Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Mol Neurosci ; 15: 841047, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35465088

RESUMO

Defective mitochondrial dynamics in axons have been linked to both developmental and late-onset neurological disorders. Axonal trafficking is in large part governed by the microtubule motors kinesin-1 and cytoplasmic dynein 1 (dynein). Dynein is the primary retrograde transport motor in axons, and mutations in dynein and many of its regulators also cause neurological diseases. Depletion of LIS1, famous for linking dynein deregulation to lissencephaly (smooth brain), in adult mice leads to severe neurological phenotypes, demonstrating post-developmental roles. LIS1 stimulates retrograde transport of acidic organelles in cultured adult rat dorsal root ganglion (DRG) axons but findings on its role in mitochondrial trafficking have been inconsistent and have not been reported for adult axons. Here we report that there is an increased number of mitochondria in cross-sections of sciatic nerve axons from adult LIS1+/- mice. This is probably related to reduced dynein activity as axons from adult rat nerves exposed to the dynein inhibitor, ciliobrevin D also had increased numbers of mitochondria. Moreover, LIS1 overexpression (OE) in cultured adult rat DRG axons stimulated retrograde mitochondrial transport while LIS1 knockdown (KD) or expression of a LIS1 dynein-binding mutant (LIS1-K147A) inhibited retrograde transport, as did KD of dynein heavy chain (DHC). These findings are consistent with our report on acidic organelles. However, KD of NDEL1, a LIS1 and dynein binding protein, or expression of a LIS1 NDEL1-binding mutant (LIS1-R212A) also dramatically impacted retrograde mitochondrial transport, which was not the case for acidic organelles. Manipulations that disrupted retrograde mitochondrial transport also increased the average length of axonal mitochondria, suggesting a role for dynein in fusion or fission events. Our data point to cargo specificity in NDEL1 function and raise the possibility that defects in the LIS1/NDEL1 dynein regulatory pathway could contribute to mitochondrial diseases with axonal pathologies.

2.
J Cell Biol ; 218(6): 1871-1890, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31068376

RESUMO

Inhibition of histone deacetylase 6 (HDAC6) was shown to support axon growth on the nonpermissive substrates myelin-associated glycoprotein (MAG) and chondroitin sulfate proteoglycans (CSPGs). Though HDAC6 deacetylates α-tubulin, we find that another HDAC6 substrate contributes to this axon growth failure. HDAC6 is known to impact transport of mitochondria, and we show that mitochondria accumulate in distal axons after HDAC6 inhibition. Miro and Milton proteins link mitochondria to motor proteins for axon transport. Exposing neurons to MAG and CSPGs decreases acetylation of Miro1 on Lysine 105 (K105) and decreases axonal mitochondrial transport. HDAC6 inhibition increases acetylated Miro1 in axons, and acetyl-mimetic Miro1 K105Q prevents CSPG-dependent decreases in mitochondrial transport and axon growth. MAG- and CSPG-dependent deacetylation of Miro1 requires RhoA/ROCK activation and downstream intracellular Ca2+ increase, and Miro1 K105Q prevents the decrease in axonal mitochondria seen with activated RhoA and elevated Ca2+ These data point to HDAC6-dependent deacetylation of Miro1 as a mediator of axon growth inhibition through decreased mitochondrial transport.


Assuntos
Desacetilase 6 de Histona/genética , Mitocôndrias/metabolismo , Neurônios/metabolismo , Proteínas rho de Ligação ao GTP/genética , Quinases Associadas a rho/genética , Acetilação/efeitos dos fármacos , Animais , Transporte Axonal/efeitos dos fármacos , Transporte Axonal/genética , Cálcio/metabolismo , Proteoglicanas de Sulfatos de Condroitina/farmacologia , Feminino , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica , Desacetilase 6 de Histona/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Glicoproteína Associada a Mielina/farmacologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
3.
Nat Commun ; 10(1): 228, 2019 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-30651536

RESUMO

Single-molecule cytoplasmic dynein function is well understood, but there are major gaps in mechanistic understanding of cellular dynein regulation. We reported a mode of dynein regulation, force adaptation, where lipid droplets adapt to opposition to motion by increasing the duration and magnitude of force production, and found LIS1 and NudEL to be essential. Adaptation reflects increasing NudEL-LIS1 utilization; here, we hypothesize that such increasing utilization reflects CDK5-mediated NudEL phosphorylation, which increases the dynein-NudEL interaction, and makes force adaptation possible. We report that CDK5, 14-3-3ε, and CDK5 cofactor KIAA0528 together promote NudEL phosphorylation and are essential for force adaptation. By studying the process in COS-1 cells lacking Tau, we avoid confounding neuronal effects of CDK5 on microtubules. Finally, we extend this in vivo regulatory pathway to lysosomes and mitochondria. Ultimately, we show that dynein force adaptation can control the severity of lysosomal tug-of-wars among other intracellular transport functions involving high force.


Assuntos
Proteínas 14-3-3/metabolismo , Quinase 5 Dependente de Ciclina/metabolismo , Dineínas do Citoplasma/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas 14-3-3/genética , Animais , Fenômenos Biomecânicos , Células COS , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Chlorocebus aethiops , Quinase 5 Dependente de Ciclina/genética , Gotículas Lipídicas/metabolismo , Lisossomos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Mitocôndrias/metabolismo , RNA Interferente Pequeno/metabolismo
4.
Mol Cell Proteomics ; 18(3): 504-519, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30587509

RESUMO

The fumarate ester dimethyl fumarate (DMF) has been introduced recently as a treatment for relapsing remitting multiple sclerosis (RRMS), a chronic inflammatory condition that results in neuronal demyelination and axonal loss. DMF is known to act by depleting intracellular glutathione and modifying thiols on Keap1 protein, resulting in the stabilization of the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. We have previously shown that DMF reacts with a wide range of protein thiols, suggesting that the complete mechanisms of action of DMF are unknown. Here, we investigated other intracellular thiol residues that may also be irreversibly modified by DMF in neurons and astrocytes. Using mass spectrometry, we identified 24 novel proteins that were modified by DMF in neurons and astrocytes, including cofilin-1, tubulin and collapsin response mediator protein 2 (CRMP2). Using an in vitro functional assay, we demonstrated that DMF-modified cofilin-1 loses its activity and generates less monomeric actin, potentially inhibiting its cytoskeletal remodeling activity, which could be beneficial in the modulation of myelination during RRMS. DMF modification of tubulin did not significantly impact axonal lysosomal trafficking. We found that the oxygen consumption rate of N1E-115 neurons and the levels of proteins related to mitochondrial energy production were only slightly affected by the highest doses of DMF, confirming that DMF treatment does not impair cellular respiratory function. In summary, our work provides new insights into the mechanisms supporting the neuroprotective and remyelination benefits associated with DMF treatment in addition to the antioxidant response by Nrf2.


Assuntos
Astrócitos/metabolismo , Cisteína/efeitos dos fármacos , Fumarato de Dimetilo/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Neurônios/metabolismo , Células 3T3-L1 , Animais , Astrócitos/citologia , Astrócitos/efeitos dos fármacos , Células Cultivadas , Cofilina 1/química , Cofilina 1/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular , Espectrometria de Massas , Camundongos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Ratos , Tubulina (Proteína)/química , Tubulina (Proteína)/metabolismo
5.
J Cell Sci ; 131(8)2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29654160

RESUMO

Localization and translation of mRNAs within different subcellular domains provides an important mechanism to spatially and temporally introduce new proteins in polarized cells. Neurons make use of this localized protein synthesis during initial growth, regeneration and functional maintenance of their axons. Although the first evidence for protein synthesis in axons dates back to 1960s, improved methodologies, including the ability to isolate axons to purity, highly sensitive RNA detection methods and imaging approaches, have shed new light on the complexity of the transcriptome of the axon and how it is regulated. Moreover, these efforts are now uncovering new roles for locally synthesized proteins in neurological diseases and injury responses. In this Cell Science at a Glance article and the accompanying poster, we provide an overview of how axonal mRNA transport and translation are regulated, and discuss their emerging links to neurological disorders and neural repair.


Assuntos
Axônios/metabolismo , Transporte de RNA/fisiologia , RNA Mensageiro/metabolismo
6.
eNeuro ; 5(1)2018.
Artigo em Inglês | MEDLINE | ID: mdl-29404402

RESUMO

LIS1 mutations cause lissencephaly (LIS), a severe developmental brain malformation. Much less is known about its role in the mature nervous system. LIS1 regulates the microtubule motor cytoplasmic dynein 1 (dynein), and as LIS1 and dynein are both expressed in the adult nervous system, Lis1 could potentially regulate dynein-dependent processes such as axonal transport. We therefore knocked out Lis1 in adult mice using tamoxifen-induced, Cre-ER-mediated recombination. When an actin promoter was used to drive Cre-ER expression (Act-Cre-ER), heterozygous Lis1 knockout (KO) caused no obvious change in viability or behavior, despite evidence of widespread recombination by a Cre reporter three weeks after tamoxifen exposure. In contrast, homozygous Lis1 KO caused the rapid onset of neurological symptoms in both male and female mice. One tamoxifen-dosing regimen caused prominent recombination in the midbrain/hindbrain, PNS, and cardiac/skeletal muscle within a week; these mice developed severe symptoms in that time frame and were killed. A different tamoxifen regimen resulted in delayed recombination in midbrain/hindbrain, but not in other tissues, and also delayed the onset of symptoms. This indicates that Lis1 loss in the midbrain/hindbrain causes the severe phenotype. In support of this, brainstem regions known to house cardiorespiratory centers showed signs of axonal dysfunction in KO animals. Transport defects, neurofilament (NF) alterations, and varicosities were observed in axons in cultured DRG neurons from KO animals. Because no symptoms were observed when a cardiac specific Cre-ER promoter was used, we propose a vital role for Lis1 in autonomic neurons and implicate defective axonal transport in the KO phenotype.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Transporte Axonal/fisiologia , Encéfalo/patologia , Células Cultivadas , Feminino , Gânglios Espinais/crescimento & desenvolvimento , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Masculino , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Doenças do Sistema Nervoso/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Fenótipo
7.
Mol Biol Cell ; 28(5): 587-599, 2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28057765

RESUMO

Diabetes is linked to an increased risk for colorectal cancer, but the mechanistic underpinnings of this clinically important effect are unclear. Here we describe an interaction between the microtubule motor cytoplasmic dynein, the adenomatous polyposis coli tumor suppressor protein (APC), and glycogen synthase kinase-3ß (GSK-3ß), which could shed light on this issue. GSK-3ß is perhaps best known for glycogen regulation, being inhibited downstream in an insulin-signaling pathway. However, the kinase is also important in many other processes. Mutations in APC that disrupt the regulation of ß-catenin by GSK-3ß cause colorectal cancer in humans. Of interest, both APC and GSK-3ß interact with microtubules and cellular membranes. We recently demonstrated that dynein is a GSK-3ß substrate and that inhibition of GSK-3ß promotes dynein-dependent transport. We now report that dynein stimulation in intestinal cells in response to acute insulin exposure (or GSK-3ß inhibition) is blocked by tumor-promoting isoforms of APC that reduce an interaction between wild-type APC and dynein. We propose that under normal conditions, insulin decreases dynein binding to APC to stimulate minus end-directed transport, which could modulate endocytic and secretory systems in intestinal cells. Mutations in APC likely impair the ability to respond appropriately to insulin signaling. This is exciting because it has the potential to be a contributing factor in the development of colorectal cancer in patients with diabetes.


Assuntos
Polipose Adenomatosa do Colo/metabolismo , Neoplasias Colorretais/metabolismo , Dineínas do Citoplasma/metabolismo , Insulina/metabolismo , Animais , Linhagem Celular , Citoplasma/metabolismo , Complicações do Diabetes/metabolismo , Feminino , Genes APC/fisiologia , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microtúbulos/metabolismo , Ligação Proteica , Transdução de Sinais , beta Catenina/metabolismo
8.
Traffic ; 16(9): 941-61, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26010407

RESUMO

Glycogen synthase kinase 3 (GSK-3) has been linked to regulation of kinesin-dependent axonal transport in squid and flies, and to indirect regulation of cytoplasmic dynein. We have now found evidence for direct regulation of dynein by mammalian GSK-3ß in both neurons and non-neuronal cells. GSK-3ß coprecipitates with and phosphorylates mammalian dynein. Phosphorylation of dynein intermediate chain (IC) reduces its interaction with Ndel1, a protein that contributes to dynein force generation. Two conserved residues, S87/T88 in IC-1B and S88/T89 in IC-2C, have been identified as GSK-3 targets by both mass spectrometry and site-directed mutagenesis. These sites are within an Ndel1-binding domain, and mutation of both sites alters the interaction of IC's with Ndel1. Dynein motility is stimulated by (i) pharmacological and genetic inhibition of GSK-3ß, (ii) an insulin-sensitizing agent (rosiglitazone) and (iii) manipulating an insulin response pathway that leads to GSK-3ß inactivation. Thus, our study connects a well-characterized insulin-signaling pathway directly to dynein stimulation via GSK-3 inhibition.


Assuntos
Dineínas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Chlorocebus aethiops , Citoplasma/metabolismo , Dineínas/química , Dineínas/genética , Quinase 3 da Glicogênio Sintase/genética , Humanos , Insulina/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Transporte Proteico , Sistemas do Segundo Mensageiro
9.
J Neurosci ; 31(47): 17207-19, 2011 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-22114287

RESUMO

Lissencephaly is a human developmental brain abnormality caused by LIS1 haploinsufficiency. This disorder is in large part attributed to altered mitosis and migration in the developing brain. LIS1 and an interacting protein, NDEL1, bind to cytoplasmic dynein, a microtubule motor protein. While the tripartite complex is clearly important for developmental events, we are intrigued by the fact that Lis1 and Ndel1 expression remain high in the adult mouse nervous system. Dynein plays a crucial role in retrograde axonal transport, a process that is used by mature neurons. Here, we monitored acidic organelles moving in axons of adult rat sensory neurons to determine whether Lis1 and Ndel1 contribute to axonal transport. Lis1 RNAi significantly reduced axon transport of these organelles. Ndel1 RNAi had little impact, but combined Lis1 and Ndel1 RNAi caused a more severe phenotype than Lis1 RNAi alone, essentially shutting down transport. Lis1 overexpression stimulated retrograde transport, while a Lis1 dynein-binding mutant severely disrupted transport. Overexpression of Ndel1 or a Lis1 Ndel1-binding mutant only mildly perturbed transport. However, expressing a mutant Ndel1 lacking key phosphorylation sites shut down transport completely, as did a dominant-negative Cdk5 construct. We propose that, in axons, unphosphorylated Ndel1 inhibits the capacity of dynein to transport acidic organelles. Phosphorylation of Ndel1 by Cdk5 not only reduces this inhibition but also allows Lis1 to further stimulate the cargo transport capacity of dynein. Our data raise the possibility that defects in a Lis1/Ndel1 regulatory switch could contribute to neurodegenerative diseases linked to axonal pathology in adults.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Axônios/metabolismo , Proteínas de Transporte/metabolismo , Quinase 5 Dependente de Ciclina/fisiologia , Dineínas/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Organelas/metabolismo , Fatores Etários , Animais , Axônios/fisiologia , Transporte Biológico/fisiologia , Células COS , Células Cultivadas , Chlorocebus aethiops , Dineínas/antagonistas & inibidores , Dineínas/metabolismo , Masculino , Camundongos , Organelas/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley
10.
J Cell Biol ; 182(6): 1063-71, 2008 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-18809722

RESUMO

Lis1 and Ndel1 are essential for animal development. They interact directly with one another and with cytoplasmic dynein. The developing brain is especially sensitive to reduced Lis1 or Ndel1 levels, as both proteins influence spindle orientation, neural cell fate decisions, and neuronal migration. We report here that Lis1 and Ndel1 reduction in a mitotic cell line impairs prophase nuclear envelope (NE) invagination (PNEI). This dynein-dependent process facilitates NE breakdown (NEBD) and occurs before the establishment of the bipolar spindle. Ndel1 phosphorylation is important for this function, regulating binding to both Lis1 and dynein. Prophase cells in the ventricular zone (VZ) of embryonic day 13.5 Lis1(+/-) mouse brains show reduced PNEI, and the ratio of prophase to prometaphase cells is increased, suggesting an NEBD delay. Moreover, prophase cells in the VZ contain elevated levels of Ndel1 phosphorylated at a key cdk5 site. Our data suggest that a delay in NEBD in the VZ could contribute to developmental defects associated with Lis1-Ndel1 disruption.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios , Membrana Nuclear/metabolismo , Células-Tronco , 1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Animais , Células COS , Proteínas de Transporte/genética , Ciclo Celular/fisiologia , Linhagem Celular , Chlorocebus aethiops , Complexo Dinactina , Dineínas/metabolismo , Feminino , Humanos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Nocodazol/metabolismo , Fosforilação , Ligação Proteica , Ratos , Células-Tronco/citologia , Células-Tronco/metabolismo , Moduladores de Tubulina/metabolismo
11.
Dev Neurosci ; 30(1-3): 157-70, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18075263

RESUMO

Hemizygous Lis1 mutations cause type 1 lissencephaly, a neuronal migration disorder in humans. The Lis1+/- mouse is a model for lissencephaly; mice exhibit neuronal migration defects but are viable and fertile. On an inbred genetic background, 20% of Lis1+/- mice develop hydrocephalus and die prematurely. Lis1 functions with the microtubule motor cytoplasmic dynein. Because dynactin, a dynein regulator, interacts with end-binding protein 1 (EB1) and beta-catenin, two known binding partners of the adenomatous polyposis coli (APC) protein, we looked for a genetic interaction between Lis1 and APC. Mice with a heterozygous truncating mutation in APC (Min mutation) do not exhibit neuronal migration defects or develop hydrocephalus. However, the presence of the APC mutation increases the migration deficit and the incidence of hydrocephalus in Lis1+/- animals. Lis1 and dynein distribution is altered in cells derived from Min mice, and both Lis1 and dynein interact with the C terminus of APC in vitro. Together, our findings point to a previously unknown interaction between APC and Lis1 during mammalian brain development.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/genética , Proteína da Polipose Adenomatosa do Colo/genética , Predisposição Genética para Doença/genética , Hidrocefalia/genética , Lisencefalia/genética , Proteínas Associadas aos Microtúbulos/genética , Mutação/genética , Animais , Animais Recém-Nascidos , Encéfalo/anormalidades , Encéfalo/citologia , Encéfalo/metabolismo , Movimento Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Dineínas/genética , Dineínas/metabolismo , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Heterozigoto , Humanos , Hidrocefalia/metabolismo , Hidrocefalia/fisiopatologia , Lisencefalia/metabolismo , Lisencefalia/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Estrutura Terciária de Proteína/genética
12.
J Cell Biol ; 178(6): 965-80, 2007 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-17785519

RESUMO

Subcellular regulation of protein synthesis requires the correct localization of messenger RNAs (mRNAs) within the cell. In this study, we investigate whether the axonal localization of neuronal mRNAs is regulated by extracellular stimuli. By profiling axonal levels of 50 mRNAs detected in regenerating adult sensory axons, we show that neurotrophins can increase and decrease levels of axonal mRNAs. Neurotrophins (nerve growth factor, brain-derived neurotrophic factor, and neurotrophin-3) regulate axonal mRNA levels and use distinct downstream signals to localize individual mRNAs. However, myelin-associated glycoprotein and semaphorin 3A regulate axonal levels of different mRNAs and elicit the opposite effect on axonal mRNA levels from those observed with neurotrophins. The axonal mRNAs accumulate at or are depleted from points of ligand stimulation along the axons. The translation product of a chimeric green fluorescent protein-beta-actin mRNA showed similar accumulation or depletion adjacent to stimuli that increase or decrease axonal levels of endogenous beta-actin mRNA. Thus, extracellular ligands can regulate protein generation within subcellular regions by specifically altering the localized levels of particular mRNAs.


Assuntos
Neurônios/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais , Actinas/genética , Actinas/metabolismo , Animais , Axônios/metabolismo , Células Cultivadas , Proteínas de Fluorescência Verde/genética , Hibridização In Situ , Glicoproteína Associada a Mielina/metabolismo , Fatores de Crescimento Neural/fisiologia , Regeneração Nervosa , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Semaforina-3A/metabolismo
13.
J Neurosci ; 26(7): 2132-9, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16481446

RESUMO

Mutations in Lis1 cause classical lissencephaly, a developmental brain abnormality characterized by defects in neuronal positioning. Over the last decade, a clear link has been forged between Lis1 and the microtubule motor cytoplasmic dynein. Substantial evidence indicates that Lis1 functions in a highly conserved pathway with dynein to regulate neuronal migration and other motile events. Yeast two-hybrid studies predict that Lis1 binds directly to dynein heavy chains (Sasaki et al., 2000; Tai et al., 2002), but the mechanistic significance of this interaction is not well understood. We now report that recombinant Lis1 binds to native brain dynein and significantly increases the microtubule-stimulated enzymatic activity of dynein in vitro. Lis1 does this without increasing the proportion of dynein that binds to microtubules, indicating that Lis1 influences enzymatic activity rather than microtubule association. Dynein stimulation in vitro is not a generic feature of microtubule-associated proteins, because tau did not stimulate dynein. To our knowledge, this is the first indication that Lis1 or any other factor directly modulates the enzymatic activity of cytoplasmic dynein. Lis1 must be able to homodimerize to stimulate dynein, because a C-terminal fragment (containing the dynein interaction site but missing the self-association domain) was unable to stimulate dynein. Binding and colocalization studies indicate that Lis1 does not interact with all dynein complexes found in the brain. We propose a model in which Lis1 stimulates the activity of a subset of motors, which could be particularly important during neuronal migration and long-distance axonal transport.


Assuntos
Dineínas/metabolismo , Proteínas Associadas aos Microtúbulos/fisiologia , Neurônios/fisiologia , 1-Alquil-2-acetilglicerofosfocolina Esterase , Processamento Alternativo , Animais , Transporte Axonal/fisiologia , Encéfalo/fisiologia , Linhagem Celular , Movimento Celular/fisiologia , Variação Genética , Camundongos , Proteínas Recombinantes/metabolismo , Spodoptera , Transfecção
14.
Trends Cell Biol ; 12(1): 28-36, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11854007

RESUMO

Cdk5, a serine/threonine kinase in the cyclin-dependent kinase (Cdk) family, is an important regulator of neuronal positioning during brain development. Cdk5 might also play a role in synaptogenesis and neurotransmission. Loss of Cdk5 in mice is perinatal lethal, and overactive Cdk5 induces apoptosis in cultured cells, indicating that strict regulation of kinase activity is crucial. Indeed, activity depends on the stability of activating partners, subcellular localization and the phosphorylation state of the enzyme itself. Deregulated kinase activity has been linked to neurodegenerative diseases such as Alzheimer's disease (AD) and amyotrophic lateral sclerosis (ALS). This review focuses on links between Cdk5 activity and components of cytoskeletal, membrane and adhesion systems that allow us to postulate a role for Cdk5 in directing intracellular traffic in neurons.


Assuntos
Quinases Ciclina-Dependentes/fisiologia , Citoesqueleto/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/fisiologia , Transdução de Sinais/fisiologia , Animais , Transporte Biológico Ativo/fisiologia , Quinase 5 Dependente de Ciclina , Quinases Ciclina-Dependentes/metabolismo , Humanos , Membranas/fisiologia , Neurotransmissores/fisiologia , Transporte Proteico/fisiologia , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...