous for the reference allele. Mutations in human RAB3GAP1 cause Warburg micro syndrome (WARBM), a severe developmental disorder characterized by abnormalities of the eye, genitals and nervous system including a predominantly axonal peripheral neuropathy. RAB3GAP1 encodes the catalytic subunit of a GTPase activator protein and guanine exchange factor for Rab3 and Rab18 respectively. Rab proteins are involved in membrane trafficking in the endoplasmic reticulum, axonal transport, autophagy and synaptic transmission. The neuronal vacuolation and membranous inclusions and vacuoles in axons seen in this canine disorder likely reflect alterations of these processes. Thus, this canine disease could serve as a model for WARBM and provide insight into its pathogenesis and treatment.


Assuntos
Mutação , Polineuropatias/genética , Síndrome de Walker-Warburg/genética , Proteínas rab3 de Ligação ao GTP/genética , Animais , Catarata/genética , Catarata/patologia , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Citoplasma/ultraestrutura , Modelos Animais de Doenças , Cães , Feminino , Músculos Laríngeos/ultraestrutura , Laringe/patologia , Masculino , Neurônios/metabolismo , Neurônios/ultraestrutura , Fenótipo , Polineuropatias/patologia , Polineuropatias/fisiopatologia , Polineuropatias/veterinária , Síndrome de Walker-Warburg/patologia , Síndrome de Walker-Warburg/fisiopatologia , Síndrome de Walker-Warburg/veterinária
15.
Mamm Genome ; 27(1-2): 8-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26662625

RESUMO

Key facets of mammalian forebrain cortical development include the radial migration of projection neurons and subsequent cellular differentiation into layer-specific subtypes. Inappropriate regulation of these processes can lead to a number of congenital brain defects in both mouse and human, including lissencephaly and intellectual disability. The genes regulating these processes are still not all identified, suggesting genetic analyses will continue to be a powerful tool in mechanistically studying the development of the cerebral cortex. Reelin is a molecule which we have understood to be critical for proper cortical development for many years. The precise mechanism of Reelin, however, is not fully understood. To address both of these unresolved issues, we report here the creation of a novel conditional allele of the Reelin gene and showcase the use of an Etv1-GFP transgenic line highlighting a subpopulation of the cortex: layer V pyramidal neurons. Together, these represent genetic tools which may facilitate the study of cortical development in a number of different ways.


Assuntos
Cerebelo/ultraestrutura , Córtex Cerebral/ultraestrutura , Efeito Fundador , Hipocampo/ultraestrutura , Camundongos Transgênicos , Células Piramidais/ultraestrutura , Alelos , Animais , Moléculas de Adesão Celular Neuronais/genética , Moléculas de Adesão Celular Neuronais/metabolismo , Movimento Celular , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Cruzamentos Genéticos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Feminino , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Células Piramidais/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína Reelina , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
16.
Methods ; 75: 79-86, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25644445

RESUMO

Autophagy is a highly dynamic process that mediates the degradation of cellular constituents inside lysosomes. It is characterized by the formation of autophagosomes, double membrane organelles that engulf cytosolic components and organelles and degrade their contents upon fusion with lysosomes. Upregulation of autophagy in response to specific stimuli can be determined by evaluating autophagic flux. This is achieved by comparing the number of autophagosomes in the absence and presence of lysosomal inhibitors. While the determination of autophagic flux in isolated cells is well-documented, few studies have described its determination in tissues or in vivo. Here, we describe the evaluation of autophagic flux both in vivo and ex vivo in several tissues, after treatment with lysosomal inhibitors and exposure to classical autophagy-inducing stimuli. This method uses LC3 lipidation, as determined by Western blot, fluorescence microscopy and flow cytometry. Our findings demonstrate that autophagic flux can be evaluated in vivo and ex vivo in several tissues.


Assuntos
Autofagia/genética , Fígado/ultraestrutura , Microscopia de Fluorescência/métodos , Retina/ultraestrutura , Animais , Cerebelo/metabolismo , Cerebelo/ultraestrutura , Leupeptinas/química , Fígado/metabolismo , Lisossomos/genética , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Camundongos , Retina/metabolismo
17.
J Neurosci ; 34(28): 9281-9, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009261

RESUMO

Neurons in the CNS do not regenerate following injury; regeneration is blocked by inhibitory proteins in myelin, such as myelin-associated glycoprotein (MAG). Elevating neuronal levels of the second messenger cAMP overcomes this blocked axonal outgrowth. One way to elevate cAMP is pretreating neurons with neurotrophins, such as brain-derived neurotrophic factor (BDNF). However, pleiotropic effects and poor bioavailability make exogenous administration of neurotrophins in vivo problematic; therefore, alternative targets must be considered. In neurons, two families of adenylyl cyclases synthesize cAMP, transmembrane adenylyl cyclases (tmACs), and soluble adenylyl cyclase (sAC). Here, we demonstrate that sAC is the essential source of cAMP for BDNF to overcome MAG-dependent inhibition of neurite outgrowth. Elevating sAC in rat and mouse neurons is sufficient to induce neurite outgrowth on myelin in vitro and promotes regeneration in vivo. These results suggest that stimulators of sAC might represent a novel therapeutic strategy to promote axonal growth and regeneration.


Assuntos
Adenilil Ciclases/química , Adenilil Ciclases/metabolismo , Axônios/fisiologia , Axônios/ultraestrutura , Cerebelo/metabolismo , Proteínas da Mielina/metabolismo , Regeneração Nervosa/fisiologia , Animais , Células CHO , Crescimento Celular , Células Cultivadas , Cerebelo/ultraestrutura , Cricetulus , Ativação Enzimática , Camundongos , Camundongos Knockout , Glicoproteína Associada a Mielina , Neurogênese/fisiologia , Ratos , Ratos Long-Evans , Solubilidade
18.
J Neurosci ; 34(22): 7412-24, 2014 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-24872547

RESUMO

Of the two members of the δ subfamily of ionotropic glutamate receptors, GluD2 is exclusively expressed at parallel fiber-Purkinje cell (PF-PC) synapses in the cerebellum and regulates their structural and functional connectivity. However, little is known to date regarding cellular and synaptic expression of GluD1 and its role in synaptic circuit formation. In the present study, we investigated this issue by producing specific and sensitive histochemical probes for GluD1 and analyzing cerebellar synaptic circuits in GluD1-knock-out mice. GluD1 was widely expressed in the adult mouse brain, with high levels in higher brain regions, including the cerebral cortex, striatum, limbic regions (hippocampus, nucleus accumbens, lateral septum, bed nucleus stria terminalis, lateral habenula, and central nucleus of the amygdala), and cerebellar cortex. In the cerebellar cortex, GluD1 mRNA was expressed at the highest level in molecular layer interneurons and its immunoreactivity was concentrated at PF synapses on interneuron somata. In GluD1-knock-out mice, the density of PF synapses on interneuron somata was significantly reduced and the size and number of interneurons were significantly diminished. Therefore, GluD1 is common to GluD2 in expression at PF synapses, but distinct from GluD2 in neuronal expression in the cerebellar cortex; that is, GluD1 in interneurons and GluD2 in PCs. Furthermore, GluD1 regulates the connectivity of PF-interneuron synapses and promotes the differentiation and/or survival of molecular layer interneurons. These results suggest that GluD1 works in concert with GluD2 for the construction of cerebellar synaptic wiring through distinct neuronal and synaptic expressions and also their shared synapse-connecting function.


Assuntos
Química Encefálica/fisiologia , Cerebelo/fisiologia , Regulação da Expressão Gênica/fisiologia , Interneurônios/metabolismo , Fibras Nervosas Mielinizadas/fisiologia , Receptores de Glutamato/biossíntese , Sinapses/fisiologia , Animais , Diferenciação Celular/fisiologia , Cerebelo/ultraestrutura , Glutamato Desidrogenase , Células HEK293 , Humanos , Interneurônios/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fibras Nervosas Mielinizadas/ultraestrutura , Rede Nervosa/fisiologia , Rede Nervosa/ultraestrutura , Receptores de Glutamato/genética , Receptores de Glutamato/fisiologia , Sinapses/ultraestrutura
19.
Neurobiol Dis ; 78: 57-67, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25843669

RESUMO

Nonsense mutations across the whole coding sequence of Syne1/Nesprin1 have been linked to autosomal recessive cerebellar ataxia Type I (ARCA1). However, nothing is known about the molecular etiology of this late-onset debilitating pathology. In this work, we report that Nesprin1 giant is specifically expressed in CNS tissues. We also identified a CNS-specific splicing event that leads to the abundant expression of a KASH-LESS variant of Nesprin1 giant (KLNes1g) in the cerebellum. KLNes1g displayed a noncanonical localization at glomeruli of cerebellar mossy fibers whereas Nesprin2 exclusively decorated the nuclear envelope of all cerebellar neurons. In immunogold electron microscopy, KLNes1g colocalized both with synaptic vesicles within mossy fibers and with dendritic membranes of cerebellar granule neurons. We further identified vesicle- and membrane-associated proteins in KLNes1g immunoprecipitates. Together, our results suggest that the loss of function of KLNes1g resulting from Nesprin1 nonsense mutations underlies the molecular etiology of ARCA1.


Assuntos
Cerebelo/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/metabolismo , Animais , Encéfalo/metabolismo , Cerebelo/ultraestrutura , Proteínas do Citoesqueleto , Camundongos , Fibras Nervosas/metabolismo , Fibras Nervosas/ultraestrutura , Neurônios/metabolismo , Neurônios/ultraestrutura
20.
J Neuroinflammation ; 12: 152, 2015 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-26310930

RESUMO

BACKGROUND: In brain inflammatory diseases, axonal damage is one of the most critical steps in the cascade that leads to permanent disability. Thus, identifying the initial events triggered by inflammation or oxidative stress that provoke axonal damage is critical for the development of neuroprotective therapies. Energy depletion due to mitochondrial dysfunction has been postulated as an important step in the damage of axons. This prompted us to study the effects of acute inflammation and oxidative stress on the morphology, transport, and function of mitochondria in axons. METHODS: Mouse cerebellar slice cultures were challenged with either lipopolysaccharide (LPS) or hydrogen peroxide (H2O2) ex vivo for 24 h. Axonal mitochondrial morphology was evaluated by transmission electron microscopy (TEM) and mitochondrial transportation by time-lapse imaging. In addition, mitochondrial function in the cerebellar slice cultures was analyzed through high-resolution respirometry assays and quantification of adenosine triphosphate (ATP) production. RESULTS: Both conditions promoted an increase in the size and complexity of axonal mitochondria evident in electron microscopy images, suggesting a compensatory response. Such compensation was reflected at the tissue level as increased respiratory activity of complexes I and IV and as a transient increase in ATP production in response to acute inflammation. Notably, time-lapse microscopy indicated that mitochondrial transport (mean velocity) was severely impaired in axons, increasing the proportion of stationary mitochondria in axons after LPS challenge. Indeed, the two challenges used produced different effects: inflammation mostly reducing retrograde transport and oxidative stress slightly enhancing retrograde transportation. CONCLUSIONS: Neuroinflammation acutely impairs axonal mitochondrial transportation, which would promote an inappropriate delivery of energy throughout axons and, by this way, contribute to axonal damage. Thus, preserving axonal mitochondrial transport might represent a promising avenue to exploit as a therapeutic target for neuroprotection in brain inflammatory diseases like multiple sclerosis.


Assuntos
Transporte Axonal/fisiologia , Axônios/ultraestrutura , Cerebelo/ultraestrutura , Mitocôndrias/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Animais Recém-Nascidos , Transporte Axonal/efeitos dos fármacos , Axônios/efeitos dos fármacos , Cerebelo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Peróxido de Hidrogênio/toxicidade , Técnicas In Vitro , Lipopolissacarídeos/toxicidade , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/ultraestrutura , Complexos Multienzimáticos/metabolismo , Técnicas de Cultura de Órgãos , Oxidantes/toxicidade , Fatores de Tempo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA