Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Neurochem ; 167(2): 296-317, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37753846

RESUMO

Mutations in PARK15, which encodes for the F-box protein FBXO7 have been associated with Parkinsonian Pyramidal syndrome, a rare and complex movement disorder with Parkinsonian symptoms, pyramidal tract signs and juvenile onset. Our previous study showed that systemic loss of Fbxo7 in mice causes motor defects and premature death. We have also demonstrated that FBXO7 has a crucial role in neurons as the specific deletion in tyrosine hydroxylase-positive or glutamatergic forebrain neurons leads to late-onset or early-onset motor dysfunction, respectively. In this study, we examined NEX-Cre;Fbxo7fl/fl mice, in which Fbxo7 was specifically deleted in glutamatergic projection neurons. The effects of FBXO7 deficiency on striatal integrity were investigated with HPLC and histological analyses. NEX-Cre;Fbxo7fl/fl mice revealed an increase in striatal dopamine concentrations, changes in the glutamatergic, GABAergic and dopaminergic pathways, astrogliosis and microgliosis and little or no neuronal loss in the striatum. To determine the effects on the integrity of the synapse, we purified synaptic membranes, subjected them to quantitative mass spectrometry analysis and found alterations in the complement system, endocytosis and exocytosis pathways. These neuropathological changes coincide with alterations in spontaneous home cage behavior. Taken together, our findings suggest that FBXO7 is crucial for corticostriatal projections and the synaptic integrity of the striatum, and consequently for proper motor control.

2.
Biochem J ; 478(12): 2179-2199, 2021 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-34060591

RESUMO

The regulation of proteasome activity is essential to cellular homeostasis and defects have been implicated in various disorders including Parkinson disease. The F-box protein FBXO7 has been implicated in early-onset parkinsonism and has previously been shown to have a regulatory role in proteasome activity and assembly. Here, we report the association of the E3 ubiquitin ligase FBXO7-SCF (SKP1, cullin-1, F-box protein) with the BAG6 complex, consisting of the subunits BAG6, GET4 and UBL4A. We identify the subunit GET4 as a direct interactor of FBXO7 and we show that the subunits GET4 and UBL4A are required for proper proteasome activity. Our findings demonstrate reduced binding of FBXO7 variants to GET4 and that FBXO7 variants bring about reduced proteasome activity. In addition, we find that GET4 is a non-proteolytic substrate of FBXO7, that binding of GET4 to BAG6 is enhanced in the presence of active FBXO7-SCF and that the cytoplasmic localization of the BAG6 complex is dependent on the E3 ubiquitin ligase activity. Taken together, our study shows that the parkinsonism-associated FBXO7 cooperates with the BAG6 complex in proteasome function and determines the subcellular localization of this complex.


Assuntos
Proteínas F-Box/metabolismo , Chaperonas Moleculares/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Frações Subcelulares/metabolismo , Ubiquitinas/metabolismo , Proteínas F-Box/genética , Células HEK293 , Humanos , Chaperonas Moleculares/genética , Mutação , Transtornos Parkinsonianos/genética , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Ubiquitinação , Ubiquitinas/genética
3.
Mol Cell Neurosci ; 112: 103602, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33581237

RESUMO

Ubiquitination is a key posttranslational modification for the controlled protein degradation and proteostasis. The substrate specificity is determined by a family of E3 ubiquitin ligases, which are encoded by more than 600 genes in the mammalian genome. Gain- or loss-of-function of a number of E3 genes results in neurodegeneration or neurodevelopmental disorders, affecting synapse function. This implies that the specific ubiquitination of synaptic substrates are of crucial importance for the normal neuronal network. In this review, we will summarize the history, current topics, and challenges in the field of ubiquitination-dependent regulations of synaptogenesis and synaptic transmission.


Assuntos
Encéfalo/enzimologia , Proteínas do Tecido Nervoso/fisiologia , Sinapses/enzimologia , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação , Animais , Encéfalo/patologia , Humanos , Camundongos , Família Multigênica , Degeneração Neural/enzimologia , Transtornos do Neurodesenvolvimento/enzimologia , Transtornos do Neurodesenvolvimento/genética , Plasticidade Neuronal , Doença de Parkinson/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Proteostase , Domínios RING Finger , Transmissão Sináptica , Ubiquitina-Proteína Ligases/classificação , Ubiquitina-Proteína Ligases/genética
4.
J Neurochem ; 155(5): 471-474, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33011998

RESUMO

Restless legs syndrome (RLS) is a movement disorder that is characterized by an uncomfortable sensation in the legs, and the urge to move the legs. Meis1 has previously identified as a risk gene for RLS. This Editorial highlights the study by Lyu and colleagues who developed a novel genetic mouse model heterozygous for Meis1 expression in neurons of the central nervous system. Using behavioral tests, the authors established hyperactivity of the mice, reminiscent of symptoms found in RLS patients. In addition, the authors took a closer look at the iron, dopaminergic, and cholinergic system of these mice.


Assuntos
Modelos Animais de Doenças , Modelos Genéticos , Proteína Meis1/deficiência , Proteína Meis1/genética , Síndrome das Pernas Inquietas/genética , Síndrome das Pernas Inquietas/metabolismo , Animais , Caenorhabditis elegans , Humanos , Camundongos , Camundongos Knockout , Síndrome das Pernas Inquietas/patologia
5.
J Neurosci ; 39(28): 5606-5626, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31085610

RESUMO

Myelination of axons facilitates the rapid propagation of electrical signals and the long-term integrity of axons. The ubiquitin-proteasome system is essential for proper protein homeostasis, which is particularly crucial for interactions of postmitotic cells. In our study, we examined how the E3 ubiquitin ligase FBXO7-SCF (SKP1, Cul1, F-box protein) expressed in myelinating cells affects the axon-myelin unit. Deletion of Fbxo7 in oligodendrocytes and Schwann cells in mice using the Cnp1-Cre driver line led to motor impairment due to hindlimb paresis. It did not result in apoptosis of myelinating cells, nor did it affect the proper myelination of axons or lead to demyelination. It however triggered axonal degeneration in the CNS and resulted in the severe degeneration of axons in the PNS, inducing a full-blown neuropathy. Both the CNS and PNS displayed inflammation, while the PNS was also characterized by fibrosis, massive infiltration of macrophages, and edema. Tamoxifen-induced deletion of Fbxo7, after myelination using the Plp1-CreERT2 line, led to a small number of degenerated axons and hence a very mild peripheral neuropathy. Interestingly, loss of Fbxo7 also resulted in reduced proteasome activity in Schwann cells but not in cerebellar granule neurons, indicating a specific sensitivity of the former cell type. Together, our results demonstrate an essential role for FBXO7 in myelinating cells to support associated axons, which is fundamental to the proper developmental establishment and the long-term integrity of the axon-myelin unit.SIGNIFICANCE STATEMENT The myelination of axons facilitates the fast propagation of electrical signals and the trophic support of the myelin-axon unit. Here, we report that deletion of Fbxo7 in myelinating cells in mice triggered motor impairment but had no effect on myelin biogenesis. Loss of Fbxo7 in myelinating glia, however, led to axonal degeneration in the CNS and peripheral neuropathy of the axonal type. In addition, we found that Schwann cells were particularly sensitive to Fbxo7 deficiency reflected by reduced proteasome activity. Based on these findings, we conclude that Fbxo7 is essential for the support of the axon-myelin unit and long-term axonal health.


Assuntos
Axônios/metabolismo , Proteínas F-Box/genética , Bainha de Mielina/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Animais , Apoptose , Axônios/patologia , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Proteínas F-Box/metabolismo , Feminino , Deleção de Genes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/patologia , Doenças do Sistema Nervoso Periférico/genética , Doenças do Sistema Nervoso Periférico/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo
6.
Bio Protoc ; 9(22): e3436, 2019 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33654932

RESUMO

In the nervous system of vertebrates, nerve impulse propagation is accelerated by the ensheathment of neuronal axons with myelin. Myelin sheaths are molecularly specialized, lipid-rich plasma membrane extensions of Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system (CNS). To visualize myelinated nerve fibers and to allow for the morphological analyses of myelin in the brain and the spinal cord, an efficient method for silver impregnation of myelin has originally been developed by Ferenc Gallyas in 1979, referred to as Gallyas silver impregnation. Gallyas' method is based on the agyrophilic characteristic of myelin to form and bind silver particles, while this process is suppressed in tissues other than myelin. The silver particles are finally enhanced in a developing step ("physical developer"). The main advantage of this method is that it efficiently visualizes both large myelinated fiber tracts and individual myelinated axons. Here we provide our laboratory protocol that is suitable for paraffin embedded sections and the use of light microscopy based on Gallyas' original protocol and subsequent modifications by Pistorio and colleagues.

7.
J Neurochem ; 144(2): 118-127, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29134665

RESUMO

Parkinson disease (PD) is, without doubt, a burden on modern society as the prevalence increases significantly with age. Owing to this growing number of PD cases, it is more critical than ever to understand the pathogenic mechanisms underlying PD to identify therapeutic targets. The discovery of genetic mutations associated with PD and parkinsonism paves the way toward this goal. Even though, familial forms of the disease represent the minority of PD cases and some forms are so rare that there are only a few affected families, the research on the associated genes is invaluable. Recent additions to PARK mutations are those in PARK15 that encodes the F-box protein O-type 7 (FBXO7). In this review, we highlight the recent research on FBXO7, which advances our knowledge of the etiopathological pathways and fills unexpected gaps therein, justifying the dedicated study of rare variants of PD.


Assuntos
Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Animais , Blefarospasmo/genética , Globo Pálido , Humanos , Camundongos , Doença de Parkinson Secundária/genética
8.
J Neurochem ; 139(2): 159-161, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27723099

RESUMO

This Editorial highlights a study by Huang and colleagues in the current issue of Journal of Neurochemistry. The authors introduce a novel ALS-FTD (amyotrophic lateral sclerosis-frontotemporal dementia) rat model to explore the role of the UBLQN2 gene that has previously been associated with familial ALS-FTD. Over-expression of ubiquilin 2 in the cortex (CTX) and hippocampus of the rat results in ubiquilin 2 aggregates and neurodegeneration together with cognitive deficits. The new rat model not only gives insight into potential molecular underpinnings of ALS-FTD, but also represents an important new tool for future research and therapeutic approaches. Read the highlighted article 'Increased Ubqln2 expression causes neuron death in transgenic rats' on page 285.


Assuntos
Esclerose Lateral Amiotrófica/genética , Degeneração Lobar Frontotemporal/genética , Neurônios/patologia , Ratos Transgênicos/genética , Ubiquitinas/genética , Esclerose Lateral Amiotrófica/patologia , Animais , Autofagia , Morte Celular , Degeneração Lobar Frontotemporal/patologia , Ratos
9.
EMBO J ; 35(18): 2008-25, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27497298

RESUMO

Mutations in the FBXO7 (PARK15) gene have been implicated in a juvenile form of parkinsonism termed parkinsonian pyramidal syndrome (PPS), characterized by Parkinsonian symptoms and pyramidal tract signs. FBXO7 (F-box protein only 7) is a subunit of the SCF (SKP1/cullin-1/F-box protein) E3 ubiquitin ligase complex, but its relevance and function in neurons remain to be elucidated. Here, we report that the E3 ligase FBXO7-SCF binds to and ubiquitinates the proteasomal subunit PSMA2. In addition, we show that FBXO7 is a proteasome-associated protein involved in proteasome assembly. In FBXO7 knockout mice, we find reduced proteasome activity and early-onset motor deficits together with premature death. In addition, we demonstrate that NEX (neuronal helix-loop-helix protein-1)-Cre-induced deletion of the FBXO7 gene in forebrain neurons or the loss of FBXO7 in tyrosine hydroxylase (TH)-positive neurons results in motor defects, reminiscent of the phenotype in PARK15 patients. Taken together, our study establishes a vital role for FBXO7 in neurons, which is required for proper motor control and accentuates the importance of FBXO7 in proteasome function.


Assuntos
Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Técnicas de Inativação de Genes , Transtornos Parkinsonianos/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Animais , Camundongos Knockout , Processamento de Proteína Pós-Traducional , Ubiquitinação
10.
PLoS One ; 10(9): e0137311, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26340347

RESUMO

The NG2 proteoglycan is characteristically expressed by oligodendrocyte progenitor cells (OPC) and also by aggressive brain tumours highly resistant to chemo- and radiation therapy. Oligodendrocyte-lineage cells are particularly sensitive to stress resulting in cell death in white matter after hypoxic or ischemic insults of premature infants and destruction of OPC in some types of Multiple Sclerosis lesions. Here we show that the NG2 proteoglycan binds OMI/HtrA2, a mitochondrial serine protease which is released from damaged mitochondria into the cytosol in response to stress. In the cytosol, OMI/HtrA2 initiates apoptosis by proteolytic degradation of anti-apoptotic factors. OPC in which NG2 has been downregulated by siRNA, or OPC from the NG2-knockout mouse show an increased sensitivity to oxidative stress evidenced by increased cell death. The proapoptotic protease activity of OMI/HtrA2 in the cytosol can be reduced by the interaction with NG2. Human glioma expressing high levels of NG2 are less sensitive to oxidative stress than those with lower NG2 expression and reducing NG2 expression by siRNA increases cell death in response to oxidative stress. Binding of NG2 to OMI/HtrA2 may thus help protect cells against oxidative stress-induced cell death. This interaction is likely to contribute to the high chemo- and radioresistance of glioma.


Assuntos
Antígenos/metabolismo , Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Proteínas Mitocondriais/metabolismo , Proteoglicanas/metabolismo , Serina Endopeptidases/metabolismo , Animais , Anticorpos Neutralizantes/farmacologia , Antígenos/genética , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Cerebelo/efeitos dos fármacos , Cerebelo/metabolismo , Cerebelo/patologia , Citosol/efeitos dos fármacos , Citosol/metabolismo , Glioblastoma/genética , Glioblastoma/patologia , Serina Peptidase 2 de Requerimento de Alta Temperatura A , Humanos , Peróxido de Hidrogênio/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Estresse Oxidativo , Cultura Primária de Células , Ligação Proteica , Proteoglicanas/antagonistas & inibidores , Proteoglicanas/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serina Endopeptidases/genética , Transdução de Sinais
11.
J Neurosci ; 35(23): 8701-17, 2015 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-26063905

RESUMO

The cerebellum is crucial for sensorimotor coordination. The cerebellar architecture not only requires proper development but also long-term integrity to ensure accurate functioning. Developmental defects such as impaired neuronal migration or neurodegeneration are thus detrimental to the cerebellum and can result in movement disorders including ataxias. In this study, we identify FBXO41 as a novel CNS-specific F-box protein that localizes to the centrosome and the cytoplasm of neurons and demonstrate that cytoplasmic FBXO41 promotes neuronal migration. Interestingly, deletion of the FBXO41 gene results in a severely ataxic gait in mice, which show delayed neuronal migration of granule neurons in the developing cerebellum in addition to deformities and degeneration of the mature cerebellum. We show that FBXO41 is a critical factor, not only for neuronal migration in the cerebellum, but also for its long-term integrity.


Assuntos
Encéfalo/patologia , Movimento Celular/genética , Proteínas F-Box/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Neurônios/patologia , Ataxias Espinocerebelares/genética , Ataxias Espinocerebelares/patologia , Animais , Animais Recém-Nascidos , Sobrevivência Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Proteínas F-Box/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Neuroglia/metabolismo , Neurônios/metabolismo , Fenótipo , Frações Subcelulares/metabolismo
12.
J Cereb Blood Flow Metab ; 35(8): 1233-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25966953

RESUMO

Traumatic brain injury causes progressive brain atrophy and cognitive decline. Surprisingly, an early treatment with erythropoietin (EPO) prevents these consequences of secondary neurodegeneration, but the mechanisms have remained obscure. Here we show by advanced imaging and innovative analytical tools that recombinant human EPO, a clinically established and neuroprotective growth factor, dampens microglial activity, as visualized also in vivo by a strongly attenuated injury-induced cellular motility.


Assuntos
Movimento Celular/efeitos dos fármacos , Eritropoetina/farmacologia , Microglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Células Cultivadas , Humanos , Proteínas Recombinantes/farmacologia
13.
J Vis Exp ; (85)2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24686379

RESUMO

Developmental events in the brain including neuronal morphogenesis and migration are highly orchestrated processes. In vitro and in vivo analyses allow for an in-depth characterization to identify pathways involved in these events. Cerebellar granule neurons (CGNs) that are derived from the developing cerebellum are an ideal model system that allows for morphological analyses. Here, we describe a method of how to genetically manipulate CGNs and how to study axono- and dendritogenesis of individual neurons. With this method the effects of RNA interference, overexpression or small molecules can be compared to control neurons. In addition, the rodent cerebellar cortex is an easily accessible in vivo system owing to its predominant postnatal development. We also present an in vivo electroporation technique to genetically manipulate the developing cerebella and describe subsequent cerebellar analyses to assess neuronal morphology and migration.


Assuntos
Movimento Celular/fisiologia , Cerebelo/fisiologia , Eletroporação/métodos , Neurônios/fisiologia , Transfecção/métodos , Animais , Axônios/fisiologia , Cerebelo/citologia , Dendritos/fisiologia , Neurônios/citologia , Ratos
14.
PLoS One ; 8(2): e57530, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23469015

RESUMO

Neuronal development requires proper migration, polarization and establishment of axons and dendrites. Growing evidence identifies the ubiquitin proteasome system (UPS) with its numerous components as an important regulator of various aspects of neuronal development. F-box proteins are interchangeable subunits of the Cullin-1 based E3 ubiquitin ligase, but only a few family members have been studied. Here, we report that the centrosomal E3 ligase FBXO31-SCF (Skp1/Cullin-1/F-box protein) regulates neuronal morphogenesis and axonal identity. In addition, we identified the polarity protein Par6c as a novel interaction partner and substrate targeted for proteasomal degradation in the control of axon but not dendrite growth. Finally, we ascribe a role for FBXO31 in dendrite growth and neuronal migration in the developing cerebellar cortex. Taken together, we uncovered the centrosomal E3 ligase FBXO31-SCF as a novel regulator of neuronal development.


Assuntos
Movimento Celular , Centrossomo/enzimologia , Proteínas F-Box/metabolismo , Morfogênese , Neurônios/citologia , Proteínas Ligases SKP Culina F-Box/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Sequência de Bases , Primers do DNA , Eletroporação , Células HEK293 , Humanos , Ubiquitinação
15.
PLoS One ; 7(11): e50735, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226367

RESUMO

Axon growth is an essential process during brain development. The E3 ubiquitin ligase Cdh1-APC has emerged as a critical regulator of intrinsic axon growth control. Here, we identified the RhoGAP p250GAP as a novel interactor of the E3 ubiquitin ligase Cdh1-APC and found that p250GAP promotes axon growth downstream of Cdh1-APC. We also report that p250GAP undergoes non-proteolytic ubiquitination and associates with the Cdh1 substrate Smurf1 to synergistically regulate axon growth. Finally, we found that in vivo knockdown of p250GAP in the developing cerebellar cortex results in impaired migration and axonal growth. Taken together, our data indicate that Cdh1-APC together with the RhoA regulators p250GAP and Smurf1 controls axon growth in the mammalian brain.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Axônios/metabolismo , Caderinas/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antígenos CD , Movimento Celular , Cerebelo/citologia , Cerebelo/crescimento & desenvolvimento , Cerebelo/metabolismo , Células HEK293 , Humanos , Camundongos , Ratos , Ubiquitinação
16.
Development ; 139(19): 3600-12, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22949615

RESUMO

Axon growth is an essential event during brain development and is extremely limited due to extrinsic and intrinsic inhibition in the adult brain. The E3 ubiquitin ligase Cdh1-anaphase promoting complex (APC) has emerged as an important intrinsic suppressor of axon growth. In this study, we identify in rodents the E3 ligase Smurf1 as a novel substrate of Cdh1-APC and that Cdh1 targets Smurf1 for degradation in a destruction box-dependent manner. We find that Smurf1 acts downstream of Cdh1-APC in axon growth and that the turnover of RhoA by Smurf1 is important in this process. In addition, we demonstrate that acute knockdown of Smurf1 in vivo in the developing cerebellar cortex results in impaired axonal growth and migration. Finally, we show that a stabilized form of Smurf1 overrides the inhibition of axon growth by myelin. Taken together, we uncovered a Cdh1-APC/Smurf1/RhoA pathway that mediates axonal growth suppression in the developing mammalian brain.


Assuntos
Axônios/fisiologia , Complexos Ubiquitina-Proteína Ligase/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Ciclossomo-Complexo Promotor de Anáfase , Animais , Animais Recém-Nascidos , Axônios/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Células HEK293 , Humanos , Neurogênese/genética , Neurogênese/fisiologia , Ratos , Ratos Wistar , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Complexos Ubiquitina-Proteína Ligase/genética , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/fisiologia
17.
Neuron ; 69(5): 930-44, 2011 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-21382553

RESUMO

Control of neuronal positioning is fundamental to normal brain development. However, the cell-intrinsic mechanisms that govern neuronal positioning remain to be elucidated. Here, we report that the spliced protein products of the transcriptional regulator SnoN, SnoN1 and SnoN2, harbor opposing functions in the coordinate regulation of neuronal branching and positioning. Knockdown of SnoN2 stimulates axon branching in primary neurons and impairs migration of granule neurons in the rat cerebellar cortex in vivo. By contrast, SnoN1 knockdown suppresses SnoN2 knockdown-induced neuronal branching and strikingly triggers excessive migration of granule neurons in the cerebellar cortex. We also find that SnoN1 forms a complex with the transcription factor FOXO1 that represses the X-linked lissencephaly gene encoding doublecortin (DCX). Accordingly, repression of DCX mediates the ability of SnoN1 to regulate branching in primary neurons and granule neuron migration in vivo. These data define an isoform-specific SnoN1-FOXO1 transcriptional complex that orchestrates neuronal branching and positioning in the brain with important implications for the study of developmental disorders of cognition and epilepsy.


Assuntos
Movimento Celular/fisiologia , Forma Celular/fisiologia , Cerebelo/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neurônios/fisiologia , Fatores de Transcrição/metabolismo , Análise de Variância , Animais , Western Blotting , Diferenciação Celular/fisiologia , Células Cultivadas , Cerebelo/citologia , Proteína Duplacortina , Imunoprecipitação , Neurônios/citologia , Ratos , Ratos Long-Evans
18.
F1000 Biol Rep ; 22010 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-20948796

RESUMO

The ubiquitin proteasome system (UPS) has drawn tremendous attention in the field of neuroscience. In recent years, we have gained insights into UPS-dependent mechanisms in brain development and disease. Several interesting studies over the past two years have highlighted the role of distinct E3 ubiquitin ligases in neurogenesis. Here, we will review the major findings in these studies and discuss their implications.

19.
Genes Dev ; 24(8): 799-813, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20395366

RESUMO

Neuronal polarity is essential for normal brain development and function. However, cell-intrinsic mechanisms that govern the establishment of neuronal polarity remain to be identified. Here, we report that knockdown of endogenous FOXO proteins in hippocampal and cerebellar granule neurons, including in the rat cerebellar cortex in vivo, reveals a requirement for the FOXO transcription factors in the establishment of neuronal polarity. The FOXO transcription factors, including the brain-enriched protein FOXO6, play a critical role in axo-dendritic polarization of undifferentiated neurites, and hence in a switch from unpolarized to polarized neuronal morphology. We also identify the gene encoding the protein kinase Pak1, which acts locally in neuronal processes to induce polarity, as a critical direct target gene of the FOXO transcription factors. Knockdown of endogenous Pak1 phenocopies the effect of FOXO knockdown on neuronal polarity. Importantly, exogenous expression of Pak1 in the background of FOXO knockdown in both primary neurons and postnatal rat pups in vivo restores the polarized morphology of neurons. These findings define the FOXO proteins and Pak1 as components of a cell-intrinsic transcriptional pathway that orchestrates neuronal polarity, thus identifying a novel function for the FOXO transcription factors in a unique aspect of neural development.


Assuntos
Polaridade Celular/fisiologia , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Neurônios/citologia , Neurônios/fisiologia , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Animais , Polaridade Celular/genética , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Hipocampo/fisiologia , Neurônios/metabolismo , Interferência de RNA , Ratos
20.
J Neurosci ; 29(13): 4312-21, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19339625

RESUMO

The transcriptional corepressor SnoN is a critical regulator of axonal morphogenesis, but how SnoN drives axonal growth is unknown. Here, we report that gene-profiling analyses in cerebellar granule neurons reveal that the large majority of genes altered upon SnoN knockdown are surprisingly downregulated, suggesting that SnoN may activate transcription in neurons. Accordingly, we find that the transcriptional coactivator p300 interacts with SnoN, and p300 plays a critical role in SnoN-induced axon growth. We also identify the gene encoding the signaling scaffold protein Ccd1 as a critical target of SnoN in neurons. Ccd1 localizes to the actin cytoskeleton, is enriched at axon terminals in neurons, and activates the axon growth-promoting kinase JNK (c-Jun N-terminal protein kinase). Knockdown of Ccd1 in neurons reduces axonal length and suppresses the ability of SnoN to promote axonal growth. Importantly, Ccd1 knockdown in rat pups profoundly impairs the formation of granule neuron parallel fiber axons in the rat cerebellar cortex in vivo. These findings define a novel SnoN-Ccd1 link that promotes axonal growth in the mammalian brain, with important implications for axonal development and regeneration.


Assuntos
Axônios/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Morfogênese/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Análise de Variância , Animais , Animais Recém-Nascidos , Células Cultivadas , Cerebelo/citologia , Chlorocebus aethiops , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Análise em Microsséries/métodos , Morfogênese/genética , Proteínas do Tecido Nervoso/genética , Interferência de RNA/fisiologia , RNA Mensageiro/metabolismo , Ratos , Ratos Long-Evans , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Transfecção/métodos , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...