Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Rep ; 42(9): 113068, 2023 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-37656624

RESUMO

Primary somatosensory axons stop regenerating as they re-enter the spinal cord, resulting in incurable sensory loss. What arrests them has remained unclear. We previously showed that axons stop by forming synaptic contacts with unknown non-neuronal cells. Here, we identified these cells in adult mice as oligodendrocyte precursor cells (OPCs). We also found that only a few axons stop regenerating by forming dystrophic endings, exclusively at the CNS:peripheral nervous system (PNS) borderline where OPCs are absent. Most axons stop in contact with a dense network of OPC processes. Live imaging, immuno-electron microscopy (immuno-EM), and OPC-dorsal root ganglia (DRG) co-culture additionally suggest that axons are rapidly immobilized by forming synapses with OPCs. Genetic OPC ablation enables many axons to continue regenerating deep into the spinal cord. We propose that sensory axons stop regenerating by encountering OPCs that induce presynaptic differentiation. Our findings identify OPCs as a major regenerative barrier that prevents intraspinal restoration of sensory circuits following spinal root injury.


Assuntos
Células Precursoras de Oligodendrócitos , Camundongos , Animais , Medula Espinal/fisiologia , Axônios/fisiologia , Raízes Nervosas Espinhais , Gânglios Espinais/fisiologia , Regeneração Nervosa/fisiologia
2.
Elife ; 102021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33942723

RESUMO

A major barrier to intraspinal regeneration after dorsal root (DR) injury is the DR entry zone (DREZ), the CNS/PNS interface. DR axons stop regenerating at the DREZ, even if regenerative capacity is increased by a nerve conditioning lesion. This potent blockade has long been attributed to myelin-associated inhibitors and (CSPGs), but incomplete lesions and conflicting reports have prevented conclusive agreement. Here, we evaluated DR regeneration in mice using novel strategies to facilitate complete lesions and analyses, selective tracing of proprioceptive and mechanoreceptive axons, and the first simultaneous targeting of Nogo/Reticulon-4, MAG, OMgp, CSPGs, and GDNF. Co-eliminating myelin inhibitors and CSPGs elicited regeneration of only a few conditioning-lesioned DR axons across the DREZ. Their absence, however, markedly and synergistically enhanced regeneration of GDNF-stimulated axons, highlighting the importance of sufficiently elevating intrinsic growth capacity. We also conclude that myelin inhibitors and CSPGs are not the primary mechanism stopping axons at the DREZ.


Assuntos
Axônios/fisiologia , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Bainha de Mielina/metabolismo , Medula Espinal/citologia , Raízes Nervosas Espinhais/patologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
3.
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
4.
Elife ; 62017 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-28124973

RESUMO

Nuclear exclusion of the transcriptional regulators and potent oncoproteins, YAP/TAZ, is considered necessary for adult tissue homeostasis. Here we show that nuclear YAP/TAZ are essential regulators of peripheral nerve development and myelin maintenance. To proliferate, developing Schwann cells (SCs) require YAP/TAZ to enter S-phase and, without them, fail to generate sufficient SCs for timely axon sorting. To differentiate, SCs require YAP/TAZ to upregulate Krox20 and, without them, completely fail to myelinate, resulting in severe peripheral neuropathy. Remarkably, in adulthood, nuclear YAP/TAZ are selectively expressed by myelinating SCs, and conditional ablation results in severe peripheral demyelination and mouse death. YAP/TAZ regulate both developmental and adult myelination by driving TEAD1 to activate Krox20. Therefore, YAP/TAZ are crucial for SCs to myelinate developing nerve and to maintain myelinated nerve in adulthood. Our study also provides a new insight into the role of nuclear YAP/TAZ in homeostatic maintenance of an adult tissue.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Bainha de Mielina/metabolismo , Fosfoproteínas/metabolismo , Células de Schwann/fisiologia , Fatores de Transcrição/metabolismo , Aciltransferases , Animais , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Camundongos , Proteínas de Sinalização YAP
5.
Exp Neurol ; 283(Pt A): 73-84, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27264357

RESUMO

Although previous studies have identified several strategies to stimulate regeneration of CNS axons, extensive regeneration and functional recovery have remained a major challenge, particularly for large diameter myelinated axons. Within the CNS, myelin is thought to inhibit axon regeneration, while modulating activity of the mTOR pathway promotes regeneration of injured axons. In this study, we examined NT-3 mediated regeneration of sensory axons through the dorsal root entry zone in a triple knockout of myelin inhibitory proteins or after activation of mTOR using a constitutively active (ca) Rheb in DRG neurons to determine the influence of environmental inhibitory or activation of intrinsic growth pathways could enhance NT-3-mediate regeneration. Loss of myelin inhibitory proteins showed modest enhancement of sensory axon regeneration. In mTOR studies, we found a dramatic age related decrease in the mTOR activation as determined by phosphorylation of the downstream marker S6 ribosomal subunit. Expression of caRheb within adult DRG neurons in vitro increased S6 phosphorylation and doubled the overall length of neurite outgrowth, which was reversed in the presence of rapamycin. In adult female rats, combined expression of caRheb in DRG neurons and NT-3 within the spinal cord increased regeneration of sensory axons almost 3 fold when compared to NT-3 alone. Proprioceptive assessment using a grid runway indicates functionally significant regeneration of large-diameter myelinated sensory afferents. Our results indicate that caRheb-induced increase in mTOR activation enhances neurotrophin-3 induced regeneration of large-diameter myelinated axons.


Assuntos
Regulação da Expressão Gênica/fisiologia , Regeneração Nervosa/fisiologia , Neurotrofina 3/metabolismo , Transdução de Sinais/fisiologia , Distúrbios Somatossensoriais/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Gânglios Espinais/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hiperalgesia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Glicoproteína Associada a Mielina/deficiência , Glicoproteína Associada a Mielina/genética , Neurotrofina 3/genética , Neurotrofina 3/uso terapêutico , Proteínas Nogo/deficiência , Proteínas Nogo/genética , Ratos , Ratos Sprague-Dawley , Sirolimo/farmacologia , Distúrbios Somatossensoriais/patologia , Distúrbios Somatossensoriais/fisiopatologia , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/terapia
6.
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
7.
Hum Mol Genet ; 20(21): 4116-31, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21816949

RESUMO

Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase B (TrkB) are widely expressed in the vertebrate nervous system and play a central role in mature neuronal function. In vitro BDNF/TrkB signaling promotes neuronal survival and can help neurons resist toxic insults. Paradoxically, BDNF/TrkB signaling has also been shown, under certain in vitro circumstances, to render neurons vulnerable to insults. We show here that in vivo conditional deletion of TrkB from mature motor neurons attenuates mutant superoxide dismutase 1 (SOD1) toxicity. Mutant SOD1 mice lacking motor neuron TrkB live a month longer than controls and retain motor function for a longer period, particularly in the early phase of the disease. These effects are subserved by slowed motor neuron loss, persistence of neuromuscular junction integrity and reduced astrocytic and microglial reactivity within the spinal cord. These results suggest that manipulation of BDNF/TrkB signaling might have therapeutic efficacy in motor neuron diseases.


Assuntos
Doença dos Neurônios Motores/enzimologia , Doença dos Neurônios Motores/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Mutação/genética , Receptor trkB/metabolismo , Superóxido Dismutase/genética , Substituição de Aminoácidos , Animais , Axônios/metabolismo , Axônios/patologia , Denervação , Progressão da Doença , Cistos Glanglionares/metabolismo , Cistos Glanglionares/patologia , Deleção de Genes , Corpos de Inclusão/metabolismo , Inflamação/complicações , Inflamação/patologia , Inflamação/fisiopatologia , Integrases/metabolismo , Interneurônios/metabolismo , Interneurônios/patologia , Longevidade , Camundongos , Camundongos Knockout , Atividade Motora , Doença dos Neurônios Motores/complicações , Doença dos Neurônios Motores/fisiopatologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/patologia , Recombinação Genética/genética , Medula Espinal/metabolismo , Medula Espinal/patologia , Superóxido Dismutase-1 , Ubiquitina/metabolismo , Ubiquitinação , Proteínas Vesiculares de Transporte de Acetilcolina/metabolismo
8.
J Cell Biol ; 180(1): 187-203, 2008 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-18195107

RESUMO

Integrin binding to matrix proteins such as fibronectin (FN) leads to formation of focal adhesion (FA) cellular contact sites that regulate migration. RhoA GTPases facilitate FA formation, yet FA-associated RhoA-specific guanine nucleotide exchange factors (GEFs) remain unknown. Here, we show that proline-rich kinase-2 (Pyk2) levels increase upon loss of focal adhesion kinase (FAK) in mouse embryonic fibroblasts (MEFs). Additionally, we demonstrate that Pyk2 facilitates deregulated RhoA activation, elevated FA formation, and enhanced cell proliferation by promoting p190RhoGEF expression. In normal MEFs, p190RhoGEF knockdown inhibits FN-associated RhoA activation, FA formation, and cell migration. Knockdown of p190RhoGEF-related GEFH1 does not affect FA formation in FAK(-/-) or normal MEFs. p190RhoGEF overexpression enhances RhoA activation and FA formation in MEFs dependent on FAK binding and associated with p190RhoGEF FA recruitment and tyrosine phosphorylation. These studies elucidate a compensatory function for Pyk2 upon FAK loss and identify the FAK-p190RhoGEF complex as an important integrin-proximal regulator of FA formation during FN-stimulated cell motility.


Assuntos
Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Adesões Focais/fisiologia , ras-GRF1/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Quinase 1 de Adesão Focal/genética , Quinase 2 de Adesão Focal/genética , Regulação da Expressão Gênica , Camundongos , Paxilina/metabolismo , Fosforilação , Tirosina/metabolismo , ras-GRF1/genética
9.
Hum Mol Genet ; 16(24): 3103-16, 2007 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-17881652

RESUMO

Mutations in neurofilament light (NFL) subunit and small heat-shock protein B1 (HSPB1) cause autosomal-dominant axonal Charcot-Marie-Tooth disease type 2E (CMT2E) and type 2F (CMT2F). Previous studies have shown that CMT mutations in NFL and HSPB1 disrupt NF assembly and cause aggregation of NFL protein. In this study, we investigate the role of aggregation of NFL protein in the neurotoxicity of CMT mutant NFL and CMT mutant HSPB1 in motor neurons. We find that expression of CMT mutant NFL leads to progressive degeneration and loss of neuronal viability of cultured motor neurons. Degenerating motor neurons show fragmentation and loss of neuritic processes associated with disruption of NF network and aggregation of NFL protein. Co-expression of wild-type HSPB1 diminishes aggregation of CMT mutant NFL, induces reversal of CMT mutant NFL aggregates and reduces CMT mutant NFL-induced loss of motor neuron viability. Like CMT mutant NFL, expression of S135F CMT mutant HSPB1 also leads to progressive degeneration of motor neurons with disruption of NF network and aggregation of NFL protein. Further studies show that wild-type and S135F mutant HSPB1 associate with wild-type and CMT mutant NFL and that S135F mutant HSPB1 has dominant effect on disruption of NF assembly and aggregation of NFL protein. Finally, we show that deletion of NFL markedly reduces degeneration and loss of motor neuron viability induced by S135F mutant HSPB1. Together, our data support the view that disruption of NF network with aggregation of NFL is a common triggering event of motor neuron degeneration in CMT2E and CMT2F disease.


Assuntos
Doença de Charcot-Marie-Tooth/genética , Proteínas de Choque Térmico/genética , Neurônios Motores/patologia , Mutação , Proteínas de Neoplasias/genética , Degeneração Neural/patologia , Rede Nervosa/química , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Doença de Charcot-Marie-Tooth/patologia , Embrião de Mamíferos , Proteínas de Choque Térmico HSP27 , Corpos de Inclusão/metabolismo , Corpos de Inclusão/patologia , Camundongos , Camundongos Knockout , Chaperonas Moleculares , Proteínas Mutantes/fisiologia , Mutação/fisiologia , Degeneração Neural/genética , Degeneração Neural/metabolismo , Rede Nervosa/metabolismo , Proteínas de Neurofilamentos/química , Transdução de Sinais/fisiologia
10.
Hum Mol Genet ; 14(23): 3643-59, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16236762

RESUMO

Abnormal protein aggregation is emerging as a common theme in the pathogenesis of neurodegenerative disease. Our previous studies have shown that overexpression of untranslated light neurofilament (NF-L) RNA causes motor neuron degeneration in transgenic mice, leads to accumulation of ubiquitinated aggregates in degenerating cultured motor neurons and triggers aggregation of NF-L protein and co-aggregation of mutant SOD1 protein in neuronal cells. Here, we report that p190RhoGEF, an RNA-binding protein that binds to a destabilizing element in NF-L mRNA, is involved in aggregation of NF-L protein and is implicated in the pathogenesis of motor neuron degeneration. We show that p190RhoGEF co-aggregates with unassembled NF-L protein and that co-aggregation is associated with down-regulation of parent NF-L mRNA in neuronal cells. Co-expression of NF-M increases NF assembly and reduces RNA-triggered aggregation as well as loss of solubility of NF-L protein. siRNA-induced down-regulation of p190RhoGEF not only reduces aggregation and promotes assembly of NF-L and NF-M, but also causes reversal of aggregation and recovery of NF assembly in transfected cells. Examination of transgenic models of motor neuron disease shows that prominent aggregates of p190RhoGEF and NF-L and down-regulation of NF-L expression occur in degenerating motor neurons of mice expressing untranslated NF-L RNA or a G93A mutant SOD1 transgene. Moreover, aggregates of p190RhoGEF and NF-L appear as early pathological changes in presymptomatic G93A mutant SOD1 transgenic mice. Together, the findings indicate that p190RhoGEF is involved in aggregation of NF-L protein and support a working hypothesis that aggregation of p190RhoGEF and NF-L is an upstream event triggering neurotoxicity in motor neuron disease.


Assuntos
Proteínas de Transporte/metabolismo , Doença dos Neurônios Motores/metabolismo , Degeneração Neural/metabolismo , Proteínas de Neurofilamentos/metabolismo , Estabilidade de RNA , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas de Transporte/análise , Proteínas de Transporte/genética , Proteínas de Ligação a DNA , Proteínas Ativadoras de GTPase , Camundongos , Camundongos Transgênicos , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/patologia , Neurônios Motores/química , Neurônios Motores/metabolismo , Degeneração Neural/genética , Proteínas de Neurofilamentos/análise , Proteínas de Neurofilamentos/genética , Interferência de RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/análise , Proteínas de Ligação a RNA/genética , Proteínas Repressoras , Solubilidade
11.
Hum Mol Genet ; 14(18): 2629-40, 2005 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-16079151

RESUMO

Mutations in Cu/Zn superoxide dismutase (SOD1) cause approximately 20% of familial amyotrophic lateral sclerosis by a toxic gain of function; however, the precise mechanisms remain unclear. Here, we report the identification of HoxB2, a homeodomain-containing transcription factor, as a G93A mutant SOD1 interactive protein in a yeast two-hybrid screen. We show that HoxB2 co-precipitates and co-localizes with mutant SOD1 in neuronal cell lines, as well as in brain and spinal cord of G93A mutant SOD1 transgenic mice. Mutagenesis further shows that this interaction is mediated by the central homeodomain of HoxB2. In motor neuron-like NSC-34 cells, overexpression of HoxB2 or its homeodomain decreases the insolubility of mutant SOD1 and inhibits G93A or G86R mutant SOD1-induced neuronal cell death. In human and mouse tissues, we show that expression of HoxB2 persists in adult spinal cord and is primarily localized in nuclei of motor neurons. In G93A transgenic mice, HoxB2 co-localizes with mutant SOD1 and is redistributed to perikarya and proximal neurites of motor neurons. In addition, there is progressive accumulation of HoxB2 and mutant SOD1 as punctate inclusions in the neuropil surrounding motor neurons. Taken together, our findings demonstrate that interaction of HoxB2 with mutant SOD1 occurs in motor neurons of G93A mutant SOD1 transgenic mice and suggest that this interaction may modulate the neurotoxicity of mutant SOD1.


Assuntos
Esclerose Lateral Amiotrófica/genética , Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Superóxido Dismutase/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/genética , Western Blotting , Linhagem Celular , Imunofluorescência , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Imunoprecipitação , Camundongos , Camundongos Transgênicos , Neurônios Motores/metabolismo , Mutagênese Sítio-Dirigida , Mutação/genética , Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Fatores de Transcrição/genética , Técnicas do Sistema de Duplo-Híbrido
12.
J Neurosci ; 25(17): 4353-64, 2005 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15858061

RESUMO

A 68 nucleotide segment of the light neurofilament (NF-L) mRNA, spanning the translation termination signal, participates in regulating the stability of the transcript in vivo. Aldolases A and C, but not B, interact specifically with this segment of the transcript in vitro. Aldolases A and C are glycolytic enzymes expressed in neural cells, and their mRNA binding activity represents a novel function of these isozymes. This unsuspected new activity was first uncovered by Northwestern blotting of a brainstem/spinal cord cDNA library. It was confirmed by two-dimensional fractionation of mouse brain cytosol followed by Northwestern hybridization and protein sequencing. Both neuronal aldolases interact specifically with the NF-L but not the heavy neurofilament mRNA, and their binding to the transcript excludes the poly(A)-binding protein (PABP) from the complex. Constitutive ectopic expression of aldolases A and C accelerates the decay of a neurofilament transgene (NF-L) driven by a tetracycline inducible system. In contrast, mutant transgenes lacking mRNA sequence for aldolase binding are stabilized. Our findings strongly suggest that aldolases A and C are regulatory components of a light neurofilament mRNA complex that modulates the stability of NF-L mRNA. This modulation likely involves endonucleolytic cleavage and a competing interaction with the PABP. Interactions of aldolases A and C in NF-L expression may be linked to regulatory pathways that maintain the highly asymmetrical form and function of large neurons.


Assuntos
Frutose-Bifosfato Aldolase/fisiologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Neurofilamentos/metabolismo , Animais , Northern Blotting/métodos , Western Blotting/métodos , Encéfalo/metabolismo , Linhagem Celular , Chlorocebus aethiops , Cromatografia Líquida de Alta Pressão/métodos , Clonagem Molecular/métodos , Eletroforese em Gel Bidimensional/métodos , Ensaio de Desvio de Mobilidade Eletroforética/métodos , Expressão Gênica/fisiologia , Biblioteca Gênica , Humanos , Imunoprecipitação/métodos , Camundongos , Dados de Sequência Molecular , Peso Molecular , Proteínas de Neurofilamentos/genética , Mapeamento de Peptídeos/métodos , Biossíntese de Proteínas/fisiologia , RNA Mensageiro/metabolismo , Proteínas Recombinantes de Fusão , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Transfecção/métodos
13.
J Neurosci ; 24(11): 2716-26, 2004 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-15028764

RESUMO

The pathogenesis of neurodegenerative diseases is believed to involve abnormal aggregation of proteins, but the mechanisms initiating protein aggregation are unclear. Here we report a novel phenomenon that could be instrumental in triggering protein aggregation in neurodegenerative diseases. We show that the 3' untranslated region (3'UTR) of a light neurofilament (NF-L) transcript enhances the reactivity of its own translated product and leads to loss of solubility and aggregation of NF-L protein and to coaggregation of mutant superoxide dismutase 1 (SOD1) protein. Full-length mouse NF-L cDNAs, with and without NF-L 3'UTR, were fused to the C terminus of a green fluorescent protein (GFP) reporter gene, and the GFP-tagged NF-L proteins were examined in transfected Neuro2a cells. The GFP-tagged NF-L protein expressed from the transgene containing NF-L 3'UTR, but not from the transgene lacking NF-L 3'UTR, colocalizes with endogenous heavy neurofilament protein and, at high-level expression, leads to loss of solubility and aggregation of GFP-tagged NF-L protein. Aggregation of GFP-tagged NF-L protein triggers coaggregation and loss of solubility of coexpressed DsRed-tagged mutant (G93A) SOD1 protein but not wild-type SOD1 protein. Deletional mutagenesis maps the RNA sequence causing aggregation of GFP-tagged NF-L protein to the proximal 45 nucleotides of NF-L 3'UTR. This is the site of a major destabilizing element in NF-L RNA and binding site for RNA-binding proteins. Our findings support a working model whereby NF-L RNA, or cognate RNA-binding factors, enhances the reactivity of NF-L protein and provides a triggering mechanism leading to aggregation of NF-L and other proteins in neurodegenerative diseases.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Proteínas de Neurofilamentos/genética , Proteínas de Neurofilamentos/metabolismo , Neurônios/metabolismo , RNA Mensageiro/fisiologia , Superóxido Dismutase/metabolismo , Regiões 3' não Traduzidas/farmacologia , Animais , Linhagem Celular , Genes Reporter/genética , Proteínas de Fluorescência Verde , Proteínas de Choque Térmico/metabolismo , Proteínas Luminescentes/genética , Substâncias Macromoleculares , Camundongos , Neuroblastoma/metabolismo , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/metabolismo , Neurônios/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/genética , Ligação Proteica/fisiologia , RNA Mensageiro/metabolismo , RNA Mensageiro/farmacologia , Receptores Androgênicos/genética , Receptores Androgênicos/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Solubilidade , Superóxido Dismutase/genética , Superóxido Dismutase-1 , Transfecção , Transgenes , Expansão das Repetições de Trinucleotídeos , Ubiquitinas/metabolismo
14.
J Neuropathol Exp Neurol ; 62(9): 936-50, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14533783

RESUMO

The mechanisms whereby mutant gene expression triggers neurodegeneration are poorly understood but have generally been attributed to translated gene products. We now demonstrate direct neuropathic effects of untranslated RNA on cultured motor neurons. We show that expression of untranslated light neurofilament (NF-L) RNA sequence in the 3'UTR of an EGFP transgene (pEGFP/NF-L RNA) or in a separate expression vector (pRc/NF-L RNA) causes dose-dependent, neuron-specific motor neuron degeneration. Neither unfused EGFP protein (pEGFP/wt) nor EGFP-tagged NF-L protein (pEGFP/NF-L protein) has similar neuropathic effects. The findings are the first demonstration of a direct RNA-mediated neurotoxic effect. Moreover, the resulting neuropathological changes show that untranslated RNA can lead to early degeneration of neuritic processes and accumulations of ubiquitinated aggregates in the perikarya and nuclei of degenerating motor neurons. The latter findings are hallmark neuropathological features of neurodegenerative diseases and their occurrence as a result of altered RNA expression raises the prospects of an RNA-mediated component in the pathogenesis of neurodegenerative states.


Assuntos
Neurônios Motores/metabolismo , Degeneração Neural/metabolismo , Proteínas de Neurofilamentos/biossíntese , RNA/biossíntese , Ubiquitinas/biossíntese , Animais , Agregação Celular/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Camundongos , Neurônios Motores/patologia , Degeneração Neural/patologia , Proteínas de Neurofilamentos/genética , RNA/metabolismo , Medula Espinal/metabolismo , Medula Espinal/patologia , Ubiquitinas/genética
15.
Brain Res Mol Brain Res ; 117(1): 27-38, 2003 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-14499478

RESUMO

p190RhoGEF is a large multi-functional protein with guanine nucleotide exchange (GEF) activity. The C-terminal region of p190RhoGEF is a highly interactive domain that binds multiple factors, including proteins with anti-apoptotic activities. We now report that transfection of EGFP-tagged p190RhoGEF protects Neuro 2a cells from stress-induced apoptosis and that anti-apoptotic activity is localized to cytoplasmic retention sequences (CRS-1 and CRS-2) in the C-terminal region of p190RhoGEF. Cytoplasmic retention is conferred to an EGFP fluorescent marker when fused to either CRS-1 or CRS-2. Both cytoplasmic retention and anti-apoptotic activity are lost by deleting CRS-1 and CRS-2 in the p190RhoGEF sequence and can be recovered by restoring either CRS-1 or CRS-2 to the EGFP-tagged sequence. Since the CRS-1 and CRS-2 contain the JIP-1 and 14-3-3 binding sites, we propose that anti-apoptotic activity may be conferred by the binding of p190RhoGEF to JIP-1 or 14-3-3, possibly by altering their interactive properties or nucleocytoplasmic movements. Taken together, our findings support a model whereby multiple interactions of p190RhoGEF confer homeostatic properties to differentiated neurons and may link neuronal homeostasis to the regulation of NF-L expression.


Assuntos
Apoptose/fisiologia , Citoplasma/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Luminescentes/metabolismo , Proteínas Nucleares/metabolismo , Animais , Sítios de Ligação , Western Blotting , Proteínas de Transporte/metabolismo , Agregação Celular , Morte Celular , Linhagem Celular , Proteínas de Ligação a DNA , Proteínas Ativadoras de GTPase , Glutationa Transferase/genética , Proteínas de Fluorescência Verde , Marcação In Situ das Extremidades Cortadas/métodos , Microscopia Confocal , Fragmentos de Peptídeos/metabolismo , Testes de Precipitina/métodos , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras , Homologia de Sequência de Aminoácidos , Transfecção , Técnicas do Sistema de Duplo-Híbrido
16.
J Biol Chem ; 278(27): 24865-73, 2003 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-12702722

RESUMO

Focal adhesion kinase (FAK) is a protein-tyrosine kinase that associates with multiple cell surface receptors and signaling proteins through which it can modulate the activity of several intracellular signaling pathways. FAK activity can influence the formation of distinct actin cytoskeletal structures such as lamellipodia and stress fibers in part through effects on small Rho GTPases, although the molecular interconnections of these events are not well defined. Here, we report that FAK interacts with p190RhoGEF, a RhoA-specific GDP/GTP exchange factor, in neuronal cells and in brain tissue extracts by co-immunoprecipitation and co-localization analyses. Using a two-hybrid assay and deletion mutagenesis, the binding site of the FAK C-terminal focal adhesion targeting (FAT) domain was identified within the C-terminal coiled-coil domain of p190RhoGEF. Binding was independent of a LD-like binding motif within p190RhoGEF, yet FAK association was disrupted by a mutation (Leu-1034 to Ser) that weakens the helical bundle structure of the FAK FAT domain. Neuro-2a cell binding to laminin increased endogenous FAK and p190RhoGEF tyrosine phosphorylation, and co-transfection of a dominant-negative inhibitor of FAK activity, termed FRNK, inhibited lamininstimulated p190RhoGEF tyrosine phosphorylation and p21 RhoA GTP binding. Overexpression of FAK in Neuro-2a cells increased both endogenous p190RhoGEF tyrosine phosphorylation and RhoA activity, whereas these events were inhibited by FRNK co-expression. Because insulin-like growth factor 1 treatment of Neuro-2a cells increased FAK tyrosine phosphorylation and enhanced p190RhoGEF-mediated activation of RhoA, our results support the conclusion that FAK association with p190RhoGEF functions as a signaling pathway downstream of integrins and growth factor receptors to stimulate Rho activity.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Proteínas de Ligação a DNA , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Proteínas Ativadoras de GTPase , Camundongos , Ligação Proteica , Proteínas Repressoras , Células Tumorais Cultivadas , Proteína rhoA de Ligação ao GTP/metabolismo
17.
J Biol Chem ; 277(45): 42701-5, 2002 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-12215442

RESUMO

The enhancement of RNA-mediated motor neuron degeneration in transgenic mice by mutating a major mRNA instability determinant in a light neurofilament (NF-L) transgene implicates cognate RNA binding factors in the pathogenesis of motor neuron degeneration. p190RhoGEF is a neuron-enriched guanine exchange factor (GEF) that binds to the NF-L-destabilizing element, to c-Jun N-terminal kinase-interactive protein-1 (JIP-1), and to 14-3-3 and may link neurofilament expression to pathways affecting neuronal homeostasis. This study was undertaken to identify additional RNA species that bind p190RhoGEF and could affect interactions of the exchange factor with NF-L transcripts. The C-terminal domain of p190RhoGEF, containing the RNA-binding site, was expressed as a glutathione S-transferase fusion protein and was used as an affinity probe to isolate interactive RNAs in rat brain extracts. As expected, NF-L mRNA was identified as an RNA specie eluted from the affinity column. In addition, BC1 RNA was also found enriched in the bound RNA fraction. BC1 is a 152-nucleotide RNA that is highly expressed but untranslated in differentiated neurons. We show that BC1 and NF-L mRNA bind to a similar site in the C-terminal domain of p190RhoGEF, and their bindings to p190RhoGEF are readily cross-competed. Moreover, we identify a novel binding site in BC1 to account for its interaction with p190RhoGEF. The findings suggest a novel role of BC1 in differentiated neurons involving RNA-protein interactions of p190RhoGEF.


Assuntos
Encéfalo/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Filamentos Intermediários/metabolismo , Proteínas de Neurofilamentos/genética , Neurônios/fisiologia , Proteínas Nucleares/metabolismo , RNA Mensageiro/genética , RNA Citoplasmático Pequeno/genética , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Ligação a DNA , Proteínas Ativadoras de GTPase , Glutationa Transferase/genética , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Degeneração Neural/genética , Sondas RNA , RNA Mensageiro/metabolismo , RNA Citoplasmático Pequeno/química , RNA Citoplasmático Pequeno/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras , Transcrição Gênica
18.
J Neurosci ; 22(17): 7662-70, 2002 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-12196589

RESUMO

Studies of experimental motor neuron degeneration attributable to expression of neurofilament light chain (NF-L) transgenes have raised the possibility that the neuropathic effects result from overexpression of NF-L mRNA, independent of NF-L protein effects (Cañete-Soler et al., 1999). The present study was undertaken to test for an RNA-mediated pathogenesis. Transgenic mice were derived using either an enhanced green fluorescent protein reporter construct or modified chimeric constructs that differ only in their 3' untranslated regions (UTRs). Motor function and spinal cord histology were normal in mice expressing the unmodified reporter transgene. In mice expressing a chimeric transgene in which sequence of NF-L 3' UTR was inserted into the 3' UTR of the reporter transgene, we observed growth retardation and reduced kinetic activity during postnatal development. Older mice developed impairment of motor function and atrophy of nerve fibers in the ventral roots. A similar but more severe phenotype was observed when the chimeric transgene contained a 36 bp c-myc insert in an mRNA destabilizing element of the NF-L sequence. Our results suggest that neuropathic effects of overexpressing NF-L can occur at the level of transgene RNA and are mediated by sequences in the NF-L 3' UTR.


Assuntos
Regiões 3' não Traduzidas/genética , Transtornos do Crescimento/genética , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/metabolismo , Proteínas de Neurofilamentos/genética , Animais , Comportamento Animal , Peso Corporal/genética , Células COS , Genes Reporter , Genes myc/genética , Transtornos do Crescimento/patologia , Proteínas Luminescentes/genética , Camundongos , Camundongos Transgênicos , Atividade Motora/genética , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/patologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Mutação , Proteínas de Neurofilamentos/biossíntese , Proteínas de Neurofilamentos/farmacologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transgenes/fisiologia , Vacúolos/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...