Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
ACS Chem Neurosci ; 2023 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-37015082

RESUMO

A hexanucleotide repeat expansion (HRE) in an intron of gene C9ORF72 is the most common cause of familial amyotrophic lateral sclerosis and frontotemporal dementia. The HRE undergoes noncanonical translation (repeat-associated non-ATG translation) resulting in the production of five distinct dipeptide repeat (DPR) proteins. Arginine-rich DPR proteins have shown to be toxic to motor neurons, and recent evidence suggests this toxicity is associated with disruption of the ubiquitin-proteasome system. Here we report the ability of known 20S proteasome activator, TCH-165, to enhance the degradation of DPR proteins and overcome proteasome impairment evoked by DPR proteins. Furthermore, the 20S activator protects rodent motor neurons from DPR protein toxicity and restores proteostasis in cortical neuron cultures. This study suggests that 20S proteasome enhancers may have therapeutic efficacy in neurodegenerative diseases that display proteostasis defects.

2.
Mol Metab ; 60: 101468, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35248787

RESUMO

OBJECTIVES: Normal cellular function requires a rate of ATP production sufficient to meet demand. In most neurodegenerative diseases (including Amyotrophic Lateral Sclerosis [ALS]), mitochondrial dysfunction is postulated raising the possibility of impaired ATP production and a need for compensatory maneuvers to sustain the ATP production/demand balance. We investigated intermediary metabolism of neurons expressing familial ALS (fALS) genes and interrogated the functional consequences of glycolysis genes in fitness assays and neuronal survival. METHODS: We created a pure neuronal model system for isotopologue investigations of fuel utilization. In a yeast platform we studied the functional contributions of glycolysis genes in a growth fitness assay iafter expressing of a fALS gene. RESULTS: We find in our rodent models of fALS, a reduction in neuronal lactate production with maintained or enhanced activity of the neuronal citric acid cycle. This rewiring of metabolism is associated with normal ATP levels, bioenergetics, and redox status, thus supporting the notion that gross mitochondrial function is not compromised in neurons soon after expressing fALS genes. Genetic loss-of-function manipulation of individual steps in the glycolysis and the pentose phosphate pathway blunt the negative phenotypes seen in various fALS models. CONCLUSIONS: We propose that neurons adjust fuel utilization in the setting of neurodegenerative disease-associated alteration in mitochondrial function in a baleful manner and targeting this process can be healthful.


Assuntos
Esclerose Lateral Amiotrófica , Doenças Neurodegenerativas , Trifosfato de Adenosina , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Humanos , Doenças Neurodegenerativas/patologia , Neurônios/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(40)2021 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-34593637

RESUMO

A hexanucleotide repeat expansion in the C9orf72 gene is the most common cause of inherited amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Unconventional translation of the C9orf72 repeat produces dipeptide repeat proteins (DPRs). Previously, we showed that the DPRs PR50 and GR50 are highly toxic when expressed in Caenorhabditis elegans, and this toxicity depends on nuclear localization of the DPR. In an unbiased genome-wide RNA interference (RNAi) screen for suppressors of PR50 toxicity, we identified 12 genes that consistently suppressed either the developmental arrest and/or paralysis phenotype evoked by PR50 expression. All of these genes have vertebrate homologs, and 7 of 12 contain predicted nuclear localization signals. One of these genes was spop-1, the C. elegans homolog of SPOP, a nuclear localized E3 ubiquitin ligase adaptor only found in metazoans. SPOP is also required for GR50 toxicity and functions in a genetic pathway that includes cul-3, which is the canonical E3 ligase partner for SPOP Genetic or pharmacological inhibition of SPOP in mammalian primary spinal cord motor neurons suppressed DPR toxicity without affecting DPR expression levels. Finally, we find that knockdown of bromodomain proteins in both C. elegans and mammalian neurons, which are known SPOP ubiquitination targets, suppresses the protective effect of SPOP inhibition. Together, these data suggest a model in which SPOP promotes the DPR-dependent ubiquitination and degradation of BRD proteins. We speculate the pharmacological manipulation of this pathway, which is currently underway for multiple cancer subtypes, could also represent an entry point for therapeutic intervention to treat C9orf72 FTD/ALS.


Assuntos
Proteína C9orf72/metabolismo , Núcleo Celular/metabolismo , Dipeptídeos/metabolismo , Ligases/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Ubiquitina/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Caenorhabditis elegans/metabolismo , Células Cultivadas , Expansão das Repetições de DNA/fisiologia , Demência Frontotemporal/metabolismo , Neurônios Motores/metabolismo , Ratos , Medula Espinal/metabolismo
4.
Nat Neurosci ; 22(7): 1196, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31164751

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Nat Neurosci ; 22(6): 875-886, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31061493

RESUMO

Misfolded protein toxicity and failure of protein quality control underlie neurodegenerative diseases including amyotrophic lateral sclerosis and frontotemporal dementia. Here, we identified Lethal(3)malignant brain tumor-like protein 1 (L3MBTL1) as a key regulator of protein quality control, the loss of which protected against the proteotoxicity of mutant Cu/Zn superoxide dismutase or C9orf72 dipeptide repeat proteins. L3MBTL1 acts by regulating p53-dependent quality control systems that degrade misfolded proteins. SET domain-containing protein 8, an L3MBTL1-associated p53-binding protein, also regulated clearance of misfolded proteins and was increased by proteotoxicity-associated stresses in mammalian cells. Both L3MBTL1 and SET domain-containing protein 8 were upregulated in the central nervous systems of mouse models of amyotrophic lateral sclerosis and human patients with amyotrophic lateral sclerosis/frontotemporal dementia. The role of L3MBTL1 in protein quality control is conserved from Caenorhabditis elegans to mammalian neurons. These results reveal a protein quality-control pathway that operates in both normal stress response and proteotoxicity-associated neurodegenerative diseases.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Proteínas Cromossômicas não Histona/metabolismo , Demência Frontotemporal/metabolismo , Demência Frontotemporal/patologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Animais , Caenorhabditis elegans , Drosophila , Humanos , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Proteínas Repressoras , Proteínas Supressoras de Tumor
6.
Mol Cell ; 71(5): 703-717.e9, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30100264

RESUMO

In amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration (FTD), cytoplasmic aggregates of hyperphosphorylated TDP-43 accumulate and colocalize with some stress granule components, but how pathological TDP-43 aggregation is nucleated remains unknown. In Drosophila, we establish that downregulation of tankyrase, a poly(ADP-ribose) (PAR) polymerase, reduces TDP-43 accumulation in the cytoplasm and potently mitigates neurodegeneration. We establish that TDP-43 non-covalently binds to PAR via PAR-binding motifs embedded within its nuclear localization sequence. PAR binding promotes liquid-liquid phase separation of TDP-43 in vitro and is required for TDP-43 accumulation in stress granules in mammalian cells and neurons. Stress granule localization initially protects TDP-43 from disease-associated phosphorylation, but upon long-term stress, stress granules resolve, leaving behind aggregates of phosphorylated TDP-43. Finally, small-molecule inhibition of Tankyrase-1/2 in mammalian cells inhibits formation of cytoplasmic TDP-43 foci without affecting stress granule assembly. Thus, Tankyrase inhibition antagonizes TDP-43-associated pathology and neurodegeneration and could have therapeutic utility for ALS and FTD.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Esclerose Lateral Amiotrófica/metabolismo , Animais , Células COS , Linhagem Celular , Núcleo Celular/metabolismo , Chlorocebus aethiops , Citoplasma/metabolismo , Drosophila , Feminino , Degeneração Lobar Frontotemporal/metabolismo , Masculino , Mamíferos/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Fosforilação/fisiologia , Ratos , Ratos Sprague-Dawley
7.
eNeuro ; 4(6)2017.
Artigo em Inglês | MEDLINE | ID: mdl-29218323

RESUMO

The dendritic tree is a key determinant of neuronal information processing. In the motor system, the dendritic tree of spinal cord neurons undergoes dramatic remodeling in an activity-dependent manner during early postnatal life. This leads to the proper segmental spinal cord connectivity that subserves normal locomotor behavior. One molecular system driving the establishment of dendrite architecture of mammalian motor neurons relies on AMPA receptors (AMPA-Rs) assembled with the GluA1 subunit, and this occurs in an NMDA receptor (NMDA-R)-independent manner. The dendrite growth promoting activity of GluA1-containing AMPA-Rs depends on its intracellular binding partner, SAP97, and SAP97's PDZ3 domain. We show here that cysteine-rich interactor of PDZ3 (CRIPT) is a bona fide SAP97 PDZ3-domain binding partner, localizes to synapses with GluA1 and SAP97 along the dendritic tree, and is a determinant of the dendritic growth of mammalian spinal cord neurons. We further show that CRIPT has a well-conserved ortholog in the nematode, Caenorhabditis elegans, and animals lacking CRIPT display decreased dendrite branching of the well-studied PVD neuron in vivo. The lack of CRIPT leads to a selective defect in touch perception, and this is rescued by expression of wild-type (WT) human CRIPT (hCRIPT) in the nervous system. This work brings new light into the molecular machinery that drives dendritic growth during development and may prove relevant to the promotion of nervous system plasticity following insult.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dendritos , Proteínas de Membrana/metabolismo , Neurogênese/fisiologia , Medula Espinal/crescimento & desenvolvimento , Medula Espinal/metabolismo , Animais , Caenorhabditis elegans , Proteína 1 Homóloga a Discs-Large , Células HEK293 , Humanos , Ratos
8.
eNeuro ; 4(1)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28197542

RESUMO

An intronic hexanucleotide repeat expansion (HRE) mutation in the C9ORF72 gene is the most common cause of familial ALS and frontotemporal dementia (FTD) and is found in ∼7% of individuals with apparently sporadic disease. Several different diamino acid peptides can be generated from the HRE by noncanonical translation (repeat-associated non-ATG translation, or RAN translation), and some of these peptides can be toxic. Here, we studied the effects of two arginine containing RAN translation products [proline/arginine repeated 20 times (PR20) and glycine/arginine repeated 20 times (GR20)] in primary rat spinal cord neuron cultures grown on an astrocyte feeder layer. We find that PR20 kills motor neurons with an LD50 of 2 µM, but in contrast to the effects of other ALS-causing mutant proteins (i.e., SOD or TDP43), PR20 does not evoke the biochemical signature of mitochondrial dysfunction, ER stress, or mTORC down-regulation. PR20 does result in a time-dependent build-up of ubiquitylated substrates, and this is associated with a reduction of flux through both autophagic and proteasomal degradation pathways. GR20, however, does not have these effects. The effects of PR20 on the proteasome are likely to be direct because (1) PR20 physically associates with proteasomes in biochemical assays, and (2) PR20 inhibits the degradation of a ubiquitylated test substrate when presented to purified proteasomes. Application of a proteasomal activator (IU1) blocks the toxic effects of PR20 on motor neuron survival. This work suggests that proteasomal activators have therapeutic potential in individuals with C9ORF72 HRE.


Assuntos
Peptídeos/farmacologia , Inibidores de Proteassoma/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Células Cultivadas , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Técnicas de Cocultura , Expansão das Repetições de DNA , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia
9.
J Neurosci ; 35(42): 14286-306, 2015 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-26490867

RESUMO

Misfolded proteins accumulate and aggregate in neurodegenerative disease. The existence of these deposits reflects a derangement in the protein homeostasis machinery. Using a candidate gene screen, we report that loss of RAD-23 protects against the toxicity of proteins known to aggregate in amyotrophic lateral sclerosis. Loss of RAD-23 suppresses the locomotor deficit of Caenorhabditis elegans engineered to express mutTDP-43 or mutSOD1 and also protects against aging and proteotoxic insults. Knockdown of RAD-23 is further neuroprotective against the toxicity of SOD1 and TDP-43 expression in mammalian neurons. Biochemical investigation indicates that RAD-23 modifies mutTDP-43 and mutSOD1 abundance, solubility, and turnover in association with altering the ubiquitination status of these substrates. In human amyotrophic lateral sclerosis spinal cord, we find that RAD-23 abundance is increased and RAD-23 is mislocalized within motor neurons. We propose a novel pathophysiological function for RAD-23 in the stabilization of mutated proteins that cause neurodegeneration. SIGNIFICANCE STATEMENT: In this work, we identify RAD-23, a component of the protein homeostasis network and nucleotide excision repair pathway, as a modifier of the toxicity of two disease-causing, misfolding-prone proteins, SOD1 and TDP-43. Reducing the abundance of RAD-23 accelerates the degradation of mutant SOD1 and TDP-43 and reduces the cellular content of the toxic species. The existence of endogenous proteins that act as "anti-chaperones" uncovers new and general targets for therapeutic intervention.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Doença dos Neurônios Motores/genética , Mutação/genética , Interferência de RNA/fisiologia , Animais , Animais Geneticamente Modificados , Animais Recém-Nascidos , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/genética , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Genótipo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Masculino , Camundongos , Atividade Motora/genética , Fotodegradação , Ratos , Ratos Sprague-Dawley
10.
J Neurosci ; 35(24): 9088-105, 2015 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-26085633

RESUMO

Mutant genes that underlie Mendelian forms of amyotrophic lateral sclerosis (ALS) and biochemical investigations of genetic disease models point to potential driver pathophysiological events involving endoplasmic reticulum (ER) stress and autophagy. Several steps in these cell biological processes are known to be controlled physiologically by small ADP-ribosylation factor (ARF) signaling. Here, we investigated the role of ARF guanine nucleotide exchange factors (GEFs), cytohesins, in models of ALS. Genetic or pharmacological inhibition of cytohesins protects motor neurons in vitro from proteotoxic insults and rescues locomotor defects in a Caenorhabditis elegans model of disease. Cytohesins form a complex with mutant superoxide dismutase 1 (SOD1), a known cause of familial ALS, but this is not associated with a change in GEF activity or ARF activation. ER stress evoked by mutant SOD1 expression is alleviated by antagonism of cytohesin activity. In the setting of mutant SOD1 toxicity, inhibition of cytohesin activity enhances autophagic flux and reduces the burden of misfolded SOD1. These observations suggest that targeting cytohesins may have potential benefits for the treatment of ALS.


Assuntos
Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Modelos Animais de Doenças , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/genética , Doença dos Neurônios Motores/genética , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/biossíntese , Células Cultivadas , Proteínas Ativadoras de GTPase/antagonistas & inibidores , Proteínas Ativadoras de GTPase/biossíntese , Proteínas Ativadoras de GTPase/genética , Fatores de Troca do Nucleotídeo Guanina/biossíntese , Células HeLa , Humanos , Camundongos , Modelos Genéticos , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/biossíntese , Superóxido Dismutase/genética
11.
Brain Res ; 1474: 40-9, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22842523

RESUMO

It has been hypothesized that, in the developing rodent hippocampus, mossy fiber terminals release GABA together with glutamate. Here, we used transgenic glutamic acid decarboxylase-67 (GAD67)-GFP expressing mice and multi-label immunohistochemistry to address whether glutamatergic and GABAergic markers are colocalized. We demonstrate that in the dentate gyrus, interneurons positive for GABA/GAD are sparsely distributed along the edge of the hilus, in a different pattern from that of the densely packed granule cells. Co-staining for synaptophysin and vesicular glutamate transporter1 (VGLUT1) in postnatal day 14 brain sections from both mice and rats showed mossy fiber terminals as a group of large (2-5 µm in diameter) VGLUT1-positive excitatory presynaptic terminals in the stratum lucidum of area CA3a/b. Furthermore, co-staining for synaptophysin and vesicular GABA transporter (VGAT) revealed a group of small-sized (∼0.5 µm in diameter) inhibitory presynaptic terminals in the same area where identified mossy fiber terminals were present. The two types of terminals appeared to be mutually exclusive, and showed no colocalization. Thus, our results do not support the hypothesis that GABA is released as a neurotransmitter from mossy fiber terminals during development.


Assuntos
Ácido Glutâmico/biossíntese , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Fibras Musgosas Hipocampais/crescimento & desenvolvimento , Fibras Musgosas Hipocampais/metabolismo , Ácido gama-Aminobutírico/biossíntese , Animais , Western Blotting , Imunofluorescência , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Microscopia Confocal , Ratos
12.
Hum Mol Genet ; 21(13): 2899-911, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22454397

RESUMO

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease affecting motor neurons. Mutations in related RNA-binding proteins TDP-43, FUS/TLS and TAF15 have been connected to ALS. These three proteins share several features, including the presence of a bioinformatics-predicted prion domain, aggregation-prone nature in vitro and in vivo and toxic effects when expressed in multiple model systems. Given these commonalities, we hypothesized that a related protein, EWSR1 (Ewing sarcoma breakpoint region 1), might also exhibit similar properties and therefore could contribute to disease. Here, we report an analysis of EWSR1 in multiple functional assays, including mutational screening in ALS patients and controls. We identified three missense variants in EWSR1 in ALS patients, which were absent in a large number of healthy control individuals. We show that disease-specific variants affect EWSR1 localization in motor neurons. We also provide multiple independent lines of in vitro and in vivo evidence that EWSR1 has similar properties as TDP-43, FUS and TAF15, including aggregation-prone behavior in vitro and ability to confer neurodegeneration in Drosophila. Postmortem analysis of sporadic ALS cases also revealed cytoplasmic mislocalization of EWSR1. Together, our studies highlight a potential role for EWSR1 in ALS, provide a collection of functional assays to be used to assess roles of additional RNA-binding proteins in disease and support an emerging concept that a class of aggregation-prone RNA-binding proteins might contribute broadly to ALS and related neurodegenerative diseases.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a Calmodulina/genética , Neurônios Motores/patologia , Proteínas de Ligação a RNA/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Animais , Animais Geneticamente Modificados , Proteínas de Ligação a Calmodulina/metabolismo , Células Cultivadas , Criança , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Drosophila/genética , Feminino , Genes Reguladores , Variação Genética , Genótipo , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios Motores/metabolismo , Mutação de Sentido Incorreto , Proteína EWS de Ligação a RNA , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Alinhamento de Sequência , Fatores Associados à Proteína de Ligação a TATA/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Adulto Jovem
13.
Proc Natl Acad Sci U S A ; 108(52): 20881-90, 2011 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-22065782

RESUMO

Amyotrophic lateral sclerosis (ALS) is a devastating and universally fatal neurodegenerative disease. Mutations in two related RNA-binding proteins, TDP-43 and FUS, that harbor prion-like domains, cause some forms of ALS. There are at least 213 human proteins harboring RNA recognition motifs, including FUS and TDP-43, raising the possibility that additional RNA-binding proteins might contribute to ALS pathogenesis. We performed a systematic survey of these proteins to find additional candidates similar to TDP-43 and FUS, followed by bioinformatics to predict prion-like domains in a subset of them. We sequenced one of these genes, TAF15, in patients with ALS and identified missense variants, which were absent in a large number of healthy controls. These disease-associated variants of TAF15 caused formation of cytoplasmic foci when expressed in primary cultures of spinal cord neurons. Very similar to TDP-43 and FUS, TAF15 aggregated in vitro and conferred neurodegeneration in Drosophila, with the ALS-linked variants having a more severe effect than wild type. Immunohistochemistry of postmortem spinal cord tissue revealed mislocalization of TAF15 in motor neurons of patients with ALS. We propose that aggregation-prone RNA-binding proteins might contribute very broadly to ALS pathogenesis and the genes identified in our yeast functional screen, coupled with prion-like domain prediction analysis, now provide a powerful resource to facilitate ALS disease gene discovery.


Assuntos
Esclerose Lateral Amiotrófica/genética , Neurônios Motores/metabolismo , Estrutura Terciária de Proteína , Proteínas de Ligação a RNA/genética , Medula Espinal/citologia , Fatores Associados à Proteína de Ligação a TATA/genética , Animais , Células Cultivadas , Biologia Computacional , Drosophila melanogaster/genética , Estudos de Associação Genética/métodos , Humanos , Imuno-Histoquímica , Mutação de Sentido Incorreto/genética , Saccharomyces cerevisiae/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo
14.
J Neurosci ; 31(1): 295-9, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21209215

RESUMO

The survival of dorsal root ganglion and sympathetic neurons is promoted whether nerve growth factor (NGF) activates TrkA receptors on the cell body or the axon. Yet other aspects of neurotrophic factor actions (i.e., ability to promote axon growth, selection of neurochemical phenotype and engagement of signaling modules) differ as a function of the location of the ligand-receptor interaction. The extent to which these observations are relevant to CNS neurons is unknown. This may be particularly relevant to neurodegenerative diseases such as amyotrophic lateral sclerosis, where beneficial axon-target interactions are disturbed early in the disease process. Here we characterize the growth of pure motor neurons in compartment cultures and show that brain-derived neurotrophic factor (BDNF) stimulation of the cell body or axons/dendrites promotes survival. Expression of G37R mutant superoxide dismutase (SOD) in motor neurons will lead to death and this depends on BDNF activation of TrkB on axons and/or dendrites. BDNF action depends upon endocytosis of the BDNF-TrkB complex and de novo protein synthesis. These results highlight the importance of signaling events occurring in axons/dendrites in mutant SOD toxicity.


Assuntos
Axônios/fisiologia , Dendritos/fisiologia , Neurônios Motores/citologia , Mutação/genética , Transdução de Sinais/genética , Superóxido Dismutase/genética , Análise de Variância , Animais , Arginina/genética , Axônios/efeitos dos fármacos , Biotinilação , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Sobrevivência Celular/genética , Células Cultivadas , Dendritos/efeitos dos fármacos , Embrião de Mamíferos , Endocitose/efeitos dos fármacos , Endocitose/genética , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Glicina/genética , Imunoprecipitação/métodos , Neurônios Motores/efeitos dos fármacos , Ratos , Receptor trkB/genética , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Simplexvirus/genética , Simplexvirus/metabolismo , Medula Espinal/citologia , Transfecção/métodos
15.
J Neurosci ; 29(25): 8236-47, 2009 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-19553463

RESUMO

Aging is a risk factor for the development of adult-onset neurodegenerative diseases. Although some of the molecular pathways regulating longevity and stress resistance in lower organisms are defined (i.e., those activating the transcriptional regulators DAF-16 and HSF-1 in Caenorhabditis elegans), their relevance to mammals and disease susceptibility are unknown. We studied the signaling controlled by the mammalian homolog of DAF-16, FOXO3a, in model systems of motor neuron disease. Neuron death elicited in vitro by excitotoxic insult or the expression of mutant SOD1, mutant p150(glued), or polyQ-expanded androgen receptor was abrogated by expression of nuclear-targeted FOXO3a. We identify a compound [Psammaplysene A (PA)] that increases nuclear localization of FOXO3a in vitro and in vivo and show that PA also protects against these insults in vitro. Administration of PA to invertebrate model systems of neurodegeneration similarly blocked neuron death in a DAF-16/FOXO3a-dependent manner. These results indicate that activation of the DAF-16/FOXO3a pathway, genetically or pharmacologically, confers protection against the known causes of motor neuron diseases.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Doença dos Neurônios Motores/tratamento farmacológico , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais , Tirosina/análogos & derivados , Animais , Western Blotting , Contagem de Células/métodos , Técnicas de Cultura de Células , Morte Celular/efeitos dos fármacos , Biologia Computacional , Modelos Animais de Doenças , Drosophila , Embrião de Mamíferos , Agonistas de Aminoácidos Excitatórios/toxicidade , Feminino , Fluorescência , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/biossíntese , Imuno-Histoquímica , Ácido Caínico/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Doença dos Neurônios Motores/metabolismo , Doença dos Neurônios Motores/patologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/citologia , Tirosina/administração & dosagem , Tirosina/farmacologia
16.
J Neurosci ; 28(41): 10220-33, 2008 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-18842882

RESUMO

Activity-dependent dendrite elaboration influences the pattern of interneuronal connectivity and network function. In the present study, we examined the mechanism by which the GluR1 subunit of AMPA receptors controls dendrite morphogenesis. GluR1 binds to SAP97, a scaffolding protein that is a component of the postsynaptic density, via its C-terminal 7 aa. We find that elimination of this interaction in vitro or in vivo (by deleting the C-terminal 7 aa of GluR1, GluR1Delta7) does not influence trafficking, processing, or cell surface GluR1 expression but does prevent translocation of SAP97 from the cytosol to membranes. GluR1 and SAP97 together at the plasma membrane promotes dendrite branching in an activity-dependent manner, although this does not require physical association. Our findings suggest that the C-terminal 7 aa of GluR1 are essential for bringing SAP97 to the plasma membrane, where it acts to translate the activity of AMPA receptors into dendrite growth.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Dendritos/fisiologia , Proteínas de Membrana/metabolismo , Receptores de AMPA/metabolismo , Medula Espinal/fisiologia , Animais , Membrana Celular/metabolismo , Células Cultivadas , Quimera , Dendritos/metabolismo , Proteína 1 Homóloga a Discs-Large , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neurônios Motores/metabolismo , Transporte Proteico/fisiologia , Medula Espinal/ultraestrutura
17.
J Neurosci ; 28(40): 9953-68, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18829953

RESUMO

Activity-dependent specification of neuronal architecture during early postnatal life is essential for refining the precision of communication between neurons. In the spinal cord under normal circumstances, the AMPA receptor subunit GluR1 is expressed at high levels by motor neurons and surrounding interneurons during this critical developmental period, although the role it plays in circuit formation and locomotor behavior is unknown. Here, we show that GluR1 promotes dendrite growth in a non-cell-autonomous manner in vitro and in vivo. The mal-development of motor neuron dendrites is associated with changes in the pattern of interneuronal connectivity within the segmental spinal cord and defects in strength and endurance. Transgenic expression of GluR1 in adult motor neurons leads to dendrite remodeling and supernormal locomotor function. GluR1 expression by neurons within the segmental spinal cord plays an essential role in formation of the neural network that underlies normal motor behavior.


Assuntos
Neurônios Motores/fisiologia , Receptores de AMPA/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Atividade Motora/fisiologia , Neurônios Motores/citologia , Rede Nervosa/citologia , Rede Nervosa/crescimento & desenvolvimento , Ratos , Ratos Sprague-Dawley , Receptores de AMPA/biossíntese , Receptores de AMPA/genética , Medula Espinal/citologia , Medula Espinal/crescimento & desenvolvimento , Xenopus laevis
18.
J Neuroinflammation ; 4: 12, 2007 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-17474992

RESUMO

BACKGROUND: Neuroinflammation has been implicated in various brain pathologies characterized by hypoxia and ischemia. Astroglia play an important role in the initiation and propagation of hypoxia/ischemia-induced inflammation by secreting inflammatory chemokines that attract neutrophils and monocytes into the brain. However, triggers of chemokine up-regulation by hypoxia/ischemia in these cells are poorly understood. Hypoxia-inducible factor-1 (HIF-1) is a dimeric transcriptional factor consisting of HIF-1alpha and HIF-1beta subunits. HIF-1 binds to HIF-1-binding sites in the target genes and activates their transcription. We have recently shown that hypoxia-induced expression of IL-1beta in astrocytes is mediated by HIF-1alpha. In this study, we demonstrate the role of HIF-1alpha in hypoxia-induced up-regulation of inflammatory chemokines, human monocyte chemoattractant protein-1 (MCP-1/CCL2) and mouse MCP-5 (Ccl12), in human and mouse astrocytes, respectively. METHODS: Primary fetal human astrocytes or mouse astrocytes generated from HIF-1alpha+/+ and HIF-1alpha+/- mice were subjected to hypoxia (<2% oxygen) or 125 muM CoCl2 for 4 h and 6 h, respectively. The expression of HIF-1alpha, MCP-1 and MCP-5 was determined by semi-quantitative RT-PCR, western blot or ELISA. The interaction of HIF-1alpha with a HIF-1-binding DNA sequence was examined by EMSA and supershift assay. HIF-1-binding sequence in the promoter of MCP-1 gene was cloned and transcriptional activation of MCP-1 by HIF-1alpha was analyzed by reporter gene assay. RESULTS: Sequence analyses identified HIF-1-binding sites in the promoters of MCP-1 and MCP-5 genes. Both hypoxia and HIF-1alpha inducer, CoCl2, strongly up-regulated HIF-1alpha expression in astrocytes. Mouse HIF-1alpha+/- astrocytes had lower basal levels of HIF-1alpha and MCP-5 expression. The up-regulation of MCP-5 by hypoxia or CoCl2 in HIF-1alpha+/+ and HIF-1alpha+/- astrocytes was correlated with the levels of HIF-1alpha in cells. Both hypoxia and CoCl2 also up-regulated HIF-1alpha and MCP-1 expression in human astrocytes. EMSA assay demonstrated that HIF-1 activated by either hypoxia or CoCl2 binds to wild-type HIF-1-binding DNA sequence, but not the mutant sequence. Furthermore, reporter gene assay demonstrated that hypoxia markedly activated MCP-1 transcription but not the mutated MCP-1 promoter in transfected astrocytes. CONCLUSION: These findings suggest that both MCP-1 and MCP-5 are HIF-1 target genes and that HIF-1alpha is involved in transcriptional induction of these two chemokines in astrocytes by hypoxia.


Assuntos
Astrócitos/metabolismo , Hipóxia Celular/fisiologia , Quimiocina CCL2/biossíntese , Regulação da Expressão Gênica/fisiologia , Fator 1 Induzível por Hipóxia/fisiologia , Proteínas Quimioatraentes de Monócitos/biossíntese , Animais , Sítios de Ligação/genética , Hipóxia Celular/genética , Células Cultivadas , Quimiocina CCL2/genética , Humanos , Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Transgênicos , Proteínas Quimioatraentes de Monócitos/genética
19.
Eur J Neurosci ; 25(7): 1987-97, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17439487

RESUMO

We have examined the participation of a neuronal nitric oxide synthase (nNOS) signaling pathway in the elaboration of motor neuron dendrites during embryonic life. During chick embryogenesis, nNOS is expressed by interneurons that surround the motor neuron pools in the ventral horn. Pseudorabies virus tracing suggests that these cells, while juxtaposed to motor neurons are not synaptically connected to them. The downstream effectors, soluble guanylyl cyclase (sGC) and protein kinase G (PKG), are found in motor neurons as well as several other populations of spinal cord cells. To determine the functional significance of the nNOS/sGC/PKG signaling pathway, pharmacological inhibitors were applied to chick embryos and the effects on motor neuron dendrites monitored. Inhibition of nNOS activity led to a lasting reduction in the overall size and degree of branching of the dendritic tree. These alterations in dendritic architecture were also seen when the activity of sGC or PKG was blocked. Our results suggest that normal motor neuron dendrite elaboration depends, in part, on the activity-dependent generation of NO by ventral horn interneurons, which then activates sGC and PKG in motor neurons.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Dendritos/fisiologia , Embrião de Mamíferos/citologia , Guanilato Ciclase/metabolismo , Neurônios Motores , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animais , Células Cultivadas , Embrião de Galinha , Proteínas Quinases Dependentes de GMP Cíclico/antagonistas & inibidores , Dendritos/ultraestrutura , Embrião de Mamíferos/fisiologia , Endotelina-1/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Guanilato Ciclase/antagonistas & inibidores , Imuno-Histoquímica , Interneurônios/citologia , Interneurônios/metabolismo , Neurônios Motores/citologia , Neurônios Motores/fisiologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Medula Espinal/citologia
20.
J Neurosci ; 26(36): 9250-63, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16957081

RESUMO

The death of motor neurons in amyotrophic lateral sclerosis (ALS) is thought to result from the interaction of a variety of factors including excitotoxicity, accumulation of toxic proteins, and abnormal axonal transport. Previously, we found that the susceptibility of motor neurons to excitotoxic insults can be limited by inhibiting signals evoked by brain-derived neurotrophic factor (BDNF) activation of the receptor tyrosine kinase B (TrkB). Here we show that this can be achieved by direct kinase inhibition or by blockade of a transactivation pathway that uses adenosine A2a receptors and src-family kinases (SFKs). Downstream signaling cascades (such as mitogen-activated protein kinase and phosphatidylinositol-3 kinase) are inhibited by these blockers. In addition to protecting motor neurons from excitotoxic insult, these agents also prevent toxicity that follows from the expression of mutant proteins (G85R superoxide dismutase 1; G59S p150(glued)) that cause familial motor neuron disease. TrkB, adenosine A2a receptors, and SFKs associate into complexes in lipid raft and nonlipid raft membranes and the signaling from lipids rafts may be particularly important because their disruption by cholesterol depletion blocks the ability of BDNF to render motor neurons vulnerable to insult. The neuroprotective versatility of Trk antagonism suggests that it may have broad utility in the treatment of ALS patients.


Assuntos
Antagonistas do Receptor A2 de Adenosina , Esclerose Lateral Amiotrófica/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Esclerose Lateral Amiotrófica/induzido quimicamente , Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/prevenção & controle , Animais , Células Cultivadas , Ácido Caínico , Neurônios Motores/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/metabolismo , Receptor A2A de Adenosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA